US20110218186A1 - Spirocyclic heterocyclic derivatives and methods of their use - Google Patents

Spirocyclic heterocyclic derivatives and methods of their use Download PDF

Info

Publication number
US20110218186A1
US20110218186A1 US13/097,940 US201113097940A US2011218186A1 US 20110218186 A1 US20110218186 A1 US 20110218186A1 US 201113097940 A US201113097940 A US 201113097940A US 2011218186 A1 US2011218186 A1 US 2011218186A1
Authority
US
United States
Prior art keywords
alkyl
compound
compounds
derivative
pain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/097,940
Inventor
Roland E. Dolle
Bertrand Le Bourdonnec
Guo-Hua Chu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Adolor Corp
Original Assignee
Adolor Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adolor Corp filed Critical Adolor Corp
Priority to US13/097,940 priority Critical patent/US20110218186A1/en
Publication of US20110218186A1 publication Critical patent/US20110218186A1/en
Assigned to ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT reassignment ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT SECURITY AGREEMENT Assignors: ADOLOR CORPORATION, CALIXA THERAPEUTICS, INC., CUBIST PHARMACEUTICALS HOLDINGS, INC., CUBIST PHARMACEUTICALS U.S., CUBIST PHARMACEUTICALS, INC.
Assigned to CUBIST PHARMACEUTICALS, INC., CALIXA THERAPEUTICS, INC., ADOLOR CORPORATION reassignment CUBIST PHARMACEUTICALS, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/20Spiro-condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/14Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/96Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings spiro-condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/10Spiro-condensed systems

Definitions

  • the invention relates to spirocyclic heterocyclic derivatives (including derivatives of spiro(2H-1-benzopyran-2,4′-piperidines), pharmaceutical compositions containing these compounds, and methods for their pharmaceutical use.
  • the spirocyclic heterocyclic derivatives are ligands of the ⁇ opioid receptor and are useful, inter alia, for treating and/or preventing pain, anxiety, gastrointestinal disorders, and other ⁇ opioid receptor-mediated conditions.
  • ⁇ , ⁇ and ⁇ There are at least three different opioid receptors ( ⁇ , ⁇ and ⁇ ) that are present in both central and peripheral nervous systems of many species, including humans. Lord, J. A. H., et al., Nature, 1977, 267, 495. Activation of the ⁇ opioid receptors induces analgesia in various animal models. Moulin, et al., Pain, 1985, 23, 213. Some work suggests that the analgesics working at ⁇ opioid receptors do not have the attendant side effects associated with ⁇ and ⁇ opioid receptor activation. Galligan, et al., J. Pharm. Exp. Ther., 1985, 229, 641. The ⁇ opioid receptor has also been identified as having a role in circulatory systems.
  • Ligands for the ⁇ receptor have also been shown to possess immunomodulatory activities. Dondio, et al., Exp. Opin. Ther. Patents, 1997, 10, 1075. Further, selective ⁇ opioid receptor agonists have been shown to promote organ and cell survival. Su, T-P, Journal of Biomedical Science, 2000, 9(3), 195-199. Ligands for the ⁇ opioid receptor may therefore find potential use as analgesics, as antihypertensive agents, as immunomodulatory agents, and/or agents for the treatment of cardiac disorders.
  • the invention is directed to compounds of formula XIV:
  • the invention is directed to compounds of formula XVII:
  • the invention is directed to compounds of formula XX:
  • the compound of formula XX is other than 4-[(4-N,N-diethylaminocarbonyl)phenyl]-spiro[2H,1-benzopyran-2,3′-pyrrolidine].
  • the invention is directed to compounds of formula XXII:
  • the invention is directed to compounds of formula XXV:
  • the invention is directed to compounds of formula XXVII:
  • the invention is directed to compounds of formula XXVIII:
  • the invention is directed to compounds of formula XXIX:
  • the invention is directed to compounds of formula XXX:
  • the aryl ring of J 2 is substituted with at least one haloalkoxy.
  • the invention is directed to compounds of formula XXXII:
  • a 2 and B 2 are each H.
  • the invention is directed to compounds of formula XXXIII:
  • the invention is directed to compounds of formula XXXIV:
  • compositions comprising:
  • a pharmaceutically acceptable carrier and a compound as described herein including, for example, a compound of formula XIV, XVII, XX, XXII, XXV, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXII, XXXIII, and/or XXXIV.
  • the invention is directed to methods of binding opioid receptors in a patient in need thereof, comprising the step of:
  • a compound as described herein including, for example, a compound of formula XIV, XVII, XX, XXII, XXV, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXII, XXXIII, and/or XXXIV.
  • the invention relates to spirocyclic heterocyclic derivatives, pharmaceutical compositions containing these compounds, and methods for their pharmaceutical use.
  • This invention is related by subject matter to co-pending U.S. application Ser. No. 12/787,906, filed May 26, 2010, application Ser. No. 12/718,439, filed Mar. 5, 2010, application Ser. No. 12/483,781, filed Jun. 12, 2009, application Ser. No. 11/960,845, filed Dec. 20, 2007, now U.S. Pat. No. 7,638,527, application Ser. No. 10/957,554, filed Oct. 1, 2004, now U.S. Pat. No. 7,338,962, and Provisional Application Ser. No. 60/507,864, filed Oct. 1, 2003, the disclosures of which are hereby incorporated herein by reference, in their entireties.
  • the spirocyclic heterocyclic derivatives are ligands of the ⁇ opioid receptor and may be useful, inter alia, in methods for treating and/or preventing diseases and conditions that may be mediated or modulated by the ⁇ opioid receptor including, for example, pain, gastrointestinal disorders, urogenital tract disorders including incontinence and overactive bladder, immunomodulatory disorders, inflammatory disorders, respiratory function disorders, anxiety, mood disorders, stress-related disorders, attention deficit hyperactivity disorders, sympathetic nervous system disorders, depression, tussis, motor disorders, traumatic injuries, especially to the central nervous system, stroke, cardiac arrhythmias, glaucoma, sexual dysfunctions, shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, rejections in organ transplants and skin grafts, and substance addiction.
  • diseases and conditions that may be mediated or modulated by the ⁇ opioid receptor
  • the spirocyclic heterocyclic derivatives are ligands of the ⁇ opioid receptor and may be useful in, inter alia, methods for improving organ and cell survival, methods for providing cardioprotection following myocardial infarction, methods for reducing the need for anesthesia, methods for producing and/or maintaining an anaesthetic state, and methods of detecting, imaging or monitoring degeneration or dysfunction of opioid receptors in a patient.
  • Alkyl refers to an optionally substituted, saturated straight, or branched, hydrocarbon having from about 1 to about 20 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), preferably from about 1 to about 8 carbon atoms, herein referred to as “lower alkyl,” more preferably from about 1 to about 6, still more preferably from about 1 to about 4, with from about 2 to about 3 being most preferred.
  • the alkyl group more preferably, has one carbon atom.
  • Alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, n-pentyl, cyclopentyl, isopentyl, neopentyl, n-hexyl, isohexyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl.
  • alkylene refers to an optionally substituted bivalent alkyl radical having the general formula —(CH 2 ) n —, where n is 1 to 10, preferably 1 to 6, with 1 to 4 being most preferred. In alternative embodiments, n is preferably 4 to 6. Non-limiting examples include methylene, dimethylene, trimethylene, tetramethylene, pentamethylene, and hexamethylene.
  • Cycloalkyl refers to an optionally substituted alkyl group having one or more rings in their structures and having from about 3 to about 20 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 3 to about 10 carbon atoms being preferred. Multi-ring structures may be bridged or fused ring structures.
  • Cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl, 2-[4-isopropyl-1-methyl-7-oxa-bicyclo[2.2.1]heptanyl], 2-[1,2,3,4-tetrahydro-naphthalenyl], and adamantyl.
  • Alkylcycloalkyl refers to an optionally substituted ring system comprising a cycloalkyl group having one or more alkyl substituents, wherein cycloalkyl and alkyl are each as previously defined.
  • exemplary alkylcycloalkyl groups include, for example, 2-methylcyclohexyl, 3,3-dimethylcyclopentyl, trans-2,3-dimethylcyclooctyl, and 4-methyldecahydronaphthalenyl.
  • Heterocycloalkyl refers to an optionally substituted ring system composed of a cycloalkyl radical wherein in at least one of the rings, one or more of the carbon atom ring members is independently replaced by a heteroatom group selected from the group consisting of O, S, N, and NH, wherein cycloalkyl is as previously defined. Heterocycloalkyl ring systems having a total of from about 5 to about 14 carbon atom ring members and heteroatom ring members (and all combinations and subcombinations of ranges and specific numbers of carbon and heteroatom ring members) are preferred. In other preferred embodiments, the heterocyclic groups may be fused to one or more aromatic rings.
  • heterocycloalkyl groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, piperazinyl, morpholinyl, piperadinyl, decahydroquinolyl, octahydrochromenyl, octahydro-cyclopenta[c]pyranyl, 1,2,3,4,-tetrahydroquinolyl, octahydro-[2]pyrindinyl, decahydro-cycloocta[c]furanyl, tetrahydroquinolyl, and imidazolidinyl.
  • Alkylheterocycloalkyl refers to an optionally substituted ring system comprising a heterocycloalkyl group having one or more alkyl substituents, wherein heterocycloalkyl and alkyl are each as previously defined.
  • exemplary alkylheterocycloalkyl groups include, for example, 2-methylpiperidinyl, 3,3-dimethylpyrrolidinyl, trans-2,3-dimethylmorpholinyl, and 4-methyldecahydroquinolinyl.
  • Alkenyl refers to an optionally substituted alkyl group having from about 2 to about 10 carbon atoms and one or more double bonds (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), wherein alkyl is as previously defined.
  • Alkynyl refers to an optionally substituted alkyl group having from about 2 to about 10 carbon atoms and one or more triple bonds (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), wherein alkyl is as previously defined.
  • Aryl refers to an optionally substituted, mono-, di-, tri-, or other multicyclic aromatic ring system having from about 5 to about 50 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 6 to about 10 carbons being preferred.
  • Non-limiting examples include, for example, phenyl, naphthyl, anthracenyl, and phenanthrenyl.
  • Alkyl refers to an optionally substituted moiety composed of an alkyl radical bearing an aryl substituent and having from about 6 to about 50 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 6 to about 10 carbon atoms being preferred.
  • Non-limiting examples include, for example, benzyl, diphenylmethyl, triphenylmethyl, phenylethyl, and diphenylethyl.
  • Halo refers to a fluoro, chloro, bromo, or iodo moiety, preferably fluoro.
  • Heteroaryl refers to an optionally substituted aryl ring system wherein in at least one of the rings, one or more of the carbon atom ring members is independently replaced by a heteroatom group selected from the group consisting of S, O, N, and NH, wherein aryl is as previously defined. Heteroaryl groups having a total of from about 5 to about 14 carbon atom ring members and heteroatom ring members (and all combinations and subcombinations of ranges and specific numbers of carbon and heteroatom ring members) are preferred.
  • heteroaryl groups include, but are not limited to, pyrryl, furyl, pyridyl, 1,2,4-thiadiazolyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, tetrazolyl, pyrazinyl, pyrimidyl, quinolyl, isoquinolyl, thiophenyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, purinyl, carbazolyl, benzimidazolyl, and isoxazolyl.
  • Heteroaryl may be attached via a carbon or a heteroatom to the rest of the molecule.
  • Heteroarylalkyl and “heteroaralkyl” each refers to an optionally substituted, heteroaryl substituted alkyl radical where heteroaryl and alkyl are as previously defined
  • Non-limiting examples include, for example, 2-(1H-pyrrol-3-yl)ethyl, 3-pyridylmethyl, 5-(2H-tetrazolyl)methyl, and 3-(pyrimidin-2-yl)-2-methylcyclopentanyl.
  • Perhaloalkyl refers to an alkyl group, wherein two or more hydrogen atoms are replaced by halo (F, Cl, Br, I) atoms, and alkyl is as previously defined.
  • exemplary perhaloalkyl groups include, for example, perhalomethyl, such as perfluoromethyl and difluoromethyl.
  • Alkoxy and “alkoxyl” refer to an optionally substituted alkyl-O— group wherein alkyl is as previously defined.
  • exemplary alkoxy and alkoxyl groups include, for example, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, and heptoxy.
  • Alkenyloxy refers to an optionally substituted alkenyl-O— group wherein alkenyl is as previously defined.
  • exemplary alkenyloxy and alkenyloxyl groups include, for example, allyloxy, butenyloxy, heptenyloxy, 2-methyl-3-buten-1-yloxy, and 2,2-dimethylallyloxy.
  • Alkynyloxy refers to an optionally substituted alkynyl-O— group wherein alkynyl is as previously defined.
  • exemplary alkynyloxy and alkynyloxyl groups include, for example, propargyloxy, butynyloxy, heptynyloxy, 2-methyl-3-butyn-1-yloxy, and 2,2-dimethylpropargyloxy.
  • Aryloxy and “aryloxyl” refer to an optionally substituted aryl-O— group wherein aryl is as previously defined.
  • Exemplary aryloxy and aryloxyl groups include, for example, phenoxy and naphthoxy.
  • aralkoxy and aralkoxyl refer to an optionally substituted aralkyl-O-group wherein aralkyl is as previously defined.
  • exemplary aralkoxy and aralkoxyl groups include, for example, benzyloxy, 1-phenylethoxy, 2-phenylethoxy, and 3-naphthylheptoxy.
  • Cycloalkoxy refers to an optionally substituted cycloalkyl-O— group wherein cycloalkyl is as previously defined.
  • exemplary cycloalkoxy groups include, for example, cyclopropanoxy, cyclobutanoxy, cyclopentanoxy, cyclohexanoxy, and cycloheptanoxy.
  • Heteroaryloxy refers to an optionally substituted heteroaryl-O— group wherein heteroaryl is as previously defined.
  • exemplary heteroaryloxy groups include, but are not limited to, pyrryloxy, furyloxyl, pyridyloxy, 1,2,4-thiadiazolyloxy, pyrimidyloxy, thienyloxy, isothiazolyloxy, imidazolyloxy, tetrazolyloxy, pyrazinyloxy, pyrimidyloxy, quinolyloxy, isoquinolyloxy, thiophenyloxy, benzothienyloxy, isobenzofuryloxy, pyrazolyloxy, indolyloxy, purinyloxy, carbazolyloxy, benzimidazolyloxy, and isoxazolyloxy.
  • Heteroaralkoxy refers to an optionally substituted heteroarylalkyl-O-group wherein heteroarylalkyl is as previously defined.
  • exemplary heteroaralkoxy groups include, but are not limited to, pyrrylethyloxy, furylethyloxy, pyridylmethyloxy, 1,2,4-thiadiazolylpropyloxy, pyrimidylmethyloxy, thienylethyloxy, isothiazolylbutyloxy, and imidazolyl-2-methylpropyloxy.
  • Heterocycloalkylaryl refers to an optionally substituted ring system composed of an aryl radical bearing a heterocycloalkyl substituent wherein heterocycloalkyl and aryl are as previously defined.
  • exemplary heterocycloalkylaryl groups include, but are not limited to, morpholinylphenyl, piperidinylnaphthyl, piperidinylphenyl, tetrahydrofuranylphenyl, and pyrrolidinylphenyl.
  • Alkylheteroaryl refers to an optionally substituted ring system composed of a heteroaryl radical bearing an alkyl substituent wherein heteroaryl and alkyl are as previously defined.
  • exemplary alkylheteroaryl groups include, but are not limited to, methylpyrryl, ethylfuryl, 2,3-dimethylpyridyl, N-methyl-1,2,4-thiadiazolyl, propylpyrimidyl, 2-butylthienyl, methylisothiazolyl, 2-ethylimidazolyl, butyltetrazolyl, 5-ethylbenzothienyl, and N-methylindolyl.
  • Alkyheteroaryl groups may be attached via a carbon or a heteroatom to the rest of the molecule.
  • Heteroarylaryl refers to an optionally substituted ring system composed of an aryl radical bearing a heteroaryl substituent wherein heteroaryl and aryl are as previously defined.
  • exemplary heteroarylaryl groups include, but are not limited to, pyrrylphenyl, furylnaphthyl, pyridylphenyl, 1,2,4-thiadiazolylnaphthyl, pyrimidylphenyl, thienylphenyl, isothiazolylnaphthyl, imidazolylphenyl, tetrazolylphenyl, pyrazinylnaphthyl, pyrimidylphenyl, quinolylphenyl, isoquinolylnaphthyl, thiophenylphenyl, benzothienylphenyl, isobenzofurylnaphthyl, pyrazolylphenyl, indolyln
  • Alkylheteroarylaryl refers to an optionally substituted ring system composed of an aryl radical bearing an alkylheteroaryl substituent and have from about 12 to about 50 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 12 to about 30 carbon atoms being preferred wherein aryl and alkylheteroaryl are as previously defined.
  • Exemplary heteroarylaryl groups include, but are not limited to, methylpyrrylphenyl, ethylfurylnaphthyl, methylethylpyridylphenyl, dimethylethylpyrimidylphenyl, and dimethylthienylphenyl.
  • substituted chemical moieties include one or more substituents that replace hydrogen.
  • substituents include, for example, halo (e.g., F, Cl, Br, I), alkyl, cycloalkyl, alkylcycloalkyl, alkenyl, alkynyl, aralkyl, aryl, heteroaryl, heteroaralkyl, spiroalkyl, heterocycloalkyl, hydroxyl (—OH), oxo ( ⁇ O), alkoxyl, aryloxyl, aralkoxyl, nitro (—NO 2 ), cyano (—CN), amino (—NH 2 ), —N-substituted amino (—NHR′′), -N,N-disubstituted amino (—N(R′′)R′′), carboxyl (—COOH), —C( ⁇ O)R′′, —OR′′, —C( ⁇ O)OR′′, —C( ⁇ O)NHSO 2 R′′, —OR
  • Aryl substituents may also include (CH 2 ) p SO 2 NR′′(CH 2 ) q and (CH 2 ) p CO 2 NR′′(CH 2 ) q , where p and q are independently integers from 0 to 3, where the methylene units are attached in a 1,2 arrangement yielding substituted aryls of the type:
  • each moiety R′′ can be, independently, any of H, alkyl, cycloalkyl, alkenyl, aryl, aralkyl, heteroaryl, or heterocycloalkyl, or when (R′′(R′′)) is attached to a nitrogen atom, R′′ and R′′ can be taken together to form a 4- to 8-membered nitrogen heterocycloalkyl ring, wherein said heterocycloalkyl ring is optionally interrupted by one or more additional —O—, —S—, —SO, —SO 2 —, —NH—, —N(alkyl)-, or —N(aryl)- groups, for example.
  • an “*” denotes the presence of a non-racemic stereoisomeric center in a molecule, wherein one stereoisomeric form (R or S) predominates, but for which the absolute configuration at this center has not been conclusively established. This is equivalently expressed by saying that the molecule's configuration at the asterisked carbon atom is either greater than 50% R or greater than 50% S. More preferably the compound, or its stereoisomeric center, is “substantially enriched”, and even more preferably is substantially enantiomerically pure.
  • the compound is “substantially enantiomerically pure”, that is, at least about 97.5%, more preferably about 99%, even more preferably about 99.5% of one stereoisomeric forms predominates.
  • a compound having one stereoisomeric center may be represented by one of two stereoisomeric forms (R or S), differing only in the spatial arrangement of atoms about a single carbon atom.
  • the “*” denotes non-equal amounts of the two isomers.
  • each center denoted by an asterisk is evaluated individually.
  • a predominance of one stereoisomeric form (R or S) occurring at least one center is considered non-racemic within the definition herein provided.
  • the range of possible non-racemic compounds extends from the point at which a stereoisomeric form predominates at a single chiral center and includes all combinations and subcombinations up to and including the compound wherein all stereoisomeric centers in the compound are each individually R or S.
  • Use of the “*” can be expressed, for example in a compound's identification number such as 4*, and indicates that the stereochemical configuration of at least one chiral center of the identified compound has not been established.
  • the specific center is identified within a structure by placing the “*” adjacent the chiral center in question, such as, for example, in the structure below.
  • the first pair of enantiomers having two chiral centers may have the configurations, for example, (R,R) and (S,S).
  • the second pair then have configurations, for example, (R,S) and (S,R).
  • the asterisks may indicate that the enantiomers have been enriched (partially resolved) or preferably fully resolved, into the individual enantiomers.
  • Ligand or “modulator” refers to a compound that binds to a receptor to form a complex, and includes, agonists, partial agonists, antagonists and inverse agonists.
  • Antagonist refers to a compound that may bind to a receptor to form a complex that may elicit a full pharmacological response, which is typically peculiar to the nature of the receptor involved and which may alter the equilibrium between inactive and active receptor.
  • Partial agonist refers to a compound that may bind to a receptor to form a complex that may elicit only a proportion of the full pharmacological response, typically peculiar to the nature of the receptor involved, even if a high proportion of the receptors are occupied by the compound.
  • Antagonist refers to a compound that may bind to a receptor to form a complex that may not elicit any response, typically in the same manner as an unoccupied receptor, and which preferably does not alter the equilibrium between inactive and active receptor.
  • “Inverse agonist” refers to a compound that may bind to a receptor to form a complex that may preferentially stabilize the inactive conformation of the receptor.
  • Prodrug refers to compounds specifically designed to maximize the amount of active species that reaches the desired site of reaction that are themselves typically inactive or minimally active for the activity desired, but through biotransformation are converted into biologically active metabolites.
  • Stepoisomers refers to compounds that have identical chemical constitution, but differ as regards the arrangement of the atoms or groups in space.
  • N-oxide refers to compounds wherein the basic nitrogen atom of either a heteroaromatic ring or tertiary amine is oxidized to give a quaternary nitrogen bearing a positive formal charge and an attached oxygen atom bearing a negative formal charge.
  • “Hydrate” refers to a compound as described herein which is associated with water in the molecular form, i.e., in which the H—OH bond is not split, and may be represented, for example, by the formula R.H2O, where R is a compound as described herein.
  • a given compound may form more than one hydrate including, for example, monohydrates (R.H2O), dihydrates (R.2H2O), trihydrates (R.3H2O), and the like.
  • haloalkoxy refers to an alkoxy group, wherein one, preferably two or more, hydrogen(s) of the alkyl moiety of said alkoxy are replaced by halo atoms, and alkoxy, alkyl, and halo are each as previously defined.
  • solvent refers to a compound of the present invention which is associated with solvent in the molecular form, i.e., in which the solvent is coordinatively bound, and may be represented, for example, by the formula R.(solvent), where R is a compound of the invention.
  • a given compound may form more than one solvate including, for example, monosolvates (R.(solvent)) or polysolvates (R.n(solvent)) wherein n is an integer >1) including, for example, disolvates (R.2(solvent)), trisolvates (R.3(solvent)), and the like, or hemisolvates, such as, for example, R.n/2(solvent), R.n/3(solvent), R.n/4(solvent) and the like wherein n is an integer.
  • Solvents herein include mixed solvents, for example, methanol/water, and as such, the solvates may incorporate one or more solvents within the solvate.
  • “Acid hydrate” refers to a complex that may be formed through association of a compound having one or more base moieties with at least one compound having one or more acid moieties or through association of a compound having one or more acid moieties with at least one compound having one or more base moieties, said complex being further associated with water molecules so as to form a hydrate, wherein said hydrate is as previously defined and R represents the complex herein described above.
  • “Pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic,
  • physiologically acceptable salts are prepared by methods known in the art, e.g., by dissolving the free amine bases with an excess of the acid in aqueous alcohol, or neutralizing a free carboxylic acid with an alkali metal base such as a hydroxide, or with an amine.
  • Certain acidic or basic compounds as described herein may exist as zwitterions. All forms of the compounds, including free acid, free base and zwitterions, are contemplated to be within the scope of the present invention. It is well known in the art that compounds containing both basic nitrogen atom and acidic groups often exist in equilibrium with their zwitterionic forms. Thus, any of the compounds described herein throughout that contain, for example, both basic nitrogen and acidic groups, also include reference to their corresponding zwitterions.
  • Effective amount refers to an amount of a compound as described herein that may be therapeutically effective to inhibit, prevent or treat the symptoms of particular disease, disorder, condition, or side effect.
  • diseases, disorders, conditions, and side effects include, but are not limited to, those pathological conditions associated with the binding of ⁇ opioid receptor (for example, in connection with the treatment and/or prevention of pain), wherein the treatment or prevention comprises, for example, agonizing the activity thereof by contacting cells, tissues or receptors with compounds as described herein.
  • the term “effective amount”, when used in connection with compounds as described herein, opioids, or opioid replacements, for example, for the treatment of pain refers to the treatment and/or prevention of the painful condition.
  • the term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of immunomodulatory disorders refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with immunomodulatory disorders and other related conditions.
  • the term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of anxiety, mood disorders, stress-related disorders, and attention deficit hyperactivity disorder refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with anxiety, mood disorders, stress-related disorders, attention deficit hyperactivity disorder and other related conditions.
  • the term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of motor disorders refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with motor disorders and other related conditions.
  • the term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of sexual dysfunction refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with sexual dysfunction and other related conditions.
  • the term “effective amount,” when used in connection with compounds useful in improving organ and cell survival, refers to refers to the maintenance and/or improvement of a minimally-acceptable level of organ or cell survival, including organ preservation.
  • the term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of myocardial infarction, refers to the minimum level of compound necessary to provide cardioprotection after myocardial infarction.
  • an effective amount when used in connection with compounds useful in the treatment and/or prevention of shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts and other related conditions.
  • an effective amount when used in connection with compounds useful in the treatment of substance addiction, refers to the treatment of symptoms, diseases, disorders, and conditions typically associated with substance addiction and other related conditions.
  • “Pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio. The term specifically encompasses veterinary uses.
  • “In combination with,” “combination therapy,” and “combination products” refer, in certain embodiments, to the concurrent administration to a patient of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII, and/or XXXIV, and one or more additional agents including, for example, an opioid, an anaesthetic agent (such as for example, an inhaled anesthetic, hypnotic, anxiolytic, neuromuscular blocker and opioid), an anti-Parkinson's agent (for example, in the case of treating or preventing a motor disorder, particularly Parkinson'
  • Dosage unit refers to physically discrete units suited as unitary dosages for the particular individual to be treated. Each unit may contain a predetermined quantity of active compound(s) calculated to produce the desired therapeutic effect(s) in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention may be dictated by (a) the unique characteristics of the active compound(s) and the particular therapeutic effect(s) to be achieved, and (b) the limitations inherent in the art of compounding such active compound(s).
  • “Pain” refers to the perception or condition of unpleasant sensory or emotional experience, associated with actual or potential tissue damage or described in terms of such damage. “Pain” includes, but is not limited to, two broad categories of pain: acute and chronic pain (Buschmann, H.; Christoph, T; Friderichs, E.; Maul, C.; Sundermann, B; eds.; Analgesics , Wiley-VCH, Verlag GMbH & Co. KgaA, Weinheim; 2002; Jain, K. K. “A Guide to Drug Evaluation for Chronic Pain”; Emerging Drugs, 5(2), 241-257 (2000)).
  • Non-limiting examples of pain include, for example, nociceptive pain, inflammatory pain, visceral pain, somatic pain, neuralgias, neuropathic pain, AIDS pain, cancer pain, phantom pain, and psychogenic pain, and pain resulting from hyperalgesia, pain caused by rheumatoid arthritis, migraine, allodynia and the like.
  • Gastrointestinal dysfunction refers collectively to maladies of the stomach, small and large intestine.
  • Non-limiting examples of gastrointestinal dysfunction include, for example, diarrhea, nausea, emesis, post-operative emesis, opioid-induced emesis, irritable bowel syndrome, opioid-bowel dysfunction, inflammatory bowel disease, colitis, increased gastric motility, increased gastric emptying, stimulation of small intestinal propulsion, stimulation of large intestinal propulsion, decreased amplitude of non-propulsive segmental contractions, disorders associated with sphincter of Oddi, disorders associated with anal sphincter tone, impaired reflex relaxation with rectal distention, disorders associated with gastric, biliary, pancreatic or intestinal secretions, changes to the absorption of water from bowel contents, gastro-esophageal reflux, gastroparesis, cramping, bloating, distension, abdominal or epigastric pain and discomfort, non-ulcerogenic dyspepsia, gastritis, or changes to the absorption of orally administered medications or
  • Urogenital tract disorders refers collectively to maladies of the urinary and genital apparati.
  • Non-limiting examples of urogenital tract disorders include incontinence (i.e., involuntary loss of urine) such as stress urinary incontinence, urge urinary incontinence and benign prostatic hyperplasia, overactive bladder disorder, urinary retention, renal colic, glomerulonephritis, and interstitial cystitis.
  • “Overactive bladder disorder” refers to a condition with symptoms of urgency with or without incontinence, and is typically associated with increased urinary frequency and nocturia. Overactive bladder disorders are typically associated with urodynamic finding of involuntary bladder contractions, generally referred to as bladder instability.
  • Immunomodulatory disorders refers collectively to maladies characterized by a compromised or over-stimulated immune system.
  • Non-limiting examples of immunomodulatory disorders include autoimmune diseases (such as arthritis, autoimmune disorders associated with skin grafts, autoimmune disorders associated with organ transplants, and autoimmune disorders associated with surgery), collagen diseases, allergies, side effects associated with the administration of an anti-tumor agent, side effects associated with the administration of an antiviral agent, multiple sclerosis and Guillain-Bane syndrome.
  • “Inflammatory disorders” refers collectively to maladies characterized by cellular events in injured tissues.
  • Non-limiting examples of inflammatory diseases include arthritis, psoriasis, asthma, and inflammatory bowel disease.
  • “Respiratory function disorders” refers to conditions in which breathing and/or airflow into the lung is compromised.
  • Non-limiting examples of respiratory function disorders include asthma, apnea, tussis, chronic obstruction pulmonary disease, and lung edema.
  • “Lung edema” refers to the presence of abnormally large amounts of fluid in the intercellular tissue spaces of the lungs.
  • “Anxiety” refers to the unpleasant emotional state consisting of psychophysiological responses to anticipation of real, unreal or imagined danger, ostensibly resulting from unrecognized intrapsychic conflict.
  • “Mood disorders” refers to disorders that have a disturbance in mood as their predominant feature, including depression, bipolar manic-depression, borderline personality disorder, and seasonal affective disorder.
  • “Depression” refers to a mental state of depressed mood characterized by feelings of sadness, despair and discouragement, including the blues, dysthymia, and major depression.
  • Stress-related disorders refer collectively to maladies characterized by a state of hyper- or hypo-arousal with hyper- and hypo-vigilance.
  • Non-limiting examples of stress-related disorders include post-traumatic stress disorder, panic disorder, generalized anxiety disorder, social phobia, and obsessive-compulsive disorder.
  • “Attention deficit hyperactivity disorder” refers to a condition characterized by an inability to control behavior due to difficulty in processing neural stimuli.
  • “Sympathetic nervous system disorders” refer collectively to maladies characterized by disturbances of the autonomic nervous system. Non-limiting examples of sympathetic nervous system disorders include hypertension, and the like.
  • Tussis refers to a coughing condition
  • antigenitussive agents refer to those materials that modulate the coughing response.
  • Motor disorders refers to involuntary manifestations of hyper or hypo muscle activity and coordination
  • Non-limiting examples of motor disorders include tremors, Parkinson's disease, tourette syndrome, parasomnias (sleep disorders) including restless leg syndrome, postoperative shivering and dyskinesia.
  • Traumatic injury of the central nervous system refers to a physical wound or injury to the spinal cord or brain.
  • “Stroke” refers to a condition due to the lack of oxygen to the brain.
  • Cardiac arrhythmia refers to a condition characterized by a disturbance in the electrical activity of the heart that manifests as an abnormality in heart rate or heart rhythm. Patients with a cardiac arrhythmia may experience a wide variety of symptoms ranging from palpitations to fainting.
  • “Glaucoma” refers collectively to eye diseases characterized by an increase in intraocular pressure that causes pathological changes in the optic disk and typical defects in the field of vision.
  • “Sexual dysfunction” refers collectively to disturbances, impairments or abnormalities of the functioning of the male or female sexual organs, including, but not limited to premature ejaculation and erectile dysfunction.
  • Cardioprotection refers to conditions or agents that protect or restore the heart from dysfunction, heart failure and reperfusion injury.
  • Myocardial infarction refers to irreversible injury to heart muscle caused by a local lack of oxygen.
  • “Addiction” refers to a pattern of compulsive substance abuse (alcohol, nicotine, or drug) characterized by a continued craving for the substance and, in some cases, the need to use the substance for effects other than its prescribed or legal use.
  • “Anaesthetic state” refers to the state of the loss of feeling or sensation, including not only the loss of tactile sensibility or of any of the other senses, but also to the loss of sensation of pain, as it is induced to permit performance of surgery or other painful procedures, and specifically including amnesia, analgesia, muscle relaxation and sedation.
  • “Improving organ and cell survival” refers to the maintenance and/or improvement of a minimally-acceptable level of organ or cell survival.
  • “Patient” refers to animals, including mammals, preferably humans.
  • “Side effect” refers to a consequence other than the one(s) for which an agent or measure is used, as the adverse effects produced by a drug, especially on a tissue or organ system other then the one sought to be benefited by its administration.
  • the term “side effect” may refer to such conditions as, for example, constipation, nausea, vomiting, dyspnea and pruritus.
  • the compounds, pharmaceutical compositions and methods of the present invention may involve a peripheral ⁇ opioid modulator compound.
  • peripheral designates that the compound acts primarily on physiological systems and components external to the central nervous system.
  • the peripheral ⁇ opioid modulator compounds employed in the methods of the present invention exhibit high levels of activity with respect to peripheral tissue, such as, gastrointestinal tissue, while exhibiting reduced, and preferably substantially no, CNS activity.
  • substantially no CNS activity means that less than about 50% of the pharmacological activity of the compounds employed in the present methods is exhibited in the CNS, preferably less than about 25%, more preferably less than about 10%, even more preferably less than about 5% and most preferably 0% of the pharmacological activity of the compounds employed in the present methods is exhibited in the CNS.
  • the ⁇ opioid modulator compound does not substantially cross the blood-brain barrier.
  • the phrase “does not substantially cross,” as used herein, means that less than about 20% by weight of the compound employed in the present methods crosses the blood-brain barrier, preferably less than about 15% by weight, more preferably less than about 10% by weight, even more preferably less than about 5% by weight and most preferably 0% by weight of the compound crosses the blood-brain barrier.
  • Selected compounds can be evaluated for CNS penetration, for example, by determining plasma and brain levels following i.v. administration.
  • the invention is directed to compounds of formula XIV:
  • W 2 is aryl or heteroaryl.
  • the aryl ring is preferably phenyl.
  • the heteroaryl ring is preferably pyridyl.
  • W 2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)).
  • W 2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)). More preferably, W 2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • W 2 is:
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • p is 1.
  • a 2 and B 2 together form a double bond.
  • X 2 is —O—.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl. In certain preferred embodiments, one of R 23 and R 24 is H and the other is alkyl.
  • the compounds of formula XIV have the following formula XV:
  • the compound of formula XIV is:
  • the invention is directed to compounds of formula XVII:
  • W 2 is aryl or heteroaryl.
  • the aryl ring is preferably phenyl.
  • the heteroaryl ring is preferably pyridyl.
  • W 2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)).
  • W 2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)). More preferably, W 2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • W 2 is:
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • X 2 is —O—.
  • a 2 and B 2 together form a double bond.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H.
  • J 2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • J 2 is substituted with 0-3 groups, preferably 0-2, more preferably 0-1 groups, selected independently from halo, hydroxy, and —S( ⁇ O) 2 -alkyl.
  • the halo group is preferably fluoro.
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, still more preferably alkyl groups of 1 to 2 carbons, yet more preferably methyl or ethyl.
  • the compounds of formula XVII have the following formula XVIII:
  • the compounds of formula XVII have the following structures:
  • the above compounds may be resolved into any of their R and S, or (R,R), (S,S), (R,S), and (S,R) enantiomers, or partially resolved into any of their non-racemic mixtures.
  • the invention is directed to compounds of formula XX:
  • the compound of formula XX is other than 4-[(4-N,N-diethylaminocarbonyl)phenyl]-spiro[2H,1-benzopyran-2,3′-pyrrolidine].
  • W 2 is aryl or heteroaryl.
  • the aryl ring is preferably phenyl.
  • the heteroaryl ring is preferably pyridyl.
  • W 2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)).
  • W 2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)). More preferably, W 2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • W 2 is:
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • a 2 and B 2 together form a double bond.
  • X 2 is —O—.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H.
  • J 2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • J 2 is substituted independently with 0-3, preferably 0-2, more preferably 0-1, hydroxy or halo groups, more preferably still, 0-1 hydroxy groups.
  • the compounds of formula XX have the following formula XXI:
  • the compounds of formula XX have the following structures:
  • the invention is directed to compounds of formula XXII:
  • W 2 is aryl or heteroaryl.
  • the aryl ring is preferably phenyl.
  • the heteroaryl ring is preferably pyridyl.
  • W 2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)).
  • W 2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C( ⁇ O)—NH 2 ), N-alkylaminocarbonyl (—C( ⁇ O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)). More preferably, W 2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • W 2 is:
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H.
  • J 2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • J 2 is substituted with 0-3 groups, preferably 0-2 groups, selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S( ⁇ O) 2 -alkyl, —S( ⁇ O) 2 —NH 2 , —S( ⁇ O) 2 —NH(alkyl), —S( ⁇ O) 2 —N(alkyl)(alkyl), carboxy (—COOH), —C( ⁇ O)—O-alkyl, and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)).
  • 0-3 groups preferably 0-2 groups, selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S( ⁇ O) 2 -alkyl, —S( ⁇ O) 2 —NH 2 , —S( ⁇ O) 2 —NH(alkyl), —S( ⁇ O) 2 —N(alkyl)(al
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 to 2 carbons being still more preferred.
  • the alkyl group is methyl or ethyl.
  • the halo group is preferably fluoro.
  • the compounds of formula XXII have the following formula XXIV:
  • the compounds of formula XXII have the following structure:
  • the compounds of formula XXII have the following structure:
  • the above compounds may be resolved into any of their R and S, or (R,R), (S,S), (R,S), and (S,R) enantiomers, or partially resolved into any of their non-racemic mixtures.
  • the invention is directed to compounds of formula
  • the compound of formula XXV is other than 4-phenyl-spiro[2H,1-benzopyran-2,4′-piperidine].
  • W 2 is aryl more preferably phenyl.
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 carbon being still more preferred. In particularly preferred embodiments, the alkyl group is methyl.
  • the aryl group is a 6-membered aryl ring, more preferably phenyl.
  • a 2 and B 2 together form a double bond.
  • X 2 is —O—.
  • p is 1.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H.
  • J 2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • J 2 is substituted with 0-3 groups, preferably 0-2, yet more preferably 0-1 groups, selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S( ⁇ O) 2 -alkyl, —S( ⁇ O) 2 —NH 2 , —S( ⁇ O) 2 —NH(alkyl), —S( ⁇ O) 2 —N(alkyl)(alkyl), carboxy (—COOH), —C( ⁇ O)—O-alkyl, and N,N-dialkylaminocarbonyl (—C( ⁇ O)—N(alkyl)(alkyl)).
  • 0-3 groups preferably 0-2, yet more preferably 0-1 groups, selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S( ⁇ O) 2 -alkyl, —S( ⁇ O) 2 —NH 2 , —S( ⁇ O) 2 —NH(alkyl), —S(
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 to 2 carbons being still more preferred.
  • the alkyl group is methyl or ethyl.
  • the halo group is preferably fluoro.
  • the compounds of formula XXV have the following formula XXVI:
  • the alkyl group is methyl or ethyl.
  • the halo group is preferably fluoro.
  • the compounds of formula XXV have the following structure:
  • the invention is directed to compounds of formula XXVII:
  • W 2 is para-dialkylaminocarbonylphenyl. As set forth above, W 2 is further optionally substituted with 1-2 groups independently selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C( ⁇ O)—NH 2 ). In preferred embodiments wherein W 2 is substituted with 1-2 groups, W 2 is preferably substituted with hydroxy.
  • W 2 is:
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H.
  • Q 1 and Q 2 are each independently H, hydroxy, alkoxy, haloalkoxy, halo, or heterocycloalkyl.
  • the alkoxy is preferably C 1 -C 3 alkoxy, more preferably C 1 alkoxy, yet more preferably, methoxy.
  • the halo is preferably fluoro.
  • the heterocycloalkyl is preferably a 5- or 6-membered ring heterocycloalkyl, more preferably pyrrolidinyl or morpholinyl.
  • Q 1 or Q 2 is haloalkoxy
  • the alkoxy is substituted with one or more, preferably two or more, fluoro atoms.
  • the compounds of formula XXVII have the structures:
  • the compound of formula XXVII has the structure:
  • the compound is substantially enantiomerically pure.
  • the invention is directed to compounds of formula XXVIII:
  • D is:
  • the alkyl group independently is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • the alkyl group independently is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1-2 carbon being still more preferred. In particularly preferred embodiments, the alkyl group is methyl or ethyl.
  • the heteroaryl group is preferably a 5-membered ring heteroaryl, more preferably a tetrazolyl ring.
  • a 2 and B 2 together form a double bond.
  • p is 1.
  • X 2 is —O—.
  • R 23 , R 24 , and R 26 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H. In certain preferred embodiments, one of R 23 and R 24 is H and the other is alkyl.
  • the compounds of formula XXVIII have the structures:
  • the invention is directed to compounds of formula XXIX:
  • W 2 is para-N(alkyl),N(alkyl-Z)-aminocarbonylaryl or para-N(alkyl),N(alkyl-Z)-aminocarbonylheteroaryl.
  • the aryl ring is preferably phenyl.
  • the heteroaryl ring is preferably pyridyl.
  • W 2 is substituted with 0-2 groups selected independently from selected independently from hydroxy and alkoxy.
  • W 2 is substituted with 0-1 groups, selected independently from hydroxy and alkoxy, more preferably hydroxy.
  • W 2 is:
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • p is 1.
  • a 2 and B 2 together form a double bond.
  • X 2 is —O—.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl. In certain preferred embodiments, one of R 23 and R 24 is H and the other is alkyl.
  • Z is alkoxy, alkylamino, or dialkylamino, preferably alkoxy.
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 carbon being still more preferred.
  • the alkyl group is methyl.
  • the compounds of formula XXIX have the structures:
  • the invention is directed to compounds of formula XXX:
  • the aryl ring of J 2 is substituted with at least one haloalkoxy.
  • W 2 is:
  • p is 1.
  • R 23 and R 24 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H. In certain preferred embodiments, one of R 23 and R 24 is H and the other is alkyl.
  • J 2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably a phenyl ring.
  • the halo group of halo or haloalkoxy is preferably fluoro.
  • the compounds of formula XXX have the following formula XXXI:
  • the compounds of formula XXX have the structures:
  • the invention is directed to compounds of formula XXXII:
  • a 2 and B 2 are each H.
  • the heteroaryl group is preferably pyridyl or thienyl.
  • the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2-3 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl or isopropyl.
  • a 2 and B 2 are each H.
  • X 2 is —O—.
  • R 23 , R 24 , and R 26 are each independently H or alkyl, preferably H or C 1 -C 3 alkyl, more preferably H or methyl, yet more preferably H. In certain preferred embodiments, one of R 23 and R 24 is H and the other is alkyl.
  • the compounds of formula XXXII have the structures:
  • the present invention is directed, in part, to compounds of formula XXXIII:
  • F 1 is heteroaryl, preferably a 5- or 6-membered heteroaryl having 1 to 4 heteroatoms, with 2 to 4 heteroatoms being more preferred. In certain more preferred embodiments, F 1 is a 5-membered heteroaryl, still more preferably a tetrazole.
  • G is C 1-6 alkylene substituted with NH 2 , NHC( ⁇ O)alkyl, NH(C(O)N(H)alkyl, or NHS( ⁇ O) 2 alkyl.
  • G is C 1-6 alkylene substituted with NH 2 .
  • G is C 1-6 alkylene substituted with NHC( ⁇ O)alkyl.
  • G is C 1-6 alkylene substituted with NHS( ⁇ O) 2 alkyl. More preferably, G is C 1-6 alkylene substituted with NH 2 .
  • F 1 -G is:
  • the compounds of formula XXXIII are selected from the group consisting of:
  • the present invention is directed, in part, to compounds of formula XXXIV:
  • F 2 is aryl or heteroaryl.
  • F 2 is aryl, preferably it is C 6-10 aryl, more preferably C 6 aryl, with phenyl being even more preferred.
  • F 2 is heteroaryl, preferably it is C 6-10 heteroaryl, with pyridyl or benzothiophenyl being more preferred.
  • Q 3 is hydroxy or alkoxy, preferably hydroxy.
  • the compound of the invention is selected from the group consisting of:
  • compositions comprising:
  • compositions comprising:
  • Compounds as described herein may be useful as analgesic agents for use during general anesthesia and monitored anesthesia care.
  • Combinations of agents with different properties are often used to achieve a balance of effects needed to maintain the anaesthetic state (e.g., amnesia, analgesia, muscle relaxation and sedation). Included in this combination are inhaled anesthetics, hypnotics, anxiolytics, neuromuscular blockers and opioids.
  • a compound as described herein may be either a compound of one of the formulae herein described, or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof.
  • prodrug is intended to include any covalently bonded carriers which release the active parent drug, for example, as according to formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV, or other formulas or compounds as described herein, such as for example, compounds of formula XXVIIA, in vivo when such prodrug is administered to a mammalian subject.
  • prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.), the compounds described herein may, if desired, be delivered in prodrug form. Thus, the present invention contemplates compositions and methods involving prodrugs.
  • Prodrugs of the compounds employed in the present invention for example formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXII, XXIII and/or XXIV, may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • prodrugs include, for example, compounds described herein in which a hydroxy, amino, or carboxy group is bonded to any group that, when the prodrug is administered to a mammalian subject, cleaves to form a free hydroxyl, free amino, or carboxylic acid, respectively.
  • Examples include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups; and alkyl, carbocyclic, aryl, and alkylaryl esters such as methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, phenyl, benzyl, and phenethyl esters, and the like.
  • alkyl, carbocyclic, aryl, and alkylaryl esters such as methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, phenyl, benzyl, and phenethyl esters, and the like.
  • Compounds described herein may contain one or more asymmetrically substituted carbon atoms, and may be isolated in optically active or racemic forms. Thus, all chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. It is well known in the art how to prepare and isolate such optically active forms. For example, mixtures of stereoisomers may be separated by standard techniques including, but not limited to, resolution of racemic forms, normal, reverse-phase, and chiral chromatography, preferential salt formation, recrystallization, and the like, or by chiral synthesis either from chiral starting materials or by deliberate synthesis of target chiral centers.
  • the compounds as herein described may be prepared in a number of ways well known to those skilled in the art.
  • the compounds can be synthesized, for example, by the methods described below, or variations thereon as appreciated by the skilled artisan. All processes disclosed in association with the present invention are contemplated to be practiced on any scale, including milligram, gram, multigram, kilogram, multikilogram or commercial industrial scale.
  • protecting groups may contain protecting groups during the course of synthesis.
  • Protecting groups are known per se as chemical functional groups that can be selectively appended to and removed from functionalities, such as hydroxyl groups and carboxyl groups. These groups are present in a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed.
  • Any of a variety of protecting groups may be employed with the present invention.
  • Preferred protecting groups include the benzyloxycarbonyl group and the tert-butyloxycarbonyl group.
  • Other preferred protecting groups that may be employed in accordance with the present invention may be described in Greene, T. W. and Wuts, P. G. M., Protective Groups in Organic Synthesis 2d. Ed., Wiley & Sons, 1991, the disclosures of which are hereby incorporated herein by reference, in their entirety.
  • the ⁇ agonist compounds as described herein may be administered by any means that results in the contact of the active agent with the agent's site of action in the body of a patient.
  • the compounds may be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents.
  • they may be administered as the sole active agent in a pharmaceutical composition, or they can be used in combination with other therapeutically active ingredients including, for example, opioid analgesic agents.
  • selected compounds as described herein may provide equivalent or even enhanced therapeutic activity such as, for example, pain ameliorization, while providing reduced adverse side effects associated with opioids, such as addiction or pruritus, by lowering the amount of opioid required to achieve a therapeutic effect.
  • the compounds are preferably combined with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice as described, for example, in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa., 1980), the disclosures of which are hereby incorporated herein by reference, in their entirety.
  • the compounds of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXII, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXII, XXXIII and/or XXXIV may be co-administered with at least one opioid, preferably a ⁇ opioid receptor modulator compound.
  • the utility of the instant combination product may be determined by those skilled in the art using established animal models.
  • Suitable opioids include, without limitation, alfentanil, allylprodine, alphaprodine, anileridine, benzyl-morphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioaphetylbutyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacylmorphan,
  • the pain ameliorating and/or opioid combination products of the present compositions may further include one or more other active ingredients that may be conventionally employed in analgesic and/or cough-cold-antitussive combination products.
  • active ingredients include, for example, aspirin, acetaminophen, phenylpropanolamine, phenylephrine, chlorpheniramine, caffeine, and/or guaifenesin.
  • Typical or conventional ingredients that may be included in the opioid component are described, for example, in the Physicians' Desk Reference, 1999, the disclosure of which is hereby incorporated herein by reference, in its entirety.
  • the opioid component may further include one or more compounds that may be designed to enhance the analgesic potency of the opioid and/or to reduce analgesic tolerance development.
  • compounds include, for example, dextromethorphan or other NMDA antagonists (Mao, M. J. et al., Pain 1996, 67, 361), L-364,718 and other CCK antagonists (Dourish, C. T. et al., Eur J Pharmacol 1988, 147, 469), NOS inhibitors (Bhargava, H. N. et al., Neuropeptides 1996, 30, 219), PKC inhibitors (Bilsky, E. J.
  • opioids optional conventional opioid components, and optional compounds for enhancing the analgesic potency of the opioid and/or for reducing analgesic tolerance development, that may be employed in the methods and compositions of the present invention, in addition to those exemplified above, would be readily apparent to one of ordinary skill in the art, once armed with the teachings of the present disclosure.
  • Parenteral administration in this respect includes administration by the following routes: intravenous, intramuscular, subcutaneous, rectal, intraocular, intrasynovial, transepithelial including transdermal, ophthalmic, sublingual and buccal; topically including ophthalmic, dermal, ocular, rectal, and nasal inhalation via insufflation aerosol.
  • the active compound may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet.
  • the active compound may be incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should preferably contain at least 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be, for example, from about 2 to about 6% of the weight of the unit.
  • the amount of active compound in such therapeutically useful compositions is preferably such that a suitable dosage will be obtained.
  • Preferred compositions or preparations according to the present invention may be prepared so that an oral dosage unit form contains from about 0.1 to about 1000 mg of active compound.
  • the tablets, troches, pills, capsules and the like may also contain one or more of the following: a binder, such as gum tragacanth, acacia, corn starch or gelatin; an excipient, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; a sweetening agent such as sucrose, lactose or saccharin; or a flavoring agent, such as peppermint, oil of wintergreen or cherry flavoring.
  • a binder such as gum tragacanth, acacia, corn starch or gelatin
  • an excipient such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin
  • a flavoring agent such
  • any material used in preparing any dosage unit form is preferably pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and formulations.
  • the active compound may also be administered parenterally or intraperitoneally.
  • Solutions of the active compound as a free base or a pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • a dispersion can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form is preferably sterile and fluid to provide easy syringability. It is preferably stable under the conditions of manufacture and storage and is preferably preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of a dispersion, and by the use of surfactants.
  • a coating such as lecithin
  • surfactants for example, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium stearate, sodium stearate, and gelatin.
  • Sterile injectable solutions may be prepared by incorporating the active compound in the required amount, in the appropriate solvent, with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions may be prepared by incorporating the sterilized active ingredient into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation may include vacuum drying and the freeze-drying technique that yield a powder of the active ingredient, plus any additional desired ingredient from the previously sterile-filtered solution thereof.
  • the therapeutic compounds of this invention may be administered to a patient alone or in combination with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier As noted above, the relative proportions of active ingredient and carrier may be determined, for example, by the solubility and chemical nature of the compound, chosen route of administration and standard pharmaceutical practice.
  • the dosage of the compounds as described herein that will be most suitable for prophylaxis or treatment will vary with the form of administration, the particular compound chosen and the physiological characteristics of the particular patient under treatment. Generally, small dosages may be used initially and, if necessary, increased by small increments until the desired effect under the circumstances is reached.
  • the therapeutic human dosage based on physiological studies using rats, may generally range from about 0.01 mg to about 100 mg/kg of body weight per day, and all combinations and subcombinations of ranges and specific dosages therein. Alternatively, the therapeutic human dosage may be from about 0.4 mg to about 10 g or higher, and may be administered in several different dosage units from once to several times a day. Generally speaking, oral administration may require higher dosages.
  • the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • the dose may also be provided by controlled release of the compound, by techniques well known to those in the art.
  • the compounds as described herein may also be formulated with other optional active ingredients, in addition to or instead of the optional opioids, and in addition to the optional pharmaceutical-acceptable carriers.
  • active ingredients include, but are not limited to, antibiotics, antivirals, antifungals, anti-inflammatories, including steroidal and non-steroidal anti-inflammatories, anesthetics and mixtures thereof.
  • additional ingredients include any of the following:
  • Aminoglycosides such as Amikacin, Apramycin, Arbekacin, Bambermycins, Butirosin, Dibekacin, Dihydrostreptomycin, Fortimicin(s), Fradiomycin, Gentamicin, Ispamicin, Kanamycin, Micronomicin, Neomycin, Neomycin Undecylenate, Netilmicin, Paromomycin, Ribostamycin, Sisomicin, Spectinomycin, Streptomycin, Streptonicozid and Tobramycin;
  • Amphenicols such as Azidamfenicol, Chloramphenicol, Chloramphenicol Palmirate, Chloramphenicol Pantothenate, Florfenicol, Thiamphenicol;
  • Ansamycins such as Rifamide, Rifampin, Rifamycin and Rifaximin;
  • Carbapenems such as Imipenem
  • Cephalosporins such as 1-Carba (dethia) Cephalosporin, Cefactor, Cefadroxil, Cefamandole, Cefatrizine, Cefazedone, Cefazolin, Cefixime, Cefmenoxime, Cefodizime, Cefonicid, Cefoperazone, Ceforanide, Cefotaxime, Cefotiam, Cefpimizole, Cefpirimide, Cefpodoxime Proxetil, Cefroxadine, Cefsulodin, Ceftazidime, Cefteram, Ceftezole, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefuroxime, Cefuzonam, Cephacetrile Sodium, Cephalexin, Cephaloglycin, Cephaloridine, Cephalosporin, Cephalothin, Cephapirin Sodium, Cephradine and Pivcefalexin;
  • Cephamycins such as Cefbuperazone, Cefmetazole, Cefminox, Cefetan and Cefoxitin;
  • Monobactams such as Aztreonam, Carumonam and Tigemonan
  • Oxacephems such as Flomoxef and Moxolactam
  • Penicillins such as Amidinocillin, Amdinocillin, Pivoxil, Amoxicillin, Ampicillan, Apalcillin, Aspoxicillin, Azidocillan, Azlocillan, Bacampicillin, Benzylpenicillinic Acid, Benzylpenicillin, Carbenicillin, Carfecillin, Carindacillin, Clometocillin, Cloxacillin, Cyclacillin, Dicloxacillin, Diphenicillin, Epicillin, Fenbenicillin, Floxicillin, Hetacillin, Lenampicillin, Metampicillin, Methicillin, Mezlocillin, Nafcillin, Oxacillin, Penamecillin, Penethamate Hydriodide, Penicillin G Benethamine, Penicillin G Benzathine, Penicillin G Benzhydrylamine, Penicillin G Calcium, Penicillin G Hydragamine, Penicillin G Potassium, Penicillin G Procaine, Penicillin N, Penicillin O, Penicillin V, Penicillin
  • Lincosumides such as Clindamycin and Lincomycin
  • Macrolides such as Azithromycin, Carbomycin, Clarithromycin, Erythromycin(s) and Derivatives, Josamycin, Leucomycins, Midecamycins, Miokamycin, Oleandomycin, Primycin, Rokitamycin, Rosaramicin, Roxithromycin, Spiramycin and Troleandomycin;
  • Polypeptides such as Amphomycin, Bacitracin, Capreomycin, Colistin, Enduracidin, Enviomycin, Fusafungine, Gramicidin(s), Gramicidin S, Mikamycin, Polymyxin, Polymyxin ⁇ -Methanesulfonic Acid, Pristinamycin, Ristocetin, Teicoplanin, Thiostrepton, Tuberactinomycin, Tyrocidine, Tyrothricin, Vancomycin, Viomycin(s), Virginiamycin and Zinc Bacitracin;
  • Tetracyclines such as Spicycline, Chlortetracycline, Clomocycline, Demeclocycline, Doxycycline, Guamecycline, Lymecycline, Meclocycline, Methacycline, Minocycline, Oxytetracycline, Penimepicycline, Pipacycline, Rolitetracycline, Sancycline, Senociclin and Tetracycline; and
  • 2,4-Diaminopyrimidines such as Brodimoprim, Tetroxoprim and Trimethoprim;
  • Nitrofurans such as Furaltadone, Furazolium, Nifuradene, Nifuratel, Nifurfoline, Nifurpirinol, Nifurprazine, Nifurtoinol and Nitrofurantoin;
  • Quinolones and analogs thereof such as Amifloxacin, Cinoxacin, Ciprofloxacin, Difloxacin, Enoxacin, Fleroxacin, Flumequine, Lomefloxacin, Miloxacin, Nalidixic Acid, Norfloxacin, Ofloxacin, Oxolinic Acid, Perfloxacin, Pipemidic Acid, Piromidic Acid, Rosoxacin, Temafloxacin and Tosufloxacin;
  • Sulfonamides such as Acetyl Sulfamethoxypyrazine, Acetyl Sulfisoxazole, Azosulfamide, Benzylsulfamide, Chloramine- ⁇ , Chloramine-T, Dichloramine-T, Formosulfathiazole, N.sup.2-Formyl-sulfisomidine, N.sup.4- ⁇ -D-Glucosylsulfanilamide, Mafenide, 4′-(Methyl-sulfamoyl)sulfanilanilide, p-Nitrosulfathiazole, Noprylsulfamide, Phthalylsulfacetamide, Phthalylsulfathiazole, Salazosulfadimidine, Succinylsulfathiazole, Sulfabenzamide, Sulfacetamide, Sulfachlorpyridazine, Sulfachrysoidine, Sulfacytine, Sulf
  • Sulfones such as Acedapsone, Acediasulfone, Acetosulfone, Dapsone, Diathymosulfone, Glucosulfone, Solasulfone, Succisulfone, Sulfanilic Acid, p-Sulfanilylbenzylamine, p,p′-sulfonyldianiline-N,N′-digalactoside, Sulfoxone and Thiazolsulfone;
  • Clofoctol Hexedine
  • Magainins Methenamine, Methenamine Anhydromethylene-citrate, Methenamine Hippurate, Methenamine Mandelate, Methenamine Sulfosalicylate, Nitroxoline, Squalamine and Xibomol.
  • Polyenes such as Amphotericin-B, Candicidin, Dermostatin, Filipin, Fungichromin, Hachimycin, Hamycin, Lucensomycin, Mepartricin, Natamycin, Nystatin, Pecilocin, Perimycin; and others, such as Azaserine, Griseofulvin, Oligomycins, PyrroInitrin, Siccanin, Tubercidin and Viridin.
  • Imidazoles such as Bifonazole, Butoconazole, Chlordantoin, Chlormidazole, Cloconazole, Clotrimazole, Econazole, Enilconazole, Finticonazole, Isoconazole, Ketoconazole, Miconazole, Omoconazole, Oxiconazole Nitrate, Sulconazole and Tioconazole;
  • Triazoles such as Fluconazole, Itraconazole, Terconazole;
  • Antiglaucoma agents such as Dapiprazoke, Dichlorphenamide, Dipivefrin and Pilocarpine.
  • aminoarylcarboxylic Acid Derivatives such as Etofenamate, Meclofenamic Acid, Mefanamic Acid, Niflumic Acid;
  • Arylacetic Acid Derivatives such as Acemetacin, Amfenac Cinmetacin, Clopirac, Diclofenac, Fenclofenac, Fenclorac, Fenclozic Acid, Fentiazac, Glucametacin, Isozepac, Lonazolac, Metiazinic Acid, Oxametacine, Proglumetacin, Sulindac, Tiaramide and Tolmetin;
  • Arylbutyric Acid Derivatives such as Butibufen and Fenbufen; Arylcarboxylic Acids such as Clidanac, Ketorolac and Tinoridine; Arylpropionic Acid Derivatives such as Bucloxic Acid, Carprofen, Fenoprofen, Flunoxaprofen, Ibuprofen, Ibuproxam, Oxaprozin, Piketoprofen, Pirprofen, Pranoprofen, Protizinic Acid and Tiaprofenic Add;
  • Pyrazolones such as Clofezone, Feprazone, Mofebutazone, Oxyphenbutazone, Phenylbutazone, Phenyl Pyrazolidininones, Suxibuzone and Thiazolinobutazone;
  • Salicylic Acid Derivatives such as Bromosaligenin, Fendosal, Glycol Salicylate, Mesalamine, 1-Naphthyl Salicylate, Olsalazine and Sulfasalazine;
  • Thiazinecarboxamides such as Droxicam, Isoxicam and Piroxicam;
  • Guanidines such as Alexidine, Ambazone, Chlorhexidine and Picloxydine;
  • Halogens/Halogen Compounds such as Bomyl Chloride, Calcium Iodate, Iodine, Iodine Monochloride, Iodine Trichloride, Iodoform, Povidone-Iodine, Sodium Hypochlorite, Sodium Iodate, Symclosene, Thymol Iodide, Triclocarban, Triclosan and Troclosene Potassium;
  • Nitrofurans such as Furazolidone, 2-(Methoxymethyl)-5-Nitrofuran, Nidroxyzone, Nifuroxime, Nifurzide and Nitrofurazone;
  • Phenols such as Acetomeroctol, Chloroxylenol, Hexachlorophene, 1-Naphthyl Salicylate, 2,4,6-Tribromo-m-cresol and 3′,4′,5-Trichlorosalicylanilide;
  • Quinolines such as Aminoquinuride, Chloroxine, Chlorquinaldol, Cloxyquin, Ethylhydrocupreine, Halquinol, Hydrastine, 8-Hydroxyquinoline and Sulfate; and
  • Purines/Pyrimidinones such as 2-Acetyl-Pyridine 5-((2-pyridylamino)thiocarbonyl) Thiocarbonohydrazone, Acyclovir, Dideoxyadenosine, Dideoxycytidine, Dideoxyinosine, Edoxudine, Floxuridine, Ganciclovir, Idoxuridine, MADU, Pyridinone, Trifluridine, Vidrarbine and Zidovudline;
  • Mild OTC over the counter analgesics, such as aspirin, acetaminophen, and ibuprophen.
  • Narcotic analgesics such as codeine.
  • Anti seizure medications such as carbamazepine, gabapentin, lamotrigine and phenyloin.
  • Anti-depressants such as amitryptiline.
  • SSRIs Selective serotonin re-uptake inhibitors
  • Tricyclics such as Imipramine, Amitriptyline, Desipramine, Nortriptyline Protriptyline, Trimipramine, Doxepin, Amoxapine, and Clomipramine.
  • MAOIs Monoamine Oxidase Inhibitors
  • Tranylcypromine Phenelzine
  • Isocarboxazid Isocarboxazid
  • Heterocyclics such as Amoxipine, Maprotiline and Trazodone.
  • Anticholinergic agents such as propantheline.
  • Antispasmodic medications such as oxybutynin, tolterodine, and flavoxate.
  • Tricyclic antidepressants such as imipramine, and doxepin.
  • Calcium channel blockers such as tolterodine.
  • Beta agonists such as terbutaline.
  • the invention is directed to methods of binding opioid receptors, preferably ⁇ opioid receptors, in a patient in need thereof, comprising the step of administering to said patient an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XVI, XXVII, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the ⁇ opioid receptors may be located in the central nervous system or located peripherally to the central nervous system.
  • the binding of the present compounds modulates the activity, preferably as an agonist, of said opioid receptors.
  • the compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV does not substantially cross the blood-brain barrier.
  • the compounds as described herein are peripherally selective.
  • the spirocyclic heterocyclic derivatives of the present invention and pharmaceutical compositions containing these compounds may be utilized in a number of ways.
  • the spirocyclic heterocyclic derivatives are ligands of the ⁇ opioid receptor and are useful, inter alia, in methods for treating and/or preventing pain, gastrointestinal disorders, urogenital tract disorders including incontinence, for example, stress urinary incontinence, urge urinary incontinence and benigh prostatic hyperplasia, and overactive bladder disorder (see, e.g., R. B. Moreland et al., Perspectives in Pharmacology , Vol. 308(3), pp. 797-804 (2004) and M. O.
  • immunomodulatory disorders inflammatory disorders, respiratory function disorders, depression, anxiety, mood disorders, stress-related disorders, sympathetic nervous system disorder, tussis, motor disorder, traumatic injury, stroke, cardiac arrhythmia, glaucoma, sexual dysfunction, shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, and substance addiction.
  • the spirocyclic heterocyclic derivatives are ligands of the ⁇ opioid receptor and are useful, inter alia, in methods for providing cardioprotection following myocardial infarction, in methods for providing and maintaining an anaesthetic state, and in methods of detecting, imaging or monitoring degeneration or dysfunction of opioid receptors in a patient.
  • methods of preventing or treating pain comprising the step of administering to said patient an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the present methods of preventing or treating pain may further comprise the administration to a patient of an effective amount of an agent for the treatment of neuralgia and/or neuropathic pain.
  • the invention is directed to methods for preventing or treating gastrointestinal dysfunction, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXIV.
  • the invention is directed to methods for preventing or treating a urogenital tract disorder, such as incontinence (including, for example, stress urinary incontinence and urge urinary incontinence, and overactive bladder), comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • a urogenital tract disorder such as incontinence (including, for example, stress urinary incontinence and urge urinary incontinence, and overactive bladder)
  • a compound as described herein including, for
  • the present methods of preventing or treating a urogenital tract disorder may further comprise the administration to a patient of an effective amount of an agent for the treatment of incontinence.
  • the invention is directed to methods of preventing or treating an immunomodulatory disorder, comprising the step of administering to a patient in need thereof an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXIV
  • Immunomodulatory disorders include, but are not limited to, autoimmune diseases, collagen diseases, allergies, side effects associated with the administration of an anti-tumor agent, and side effects associated with the administration of an antiviral agent.
  • Autoimmune diseases include, but are not limited to, arthritis, autoimmune disorders associated with skin grafts, autoimmune disorders associated with
  • the invention is directed to methods of preventing or treating an inflammatory disorder, comprising the step of administering to a patient in need thereof an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • Inflammatory disorders include, but are not limited to, arthritis, psoriasis, asthma, or inflammatory bowel disease.
  • the invention is directed to methods of preventing or treating a respiratory function disorder, comprising the step of administering to a patient in need thereof an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXIV.
  • Respiratory function disorders include but are not limited to asthma or lung edema.
  • the invention is directed to methods for preventing or treating anxiety, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating a mood disorder, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the present methods of preventing or treating a mood disorder may further comprise the administration to a patient of an effective amount of an agent for the treatment of depression.
  • the invention is directed to methods for preventing or treating a stress-related disorder, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXIV.
  • Stress-related disorders include, but are not limited to, post-traumatic stress disorder, panic disorder, generalized anxiety disorder, social phobia, and obsessive compulsive disorder.
  • the invention is directed to methods for preventing or treating attention deficit hyperactivity disorder, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating sympathetic nervous system disorders, including hypertension, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating tussis, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating a motor disorder, including tremors, Parkinson's disease, Tourette's syndrome and dyskenesia, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XVI, XXVII, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the present methods of preventing or treating a motor disorder may further comprise the administration to a patient of an effective amount of an agent for the treatment of Parkinson's disease.
  • the invention is directed to methods for treating a traumatic injury to the central nervous system, including the spinal cord or brain, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XVI, XXVII, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating stroke, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating cardiac arrhythmia, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXIV.
  • the invention is directed to methods for preventing or treating glaucoma, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for preventing or treating sexual dysfunction, including premature ejaculation, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XVI, XXVII, XXVIIA, XVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for treating a condition selected from the group consisting of shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, comprising the step of administering to a patient in need of such treatment an effective amount of a compound of the invention including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for treating substance addiction, including addictions to alcohol, nicotine or drugs such as opioids, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for improving organ and cell survival, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for providing cardioprotection following myocardial infarction, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XVIII, XXIX, XX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods for reducing the need for anesthesia, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXXIV.
  • the invention is directed to methods of producing or maintaining an anesthetic state, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, XXI, XXII, XXIII and/or XXIV.
  • the method may further comprise the step of administering to said patient an anesthetic agent, which may be co-administered with compound(s) of the invention.
  • anesthetic agent include, for example, an inhaled anaesthetic, a hypnotic, an anxiolytic, a neuromuscular blocker and an opioid.
  • compounds of the invention may be useful as analgesic agents for use during general anesthesia and monitored anesthesia care. Combinations of agents with different properties may be used to achieve a balance of effects needed to maintain the anaesthetic state.
  • Additional diseases and/or disorders which may be treated and/or prevented with the compounds and pharmaceutical compositions of the present invention include those described, for example, in WO2004/062562 A2, WO 2004/063157 A1, WO 2004/063193 A1, WO 2004/041801 A1, WO 2004/041784 A1, WO 2004/041800 A1, WO 2004/060321 A2, WO 2004/035541 A1, WO 2004/035574 A2, WO 2004041802 A1, US 2004082612 A1, WO 2004026819 A2, WO 2003057223 A1, WO 2003037342 A1, WO 2002094812 A1, WO 2002094810 A1, WO 2002094794 A1, WO 2002094786 A1, WO 2002094785 A1, WO 2002094784 A1, WO 2002094782 A1, WO 2002094783 A1, WO 2002094811 A1, the disclosures of each of which are hereby incorporated herein by reference in their entireties.
  • the present invention is directed to radiolabeled derivatives and isotopically labeled derivatives of compounds as described herein including, for example, compounds of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVIIA, XXVIII, XXIX, XXX, MI, XXXII, XXIII and/or XXIV.
  • Suitable labels include, for example, 2 H, 3 H, 11 C, 13 C, 13 N, 15 N, 15 O, 18 O, 18 F and 34 S.
  • Such labeled derivatives may be useful for biological studies, for example, using positron emission tomography, for metabolite identification studies and the like.
  • diagnostic imaging methods may comprise, for example, administering to a patient a radiolabeled derivative or isotopically labeled derivative of a compound of the invention, and imaging the patient, for example, by application of suitable energy, such as in positron emission tomography.
  • Isotopically- and radio-labeled derivatives may be prepared utilizing techniques well known to the ordinarily skilled artisan.
  • the ether derivatives 2.9 were also obtained from the phenols 2.7 using the Mitsunobu conditions, i.e., condensation of the phenols 2.7 with the appropriate alcohol (2.8d, 2.8e) in the presence of triphenylphosphine and diisopropyl azodicarboxylate (DIAD) (method 2B). Treatment of the Boc derivatives 2.9 with hydrochloric acid provided the final compounds 2C-F.
  • the dimethylamide derivative 10.4b (R 1 ⁇ H, R 2 ⁇ CH 3 ) was obtained by heating a mixture of the ester 10.2 with methylamine (3.4b) in methanol in a sealed tube. Treatment of the Boc derivatives 10.2, 10.3 and 10.4 with hydrochloric acid provided the final compounds 10A-10I. Treatment of the ester 10.2 with lithium borohydride in tetrahydrofuran provided the primary alcohol 10.5 which was converted to the compound 10J under acidic conditions.
  • Treatment of the Boc derivatives 11.9 with hydrochloric acid provided the final compounds 11C-11F.
  • Treatment of the phenol 11.2 with methyl chlorodifluoroacetate (9.7) in N,N-dimethylformamide in the presence of cesium carbonate provided the ether derivative 11.11.
  • Conversion of the ketone 11.11 to the enol triflate derivative 11.12 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent.
  • Acidic hydrolysis of 19.6 provided the aniline derivative 19.7 which reacted with acyl chlorides 19.8a, 19.8b, isopropylsulfonyl chloride (6.5b) or ethyl isocyanate (19.11) to give the corresponding amide derivatives 19.9, sulfonamide derivative 19.10 or urea derivative 19.12, respectively.
  • the derivatives 19.9, 19.10 and 19.12 were converted to compounds 19A-19D by treatment with iodotrimethylsilane.
  • the synthesis of compounds 20A-20R is outlined in Scheme 20.
  • the tertiary amine derivatives compounds 20A-20R were obtained from the secondary amines of general formula 20I, by reductive amination methods (methods 20A or 20B) using the aldehydes 20.1a-20.1d and sodium cyanoborohydride as reducing agent or by alkylation method (method 20C) using the bromides 2.8a, 20.2a-e as the alkylating reagent.
  • the 2′-hydroxyacetophenone 1.1a was condensed with 21.4 in refluxing methanol in the presence of pyrrolidine to provide the racemic ketone 21.5. Conversion of 21.5 to the enol triflate derivative 21.6 was achieved using the triflating reagent N-phenylbis(trifluoromethanesulphonimide) 1.4.
  • the derivatives 33.1 used in the Suzuki coupling step were either obtained from commercial sources (33.1a-e,l,m) or prepared as follows. Coupling of 5-bromopyridine-3-carboxylic acid (33.3) or 6-bromopyridine-2-carboxylic acid (33.4) with diethylamine (1.12) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the diethylaminocarbonyl derivative derivatives 33.1f and 33.1g, respectively.
  • TBTU O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate
  • the compound 38B (racemic mixture) was obtained by hydrogenation of the unsaturated derivative 38A in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon. Conversion of the enol triflate 38.7 to the corresponding boron derivative 38.9 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct ⁇ [Pd(dppf)Cl 2 .CH 2 Cl 2 ] ⁇ .
  • the enantiomers 42.2 and 42.3 were converted to compounds 42A and 42B, respectively under acidic conditions. Conversion of the enol triflate 21.6 to the corresponding boron derivative 42.4 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct ⁇ [Pd(dppf)Cl 2 .CH 2 Cl 2 ] ⁇ .
  • the enantiomers 43.5 and 43.6 were converted to compounds 43A and 43B, respectively under acidic conditions.
  • the 2′-5′-dihydroxyacetophenone derivative 2.1 was condensed with tert-butyl 4-oxoazepane-1-carboxylate 21.4 in refluxing methanol in the presence of pyrrolidine to provide the derivative 45.4 which was converted to the silyl ether derivative 45.5 using tert-butyldimethylsilyl chloride 2.3. Conversion of the ketone 45.5 to the enol triflate derivative 45.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent.
  • the 2′-6′-dihydroxyacetophenone derivative 11.1 was condensed with tert-butyl 3-oxopyrrolidine-1-carboxylate 23.1a in refluxing methanol in the presence of pyrrolidine to provide the derivative 47.4 which was converted to the methoxymethyl (MOM) ether derivative 47.5 using chloro(methoxy)methane (11.3). Conversion of the ketone 47.5 to the enol triflate derivative 47.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent.
  • the 2′-5′-dihydroxyacetophenone derivative 2.1 was condensed with tert-butyl 3-oxopyrrolidine-1-carboxylate 23.1a in refluxing methanol in the presence of pyrrolidine to provide the derivative 47.9 which was converted to the silyl ether derivative 47.10 using tert-butyldimethylsilyl chloride 2.3. Conversion of the ketone 47.10 to the enol triflate derivative 47.11 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent.
  • the derivatives 58.1a and 58.1b were then converted to the final products 58A and 58B, respectively, under acidic conditions.
  • TBAF tetrabutylammonium fluoride
  • the Boc protecting group of 59.9 was removed using hydrochloric acid to generate the compounds 59A-59D.
  • Treatment of the amines 59.9 with methane sulfonyl chloride in dichloromethane in the presence of triethylamine provided the sulfonamides 59.11, which were converted to the final products 59I-59L under acidic conditions.
  • the compounds of the invention have the potential for chirality but were prepared in racemic form.
  • the racemic mixtures of intermediates or final products initially prepared as their racemates may be partially or completely resolved into any, some, or all of the enantiomers contained in the racemate as described herein, for example in the separation of intermediates leading to 39F and 39G.
  • racemic mixtures, mixtures enriched in or more stereoisomers, and pure enantiomers are considered to be within the ambit of the present invention.
  • 51A and 51B are chirally resolved products derived from 51C. Their absolute stereochemistries have not been conclusively established.
  • 52B and 52C are diastereomeric with respect to one another. 52B and 52C are chirally pure. Their absolute stereochemistry has not been conclusively established.
  • 21B and 21C are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 21D and 21E are diastereomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 24B and 24C are geometric isomers with respect to one another (wherein the hydroxyl is either equatorial or axial), but the conformation of each has not been conclusively established.
  • 24D and 24E are geometric isomers with respect to one another (wherein the hydroxyl is either equatorial or axial), but the conformation of each has not been conclusively established.
  • 24F and 24G are geometric isomers with respect to one another (wherein the hydroxyl is either equatorial or axial), but the conformation of each has not been conclusively established.
  • 27B and 27C are enantiomeric with respect to one another, and their absolute stereochemistry has been conclusively established using X-ray crystallography.
  • 27E and 27F are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27I and 27J are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27L and 27M are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27O and 27P are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27R and 27S are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27U and 27V are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 39B and 39C are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 39F and 39G are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 43A and 43B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 43D and 43E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 44A and 44B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 44D and 44E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 45A and 45B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 45D and 45E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 46A and 46B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 51A and 51B are resolvable by chiral column chromatography as individual peaks with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 61A and 61B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • LC-MS data were obtained using a Thermo-Finnigan Surveyor HPLC and a Thermo-Finnigan AQA MS using either positive or negative electrospray ionization.
  • Program (positive) Solvent A: 10 mM ammonium acetate, pH 4.5, 1% acetonitrile; solvent B: acetonitrile; column: Michrom Bioresources Magic C18 Macro Bullet, detector: PDA ⁇ 220-300 nm. Gradient: 96% A-100% B in 3.2 minutes, hold 100% B for 0.4 minutes.
  • the compounds set forth in Tables 1 and 2 are actual examples.
  • the compounds set forth in Table 3 include actual and prophetic examples for which schemes and general procedure descriptions are set forth herein.
  • Method 1A Pyrrolidine (6.12 mL, 73.38 mmol, 2.0 eq) was added at room temperature to 1.2 (7.31 g, 36.69 mmol, 1.0 eq) and 1.1a (5.00 g, 36.69 mmol, 1.0 eq). The solution was stirred overnight at room temperature and then concentrated under reduced pressure. Diethyl ether (500 mL) was added. The organic mixture was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. Hexane (300 mL) was added to the mixture. The resulting precipitate was collected by filtration, washed with hexane and used for the next step without further purification. Yield: 68%
  • Method 1B Pyrrolidine (42 mL, 73.38, 2.0 eq) was added drop wise at room temperature to a solution of 1.2 (49.8 g, 0.249 mol, 1.0 eq) and 1.1a (34 g, 0.184 mol, 1.0 eq) in anhydrous methanol (400 mL). The solution was refluxed overnight and then concentrated under reduced pressure. Diethyl ether (500 mL) was added. The organic mixture was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. Hexane (300 mL) was added to the mixture. The resulting precipitate was collected by filtration, washed with hexane, and used for the next step without further purification.
  • the mixture was then poured into ice water and the two phases were separated.
  • the organic phase was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate.
  • the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Method 1C To a solution of 1.5a (15 g, 33.37 mmol, 1.0 eq) in dimethoxyethane (100 mL) was added sequentially a 2N aqueous solution of sodium carbonate (50.06 mL, 100.12 mmol, 3.0 eq), lithium chloride (4.24 g, 100.12 mmol, 3.0 eq), 1.6 (8.12 g, 36.71 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.77 g, 0.67 mmol, 0.02 eq). The mixture was refluxed for 10 h under nitrogen.
  • Method 1D To a solution of 1.5a (10 g, 22.25 mmol, 1.0 eq) in dimethoxyethane (67 mL) was added sequentially a 2N aqueous solution of sodium carbonate (33.37 mL, 66.75 mmol, 3.0 eq), lithium chloride (2.83 g, 66.75 mmol, 3.0 eq), 1.6 (4.40 g, 24.47 mmol, 1.1 eq) and palladium, 10 weight % (dry basis) on activated carbon, wet, Degussa type E101 NE/W (0.24 g, 0.11 mmol, 0.005 eq). The mixture was refluxed for 2 h under nitrogen.
  • Method 1E A 2.0M solution of hydrochloric acid in diethyl ether (34.6 mL, 69.24 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 1.8a (6.00 g, 12.59 mmol, 1.0 eq) in anhydrous dichloromethane (70 mL). The mixture was warmed to room temperature and stiffing was continued for an additional 10 h. Diethyl ether (100 mL) was added to the solution and the resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 99%
  • Method 1F Trifluoroacetic acid (10.33 mL, 134.09 mmol, 5.5 eq) was added drop wise to a cold (0° C.) solution of 1.8a (11.62 g, 24.38 mmol, 1.0 eq) in anhydrous dichloromethane (50 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h. The mixture was then concentrated under reduced pressure. A saturated solution of sodium bicarbonate (100 mL) was added to the mixture, which was extracted with dichloromethane. The organic phase was separated, washed with brine, dried over sodium sulfate and concentrated under reduced pressure.
  • Step 1.1 1.1a was replaced by 1.1b and Method 1B was used.
  • Step 1.3 Method 1C was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1c and Method 1A was used.
  • Step 1.3 Method 1C was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1d and Method 1B was used.
  • Step 1.3 Method 1D was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1e and Method 1A was used.
  • Step 1.3 Method 1D was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1f and Method 1B was used.
  • Step 1.3 Method 1C was used.
  • Step 1.4 Method 1F was used.
  • Step 1.1 1.1a was replaced by 1.1 g and Method 1B was used.
  • Step 1.3 Method 1C was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1h and Method 1B was used.
  • Step 1.3 Method 1D was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1i and Method 1B was used.
  • Step 1.3 Method 1D was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1j and Method 1A was used.
  • Step 1.3 Method 1D was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1k and Method 1B was used.
  • Step 1.3 Method 1C was used.
  • Step 1.4 Method 1F was used.
  • Step 1.1 1.1a was replaced by 1.11 and Method 1B was used.
  • Step 1.3 Method 1D was used.
  • Step 1.4 Method 1E was used.
  • Step 1.1 1.1a was replaced by 1.1m and Method 1B was used.
  • Step 1.3 Method 1C was used.
  • Step 1.4 Method 1E was used.

Abstract

Spirocyclic heterocyclic derivatives, pharmaceutical compositions containing these compounds, and methods for their pharmaceutical use are disclosed. In certain embodiments, the spirocyclic heterocyclic derivatives are ligands of the δ opioid receptor and may be useful, inter alia, for treating and/or preventing pain, anxiety, gastrointestinal disorders, and other δ opioid receptor-mediated conditions.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. patent application Ser. No. 12/542,302, filed Aug. 17, 2009, now allowed, which is a divisional of U.S. patent application Ser. No. 11/393,133, filed Mar. 30, 2006, now U.S. Pat. No. 7,598,261, which claims priority to U.S. Provisional Application Ser. No. 60/667,177, filed Mar. 31, 2005, the disclosures of which are each hereby incorporated herein by reference in their entireties.
  • FIELD OF THE INVENTION
  • The invention relates to spirocyclic heterocyclic derivatives (including derivatives of spiro(2H-1-benzopyran-2,4′-piperidines), pharmaceutical compositions containing these compounds, and methods for their pharmaceutical use. In certain embodiments, the spirocyclic heterocyclic derivatives are ligands of the δ opioid receptor and are useful, inter alia, for treating and/or preventing pain, anxiety, gastrointestinal disorders, and other δ opioid receptor-mediated conditions.
  • BACKGROUND OF THE INVENTION
  • There are at least three different opioid receptors (μ, δ and κ) that are present in both central and peripheral nervous systems of many species, including humans. Lord, J. A. H., et al., Nature, 1977, 267, 495. Activation of the δ opioid receptors induces analgesia in various animal models. Moulin, et al., Pain, 1985, 23, 213. Some work suggests that the analgesics working at δ opioid receptors do not have the attendant side effects associated with μ and κ opioid receptor activation. Galligan, et al., J. Pharm. Exp. Ther., 1985, 229, 641. The δ opioid receptor has also been identified as having a role in circulatory systems. Ligands for the δ receptor have also been shown to possess immunomodulatory activities. Dondio, et al., Exp. Opin. Ther. Patents, 1997, 10, 1075. Further, selective δ opioid receptor agonists have been shown to promote organ and cell survival. Su, T-P, Journal of Biomedical Science, 2000, 9(3), 195-199. Ligands for the δ opioid receptor may therefore find potential use as analgesics, as antihypertensive agents, as immunomodulatory agents, and/or agents for the treatment of cardiac disorders.
  • Numerous selective δ opioid ligands are peptidic in nature and thus are unsuitable for administration by systemic routes. Several non-peptidic δ opioid receptor ligands have been developed. See, for example, E. J. Bilsky, et al., Journal of Pharmacology and Experimental Therapeutics, 1995, 273(1), 359-366; WO 93/15062, WO 95/04734, WO 95/31464, WO 96/22276, WO 97/10216, WO 01/46192, WO 02/094794, WO 02/094810, WO 02/094811, WO 02/094812, WO 02/48122, WO 03/029215, WO 03/033486, JP-4275288, EP-A-0,864,559, U.S. Pat. No. 5,354,863, U.S. Pat. No. 6,200,978, U.S. Pat. No. 6,436,959 and US 2003/0069241.
  • While there are a large number of non-peptidic δ opioid receptor modulators, there is still an unfulfilled need for compounds with selective δ opioid receptor activity that may be used in methods to provide beneficial pharmaceutical characteristics while minimizing undesirable side effects. The present invention is directed to these, as well as other important ends.
  • SUMMARY OF THE INVENTION
  • In one embodiment, the invention is directed to compounds of formula XIV:
  • Figure US20110218186A1-20110908-C00001
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl, provided that at least one of R23 and R24 is alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In another embodiment, the invention is directed to compounds of formula XVII:
  • Figure US20110218186A1-20110908-C00002
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 0-3 groups selected independently from halo, hydroxy, and —S(═O)2-alkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when W2 is para-diethylaminocarbonylphenyl, X2 is O, and A2 and B2 together form a double bond, then the aryl ring of J2 is substituted with at least one group selected independently from halo and —S(═O)2-alkyl in which the alkyl group is C2-C6 alkyl;
      • when W2 is para-diethylaminocarbonylphenyl, X2 is O, and A2 and B2 are each H, then the aryl ring of J2 is substituted with 1-3 groups selected independently from halo, hydroxy, and —S(═O)2-alkyl; and
      • the compound of formula XVII is other than:
  • Figure US20110218186A1-20110908-C00003
  • In yet another embodiment, the invention is directed to compounds of formula XX:
  • Figure US20110218186A1-20110908-C00004
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted independently with 0-3 hydroxy or halo groups;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that the compound of formula XX is other than 4-[(4-N,N-diethylaminocarbonyl)phenyl]-spiro[2H,1-benzopyran-2,3′-pyrrolidine].
  • In still another embodiment, the invention is directed to compounds of formula XXII:
  • Figure US20110218186A1-20110908-C00005
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from heteroaryl, hydroxy, carboxy (—COOH), —C(═O)-alkyl, —C(═O)-aryl, —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl); aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 0-3 groups selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), carboxy (—COOH), —C(═O)—O-alkyl, and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when W2 is para-diethylaminocarbonylphenyl, para-prop-2-ylaminocarbonylphenyl, or para-pent-3-ylaminocarbonylphenyl, R23 and R24 are each H, and A and B are each H or together form a double bond, then J2 is other than unsubstituted phenyl or anisyl; and
      • when W2 is:
  • Figure US20110218186A1-20110908-C00006
      • R23 and R24 are each H, and A and B together form a double bond; then J2 is other than unsubstituted phenyl.
  • In another embodiment, the invention is directed to compounds of formula XXV:
  • Figure US20110218186A1-20110908-C00007
  • wherein:
      • W2 is aryl optionally substituted with —C(═O)-alkyl or —C(═O)-aryl;
      • R23 and R24 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 0-3 groups selected independently from hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—N(alkyl)(alkyl), carboxy (—COOH), and —C(═O)—O-alkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
      • provided that the compound of formula XXV is other than 4-phenyl-spiro[2H,1-benzopyran-2,4′-piperidine].
  • In yet another embodiment, the invention is directed to compounds of formula XXVII:
  • Figure US20110218186A1-20110908-C00008
  • wherein:
      • W2 is para-dialkylaminocarbonylphenyl, the phenyl group of which is further optionally substituted with 1-2 groups independently selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2);
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • Q1 and Q2 are each independently H, hydroxy, alkoxy, haloalkoxy, halo, or heterocycloalkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when one of Q1 and Q2 is hydroxy and the other is H, or both Q1 and Q2 are hydroxy, then the phenyl group of W2 is further substituted with 1-2 groups selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2);
      • when Q1, Q2, R23, and R24 are each H and the phenyl group of W2 is further substituted with one hydroxy, then A2 and B2 are each H;
      • when W2 is para-dialkylaminocarbonylphenyl, then at least one of Q1, Q2, R23, and R24 is other than H;
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, and Q2 is halo, then Q1 is other than H or hydroxy;
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, Q1 is methoxy or cyclopropylmethoxy and Q2 is H, then A2 and B2 are each H; and
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, and Q1 is H or OH, then Q2 is other than methoxy or cyclopropylmethoxy.
  • In still another embodiment, the invention is directed to compounds of formula XXVIII:
  • Figure US20110218186A1-20110908-C00009
  • wherein:
      • D is:
  • Figure US20110218186A1-20110908-C00010
      • K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
      • R23, R24, and R26 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
  • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In another embodiment, the invention is directed to compounds of formula XXIX:
  • Figure US20110218186A1-20110908-C00011
  • wherein:
      • W2 is para-N(alkyl),N(alkyl-Z)aminocarbonylaryl or para-N(alkyl),N(alkyl-Z)aminocarbonylheteroaryl, wherein the aryl or heteroaryl ring of W2 is substituted with 0-2 groups selected independently from hydroxy and alkoxy;
      • Z is alkoxy, alkylamino, or dialkylamino;
      • R23 and R24 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In yet another embodiment, the invention is directed to compounds of formula XXX:
  • Figure US20110218186A1-20110908-C00012
  • wherein:
      • W2 is:
  • Figure US20110218186A1-20110908-C00013
      • R23 and R24 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 1-3 groups selected independently from halo or haloalkoxy;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
      • provided that when W2 is:
  • Figure US20110218186A1-20110908-C00014
  • then the aryl ring of J2 is substituted with at least one haloalkoxy.
  • In another embodiment, the invention is directed to compounds of formula XXXII:
  • Figure US20110218186A1-20110908-C00015
  • wherein:
      • D is N(alkyl),N(alkyl)aminocarbonylheteroaryl;
      • R23, R24, and R26 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that when D is:
  • Figure US20110218186A1-20110908-C00016
  • and X2 is —O—, then A2 and B2 are each H.
  • In one embodiment, the invention is directed to compounds of formula XXXIII:
  • Figure US20110218186A1-20110908-C00017
  • wherein:
      • F1 is heteroaryl; and
      • G is C1-6alkylene substituted with NH2, NHC(═O)alkyl, NH(C(O)N(H)alkyl, or NHS(═O)2alkyl;
  • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In another embodiment, the invention is directed to compounds of formula XXXIV:
  • Figure US20110218186A1-20110908-C00018
  • wherein:
      • F2 is aryl or heteroaryl; and
      • Q3 is hydroxy or alkoxy;
  • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In still another embodiment, the invention is directed to pharmaceutical compositions, comprising:
  • a pharmaceutically acceptable carrier; and a compound as described herein including, for example, a compound of formula XIV, XVII, XX, XXII, XXV, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXII, XXXIII, and/or XXXIV.
  • In still another embodiment, the invention is directed to methods of binding opioid receptors in a patient in need thereof, comprising the step of:
  • administering to said patient an effective amount of a compound as described herein including, for example, a compound of formula XIV, XVII, XX, XXII, XXV, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXII, XXXIII, and/or XXXIV.
  • These and other embodiments of the invention will become more apparent from the following detailed description.
  • DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
  • The invention relates to spirocyclic heterocyclic derivatives, pharmaceutical compositions containing these compounds, and methods for their pharmaceutical use. This invention is related by subject matter to co-pending U.S. application Ser. No. 12/787,906, filed May 26, 2010, application Ser. No. 12/718,439, filed Mar. 5, 2010, application Ser. No. 12/483,781, filed Jun. 12, 2009, application Ser. No. 11/960,845, filed Dec. 20, 2007, now U.S. Pat. No. 7,638,527, application Ser. No. 10/957,554, filed Oct. 1, 2004, now U.S. Pat. No. 7,338,962, and Provisional Application Ser. No. 60/507,864, filed Oct. 1, 2003, the disclosures of which are hereby incorporated herein by reference, in their entireties.
  • In certain embodiments, the spirocyclic heterocyclic derivatives are ligands of the δ opioid receptor and may be useful, inter alia, in methods for treating and/or preventing diseases and conditions that may be mediated or modulated by the δ opioid receptor including, for example, pain, gastrointestinal disorders, urogenital tract disorders including incontinence and overactive bladder, immunomodulatory disorders, inflammatory disorders, respiratory function disorders, anxiety, mood disorders, stress-related disorders, attention deficit hyperactivity disorders, sympathetic nervous system disorders, depression, tussis, motor disorders, traumatic injuries, especially to the central nervous system, stroke, cardiac arrhythmias, glaucoma, sexual dysfunctions, shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, rejections in organ transplants and skin grafts, and substance addiction. In certain other embodiments, the spirocyclic heterocyclic derivatives are ligands of the δ opioid receptor and may be useful in, inter alia, methods for improving organ and cell survival, methods for providing cardioprotection following myocardial infarction, methods for reducing the need for anesthesia, methods for producing and/or maintaining an anaesthetic state, and methods of detecting, imaging or monitoring degeneration or dysfunction of opioid receptors in a patient.
  • As employed above and throughout the disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings.
  • “Alkyl” refers to an optionally substituted, saturated straight, or branched, hydrocarbon having from about 1 to about 20 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), preferably from about 1 to about 8 carbon atoms, herein referred to as “lower alkyl,” more preferably from about 1 to about 6, still more preferably from about 1 to about 4, with from about 2 to about 3 being most preferred. In certain alternative preferred embodiments, the alkyl group, more preferably, has one carbon atom. Alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, n-pentyl, cyclopentyl, isopentyl, neopentyl, n-hexyl, isohexyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl.
  • As used herein, “alkylene” refers to an optionally substituted bivalent alkyl radical having the general formula —(CH2)n—, where n is 1 to 10, preferably 1 to 6, with 1 to 4 being most preferred. In alternative embodiments, n is preferably 4 to 6. Non-limiting examples include methylene, dimethylene, trimethylene, tetramethylene, pentamethylene, and hexamethylene.
  • “Cycloalkyl” refers to an optionally substituted alkyl group having one or more rings in their structures and having from about 3 to about 20 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 3 to about 10 carbon atoms being preferred. Multi-ring structures may be bridged or fused ring structures. Cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl, 2-[4-isopropyl-1-methyl-7-oxa-bicyclo[2.2.1]heptanyl], 2-[1,2,3,4-tetrahydro-naphthalenyl], and adamantyl.
  • “Alkylcycloalkyl” refers to an optionally substituted ring system comprising a cycloalkyl group having one or more alkyl substituents, wherein cycloalkyl and alkyl are each as previously defined. Exemplary alkylcycloalkyl groups include, for example, 2-methylcyclohexyl, 3,3-dimethylcyclopentyl, trans-2,3-dimethylcyclooctyl, and 4-methyldecahydronaphthalenyl.
  • “Heterocycloalkyl” refers to an optionally substituted ring system composed of a cycloalkyl radical wherein in at least one of the rings, one or more of the carbon atom ring members is independently replaced by a heteroatom group selected from the group consisting of O, S, N, and NH, wherein cycloalkyl is as previously defined. Heterocycloalkyl ring systems having a total of from about 5 to about 14 carbon atom ring members and heteroatom ring members (and all combinations and subcombinations of ranges and specific numbers of carbon and heteroatom ring members) are preferred. In other preferred embodiments, the heterocyclic groups may be fused to one or more aromatic rings. Exemplary heterocycloalkyl groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, piperazinyl, morpholinyl, piperadinyl, decahydroquinolyl, octahydrochromenyl, octahydro-cyclopenta[c]pyranyl, 1,2,3,4,-tetrahydroquinolyl, octahydro-[2]pyrindinyl, decahydro-cycloocta[c]furanyl, tetrahydroquinolyl, and imidazolidinyl.
  • “Alkylheterocycloalkyl” refers to an optionally substituted ring system comprising a heterocycloalkyl group having one or more alkyl substituents, wherein heterocycloalkyl and alkyl are each as previously defined. Exemplary alkylheterocycloalkyl groups include, for example, 2-methylpiperidinyl, 3,3-dimethylpyrrolidinyl, trans-2,3-dimethylmorpholinyl, and 4-methyldecahydroquinolinyl.
  • “Alkenyl” refers to an optionally substituted alkyl group having from about 2 to about 10 carbon atoms and one or more double bonds (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), wherein alkyl is as previously defined.
  • “Alkynyl” refers to an optionally substituted alkyl group having from about 2 to about 10 carbon atoms and one or more triple bonds (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), wherein alkyl is as previously defined.
  • “Aryl” refers to an optionally substituted, mono-, di-, tri-, or other multicyclic aromatic ring system having from about 5 to about 50 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 6 to about 10 carbons being preferred. Non-limiting examples include, for example, phenyl, naphthyl, anthracenyl, and phenanthrenyl.
  • “Aralkyl” refers to an optionally substituted moiety composed of an alkyl radical bearing an aryl substituent and having from about 6 to about 50 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 6 to about 10 carbon atoms being preferred. Non-limiting examples include, for example, benzyl, diphenylmethyl, triphenylmethyl, phenylethyl, and diphenylethyl.
  • “Halo” refers to a fluoro, chloro, bromo, or iodo moiety, preferably fluoro.
  • “Heteroaryl” refers to an optionally substituted aryl ring system wherein in at least one of the rings, one or more of the carbon atom ring members is independently replaced by a heteroatom group selected from the group consisting of S, O, N, and NH, wherein aryl is as previously defined. Heteroaryl groups having a total of from about 5 to about 14 carbon atom ring members and heteroatom ring members (and all combinations and subcombinations of ranges and specific numbers of carbon and heteroatom ring members) are preferred. Exemplary heteroaryl groups include, but are not limited to, pyrryl, furyl, pyridyl, 1,2,4-thiadiazolyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, tetrazolyl, pyrazinyl, pyrimidyl, quinolyl, isoquinolyl, thiophenyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, purinyl, carbazolyl, benzimidazolyl, and isoxazolyl. Heteroaryl may be attached via a carbon or a heteroatom to the rest of the molecule.
  • “Heteroarylalkyl” and “heteroaralkyl” each refers to an optionally substituted, heteroaryl substituted alkyl radical where heteroaryl and alkyl are as previously defined Non-limiting examples include, for example, 2-(1H-pyrrol-3-yl)ethyl, 3-pyridylmethyl, 5-(2H-tetrazolyl)methyl, and 3-(pyrimidin-2-yl)-2-methylcyclopentanyl.
  • “Perhaloalkyl” refers to an alkyl group, wherein two or more hydrogen atoms are replaced by halo (F, Cl, Br, I) atoms, and alkyl is as previously defined. Exemplary perhaloalkyl groups include, for example, perhalomethyl, such as perfluoromethyl and difluoromethyl.
  • “Alkoxy” and “alkoxyl” refer to an optionally substituted alkyl-O— group wherein alkyl is as previously defined. Exemplary alkoxy and alkoxyl groups include, for example, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, and heptoxy.
  • “Alkenyloxy” refers to an optionally substituted alkenyl-O— group wherein alkenyl is as previously defined. Exemplary alkenyloxy and alkenyloxyl groups include, for example, allyloxy, butenyloxy, heptenyloxy, 2-methyl-3-buten-1-yloxy, and 2,2-dimethylallyloxy.
  • “Alkynyloxy” refers to an optionally substituted alkynyl-O— group wherein alkynyl is as previously defined. Exemplary alkynyloxy and alkynyloxyl groups include, for example, propargyloxy, butynyloxy, heptynyloxy, 2-methyl-3-butyn-1-yloxy, and 2,2-dimethylpropargyloxy.
  • “Aryloxy” and “aryloxyl” refer to an optionally substituted aryl-O— group wherein aryl is as previously defined. Exemplary aryloxy and aryloxyl groups include, for example, phenoxy and naphthoxy.
  • “Aralkoxy” and “aralkoxyl” refer to an optionally substituted aralkyl-O-group wherein aralkyl is as previously defined. Exemplary aralkoxy and aralkoxyl groups include, for example, benzyloxy, 1-phenylethoxy, 2-phenylethoxy, and 3-naphthylheptoxy.
  • “Cycloalkoxy” refers to an optionally substituted cycloalkyl-O— group wherein cycloalkyl is as previously defined. Exemplary cycloalkoxy groups include, for example, cyclopropanoxy, cyclobutanoxy, cyclopentanoxy, cyclohexanoxy, and cycloheptanoxy.
  • “Heteroaryloxy” refers to an optionally substituted heteroaryl-O— group wherein heteroaryl is as previously defined. Exemplary heteroaryloxy groups include, but are not limited to, pyrryloxy, furyloxyl, pyridyloxy, 1,2,4-thiadiazolyloxy, pyrimidyloxy, thienyloxy, isothiazolyloxy, imidazolyloxy, tetrazolyloxy, pyrazinyloxy, pyrimidyloxy, quinolyloxy, isoquinolyloxy, thiophenyloxy, benzothienyloxy, isobenzofuryloxy, pyrazolyloxy, indolyloxy, purinyloxy, carbazolyloxy, benzimidazolyloxy, and isoxazolyloxy.
  • “Heteroaralkoxy” refers to an optionally substituted heteroarylalkyl-O-group wherein heteroarylalkyl is as previously defined. Exemplary heteroaralkoxy groups include, but are not limited to, pyrrylethyloxy, furylethyloxy, pyridylmethyloxy, 1,2,4-thiadiazolylpropyloxy, pyrimidylmethyloxy, thienylethyloxy, isothiazolylbutyloxy, and imidazolyl-2-methylpropyloxy.
  • “Heterocycloalkylaryl” refers to an optionally substituted ring system composed of an aryl radical bearing a heterocycloalkyl substituent wherein heterocycloalkyl and aryl are as previously defined. Exemplary heterocycloalkylaryl groups include, but are not limited to, morpholinylphenyl, piperidinylnaphthyl, piperidinylphenyl, tetrahydrofuranylphenyl, and pyrrolidinylphenyl.
  • “Alkylheteroaryl” refers to an optionally substituted ring system composed of a heteroaryl radical bearing an alkyl substituent wherein heteroaryl and alkyl are as previously defined. Exemplary alkylheteroaryl groups include, but are not limited to, methylpyrryl, ethylfuryl, 2,3-dimethylpyridyl, N-methyl-1,2,4-thiadiazolyl, propylpyrimidyl, 2-butylthienyl, methylisothiazolyl, 2-ethylimidazolyl, butyltetrazolyl, 5-ethylbenzothienyl, and N-methylindolyl. Alkyheteroaryl groups may be attached via a carbon or a heteroatom to the rest of the molecule.
  • “Heteroarylaryl” refers to an optionally substituted ring system composed of an aryl radical bearing a heteroaryl substituent wherein heteroaryl and aryl are as previously defined. Exemplary heteroarylaryl groups include, but are not limited to, pyrrylphenyl, furylnaphthyl, pyridylphenyl, 1,2,4-thiadiazolylnaphthyl, pyrimidylphenyl, thienylphenyl, isothiazolylnaphthyl, imidazolylphenyl, tetrazolylphenyl, pyrazinylnaphthyl, pyrimidylphenyl, quinolylphenyl, isoquinolylnaphthyl, thiophenylphenyl, benzothienylphenyl, isobenzofurylnaphthyl, pyrazolylphenyl, indolylnaphthyl, purinylphenyl, carbazolylnaphthyl, benzimidazolylphenyl, and isoxazolylphenyl. Heteroarylaryl may be attached via a carbon or a heteroatom to the rest of the molecule.
  • “Alkylheteroarylaryl” refers to an optionally substituted ring system composed of an aryl radical bearing an alkylheteroaryl substituent and have from about 12 to about 50 carbon atoms (and all combinations and subcombinations of ranges and specific numbers of carbon atoms therein), with from about 12 to about 30 carbon atoms being preferred wherein aryl and alkylheteroaryl are as previously defined. Exemplary heteroarylaryl groups include, but are not limited to, methylpyrrylphenyl, ethylfurylnaphthyl, methylethylpyridylphenyl, dimethylethylpyrimidylphenyl, and dimethylthienylphenyl.
  • Typically, substituted chemical moieties include one or more substituents that replace hydrogen. Exemplary substituents include, for example, halo (e.g., F, Cl, Br, I), alkyl, cycloalkyl, alkylcycloalkyl, alkenyl, alkynyl, aralkyl, aryl, heteroaryl, heteroaralkyl, spiroalkyl, heterocycloalkyl, hydroxyl (—OH), oxo (═O), alkoxyl, aryloxyl, aralkoxyl, nitro (—NO2), cyano (—CN), amino (—NH2), —N-substituted amino (—NHR″), -N,N-disubstituted amino (—N(R″)R″), carboxyl (—COOH), —C(═O)R″, —OR″, —C(═O)OR″, —C(═O)NHSO2R″, —NHC(═O)R″, aminocarbonyl (—C(═O)NH2), —N-substituted aminocarbonyl (—C(═O)NHR″), -N,N-disubstituted aminocarbonyl (—C(═O)N(R″)R″), thiol, thiolato (SR″), sulfonic acid and its esters (SO3R″), phosphonic acid and its mono-esters (P(═O)OR″OH) and di-esters (P(═O)OR″OR″), S(═O)2R″, S(═O)2NH2, S(═O)2NHR″, S(═O)2NR″R″, SO2NHC(═O)R″, NHS(═O)2R″, NR″S(═O)2R″, CF3, CF2CF3, NHC(═O)NHR″, NHC(═O)NR″R″, NR″C(═O)NHR″, NR″C(═O)NR″R″, NR″C(═O)R″, NR″C(═N—CN)NR″R″, and the like. Aryl substituents may also include (CH2)pSO2NR″(CH2)q and (CH2)pCO2NR″(CH2)q, where p and q are independently integers from 0 to 3, where the methylene units are attached in a 1,2 arrangement yielding substituted aryls of the type:
  • Figure US20110218186A1-20110908-C00019
  • In relation to the aforementioned substituents, each moiety R″ can be, independently, any of H, alkyl, cycloalkyl, alkenyl, aryl, aralkyl, heteroaryl, or heterocycloalkyl, or when (R″(R″)) is attached to a nitrogen atom, R″ and R″ can be taken together to form a 4- to 8-membered nitrogen heterocycloalkyl ring, wherein said heterocycloalkyl ring is optionally interrupted by one or more additional —O—, —S—, —SO, —SO2—, —NH—, —N(alkyl)-, or —N(aryl)- groups, for example.
  • As used herein, an “*” denotes the presence of a non-racemic stereoisomeric center in a molecule, wherein one stereoisomeric form (R or S) predominates, but for which the absolute configuration at this center has not been conclusively established. This is equivalently expressed by saying that the molecule's configuration at the asterisked carbon atom is either greater than 50% R or greater than 50% S. More preferably the compound, or its stereoisomeric center, is “substantially enriched”, and even more preferably is substantially enantiomerically pure.
  • As used herein, the term “substantially enriched”, when referring to a stereoisomer or stereoisomeric center, denotes that at least about 60%, preferably about 70%, more preferably about 80%, still more preferably about 90% of one stereoisomer or one stereoisomeric center predominating in the mixture, with at least about 95% of one stereoisomer or one stereoisomeric center being even more preferred. In some preferred embodiments, the compound is “substantially enantiomerically pure”, that is, at least about 97.5%, more preferably about 99%, even more preferably about 99.5% of one stereoisomeric forms predominates. For example, a compound having one stereoisomeric center may be represented by one of two stereoisomeric forms (R or S), differing only in the spatial arrangement of atoms about a single carbon atom. The “*” denotes non-equal amounts of the two isomers. When a compound has two or more stereoisomeric centers, each center denoted by an asterisk is evaluated individually. A predominance of one stereoisomeric form (R or S) occurring at least one center is considered non-racemic within the definition herein provided. The range of possible non-racemic compounds extends from the point at which a stereoisomeric form predominates at a single chiral center and includes all combinations and subcombinations up to and including the compound wherein all stereoisomeric centers in the compound are each individually R or S.
  • Use of the “*” can be expressed, for example in a compound's identification number such as 4*, and indicates that the stereochemical configuration of at least one chiral center of the identified compound has not been established. The specific center is identified within a structure by placing the “*” adjacent the chiral center in question, such as, for example, in the structure below.
  • Figure US20110218186A1-20110908-C00020
  • In some compounds, several chiral centers may be present. The presence of two asterisks “*” in a single structure indicates that two racemic pairs may be present, but that each pair is diastereomeric relative to the other pair. As such, the first pair of enantiomers having two chiral centers may have the configurations, for example, (R,R) and (S,S). The second pair then have configurations, for example, (R,S) and (S,R). Alternatively, where only two stereoisomers bearing an enantiomeric relationship to each other are initially provided, such as the (R,R) and (S,S) pair, the asterisks may indicate that the enantiomers have been enriched (partially resolved) or preferably fully resolved, into the individual enantiomers.
  • “Ligand” or “modulator” refers to a compound that binds to a receptor to form a complex, and includes, agonists, partial agonists, antagonists and inverse agonists.
  • “Agonist” refers to a compound that may bind to a receptor to form a complex that may elicit a full pharmacological response, which is typically peculiar to the nature of the receptor involved and which may alter the equilibrium between inactive and active receptor.
  • “Partial agonist” refers to a compound that may bind to a receptor to form a complex that may elicit only a proportion of the full pharmacological response, typically peculiar to the nature of the receptor involved, even if a high proportion of the receptors are occupied by the compound.
  • “Antagonist” refers to a compound that may bind to a receptor to form a complex that may not elicit any response, typically in the same manner as an unoccupied receptor, and which preferably does not alter the equilibrium between inactive and active receptor.
  • “Inverse agonist” refers to a compound that may bind to a receptor to form a complex that may preferentially stabilize the inactive conformation of the receptor.
  • “Prodrug” refers to compounds specifically designed to maximize the amount of active species that reaches the desired site of reaction that are themselves typically inactive or minimally active for the activity desired, but through biotransformation are converted into biologically active metabolites.
  • “Stereoisomers” refers to compounds that have identical chemical constitution, but differ as regards the arrangement of the atoms or groups in space.
  • “N-oxide” refers to compounds wherein the basic nitrogen atom of either a heteroaromatic ring or tertiary amine is oxidized to give a quaternary nitrogen bearing a positive formal charge and an attached oxygen atom bearing a negative formal charge.
  • “Hydrate” refers to a compound as described herein which is associated with water in the molecular form, i.e., in which the H—OH bond is not split, and may be represented, for example, by the formula R.H2O, where R is a compound as described herein. A given compound may form more than one hydrate including, for example, monohydrates (R.H2O), dihydrates (R.2H2O), trihydrates (R.3H2O), and the like.
  • As used herein, “haloalkoxy” refers to an alkoxy group, wherein one, preferably two or more, hydrogen(s) of the alkyl moiety of said alkoxy are replaced by halo atoms, and alkoxy, alkyl, and halo are each as previously defined.
  • “Solvate” refers to a compound of the present invention which is associated with solvent in the molecular form, i.e., in which the solvent is coordinatively bound, and may be represented, for example, by the formula R.(solvent), where R is a compound of the invention. A given compound may form more than one solvate including, for example, monosolvates (R.(solvent)) or polysolvates (R.n(solvent)) wherein n is an integer >1) including, for example, disolvates (R.2(solvent)), trisolvates (R.3(solvent)), and the like, or hemisolvates, such as, for example, R.n/2(solvent), R.n/3(solvent), R.n/4(solvent) and the like wherein n is an integer. Solvents herein include mixed solvents, for example, methanol/water, and as such, the solvates may incorporate one or more solvents within the solvate.
  • “Acid hydrate” refers to a complex that may be formed through association of a compound having one or more base moieties with at least one compound having one or more acid moieties or through association of a compound having one or more acid moieties with at least one compound having one or more base moieties, said complex being further associated with water molecules so as to form a hydrate, wherein said hydrate is as previously defined and R represents the complex herein described above.
  • “Pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like. These physiologically acceptable salts are prepared by methods known in the art, e.g., by dissolving the free amine bases with an excess of the acid in aqueous alcohol, or neutralizing a free carboxylic acid with an alkali metal base such as a hydroxide, or with an amine.
  • Compounds described herein throughout can be used or prepared in alternate forms. For example, many amino-containing compounds can be used or prepared as an acid addition salt. Often such salts improve isolation and handling properties of the compound. For example, depending on the reagents, reaction conditions and the like, compounds as described herein can be used or prepared, for example, as their hydrochloride or tosylate salts. Isomorphic crystalline forms, all chiral and racemic forms, N-oxide, hydrates, solvates, and acid salt hydrates, are also contemplated to be within the scope of the present invention.
  • Certain acidic or basic compounds as described herein may exist as zwitterions. All forms of the compounds, including free acid, free base and zwitterions, are contemplated to be within the scope of the present invention. It is well known in the art that compounds containing both basic nitrogen atom and acidic groups often exist in equilibrium with their zwitterionic forms. Thus, any of the compounds described herein throughout that contain, for example, both basic nitrogen and acidic groups, also include reference to their corresponding zwitterions.
  • “Effective amount” refers to an amount of a compound as described herein that may be therapeutically effective to inhibit, prevent or treat the symptoms of particular disease, disorder, condition, or side effect. Such diseases, disorders, conditions, and side effects include, but are not limited to, those pathological conditions associated with the binding of δ opioid receptor (for example, in connection with the treatment and/or prevention of pain), wherein the treatment or prevention comprises, for example, agonizing the activity thereof by contacting cells, tissues or receptors with compounds as described herein. Thus, for example, the term “effective amount”, when used in connection with compounds as described herein, opioids, or opioid replacements, for example, for the treatment of pain, refers to the treatment and/or prevention of the painful condition. The term “effective amount,” when used in connection with compounds active against gastrointestinal dysfunction, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with gastrointestinal dysfunction. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of urogenital tract disorders, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with urogenital tract disorders and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of immunomodulatory disorders, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with immunomodulatory disorders and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of inflammatory disorders, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with inflammatory disorders and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of respiratory function disorders, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with respiratory function disorders and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of anxiety, mood disorders, stress-related disorders, and attention deficit hyperactivity disorder, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with anxiety, mood disorders, stress-related disorders, attention deficit hyperactivity disorder and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of sympathetic nervous system disorders, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with sympathetic nervous system disorders and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of tussis, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with tussis and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of motor disorders, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with motor disorders and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment of traumatic injuries of the central nervous system, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with the central nervous system and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of stroke, cardiac arrhythmia or glaucoma, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with stroke, cardiac arrhythmia, glaucoma and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of sexual dysfunction, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with sexual dysfunction and other related conditions. The term “effective amount,” when used in connection with compounds useful in improving organ and cell survival, refers to refers to the maintenance and/or improvement of a minimally-acceptable level of organ or cell survival, including organ preservation. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of myocardial infarction, refers to the minimum level of compound necessary to provide cardioprotection after myocardial infarction. The term “effective amount,” when used in connection with compounds useful in the treatment and/or prevention of shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, refers to the treatment and/or prevention of symptoms, diseases, disorders, and conditions typically associated with shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts and other related conditions. The term “effective amount,” when used in connection with compounds useful in the treatment of substance addiction, refers to the treatment of symptoms, diseases, disorders, and conditions typically associated with substance addiction and other related conditions. The term “effective amount,” when used in connection with compounds useful in reducing the need for anesthesia or producing and/or maintaining an anesthetic state, refers to the production and/or maintenance of a minimally-acceptable anesthetic state.
  • “Pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio. The term specifically encompasses veterinary uses.
  • “In combination with,” “combination therapy,” and “combination products” refer, in certain embodiments, to the concurrent administration to a patient of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII, and/or XXXIV, and one or more additional agents including, for example, an opioid, an anaesthetic agent (such as for example, an inhaled anesthetic, hypnotic, anxiolytic, neuromuscular blocker and opioid), an anti-Parkinson's agent (for example, in the case of treating or preventing a motor disorder, particularly Parkinson's disease), an antidepressant (for example, in the case of treating or preventing a mood disorder, particularly depression), an agent for the treatment of incontinence (for example, in the case of treating or preventing a urogenital tract disorder), an agent for the treatment of pain, including neuralgias or neuropathic pain, and/or other optional ingredients (including, for example, antibiotics, antivirals, antifungals, anti-inflammatories, anesthetics and mixtures thereof). When administered in combination, each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • “Dosage unit” refers to physically discrete units suited as unitary dosages for the particular individual to be treated. Each unit may contain a predetermined quantity of active compound(s) calculated to produce the desired therapeutic effect(s) in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention may be dictated by (a) the unique characteristics of the active compound(s) and the particular therapeutic effect(s) to be achieved, and (b) the limitations inherent in the art of compounding such active compound(s).
  • “Pain” refers to the perception or condition of unpleasant sensory or emotional experience, associated with actual or potential tissue damage or described in terms of such damage. “Pain” includes, but is not limited to, two broad categories of pain: acute and chronic pain (Buschmann, H.; Christoph, T; Friderichs, E.; Maul, C.; Sundermann, B; eds.; Analgesics, Wiley-VCH, Verlag GMbH & Co. KgaA, Weinheim; 2002; Jain, K. K. “A Guide to Drug Evaluation for Chronic Pain”; Emerging Drugs, 5(2), 241-257 (2000)). Non-limiting examples of pain include, for example, nociceptive pain, inflammatory pain, visceral pain, somatic pain, neuralgias, neuropathic pain, AIDS pain, cancer pain, phantom pain, and psychogenic pain, and pain resulting from hyperalgesia, pain caused by rheumatoid arthritis, migraine, allodynia and the like.
  • “Gastrointestinal dysfunction” refers collectively to maladies of the stomach, small and large intestine. Non-limiting examples of gastrointestinal dysfunction include, for example, diarrhea, nausea, emesis, post-operative emesis, opioid-induced emesis, irritable bowel syndrome, opioid-bowel dysfunction, inflammatory bowel disease, colitis, increased gastric motility, increased gastric emptying, stimulation of small intestinal propulsion, stimulation of large intestinal propulsion, decreased amplitude of non-propulsive segmental contractions, disorders associated with sphincter of Oddi, disorders associated with anal sphincter tone, impaired reflex relaxation with rectal distention, disorders associated with gastric, biliary, pancreatic or intestinal secretions, changes to the absorption of water from bowel contents, gastro-esophageal reflux, gastroparesis, cramping, bloating, distension, abdominal or epigastric pain and discomfort, non-ulcerogenic dyspepsia, gastritis, or changes to the absorption of orally administered medications or nutritive substances.
  • “Urogenital tract disorders” refers collectively to maladies of the urinary and genital apparati. Non-limiting examples of urogenital tract disorders include incontinence (i.e., involuntary loss of urine) such as stress urinary incontinence, urge urinary incontinence and benign prostatic hyperplasia, overactive bladder disorder, urinary retention, renal colic, glomerulonephritis, and interstitial cystitis.
  • “Overactive bladder disorder” refers to a condition with symptoms of urgency with or without incontinence, and is typically associated with increased urinary frequency and nocturia. Overactive bladder disorders are typically associated with urodynamic finding of involuntary bladder contractions, generally referred to as bladder instability.
  • “Immunomodulatory disorders” refers collectively to maladies characterized by a compromised or over-stimulated immune system. Non-limiting examples of immunomodulatory disorders include autoimmune diseases (such as arthritis, autoimmune disorders associated with skin grafts, autoimmune disorders associated with organ transplants, and autoimmune disorders associated with surgery), collagen diseases, allergies, side effects associated with the administration of an anti-tumor agent, side effects associated with the administration of an antiviral agent, multiple sclerosis and Guillain-Bane syndrome.
  • “Inflammatory disorders” refers collectively to maladies characterized by cellular events in injured tissues. Non-limiting examples of inflammatory diseases include arthritis, psoriasis, asthma, and inflammatory bowel disease.
  • “Respiratory function disorders” refers to conditions in which breathing and/or airflow into the lung is compromised. Non-limiting examples of respiratory function disorders include asthma, apnea, tussis, chronic obstruction pulmonary disease, and lung edema.
  • “Lung edema” refers to the presence of abnormally large amounts of fluid in the intercellular tissue spaces of the lungs.
  • “Anxiety” refers to the unpleasant emotional state consisting of psychophysiological responses to anticipation of real, unreal or imagined danger, ostensibly resulting from unrecognized intrapsychic conflict.
  • “Mood disorders” refers to disorders that have a disturbance in mood as their predominant feature, including depression, bipolar manic-depression, borderline personality disorder, and seasonal affective disorder.
  • “Depression” refers to a mental state of depressed mood characterized by feelings of sadness, despair and discouragement, including the blues, dysthymia, and major depression.
  • “Stress-related disorders” refer collectively to maladies characterized by a state of hyper- or hypo-arousal with hyper- and hypo-vigilance. Non-limiting examples of stress-related disorders include post-traumatic stress disorder, panic disorder, generalized anxiety disorder, social phobia, and obsessive-compulsive disorder.
  • “Attention deficit hyperactivity disorder” refers to a condition characterized by an inability to control behavior due to difficulty in processing neural stimuli.
  • “Sympathetic nervous system disorders” refer collectively to maladies characterized by disturbances of the autonomic nervous system. Non-limiting examples of sympathetic nervous system disorders include hypertension, and the like.
  • “Tussis” refers to a coughing condition, and “antitussive” agents refer to those materials that modulate the coughing response.
  • “Motor disorders” refers to involuntary manifestations of hyper or hypo muscle activity and coordination Non-limiting examples of motor disorders include tremors, Parkinson's disease, tourette syndrome, parasomnias (sleep disorders) including restless leg syndrome, postoperative shivering and dyskinesia.
  • “Traumatic injury of the central nervous system” refers to a physical wound or injury to the spinal cord or brain.
  • “Stroke” refers to a condition due to the lack of oxygen to the brain.
  • “Cardiac arrhythmia” refers to a condition characterized by a disturbance in the electrical activity of the heart that manifests as an abnormality in heart rate or heart rhythm. Patients with a cardiac arrhythmia may experience a wide variety of symptoms ranging from palpitations to fainting.
  • “Glaucoma” refers collectively to eye diseases characterized by an increase in intraocular pressure that causes pathological changes in the optic disk and typical defects in the field of vision.
  • “Sexual dysfunction” refers collectively to disturbances, impairments or abnormalities of the functioning of the male or female sexual organs, including, but not limited to premature ejaculation and erectile dysfunction.
  • “Cardioprotection” refers to conditions or agents that protect or restore the heart from dysfunction, heart failure and reperfusion injury.
  • “Myocardial infarction” refers to irreversible injury to heart muscle caused by a local lack of oxygen.
  • “Addiction” refers to a pattern of compulsive substance abuse (alcohol, nicotine, or drug) characterized by a continued craving for the substance and, in some cases, the need to use the substance for effects other than its prescribed or legal use.
  • “Anaesthetic state” refers to the state of the loss of feeling or sensation, including not only the loss of tactile sensibility or of any of the other senses, but also to the loss of sensation of pain, as it is induced to permit performance of surgery or other painful procedures, and specifically including amnesia, analgesia, muscle relaxation and sedation.
  • “Improving organ and cell survival” refers to the maintenance and/or improvement of a minimally-acceptable level of organ or cell survival.
  • “Patient” refers to animals, including mammals, preferably humans.
  • “Side effect” refers to a consequence other than the one(s) for which an agent or measure is used, as the adverse effects produced by a drug, especially on a tissue or organ system other then the one sought to be benefited by its administration. In the case, for example, of opioids, the term “side effect” may refer to such conditions as, for example, constipation, nausea, vomiting, dyspnea and pruritus.
  • When any variable occurs more than one time in any constituent or in any formula, its definition in each occurrence is independent of its definition at every other occurrence. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • It is believed the chemical formulas and names used herein correctly and accurately reflect the underlying chemical compounds. However, the nature and value of the present invention does not depend upon the theoretical correctness of these formulae, in whole or in part. Thus it is understood that the formulas used herein, as well as the chemical names attributed to the correspondingly indicated compounds, are not intended to limit the invention in any way, including restricting it to any specific tautomeric form or to any specific optical or geometric isomer, except where such stereochemistry is clearly defined.
  • In certain preferred embodiments, the compounds, pharmaceutical compositions and methods of the present invention may involve a peripheral δ opioid modulator compound. The term “peripheral” designates that the compound acts primarily on physiological systems and components external to the central nervous system. In preferred form, the peripheral δ opioid modulator compounds employed in the methods of the present invention exhibit high levels of activity with respect to peripheral tissue, such as, gastrointestinal tissue, while exhibiting reduced, and preferably substantially no, CNS activity. The phrase “substantially no CNS activity,” as used herein, means that less than about 50% of the pharmacological activity of the compounds employed in the present methods is exhibited in the CNS, preferably less than about 25%, more preferably less than about 10%, even more preferably less than about 5% and most preferably 0% of the pharmacological activity of the compounds employed in the present methods is exhibited in the CNS.
  • Furthermore, it is preferred in certain embodiments of the invention that the δ opioid modulator compound does not substantially cross the blood-brain barrier. The phrase “does not substantially cross,” as used herein, means that less than about 20% by weight of the compound employed in the present methods crosses the blood-brain barrier, preferably less than about 15% by weight, more preferably less than about 10% by weight, even more preferably less than about 5% by weight and most preferably 0% by weight of the compound crosses the blood-brain barrier. Selected compounds can be evaluated for CNS penetration, for example, by determining plasma and brain levels following i.v. administration.
  • Accordingly, in one embodiment, the invention is directed to compounds of formula XIV:
  • Figure US20110218186A1-20110908-C00021
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl, provided that at least one of R23 and R24 is alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In preferred embodiments of formula XIV compounds, W2 is aryl or heteroaryl. When W2 is aryl, the aryl ring is preferably phenyl. When W2 is heteroaryl, the heteroaryl ring is preferably pyridyl.
  • As set forth above, W2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In preferred embodiments, W2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). More preferably, W2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • In preferred embodiments of formula XIV compounds, W2 is:
  • Figure US20110218186A1-20110908-C00022
  • more preferably:
  • Figure US20110218186A1-20110908-C00023
  • In embodiments in which W2 is substituted with N-alkylaminocarbonyl (—C(═O)—NH(alkyl)) or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XIV compounds, p is 1.
  • In preferred embodiments of formula XIV compounds, A2 and B2 together form a double bond.
  • In preferred embodiments of formula XIV compounds, X2 is —O—.
  • In preferred embodiments of formula XIV compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl. In certain preferred embodiments, one of R23 and R24 is H and the other is alkyl.
  • In preferred embodiments, the compounds of formula XIV have the following formula XV:
  • Figure US20110218186A1-20110908-C00024
  • with compounds of formula XVI being even more preferred:
  • Figure US20110218186A1-20110908-C00025
  • In preferred embodiments, the compound of formula XIV is:
  • Figure US20110218186A1-20110908-C00026
  • more preferably:
  • Figure US20110218186A1-20110908-C00027
  • In an alternative embodiment, the invention is directed to compounds of formula XVII:
  • Figure US20110218186A1-20110908-C00028
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 0-3 groups selected independently from halo, hydroxy, and —S(═O)2-alkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when W2 is para-diethylaminocarbonylphenyl, X2 is O, and A2 and B2 together form a double bond, then the aryl ring of J2 is substituted with at least one group selected independently from halo, and —S(═O)2-alkyl in which the alkyl group is C2-C6 alkyl;
      • when W2 is para-diethylaminocarbonylphenyl, X2 is O, and A2 and B2 are each H, then the aryl ring of J2 is substituted with 1-3 groups selected independently from halo, hydroxy, and —S(═O)2-alkyl; and
      • the compound of formula XVII is other than:
  • Figure US20110218186A1-20110908-C00029
  • In preferred embodiments of formula XVII compounds, W2 is aryl or heteroaryl. When W2 is aryl, the aryl ring is preferably phenyl. When W2 is heteroaryl, the heteroaryl ring is preferably pyridyl.
  • As set forth above, W2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In preferred embodiments, W2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). More preferably, W2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • In preferred embodiments of formula XVII compounds, W2 is:
  • Figure US20110218186A1-20110908-C00030
  • In embodiments in which W2 is substituted with N-alkylaminocarbonyl (—C(═O)—NH(alkyl)) or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XVII compounds, X2 is —O—.
  • In preferred embodiments of formula XVII compounds, A2 and B2 together form a double bond.
  • In preferred embodiments of formula XVII compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H.
  • In preferred embodiments of formula XVII compounds, J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • As set forth above, J2 is substituted with 0-3 groups, preferably 0-2, more preferably 0-1 groups, selected independently from halo, hydroxy, and —S(═O)2-alkyl. In embodiments wherein J2 is substituted with halo, the halo group is preferably fluoro. In embodiments wherein J2 is substituted with —S(═O)2-alkyl, the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, still more preferably alkyl groups of 1 to 2 carbons, yet more preferably methyl or ethyl.
  • In preferred embodiments, the compounds of formula XVII have the following formula XVIII:
  • Figure US20110218186A1-20110908-C00031
  • wherein:
      • Q1 and Q2 are each independently H, halo, hydroxy, or —S(═O)2-alkyl. In embodiments wherein Q1 or Q2 is halo, the halo group is preferably fluoro. In embodiments wherein Q1 or Q2 is —S(═O)2-alkyl, the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, still more preferably alkyl groups of 1 to 2 carbons, yet more preferably methyl or ethyl. Still more preferably, the compounds of formula XVIII have the following formula XIX:
  • Figure US20110218186A1-20110908-C00032
  • In preferred embodiments, the compounds of formula XVII have the following structures:
  • Figure US20110218186A1-20110908-C00033
    Figure US20110218186A1-20110908-C00034
    Figure US20110218186A1-20110908-C00035
    Figure US20110218186A1-20110908-C00036
  • In certain embodiments, the above compounds may be resolved into any of their R and S, or (R,R), (S,S), (R,S), and (S,R) enantiomers, or partially resolved into any of their non-racemic mixtures.
  • More preferably, the compounds of formula XVII have the following structures:
  • Figure US20110218186A1-20110908-C00037
    Figure US20110218186A1-20110908-C00038
    Figure US20110218186A1-20110908-C00039
  • yet more preferably:
  • Figure US20110218186A1-20110908-C00040
    Figure US20110218186A1-20110908-C00041
  • still more preferably:
  • Figure US20110218186A1-20110908-C00042
  • even more preferably:
  • Figure US20110218186A1-20110908-C00043
  • In certain embodiments, the invention is directed to compounds of formula XX:
  • Figure US20110218186A1-20110908-C00044
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted independently with 0-3 hydroxy or halo groups;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that the compound of formula XX is other than 4-[(4-N,N-diethylaminocarbonyl)phenyl]-spiro[2H,1-benzopyran-2,3′-pyrrolidine].
  • In preferred embodiments of formula XX compounds, W2 is aryl or heteroaryl. When W2 is aryl, the aryl ring is preferably phenyl. When W2 is heteroaryl, the heteroaryl ring is preferably pyridyl.
  • As set forth above, W2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In preferred embodiments, W2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). More preferably, W2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • In preferred embodiments of formula XX compounds, W2 is:
  • Figure US20110218186A1-20110908-C00045
  • In embodiments in which W2 is substituted with N-alkylaminocarbonyl (—C(═O)—NH(alkyl)) or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XX compounds, A2 and B2 together form a double bond.
  • In preferred embodiments of formula XX compounds, X2 is —O—.
  • In preferred embodiments of formula XX compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H.
  • In preferred embodiments of formula XX compounds, J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • As set forth above, J2 is substituted independently with 0-3, preferably 0-2, more preferably 0-1, hydroxy or halo groups, more preferably still, 0-1 hydroxy groups.
  • In preferred embodiments, the compounds of formula XX have the following formula XXI:
  • Figure US20110218186A1-20110908-C00046
  • wherein:
      • Q1 and Q2 are each independently H, hydroxy, or halo. In embodiments wherein at least one of Q1 and Q2 is halo, the halo is preferably fluoro.
  • In preferred embodiments, the compounds of formula XX have the following structures:
  • Figure US20110218186A1-20110908-C00047
  • In an alternative embodiment, the invention is directed to compounds of formula XXII:
  • Figure US20110218186A1-20110908-C00048
  • wherein:
      • W2 is aryl or heteroaryl, wherein the aryl or heteroaryl is substituted with 0-3 groups selected independently from heteroaryl, hydroxy, carboxy (—COOH), —C(═O)-alkyl, —C(═O)-aryl, —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl); aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 0-3 groups selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), carboxy (—COOH), —C(═O)—O-alkyl, and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl));
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when W2 is para-diethylaminocarbonylphenyl, para-prop-2-ylaminocarbonylphenyl, or para-pent-3-ylaminocarbonylphenyl, R23 and R24 are each H, and A and B are each H or together form a double bond, then J2 is other than unsubstituted phenyl or anisyl; and
      • when W2 is:
  • Figure US20110218186A1-20110908-C00049
      • R23 and R24 are each H, and A and B together form a double bond; then J2 is other than unsubstituted phenyl.
  • In preferred embodiments of formula XXII compounds, W2 is aryl or heteroaryl. When W2 is aryl, the aryl ring is preferably phenyl. When W2 is heteroaryl, the heteroaryl ring is preferably pyridyl.
  • As set forth above, W2 is substituted with 0-3 groups selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In preferred embodiments, W2 is substituted with 1-2 groups, selected independently from hydroxy, aminocarbonyl (—C(═O)—NH2), N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). More preferably, W2 is substituted with N,N-dialkylaminocarbonyl and/or hydroxyl.
  • In preferred embodiments of formula XXII compounds, W2 is:
  • Figure US20110218186A1-20110908-C00050
  • In embodiments in which W2 is substituted with N-alkylaminocarbonyl (—C(═O)—NH(alkyl)) or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XXII compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H.
  • In preferred embodiments of formula XXII compounds, J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • As set forth above, J2 is substituted with 0-3 groups, preferably 0-2 groups, selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), carboxy (—COOH), —C(═O)—O-alkyl, and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In embodiments wherein J2 is substituted with alkoxy, —S(═O)2-alkyl, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—O-alkyl, or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 to 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is methyl or ethyl. In embodiments wherein J2 is substituted with halo, the halo group is preferably fluoro.
  • In preferred embodiments of formula XXII compounds, the compounds have the following formula XXIII:
  • Figure US20110218186A1-20110908-C00051
  • wherein:
      • Q1 and Q2 are each independently H, halo, heterocycloalkyl, hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), carboxy (—COOH), —C(═O)—O-alkyl, or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In certain preferred embodiments, at least one of Q1 or Q2 is H, more preferably, one of Q1 or Q2 is H. In embodiments wherein Q1 or Q2 is alkoxy, —S(═O)2-alkyl, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—O-alkyl, or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 to 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is methyl or ethyl. In embodiments wherein Q1 or Q2 is halo, the halo group is preferably fluoro. In certain preferred embodiments, one of Q1 or Q2 is H.
  • In preferred embodiments, the compounds of formula XXII have the following formula XXIV:
  • Figure US20110218186A1-20110908-C00052
  • In preferred embodiments, the compounds of formula XXII have the following structure:
  • Figure US20110218186A1-20110908-C00053
    Figure US20110218186A1-20110908-C00054
    Figure US20110218186A1-20110908-C00055
    Figure US20110218186A1-20110908-C00056
    Figure US20110218186A1-20110908-C00057
  • In preferred embodiments, the compounds of formula XXII have the following structure:
  • Figure US20110218186A1-20110908-C00058
  • In certain embodiments, the above compounds may be resolved into any of their R and S, or (R,R), (S,S), (R,S), and (S,R) enantiomers, or partially resolved into any of their non-racemic mixtures.
  • More preferably, compounds of formula XXII have the following structures:
  • Figure US20110218186A1-20110908-C00059
  • still more preferably
  • Figure US20110218186A1-20110908-C00060
  • In an alternative embodiment, the invention is directed to compounds of formula
  • Figure US20110218186A1-20110908-C00061
  • wherein:
      • W2 is aryl optionally substituted with —C(═O)-alkyl or —C(═O)-aryl;
      • R23 and R24 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 0-3 groups selected independently from hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—N(alkyl)(alkyl), carboxy (—COOH), and —C(═O)—O-alkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that the compound of formula XXV is other than 4-phenyl-spiro[2H,1-benzopyran-2,4′-piperidine].
  • In preferred embodiments of compounds of formula XXV, W2 is aryl more preferably phenyl.
  • In embodiments wherein W2 is substituted with —C(═O)-alkyl, the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 carbon being still more preferred. In particularly preferred embodiments, the alkyl group is methyl.
  • In embodiments wherein W2 is substituted with or —C(═O)-aryl, preferably the aryl group is a 6-membered aryl ring, more preferably phenyl.
  • In preferred embodiments of formula XXV compounds, A2 and B2 together form a double bond.
  • In preferred embodiments of formula XXV compounds, X2 is —O—.
  • In preferred embodiments of formula XXV compounds, p is 1.
  • In preferred embodiments of formula XXV compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H.
  • In preferred embodiments of formula XXV compounds, J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably phenyl.
  • As set forth above, J2 is substituted with 0-3 groups, preferably 0-2, yet more preferably 0-1 groups, selected independently from halo, heterocycloalkyl, hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), carboxy (—COOH), —C(═O)—O-alkyl, and N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)). In embodiments wherein J2 is substituted with alkoxy, —S(═O)2-alkyl, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—O-alkyl, or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 to 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is methyl or ethyl. In embodiments wherein J2 is substituted with halo, the halo group is preferably fluoro.
  • In preferred embodiments, the compounds of formula XXV have the following formula XXVI:
  • Figure US20110218186A1-20110908-C00062
  • wherein:
      • Q1 and Q2 are each independently H, hydroxy, alkoxy, —S(═O)2-alkyl, —S(═O)2—NH2, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—N(alkyl)(alkyl), carboxy (—COOH), or —C(═O)—O-alkyl. In embodiments wherein Q1 or Q2 is alkoxy, —S(═O)2-alkyl, —S(═O)2—NH(alkyl), —S(═O)2—N(alkyl)(alkyl), —C(═O)—O-alkyl, or N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 to 2 carbons being still more preferred.
  • In particularly preferred embodiments, the alkyl group is methyl or ethyl. In embodiments wherein Q1 or Q2 is halo, the halo group is preferably fluoro.
  • In preferred embodiments, the compounds of formula XXV have the following structure:
  • Figure US20110218186A1-20110908-C00063
    Figure US20110218186A1-20110908-C00064
    Figure US20110218186A1-20110908-C00065
  • In an alternative embodiment, the invention is directed to compounds of formula XXVII:
  • Figure US20110218186A1-20110908-C00066
  • wherein:
      • W2 is para-dialkylaminocarbonylphenyl, the phenyl group of which is further optionally substituted with 1-2 groups independently selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2);
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • Q1 and Q2 are each independently H, hydroxy, alkoxy, haloalkoxy, halo, or heterocycloalkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when one of Q1 and Q2 is hydroxy and the other is H, or both Q1 and Q2 are hydroxy, then the phenyl group of W2 is further substituted with 1-2 groups selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2);
      • when Q1, Q2, R23, and R24 are each H and the phenyl group of W2 is further substituted with one hydroxy, then A2 and B2 are each H;
      • when W2 is para-dialkylaminocarbonylphenyl, then at least one of Q1, Q2, R23, and R24 is other than H;
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, and Q2 is halo, then Q1 is other than H or hydroxy;
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, Q1 is methoxy or cyclopropylmethoxy and Q2 is H, then A2 and B2 are each H; and
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, and Q1 is H or OH, then Q2 is other than methoxy or cyclopropylmethoxy
  • In preferred embodiments of formula XXVII compounds, W2 is para-dialkylaminocarbonylphenyl. As set forth above, W2 is further optionally substituted with 1-2 groups independently selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2). In preferred embodiments wherein W2 is substituted with 1-2 groups, W2 is preferably substituted with hydroxy.
  • In preferred embodiments of formula XXVII compounds, W2 is:
  • Figure US20110218186A1-20110908-C00067
  • In embodiments in which W2 is substituted with N,N-dialkylaminocarbonyl (—C(═O)—N(alkyl)(alkyl)), the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XXVII compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H.
  • In preferred embodiments of formula XXVII compounds, Q1 and Q2 are each independently H, hydroxy, alkoxy, haloalkoxy, halo, or heterocycloalkyl. In embodiments where Q1 or Q2 is alkoxy, the alkoxy is preferably C1-C3 alkoxy, more preferably C1 alkoxy, yet more preferably, methoxy. In embodiments where Q1 or Q2 is halo, the halo is preferably fluoro. In embodiments where Q1 or Q2 is heterocycloalkyl, the heterocycloalkyl is preferably a 5- or 6-membered ring heterocycloalkyl, more preferably pyrrolidinyl or morpholinyl. In embodiments where Q1 or Q2 is haloalkoxy, the alkoxy is substituted with one or more, preferably two or more, fluoro atoms.
  • In preferred embodiments, the compounds of formula XXVII, the compounds have the structures:
  • Figure US20110218186A1-20110908-C00068
    Figure US20110218186A1-20110908-C00069
  • more preferably:
  • Figure US20110218186A1-20110908-C00070
  • In some preferred embodiments, the compound of formula XXVII has the structure:
  • Figure US20110218186A1-20110908-C00071
  • wherein denotes “*” a chiral center, as described herein above. In certain preferred embodiments, the compound is substantially enantiomerically pure.
  • In an alternative embodiment, the invention is directed to compounds of formula XXVIII:
  • Figure US20110218186A1-20110908-C00072
  • wherein:
      • D is:
  • Figure US20110218186A1-20110908-C00073
      • K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
      • R23, R24, and R26 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
  • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In preferred embodiments of formula XXVIII compounds, D is:
  • Figure US20110218186A1-20110908-C00074
  • more preferably:
  • Figure US20110218186A1-20110908-C00075
  • In preferred embodiments of formula XXVIII compounds wherein K is —S(═O)2—N(alkyl)(alkyl), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl)), the alkyl group independently is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XXVIII compounds wherein K is —C(═O)—O-alkyl or alkyltetrazolyl, the alkyl group independently is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1-2 carbon being still more preferred. In particularly preferred embodiments, the alkyl group is methyl or ethyl.
  • In preferred embodiments of formula XXVIII compounds wherein K is heteroaryl or alkylheteroaryl, the heteroaryl group is preferably a 5-membered ring heteroaryl, more preferably a tetrazolyl ring.
  • In preferred embodiments of formula XXVIII compounds, A2 and B2 together form a double bond.
  • In preferred embodiments of formula XXVIII compounds, p is 1.
  • In preferred embodiments of formula XXVIII compounds, X2 is —O—.
  • In preferred embodiments of formula XXVIII compounds, R23, R24, and R26 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H. In certain preferred embodiments, one of R23 and R24 is H and the other is alkyl.
  • In preferred embodiments, the compounds of formula XXVIII, have the structures:
  • Figure US20110218186A1-20110908-C00076
    Figure US20110218186A1-20110908-C00077
  • more preferably:
  • Figure US20110218186A1-20110908-C00078
  • In an alternative embodiment, the invention is directed to compounds of formula XXIX:
  • Figure US20110218186A1-20110908-C00079
  • wherein:
      • W2 is para-N(alkyl),N(alkyl-Z)aminocarbonylaryl or para-N(alkyl),N(alkyl-Z)aminocarbonylheteroaryl, wherein the aryl or heteroaryl ring of W2 is substituted with 0-2 groups selected independently from hydroxy and alkoxy;
      • Z is alkoxy, alkylamino, or dialkylamino;
      • R23 and R24 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
  • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In preferred embodiments of formula XXIX compounds, W2 is para-N(alkyl),N(alkyl-Z)-aminocarbonylaryl or para-N(alkyl),N(alkyl-Z)-aminocarbonylheteroaryl. When W2 is para-N(alkyl),N(alkyl-Z)aminocarbonylaryl, the aryl ring is preferably phenyl. When para-N(alkyl),N(alkyl-Z)aminocarbonylheteroaryl, the heteroaryl ring is preferably pyridyl.
  • As set forth above, W2 is substituted with 0-2 groups selected independently from selected independently from hydroxy and alkoxy. In preferred embodiments, W2 is substituted with 0-1 groups, selected independently from hydroxy and alkoxy, more preferably hydroxy.
  • In preferred embodiments of formula XXIX compounds, W2 is:
  • Figure US20110218186A1-20110908-C00080
  • more preferably:
  • Figure US20110218186A1-20110908-C00081
  • more preferably still:
  • Figure US20110218186A1-20110908-C00082
  • In embodiments in which W2 is para-N(alkyl),N(alkyl-Z)aminocarbonylaryl or para-N(alkyl),N(alkyl-Z)aminocarbonylheteroaryl, the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl.
  • In preferred embodiments of formula XXIX compounds, p is 1.
  • In preferred embodiments of formula XXIX compounds, A2 and B2 together form a double bond.
  • In preferred embodiments of formula XXIX compounds, X2 is —O—.
  • In preferred embodiments of formula XXIX compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl. In certain preferred embodiments, one of R23 and R24 is H and the other is alkyl.
  • In preferred embodiments of formula XXIX compounds, Z is alkoxy, alkylamino, or dialkylamino, preferably alkoxy. In embodiments wherein Z is alkoxy, alkylamino, or dialkylamino the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 1 carbon being still more preferred. In particularly preferred embodiments, the alkyl group is methyl.
  • In certain embodiments, the compounds of formula XXIX have the structures:
  • Figure US20110218186A1-20110908-C00083
  • more preferably:
  • Figure US20110218186A1-20110908-C00084
  • In an alternative embodiment, the invention is directed to compounds of formula XXX:
  • Figure US20110218186A1-20110908-C00085
  • wherein:
      • W2 is:
  • Figure US20110218186A1-20110908-C00086
      • R23 and R24 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond;
      • X2 is —CH2— or —O—; and
      • J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring substituted with 1-3 groups selected independently from halo and haloalkoxy;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
      • provided that when W2 is:
  • Figure US20110218186A1-20110908-C00087
  • then the aryl ring of J2 is substituted with at least one haloalkoxy.
  • In preferred embodiments of compounds of formula XXX, W2 is:
  • Figure US20110218186A1-20110908-C00088
  • In preferred embodiments of formula XXX compounds, p is 1.
  • In preferred embodiments of formula XXX compounds, R23 and R24 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H. In certain preferred embodiments, one of R23 and R24 is H and the other is alkyl.
  • In preferred embodiments of formula XXX compounds, J2 when taken together with the carbon atoms to which it is attached forms a 6-membered aryl ring, preferably a phenyl ring. In embodiments wherein J2 is substituted with 1-3 groups selected independently from halo and haloalkoxy, the halo group of halo or haloalkoxy is preferably fluoro.
  • In preferred embodiments, the compounds of formula XXX have the following formula XXXI:
  • Figure US20110218186A1-20110908-C00089
  • wherein:
      • Q1 and Q2 are each independently H, halo, or haloalkoxy, provided that at least one of Q1 and Q2 is other than H. In embodiments wherein Q1 or Q2 is halo or haloalkoxy, the halo group of halo or haloalkoxy is preferably fluoro.
  • In preferred embodiments, the compounds of formula XXX have the structures:
  • Figure US20110218186A1-20110908-C00090
  • more preferably:
  • Figure US20110218186A1-20110908-C00091
  • In an alternative embodiment, the invention is directed to compounds of formula XXXII:
  • Figure US20110218186A1-20110908-C00092
  • wherein:
      • D is N(alkyl),N(alkyl)aminocarbonylheteroaryl;
      • R23, R24, and R26 are each independently H or alkyl;
      • p is 1 or 2;
      • A2 and B2 are each H, or together form a double bond; and
      • X2 is —CH2— or —O—;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that when D is:
  • Figure US20110218186A1-20110908-C00093
  • and X2 is —O—, then A2 and B2 are each H.
  • In embodiments of formula XXXII compounds wherein D is N(alkyl),N(alkyl)aminocarbonylheteroaryl, the heteroaryl group is preferably pyridyl or thienyl.
  • In embodiments of formula XXXII compounds wherein D is N(alkyl),N(alkyl)aminocarbonylheteroaryl, the alkyl group is preferably lower alkyl, with alkyl groups of 1 to 3 carbons being more preferred, and with alkyl groups of 2-3 carbons being still more preferred. In particularly preferred embodiments, the alkyl group is ethyl or isopropyl.
  • In preferred embodiments of formula XXXII compounds, A2 and B2 are each H.
  • In preferred embodiments of formula XXXII compounds, X2 is —O—.
  • In preferred embodiments of formula XXXII compounds, R23, R24, and R26 are each independently H or alkyl, preferably H or C1-C3 alkyl, more preferably H or methyl, yet more preferably H. In certain preferred embodiments, one of R23 and R24 is H and the other is alkyl.
  • In certain embodiments, the compounds of formula XXXII, have the structures:
  • Figure US20110218186A1-20110908-C00094
  • preferably:
  • Figure US20110218186A1-20110908-C00095
  • In another embodiment, the present invention is directed, in part, to compounds of formula XXXIII:
  • Figure US20110218186A1-20110908-C00096
  • wherein:
      • F1 is heteroaryl; and
      • G is C1-6alkylene substituted with NH2, NHC(═O)alkyl, NH(C(O)N(H)alkyl, or NHS(═O)2alkyl;
        or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In formula XXXIII above, F1 is heteroaryl, preferably a 5- or 6-membered heteroaryl having 1 to 4 heteroatoms, with 2 to 4 heteroatoms being more preferred. In certain more preferred embodiments, F1 is a 5-membered heteroaryl, still more preferably a tetrazole.
  • Also in formula XXXIII above, G is C1-6alkylene substituted with NH2, NHC(═O)alkyl, NH(C(O)N(H)alkyl, or NHS(═O)2alkyl. In preferred embodiments, G is C1-6alkylene substituted with NH2. In other preferred embodiments, G is C1-6alkylene substituted with NHC(═O)alkyl. In still other preferred embodiments, G is C1-6alkylene substituted with NHS(═O)2alkyl. More preferably, G is C1-6alkylene substituted with NH2.
  • In some preferred embodiments of compounds of formula XXXIII, F1-G is:
  • Figure US20110218186A1-20110908-C00097
  • In certain preferred embodiments, the compounds of formula XXXIII are selected from the group consisting of:
  • Figure US20110218186A1-20110908-C00098
    Figure US20110218186A1-20110908-C00099
    Figure US20110218186A1-20110908-C00100
    Figure US20110218186A1-20110908-C00101
    Figure US20110218186A1-20110908-C00102
  • In one embodiment, the present invention is directed, in part, to compounds of formula XXXIV:
  • Figure US20110218186A1-20110908-C00103
  • wherein:
      • F2 is aryl or heteroaryl; and
      • Q3 is hydroxy or alkoxy;
        or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • In formula XXXIV above, F2 is aryl or heteroaryl. When F2 is aryl, preferably it is C6-10aryl, more preferably C6aryl, with phenyl being even more preferred. When F2 is heteroaryl, preferably it is C6-10heteroaryl, with pyridyl or benzothiophenyl being more preferred.
  • Also in formula XXXIV above, Q3 is hydroxy or alkoxy, preferably hydroxy.
  • In another embodiment, the compound of the invention is selected from the group consisting of:
  • Figure US20110218186A1-20110908-C00104
    Figure US20110218186A1-20110908-C00105
    Figure US20110218186A1-20110908-C00106
  • more preferably from the group consisting of:
  • Figure US20110218186A1-20110908-C00107
  • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
  • The present invention is further directed to pharmaceutical compositions, comprising:
      • a pharmaceutically acceptable carrier; and an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV. In certain embodiments, the pharmaceutical composition further comprises an effective amount of at least one opioid.
  • In a preferred embodiment, the invention is directed to pharmaceutical compositions, comprising:
      • a pharmaceutically acceptable carrier; and an effective amount of a compound of formula XXVIIA:
  • Figure US20110218186A1-20110908-C00108
  • wherein:
      • W2 is para-dialkylaminocarbonylphenyl, the phenyl group of which is further optionally substituted with 1-2 groups independently selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2);
      • R23 and R24 are each independently H or alkyl;
      • A2 and B2 are each H, or together form a double bond;
      • Q1 and Q2 are each independently H, hydroxy, alkoxy, haloalkoxy, halo, or heterocycloalkyl;
      • or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
  • provided that:
      • when one of Q1 and Q2 is hydroxy and the other is H, then the phenyl group of W2 is further substituted with 1-2 groups selected from tetrazolyl, N-alkyltetrazolyl, hydroxy, carboxy (—COOH), and aminocarbonyl (—C(═O)—NH2);
      • when Q1, Q2, R23, and R24 are each H and the phenyl group of W2 is further substituted with one hydroxy, then A2 and B2 are each H;
      • when W2 is para-dialkylaminocarbonylphenyl, then at least one of Q1, Q2, R23, and R24 is other than H;
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, and Q2 is halo, then Q1 is other than H;
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, Q1 is methoxy or cyclopropylmethoxy, and Q2 is H, then A2 and B2 are each H; and
      • when W2 is para-dialkylaminocarbonylphenyl, R23 and R24 are each H, and Q1 is H, then Q2 is other than methoxy or cyclopropylmethoxy.
  • Compounds as described herein may be useful as analgesic agents for use during general anesthesia and monitored anesthesia care. Combinations of agents with different properties are often used to achieve a balance of effects needed to maintain the anaesthetic state (e.g., amnesia, analgesia, muscle relaxation and sedation). Included in this combination are inhaled anesthetics, hypnotics, anxiolytics, neuromuscular blockers and opioids.
  • In any of the above teachings, a compound as described herein may be either a compound of one of the formulae herein described, or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, N-oxide or isomorphic crystalline form thereof.
  • The compounds employed in the methods and compositions of the present invention may exist in prodrug form. As used herein, “prodrug” is intended to include any covalently bonded carriers which release the active parent drug, for example, as according to formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV, or other formulas or compounds as described herein, such as for example, compounds of formula XXVIIA, in vivo when such prodrug is administered to a mammalian subject. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.), the compounds described herein may, if desired, be delivered in prodrug form. Thus, the present invention contemplates compositions and methods involving prodrugs. Prodrugs of the compounds employed in the present invention, for example formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV, may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • Accordingly, prodrugs include, for example, compounds described herein in which a hydroxy, amino, or carboxy group is bonded to any group that, when the prodrug is administered to a mammalian subject, cleaves to form a free hydroxyl, free amino, or carboxylic acid, respectively. Examples include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups; and alkyl, carbocyclic, aryl, and alkylaryl esters such as methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, phenyl, benzyl, and phenethyl esters, and the like.
  • Compounds described herein may contain one or more asymmetrically substituted carbon atoms, and may be isolated in optically active or racemic forms. Thus, all chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. It is well known in the art how to prepare and isolate such optically active forms. For example, mixtures of stereoisomers may be separated by standard techniques including, but not limited to, resolution of racemic forms, normal, reverse-phase, and chiral chromatography, preferential salt formation, recrystallization, and the like, or by chiral synthesis either from chiral starting materials or by deliberate synthesis of target chiral centers.
  • The compounds as herein described may be prepared in a number of ways well known to those skilled in the art. The compounds can be synthesized, for example, by the methods described below, or variations thereon as appreciated by the skilled artisan. All processes disclosed in association with the present invention are contemplated to be practiced on any scale, including milligram, gram, multigram, kilogram, multikilogram or commercial industrial scale.
  • As will be readily understood, functional groups present may contain protecting groups during the course of synthesis. Protecting groups are known per se as chemical functional groups that can be selectively appended to and removed from functionalities, such as hydroxyl groups and carboxyl groups. These groups are present in a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Any of a variety of protecting groups may be employed with the present invention. Preferred protecting groups include the benzyloxycarbonyl group and the tert-butyloxycarbonyl group. Other preferred protecting groups that may be employed in accordance with the present invention may be described in Greene, T. W. and Wuts, P. G. M., Protective Groups in Organic Synthesis 2d. Ed., Wiley & Sons, 1991, the disclosures of which are hereby incorporated herein by reference, in their entirety.
  • The δ agonist compounds as described herein may be administered by any means that results in the contact of the active agent with the agent's site of action in the body of a patient. The compounds may be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. For example, they may be administered as the sole active agent in a pharmaceutical composition, or they can be used in combination with other therapeutically active ingredients including, for example, opioid analgesic agents. In such combinations, selected compounds as described herein may provide equivalent or even enhanced therapeutic activity such as, for example, pain ameliorization, while providing reduced adverse side effects associated with opioids, such as addiction or pruritus, by lowering the amount of opioid required to achieve a therapeutic effect.
  • The compounds are preferably combined with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice as described, for example, in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa., 1980), the disclosures of which are hereby incorporated herein by reference, in their entirety.
  • In addition to the pharmaceutical carrier, the compounds of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV, may be co-administered with at least one opioid, preferably a μ opioid receptor modulator compound. In certain embodiments, the combination of the compounds of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV, with at least one opioid, preferably a μ opioid receptor modulator compound, provides a synergistic analgesic effect. The utility of the instant combination product may be determined by those skilled in the art using established animal models. Suitable opioids include, without limitation, alfentanil, allylprodine, alphaprodine, anileridine, benzyl-morphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioaphetylbutyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacylmorphan, lofentanil, loperamide, meperidine (pethidine), meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbuphine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, normorphine, norpinanone, opium, oxycodone, oxymorphone, papavereturn, pentazocine, phenadoxone, phenomorphan, phanazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propiram, propoxyphene, sulfentanil, tilidine, tramadol, diastereoisomers thereof, pharmaceutically acceptable salts thereof, complexes thereof; and mixtures thereof.
  • The pain ameliorating and/or opioid combination products of the present compositions may further include one or more other active ingredients that may be conventionally employed in analgesic and/or cough-cold-antitussive combination products. Such conventional ingredients include, for example, aspirin, acetaminophen, phenylpropanolamine, phenylephrine, chlorpheniramine, caffeine, and/or guaifenesin. Typical or conventional ingredients that may be included in the opioid component are described, for example, in the Physicians' Desk Reference, 1999, the disclosure of which is hereby incorporated herein by reference, in its entirety.
  • In addition, the opioid component may further include one or more compounds that may be designed to enhance the analgesic potency of the opioid and/or to reduce analgesic tolerance development. Such compounds include, for example, dextromethorphan or other NMDA antagonists (Mao, M. J. et al., Pain 1996, 67, 361), L-364,718 and other CCK antagonists (Dourish, C. T. et al., Eur J Pharmacol 1988, 147, 469), NOS inhibitors (Bhargava, H. N. et al., Neuropeptides 1996, 30, 219), PKC inhibitors (Bilsky, E. J. et al., J Pharmacol Exp Ther 1996, 277, 484), and dynorphin antagonists or antisera (Nichols, M. L. et al., Pain 1997, 69, 317). The disclosures of each of the foregoing documents are hereby incorporated herein by reference, in their entireties.
  • Other opioids, optional conventional opioid components, and optional compounds for enhancing the analgesic potency of the opioid and/or for reducing analgesic tolerance development, that may be employed in the methods and compositions of the present invention, in addition to those exemplified above, would be readily apparent to one of ordinary skill in the art, once armed with the teachings of the present disclosure.
  • Compounds as described herein can be administered to a mammalian host in a variety of forms adapted to the chosen route of administration, e.g., orally or parenterally. Parenteral administration in this respect includes administration by the following routes: intravenous, intramuscular, subcutaneous, rectal, intraocular, intrasynovial, transepithelial including transdermal, ophthalmic, sublingual and buccal; topically including ophthalmic, dermal, ocular, rectal, and nasal inhalation via insufflation aerosol.
  • The active compound may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compound may be incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should preferably contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be, for example, from about 2 to about 6% of the weight of the unit. The amount of active compound in such therapeutically useful compositions is preferably such that a suitable dosage will be obtained. Preferred compositions or preparations according to the present invention may be prepared so that an oral dosage unit form contains from about 0.1 to about 1000 mg of active compound.
  • The tablets, troches, pills, capsules and the like may also contain one or more of the following: a binder, such as gum tragacanth, acacia, corn starch or gelatin; an excipient, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; a sweetening agent such as sucrose, lactose or saccharin; or a flavoring agent, such as peppermint, oil of wintergreen or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form is preferably pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and formulations.
  • The active compound may also be administered parenterally or intraperitoneally. Solutions of the active compound as a free base or a pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. A dispersion can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical forms suitable for injectable use include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form is preferably sterile and fluid to provide easy syringability. It is preferably stable under the conditions of manufacture and storage and is preferably preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of a dispersion, and by the use of surfactants. The prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions may be achieved by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions may be prepared by incorporating the active compound in the required amount, in the appropriate solvent, with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions may be prepared by incorporating the sterilized active ingredient into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation may include vacuum drying and the freeze-drying technique that yield a powder of the active ingredient, plus any additional desired ingredient from the previously sterile-filtered solution thereof.
  • The therapeutic compounds of this invention may be administered to a patient alone or in combination with a pharmaceutically acceptable carrier. As noted above, the relative proportions of active ingredient and carrier may be determined, for example, by the solubility and chemical nature of the compound, chosen route of administration and standard pharmaceutical practice.
  • The dosage of the compounds as described herein that will be most suitable for prophylaxis or treatment will vary with the form of administration, the particular compound chosen and the physiological characteristics of the particular patient under treatment. Generally, small dosages may be used initially and, if necessary, increased by small increments until the desired effect under the circumstances is reached. The therapeutic human dosage, based on physiological studies using rats, may generally range from about 0.01 mg to about 100 mg/kg of body weight per day, and all combinations and subcombinations of ranges and specific dosages therein. Alternatively, the therapeutic human dosage may be from about 0.4 mg to about 10 g or higher, and may be administered in several different dosage units from once to several times a day. Generally speaking, oral administration may require higher dosages.
  • It will be further appreciated that the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • The dose may also be provided by controlled release of the compound, by techniques well known to those in the art.
  • The compounds as described herein may also be formulated with other optional active ingredients, in addition to or instead of the optional opioids, and in addition to the optional pharmaceutical-acceptable carriers. Other active ingredients include, but are not limited to, antibiotics, antivirals, antifungals, anti-inflammatories, including steroidal and non-steroidal anti-inflammatories, anesthetics and mixtures thereof. Such additional ingredients include any of the following:
  • a. Antibacterial agents
  • Aminoglycosides, such as Amikacin, Apramycin, Arbekacin, Bambermycins, Butirosin, Dibekacin, Dihydrostreptomycin, Fortimicin(s), Fradiomycin, Gentamicin, Ispamicin, Kanamycin, Micronomicin, Neomycin, Neomycin Undecylenate, Netilmicin, Paromomycin, Ribostamycin, Sisomicin, Spectinomycin, Streptomycin, Streptonicozid and Tobramycin;
  • Amphenicols, such as Azidamfenicol, Chloramphenicol, Chloramphenicol Palmirate, Chloramphenicol Pantothenate, Florfenicol, Thiamphenicol;
  • Ansamycins, such as Rifamide, Rifampin, Rifamycin and Rifaximin;
  • β-Lactams;
  • Carbapenems, such as Imipenem;
  • Cephalosporins, such as 1-Carba (dethia) Cephalosporin, Cefactor, Cefadroxil, Cefamandole, Cefatrizine, Cefazedone, Cefazolin, Cefixime, Cefmenoxime, Cefodizime, Cefonicid, Cefoperazone, Ceforanide, Cefotaxime, Cefotiam, Cefpimizole, Cefpirimide, Cefpodoxime Proxetil, Cefroxadine, Cefsulodin, Ceftazidime, Cefteram, Ceftezole, Ceftibuten, Ceftizoxime, Ceftriaxone, Cefuroxime, Cefuzonam, Cephacetrile Sodium, Cephalexin, Cephaloglycin, Cephaloridine, Cephalosporin, Cephalothin, Cephapirin Sodium, Cephradine and Pivcefalexin;
  • Cephamycins such as Cefbuperazone, Cefmetazole, Cefminox, Cefetan and Cefoxitin;
  • Monobactams such as Aztreonam, Carumonam and Tigemonan;
  • Oxacephems such as Flomoxef and Moxolactam;
  • Penicillins such as Amidinocillin, Amdinocillin, Pivoxil, Amoxicillin, Ampicillan, Apalcillin, Aspoxicillin, Azidocillan, Azlocillan, Bacampicillin, Benzylpenicillinic Acid, Benzylpenicillin, Carbenicillin, Carfecillin, Carindacillin, Clometocillin, Cloxacillin, Cyclacillin, Dicloxacillin, Diphenicillin, Epicillin, Fenbenicillin, Floxicillin, Hetacillin, Lenampicillin, Metampicillin, Methicillin, Mezlocillin, Nafcillin, Oxacillin, Penamecillin, Penethamate Hydriodide, Penicillin G Benethamine, Penicillin G Benzathine, Penicillin G Benzhydrylamine, Penicillin G Calcium, Penicillin G Hydragamine, Penicillin G Potassium, Penicillin G Procaine, Penicillin N, Penicillin O, Penicillin V, Penicillin V Benzathine, Penicillin V Hydrabamine, Penimepicycline, Phenethicillin, Piperacillin, Pivapicillin, Propicillin, Quinacillin, Sulbenicillin, Talampicillin, Temocillin and Ticarcillin;
  • Lincosumides such as Clindamycin and Lincomycin;
  • Macrolides such as Azithromycin, Carbomycin, Clarithromycin, Erythromycin(s) and Derivatives, Josamycin, Leucomycins, Midecamycins, Miokamycin, Oleandomycin, Primycin, Rokitamycin, Rosaramicin, Roxithromycin, Spiramycin and Troleandomycin;
  • Polypeptides such as Amphomycin, Bacitracin, Capreomycin, Colistin, Enduracidin, Enviomycin, Fusafungine, Gramicidin(s), Gramicidin S, Mikamycin, Polymyxin, Polymyxin β-Methanesulfonic Acid, Pristinamycin, Ristocetin, Teicoplanin, Thiostrepton, Tuberactinomycin, Tyrocidine, Tyrothricin, Vancomycin, Viomycin(s), Virginiamycin and Zinc Bacitracin;
  • Tetracyclines such as Spicycline, Chlortetracycline, Clomocycline, Demeclocycline, Doxycycline, Guamecycline, Lymecycline, Meclocycline, Methacycline, Minocycline, Oxytetracycline, Penimepicycline, Pipacycline, Rolitetracycline, Sancycline, Senociclin and Tetracycline; and
  • others such as Cycloserine, Mupirocin, Tuberin.
  • b. Synthetic Antibacterials
  • 2,4-Diaminopyrimidines such as Brodimoprim, Tetroxoprim and Trimethoprim;
  • Nitrofurans such as Furaltadone, Furazolium, Nifuradene, Nifuratel, Nifurfoline, Nifurpirinol, Nifurprazine, Nifurtoinol and Nitrofurantoin;
  • Quinolones and analogs thereof, such as Amifloxacin, Cinoxacin, Ciprofloxacin, Difloxacin, Enoxacin, Fleroxacin, Flumequine, Lomefloxacin, Miloxacin, Nalidixic Acid, Norfloxacin, Ofloxacin, Oxolinic Acid, Perfloxacin, Pipemidic Acid, Piromidic Acid, Rosoxacin, Temafloxacin and Tosufloxacin;
  • Sulfonamides such as Acetyl Sulfamethoxypyrazine, Acetyl Sulfisoxazole, Azosulfamide, Benzylsulfamide, Chloramine-β, Chloramine-T, Dichloramine-T, Formosulfathiazole, N.sup.2-Formyl-sulfisomidine, N.sup.4-β-D-Glucosylsulfanilamide, Mafenide, 4′-(Methyl-sulfamoyl)sulfanilanilide, p-Nitrosulfathiazole, Noprylsulfamide, Phthalylsulfacetamide, Phthalylsulfathiazole, Salazosulfadimidine, Succinylsulfathiazole, Sulfabenzamide, Sulfacetamide, Sulfachlorpyridazine, Sulfachrysoidine, Sulfacytine, Sulfadiazine, Sulfadicramide, Sulfadimethoxine, Sulfadoxine, Sulfaethidole, Sulfaguanidine, Sulfaguanol, Sulfalene, Sulfaloxic Acid, Sulfamerazine, Sulfameter, Sulfamethazine, Sulfamethizole, Sulfamethomidine, Sulfamethoxazole, Sulfamethoxypyridazine, Sulfametrole, sulfamidochrysoidine, Sulfamoxole, Sulfanilamide, Sulfanilamidomethanesulfonic Acid Triethanolamine Salt, 4-Sulfanilamidosalicyclic Acid, N4-Sulfanilylsulfanilamide, Sulfanilylurea, N-Sulfanilyl-3,4-xylamide, Sulfanitran, Sulfaperine, Sulfaphenazole, Sulfaproxyline, Sulfapyrazine, Sulfapyridine, Sulfasomizole, Sulfasymazine, Sulfathiazole, Sulfathiourea, Sulfatolamide, Sulfisomidine and Sulfisoxazole;
  • Sulfones, such as Acedapsone, Acediasulfone, Acetosulfone, Dapsone, Diathymosulfone, Glucosulfone, Solasulfone, Succisulfone, Sulfanilic Acid, p-Sulfanilylbenzylamine, p,p′-sulfonyldianiline-N,N′-digalactoside, Sulfoxone and Thiazolsulfone;
  • Others such as Clofoctol, Hexedine, Magainins, Methenamine, Methenamine Anhydromethylene-citrate, Methenamine Hippurate, Methenamine Mandelate, Methenamine Sulfosalicylate, Nitroxoline, Squalamine and Xibomol.
  • c. Antifungal (Antibiotics)
  • Polyenes such as Amphotericin-B, Candicidin, Dermostatin, Filipin, Fungichromin, Hachimycin, Hamycin, Lucensomycin, Mepartricin, Natamycin, Nystatin, Pecilocin, Perimycin; and others, such as Azaserine, Griseofulvin, Oligomycins, PyrroInitrin, Siccanin, Tubercidin and Viridin.
  • d. Antifungal (Synthetic)
  • Allylamines such as Naftifine and terbinafine;
  • Imidazoles such as Bifonazole, Butoconazole, Chlordantoin, Chlormidazole, Cloconazole, Clotrimazole, Econazole, Enilconazole, Finticonazole, Isoconazole, Ketoconazole, Miconazole, Omoconazole, Oxiconazole Nitrate, Sulconazole and Tioconazole;
  • Triazoles such as Fluconazole, Itraconazole, Terconazole;
  • Others such as Acrisorcin, Amorolfine, Biphenamine, Bromosalicylchloranilide, Buclosamide, Chlophenesin, Ciclopirox, Cloxyquin, Coparaffinate, Diamthazole, Dihydrochloride, Exalamide, Flucytosine, Halethazole, Hexetidine, Loflucarban, Nifuratel, Potassium Iodide, Propionic Acid, Pyrithione, Salicylanilide, Sulbentine, Tenonitrozole, Tolciclate, Tolindate, Tolnaftate, Tricetin, Ujothion, and Undecylenic Acid.
  • e. Antiglaucoma Agents
  • Antiglaucoma agents, such as Dapiprazoke, Dichlorphenamide, Dipivefrin and Pilocarpine.
  • f. Anti-Inflammatory Agents
  • Corticosteroids, aminoarylcarboxylic Acid Derivatives such as Etofenamate, Meclofenamic Acid, Mefanamic Acid, Niflumic Acid;
  • Arylacetic Acid Derivatives such as Acemetacin, Amfenac Cinmetacin, Clopirac, Diclofenac, Fenclofenac, Fenclorac, Fenclozic Acid, Fentiazac, Glucametacin, Isozepac, Lonazolac, Metiazinic Acid, Oxametacine, Proglumetacin, Sulindac, Tiaramide and Tolmetin;
  • Arylbutyric Acid Derivatives such as Butibufen and Fenbufen; Arylcarboxylic Acids such as Clidanac, Ketorolac and Tinoridine; Arylpropionic Acid Derivatives such as Bucloxic Acid, Carprofen, Fenoprofen, Flunoxaprofen, Ibuprofen, Ibuproxam, Oxaprozin, Piketoprofen, Pirprofen, Pranoprofen, Protizinic Acid and Tiaprofenic Add;
  • Pyrazoles such as Mepirizole;
  • Pyrazolones such as Clofezone, Feprazone, Mofebutazone, Oxyphenbutazone, Phenylbutazone, Phenyl Pyrazolidininones, Suxibuzone and Thiazolinobutazone;
  • Salicylic Acid Derivatives such as Bromosaligenin, Fendosal, Glycol Salicylate, Mesalamine, 1-Naphthyl Salicylate, Olsalazine and Sulfasalazine;
  • Thiazinecarboxamides such as Droxicam, Isoxicam and Piroxicam;
  • Others such as e-Acetamidocaproic Acid, S-Adenosylmethionine, 3-Amino-4-hydroxybutyric Acid, Amixetrine, Bendazac, Bucolome, Carbazones, Difenpiramide, Ditazol, Guaiazulene, Heterocyclic Aminoalkyl Esters of Mycophenolic Acid and Derivatives, Nabumetone, Nimesulide, Orgotein, Oxaceprol, Oxazole Derivatives, Paranyline, Pifoxime, 2-substituted-4,6-di-tertiary-butyl-s-hydroxy-1,3-pyrimidines, Proquazone and Tenidap.
  • g. Antiseptics
  • Guanidines such as Alexidine, Ambazone, Chlorhexidine and Picloxydine;
  • Halogens/Halogen Compounds such as Bomyl Chloride, Calcium Iodate, Iodine, Iodine Monochloride, Iodine Trichloride, Iodoform, Povidone-Iodine, Sodium Hypochlorite, Sodium Iodate, Symclosene, Thymol Iodide, Triclocarban, Triclosan and Troclosene Potassium;
  • Nitrofurans such as Furazolidone, 2-(Methoxymethyl)-5-Nitrofuran, Nidroxyzone, Nifuroxime, Nifurzide and Nitrofurazone;
  • Phenols such as Acetomeroctol, Chloroxylenol, Hexachlorophene, 1-Naphthyl Salicylate, 2,4,6-Tribromo-m-cresol and 3′,4′,5-Trichlorosalicylanilide;
  • Quinolines such as Aminoquinuride, Chloroxine, Chlorquinaldol, Cloxyquin, Ethylhydrocupreine, Halquinol, Hydrastine, 8-Hydroxyquinoline and Sulfate; and
  • others, such as Boric Acid, Chloroazodin, m-Cresyl Acetate, Cupric sulfate and Ichthammol.
  • h. Antivirals
  • Purines/Pyrimidinones, such as 2-Acetyl-Pyridine 5-((2-pyridylamino)thiocarbonyl) Thiocarbonohydrazone, Acyclovir, Dideoxyadenosine, Dideoxycytidine, Dideoxyinosine, Edoxudine, Floxuridine, Ganciclovir, Idoxuridine, MADU, Pyridinone, Trifluridine, Vidrarbine and Zidovudline;
  • others such as Acetylleucine Monoethanolamine, Acridinamine, Alkylisooxazoles, Amantadine, Amidinomycin, Cuminaldehyde Thiosemicarbzone, Foscamet Sodium, Kethoxal, Lysozyme, Methisazone, Moroxydine, Podophyllotoxin, Ribavirin, Rimantadine, Stallimycin, Statolon, Thymosins, Tromantadine and Xenazoic Acid.
  • i. Agents for Neuralgia/Neuropathic Pain
  • Mild OTC (over the counter) analgesics, such as aspirin, acetaminophen, and ibuprophen.
  • Narcotic analgesics, such as codeine.
  • Anti seizure medications, such as carbamazepine, gabapentin, lamotrigine and phenyloin.
  • Anti-depressants, such as amitryptiline.
  • j. Agents for the Treatment of Depression
  • Selective serotonin re-uptake inhibitors (SSRIs), such as Fluoxetine, Paroxetine, Fluvoxamine, Citaprolam, and Sertraline.
  • Tricyclics, such as Imipramine, Amitriptyline, Desipramine, Nortriptyline Protriptyline, Trimipramine, Doxepin, Amoxapine, and Clomipramine.
  • Monoamine Oxidase Inhibitors (MAOIs), such as Tranylcypromine, Phenelzine, and Isocarboxazid.
  • Heterocyclics, such as Amoxipine, Maprotiline and Trazodone.
  • others such as Venlafaxine, Nefazodone and Mirtazapine.
  • k. Agents for the treatment of Incontinence
  • Anticholinergic agents such as propantheline.
  • Antispasmodic medications such as oxybutynin, tolterodine, and flavoxate.
  • Tricyclic antidepressants such as imipramine, and doxepin.
  • Calcium channel blockers such as tolterodine.
  • Beta agonists such as terbutaline.
  • 1. Anti-Parkinson's Agents
  • Deprenyl, Amantadine, Levodopa, and Carbidopa.
  • In yet another embodiment, the invention is directed to methods of binding opioid receptors, preferably δ opioid receptors, in a patient in need thereof, comprising the step of administering to said patient an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV. The δ opioid receptors may be located in the central nervous system or located peripherally to the central nervous system. In certain preferred embodiments, the binding of the present compounds modulates the activity, preferably as an agonist, of said opioid receptors. In certain preferred embodiments, the compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV does not substantially cross the blood-brain barrier. Preferably, the compounds as described herein are peripherally selective.
  • The spirocyclic heterocyclic derivatives of the present invention and pharmaceutical compositions containing these compounds may be utilized in a number of ways. In certain embodiments, the spirocyclic heterocyclic derivatives are ligands of the δ opioid receptor and are useful, inter alia, in methods for treating and/or preventing pain, gastrointestinal disorders, urogenital tract disorders including incontinence, for example, stress urinary incontinence, urge urinary incontinence and benigh prostatic hyperplasia, and overactive bladder disorder (see, e.g., R. B. Moreland et al., Perspectives in Pharmacology, Vol. 308(3), pp. 797-804 (2004) and M. O. Fraser, Annual Reports in Medicinal Chemistry, Chapter 6, pp. 51-60 (2003), the disclosures of which are hereby incorporated herein by reference, in their entireties), immunomodulatory disorders, inflammatory disorders, respiratory function disorders, depression, anxiety, mood disorders, stress-related disorders, sympathetic nervous system disorder, tussis, motor disorder, traumatic injury, stroke, cardiac arrhythmia, glaucoma, sexual dysfunction, shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, and substance addiction. In certain other embodiments, the spirocyclic heterocyclic derivatives are ligands of the δ opioid receptor and are useful, inter alia, in methods for providing cardioprotection following myocardial infarction, in methods for providing and maintaining an anaesthetic state, and in methods of detecting, imaging or monitoring degeneration or dysfunction of opioid receptors in a patient.
  • Thus, in accordance with preferred embodiments of the invention, there are provided methods of preventing or treating pain, comprising the step of administering to said patient an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In certain preferred embodiments, the present methods of preventing or treating pain may further comprise the administration to a patient of an effective amount of an agent for the treatment of neuralgia and/or neuropathic pain.
  • In another embodiment, the invention is directed to methods for preventing or treating gastrointestinal dysfunction, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating a urogenital tract disorder, such as incontinence (including, for example, stress urinary incontinence and urge urinary incontinence, and overactive bladder), comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In certain preferred embodiments, the present methods of preventing or treating a urogenital tract disorder may further comprise the administration to a patient of an effective amount of an agent for the treatment of incontinence.
  • In another embodiment, the invention is directed to methods of preventing or treating an immunomodulatory disorder, comprising the step of administering to a patient in need thereof an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV Immunomodulatory disorders include, but are not limited to, autoimmune diseases, collagen diseases, allergies, side effects associated with the administration of an anti-tumor agent, and side effects associated with the administration of an antiviral agent. Autoimmune diseases include, but are not limited to, arthritis, autoimmune disorders associated with skin grafts, autoimmune disorders associated with organ transplants, and autoimmune disorders associated with surgery.
  • In another embodiment, the invention is directed to methods of preventing or treating an inflammatory disorder, comprising the step of administering to a patient in need thereof an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV. Inflammatory disorders include, but are not limited to, arthritis, psoriasis, asthma, or inflammatory bowel disease.
  • In another embodiment, the invention is directed to methods of preventing or treating a respiratory function disorder, comprising the step of administering to a patient in need thereof an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV. Respiratory function disorders include but are not limited to asthma or lung edema.
  • In another embodiment, the invention is directed to methods for preventing or treating anxiety, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating a mood disorder, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In certain preferred embodiments, the present methods of preventing or treating a mood disorder may further comprise the administration to a patient of an effective amount of an agent for the treatment of depression.
  • In another embodiment, the invention is directed to methods for preventing or treating a stress-related disorder, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV. Stress-related disorders include, but are not limited to, post-traumatic stress disorder, panic disorder, generalized anxiety disorder, social phobia, and obsessive compulsive disorder.
  • In another embodiment, the invention is directed to methods for preventing or treating attention deficit hyperactivity disorder, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating sympathetic nervous system disorders, including hypertension, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating tussis, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating a motor disorder, including tremors, Parkinson's disease, Tourette's syndrome and dyskenesia, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In certain preferred embodiments, the present methods of preventing or treating a motor disorder may further comprise the administration to a patient of an effective amount of an agent for the treatment of Parkinson's disease.
  • In another embodiment, the invention is directed to methods for treating a traumatic injury to the central nervous system, including the spinal cord or brain, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating stroke, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating cardiac arrhythmia, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating glaucoma, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for preventing or treating sexual dysfunction, including premature ejaculation, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for treating a condition selected from the group consisting of shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, comprising the step of administering to a patient in need of such treatment an effective amount of a compound of the invention including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for treating substance addiction, including addictions to alcohol, nicotine or drugs such as opioids, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for improving organ and cell survival, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • Techniques for evaluating and/or employing the present compounds in methods for improving organ and cell survival and organ preservation are described, for example, in C. V. Borlongan et al., Frontiers in Bioscience (2004), 9(Suppl.), 3392-3398, Su, Journal of Biomedical Science (Base1) (2000), 7(3), 195-199, and U.S. Pat. No. 5,656,420, the disclosures of each of which are hereby incorporated herein by reference in their entireties.
  • In another embodiment, the invention is directed to methods for providing cardioprotection following myocardial infarction, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods for reducing the need for anesthesia, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXXIV.
  • In another embodiment, the invention is directed to methods of producing or maintaining an anesthetic state, comprising the step of administering to a patient in need of such treatment an effective amount of a compound as described herein including, for example, a compound of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, XXXI, XXXII, XXXIII and/or XXIV. The method may further comprise the step of administering to said patient an anesthetic agent, which may be co-administered with compound(s) of the invention. Suitable anesthetic agents include, for example, an inhaled anaesthetic, a hypnotic, an anxiolytic, a neuromuscular blocker and an opioid. Thus, in the present embodiment, compounds of the invention may be useful as analgesic agents for use during general anesthesia and monitored anesthesia care. Combinations of agents with different properties may be used to achieve a balance of effects needed to maintain the anaesthetic state.
  • Additional diseases and/or disorders which may be treated and/or prevented with the compounds and pharmaceutical compositions of the present invention include those described, for example, in WO2004/062562 A2, WO 2004/063157 A1, WO 2004/063193 A1, WO 2004/041801 A1, WO 2004/041784 A1, WO 2004/041800 A1, WO 2004/060321 A2, WO 2004/035541 A1, WO 2004/035574 A2, WO 2004041802 A1, US 2004082612 A1, WO 2004026819 A2, WO 2003057223 A1, WO 2003037342 A1, WO 2002094812 A1, WO 2002094810 A1, WO 2002094794 A1, WO 2002094786 A1, WO 2002094785 A1, WO 2002094784 A1, WO 2002094782 A1, WO 2002094783 A1, WO 2002094811 A1, the disclosures of each of which are hereby incorporated herein by reference in their entireties.
  • In certain embodiments, the present invention is directed to radiolabeled derivatives and isotopically labeled derivatives of compounds as described herein including, for example, compounds of formula XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIIA, XXVIII, XXIX, XXX, MI, XXXII, XXXIII and/or XXIV. Suitable labels include, for example, 2H, 3H, 11C, 13C, 13N, 15N, 15O, 18O, 18F and 34S. Such labeled derivatives may be useful for biological studies, for example, using positron emission tomography, for metabolite identification studies and the like. Such diagnostic imaging methods may comprise, for example, administering to a patient a radiolabeled derivative or isotopically labeled derivative of a compound of the invention, and imaging the patient, for example, by application of suitable energy, such as in positron emission tomography. Isotopically- and radio-labeled derivatives may be prepared utilizing techniques well known to the ordinarily skilled artisan.
  • The present invention will now be illustrated by reference to the following specific, non-limiting examples. Those skilled in the art of organic synthesis may be aware of still other synthetic routes to the invention compounds. The reagents and intermediates used herein are commercially available or may be prepared according to standard literature procedures.
  • Methods of Preparation
  • The examples listed in Tables 1, 2, and 3 may be prepared according to Schemes 1-57. The synthesis of compounds 1A-1U is outlined in Scheme 1. The 2′-hydroxyacetophenone derivatives 1.1a-1.1m were condensed with 1-Boc-4-piperidone 1.2 in neat pyrrolidine (method 1A) at room temperature or in refluxing methanol in the presence of pyrrolidine (method 1B) to provide N-Boc-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one derivatives 1.3. Conversion of the ketones 1.3 to the enol triflate derivatives 1.5 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 1.5 with either 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 (commercially available from Combi-Blocks Inc.) or 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0) (method 1C) or palladium, 10 wt. % (dry basis) on activated carbon (method 1D), lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 1.8 which were converted to the final products (compounds 1A-1T) under acidic conditions (method 1E: anhydrous HCl, diethyl ether, room temperature or method 1F: neat trifluoroacetic acid, room temperature). Demethylation of compound 1G using boron tribromide provided the corresponding phenolic derivative (compound 1U). The boronate derivative 1.7 was prepared in 4 steps from 2,5-dibromopyridine 1.9. Treatment of 2,5-dibromopyridine with n-butyllithium provided the corresponding lithiated derivative, which reacted with carbon dioxide to provide 5-bromopyridine-2-carboxylic acid 1.10. Treatment of the carboxylic acid derivative 1.10 with oxalyl chloride furnished the acyl chloride 1.11, which reacted with diethylamine 1.12 to provide 5-bromo-2-(N,N-diethylaminocarbonyl)-pyridine 1.13. Conversion of the aryl bromide 1.13 to the corresponding boron derivative 1.7 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro [1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, abbreviated as [Pd(dppf)Cl2.CH2Cl2].
  • The synthesis of compounds 2A-2F is outlined in Scheme 2. The 2′-5′-dihydroxyacetophenone derivative 2.1 was condensed with 1-Boc-4-piperidone 1.2 in refluxing methanol in the presence of pyrrolidine to provide N-Boc-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one derivative 2.2 which was converted to the silyl ether derivative 2.4 using tert-butyldimethylsilyl chloride 2.3. Conversion of the ketone 2.4 to the enol triflate derivative 2.5 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 2.5 with either 4-(N,N-diethylaminocarbonyl)-phenyl boronic acid 1.6 or 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0) (method 1C) or palladium, 10 wt. % (dry basis) on activated carbon (method 1D), lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 2.6. Removal of the silyl protecting group of 2.6 using a solution of tetrabutylammonium fluoride (TBAF) in tetrahydrofuran gave the phenolic derivatives 2.7 which were converted to the final products compounds 2A and 2B under acidic conditions. Preparation of each of the ether derivatives 2.9 from the phenols 2.7 was achieved by alkylation reaction using the appropriate alkyl bromide (2.8a, 2.8b) (method 2A) or alkyl iodide (2.8c) reagent (method 2C). In some instances, the ether derivatives 2.9 were also obtained from the phenols 2.7 using the Mitsunobu conditions, i.e., condensation of the phenols 2.7 with the appropriate alcohol (2.8d, 2.8e) in the presence of triphenylphosphine and diisopropyl azodicarboxylate (DIAD) (method 2B). Treatment of the Boc derivatives 2.9 with hydrochloric acid provided the final compounds 2C-F.
  • The synthesis of compounds 3A-AC is outlined in Scheme 3. Conversion of the phenols 2.7 to the triflate derivatives 3.1 was achieved using the triflating reagent N-phenylbis(trifluoromethanesulphonimide) 1.4. Palladium catalyzed carbonylation of 3.1, conducted in methanol or in a mixture dimethylsulfoxide/methanol using palladium (II) acetate, 1,1′-bis(diphenylphosphino)ferrocene (dppf) and carbon monoxide, provided the methyl esters 3.2 which were hydrolyzed under basic conditions to give the carboxylic acid derivatives 3.3. Coupling of the carboxylic acids 3.3 with various amines (3.4a-3.4q) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the primary, secondary, and tertiary amides 3.5. Treatment of the Boc derivatives 3.2, 3.3 and 3.5 with hydrochloric acid provided the final compounds 3A-3Y. Suzuki type coupling of the triflate derivative 3.1a (X═CH) with various organoboron reagents (3.6a-3.6d) in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), and/or dichloro[1,1′-bis(diphenylphosphino) ferrocene]palladium(II) dichloromethane, [Pd(dppf)Cl2CH2Cl2], lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 3.7 which were converted to the final products (compounds 3Z-3AC) under acidic conditions.
  • The synthesis of compounds 4A-4I is outlined in Scheme 4. Treatment of compound 1A with trifluoroacetic anhydride in tetrahydrofuran in the presence of triethylamine provided the trifluoroacetamide derivative 4.2 which was converted to the sulfonyl chloride 4.4 using sulfur trioxide N,N-dimethylformamide complex (4.3) as sulfating agent. Condensation of 4.4 with various primary and secondary amines (3.4, 4.5) afforded the sulfonamide derivatives 4.6 which were converted to the compounds 4A-4G under basic conditions. Treatment of the sulfonyl chloride 4.4 with ammonium hydroxide in acetonitrile provided the sulfonamide compound 411, which was further protected as its tert-butyloxycarbonyl (Boc) derivative 4.8 buy treatment with tert-butyloxycarbonyl anhydride (4.7). Acetylation of 4.8 using acetic anhydride (4.9) gave the acetylsulfonamide derivative 4.10 which was converted to compound 4I by treatment with iodotrimethylsilane.
  • The synthesis of compound 5A is described in Scheme 5. Condensation of hydrazine hydrate (5.1) with the sulfonyl chloride derivative 4.4 provided the sulfonyl hydrazide 5.2, which was converted to the sulfone 5.3 by treatment with methyl iodide (2.8c) in the presence of sodium acetate. Deprotection of the trifluoroacetamide protecting group of 5.3 under basic conditions (potassium carbonate, methanol/tetrahydrofuran/water) provided the final compound 5A.
  • The synthesis of compounds 6A-6E is described in Scheme 6. Nitration of the trifluoroacetamide 4.2 using nitronium tetrafluoroborate complex (6.1) as nitrating reagent provided predominantly the mono-nitro isomer 6.2. Reduction of the nitro functionality of 6.2 using tin(II) chloride dihydrate (6.3) gave the aniline derivative 6.4, which reacted with the sulfonyl chloride derivatives 6.5 or with acetyl chloride (6.7) to provide the sulfonamides 6.6 or the acetamide 6.8, respectively. Deprotection of the trifluoroacetamide protecting group of 6.2, 6.4, 6.6 and 6.8 under basic conditions (potassium carbonate, methanol/tetrahydrofuran/water) provided the final compounds (compounds 6A-6E).
  • The synthesis of compounds 7A-7E is described in Scheme 7. Buchwald type coupling of the triflate derivative 3.1a with diphenylmethanimine (7.1) in toluene in the presence of tris(dibenzylideneacetone)dipalladium (0) [Pd2(dba)3], 1,1′-bis(diphenylphosphino)ferrocene (dppf) and sodium tert-butoxide afforded the benzophenone imine derivative 7.2, which was converted to the aniline 7.3 by treatment with hydroxylamine hydrochloride in the presence of sodium acetate. Treatment of 7.3 with methanesulfonyl chloride (7.4) in dichloromethane in the presence of triethylamine provided the bis-methanesulfonamide 7.5, which was hydrolyzed to the mono methanesulfonamide derivative 7.6 under basic conditions. Deprotection of the tert-butyloxycarbonyl protecting group of 7.6 under acidic conditions provided the final compound 7A. Compound 7B was obtained in two steps from 7.6. Alkylation of 7.6 with methyl iodide (2.8c) in tetrahydrofuran in the presence of sodium hydride provided the N-methylsulfonamide 7.7, which was converted to compound 7B under acidic conditions. Treatment of the aniline derivative 6.4 with methanesulfonyl chloride (7.4) in dichloromethane in the presence of triethylamine provided the bis-methanesulfonamide 7.8, which was hydrolyzed to the mono-methanesulfonamide derivative compound 7A under basic conditions. During the course of this reaction, the N-methyl piperidine derivative compound 7C was identified as a side product. The separation of the mixture containing compounds 7A and 7C was achieved by first treating the mixture of compounds 7A/7C with tert-butyloxycarbonyl anhydride (4.7) which provided the Boc derivative 7.6 and unreacted compound 7C, followed by purification of compound 7C using flash column chromatography. Buchwald type coupling of the triflate derivative 3.1a with pyrrolidine (3.4 k) or morpholine (3.4p) in ethylene glycol dimethyl ether in the presence of tris(dibenzylideneacetone)dipalladium (0) [Pd2(dba)3], the phosphine ligand 2-(di-t-butylphosphino)biphenyl 7.9 and potassium phosphate afforded the derivatives 7.10, which were converted to compounds 7D,E under acidic conditions.
  • The synthesis of compounds 8A-8F is outlined in Scheme 8. The 2′-3′-dihydroxyacetophenone derivative 8.1 was condensed with 1-Boc-4-piperidone 1.2 in refluxing methanol in the presence of pyrrolidine to provide the N-Boc-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one derivative 8.2 which was converted to the silyl ether derivative 8.3 using tert-butyldimethylsilyl chloride 2.3. The ketone 8.3 was converted to the enol triflate derivative 8.4 using the triflating reagent N-phenylbis(trifluoromethanesulphonimide) 1.4. Suzuki type coupling of the enol triflate derivative 8.4 with either 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 or 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of palladium, 10 wt. % (dry basis) on activated carbon, lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 8.5. Removal of the silyl protecting group of 8.5 using a solution of tetrabutylammonium fluoride (TBAF) in tetrahydrofuran gave the phenolic derivatives 8.6 which were converted to the final products (compounds 8A and 8B) under acidic conditions. Preparation of the ether derivatives 8.7 from the phenols 8.6 was achieved by alkylation using the appropriate alkyl bromide (2.8a) or methyl iodide (2.8c) reagent. Treatment of the Boc derivatives 8.7 with hydrochloric acid provided the final compounds 8C-8F.
  • The synthesis of compounds 9A-9B is outlined in Scheme 9. The 2′-4′-dihydroxyacetophenone derivative 9.1 was condensed with 1-Boc-4-piperidone 1.2 in refluxing methanol in the presence of pyrrolidine to provide the N-Boc-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one derivative 9.2 which was converted to the silyl ether derivative 9.3 using tert-butyldimethylsilyl chloride 2.3. Conversion of the ketone 9.3 to the enol triflate derivative 9.4 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 9.4 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the phenolic derivative 9.5 (simultaneous removal of the silyl protecting group occurred under the Suzuki coupling conditions). Alkylation of the phenol 9.5 with (bromomethyl)cyclopropane (2.8a) in acetone in the presence of potassium carbonate provided the ether derivative 9.6 which was converted to compound 9A under acidic conditions. Treatment of the phenol 9.5 with methyl chlorodifluoroacetate (9.7) in N,N-dimethylformamide in the presence of cesium carbonate provided the ether derivative 9.8 which was converted to compound 9B under acidic conditions.
  • The synthesis of compounds 10A-10J is outlined in Scheme 10. Conversion of the phenol 9.5 to the triflate derivative 10.1 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Palladium catalyzed carbonylation of 10.1, conducted in a mixture N,N-dimethylformamide/methanol using palladium (II) acetate, 1,1′-bis(diphenylphosphino)ferrocene (dppf), and carbon monoxide, provided the methyl ester 10.2 which was hydrolyzed under basic conditions to give the carboxylic acid derivative 10.3. Coupling of the carboxylic acid 10.3 with various amines (3.4a,c,j,k,p; 1.12) using either O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU) (method 10B) or O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (method 10A) as coupling agents afforded the primary, secondary, and tertiary amides 10.4. The dimethylamide derivative 10.4b (R1═H, R2═CH3) was obtained by heating a mixture of the ester 10.2 with methylamine (3.4b) in methanol in a sealed tube. Treatment of the Boc derivatives 10.2, 10.3 and 10.4 with hydrochloric acid provided the final compounds 10A-10I. Treatment of the ester 10.2 with lithium borohydride in tetrahydrofuran provided the primary alcohol 10.5 which was converted to the compound 10J under acidic conditions.
  • The synthesis of compounds 11A-11I is outlined in Scheme 11. The 2′-6′-dihydroxyacetophenone derivative 11.1 was condensed with 1-Boc-4-piperidone 1.2 in refluxing methanol in the presence of pyrrolidine to provide the N-Boc-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one derivative 11.2 which was converted to the methoxymethyl (MOM) ether derivative 11.4 using chloro(methoxy)methane (11.3). Conversion of the ketone 11.4 to the enol triflate derivative 11.5 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 11.5 with either 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 or 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 11.6. Removal of the MOM and the Boc protecting groups of 11.6 in methanol at room temperature in the presence of hydrochloric acid (anhydrous solution in dioxane) afforded the phenolic compounds 11A and 11B which were converted to the corresponding Boc derivatives 11.7 by treatment with tert-butyloxycarbonyl anhydride (4.7). Preparation of the ether derivatives 11.9a [X═CH; R═CH2c(C3H5)], 11.9b [X═N; R═CH2c(C3H5)] and 11.9d [X═N; R=c(C5H9)] from the corresponding phenols 11.7a [X═CH] or 11.7b [X═N] was achieved using the Mitsunobu conditions, i.e., condensation of the phenols 11.7a or 11.7b with cyclopropylmethanol (2.8e) or cyclopentanol (11.10) in dichloromethane in the presence of triphenylphosphine and diethyl azodicarboxylate (DEAD). The cyclobutyl ether 11.9c [X═CH; R=c(C4H7)] was obtained by alkylation of the corresponding phenol 11.7a [X═CH] with bromocyclobutane in acetone in the presence of potassium carbonate. Treatment of the Boc derivatives 11.9 with hydrochloric acid provided the final compounds 11C-11F. Treatment of the phenol 11.2 with methyl chlorodifluoroacetate (9.7) in N,N-dimethylformamide in the presence of cesium carbonate provided the ether derivative 11.11. Conversion of the ketone 11.11 to the enol triflate derivative 11.12 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 11.12 with either 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 or 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, potassium phosphate, and potassium bromide, afforded compounds 11.13. Removal of the Boc protecting group of 11.13 in dichloromethane at room temperature in the presence of hydrochloric acid (anhydrous solution in diethyl ether) afforded the compounds 11G and 11H. Conversion of the aryl bromide 32.2b to the corresponding boron derivative 11.14 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct. Suzuki type coupling of the enol triflate derivative 11.5 with the boron derivative 11.14 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), potassium bromide and potassium phosphate afforded compound 11.15. Removal of the Boc and the MOM protecting groups of 11.15 in methanol at room temperature in the presence of hydrochloric acid (anhydrous solution in diethyl ether) afforded the compound 11I.
  • The synthesis of compounds 12A-12L is outlined in Scheme 12. Conversion of the phenol 11.2 to the triflate derivative 12.1 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Palladium catalyzed Negishi-type coupling of 12.1 with methylzinc chloride (12.2a), propylzinc bromide (12.2b), or butylzinc bromide (12.2c), conducted in tetrahydrofuran using tetrakis triphenylphosphine palladium (0) as catalyst, provided the ketones 12.3. Conversion of the ketones 12.3 to the enol triflate derivatives 12.4 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 12.4 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 or 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 using either method 1C (tetrakis triphenylphosphine palladium (0), lithium chloride, aqueous solution of sodium carbonate, ethylene glycol dimethyl ether) or method 12A (tetrakis triphenylphosphine palladium (0), potassium bromide, potassium phosphate, dioxane) afforded compounds 12.5. Removal of the Boc protecting group of 12.5 in dichloromethane at room temperature in the presence of hydrochloric acid (anhydrous solution in diethyl ether) afforded compounds 12A and 12H-12L. Palladium catalyzed carbonylation of 12.1, conducted in a mixture N,N-dimethylformamide/methanol using palladium (II) acetate, 1,3-bis(diphenylphosphino)propane (dppp) and carbon monoxide, provided the methyl ester 12.6 which was hydrolyzed under basic conditions (lithium hydroxide, methanol/tetrahydrofuran) to give the carboxylic acid derivative 12.7. Coupling of the carboxylic acid 12.7 with dimethylamine (3.4j) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the dimethylaminocarbonyl derivative 12.8. Conversion of 12.8 to the enol triflate derivative 12.9 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 12.9 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 12.10. Removal of the Boc protecting group of 12.10 in dichloromethane at room temperature in the presence of hydrochloric acid (anhydrous solution in diethyl ether) afforded compound 12G (R1═R2═CH3). Conversion of 12.6 to the enol triflate derivative 12.11 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 12.11 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the ester 12.12 which was hydrolyzed under basic conditions (potassium tert-butoxide, diethyl ether, water) to give the carboxylic acid 12.13. Coupling of the carboxylic acid 12.13 with various amines (12.15 or 3.4b-3.4d) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the primary and secondary aminocarbonyl derivatives 12.14. Treatment of the Boc derivatives 12.13 and 12.14 with hydrochloric acid provided the final compounds 12B-12F.
  • The synthesis of compounds 13A-13S is outlined in Scheme 13. The 2′-hydroxyacetophenone derivative 1.1a was condensed with 1-Boc-4-piperidone 1.2 in refluxing methanol in the presence of pyrrolidine to provide N-Boc-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one 1.3a. Conversion of 1.3a to the enol triflate derivative 1.5a was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 1.5a with 4-(methoxycarbonyl)phenylboronic acid (13.1) in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the ester 13.2 which was hydrolyzed under basic conditions (lithium hydroxide, methanol/tetrahydrofuran/water) to give the carboxylic acid 13.3. Coupling of the carboxylic acid 13.3 with various amines (3.4a-3.4c, 3.4e, 3.4j-3.4 k, 3.4o-3.4q; 13.4a-13.4h) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the primary, secondary, and tertiary aminocarbonyl derivatives 13.5. Treatment of the Boc derivatives 13.3 and 13.5 with hydrochloric acid provided the final compounds 13A-13R. Hydrolysis of compound 130 under basic conditions (sodium hydroxide, ethanol/tetrahydrofuran) provided the carboxylic acid compound 13S.
  • The synthesis of compounds 14A-14C is outlined in Scheme 14. Suzuki type coupling of the enol triflate derivative 1.5a with 4-cyanophenylboronic acid (14.1) in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the cyanide 14.2 which was converted to the tetrazole 14.4 using sodium azide (14.3) and zinc bromide in a solution isopropanol/water. Alkylation of 14.4 with methyl iodide (2.8c) in N,N-dimethylformamide in the presence of triethylamine afforded the two regioisomers 14.5 (major isomer) and 14.6 (minor isomer) separated by silica gel column chromatography. The Boc protecting group of 14.4, 14.5, and 14.6 was removed using hydrochloric acid to generate the compounds 14A-14C. Alternatively, the Boc protecting group of 14.4 was also removed using trifluoroacetic acid to give 14A.
  • The synthesis of compounds 15A-15N is outlined in Scheme 15. Alkylation of 14.4 with the alkyl bromide derivatives 15.1a-15.1e in N,N-dimethylformamide in the presence of triethylamine afforded the regioisomers 15.2 (major isomers) and 15.3 (minor isomers) separated by silica gel column chromatography. The Boc protecting group of 15.2 and 15.3 was removed using hydrochloric acid to generate the compounds 15A-15J. Hydrolysis of compounds 15A or 15C-15E under basic conditions (sodium hydroxide, methanol (or ethanol)/tetrahydrofuran/water) provided the corresponding carboxylic acids compounds 15K-15N, respectively. In some instances, compounds 15K-15N were also obtained in two steps from 15.2, i.e. by basic hydrolysis of the ester functionality of 15.2 followed by deprotection of the Boc derivatives 15.4 under acidic conditions.
  • The synthesis of compounds 16A-16C is outlined in Scheme 16. Suzuki type coupling of the enol triflate derivative 1.5a with 3-cyanophenylboronic acid (16.1) in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the cyanide 16.2 which was converted to the tetrazole 16.3 using sodium azide (14.3) and zinc bromide in a solution isopropanol/water. Alkylation of 16.3 with methyl iodide (2.8c) in N,N-dimethylformamide in the presence of triethylamine afforded the two regioisomers 16.4 (major isomer) and 16.5 (minor isomer) separated by silica gel column chromatography. The Boc protecting group of 16.3, 16.4, and 16.5 was removed using hydrochloric acid to generate the compounds 16A-16C.
  • The synthesis of compounds 17A-17F is outlined in Scheme 17. Alkylation of 16.3 with the alkyl bromide derivatives 15.1a or 15.1c in N,N-dimethylformamide in the presence of triethylamine afforded the regioisomers 17.1 (major isomers) and 17.2 (minor isomers) separated by silica gel column chromatography. Alkylation of 16.3 with 4-(2-bromoethyl)morpholine (17.3) in N,N-dimethylformamide in the presence of triethylamine afforded the isomer 17.4. The Boc protecting group of 17.1, 17.2, and 17.4 was removed using hydrochloric acid to generate the compounds 17A-17D. Hydrolysis of compounds 17A and 17B under basic conditions (sodium hydroxide, methanol/tetrahydrofuran/water) provided the corresponding carboxylic acids compound 17E and compound 17F, respectively. In some instances compounds 17E and 17F could also be obtained in two steps from 17.1, i.e. by basic hydrolysis of the ester functionality of 17.1 followed by deprotection of the Boc derivatives 17.5 under acidic conditions.
  • The synthesis of compounds 18A-18C is outlined in Scheme 18. Coupling of the carboxylic acid 13.3 with ammonium chloride (3.4a) in acetonitrile in the presence of diisopropylethylamine using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the primary aminocarbonyl derivative 13.5a which was converted to the thioamide 18.2 using the Lawesson's reagent (18.1) [2,4-bis(4-methoxyphenyl)-1,3-dithia-2,4-diphosphetane-2,4-disulfide]. Condensation of the thioamide 18.2 with 1-bromo-3,3-dimethylbutan-2-one (18.3a) or 2-bromo-1-phenylethanone (18.3b) afforded the thiazole derivatives 18.4 which were converted to the final compounds (compounds 18A and 18B) under acidic conditions. Condensation of the nitrile derivative 14.2 with hydroxylamine hydrochloride (18.5) in ethanol in the presence of triethylamine afforded the N-hydroxybenzamidine derivative 18.6 which reacted with acetyl chloride (6.7) in refluxing pyridine to give the 1,2,4-oxadiazole derivative 18.7. Deprotection of the Boc functionality of 18.7 under acidic conditions afforded compound 18C.
  • The synthesis of compound 19A-19D is outlined in Scheme 19. The 2′-hydroxyacetophenone 1.1a was condensed with benzyl 4-oxopiperidine-1-carboxylate (19.1) in refluxing methanol in the presence of pyrrolidine to provide N-Cbz-spiro[2H-1-benzopyran-2,4′-piperidine]-4(3H)-one (19.2). Conversion of the ketone 19.2 to the enol triflate derivative 19.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Conversion of the enol triflate 19.3 to the corresponding boron derivative 19.4 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, abbreviated as [Pd(dppf)Cl2.CH2Cl2]. Suzuki type coupling of the boronate derivative 19.4 with tert-butyl 4-bromophenylcarbamate 19.5 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the tert-butyloxycarbonyl (Boc) protected aniline derivative 19.6. Acidic hydrolysis of 19.6 provided the aniline derivative 19.7 which reacted with acyl chlorides 19.8a, 19.8b, isopropylsulfonyl chloride (6.5b) or ethyl isocyanate (19.11) to give the corresponding amide derivatives 19.9, sulfonamide derivative 19.10 or urea derivative 19.12, respectively. The derivatives 19.9, 19.10 and 19.12 were converted to compounds 19A-19D by treatment with iodotrimethylsilane.
  • The synthesis of compounds 20A-20R is outlined in Scheme 20. The tertiary amine derivatives compounds 20A-20R were obtained from the secondary amines of general formula 20I, by reductive amination methods (methods 20A or 20B) using the aldehydes 20.1a-20.1d and sodium cyanoborohydride as reducing agent or by alkylation method (method 20C) using the bromides 2.8a, 20.2a-e as the alkylating reagent.
  • The synthesis of compounds 21A-21F is outlined in Scheme 21. Condensation of 1-Boc-4-piperidone 1.2 with ethyl diazoacetate (21.1) in the presence of boron trifluoride diethyl etherate provided 1-tert-butyl 4-ethyl 3-oxoazepane-1,4-dicarboxylate in equilibrium with its enol form (21.2). Ester hydrolysis followed by decarboxylation of 21.2 under acidic conditions provided the azepan-3-one (21.3), which was protected as its Boc derivative 21.4 by treatment with tert-butyloxycarbonyl anhydride (4.7). The 2′-hydroxyacetophenone 1.1a was condensed with 21.4 in refluxing methanol in the presence of pyrrolidine to provide the racemic ketone 21.5. Conversion of 21.5 to the enol triflate derivative 21.6 was achieved using the triflating reagent N-phenylbis(trifluoromethanesulphonimide) 1.4. Suzuki type coupling of the enol triflate derivative 21.6 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid (1.6) in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the racemic derivative 21.7, which was hydrolyzed under acidic conditions to give the compound 21A (racemic mixture). The two enantiomers derived from 21.7, i.e. compounds 21.7a and 21.7b, were separated by chiral HPLC. The pure enantiomers 21.7a and 21.7b were converted to compounds 21B and 21C, respectively under acidic conditions. Palladium catalyzed hydrogenation of compounds 21B and 21C afforded compounds 21D (diastereoisomeric mixture) and 21E (diastereoisomeric mixture), respectively. Treatment of compound 21A with benzyl chloroformate (21.8) in dichloromethane in the presence of triethylamine provided the Cbz-protected derivative 21.9, which was converted to the sulfonyl chloride 21.10 using sulfur trioxide N,N-dimethylformamide complex (4.3) as sulfating agent. Condensation of 21.10 with ethylamine (3.4c) in dichloromethane in the presence of triethylamine, afforded the ethyl sulfonamide derivative 21.11 which was converted to compound 21F by treatment with iodotrimethylsilane.
  • The synthesis of compounds 22A-22F is outlined in Scheme 22. Treatment of compound 21B (most active enantiomer) with trifluoroacetic anhydride (4.1) in tetrahydrofuran in the presence of triethylamine provided the trifluoroacetamide derivative 22.1 which was converted to the sulfonyl chloride 22.2 using sulfur trioxide N,N-dimethylformamide complex (4.3) as sulfating agent. Condensation of 22.2 with various primary amines (3.4b, 3.4c, 3.4d, 3.4 g) afforded the sulfonamide derivatives 22.3 which were converted to compounds 22A-22D under basic conditions. Condensation of hydrazine hydrate (5.1) with the sulfonyl chloride derivative 22.2 provided the sulfonyl hydrazide 22.4, which was converted to the sulfones 22.5 and 22.7 by treatment with methyl iodide (2.8c) and ethyl iodide (22.6), respectively, in the presence of sodium acetate. Deprotection of the trifluoroacetamide protecting group of 22.5 and 22.7 under basic conditions (potassium carbonate, methanol, water) provided the corresponding methyl sulfone (compound 22E) and ethyl sulfone (compound 22F) derivatives.
  • The synthesis of compounds 23A-23C is outlined in Scheme 23. The 2′-hydroxyacetophenone 1.1a was condensed with tert-butyl 3-oxopyrrolidine-1-carboxylate (23.1a) or tert-butyl 3-oxopiperidine-1-carboxylate (23.1b) in refluxing methanol in the presence of pyrrolidine to provide the racemic ketones 23.2a (n=0) and 23.2b (n=1), respectively. Conversion of the ketones 23.2 to the enol triflate derivatives 23.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 23.3 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the Boc derivatives 23.4 which were converted to the final products compounds 23A and 23B (racemic mixtures) under acidic conditions. The 2′-hydroxyacetophenone 1.1a was also condensed with 1-Boc-4-nortropinone (23.5) in refluxing methanol in the presence of pyrrolidine to provide the ketone 23.6. Conversion of the ketone 23.6 to the enol triflate derivative 23.7 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 23.7 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the Boc derivative 23.8 which was converted to the final product compound 23C under acidic conditions.
  • The synthesis of compounds 24A-24G is outlined in Scheme 24. The 2′-hydroxyacetophenone 1.1a was condensed with 1,4-cyclohexanedione mono-ethylene ketal (24.1) in refluxing methanol in the presence of pyrrolidine to provide the ketone 24.2. Conversion of the ketone 24.2 to the enol triflate derivative 24.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 24.3 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 24.4 which was converted to the ketone compound 24A under acidic conditions. The reduction of the ketone compound 24A, conducted in tetrahydrofuran in the presence of sodium borohydride, provided the corresponding alcohol derivatives compounds 24B and 24C. Treatment of the ketone compound 24A with propylamine (3.4d) or dimethylamine (3.4j) under reductive amination conditions using sodium cyanoborohydride as reducing agent, provided the amines compounds 24D-24G.
  • The synthesis of compound 25A is outlined in Scheme 25. The 2′-hydroxyacetophenone 1.1a was also condensed with tetrahydropyran-4-one (25.1) in refluxing methanol in the presence of pyrrolidine to provide the ketone 25.2. Conversion of the ketone 25.2 to the enol triflate derivative 25.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 25.3 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 25A.
  • The synthesis of compounds 26A-26B is outlined in Scheme 26. Palladium catalyzed Negishi-type coupling of 1.5a with 4-cyanobenzylzinc bromide (26.1) conducted in tetrahydrofuran using tetrakis triphenylphosphine palladium (0) as catalyst, provided the nitrile 26.2. Acidic hydroysis of the nitrile 26.2 provided the carboxylic acid derivatives 26.3a and 26.3b (compounds 26.3a and 26.3b were separated by column chromatography; however, the following step was conducted using the mixture 26.3a/26.3b). Treatment of the mixture 26.3a/26.3b with methanol in the presence of hydrochloric acid afforded the piperidine esters 26.4a/26.4b which were converted to the corresponding Boc derivatives 26.5a/26.5b by treatment with tert-butyloxycarbonyl anhydride (4.7). Hydrolysis of the esters 26.5a/26.5b in basic conditions gave the carboxylic acid derivatives 26.6a/26.6b. Coupling of the carboxylic acid derivatives 26.6a/26.6b with diethylamine (1.12) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the dimethylaminocarbonyl derivatives 26.7a/26.7b. Removal of the Boc protecting group of 26.7a/26.7b in dichloromethane at room temperature in the presence of hydrochloric acid (anhydrous solution in dioxane) afforded compounds 26A and 26.8 which were separated by column chromatography. Palladium catalyzed hydrogenation of compound 26.8 afforded compound 26B.
  • The synthesis of compounds 27A-27W is outlined in Scheme 27. The saturated derivatives (compounds 27A, 27D, 27G, 27H, 27K, 27N, and 27W, as their racemic mixtures) were obtained by hydrogenation of the unsaturated analogs (compounds 1A, 1D, 2C, 1N, 1O, 1S, and 1E), respectively, in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon (method 27A) or palladium hydroxide, 20 wt. % Pd (dry basis) on carbon (Pearlman's catalyst (method 27B)). Hydrogenation of 11.6a in methanol in the presence of palladium hydroxide, 20 wt. % Pd (dry basis) on carbon (Pearlman's catalyst) provided the saturated derivative 27.1. Acidic hydrolysis of 27.1 provided compound 27T. Hydrolysis of 2.7a in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon, provided the saturated derivative 27.6. Acidic hydrolysis of 27.6 provided the compound 27Q. Chiral separation of the enantiomers derived from 27.1 provided compounds 27.4 and 27.5. The enantiomers 27.4 and 27.5 were converted to compounds 27U and 27V, respectively under acidic conditions. Chiral separation of the enantiomers derived from each of the racemic compounds (compounds 27A, 27 D, 27G, 27H, 27K, 27N, 27Q and 27W) provided compounds 27B, 27E, 27I, 27L, 27O, 27R (pure enantiomer) and compounds 27C, 27F, 27J, 27M, 27P, 27S (pure enantiomer). Condensation of compound 27B with (1S)-(+)-10-camphorsulfonyl chloride (27.2) (used as chiral resolving agent) in dichloromethane in the presence of triethylamine provided the chiral sulfonamide derivative 27.3. The absolute configuration of 27.3 was determined by X-ray crystallography, therefore establishing the absolute configuration of compound 27B, and therefore by inference, its enantiomer, compound 29C.
  • The synthesis of compounds 28A-28E is outlined in Scheme 28. Condensation of benzyl 4-oxopiperidine-1-carboxylate (19.1) with ethyl cyanoacetate (28.1) in the presence of acetic acid and ammonium acetate gave the unsaturated ester 28.2. Compound 28.2 was subjected to conjugate addition by reaction with organo cuprate reagents derived from benzyl or methoxybenzyl magnesium chloride (28.3a and 28.3b, respectively) and copper (I) cyanide to yield the cyano esters 28.4. Treatment of the conjugate addition product 28.4a (Rv═H) with concentrated sulfuric acid at 90° C. provided the amino ketone 28.5. Treatment of 28.5 with benzyl chloroformate (21.8) in dichloromethane in the presence of triethylamine provided the corresponding Cbz-protected derivative 28.6a (Rv═H). Decarboxylation of 28.4b (Rv═OCH3) by treatment with sodium chloride in dimethylsulfoxide containing small amount of water at 160° C. afforded the nitrile 28.9. Hydrolysis of the nitrile functionality of 28.9 to the methyl ester group by treatment with methanol in the presence of sulfuric acid provided the corresponding piperidine derivative (Cleavage of the Cbz protecting group of 28.9 occurred during the course of the hydrolysis). Treatment of the piperidine derivative with benzyl chloroformate afforded the compound 28.10. The ester 28.10 was hydrolyzed with lithium hydroxide to furnish the carboxylic acid 28.11. Treatment of the acid 28.11 with oxalyl chloride followed by reaction of the resulting acyl chloride with aluminum chloride yielded the corresponding spiro piperidine derivative which was further protected as its CBz derivative 28.6b (Rv═OCH3) by treatment with benzylchloroformate. Conversion of the ketones 28.6 to the enol triflate derivatives 28.7 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 28.7 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivatives 28.8 which were converted to the compounds 28A and 28B by treatment with iodotrimethylsilane. The compounds 28C and 28D (racemic mixtures) were obtained by hydrogenation of unsaturated derivatives 28.8 in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon. Suzuki type coupling of the enol triflate derivative 28.7a (Rv═H) with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 28.12 which was converted to the compound 28E by treatment with iodotrimethylsilane.
  • The synthesis of compounds 29A-29D is outlined in Scheme 29. The Negishi coupling of the enol triflate 28.7a with 4-(ethoxycarbonyl)phenylzinc iodide (29.1) in tetrahydrofuran in the presence of tetrakis triphenylphosphine palladium (0) gave the ester 29.2, which was hydrolyzed with lithium hydroxide to afford the carboxylic acid 29.3. Coupling of the carboxylic acid 29.3 with isopropylamine (3.4h) or 1-ethylpropylamine (29.4) using 2-chloro-1-methylpyridinium iodide (Mukaiyama acylating reagent) as coupling agent afforded the secondary aminocarbonyl derivatives 29.5, which were converted to the compounds 29A and 29B by treatment with iodotrimethylsilane. Curtius rearrangement of the carboxylic acid 29.3 by reaction with diphenylphosphoryl azide (29.6) in the presence of tert-butyl alcohol provided the tert-butyloxycarbonyl (Boc) protected aniline derivative 29.7. Acidic hydrolysis of 29.7 provided the aniline derivative 29.8 which reacted with propionyl chloride 29.9 or methanesulfonyl chloride (7.4) to give the corresponding amide derivative 29.10 or sulfonamide derivative 29.11, respectively. The derivatives 29.10 and 29.11 were converted to compounds 29C and 29D, respectively, by treatment with iodotrimethylsilane.
  • The synthesis of compound 30A is outlined in Scheme 30. Wittig type condensation of 1-benzoyl-4-piperidone (30.1) with methyl(triphenylphosphoranylidene)acetate (30.2) in toluene gave the unsaturated ester 30.3. Compound 30.3 was subjected to conjugate addition by reaction with benzenethiol (30.4) to yield the thioether 30.5. Treatment of the conjugate addition product 30.5 with concentrated sulfuric acid provided the cyclized product 30.6, which was converted to the sulfone 30.7 by oxidation using a solution of hydrogen peroxide in glacial acetic acid. Acidic hydrolysis of 30.7 provided the amine 30.8, which was treated with tert-butyloxycarbonyl anhydride (4.7) to give the Boc protected derivative 30.9. Conversion of the ketone 30.9 to the enol triflate derivative 30.10 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 30.10 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 30.11 which was converted to compound 30A under acidic conditions.
  • The synthesis of compounds 31A-31AA is outlined in Scheme 31. Suzuki type coupling of the enol triflate derivative 1.5a with the commercially available boronic acid derivatives 13.1, 14.1, 16.1 or 31.1a-31.1u in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 13.2, 14.2, 16.2 and 31.2, respectively. Compounds 13.2, 14.2, 16.2 and 31.2 were converted to the final products compounds 31A-31X under acidic conditions (method 1E: anhydrous HCl, diethyl ether, room temperature or method 1F: neat trifluoroacetic acid (with optional dichloromethane), room temperature or method 31A: anhydrous HCl, methanol, dioxane, reflux). Treatment of the nitrile 16.2 with lithium aluminum hydride in tetrahydrofuran provided the diamine compound 31Y, which reacted with acetyl chloride (6.7) or methanesulfonyl chloride (7.4) to give the corresponding amide derivative compounds 31Z or the sulfonamide derivative compound 31AA, respectively.
  • The synthesis of compounds 32A-32Z is outlined in Scheme 32. Conversion of the enol triflate 1.5a to the corresponding boron derivative 32.1 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, abbreviated as [Pd(dppf)Cl2.CH2Cl2]. Suzuki type coupling of the boronate derivative 32.1 with various aryl bromide derivatives 32.2 under different conditions [method 1C: ethylene glycol dimethyl ether, tetrakis triphenylphosphine palladium (0), lithium chloride, aqueous solution of sodium carbonate; method 1D: ethylene glycol dimethyl ether, palladium, 10 wt. % (dry basis) on activated carbon, lithium chloride, aqueous solution of sodium carbonate; method 12A: tetrakis triphenylphosphine palladium (0), potassium bromide, potassium phosphate, dioxane] afforded the derivatives 32.3, which were converted to compounds 32A-32I or 32K-32Z under acidic conditions. The tert-butyl sulfonamide derivative compound 32.3b was converted to the sulfonamide compound 32J by treatment with trifluoroacetic acid. The derivatives 32.2 used in the Suzuki coupling step were prepared as follows. Coupling of the carboxylic acid 32.4 with diethylamine (1.12) using 2-chloro-1-methylpyridinium iodide (Mukaiyama acylating reagent) as coupling agent afforded 2-(4-bromophenyl)-N,N-diethylacetamide (32.2a). The sulfone derivatives 32.2j-32.2p were obtained in two steps from 4-bromobenzenethiol (32.7). Alkylation of 32.7 with the alkyl bromide derivatives 20.2, 2.8 or 32.8 in acetonitrile in the presence of triethylamine (method 32A) or in N,N-dimethylformamide in the presence of sodium hydride (method 32B) provided the thioether derivatives 32.9, which were oxidized to the sulfone derivatives 32.2j-32.2p in glacial acetic acid in the presence of an aqueous solution of hydrogen peroxide. Coupling of 4-bromobenzene-1-sulfonyl chloride (32.5) with various amines (3.4, 1.12, 13.4 or 32.6) in tetrahydrofuran in the presence of triethylamine provided the sulfonamides 32.2b-32.2i. Acylation of N-methyl-4-bromoaniline (32.10) with various acyl chloride derivatives (19.8, 32.11 or 6.7) in dichloromethane in the presence of triethylamine provided the amides 32.2q-32.2u, 32.2x, 32.2y. The aryl bromides 32.2v and 32.2w are commercially available.
  • The synthesis of compounds 33A-33N is outlined in Scheme 33. Suzuki type coupling of the boronate derivative 32.1 with various aryl bromide derivatives 33.1 under different conditions [method 1C: ethylene glycol dimethyl ether, tetrakis triphenylphosphine palladium (0), lithium chloride, aqueous solution of sodium carbonate; method 1D: ethylene glycol dimethyl ether, palladium, 10 wt. % (dry basis) on activated carbon, lithium chloride, aqueous solution of sodium carbonate; method 33A: ethylene glycol dimethyl ether, dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, abbreviated as [Pd(dppf)Cl2.CH2Cl2], lithium chloride, potassium phosphate; method 33B: dioxane, tetrakis triphenylphosphine palladium (0), potassium bromide, potassium phosphate] afforded the derivatives 33.2, which were converted to compounds 33A-33K, 33M and 33N under acidic conditions. The derivatives 33.1 used in the Suzuki coupling step were either obtained from commercial sources (33.1a-e,l,m) or prepared as follows. Coupling of 5-bromopyridine-3-carboxylic acid (33.3) or 6-bromopyridine-2-carboxylic acid (33.4) with diethylamine (1.12) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the diethylaminocarbonyl derivative derivatives 33.1f and 33.1g, respectively. Treatment of 2,5-dibromopyridine (1.9) with n-butyllithium provided the corresponding lithiated derivative, which reacted with carbon dioxide to provide 5-bromopyridine-2-carboxylic acid 1.10. The carboxylic acid 1.10 was also obtained by acidic hydrolysis of commercially available 5-bromopyridine-2-carbonitrile (33.1e). Treatment of the carboxylic acid derivative 1.10 with oxalyl chloride furnished the acyl chloride 1.11, which reacted with dimethylamine (3.4j), ethylamine (3.4c) or methylamine (3.4b) to provide the corresponding aminocarbonyl derivatives 33.1h, 33.1i and 33.1j, respectively. Treatment of commercially available 5-bromo-2-iodopyrimidine (33.5) with n-butyllithium provided the corresponding lithiated derivative, which reacted with carbon dioxide to provide 5-bromopyrimidine-2-carboxylic acid (33.6). Treatment of the carboxylic acid derivative 33.6 with oxalyl chloride furnished the acyl chloride 33.7, which reacted with diethylamine 1.12 to provide 5-bromo-2-(N,N-diethylaminoarbonyl)-pyrimidine 33.1k.
  • Hydrolysis of the nitrile derivative 33.2a under acidic conditions provided the carboxylic acid derivative compound 33E and compound 33L. Compound 33E and compound 33L were readily separated by column chromatography.
  • The synthesis of compounds 34A-34P is outlined in Scheme 34. Suzuki type coupling of the boronate derivative 32.1 with various aryl bromide derivatives 34.1 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compounds 34.2 which were converted to the final products compounds 34A-34P under acidic conditions. The derivatives 34.1 used in the Suzuki coupling step were prepared as follow. Coupling of 6-bromopyridine-3-carboxylic acid (34.3), 5-bromothiophene-2-carboxylic acid (34.4), 4-bromothiophene-2-carboxylic acid (34.7) or 5-bromofuran-2-carboxylic acid (34.6) with diethylamine (1.12) or diisopropylamine (3.4o) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the diethylaminocarbonyl derivatives 34.1a-d, f-i. Coupling of 5-bromothiophene-2-sulfonyl chloride (34.5) with diethylamine (1.12) in acetonitrile in the presence of triethylamine provided the sulfonamide 34.1e. Coupling of the commercially available carboxylic acid derivatives 34.8a-34.8f and 34.9 with diethylamine (1.12) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the corresponding diethylaminocarbonyl derivatives 34.1j-34.1o and 34.1p.
  • The synthesis of compounds 35A and 35B is outlined in Scheme 35. Iodination of 3-hydroxybenzoic acid (35.1) afforded 3-hydroxy-4-iodobenzoic acid (35.2), which was converted to the methyl ester 35.3 under standard esterification conditions. Alkylation of the phenolic derivative 35.3 with methyl iodide (2.8c) in acetone in the presence of potassium carbonate afforded the methyl ether 35.4, which was converted to the carboxylic acid 35.5 in the presence of lithium hydroxide. Coupling of the carboxylic acid derivatives 35.5 with diethylamine (1.12) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the corresponding diethylaminocarbonyl derivative 35.6. Demethylation of 35.6 using boron tribromide afforded the phenolic derivative 35.7 which was converted to the methyloxymethyl (MOM) ether derivative 35.8 using chloro(methoxy)methane 11.3. Suzuki type coupling of the boronate derivative 32.1 with 35.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 35.9 which was converted to the final product compound 35A under acidic conditions. Suzuki type coupling of the boronate derivative 32.1 with 35.8 in ethylene glycol dimethyl ether in the presence of palladium, 10 wt. % (dry basis) on activated carbon, lithium chloride, and an aqueous solution of sodium carbonate afforded compound 35.10 which was converted to the final product compound 35B under acidic conditions.
  • The synthesis of compounds 36A and 36B is outlined in Scheme 36. Coupling of 4-bromo-2-hydroxybenzoic acid (36.3) [obtained from 4-amino-2-hydroxybenzoic acid (36.1) under Sandmeyer conditions] with diethylamine (1.12) using 0-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU) as coupling agent afforded the corresponding diethylaminocarbonyl derivative 36.4. Suzuki type coupling of the boronate derivative 32.1 with 36.4 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 36.5 which was converted to the final product (compound 36A) under acidic conditions. Compound 36B was obtained in 7 steps from 2-(3-methoxyphenyl)ethanamine (36.6). Coupling of 36.6 with ethyl chloroformate (36.7) afforded the ethyl carbamate derivative 36.8 which was cyclized to 3,4-dihydro-6-methoxyisoquinolin-1-(2H)-one (36.9) in the presence of polyphosphoric acid. Alkylation of 36.9 with ethyl iodide (36.10) in tetrahydrofuran in the presence of sodium hydride, afforded the methyl ether 36.11, which was converted to the phenolic derivative 36.12 by treatment with boron tribromide. Condensation of 36.12 with trifluoromethanesulfonic anhydride (36.13) in dichloromethane in the presence of pyridine afforded the triflate derivative 36.14. Suzuki type coupling of the boronate derivative 32.1 with 36.14 in N,N-dimethylformamide in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, abbreviated as [Pd(dppf)Cl2.CH2Cl2], and potassium acetate afforded compound 36.15 which was converted to the final product (compound 36B) under acidic conditions.
  • The synthesis of compounds 37A-37B is outlined in Scheme 37. The 2′-hydroxyacetophenone 1.1a was condensed with 1-benzyl-3-methylpiperidin-4-one (37.1) (racemic mixture) in refluxing methanol in the presence of pyrrolidine to provide the racemic ketones 37.2 and 37.3. The diastereoisomers 37.2 and 37.3 were separated by column chromatography. Palladium catalyzed hydrogenation of 37.2 afforded the piperidine derivative 37.4, which was converted to 37.5 by treatment with tert-butyloxycarbonyl anhydride (4.7). Conversion of the ketone 37.5 to the enol triflate derivative 37.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 37.6 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the Boc derivative 37.7, which was converted to the final product compound 37A (racemic mixture) under acidic conditions. Similarly, palladium catalyzed hydrogenation of 37.3 afforded the piperidine derivative 37.8, which was converted to 37.9 by treatment with tert-butyloxycarbonyl anhydride (4.7). Conversion of the ketone 37.9 to the enol triflate derivative 37.10 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 37.10 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the Boc derivative 37.11, which was converted to the final product compound 37B (racemic mixture) under acidic conditions.
  • The synthesis of compounds 38A-38D is outlined in Scheme 38. Condensation of benzyl 4-oxopiperidine-1-carboxylate (19.1) with 2,2-dimethyl-1,3-dioxane-4,6-dione (Meldrum's acid; 38.1) in the presence of pyridine and piperidine gave the derivative 38.2. Compound 38.2 was subjected to conjugate addition by reaction with organocuprate reagents derived from 4-fluorobenzyl magnesium chloride (38.3) and copper (I) iodide to yield, upon work-up, the sodium salt 38.4. Hydrolysis and decarboxylation of the conjugate addition product 38.4 proceeded by heating a solution of 38.4 in N,N-dimethylformamide in the presence of water. Treatment of the corresponding carboxylic acid derivative 38.5 with oxalyl chloride followed by reaction of the resulting acyl chloride with aluminum chloride yielded the corresponding spiro piperidine derivative which was further protected as its tert-butyloxycarbonyl (Boc) derivative 38.6 by treatment with tert-butyloxycarbonyl anhydride (4.7). Conversion of the ketone 38.6 to the enol triflate derivative 38.7 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 38.7 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}, and an aqueous solution of potassium carbonate afforded the derivative 38.8 which was converted to the compound 38A under acidic conditions. The compound 38B (racemic mixture) was obtained by hydrogenation of the unsaturated derivative 38A in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon. Conversion of the enol triflate 38.7 to the corresponding boron derivative 38.9 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}. Suzuki type coupling of the boron derivative 38.9 with the aryl iodide derivative 35.8 in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}, and an aqueous solution of potassium carbonate afforded the derivative 38.10 which was converted to the compound 38C under acidic conditions. Hydrogenation of the unsaturated derivatives 38.10 in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon provided the compound 38.11, which was converted to the compound 38D under acidic conditions.
  • The synthesis of compounds 39A-39G is outlined in Scheme 39. Compound 38.2 was subjected to conjugate addition by reaction with organocuprate reagents derived from benzyl magnesium chloride (28.3a) and copper (I) iodide to yield, upon work-up, the sodium salt 39.1. Hydrolysis and decarboxylation of the conjugate addition product 39.1 proceeded by heating a solution of 39.1 in N,N-dimethylformamide in the presence of water. Treatment of the corresponding carboxylic acid derivative 39.2 with oxalyl chloride followed by reaction of the resulting acyl chloride with aluminum chloride yielded the corresponding spiro piperidine derivative which was further protected as its CBz derivative 28.6a (Rv═H) by treatment with benzylchloroformate. Conversion of the ketone 28.6a to the enol triflate derivative 28.7a was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 28.7a 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 28.12 which was converted to the compound 39A by hydrogenation in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon. Treatment of compound 39A with tert-butyloxycarbonyl anhydride (4.7) provided the Boc derivative 39.3. Chiral separation of the enantiomers derived from 39.3 provided compounds 39.4 and 39.5. The enantiomers 39.4 and 39.5 were converted to compounds 39B and 39C, respectively under acidic conditions. Suzuki type coupling of the enol triflate derivatives 28.7a with N,N-diethyl-3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzamide 39.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 39.7 which was converted to the compound 39D by treatment with iodotrimethylsilane followed by acidic cleavage of the MOM protecting group. Hydrogenation of 39.7 in methanol, in the presence of palladium, 10 wt. % (dry basis) on activated carbon, followed by acidic cleavage of the MOM protecting group, afforded compound 39E. Treatment of compound 39E with tert-butyloxycarbonyl anhydride (4.7) provided the Boc derivative 39.8. Chiral separation of the enantiomers derived from 39.8 provided compounds 39.9 and 39.10. The enantiomers 39.9 and 39.10 were converted to compounds 39F and 39G, respectively under acidic conditions.
  • The synthesis of compounds 40A-40C is outlined in Scheme 40. Compound 38.2 was subjected to conjugate addition by reaction with organocuprate reagents derived from 4-methoxybenzyl magnesium chloride (28.3b) and copper (I) iodide to yield, upon work-up, the sodium salt 40.1. Hydrolysis and decarboxylation of the conjugate addition product 40.1 proceeded by heating a solution of 40.1 in N,N-dimethylformamide in the presence of water. Treatment of the corresponding carboxylic acid derivative 28.11 with oxalyl chloride followed by reaction of the resulting acyl chloride with aluminum chloride yielded the corresponding spiro piperidine derivative which was further protected as its CBz derivative 28.6b (Rv═OCH3) by treatment with benzylchloroformate. Conversion of the ketone 28.6b to the enol triflate derivative 28.7b was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 28.7b with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 40.2 which was converted to the compound 40A by treatment with iodotrimethylsilane. Hydrogenation of 40.2 in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon afforded compound 40B (racemic mixture). Suzuki type coupling of the enol triflate derivatives 28.7b with N,N-diethyl-3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzamide 39.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded the derivative 40.3 which was converted to the compound 40C by treatment with iodotrimethylsilane.
  • The synthesis of compounds 41A-41E is outlined in Scheme 41. Coupling of the carboxylic acid 13.3 with N-ethyl-2-methoxyethanamine (41.1) using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the tertiary aminocarbonyl derivative 41.2, which was converted to compound 41A under acidic conditions. Coupling of the carboxylic acid 13.3 with N-ethyl-N,N-dimethylethane-1,2-diamine (41.3) using TBTU as coupling agent afforded the tertiary aminocarbonyl derivative 41.4, which was converted to compound 41B under acidic conditions. Coupling of the carboxylic acid 13.3 with N-(3-(ethylamino)propyl)-2,2,2-trifluoroacetamide (41.7) (obtained by treatment of N-ethylpropane-1,3-diamine 41.5 with ethyl 2,2,2-trifluoroacetate 41.6) using TBTU as coupling agent afforded the tertiary aminocarbonyl derivative 41.8, which was converted to 41.9 under basic conditions. Treatment of 41.9 with 2-nitrobenzene-1-sulfonyl chloride (41.10) afforded the sulfonamide derivative 41.11, which was converted to 41.12 by treatment with methyl iodide. Thiophenol-mediated deprotection of 41.12 afforded the derivative 41.13, which was converted to the final compound 41C under acidic conditions. Coupling of the carboxylic acid 13.3 with N,N-dimethylpropane-1,3-diamine (41.14) using TBTU as coupling agent afforded the tertiary aminocarbonyl derivative 41.15, which was converted to compound 41D under acidic conditions. Coupling of the carboxylic acid 13.3 with N-(2-(ethylamino)ethyl)-2,2,2-trifluoroacetamide (41.17) (obtained by treatment of N-ethylethane-1,2-diamine 41.16 with ethyl 2,2,2-trifluoroacetate 41.6) using TBTU as coupling agent afforded the tertiary aminocarbonyl derivative 41.18, which was converted to 41.19 under basic conditions. Treatment of compound 41.19 with tert-butyloxycarbonyl anhydride (4.7) provided the bis-Boc derivative 41.20. Treatment of 41.20 with methyl iodide in the presence of sodium hydride afforded the derivative 41.21, which was converted to the compound 41E under acidic conditions.
  • The synthesis of compounds 42A-421 is outlined in Scheme 42. Suzuki type coupling of the enol triflate derivatives 21.6 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 42.1 (racemic mixture) which was converted to compound 42C under acidic conditions. Chiral separation of the enantiomers derived from 42.1 provided compounds 42.2 and 42.3. The enantiomers 42.2 and 42.3 were converted to compounds 42A and 42B, respectively under acidic conditions. Conversion of the enol triflate 21.6 to the corresponding boron derivative 42.4 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}. Suzuki type coupling of the boron derivative 42.4 with the aryl bromide 34.1a in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 42.5 (racemic mixture), which was converted to compound 42F under acidic conditions. Chiral separation of the enantiomers derived from 42.5 provided compounds 42.6 and 42.7. The enantiomers 42.6 and 42.7 were converted to compounds 42D and 42E, respectively under acidic conditions. Suzuki type coupling of the boron derivative 42.4 with the aryl iodide 35.8 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 42.8 (racemic mixture), which was converted to compound 42I under acidic conditions. Chiral separation of the enantiomers derived from 42.8 provided compounds 42.9 and 42.10. The enantiomers 42.9 and 42.10 were converted to compounds 42G and 42H, respectively under acidic conditions.
  • The synthesis of compounds 43A-43F is outlined in Scheme 43. The 2′-6′-dihydroxyacetophenone derivative 11.1 was condensed with tert-butyl 4-oxoazepane-1-carboxylate 21.4 in refluxing methanol in the presence of pyrrolidine to provide the derivative 43.1 which was converted to the methoxymethyl (MOM) ether derivative 43.2 using chloro(methoxy)methane (11.3). Conversion of the ketone 43.2 to the enol triflate derivative 43.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 43.3 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 43.4 (racemic mixture). Removal of the MOM and the Boc protecting groups of 43.4 in methanol at room temperature in the presence of hydrochloric acid (anhydrous solution in dichloromethane) afforded compound 43C (racemic mixture). Chiral separation of the enantiomers derived from 43.4 provided compounds 43.5 and 43.6. The enantiomers 43.5 and 43.6 were converted to compounds 43A and 43B, respectively under acidic conditions. Suzuki type coupling of the enol triflate derivative 43.3 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 43.7 (racemic mixture). Removal of the MOM and the Boc protecting groups of 43.7 in methanol at room temperature in the presence of hydrochloric acid (anhydrous solution in dichloromethane) afforded compound 43F (racemic mixture). Chiral separation of the enantiomers derived from 43.7 provided compounds 43.8 and 43.9. The enantiomers 43.8 and 43.9 were converted to compounds 43D and 43E, respectively under acidic conditions.
  • The synthesis of compounds 44A-44F is outlined in Scheme 44. The 5′-fluoro-2′-hydroxy-acetophenone derivative 1.1d was condensed with tert-butyl 4-oxoazepane-1-carboxylate 21.4 in refluxing methanol in the presence of pyrrolidine to provide the derivative 44.1. Conversion of the ketone 44.1 to the enol triflate derivative 44.2 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 44.2 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 44.3 (racemic mixture) which was converted to compound 44C under acidic conditions. Chiral separation of the enantiomers derived from 44.3 provided compounds 44.4 and 44.5. The enantiomers 44.4 and 44.5 were converted to compounds 44A and 44B, respectively under acidic conditions. Conversion of the enol triflate 44.2 to the corresponding boron derivative 44.6 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}. Suzuki type coupling of the boron derivative 44.6 with the aryl bromide 34.1a in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 44.7 (racemic mixture), which was converted to compound 44F under acidic conditions. Chiral separation of the enantiomers derived from 44.7 provided compounds 44.8 and 44.9. The enantiomers 44.8 and 44.9 were converted to compounds 44D and 44E, respectively under acidic conditions.
  • The synthesis of compounds 45A-45F is outlined in Scheme 45. Suzuki type coupling of the boron derivative 44.6 with the aryl iodide 35.8 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 45.1 (racemic mixture), which was converted to compound 45C under acidic conditions. Chiral separation of the enantiomers derived from 45.1 provided compounds 45.2 and 45.3. The enantiomers 45.2 and 45.3 were converted to compounds 45A and 45B, respectively under acidic conditions. The 2′-5′-dihydroxyacetophenone derivative 2.1 was condensed with tert-butyl 4-oxoazepane-1-carboxylate 21.4 in refluxing methanol in the presence of pyrrolidine to provide the derivative 45.4 which was converted to the silyl ether derivative 45.5 using tert-butyldimethylsilyl chloride 2.3. Conversion of the ketone 45.5 to the enol triflate derivative 45.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 45.6 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 45.7 (racemic mixture), which was converted to compound 45F under acidic conditions. Chiral separation of the enantiomers derived from 45.7 provided compounds 45.8 and 45.9. The enantiomers 45.8 and 45.9 were converted to compounds 45D and 45E, respectively under acidic conditions.
  • The synthesis of compounds 46A-46C is outlined in Scheme 46. Suzuki type coupling of the enol triflate derivative 45.6 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 46.1 (racemic mixture), which was converted to compound 46C under acidic conditions. Treatment of 46.1 with chloro(methoxy)methane (11.3) provided the methoxymethyl (MOM) ether derivative 46.2 (racemic mixture). Chiral separation of the enantiomers derived from 46.2 provided compounds 46.3 and 46.4. The enantiomers 46.3 and 46.4 were converted to compounds 46A and 46B, respectively under acidic conditions.
  • The synthesis of compounds 47A-47F is outlined in Scheme 47. Suzuki type coupling of the enol triflate derivative 23.3a with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 47.1 (racemic mixture) which was converted to compound 47A under acidic conditions. Conversion of the enol triflate 23.3a to the corresponding boron derivative 47.2 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}. Suzuki type coupling of the boron derivative 47.2 with the aryl iodide 35.8 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 47.3 (racemic mixture), which was converted to compound 47B under acidic conditions. The 2′-6′-dihydroxyacetophenone derivative 11.1 was condensed with tert-butyl 3-oxopyrrolidine-1-carboxylate 23.1a in refluxing methanol in the presence of pyrrolidine to provide the derivative 47.4 which was converted to the methoxymethyl (MOM) ether derivative 47.5 using chloro(methoxy)methane (11.3). Conversion of the ketone 47.5 to the enol triflate derivative 47.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 47.6 with compound 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 47.7 (racemic mixture). Removal of the MOM and the Boc protecting groups of 47.7 in methanol at room temperature in the presence of hydrochloric acid afforded compound 47C (racemic mixture). Suzuki type coupling of the enol triflate derivative 47.6 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 47.8 (racemic mixture). Removal of the MOM and the Boc protecting groups of 47.8 in methanol at room temperature in the presence of hydrochloric acid afforded compound 47D (racemic mixture). The 2′-5′-dihydroxyacetophenone derivative 2.1 was condensed with tert-butyl 3-oxopyrrolidine-1-carboxylate 23.1a in refluxing methanol in the presence of pyrrolidine to provide the derivative 47.9 which was converted to the silyl ether derivative 47.10 using tert-butyldimethylsilyl chloride 2.3. Conversion of the ketone 47.10 to the enol triflate derivative 47.11 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 47.11 with compound 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 47.12 (racemic mixture), which was converted to compound 47E under acidic conditions. Suzuki type coupling of the enol triflate derivative 47.11 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 47.13 (racemic mixture), which was converted to compound 47F under acidic conditions.
  • The synthesis of compounds 48A-48F is outlined in Scheme 48. Suzuki type coupling of the enol triflate derivative 1.5f with 4-cyanophenylboronic acid (14.1) in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, and an aqueous solution of potassium carbonate afforded the cyanide 48.1 which was converted to the tetrazole 48.2 using sodium azide (14.3) and zinc bromide in a solution isopropanol/water. Alkylation of 48.2 with methyl iodide (2.8c) in N,N-dimethylformamide in the presence of triethylamine afforded the two regioisomers 48.3 (major isomer) and 48.4 (minor isomer) separated by silica gel column chromatography. The Boc protecting group of 48.2, 48.3, and 48.4 was removed using hydrochloric acid to generate the compounds 48A-48C. Conversion of the enol triflate 1.5f to the corresponding boron derivative 48.5 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}. Suzuki type coupling of the boron derivative 48.5 with the aryl bromide 34.1a in dioxane in the presence of tetrakis triphenylphosphine palladium (0), potassium bromide and potassium phosphate afforded compound 48.8 (racemic mixture), which was converted to compound 48F under acidic conditions. Suzuki type coupling of the boron derivative 48.5 with the bromothiophene derivatives 34.1c and 34.1d in dioxane in the presence of and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct, and an aqueous solution o potassium carbonate afforded compound 48.6 and 48.7 respectively. Compounds 48.6 and 48.7 were converted to compound 48D and 48E, respectively, under acidic conditions.
  • The synthesis of compounds 49A-49D is outlined in Scheme 49. The 6′-fluoro-2′-hydroxy-acetophenone derivative 49.1 was condensed with 1-Boc-4-piperidone 1.2 in methanol at 0° C. in the presence of pyrrolidine to provide the derivative 49.2. Conversion of the ketone 49.2 to the enol triflate derivative 49.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 49.3 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 49.4 which was converted to compound 49A under acidic conditions. Treatment of 49.2 with pyrrolidine afforded compound 49.5. Conversion of the ketone 49.5 to the enol triflate derivative 49.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 49.6 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride, and an aqueous solution of sodium carbonate afforded compound 49.7 which was converted to compound 49B under acidic conditions. Conversion of the enol triflate 1.5d to the corresponding boron derivative 49.8 was achieved using 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane 1.14 and dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct {[Pd(dppf)Cl2.CH2Cl2]}. Suzuki type coupling of the boron derivative 49.8 with the aryl iodide 35.8 in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct and an aqueous solution of potassium carbonate afforded compound 49.9, which was converted to compound 49C under acidic conditions. Hydrogenation of the unsaturated derivative 49C in methanol in the presence of palladium, 10 wt. % (dry basis) on activated carbon provided the compound 49D (racemic mixture).
  • The synthesis of compounds 50A-50D is outlined in Scheme 50. Suzuki type coupling of the enol triflate derivative 2.5 with phenyl boronic acid 31.1g in ethylene glycol dimethyl ether, in the presence of palladium, 10 wt. % (dry basis) on activated carbon, lithium chloride, and an aqueous solution of sodium carbonate afforded compound 50.1. Conversion of the phenol 50.1 to the triflate derivative 50.2 was achieved using the triflating reagent N-phenylbis(trifluoromethanesulphonimide) 1.4. Palladium catalyzed carbonylation of 50.2, conducted in a mixture of dimethylsulfoxide/methanol using palladium (II) acetate, 1,1′-bis(diphenylphosphino)ferrocene (dppf) and carbon monoxide, provided the methyl ester 50.3 which was hydrolyzed under basic conditions to give the carboxylic acid derivatives 50.4. Coupling of the carboxylic acid 50.4 with diethylamine (1.12) using TBTU as coupling agent afforded the tertiary amide 50.5. Treatment of the Boc derivatives 50.3, 50.4, 50.5 and 50.1 with hydrochloric acid provided the final compounds 50A-50D, respectively.
  • The synthesis of compounds 51A-51C is outlined in Scheme 51. The 2′-hydroxyacetophenone 1.1a was condensed with tert-butyl 2-methyl-4-oxopiperidine-1-carboxylate (51.1) (racemic mixture) in refluxing methanol in the presence of pyrrolidine to provide the racemic ketone 51.2. Conversion of the ketone 51.2 to the enol triflate derivative 51.3 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivative 51.3 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), potassium bromide, and potassium phosphate afforded the Boc derivative 51.4 (product eluted as a single peak), which was converted to compound 51C under acidic conditions. Chiral separation of the intermediate 51.4 provided resolved products 51.5 and 51.6. The resolved products 51.5 and 51.6 were converted to compounds 51A and 51B, respectively under acidic conditions.
  • The synthesis of compounds 52A-52F is outlined in Scheme 52. Treatment of compound 21C with trifluoroacetic anhydride (4.1) in tetrahydrofuran in the presence of triethylamine provided the trifluoroacetamide derivative 52.1 which was converted to the sulfonyl chloride 52.2 using sulfur trioxide N,N-dimethylformamide complex (4.3) as sulfating agent. Condensation of hydrazine hydrate (5.1) with the sulfonyl chloride derivative 52.2 provided the sulfonyl hydrazide 52.3, which was converted to the sulfone 52.4 by treatment with methyl iodide (2.8c) in the presence of sodium acetate. Deprotection of the trifluoroacetamide protecting group of 52.4 under basic conditions (potassium carbonate, methanol, water) provided the methyl sulfonyl analog (compound 52A) [Note: compound 52A (derived from compound 21C) and 22E (derived from compound 21B) are enantiomeric with respect to one another]. Palladium catalyzed hydrogenation of compound 22E affords compounds 52B and 52C, respectively. Palladium catalyzed hydrogenation of compounds 35B, 1Q, and 1F affords compounds 52D, 52E, and 52F, respectively.
  • The synthesis of compounds 53A-53F is outlined in Scheme 53. Treatment of compounds 48.2 and 48.3 with hydrobromic acid provided the phenolic derivatives 53A and 53B, respectively. Suzuki type coupling of the enol triflate derivative 11.5 with 4-(carboxy)phenylboronic acid (53.1) in dioxane in the presence of tetrakis triphenylphosphine palladium (0), and an aqueous solution of potassium carbonate afforded the carboxylic acid derivative 53.2 which was converted to 53D under acidic conditions. The carboxylic acid derivative 53D was converted to its methyl ester analog 53C under classical esterification conditions, i.e. in the presence of concentrated hydrochloric acid and methanol. Coupling of the carboxylic acid 53.2 with ammonium chloride (3.4a) in acetonitrile, in the presence of N,N-diisopropylethylamine (Hunig's base), using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent affords the primary aminocarbonyl derivative 53.3. Coupling of the carboxylic acid 53.2 with ethylamine (3.4c) in acetonitrile, in the presence of N,N-diisopropylethylamine (Hunig's base), using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) as coupling agent afforded the secondary aminocarbonyl derivative 53.4. Treatment of the derivative 53.4 with hydrochloric acid provided the final compounds 53F. Treatment of the derivative 53.3 with hydrochloric acid provides the final compound 53E.
  • The synthesis of compounds 54A and 54B is outlined in Scheme 54. Condensation of benzyl 4-oxoazepane-1-carboxylate (54.1) (obtained by treatment of azepan-4-one 21.3 with benzylchloroformate) with 2,2-dimethyl-1,3-dioxane-4,6-dione (Meldrum's acid; 38.1) in the presence of pyridine and piperidine gave the derivative 54.2. The compound 54.2 was subjected to conjugate addition by reaction with organo cuprate reagents derived from benzyl magnesium chloride (28.3a) and copper (I) iodide to yield, upon work-up, the sodium salt 54.3. Hydrolysis and decarboxylation of the conjugate addition product 54.3 proceeded by heating a solution of 54.3 in N,N-dimethylformamide in the presence of water. Treatment of the corresponding carboxylic acid derivative 54.4 with oxalyl chloride followed by reaction of the resulting acyl chloride with aluminum chloride yielded the corresponding spiro piperidine derivative which was further protected as its tert-butyloxycarbonyl (Boc) derivative 54.5 by treatment with tert-butyloxycarbonyl anhydride (4.7). Conversion of the ketone 54.5 to the enol triflate derivative 54.6 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 54.6 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in dioxane in the presence of tetrakis triphenylphosphine palladium (0) and an aqueous solution of potassium carbonate afforded compound 54.7, which was converted to compound 54A under acidic conditions. Suzuki type coupling of the enol triflate derivative 54.6 with 2-(N,N-diethylaminocarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan-2-yl)pyridine 1.7 in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct and an aqueous solution of potassium carbonate afforded compound 54.8, which was converted to compound 54B under acidic conditions.
  • The synthesis of compounds 55A-C is outlined in Scheme 55. The compound 38.2 was subjected to conjugate addition by reaction with organo cuprate reagents derived from 3,5-dimethoxybenzyl magnesium bromide (55.1) and copper (I) iodide to yield, upon work-up, the sodium salt 55.2. Hydrolysis and decarboxylation of the conjugate addition product 55.2 proceeded by heating a solution of 55.2 in N,N-dimethylformamide in the presence of water. Treatment of the corresponding carboxylic acid derivative 55.3 with oxalyl chloride followed by reaction of the resulting acyl chloride with aluminum chloride yielded the corresponding spiro piperidine derivative, which was further protected as its CBz derivative 55.4 by treatment with benzylchloroformate. Conversion of the ketone 55.4 to the enol triflate derivative 55.5 was achieved using N-phenylbis(trifluoromethanesulphonimide) 1.4 as triflating reagent. Suzuki type coupling of the enol triflate derivatives 55.5 with 4-(N,N-diethylaminocarbonyl)phenyl boronic acid 1.6 in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0), lithium chloride and an aqueous solution of sodium carbonate afforded compound 55.6 which was converted to either the compound 55C by treatment with iodotrimethylsilane or the compound 55A by treatment with boron tribromide. Treatment of 55A with tert-butyloxycarbonyl anhydride (4.7) provided the corresponding Boc derivative 55.7. Condensation of 55.7 with N-phenylbis(trifluoromethanesulphonimide) 1.4 provided the corresponding monotriflate derivative 55.8 which was converted to compound 55.9 by treatment with triethylsilane in the presence of 1,3-bis(diphenylphosphino)propane (dppp) and palladium (II) acetate. Acidic deprotection of the Boc-protecting group of 55.9 provided the compound 55B.
  • The synthesis of compounds 56A-56D is outlined in Scheme 56. Boron tribromide-mediated demethylation of 28.8b provided the phenolic derivative 56A, which was converted to 56B under hydrogenation conditions. Similarly, boron tribromide-mediated demethylation of 40.3 provided the phenolic derivative 56C, which was converted to 56D under hydrogenation conditions.
  • The synthesis of compounds 57A-57D is outlined in Scheme 57. Treatment of compound 31J with trifluoroacetic anhydride (4.1) in tetrahydrofuran in the presence of triethylamine provided the trifluoroacetamide derivative 57.1 which was converted to the sulfonyl chloride 57.2 using sulfur trioxide N,N-dimethylformamide complex (4.3) as sulfonating agent. Condensation of 57.2 with methylamine (3.4b) and dimethylamine (3.4j) affords the corresponding sulfonamide derivatives 57.3 which are converted to compounds 57A, 57B under basic conditions. Condensation of 57.2 with ammonium hydroxide yields the compound 57C. Condensation of hydrazine hydrate (5.1) with the sulfonyl chloride derivative 57.2 provided the sulfonyl hydrazide 57.4, which was converted to the sulfone 57.5 by treatment with methyl iodide (2.8c) in the presence of sodium acetate. Deprotection of the trifluoroacetamide protecting group of 57.5 under basic conditions (potassium carbonate, methanol, water) provided the compound 57D.
  • The synthesis of compounds 58A-58D is outlined in Scheme 58. Suzuki type coupling of the enol triflate derivative 2.5 with either 3-(1,3,2-dioxaborinan-2-yl)pyridine (3.6a) or benzo[b]thiophen-2-ylboronic acid (31.1n) in dioxane in the presence of dichloro[1,1′-bis(diphenylphosphino)ferrocene]palladium(H) dichloromethane adduct (abbreviated as [Pd(dppf)Cl2.CH2Cl2]) and an aqueous solution of potassium carbonate afforded compounds 58.1. The derivatives 58.1a and 58.1b were then converted to the final products 58A and 58B, respectively, under acidic conditions. Suzuki type coupling of the enol triflate derivative 2.5 with phenyl boronic acid (31.1g) in ethylene glycol dimethyl ether in the presence of tetrakis triphenylphosphine palladium (0) and an aqueous solution of potassium carbonate afforded compound 58.2. Removal of the silyl protecting group of 58.2 using a solution of tetrabutylammonium fluoride (TBAF) in tetrahydrofuran gave the phenolic derivatives 58.3. Preparation of each of the ether derivatives 58.5 from the phenol 58.3 was achieved by alkylation reaction using the appropriate alkyl iodide or alky chloride (2.8c, 58.4) reagent. Treatment of the Boc derivatives 58.5 with hydrochloric acid provided the final compounds 58C-D.
  • The synthesis of compounds 59A-59L is outlined in Scheme 59. The alkyl bromide derivatives used as starting materials for the synthesis of compounds 59A-59L were either commercially available (59.2a, 59.2b) or prepared from the corresponding alcohols (preparation of 59.2c and 59.2d). Treatment of the amines 59.2 with ethyl 2,2,2-trifluoroacetate (59.3) in methanol in the presence of triethylamine provided the trifluoroacetamide derivatives 59.4. Alkylation of 5-(4-bromophenyl)-2H-tetrazole (59.6) (obtained by treatment of 4-bromobenzonitrile (59.5) with sodium azide and ammonium chloride in N,N-dimethylformamide) with the alkyl bromide derivatives 59.4, provided the derivatives 59.7. Suzuki type coupling of the boronate derivative 32.1 with the aryl bromide derivatives 59.7 in dioxane in the presence of tetrakis triphenylphosphine palladium (0) and an aqueous solution of potassium carbonate afforded the compounds 59.8 which were converted to the derivatives 59.9 under basic conditions. The Boc protecting group of 59.9 was removed using hydrochloric acid to generate the compounds 59A-59D. Treatment of the amines 59.9 with acetic anhydride in dichloromethane in the presence of triethylamine provided the acetamides 59.10, which were converted to the final products 59E-59H under acidic conditions. Treatment of the amines 59.9 with methane sulfonyl chloride in dichloromethane in the presence of triethylamine provided the sulfonamides 59.11, which were converted to the final products 59I-59L under acidic conditions.
  • The synthesis of compounds 60A-60C is outlined in Scheme 60. Treatment of the amine 60.1 with ethyl 2,2,2-trifluoroacetate (59.3) in methanol in the presence of triethylamine provided the trifluoroacetamide derivative 60.2. Treatment of the alcohol 60.2 with methane sulfonyl chloride in dichloromethane in the presence of triethylamine provided the mesylate derivative 60.3. Alkylation of 544-bromophenyl)-2H-tetrazole (59.6) with the mesylate derivative 60.3, provided the derivative 60.4. Suzuki type coupling of the boronate derivative 32.1 with the aryl bromide derivative 60.4 in dioxane in the presence of tetrakis triphenylphosphine palladium (0) and an aqueous solution of potassium carbonate afforded the compound 60.5 which was converted to the derivative 60.6 under basic conditions. The Boc protecting group of 60.6 was removed using hydrochloric acid to generate the compound 60A. Treatment of the amine 60.6 with acetic anhydride in dichloromethane in the presence of triethylamine provided the acetamide 60.7, which was converted to the final products 60B under acidic conditions. Treatment of the amine 60.6 with methane sulfonyl chloride in dichloromethane in the presence of triethylamine provided the sulfonamide 60.8, which was converted to the final products 60C under acidic conditions.
  • The synthesis of compounds 61A-61B is outlined in Scheme 61. Chiral separation of the enantiomers derived from 61.1 (obtained by hydrogenation of 49.9) provided compounds 61.2a and 61.2b, which were converted to the final products 61A and 61B, respectively, under acidic conditions. Condensation of compound 21C with 6-bromo-2-naphthoic acid (61.3) (used as chiral resolving agent) in acetonitrile in the presence of TBTU and diisopropylethylamine (Hunig's base) provided the amide derivative 61.4. The absolute configuration of 61.4 was determined by X-ray crystallography, therefore establishing the absolute configuration of compound 21C, and therefore by inference, its enantiomer, compound 21B (see also Scheme 21).
  • In some instances, the compounds of the invention have the potential for chirality but were prepared in racemic form. As one skilled in the art would readily recognize, the racemic mixtures of intermediates or final products initially prepared as their racemates may be partially or completely resolved into any, some, or all of the enantiomers contained in the racemate as described herein, for example in the separation of intermediates leading to 39F and 39G. As such, racemic mixtures, mixtures enriched in or more stereoisomers, and pure enantiomers are considered to be within the ambit of the present invention.
  • Figure US20110218186A1-20110908-C00109
    Figure US20110218186A1-20110908-C00110
  • Figure US20110218186A1-20110908-C00111
    Figure US20110218186A1-20110908-C00112
  • Figure US20110218186A1-20110908-C00113
    Figure US20110218186A1-20110908-C00114
  • Figure US20110218186A1-20110908-C00115
    Figure US20110218186A1-20110908-C00116
    Figure US20110218186A1-20110908-C00117
  • Figure US20110218186A1-20110908-C00118
  • Figure US20110218186A1-20110908-C00119
    Figure US20110218186A1-20110908-C00120
  • Figure US20110218186A1-20110908-C00121
    Figure US20110218186A1-20110908-C00122
    Figure US20110218186A1-20110908-C00123
  • Figure US20110218186A1-20110908-C00124
    Figure US20110218186A1-20110908-C00125
  • Figure US20110218186A1-20110908-C00126
    Figure US20110218186A1-20110908-C00127
  • Figure US20110218186A1-20110908-C00128
    Figure US20110218186A1-20110908-C00129
  • Figure US20110218186A1-20110908-C00130
    Figure US20110218186A1-20110908-C00131
    Figure US20110218186A1-20110908-C00132
  • Figure US20110218186A1-20110908-C00133
    Figure US20110218186A1-20110908-C00134
    Figure US20110218186A1-20110908-C00135
  • Figure US20110218186A1-20110908-C00136
    Figure US20110218186A1-20110908-C00137
  • Figure US20110218186A1-20110908-C00138
  • Figure US20110218186A1-20110908-C00139
  • Figure US20110218186A1-20110908-C00140
  • Figure US20110218186A1-20110908-C00141
  • Figure US20110218186A1-20110908-C00142
    Figure US20110218186A1-20110908-C00143
  • Figure US20110218186A1-20110908-C00144
    Figure US20110218186A1-20110908-C00145
  • Figure US20110218186A1-20110908-C00146
  • Figure US20110218186A1-20110908-C00147
    Figure US20110218186A1-20110908-C00148
    Figure US20110218186A1-20110908-C00149
  • Figure US20110218186A1-20110908-C00150
    Figure US20110218186A1-20110908-C00151
  • Figure US20110218186A1-20110908-C00152
    Figure US20110218186A1-20110908-C00153
    Figure US20110218186A1-20110908-C00154
  • Figure US20110218186A1-20110908-C00155
  • Figure US20110218186A1-20110908-C00156
  • Figure US20110218186A1-20110908-C00157
    Figure US20110218186A1-20110908-C00158
  • Figure US20110218186A1-20110908-C00159
    Figure US20110218186A1-20110908-C00160
    Figure US20110218186A1-20110908-C00161
  • Figure US20110218186A1-20110908-C00162
    Figure US20110218186A1-20110908-C00163
    Figure US20110218186A1-20110908-C00164
  • Figure US20110218186A1-20110908-C00165
    Figure US20110218186A1-20110908-C00166
  • Figure US20110218186A1-20110908-C00167
    Figure US20110218186A1-20110908-C00168
    Figure US20110218186A1-20110908-C00169
  • Figure US20110218186A1-20110908-C00170
    Figure US20110218186A1-20110908-C00171
    Figure US20110218186A1-20110908-C00172
  • Figure US20110218186A1-20110908-C00173
    Figure US20110218186A1-20110908-C00174
    Figure US20110218186A1-20110908-C00175
    Figure US20110218186A1-20110908-C00176
    Figure US20110218186A1-20110908-C00177
  • Figure US20110218186A1-20110908-C00178
    Figure US20110218186A1-20110908-C00179
  • Figure US20110218186A1-20110908-C00180
    Figure US20110218186A1-20110908-C00181
    Figure US20110218186A1-20110908-C00182
  • Figure US20110218186A1-20110908-C00183
    Figure US20110218186A1-20110908-C00184
  • Figure US20110218186A1-20110908-C00185
    Figure US20110218186A1-20110908-C00186
    Figure US20110218186A1-20110908-C00187
  • Figure US20110218186A1-20110908-C00188
    Figure US20110218186A1-20110908-C00189
  • 37A and 37B are diastereomeric with respect to one another, but each is a racemic mixture of its two possible enantiomers. Their absolute stereochemistry has not been conclusively established.
  • Figure US20110218186A1-20110908-C00190
    Figure US20110218186A1-20110908-C00191
  • Figure US20110218186A1-20110908-C00192
    Figure US20110218186A1-20110908-C00193
    Figure US20110218186A1-20110908-C00194
    Figure US20110218186A1-20110908-C00195
  • Figure US20110218186A1-20110908-C00196
    Figure US20110218186A1-20110908-C00197
  • Figure US20110218186A1-20110908-C00198
    Figure US20110218186A1-20110908-C00199
    Figure US20110218186A1-20110908-C00200
    Figure US20110218186A1-20110908-C00201
    Figure US20110218186A1-20110908-C00202
    Figure US20110218186A1-20110908-C00203
  • Figure US20110218186A1-20110908-C00204
    Figure US20110218186A1-20110908-C00205
    Figure US20110218186A1-20110908-C00206
  • Figure US20110218186A1-20110908-C00207
    Figure US20110218186A1-20110908-C00208
  • Figure US20110218186A1-20110908-C00209
    Figure US20110218186A1-20110908-C00210
    Figure US20110218186A1-20110908-C00211
  • Figure US20110218186A1-20110908-C00212
    Figure US20110218186A1-20110908-C00213
    Figure US20110218186A1-20110908-C00214
  • Figure US20110218186A1-20110908-C00215
    Figure US20110218186A1-20110908-C00216
  • Figure US20110218186A1-20110908-C00217
    Figure US20110218186A1-20110908-C00218
    Figure US20110218186A1-20110908-C00219
    Figure US20110218186A1-20110908-C00220
    Figure US20110218186A1-20110908-C00221
    Figure US20110218186A1-20110908-C00222
    Figure US20110218186A1-20110908-C00223
  • Figure US20110218186A1-20110908-C00224
    Figure US20110218186A1-20110908-C00225
    Figure US20110218186A1-20110908-C00226
  • Figure US20110218186A1-20110908-C00227
    Figure US20110218186A1-20110908-C00228
    Figure US20110218186A1-20110908-C00229
    Figure US20110218186A1-20110908-C00230
  • Figure US20110218186A1-20110908-C00231
    Figure US20110218186A1-20110908-C00232
  • Figure US20110218186A1-20110908-C00233
  • 51A and 51B are chirally resolved products derived from 51C. Their absolute stereochemistries have not been conclusively established.
  • Figure US20110218186A1-20110908-C00234
    Figure US20110218186A1-20110908-C00235
    Figure US20110218186A1-20110908-C00236
    Figure US20110218186A1-20110908-C00237
  • 52B and 52C are diastereomeric with respect to one another. 52B and 52C are chirally pure. Their absolute stereochemistry has not been conclusively established.
  • Figure US20110218186A1-20110908-C00238
    Figure US20110218186A1-20110908-C00239
  • Figure US20110218186A1-20110908-C00240
    Figure US20110218186A1-20110908-C00241
    Figure US20110218186A1-20110908-C00242
    Figure US20110218186A1-20110908-C00243
  • Figure US20110218186A1-20110908-C00244
    Figure US20110218186A1-20110908-C00245
    Figure US20110218186A1-20110908-C00246
    Figure US20110218186A1-20110908-C00247
  • Figure US20110218186A1-20110908-C00248
    Figure US20110218186A1-20110908-C00249
  • Figure US20110218186A1-20110908-C00250
    Figure US20110218186A1-20110908-C00251
    Figure US20110218186A1-20110908-C00252
  • Figure US20110218186A1-20110908-C00253
    Figure US20110218186A1-20110908-C00254
  • Figure US20110218186A1-20110908-C00255
    Figure US20110218186A1-20110908-C00256
    Figure US20110218186A1-20110908-C00257
  • Figure US20110218186A1-20110908-C00258
    Figure US20110218186A1-20110908-C00259
    Figure US20110218186A1-20110908-C00260
  • Figure US20110218186A1-20110908-C00261
    Figure US20110218186A1-20110908-C00262
  • C. TABLE 1
    Example Structure [M + H]+
    1A
    Figure US20110218186A1-20110908-C00263
    377.4
    1B
    Figure US20110218186A1-20110908-C00264
    407.1
    1C
    Figure US20110218186A1-20110908-C00265
    411.2
    1D
    Figure US20110218186A1-20110908-C00266
    395.2
    1E
    Figure US20110218186A1-20110908-C00267
    391.3
    1F
    Figure US20110218186A1-20110908-C00268
    407.2
    1G
    Figure US20110218186A1-20110908-C00269
    407.1
    1H
    Figure US20110218186A1-20110908-C00270
    427.4
    1I
    Figure US20110218186A1-20110908-C00271
    427.4
    1J
    Figure US20110218186A1-20110908-C00272
    405.4
    1K
    Figure US20110218186A1-20110908-C00273
    413.2
    1L
    Figure US20110218186A1-20110908-C00274
    405.4
    1M
    Figure US20110218186A1-20110908-C00275
    391.0
    1N
    Figure US20110218186A1-20110908-C00276
    378.4
    1O
    Figure US20110218186A1-20110908-C00277
    396.3
    1P
    Figure US20110218186A1-20110908-C00278
    392.3
    1Q
    Figure US20110218186A1-20110908-C00279
    408.3
    1R
    Figure US20110218186A1-20110908-C00280
    428.3
    1S
    Figure US20110218186A1-20110908-C00281
    406.3
    1T
    Figure US20110218186A1-20110908-C00282
    406.4
    1U
    Figure US20110218186A1-20110908-C00283
    393.2
    2A
    Figure US20110218186A1-20110908-C00284
    393.3
    2B
    Figure US20110218186A1-20110908-C00285
    394  
    2C
    Figure US20110218186A1-20110908-C00286
    447.1
    2D
    Figure US20110218186A1-20110908-C00287
    461.1
    2E
    Figure US20110218186A1-20110908-C00288
    448.3
    2F
    Figure US20110218186A1-20110908-C00289
    408.3
    3A
    Figure US20110218186A1-20110908-C00290
    435.0
    3B
    Figure US20110218186A1-20110908-C00291
    421.0
    3C
    Figure US20110218186A1-20110908-C00292
    422.2
    3D
    Figure US20110218186A1-20110908-C00293
    420.0
    3E
    Figure US20110218186A1-20110908-C00294
    434.3
    3F
    Figure US20110218186A1-20110908-C00295
    448.4
    3G
    Figure US20110218186A1-20110908-C00296
    462.4
    3H
    Figure US20110218186A1-20110908-C00297
    476.5
    3I
    Figure US20110218186A1-20110908-C00298
    490.6
    3J
    Figure US20110218186A1-20110908-C00299
    474.4
    3K
    Figure US20110218186A1-20110908-C00300
    462.5
    3L
    Figure US20110218186A1-20110908-C00301
    490.5
    3M
    Figure US20110218186A1-20110908-C00302
    448.4
    3N
    Figure US20110218186A1-20110908-C00303
    474.5
    3O
    Figure US20110218186A1-20110908-C00304
    490.3
    3P
    Figure US20110218186A1-20110908-C00305
    490.5
    3Q
    Figure US20110218186A1-20110908-C00306
    502.5
    3R
    Figure US20110218186A1-20110908-C00307
    476.5
    3S
    Figure US20110218186A1-20110908-C00308
    504.4
    3T
    Figure US20110218186A1-20110908-C00309
    490.1
    3U
    Figure US20110218186A1-20110908-C00310
    488.4
    3V
    Figure US20110218186A1-20110908-C00311
    421.3
    3W
    Figure US20110218186A1-20110908-C00312
    435.3
    3X
    Figure US20110218186A1-20110908-C00313
    449.3
    3Y
    Figure US20110218186A1-20110908-C00314
    449.3
    3Z
    Figure US20110218186A1-20110908-C00315
    454.0
    3AA
    Figure US20110218186A1-20110908-C00316
    459.3
    3AB
    Figure US20110218186A1-20110908-C00317
    454.4
    3AC
    Figure US20110218186A1-20110908-C00318
    455.4
    4A
    Figure US20110218186A1-20110908-C00319
    470.2
    4B
    Figure US20110218186A1-20110908-C00320
    484.3
    4C
    Figure US20110218186A1-20110908-C00321
    498.3
    4D
    Figure US20110218186A1-20110908-C00322
    510.3
    4E
    Figure US20110218186A1-20110908-C00323
    498.3
    4F
    Figure US20110218186A1-20110908-C00324
    484.1
    4G
    Figure US20110218186A1-20110908-C00325
    496.2
    4H
    Figure US20110218186A1-20110908-C00326
    456.0
    4I
    Figure US20110218186A1-20110908-C00327
    498.3
    5A
    Figure US20110218186A1-20110908-C00328
    455.2
    6A
    Figure US20110218186A1-20110908-C00329
    422.3
    6B
    Figure US20110218186A1-20110908-C00330
    392.2
    6C
    Figure US20110218186A1-20110908-C00331
    484.2
    6D
    Figure US20110218186A1-20110908-C00332
    498.2
    6E
    Figure US20110218186A1-20110908-C00333
    434.2
    7A
    Figure US20110218186A1-20110908-C00334
    470.4
    7B
    Figure US20110218186A1-20110908-C00335
    484.2
    7C
    Figure US20110218186A1-20110908-C00336
    484.2
    8A
    Figure US20110218186A1-20110908-C00337
    393.4
    8B
    Figure US20110218186A1-20110908-C00338
    394.2
    8C
    Figure US20110218186A1-20110908-C00339
    447.3
    8D
    Figure US20110218186A1-20110908-C00340
    407.3
    8E
    Figure US20110218186A1-20110908-C00341
    448.3
    8F
    Figure US20110218186A1-20110908-C00342
    408.4
    9A
    Figure US20110218186A1-20110908-C00343
    447.3
    9B
    Figure US20110218186A1-20110908-C00344
    443.4
    10A
    Figure US20110218186A1-20110908-C00345
    435.3
    10B
    Figure US20110218186A1-20110908-C00346
    421.3
    10C
    Figure US20110218186A1-20110908-C00347
    420.3
    10D
    Figure US20110218186A1-20110908-C00348
    434.3
    10E
    Figure US20110218186A1-20110908-C00349
    448.3
    10F
    Figure US20110218186A1-20110908-C00350
    448.3
    10G
    Figure US20110218186A1-20110908-C00351
    476.2
    10H
    Figure US20110218186A1-20110908-C00352
    474.3
    10I
    Figure US20110218186A1-20110908-C00353
    490.2
    10J
    Figure US20110218186A1-20110908-C00354
    407.4
    11A
    Figure US20110218186A1-20110908-C00355
    393.0
    11B
    Figure US20110218186A1-20110908-C00356
    394.3
    11C
    Figure US20110218186A1-20110908-C00357
    447.4
    11D
    Figure US20110218186A1-20110908-C00358
    448.4
    11E
    Figure US20110218186A1-20110908-C00359
    447.3
    11F
    Figure US20110218186A1-20110908-C00360
    462.4
    12A
    Figure US20110218186A1-20110908-C00361
    391.4
    12B
    Figure US20110218186A1-20110908-C00362
    421.3
    12C
    Figure US20110218186A1-20110908-C00363
    420.3
    12D
    Figure US20110218186A1-20110908-C00364
    434.3
    12E
    Figure US20110218186A1-20110908-C00365
    448.4
    12F
    Figure US20110218186A1-20110908-C00366
    462.4
    12G
    Figure US20110218186A1-20110908-C00367
    448.4
    12H
    Figure US20110218186A1-20110908-C00368
    392.4
    12I
    Figure US20110218186A1-20110908-C00369
    419.4
    12J
    Figure US20110218186A1-20110908-C00370
    433.4
    12K
    Figure US20110218186A1-20110908-C00371
    420.4
    12L
    Figure US20110218186A1-20110908-C00372
    434.3
    13A
    Figure US20110218186A1-20110908-C00373
    322.1
    13B
    Figure US20110218186A1-20110908-C00374
    321.1
    13C
    Figure US20110218186A1-20110908-C00375
    335.2
    13D
    Figure US20110218186A1-20110908-C00376
    349.2
    13E
    Figure US20110218186A1-20110908-C00377
    377.2
    13F
    Figure US20110218186A1-20110908-C00378
    349.1
    13G
    Figure US20110218186A1-20110908-C00379
    375.1
    13H
    Figure US20110218186A1-20110908-C00380
    405.3
    13I
    Figure US20110218186A1-20110908-C00381
    391.1
    13J
    Figure US20110218186A1-20110908-C00382
    389.1
    13K
    Figure US20110218186A1-20110908-C00383
    403.3
    13L
    Figure US20110218186A1-20110908-C00384
    423.1
    13M
    Figure US20110218186A1-20110908-C00385
    417.2
    13N
    Figure US20110218186A1-20110908-C00386
    425.2
    13O
    Figure US20110218186A1-20110908-C00387
    461.2
    13P
    Figure US20110218186A1-20110908-C00388
    421.2
    13Q
    Figure US20110218186A1-20110908-C00389
    404.3
    13R
    Figure US20110218186A1-20110908-C00390
    501.2
    13S
    Figure US20110218186A1-20110908-C00391
    433.1
    14A
    Figure US20110218186A1-20110908-C00392
    346.1
    14B
    Figure US20110218186A1-20110908-C00393
    360.1
    14C
    Figure US20110218186A1-20110908-C00394
    360.2
    15A
    Figure US20110218186A1-20110908-C00395
    418.1
    15B
    Figure US20110218186A1-20110908-C00396
    432.2
    15C
    Figure US20110218186A1-20110908-C00397
    460.2
    15D
    Figure US20110218186A1-20110908-C00398
    474.2
    15E
    Figure US20110218186A1-20110908-C00399
    488.2
    15F
    Figure US20110218186A1-20110908-C00400
    418.2
    15G
    Figure US20110218186A1-20110908-C00401
    432.1
    15H
    Figure US20110218186A1-20110908-C00402
    460.2
    15I
    Figure US20110218186A1-20110908-C00403
    474.3
    15J
    Figure US20110218186A1-20110908-C00404
    488.3
    15K
    Figure US20110218186A1-20110908-C00405
    404.1
    15L
    Figure US20110218186A1-20110908-C00406
    432.1
    15M
    Figure US20110218186A1-20110908-C00407
    446.2
    15N
    Figure US20110218186A1-20110908-C00408
    460.2
    16A
    Figure US20110218186A1-20110908-C00409
    346.1
    16B
    Figure US20110218186A1-20110908-C00410
    360.1
    16C
    Figure US20110218186A1-20110908-C00411
    360.1
    17A
    Figure US20110218186A1-20110908-C00412
    418.1
    17B
    Figure US20110218186A1-20110908-C00413
    460.2
    17C
    Figure US20110218186A1-20110908-C00414
    418.1
    17D
    Figure US20110218186A1-20110908-C00415
    459.2
    17E
    Figure US20110218186A1-20110908-C00416
    404.1
    17F
    Figure US20110218186A1-20110908-C00417
    432.1
    18A
    Figure US20110218186A1-20110908-C00418
    417.3
    18B
    Figure US20110218186A1-20110908-C00419
    437.1
    18C
    Figure US20110218186A1-20110908-C00420
    360.3
    19A
    Figure US20110218186A1-20110908-C00421
    363.4
    19B
    Figure US20110218186A1-20110908-C00422
    391.4
    19C
    Figure US20110218186A1-20110908-C00423
    399.3
    19D
    Figure US20110218186A1-20110908-C00424
    364.4
    20A
    Figure US20110218186A1-20110908-C00425
    391.2
    20B
    Figure US20110218186A1-20110908-C00426
    407.3
    20C
    Figure US20110218186A1-20110908-C00427
    408.3
    20D
    Figure US20110218186A1-20110908-C00428
    434.4
    20E
    Figure US20110218186A1-20110908-C00429
    448.5
    20F
    Figure US20110218186A1-20110908-C00430
    462.5
    20G
    Figure US20110218186A1-20110908-C00431
    435.4
    20H
    Figure US20110218186A1-20110908-C00432
    470.3
    20I
    Figure US20110218186A1-20110908-C00433
    405.4
    20J
    Figure US20110218186A1-20110908-C00434
    419.4
    20K
    Figure US20110218186A1-20110908-C00435
    447.5
    20L
    Figure US20110218186A1-20110908-C00436
    445.4
    20M
    Figure US20110218186A1-20110908-C00437
    431.0
    20N
    Figure US20110218186A1-20110908-C00438
    405.0
    20O
    Figure US20110218186A1-20110908-C00439
    419.1
    20P
    Figure US20110218186A1-20110908-C00440
    467.3
    20Q
    Figure US20110218186A1-20110908-C00441
    481.3
    20R
    Figure US20110218186A1-20110908-C00442
    495.3
    21A
    Figure US20110218186A1-20110908-C00443
    391.2
    21B
    Figure US20110218186A1-20110908-C00444
    391.3
    21C
    Figure US20110218186A1-20110908-C00445
    391.3
    21D
    Figure US20110218186A1-20110908-C00446
    393.3
    21E
    Figure US20110218186A1-20110908-C00447
    393.3
    21F
    Figure US20110218186A1-20110908-C00448
    498.5
    22A
    Figure US20110218186A1-20110908-C00449
    484.2
    22B
    Figure US20110218186A1-20110908-C00450
    498.3
    22C
    Figure US20110218186A1-20110908-C00451
    512.4
    22D
    Figure US20110218186A1-20110908-C00452
    524.3
    22E
    Figure US20110218186A1-20110908-C00453
    469.2
    23A
    Figure US20110218186A1-20110908-C00454
    363.2
    23B
    Figure US20110218186A1-20110908-C00455
    377.0
    23C
    Figure US20110218186A1-20110908-C00456
    403.2
    24A
    Figure US20110218186A1-20110908-C00457
    390.2
    24B
    Figure US20110218186A1-20110908-C00458
    392.2
    24C
    Figure US20110218186A1-20110908-C00459
    392.2
    24D
    Figure US20110218186A1-20110908-C00460
    433.2
    24E
    Figure US20110218186A1-20110908-C00461
    433.2
    24F
    Figure US20110218186A1-20110908-C00462
    419.2
    24G
    Figure US20110218186A1-20110908-C00463
    419.2
    25A
    Figure US20110218186A1-20110908-C00464
    378.2
    26A
    Figure US20110218186A1-20110908-C00465
    391.0
    26B
    Figure US20110218186A1-20110908-C00466
    393.0
    27A
    Figure US20110218186A1-20110908-C00467
    379.1
    27B
    Figure US20110218186A1-20110908-C00468
    379.4
    27C
    Figure US20110218186A1-20110908-C00469
    379.4
    27D
    Figure US20110218186A1-20110908-C00470
    397.3
    27E
    Figure US20110218186A1-20110908-C00471
    397.4
    27F
    Figure US20110218186A1-20110908-C00472
    397.3
    27G
    Figure US20110218186A1-20110908-C00473
    449.3
    27H
    Figure US20110218186A1-20110908-C00474
    380.2
    27I
    Figure US20110218186A1-20110908-C00475
    380.2
    27J
    Figure US20110218186A1-20110908-C00476
    380.2
    27K
    Figure US20110218186A1-20110908-C00477
    398.3
    27L
    Figure US20110218186A1-20110908-C00478
    398.3
    27M
    Figure US20110218186A1-20110908-C00479
    398.3
    27N
    Figure US20110218186A1-20110908-C00480
    408.3
    27O
    Figure US20110218186A1-20110908-C00481
    408.3
    27P
    Figure US20110218186A1-20110908-C00482
    408.3
    27Q
    Figure US20110218186A1-20110908-C00483
    395.4
    27R
    Figure US20110218186A1-20110908-C00484
    395.1
    27S
    Figure US20110218186A1-20110908-C00485
    395.1
    27T
    Figure US20110218186A1-20110908-C00486
    395.3
    27U
    Figure US20110218186A1-20110908-C00487
    395.1
    27V
    Figure US20110218186A1-20110908-C00488
    395.1
    27W
    Figure US20110218186A1-20110908-C00489
    393.4
    28A
    Figure US20110218186A1-20110908-C00490
    375.1
    28B
    Figure US20110218186A1-20110908-C00491
    405.1
    28C
    Figure US20110218186A1-20110908-C00492
    377.1
    28D
    Figure US20110218186A1-20110908-C00493
    407.3
    28E
    Figure US20110218186A1-20110908-C00494
    376.4
    29A
    Figure US20110218186A1-20110908-C00495
    361.0
    29B
    Figure US20110218186A1-20110908-C00496
    389.1
    29C
    Figure US20110218186A1-20110908-C00497
    347.0
    29D
    Figure US20110218186A1-20110908-C00498
    368.9
    30A
    Figure US20110218186A1-20110908-C00499
    425.3
    31A
    Figure US20110218186A1-20110908-C00500
    336.0
    31B
    Figure US20110218186A1-20110908-C00501
    303.1
    31C
    Figure US20110218186A1-20110908-C00502
    303.1
    31D
    Figure US20110218186A1-20110908-C00503
    377.4
    31E
    Figure US20110218186A1-20110908-C00504
    356.1
    31F
    Figure US20110218186A1-20110908-C00505
    317.0
    31G
    Figure US20110218186A1-20110908-C00506
    308.0
    31H
    Figure US20110218186A1-20110908-C00507
    292.1
    31I
    Figure US20110218186A1-20110908-C00508
    346.1
    31J
    Figure US20110218186A1-20110908-C00509
    278.1
    31K
    Figure US20110218186A1-20110908-C00510
    294.0
    31L
    Figure US20110218186A1-20110908-C00511
    308.0
    31M
    Figure US20110218186A1-20110908-C00512
    294.0
    31N
    Figure US20110218186A1-20110908-C00513
    414.1
    31O
    Figure US20110218186A1-20110908-C00514
    308.0
    31P
    Figure US20110218186A1-20110908-C00515
    294.0
    31Q
    Figure US20110218186A1-20110908-C00516
    333.9
    31R
    Figure US20110218186A1-20110908-C00517
    318.1
    31S
    Figure US20110218186A1-20110908-C00518
    279.1
    31T
    Figure US20110218186A1-20110908-C00519
    283.9
    31U
    Figure US20110218186A1-20110908-C00520
    284.1
    31V
    Figure US20110218186A1-20110908-C00521
    268.1
    31W
    Figure US20110218186A1-20110908-C00522
    457.1
    31X
    Figure US20110218186A1-20110908-C00523
    308.8
    31Y
    Figure US20110218186A1-20110908-C00524
    321.1
    31Z
    Figure US20110218186A1-20110908-C00525
    363.1
    31AA
    Figure US20110218186A1-20110908-C00526
    399.1
    32A
    Figure US20110218186A1-20110908-C00527
    391.3
    32B
    Figure US20110218186A1-20110908-C00528
    454.0
    32C
    Figure US20110218186A1-20110908-C00529
    385.3
    32D
    Figure US20110218186A1-20110908-C00530
    413.3
    32E
    Figure US20110218186A1-20110908-C00531
    459.3
    32F
    Figure US20110218186A1-20110908-C00532
    413.3
    32G
    Figure US20110218186A1-20110908-C00533
    399.4
    32H
    Figure US20110218186A1-20110908-C00534
    441.4
    32I
    Figure US20110218186A1-20110908-C00535
    453.3
    32J
    Figure US20110218186A1-20110908-C00536
    357.4
    32K
    Figure US20110218186A1-20110908-C00537
    370.2
    32L
    Figure US20110218186A1-20110908-C00538
    384.2
    32M
    Figure US20110218186A1-20110908-C00539
    396.2
    32N
    Figure US20110218186A1-20110908-C00540
    412.2
    32O
    Figure US20110218186A1-20110908-C00541
    412.2
    32P
    Figure US20110218186A1-20110908-C00542
    384.2
    32Q
    Figure US20110218186A1-20110908-C00543
    426.2
    32R
    Figure US20110218186A1-20110908-C00544
    377.3
    32S
    Figure US20110218186A1-20110908-C00545
    405.4
    32T
    Figure US20110218186A1-20110908-C00546
    391.3
    32U
    Figure US20110218186A1-20110908-C00547
    349.2
    32V
    Figure US20110218186A1-20110908-C00548
    405.3
    32W
    Figure US20110218186A1-20110908-C00549
    361.2
    32X
    Figure US20110218186A1-20110908-C00550
    361.3
    32Y
    Figure US20110218186A1-20110908-C00551
    377.4
    32Z
    Figure US20110218186A1-20110908-C00552
    391.4
    33A
    Figure US20110218186A1-20110908-C00553
    284.9
    33B
    Figure US20110218186A1-20110908-C00554
    279.9
    33C
    Figure US20110218186A1-20110908-C00555
    282.0
    33D
    Figure US20110218186A1-20110908-C00556
    362.9
    33E
    Figure US20110218186A1-20110908-C00557
    303.9
    33F
    Figure US20110218186A1-20110908-C00558
    378.3
    33G
    Figure US20110218186A1-20110908-C00559
    378.2
    33H
    Figure US20110218186A1-20110908-C00560
    350.2
    33I
    Figure US20110218186A1-20110908-C00561
    350.2
    33J
    Figure US20110218186A1-20110908-C00562
    336.2
    33K
    Figure US20110218186A1-20110908-C00563
    379.3
    33L
    Figure US20110218186A1-20110908-C00564
    321.9
    34A
    Figure US20110218186A1-20110908-C00565
    378.4
    34B
    Figure US20110218186A1-20110908-C00566
    406.4
    34C
    Figure US20110218186A1-20110908-C00567
    383.3
    34D
    Figure US20110218186A1-20110908-C00568
    411.4
    34E
    Figure US20110218186A1-20110908-C00569
    419.2
    34F
    Figure US20110218186A1-20110908-C00570
    367.3
    34G
    Figure US20110218186A1-20110908-C00571
    395.5
    34H
    Figure US20110218186A1-20110908-C00572
    383.4
    34I
    Figure US20110218186A1-20110908-C00573
    411.4
    34J
    Figure US20110218186A1-20110908-C00574
    395.0
    34K
    Figure US20110218186A1-20110908-C00575
    395.0
    34L
    Figure US20110218186A1-20110908-C00576
    391.0
    34M
    Figure US20110218186A1-20110908-C00577
    391.0
    34N
    Figure US20110218186A1-20110908-C00578
    413.0
    34O
    Figure US20110218186A1-20110908-C00579
    411.0
    34P
    Figure US20110218186A1-20110908-C00580
    377.4
    35A
    Figure US20110218186A1-20110908-C00581
    407.0
    35B
    Figure US20110218186A1-20110908-C00582
    393.3
    36A
    Figure US20110218186A1-20110908-C00583
    393.4
    36B
    Figure US20110218186A1-20110908-C00584
    375.3
    37A
    Figure US20110218186A1-20110908-C00585
    391.3
    37B
    Figure US20110218186A1-20110908-C00586
    391.3
  • 21B and 21C are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 21D and 21E are diastereomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 24B and 24C are geometric isomers with respect to one another (wherein the hydroxyl is either equatorial or axial), but the conformation of each has not been conclusively established.
  • 24D and 24E are geometric isomers with respect to one another (wherein the hydroxyl is either equatorial or axial), but the conformation of each has not been conclusively established.
  • 24F and 24G are geometric isomers with respect to one another (wherein the hydroxyl is either equatorial or axial), but the conformation of each has not been conclusively established.
  • 27B and 27C are enantiomeric with respect to one another, and their absolute stereochemistry has been conclusively established using X-ray crystallography.
  • 27E and 27F are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27I and 27J are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27L and 27M are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27O and 27P are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27R and 27S are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 27U and 27V are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 37A and 37B are diastereomeric with respect to one another, but each is a racemic mixture of its two possible enantiomers. Their absolute stereochemistry has not been conclusively established.
  • TABLE 2
    Example Structure [M + H]+
    7D
    Figure US20110218186A1-20110908-C00587
    446.4
    7E
    Figure US20110218186A1-20110908-C00588
    462.4
    11G
    Figure US20110218186A1-20110908-C00589
    443.3
    11H
    Figure US20110218186A1-20110908-C00590
    444.3
    11I
    Figure US20110218186A1-20110908-C00591
    429.3
    22F
    Figure US20110218186A1-20110908-C00592
    483.2
    33M
    Figure US20110218186A1-20110908-C00593
    320.1
    33N
    Figure US20110218186A1-20110908-C00594
    382.4
    38A
    Figure US20110218186A1-20110908-C00595
    394.3
    38B
    Figure US20110218186A1-20110908-C00596
    396.4
    38C
    Figure US20110218186A1-20110908-C00597
    409.7
    38D
    Figure US20110218186A1-20110908-C00598
    411.7
    39A
    Figure US20110218186A1-20110908-C00599
    378.3
    39B
    Figure US20110218186A1-20110908-C00600
    378.8
    39C
    Figure US20110218186A1-20110908-C00601
    378.8
    39D
    Figure US20110218186A1-20110908-C00602
    391.4
    39E
    Figure US20110218186A1-20110908-C00603
    393.4
    39F
    Figure US20110218186A1-20110908-C00604
    393.8
    39G
    Figure US20110218186A1-20110908-C00605
    393.8
    40A
    Figure US20110218186A1-20110908-C00606
    406.3
    40B
    Figure US20110218186A1-20110908-C00607
    408.5
    40C
    Figure US20110218186A1-20110908-C00608
    421.3
    41A
    Figure US20110218186A1-20110908-C00609
    407.4
    41B
    Figure US20110218186A1-20110908-C00610
    420.3
    41C
    Figure US20110218186A1-20110908-C00611
    420.3
    41D
    Figure US20110218186A1-20110908-C00612
    406.4
    41E
    Figure US20110218186A1-20110908-C00613
    406.8
    42A
    Figure US20110218186A1-20110908-C00614
    392.3
    42B
    Figure US20110218186A1-20110908-C00615
    392.3
    42C
    Figure US20110218186A1-20110908-C00616
    392.4
    42D
    Figure US20110218186A1-20110908-C00617
    392.8
    42E
    Figure US20110218186A1-20110908-C00618
    392.8
    42F
    Figure US20110218186A1-20110908-C00619
    392.4
    42G
    Figure US20110218186A1-20110908-C00620
    407.7
    42H
    Figure US20110218186A1-20110908-C00621
    407.8
    42I
    Figure US20110218186A1-20110908-C00622
    407.4
    43A
    Figure US20110218186A1-20110908-C00623
    407.3
    43B
    Figure US20110218186A1-20110908-C00624
    407.3
    43C
    Figure US20110218186A1-20110908-C00625
    407.4
    43D
    Figure US20110218186A1-20110908-C00626
    408.3
    43E
    Figure US20110218186A1-20110908-C00627
    408.3
    43F
    Figure US20110218186A1-20110908-C00628
    408.4
    44A
    Figure US20110218186A1-20110908-C00629
    410.3
    44B
    Figure US20110218186A1-20110908-C00630
    410.8
    44C
    Figure US20110218186A1-20110908-C00631
    410.4
    44D
    Figure US20110218186A1-20110908-C00632
    410.8
    44E
    Figure US20110218186A1-20110908-C00633
    410.8
    44F
    Figure US20110218186A1-20110908-C00634
    410.4
    45A
    Figure US20110218186A1-20110908-C00635
    425.8
    45B
    Figure US20110218186A1-20110908-C00636
    425.8
    45C
    Figure US20110218186A1-20110908-C00637
    425.4
    45D
    Figure US20110218186A1-20110908-C00638
    408.3
    45E
    Figure US20110218186A1-20110908-C00639
    408.8
    45F
    Figure US20110218186A1-20110908-C00640
    408.4
    46A
    Figure US20110218186A1-20110908-C00641
    407.4
    46B
    Figure US20110218186A1-20110908-C00642
    407.4
    46C
    Figure US20110218186A1-20110908-C00643
    407.4
    47A
    Figure US20110218186A1-20110908-C00644
    364.7
    47B
    Figure US20110218186A1-20110908-C00645
    379.8
    47C
    Figure US20110218186A1-20110908-C00646
    379.3
    47D
    Figure US20110218186A1-20110908-C00647
    380.4
    47E
    Figure US20110218186A1-20110908-C00648
    379.8
    47F
    Figure US20110218186A1-20110908-C00649
    380.7
    48A
    Figure US20110218186A1-20110908-C00650
    376.3
    48B
    Figure US20110218186A1-20110908-C00651
    390.5
    48C
    Figure US20110218186A1-20110908-C00652
    390.4
    48D
    Figure US20110218186A1-20110908-C00653
    413.7
    48E
    Figure US20110218186A1-20110908-C00654
    441.4
    48F
    Figure US20110218186A1-20110908-C00655
    408.4
    49A
    Figure US20110218186A1-20110908-C00656
    395.3
    49B
    Figure US20110218186A1-20110908-C00657
    446.8
    49C
    Figure US20110218186A1-20110908-C00658
    411.7
    49D
    Figure US20110218186A1-20110908-C00659
    413.7
    50A
    Figure US20110218186A1-20110908-C00660
    336.4
    50B
    Figure US20110218186A1-20110908-C00661
    322.1
    50C
    Figure US20110218186A1-20110908-C00662
    377.7
    50D
    Figure US20110218186A1-20110908-C00663
    294.1
    51A
    Figure US20110218186A1-20110908-C00664
    391.4
    51B
    Figure US20110218186A1-20110908-C00665
    391.4
    51C
    Figure US20110218186A1-20110908-C00666
    391.4
  • TABLE 3
    Exam- [M +
    ple Structure H]+
    52A
    Figure US20110218186A1-20110908-C00667
    469.4
    52B
    Figure US20110218186A1-20110908-C00668
    52C
    Figure US20110218186A1-20110908-C00669
    52D
    Figure US20110218186A1-20110908-C00670
    52E
    Figure US20110218186A1-20110908-C00671
    52F
    Figure US20110218186A1-20110908-C00672
    53A
    Figure US20110218186A1-20110908-C00673
    360.8 (note: [M − H])
    53B
    Figure US20110218186A1-20110908-C00674
    376.8
    53C
    Figure US20110218186A1-20110908-C00675
    352.7
    53D
    Figure US20110218186A1-20110908-C00676
    338.7
    53E
    Figure US20110218186A1-20110908-C00677
    53F
    Figure US20110218186A1-20110908-C00678
    365.8
    54A
    Figure US20110218186A1-20110908-C00679
    389.9
    54B
    Figure US20110218186A1-20110908-C00680
    390.8
    55A
    Figure US20110218186A1-20110908-C00681
    407.5
    55B
    Figure US20110218186A1-20110908-C00682
    391.5
    55C
    Figure US20110218186A1-20110908-C00683
    435.7
    56A
    Figure US20110218186A1-20110908-C00684
    391.4
    56B
    Figure US20110218186A1-20110908-C00685
    393.5
    56C
    Figure US20110218186A1-20110908-C00686
    392.4
    56D
    Figure US20110218186A1-20110908-C00687
    394.5
    57A
    Figure US20110218186A1-20110908-C00688
    57B
    Figure US20110218186A1-20110908-C00689
    57C
    Figure US20110218186A1-20110908-C00690
    57D
    Figure US20110218186A1-20110908-C00691
    356.3
    58A
    Figure US20110218186A1-20110908-C00692
    295.3
    58B
    Figure US20110218186A1-20110908-C00693
    350.3
    58C
    Figure US20110218186A1-20110908-C00694
    308.2
    58D
    Figure US20110218186A1-20110908-C00695
    348.2
    59A
    Figure US20110218186A1-20110908-C00696
    389.4
    59B
    Figure US20110218186A1-20110908-C00697
    403.4
    59C
    Figure US20110218186A1-20110908-C00698
    431.5
    59D
    Figure US20110218186A1-20110908-C00699
    445.5
    59E
    Figure US20110218186A1-20110908-C00700
    431.5
    59F
    Figure US20110218186A1-20110908-C00701
    445.5
    59G
    Figure US20110218186A1-20110908-C00702
    473.5
    59H
    Figure US20110218186A1-20110908-C00703
    487.5
    59I
    Figure US20110218186A1-20110908-C00704
    467.3
    59J
    Figure US20110218186A1-20110908-C00705
    481.5
    59K
    Figure US20110218186A1-20110908-C00706
    509.5
    59L
    Figure US20110218186A1-20110908-C00707
    523.6
    60A
    Figure US20110218186A1-20110908-C00708
    417.5
    60B
    Figure US20110218186A1-20110908-C00709
    459.5
    60C
    Figure US20110218186A1-20110908-C00710
    495.5
    61A
    Figure US20110218186A1-20110908-C00711
    413.9
    61B
    Figure US20110218186A1-20110908-C00712
    413.8
  • 39B and 39C are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 39F and 39G are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 42A and 42B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 42D and 42E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 42G and 42H are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 43A and 43B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 43D and 43E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 44A and 44B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 44D and 44E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 45A and 45B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 45D and 45E are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 46A and 46B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 51A and 51B are resolvable by chiral column chromatography as individual peaks with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • 61A and 61B are enantiomeric with respect to one another, but their absolute stereochemistry has not been conclusively established.
  • EXPERIMENTAL SECTION Introduction
  • Materials: All chemicals were reagent grade and used without further purification.
  • Analytical: Thin-layer chromatography (TLC) was performed on silica gel 60 flexible backed plates (250 microns) from Alltech and visualized by UV 254 irradiation and iodine. Flash chromatography was conducted using the ISCO CombiFlash with RediSep silica gel cartridges (4 g, 12 g, 40 g, 120 g). Flash chromatography was also conducted with silica gel (200-400 mesh, 60 Å, Aldrich). Chromatographic elution solvent systems are reported as volume:volume ratios. All 1H NMR spectra were recorded at ambient temperature on a Bruker-400 MHz spectrometer. They are reported in ppm on the δ scale, from TMS. LC-MS data were obtained using a Thermo-Finnigan Surveyor HPLC and a Thermo-Finnigan AQA MS using either positive or negative electrospray ionization. Program (positive) Solvent A: 10 mM ammonium acetate, pH 4.5, 1% acetonitrile; solvent B: acetonitrile; column: Michrom Bioresources Magic C18 Macro Bullet, detector: PDA λ=220-300 nm. Gradient: 96% A-100% B in 3.2 minutes, hold 100% B for 0.4 minutes. Program (negative) Solvent A: 1 mM ammonium acetate, pH 4.5, 1% acetonitrile; solvent B: acetonitrile; column: Michrom Bioresources Magic C18 Macro Bullet, detector: PDA λ=220-300 nm. Gradient: 96% A-100% B in 3.2 minutes, hold 100% B for 0.4 minutes.
  • The compounds set forth in Tables 1 and 2 are actual examples. The compounds set forth in Table 3 include actual and prophetic examples for which schemes and general procedure descriptions are set forth herein.
  • Example 1A Preparation of 1.3a
  • Method 1A: Pyrrolidine (6.12 mL, 73.38 mmol, 2.0 eq) was added at room temperature to 1.2 (7.31 g, 36.69 mmol, 1.0 eq) and 1.1a (5.00 g, 36.69 mmol, 1.0 eq). The solution was stirred overnight at room temperature and then concentrated under reduced pressure. Diethyl ether (500 mL) was added. The organic mixture was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. Hexane (300 mL) was added to the mixture. The resulting precipitate was collected by filtration, washed with hexane and used for the next step without further purification. Yield: 68%
  • Method 1B: Pyrrolidine (42 mL, 73.38, 2.0 eq) was added drop wise at room temperature to a solution of 1.2 (49.8 g, 0.249 mol, 1.0 eq) and 1.1a (34 g, 0.184 mol, 1.0 eq) in anhydrous methanol (400 mL). The solution was refluxed overnight and then concentrated under reduced pressure. Diethyl ether (500 mL) was added. The organic mixture was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. Hexane (300 mL) was added to the mixture. The resulting precipitate was collected by filtration, washed with hexane, and used for the next step without further purification.
  • Yield: 72%
  • 1H NMR (400 MHz, CDCl3) δ 7.86 (d, 1H), 7.50 (t, 1H), 7.00 (m, 2H), 3.87 (m, 2H), 3.22 (m, 2H), 2.72 (s, 2H), 2.05 (d, 2H), 1.61 (m, 2H), 1.46 (s, 9H)
  • Mass Spectral Analysis m/z=318.0 (M+H)+
  • Preparation of 1.5a
  • To a solution of 1.3a (25 g, 0.078 mol, 1.0 eq) in tetrahydrofuran (250 mL) at −78° C. under nitrogen was added drop wise a 1.0M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (94.5 mL, 0.095 mol, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (33.8 g, 0.095 mol, 1.2 eq) in tetrahydrofuran (150 mL) was added drop wise. The mixture was warmed slowly to room temperature and stiffing was continued for a further 12 h. The mixture was then poured into ice water and the two phases were separated. The organic phase was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 70%
  • 1H NMR (400 MHz, DMSO d6) 7.45-7.20 (m, 2H), 7.00 (m, 2H), 6.15 (s, 1H), 3.70 (m, 2H), 3.20 (m, 2H), 1.90 (m, 2H), 1.75 (m, 2H), 1.40 (s, 9H)
  • Mass Spectral Analysis m/z=450.1 (M+H)+
  • Preparation of 1.8a
  • Method 1C: To a solution of 1.5a (15 g, 33.37 mmol, 1.0 eq) in dimethoxyethane (100 mL) was added sequentially a 2N aqueous solution of sodium carbonate (50.06 mL, 100.12 mmol, 3.0 eq), lithium chloride (4.24 g, 100.12 mmol, 3.0 eq), 1.6 (8.12 g, 36.71 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.77 g, 0.67 mmol, 0.02 eq). The mixture was refluxed for 10 h under nitrogen. The mixture was then cooled to room temperature and water (250 mL) was added. The mixture was extracted with ethyl acetate. The organic layer was further washed with brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 73%
  • Method 1D: To a solution of 1.5a (10 g, 22.25 mmol, 1.0 eq) in dimethoxyethane (67 mL) was added sequentially a 2N aqueous solution of sodium carbonate (33.37 mL, 66.75 mmol, 3.0 eq), lithium chloride (2.83 g, 66.75 mmol, 3.0 eq), 1.6 (4.40 g, 24.47 mmol, 1.1 eq) and palladium, 10 weight % (dry basis) on activated carbon, wet, Degussa type E101 NE/W (0.24 g, 0.11 mmol, 0.005 eq). The mixture was refluxed for 2 h under nitrogen. The mixture was then cooled to room temperature and diluted with dichloromethane (350 mL). The mixture was filtered through a celite plug and dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was triturated with diethyl ether. The precipitate was collected by filtration. Yield: 60%
  • 1H NMR (400 MHz, CDCl3) δ 7.35 (m, 4H), 7.15 (t, 1H), 7.00-6.80 (m, 3H), 5.55 (s, 1H), 3.85 (m, 2H), 3.55 (m, 2H), 3.30 (m, 4H), 2.00 (m, 2H), 1.65 (m, 2H), 1.40 (s, 9H); 1.20 (m, 6H)
  • Mass Spectral Analysis m/z=477.2 (M+H)+
  • Preparation of 1A
  • Method 1E: A 2.0M solution of hydrochloric acid in diethyl ether (34.6 mL, 69.24 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 1.8a (6.00 g, 12.59 mmol, 1.0 eq) in anhydrous dichloromethane (70 mL). The mixture was warmed to room temperature and stiffing was continued for an additional 10 h. Diethyl ether (100 mL) was added to the solution and the resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 99%
  • Method 1F: Trifluoroacetic acid (10.33 mL, 134.09 mmol, 5.5 eq) was added drop wise to a cold (0° C.) solution of 1.8a (11.62 g, 24.38 mmol, 1.0 eq) in anhydrous dichloromethane (50 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h. The mixture was then concentrated under reduced pressure. A saturated solution of sodium bicarbonate (100 mL) was added to the mixture, which was extracted with dichloromethane. The organic phase was separated, washed with brine, dried over sodium sulfate and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added drop wise a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (36.5 mL, 0.073 mol, 3.0 eq). The mixture was then stirred for 1 h at room temperature and concentrated under reduced pressure. Diethyl ether was added. The resulting precipitate was collected by vacuum filtration and washed with diethyl ether. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.06 (m, 2H), 7.43 (s, 4H), 7.27 (t, 1H), 7.00 (m, 3H), 5.95 (s, 1H), 3.45 (m, 2H), 3.23 (m, 6H), 2.00 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=377.4 (M+H)+
  • Elemental analysis:
  • C24H28N2O2, 1HCl
  • Theory: % C, 69.80; % H, 7.08; % N, 6.78.
  • Found: % C, 69.73; % H, 7.04; % N, 6.81.
  • Example 1B
  • 1B was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1b and Method 1B was used.
  • Step 1.3: Method 1C was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.97 (m, 2H), 7.42 (m, 4H), 6.98 (m, 1H), 6.86 (m, 1H), 6.49 (m, 1H), 5.99 (s, 1H), 3.62 (m, 3H), 3.50 (m, 2H), 3.21 (m, 6H), 2.06 (m, 4H), 1.11 (m, 6H) Mass Spectral Analysis m/z=407.1 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 1.25H2O
  • Theory: % C, 64.51; % H, 7.25; % N, 6.02.
  • Found: % C, 64.53; % H, 7.11; % N, 5.89.
  • Example 1C
  • 1C was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1c and Method 1A was used.
  • Step 1.3: Method 1C was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.05 (m, 1.5H), 7.45 (s, 4H), 7.30 (d, 1H), 7.10 (d, 1H), 6.90 (s, 1H), 6.00 (s, 1H), 3.1-3.55 (m, 8H), 2.05 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=411.2 (M+H)+
  • Example 1D
  • 1D was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1d and Method 1B was used.
  • Step 1.3: Method 1D was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.95 (m, 1H), 7.40 (s, 4H), 7.10 (m, 2H), 6.70 (m, 1H), 6.05 (s, 1H), 3.10-3.50 (m, 8H), 2.00 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=395.2 (M+H)+
  • Example 1E
  • 1E was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1e and Method 1A was used.
  • Step 1.3: Method 1D was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (brm, 1H), 7.42 (s, 4H), 7.07 (dd, 1H), 6.94 (d, 1H), 6.79 (d, 1H), 5.92 (s, 1H), 3.45 (brs, 2H), 3.22 (brm, 6H), 2.18 (s, 3H), 2.08 (m, 2H), 1.97 (m, 2H), 1.12 (brd, 6H)+
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 1.5H2O
  • Theory: % C, 66.13; % H, 7.55; % N, 6.17.
  • Found: % C, 65.73; % H, 7.38; % N, 6.05.
  • Example 1F
  • 1F was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1f and Method 1B was used.
  • Step 1.3: Method 1C was used.
  • Step 1.4: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (m, 2H), 7.25 (m, 5H), 6.71 (m, 1H), 6.64 (m, 1H), 5.81 (s, 1H), 3.45 (m, 2H), 3.39 (m, 3H), 3.20 (m, 6H), 2.00 (m, 4H), 1.09 (m, 6H)
  • Mass Spectral Analysis m/z=407.2 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 2H2O
  • Theory: % C, 62.69; % H, 7.36; % N, 5.85.
  • Found: % C, 62.78; % H, 6.90; % N, 5.61.
  • Example 1G
  • 1G was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1 g and Method 1B was used.
  • Step 1.3: Method 1C was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.95 (m, 1H), 8.85 (m, 1H), 7.38 (m, 4H), 6.89 (m, 1H), 6.68 (m, 1H), 6.54 (m, 1H), 5.78 (s, 1H), 3.76 (m, 3H), 3.45 (m, 2H), 3.21 (m, 6H), 2.09 (m, 2H), 1.98 (m, 2H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=407.1 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 0.5H2O
  • Theory: % C, 66.43; % H, 7.14; % N, 6.20.
  • Found: % C, 66.25; % H, 7.19; % N, 6.11.
  • Example 111
  • 111 was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1h and Method 1B was used.
  • Step 1.3: Method 1D was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.80 (brm, 1H), 8.33 (d, 1H), 7.90 (m, 1H), 7.58 (m, 2H), 7.51 (d, 1H), 7.46 (d, 4H), 7.16 (d, 1H), 5.97 (s, 1H), 3.46 (brs, 2H), 3.30 (brm, 6H), 2.25 (d, 2H), 2.05 (m, 2H), 1.13 (brd, 6H)
  • Mass Spectral Analysis m/z=427.4 (M+H)+
  • Elemental analysis:
  • C28H30N2O2, 1HCl, 1.5H2O
  • Theory: % C, 68.63; % H, 6.99; % N, 5.72.
  • Found: % C, 68.96; % H, 6.82; % N, 5.75.
  • Example 1I
  • 1I was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1i and Method 1B was used.
  • Step 1.3: Method 1D was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (brm, 1H), 7.94 (d, 1H), 7.87 (d, 1H), 7.37 (m, 3H), 7.28 (t, 1H), 7.24 (d, 2H), 7.10 (t, 1H), 6.96 (d, 1H), 6.04 (s, 1H), 3.44 (brs, 2H), 3.23 (brs, 6H), 2.09 (brm, 4H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=427.4 (M+H)+
  • Elemental analysis:
  • C28H30N2O2, 1HCl, 0.67H2O
  • Theory: % C, 70.80; % H, 6.86; % N, 5.90.
  • Found: % C, 70.57; % H, 6.72; % N, 5.83.
  • Example 1J
  • 1J was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1j and Method 1A was used.
  • Step 1.3: Method 1D was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.09 (brm, 1H), 7.41 (s, 4H), 6.87 (s, 1H), 6.75 (s, 1H), 5.84 (s, 1H), 3.45 (brs, 2H), 3.20 (brm, 6H), 2.19 (s, 3H), 2.08 (s, 3H), 2.05 (m, 2H), 1.97 (m, 2H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=405.4 (M+H)+
  • Elemental analysis:
  • C26H32N2O2, 1HCl, 0.5H2O
  • Theory: % C, 69.39; % H, 7.62; % N, 6.22.
  • Found: % C, 69.22; % H, 7.49; % N, 6.24.
  • Example 1K
  • 1K was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1k and Method 1B was used.
  • Step 1.3: Method 1C was used.
  • Step 1.4: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.25 (m, 1H), 7.40 (m, 4H), 7.35 (m, 1H), 6.61 (s, 1H), 3.25 (m, 8H), 2.06 (m, 4H), 1.02 (m, 6H) Mass Spectral Analysis m/z=413.2 (M+H)+
  • Example 1L
  • 1L was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.11 and Method 1B was used.
  • Step 1.3: Method 1D was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.84 (brs, 1H), 7.41 (d, 4H), 6.96 (s, 1H), 6.61 (s, 1H), 5.86 (s, 1H), 3.45 (brs, 2H), 3.20 (brm, 6H), 2.23 (s, 3H), 2.13 (s, 3H), 2.08 (m, 2H), 1.96 (m, 2H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=405.4 (M+H)+
  • Elemental analysis:
  • C26H32N2O2, 1HCl, 0.5H2O
  • Theory: % C, 69.39; % H, 7.62; % N, 6.22.
  • Found: % C, 69.69; % H, 7.56; % N, 6.28.
  • Example 1M
  • 1M was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1m and Method 1B was used.
  • Step 1.3: Method 1C was used.
  • Step 1.4: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.05 (m, 2H), 7.46 (m, 2H), 7.20 (m, 3H), 7.01 (m, 1H), 6.82 (m, 1H), 6.48 (m, 1H), 3.45 (m, 2H), 3.28 (m, 6H), 2.24 (m, 2H), 2.06 (m, 2H), 1.60 (m, 3H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=391.0 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.25H2O
  • Theory: % C, 69.59; % H, 7.36; % N, 6.49.
  • Found: % C, 69.25; % H, 7.29; % N, 6.58.
  • Example 1N Preparation of 1.10
  • To an oven-dried 2-necked 500 mL flask charged with anhydrous toluene (90 mL) at −78° C. was added n-butyl lithium (2.5 M solution in hexane, 40 mL, 0.1 mol, 1.0 eq). A solution of 2,5-dibromo-pyridine (1.9) (23.69 g, 0.1 mol, 1.0 eq) in anhydrous toluene (50 mL) was added dropwise. The reaction mixture was stirred at −78° C. for 2 h and then poured onto freshly crushed dry-ice (˜500 g). The dry-ice mixture was then left at room temperature for 10 h. The volatiles were removed under reduced pressure and the residue was dissolved in water. The insoluble solids were filtered and the filtrate was acidified to pH 2, at which point a light brown solid precipitated out. The solids were collected by filtration and recrystallized from acetic acid (500 mL). This provided 1.10 isolated as its acetic acid salt. Yield: 74%
  • 1H NMR (400 MHz, DMSO d6) δ 8.84 (d, 1H), 8.25 (dd, 1H), 7.98 (d, 1H)
  • Mass Spectral Analysis m/z=202.06 (M+H)+
  • Preparation of 1.11
  • To a suspension of 5-bromo-pyridine-2-carboxylic acid (1.10) (808 mg, 3.01 mmol, 1.0 eq) in dry dichloromethane (5 mL) was added oxalyl chloride (0.34 mL, 3.96 mmol, 1.3 eq) followed by 2 drops of N,N-dimethylformamide. The reaction mixture was heated under reflux for 1 h. After cooling to room temperature, the mixture was concentrated under reduced pressure to provide the crude product 1.11, which was used for the next step without purification.
  • Preparation of 1.13
  • To a suspension of 1.11 (crude, as of 3.01 mmol, 1.0 eq) in dry tetrahydrofuran (5 mL) was added N,N-diethylamine (1.12) (1.56 mL, 15.08 mmol, 5.0 eq) drop wise. The reaction mixture was stirred at room temperature for 2 h. Ethyl acetate (20 mL) was added and the mixture was washed with water (20 mL), saturated aqueous sodium bicarbonate (30 mL), 1M aqueous hydrochloric acid (20 mL) and brine. The organics were dried over sodium sulfate, filtered and concentrated under reduced pressure to give a red/brown crystalline solid.
  • Yield: 88% over two steps
  • 1H NMR (400 MHz, CDCl3) δ 8.64 (d, 1H), 7.91 (dd, 1H), 7.53 (d, 1H), 3.56 (q, 2H), 3.39 (q, 2H), 1.27 (t, 3H), 1.17 (t, 3H)
  • Mass Spectral Analysis m/z=257.15 (M+H)+
  • Preparation of 1.7
  • To a solution of bis(pinacolato)diboron (1.14) (2.18 g, 8.6 mmol, 1.2 eq) in N,N-dimethylformamide (10 mL) at 0° C. was added potassium acetate (2.3 g, 23.4 mmol, 3.0 eq), 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (171 mg, 0.23 mmol, 0.03 eq). The reaction mixture was heated at 80° C. at which point a solution of 1.13 (2.0 g, 7.8 mmol, 1.0 eq) in N,N-dimethylformamide (10 mL) was added dropwise. The reaction mixture was stirred at 80° C. for another 10 h. Ethyl acetate (75 mL) and water (50 mL) were added and the two phases were separated. The organic phase was washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a dark brown oil, which solidified to needles. The crude product was triturated with hexane. The resulting solid was collected by filtration. Yield: 52%
  • 1H NMR (400 MHz, CDCl3) δ 8.92 (d, 1H), 8.14 (dd, 1H), 7.53 (d, 1H), 3.55 (q, 2H), 3.32 (q, 2H), 1.36 (s, 12H), 1.27 (t, 3H), 1.12 (t, 3H)
  • Preparation of 1.8b
  • To a solution of 1.5a (1.48 g, 3.29 mmol, 1.0 eq) in dimethoxyethane (DME) (20 mL) under nitrogen was added sequentially a 2M aqueous solution of sodium carbonate (4.94 mL, 9.87 mmol, 3.0 eq), lithium chloride (0.42 g, 9.87 mmol, 3.0 eq), palladium (70 mg, 10 wt. % (dry basis) on activated carbon, 0.033 mmol, 0.01 eq), and 1.7 (1.0 g, 3.29 mmol, 1.0 eq). The mixture was heated under reflux for 10 h. Dichloromethane (200 mL) was added to dilute the reaction mixture and palladium(0) on carbon was filtered off on a celite pad. The filtrate was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 76%
  • 1H NMR (400 MHz, CDCl3) δ 8.56 (dd, 1H), 7.75 (dd, 1H), 7.64 (dd, 1H), 7.22 (m, 1H), 6.99-6.85 (m, 3H), 5.62 (s, 1H), 3.88 (m, 2H), 3.59 (q, 2H), 3.45 (q, 2H), 3.34 (m, 2H), 2.06 (m, 2H), 1.69 (m, 2H), 1.48 (s, 9H), 1.29 (t, 3H), 1.20 (t, 3H)
  • Mass Spectral Analysis m/z=478.0 (M+H)+
  • Preparation of 1N
  • To a cold (0° C.) solution of 1.8b (2 g, 4.18 mmol, 1.0 eq) in anhydrous dichloromethane (20 mL) was slowly added a 4.0 M solution of hydrogen chloride in dioxane (5.2 mL, 20.8 mmol, 5.0 eq). The reaction mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure. The resulting foamy solids were soaked in diethyl ether to give the fine powders, which were collected by filtration and washed sequentially with ethyl acetate and diethyl ether.
  • Yield: 95%
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (m, 2H), 8.60 (d, 1H), 7.90 (dd, 1H), 7.61 (d, 1H), 7.29 (m, 1H), 7.06 (d, 1H), 6.98 (m, 2H), 6.09 (s, 1H), 3.47 (q, 2H), 3.35-3.13 (m, 6H), 2.06 (m, 4H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=378.4 (M+H)+
  • Elemental analysis:
  • C23H27N3O2, 2HCl, 0.5H2O
  • Theory: % C, 60.13; % H, 6.58; % N, 9.15.
  • Found: % C, 60.34; % H, 6.60; % N, 9.10.
  • Example 1O
  • 1O was obtained according to a procedure similar to the one described for 1N, with the following exception:
  • Step 1.1: 1.1a was replaced by 1.1d.
  • 1H NMR (400 MHz, DMSO d6) δ 8.96 (m, 1H), 8.62 (d, 1H), 7.92 (dd, 1H), 7.61 (d, 1H), 7.12 (m, 2H), 6.78 (dd, 1H), 6.20 (s, 1H), 3.47 (q, 2H), 3.30 (q, 2H), 3.24 (m, 4H), 2.05 (m, 4H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=396.3 (M+H)+
  • Elemental analysis:
  • C23H26FN3O2, 1.05HCl, 1H2O
  • Theory: % C, 61.15; % H, 6.48; % N, 9.30; % Cl, 8.24.
  • Found: % C, 61.11; % H, 6.44; % N, 9.18; % Cl, 8.28.
  • Example 1P
  • 1P was obtained according to a procedure similar to the one described for 1N, with the following exception:
  • Step 1.1: 1.1a was replaced by 1.1e.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brm, 1H), 8.60 (d, 1H), 7.89 (dd, 1H), 7.61 (d, 1H), 7.09 (dd, 1H), 6.96 (d, 1H), 6.77 (s, 1H), 6.07 (s, 1H), 3.47 (q, 2H), 3.30 (q, 2H), 2.21 (brm, 4H), 2.18 (s, 3H), 2.04 (brm, 4H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=392.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O2, 2HCl
  • Theory: % C, 62.07; % H, 6.73; % N, 9.05; % Cl, 15.27.
  • Found: % C, 61.81; % H, 6.69; % N, 8.95; % Cl, 15.42.
  • Example 1Q
  • 1Q was obtained according to a procedure similar to the one described for 1N, with the following exceptions:
  • Step 1.1: 1.1a was replaced by 1.1f and Method 1A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (m, 2H), 8.38 (m, 1H), 7.69 (m, 1H), 7.48 (m, 1H), 7.28 (m, 1H), 6.75 (m, 1H), 6.69 (m, 1H), 5.99 (s, 1H), 3.40 (m, 5H), 3.26 (m, 6H), 2.08 (m, 4H), 1.20 (m, 3H), 1.10 (m, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 1HCl, 0.25H2O
  • Theory: % C, 64.28; % H, 6.85; % N, 9.37; % Cl, 7.91.
  • Found: % C, 64.07; % H, 6.84; % N, 9.23; % Cl, 8.18.
  • Example 1R
  • 1R was obtained according to a procedure similar to the one described for 1N, with the following exception:
  • Step 1.1: 1.1a was replaced by 1.1 h.
  • 1H NMR (400 MHz, DMSO d6) δ 9.06 (brs, 0.5H), 8.90 (brs, 0.5H), 8.65 (d, 1H), 8.33 (d, 1H), 7.95 (dd, 1H), 7.91 (m, 1H), 7.64 (d, 1H), 7.59 (m, 2H), 7.53 (d, 1H), 7.14 (d, 1H), 6.11 (s, 1H), 3.48 (q, 2H), 3.32 (brm, 6H), 2.26 (d, 2H), 2.10 (m, 2H), 1.18 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=428.3 (M+H)+
  • Elemental analysis:
  • C27H29N3O2, 1.8HCl, 1H2O
  • Theory: % C, 63.44; % H, 6.47; % N, 8.22; % Cl, 12.48.
  • Found: % C, 63.36; % H, 6.22; % N, 8.14; % Cl, 12.87.
  • Example 1S
  • 1S was obtained according to a procedure similar to the one described for 1N, with the following exception:
  • Step 1.1: 1.1a was replaced by 1.1j.
  • 1H NMR (400 MHz, DMSO d6) δ 8.89 (brm, 2H), 8.59 (d, 1H), 7.88 (dd, 1H), 7.61 (d, 1H), 6.89 (s, 1H), 6.73 (s, 1H), 5.99 (s, 1H), 3.47 (q, 2H), 3.30 (q, 2H), 3.20 (brm, 4H), 2.20 (s, 3H), 2.09 (s, 3H), 2.06 (m, 2H), 1.97 (m, 2H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=406.3 (M+H)+
  • Elemental analysis:
  • C25H31N3O2, 2HCl, 2H2O
  • Theory: % C, 58.36; % H, 7.25; % N, 8.17% Cl, 13.78
  • Found: % C, 58.45; % H, 7.16; % N, 8.16; % Cl, 13.68.
  • Example 1T
  • 1T was obtained according to a procedure similar to the one described for 1N, with the following exception:
  • Step 1.1: 1.1a was replaced by 1.11.
  • 1H NMR (400 MHz, DMSO d6) δ 9.02 (brm, 1H), 8.56 (d, 1H), 7.87 (dd, 1H), 7.61 (d, 1H), 6.98 (s, 1H), 6.59 (s, 1H), 6.01 (s, 1H), 3.47 (q, 2H), 3.30 (q, 2H), 3.25 (m, 2H), 3.14 (brs, 2H), 2.24 (s, 3H), 2.15 (s, 3H), 2.09 (m, 2H), 2.02 (m, 2H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=406.4 (M+H)+
  • Elemental analysis:
  • C25H31N3O2, 1.9HCl, 0.5H2O
  • Theory: % C, 62.06; % H, 7.06; % N, 8.69; % Cl, 13.92.
  • Found: % C, 61.90; % H, 7.03; % N, 8.45; % Cl, 13.85.
  • Example 1U Preparation of 1U
  • A solution of 1G (1.00 g, 2.46 mmol, 1.0 eq) in dichloromethane (12 mL) was added drop wise to a cold (−78° C.) solution of boron tribromide, 1.0M, in anhydrous dichloromethane (13.53 mL, 13.53 mmol, 5.5 eq). The mixture was warmed to room temperature and stiffing was continued for an additional 1 h. Water (1.2 mL) was added drop wise to the cooled (0° C.) reaction mixture and then a saturated solution of sodium bicarbonate (3.7 mL) was added. The resulting mixture was stirred for 1 h at room temperature. A saturated solution of sodium bicarbonate was added to the mixture until the solution was basic when tested with pH paper. The phases were separated and the aqueous phase was extracted with dichloromethane. The organic phases were combined and washed with brine. A gummy residue stuck to the walls of the separatory funnel. It was dissolved in methanol and combined with the dichloromethane extracts. The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 79%
  • 1H NMR (400 MHz, DMSO d6) δ 9.66 (m, 1H), 7.37 (m, 4H), 6.77 (m, 1H), 6.32 (m, 2H), 5.62 (s, 1H), 3.32 (m, 5H), 2.89 (m, 2H), 2.76 (m, 2H), 1.78 (m, 2H), 1.67 (m, 2H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=393.2 (M+H)+
  • Elemental analysis:
  • C24H28N2O3, 0.5H2O
  • Theory: % C, 71.80; % H, 7.28; % N, 6.98.
  • Found: % C, 71.79; % H, 7.13; % N, 6.94.
  • Example 2A Preparation of 2.2
  • Pyrrolidine (104 mL, 1.256 mol, 2.0 eq) was added at room temperature to 1.2 (125.2 g, 0.628 mol, 1.0 eq) and 2.1 (95.6 g, 0.628 mol, 1.0 eq). The solution was stirred at 70° C. for 30 min and then cooled to room temperature and stirred for 48 h. The mixture was then concentrated under reduced pressure and ethyl acetate (800 mL) was added. The organic mixture was washed with a 1N aqueous solution of hydrochloric acid, water, brine and dried over sodium sulfate. Diethyl ether (500 mL) was added to the organics and the mixture was stirred overnight at room temperature. The resulting precipitate was collected by filtration, washed with hexane and used for the next step without further purification. Yield: 75%
  • 1H NMR (400 MHz, CDCl3) δ 7.31 (d, 1H), 7.08 (m, 1H), 6.87 (d, 1H), 6.06 (s, 1H), 3.86 (br s, 2H), 3.19 (br s, 2H), 2.69 (s, 2H), 2.02 (m, 2H), 1.58 (m, 2H), 1.47 (s, 9H)
  • Mass Spectral Analysis m/z=332.4 (M−H)
  • Preparation of 2.4
  • To a solution of 2.3 (2.17 g, 14.4 mmol, 1.2 eq) and imidazole (2.04 g, 30.03 mmol, 2.5 eq) in dimethylformamide (20 mL) at room temperature under nitrogen was added drop wise a solution of 2.2 (4 g, 12.01 mmol, 1.0 eq) in dimethylformamide (15 mL). The mixture was stirred overnight at room temperature and then diluted with ethyl acetate. The organics were washed with water, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was triturated with methanol and then isolated using vacuum filtration and used without further purification. Yield: 76%
  • 1H NMR (400 MHz, DMSO d6) δ 7.10 (m, 2H), 6.99 (d, 1H), 3.70 (m, 2H), 3.11 (brs, 2H), 2.81 (s, 2H), 1.84 (m, 2H), 1.60 (m, 2H), 1.40 (s, 9H), 0.94 (s, 9H), 0.17 (s, 6H)
  • Preparation of 2.5
  • To a solution of 2.4 (4 g, 8.94 mmol, 1.0 eq) in tetrahydrofuran (20 mL) at −78° C. under nitrogen was added drop wise a 1.0M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (6.2 mL, 10.72 mmol, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (3.83 g, 10.72 mmol, 1.2 eq) in tetrahydrofuran (20 mL) was added drop wise. The mixture was stirred and allowed to warm slowly to room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 90.5%
  • 1H NMR (400 MHz, CDCl3) δ 6.76 (m, 3H), 5.56 (s, 1H), 3.85 (br s, 2H), 3.26 (m, 2H), 2.05 (m, 2H), 1.65 (m, 2H), 1.47 (s, 9H), 0.97 (s, 9H), 0.18 (s, 6H)
  • Preparation of 2.6a
  • To a solution of 2.5 (4.47 g, 7.71 mmol, 1.0 eq) in dimethoxyethane (35 mL) was added sequentially a 2N aqueous solution of sodium carbonate (11.6 mL, 23.13 mmol, 3.0 eq), lithium chloride (0.98 g, 23.13 mmol, 3.0 eq), 1.6 (1.87 g, 8.48 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.18 g, 0.15 mmol, 0.02 eq). The mixture was refluxed for 4 h under nitrogen. The mixture was then cooled to room temperature and water was added. The mixture was extracted with ethyl acetate. The organic layer was further washed with a 2N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was triturated with hexanes and used without further purification. Yield: 84%
  • 1H NMR (400 MHz, DMSO d6) δ 7.39 (m, 4H), 6.87 (d, 1H), 6.69 (m, 1H), 6.37 (d, 1H), 5.89 (s, 1H), 3.71 (m, 2H), 3.45 (brs, 2H), 3.23 (m, 4H), 1.85 (m, 2H), 1.70 (m, 2H), 1.41 (s, 9H); 1.10 (m, 6H), 0.87 (s, 9H), 0.08 (s, 6H)
  • Mass Spectral Analysis m/z=607.0 (M+H)+
  • Preparation of 2.7a
  • To a solution of 2.6a (0.50 g, 0.82 mmol, 1.0 eq) in tetrahydrofuran (10 mL) was added a 1N solution of tetrabutylammonium fluoride (2.5 mL, 2.47 mmol, 3.0 eq) in tetrahydrofuran at 0° C. The mixture was stirred for 1 h at room temperature under nitrogen. The mixture was diluted with ethyl acetate. The organic layer was washed with a saturated solution of aqueous sodium bicarbonate, brine, a 1N solution of hydrochloric acid and brine. The solution was then dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was triturated with a diethyl ether/hexanes mixture (3:7) and used without further purification.
  • Yield: 74%
  • 1H NMR (400 MHz, CDCl3) δ 7.34 (s, 4H), 6.80 (d, 1H), 6.67 (m, 1H), 6.49 (d, 1H), 5.87 (s, 1H), 5.57 (s, 1H), 3.84 (brs, 2H), 3.56 (brs, 2H), 3.30 (brs, 4H), 2.00 (m, 2H), 1.64 (m, 2H), 1.47 (s, 9H), 1.20 (m, 6H)
  • Mass Spectral Analysis m/z=493.0 (M+H)+
  • Preparation of 2A
  • A 2.0M solution of hydrochloric acid in diethyl ether (1.7 mL, 3.35 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 2.7a (0.30 g, 0.61 mmol, 1.0 eq) in anhydrous dichloromethane (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h. Diethyl ether (100 mL) was added to the solution. The resulting precipitate was collected by filtration and washed with diethyl ether. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity).
  • Yield: 50%
  • 1H NMR (400 MHz, DMSO d6) δ 9.03 (m, 2H), 7.42 (s, 4H), 6.85 (d, 1H), 6.64 (m, 1H), 6.42 (d, 1H), 5.91 (s, 1H), 3.49 (m, 4H), 3.21 (m, 5H), 2.08 (m, 2H), 1.96 (m, 2H), 1.13 (m, 6H)
  • Mass Spectral Analysis m/z=393.3 (M+H)+
  • Elemental analysis:
  • C24H28N2O2, 1HCl, 1H2O
  • Theory: % C, 64.49; % H, 6.99; % N, 6.27.
  • Found: % C, 64.59; % H, 6.67; % N, 6.26.
  • Example 2B
  • 2B was obtained according to a procedure similar to the one described for 2A, with the following exception:
  • Step 2.4: 1.6 was replaced by 1.7.
  • 1H NMR (400 MHz, DMSO d6) δ 8.94 (brm, 2H), 8.59 (s, 1H), 7.90 (dd, 1H), 7.62 (d, 1H), 6.88 (d, 1H), 6.67 (dd, 1H), 6.38 (d, 1H), 6.06 (s, 1H), 3.47 (q, 2H), 3.22 (m, 6H), 2.07 (m, 2H), 1.97 (m, 2H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=394 (M+H)+
  • Elemental analysis:
  • C23H27N3O3, 2HCl, 1.25H2O
  • Theory: % C, 56.50; % H, 6.49; % N, 8.59; % Cl, 14.50.
  • Found: % C, 56.55; % H, 6.46; % N, 8.39; % Cl, 14.49.
  • Example 2C Preparation of 2.9a
  • A mixture of 2.7a (0.210 g, 0.00042 mol, 1.0 eq), cyclopropylmethyl bromide (2.8a) (0.12 mL, 0.0012 mol, 2.95 eq) and potassium carbonate (0.350 g, 0.0025 mole, 6.0 eq) in N,N-dimethylformamide (5 mL) was stirred for 48 h at 80° C. The mixture was cooled to room temperature, poured into water (50 mL) and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 96%
  • Mass Spectral Analysis m/z=547.12 (M+H)+
  • Preparation of 2C
  • To a cold (0° C.) solution of 2.9a (0.200 g, 0.00036 mol, 1.0 eq) in anhydrous dichloromethane (10 mL) was added drop wise a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (1.8 mL, 0.0036 mole, 10.0 eq). The mixture was warmed slowly to room temperature and stirring was continued for 12 h at room temperature. The mixture was concentrated under reduced pressure. Diethyl ether was then added to the mixture, which was stirred for 1 h at room temperature. The precipitate was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 63%
  • 1H NMR (400 MHz, DMSO d6) δ 8.85 (m, 1H), 7.40 (s, 4H), 6.97 (d, 1H), 6.80 (m, 1H), 6.45 (d, 1H), 5.95 (s, 1H), 3.65 (d, 2H), 3.10-3.50 (m, 8H), 2.00 (m, 4H), 1.10 (m, 7H), 0.50 (m, 2H), 0.20 (m, 2H)
  • Mass Spectral Analysis m/z=447.1 (M+H)+
  • Example 2D
  • 2D was obtained according to a procedure similar to the one described for 2C, with the following exception:
  • Step 2.7: 2.8a was replaced by 2.8b (method 2A).
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (s, 1H), 7.45 (s, 4H), 7.00 (m, 1H), 6.80 (m, 1H), 6.45 (m, 1H), 6.00 (s, 1H), 4.55 (m, 1H), 3.10-3.55 (m, 8H), 2.00 (m, 4H), 1.80 (m, 2H), 1.60 (m, 4H), 1.50 (m, 2H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=461.1 (M+H)+
  • Example 2E Preparation of 2.7b
  • Intermediate 2.7b was obtained according to a procedure similar to the one described for 2.7a (see 2A), except 1.6 was replaced by 1.7 in Step 2.4.
  • Preparation of 2.9b
  • To a solution of 2.7b (1.0 g, 2.03 mmol, 1.0 eq), 2.8e (0.29 g, 4.06 mmol, 2.0 eq), triphenylphosphine (1.06 g, 4.06 mmol, 2.0 eq) and triethylamine (0.82 g, 8.12 mmol, 4.0 eq) in tetrahydrofuran (50 mL) at 0° C. was added diisopropyl azodicarboxylate (DIAD) (0.82 g, 4.06 mmol, 2.0 eq). The mixture was warmed to room temperature and stirred for 48 h at room temperature. Methylene chloride was added and the crude mixture was washed with water, concentrated under reduced pressure and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 45%
  • 1H NMR (400 MHz, CDCl3) δ 8.56 (s, 1H), 7.76 (dd, 1H), 7.64 (d, 1H), 6.89 (d, 1H), 6.78 (m, 1H), 6.50 (d, 1H), 5.65 (s, 1H), 3.86 (brm, 2H), 3.62 (m, 4H), 3.45 (q, 2H), 3.32 (brm, 2H), 2.05 (brm, 2H), 1.67 (brm, 2H), 1.48 (s, 9H), 1.30 (m, 4H), 1.21 (t, 3H), 0.60 (m, 2H), 0.30 (m, 2H)
  • Mass Spectral Analysis m/z=548.4 (M+H)+
  • Preparation of 2E
  • To a solution of 2.9b (0.50 g, 0.913 mmol, 1.0 eq) in methylene chloride (3 mL) was slowly added an excess of a 1.0M solution of anhydrous hydrochloric acid in diethyl ether. The mixture was stirred for 16 h at room temperature and then concentrated under reduced pressure. This mixture (0.41 g) was purified by HPLC using a 20×150 mm XTerra Reversed Phase-HPLC column (eluent: 95:5 A:B to 1:99 A:B where A is 0.1% ammonia in Milli-Q water and B is acetonitrile). After HPLC purification, the pure product (0.10 g, 0.22 mmol, 1.0 eq) was obtained as the free amine, which was dissolved in methanol (10 mL) at 0° C. under nitrogen and treated with a 1.0M solution of anhydrous hydrochloric acid in diethyl ether (0.47 mL, 0.47 mmol, 2.1 eq). The mixture was stirred at 0° C. for 30 min. The mixture was concentrated under reduced pressure and dried under vacuum. Yield: 26%
  • 1H NMR (400 MHz, CDCl3) δ 9.75 (brs, 1H), 9.33 (brs, 1H), 9.18 (s, 1H), 8.45 (brd, 1H), 7.96 (brd, 1H), 6.94 (d, 1H), 6.80 (m, 1H), 6.42 (brm, 2H), 3.66 (brm, 4H), 3.46 (brm, 6H), 2.30 (brm, 4H), 1.35 (t, 3H), 1.22 (brm, 4H), 0.62 (m, 2H), 0.31 (m, 2H)
  • Mass Spectral Analysis m/z=448.3 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 1.75HCl, 1.5H2O
  • Theory: % C, 60.23; % H, 7.07; % N, 7.80; % Cl, 11.52.
  • Found: % C, 60.50; % H, 6.99; % N, 7.77; % Cl, 11.38.
  • Example 2F
  • 2F was obtained according to a procedure similar to the one described for 2E, with the following exception:
  • Step 2.7: 2.8e was replaced by 2.8d.
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (brs, 2H), 8.62 (d, 1H), 7.93 (dd, 1H), 7.61 (d, 1H), 7.03 (d, 1H), 6.89 (dd, 1H), 6.47 (d, 1H), 6.13 (s, 1H), 3.64 (s, 3H), 3.47 (q, 2H), 3.24 (m, 6H), 2.05 (brm, 4H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 1.25HCl, 1.25H2O
  • Theory: % C, 60.61; % H, 6.94; % N, 8.84% Cl, 9.32
  • Found: % C, 60.69; % H, 6.87; % N, 8.66; % Cl, 9.35.
  • Note: 2F was also obtained according to a procedure similar to the one described for 2C with the following exceptions:
  • Step 2.7: 2.8a was replaced by 2.8c and method 2C was used (alkylation reaction conducted in acetone instead of N,N-dimethylformamide).
  • Example 3A Preparation of 3.1a
  • To a cold (0° C.) solution of 2.7a (2.5 g, 0.0050 mol, 1.0 eq) in anhydrous dichloromethane (100 mL), was added N-triphenyltrifluoromethane sulfonimide (1.4) (2 g, 0.0055 mol, 1.1 eq) followed by addition of triethylamine (0.85 mL, 0.060 mol, 1.2 eq). The mixture was allowed to warm slowly to room temperature and stirring was continued for 12 h. The mixture was diluted with ethyl acetate and washed successively with water, aqueous 1N NaOH, water, and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 78%
  • Mass Spectral Analysis m/z=666.06 (M+H+CH3CN)+
  • Preparation of 3.2a
  • To a stirred solution of 3.1a (2.5 g, 0.040 mol, 1.0 eq) in a mixture of methanol (30 mL) and dimethylsulfoxide (40 mL) was added triethylamine (1.23 mL, 0.088 mol, 2.2 eq). Carbon monoxide gas was bubbled through the mixture for 5 min. To the mixture was added palladium (II) acetate (0.090 g, 0.00040 mol, 0.1 eq) followed by 1,1′-bis(diphenylphosphino)ferrocene (0.443 g, 0.00080 mol, 0.2 eq). Carbon monoxide gas was bubbled through the mixture for 15 min and the mixture was then stirred under an atmosphere of carbon monoxide and heated at 65° C. overnight. The mixture was cooled to room temperature and poured into water. The mixture was extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over sodium sulfate and filtered. Evaporation of the solvent under reduced pressure afforded a dark oil. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 75%
  • Mass Spectral Analysis m/z=576.08 (M+H+CH3CN)+
  • Preparation of 3A
  • To a cold (0° C.) solution of 3.2a (0.140 g, 0.00026 mole, 1.0 eq) in anhydrous dichloromethane (10 mL) was added drop wise a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (2.6 mL, 0.0026 mole, 10.0 eq). The mixture was warmed slowly to room temperature and stirring was continued for 12 h at room temperature. An additional 1.0 mL of a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether was added to the reaction mixture, which was allowed to stir for an additional 12 h at room temperature. The mixture was concentrated under reduced pressure. Diethyl ether was then added to the mixture, which was stirred for 2 h at room temperature. The resulting precipitate was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 53%
  • 1H NMR (400 MHz, DMSO d6) δ 9.08 (m, 2H), 7.90 (d, 1H), 7.60 (s, 1H), 7.40 (s, 4H), 7.15 (d, 1H), 6.00 (s, 1H), 3.70 (s, 3H), 3.10-3.50 (m, 8H), 2.1 (m, 4H), 1.10 (m, 6H) Mass Spectral Analysis m/z=435.0 (M+H)+
  • Example 3B Preparation of 3.3a
  • To a cold (0° C.) solution of 3.2a (1.41 g, 0.0026 mol, 1.0 eq) in tetrahydrofuran (20 mL), was added a solution of lithium hydroxide monohydrate (0.332 g, 0.0079 mole, 3.0 eq) in water (3 mL). Methanol (6 mL) was then added to the reaction mixture, which was stirred at room temperature for 12 h. A solution of lithium hydroxide monohydrate (0.165 g, 0.0058 mole, 1.5 eq) in water (3 mL) was added to the reaction mixture, which was stirred for an additional 12 h at room temperature. The mixture was concentrated under reduced pressure and the residue was dissolved in ethyl acetate. The organic solution was dried over sodium sulfate and filtered. Evaporation of the filtrate provided a solid, which was triturated in hexane. The precipitate was collected by filtration. Yield: 85%
  • Mass Spectral Analysis m/z=562.08 (M+H+CH3CN)+
  • Preparation of 3B
  • To a cold (0° C.) solution of 3.3a (0.200 g, 0.00038 mole, 1.0 eq) in anhydrous dichloromethane (10 mL) was added drop wise a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (1.9 mL, 0.0038 mole, 10 eq). The mixture was warmed slowly to room temperature and stirring was continued for 12 h at room temperature. The desired product precipitates from the reaction mixture. The precipitate was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (m, 1.5H), 7.85 (d, 1H), 7.60 (s, 1H), 7.40 (s, 4H), 7.10 (d, 1H), 6.00 (s, 1H), 3.10-3.55 (m, 8H), 2.10 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=421.0 (M+H)+
  • Example 3C
  • 3C was obtained according to a procedure similar to the one described for 3B, with the following exception:
  • Step 3.1: 2.7a (X═H) was replaced by 2.7b (X═N).
  • 1H NMR (400 MHz, DMSO d6) δ 9.02 (brm, 2H), 8.64 (d, 1H), 7.94 (dd, 1H), 7.87 (dd, 1H), 7.66 (d, 1H), 7.52 (d, 1H), 7.16 (d, 1H), 6.19 (s, 1H), 3.48 (q, 2H), 3.25 (brm, 6H), 2.10 (brm, 4H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=422.2 (M+H)+
  • Example 3D
  • 3D was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4a.
  • 1H NMR (400 MHz, DMSO d6) δ 9.33 (m, 2H), 7.83 (m, 2H), 7.54 (m, 1H), 7.42 (m, 4H), 7.22 (m, 1H), 7.10 (m, 1H), 6.01 (s, 1H), 5.60 (m, 2H), 3.42 (m, 2H), 3.25 (m, 4H), 2.11 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=420.0 (M+H)+
  • Elemental analysis:
  • C25H29N3O3, 1HCl, 3H2O
  • Theory: % C, 58.87; % H, 7.11; % N, 8.24.
  • Found: % C, 58.85; % H, 6.74; % N, 8.03.
  • Example 3E Preparation of 3.5a
  • O-Benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (244.2 mg, 0.76 mmol, 1.1 eq) was added to a cooled (0° C.) solution of 3.3a (360.0 mg, 0.69 mmol, 1.0 eq), 3.4b (256.8 mg, 3.80 mmol, 5.5 eq), and N,N-diisopropylethylamine (1.06 mL, 6.08 mmol, 7.7 eq) in acetonitrile (8 mL). The solution was stirred overnight at room temperature and then concentrated under reduced pressure. Ethyl acetate (10 mL) and a saturated aqueous solution of sodium bicarbonate (10 mL) were added to the crude product and the mixture was stirred for 20 min. The phases were separated and the organic phase was washed with an aqueous saturated solution of sodium bicarbonate and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 68%
  • 1H NMR (400 MHz, DMSO d6) δ 8.28 (m, 1H), 7.70 (m, 1H), 7.50 (m, 1H), 7.42 (m, 4H), 7.04 (d, 1H), 5.94 (s, 1H), 3.72 (m, 2H), 3.45 (br s, 2H), 3.25 (m, 4H), 2.70 (d, 3H), 1.89 (m, 2H), 1.74 (m, 2H), 1.42 (s, 9H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=534.3 (M+H)+
  • Preparation of 3E
  • A 2.0M solution of hydrochloric acid in diethyl ether (1.30 mL, 2.57 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 3.5a (0.25 g, 0.47 mmol, 1.0 eq) in anhydrous dichloromethane (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h. Diethyl ether (100 mL) was added to the solution. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.14 (m, 2H), 8.34 (m, 1H), 7.77 (d, 1H), 7.54 (s, 1H), 7.44 (s, 4H), 7.12 (d, 1H), 6.01 (s, 1H), 3.63 (brs, 2H), 3.45 (brs, 2H), 3.24 (m, 4H), 2.69 (d, 3H), 2.09 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=434.3 (M+H)+
  • Elemental analysis:
  • C26H31N3O3, 1HCl, 1.25H2O
  • Theory: % C, 63.40; % H, 7.06; % N, 8.53.
  • Found: % C, 63.13; % H, 6.94; % N, 8.39.
  • Example 3F
  • 3F was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (m, 2H), 8.37 (m, 1H), 7.79 (m, 1H), 7.55 (m, 1H), 7.44 (m, 4H), 7.10 (m, 1H), 6.01 (s, 1H), 3.61 (m, 2H), 3.45 (m, 2H), 3.22 (m, 6H), 2.10 (m, 4H), 1.10 (m, 9H)
  • Mass Spectral Analysis m/z=448.4 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 1HCl, 1H2O
  • Theory: % C, 64.59; % H, 7.23; % N, 8.37.
  • Found: % C, 64.70; % H, 7.16; % N, 8.30.
  • Example 3G
  • 3G was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4d.
  • 1H NMR (400 MHz, DMSO d6) δ 9.16 (m, 2H), 8.36 (m, 1H), 7.78 (m, 1H), 7.55 (m, 1H), 7.44 (m, 4H), 7.10 (m, 1H), 6.00 (s, 1H), 3.44 (m, 2H), 3.20 (m, 8H), 2.10 (m, 4H), 1.45 (m, 2H), 1.12 (m, 6H), 0.80 (m, 3H)
  • Mass Spectral Analysis m/z=462.4 (M+H)+
  • Elemental analysis:
  • C28H35N3O3, 1HCl, 1.5H2O
  • Theory: % C, 64.05; % H, 7.49; % N, 8.00.
  • Found: % C, 63.76; % H, 7.41; % N, 7.76.
  • Example 3H
  • 3H was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4e.
  • 1H NMR (400 MHz, DMSO d6) δ 9.23 (m, 2H), 8.36 (m, 1H), 7.79 (m, 1H), 7.55 (m, 1H), 7.45 (m, 4H), 7.12 (m, 1H), 6.01 (s, 1H), 3.45 (m, 2H), 3.24 (m, 6H), 3.01 (m, 2H), 2.06 (m, 4H), 1.76 (m, 1H), 1.11 (m, 6H), 0.81 (m, 6H)
  • Mass Spectral Analysis m/z=476.5 (M+H)+
  • Elemental analysis:
  • C29H37N3O3, 1HCl, 1.5H2O
  • Theory: % C, 64.61; % H, 7.67; % N, 7.79.
  • Found: % C, 64.94; % H, 7.39; % N, 7.77.
  • Example 3I
  • 3I was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4f.
  • 1H NMR (400 MHz, DMSO d6) δ 9.14 (brs, 2H), 8.23 (m, 1H), 7.80 (m, 1H), 7.54 (m, 1H), 7.44 (m, 4H), 7.11 (m, 1H), 6.02 (s, 1H), 3.45 (m, 2H), 3.23 (m, 6H), 3.01 (m, 2H), 2.10 (m, 4H), 1.12 (m, 6H), 0.83 (m, 9H)
  • Mass Spectral Analysis m/z=490.6 (M+H)+
  • Elemental analysis:
  • C30H39N3O3, 1HCl, 0.75H2O
  • Theory: % C, 66.77; % H, 7.75; % N, 7.79.
  • Found: % C, 66.63; % H, 7.64; % N, 7.77.
  • Example 3J
  • 3J was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4 g.
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (m, 2H), 8.45 (m, 1H), 7.80 (m, 1H), 7.54 (m, 1H), 7.44 (m, 4H), 7.11 (m, 1H), 6.01 (s, 1H), 3.45 (m, 2H), 3.24 (m, 6H), 3.09 (m, 2H), 2.11 (m, 4H), 1.12 (m, 6H), 0.96 (m, 1H), 0.36 (m, 2H), 0.16 (m, 2H)
  • Mass Spectral Analysis m/z=474.4 (M+H)+
  • Elemental analysis:
  • C29H35N3O3, 1HCl, 1.75H2O
  • Theory: % C, 64.31; % H, 7.35; % N, 7.76.
  • Found: % C, 64.69; % H, 7.17; % N, 7.66.
  • Example 3K
  • 3K was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4 h.
  • 1H NMR (400 MHz, DMSO d6) δ 9.36 (m, 2H), 8.13 (m, 1H), 7.82 (m, 1H), 7.54 (m, 1H), 7.44 (m, 4H), 7.11 (m, 1H), 6.00 (s, 1H), 4.01 (m, 1H), 3.45 (m, 2H), 3.22 (m, 6H), 2.10 (m, 4H), 1.15 (m, 12H)
  • Mass Spectral Analysis m/z=462.5 (M+H)+
  • Elemental analysis:
  • C28H35N3O3, 1HCl, 2.25H2O
  • Theory: % C, 62.44; % H, 7.58; % N, 7.80.
  • Found: % C, 62.42; % H, 7.58; % N, 8.08.
  • Example 3L
  • 3L was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4i.
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (m, 2H), 8.34 (m, 1H), 7.78 (m, 1H), 7.54 (m, 1H), 7.44 (m, 4H), 7.11 (m, 1H), 6.00 (s, 1H), 3.45 (m, 2H), 3.20 (m, 8H), 2.08 (m, 4H), 1.45 (m, 2H), 1.25 (m, 4H), 1.11 (m, 6H), 0.84 (m, 3H)
  • Mass Spectral Analysis m/z=490.5 (M+H)+
  • Elemental analysis:
  • C30H39N3O3, 1HCl, 1.5H2O
  • Theory: % C, 65.14; % H, 7.84; % N, 7.60.
  • Found: % C, 65.38; % H, 7.60; % N, 7.64.
  • Example 3M
  • 3M was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4j.
  • 1H NMR (400 MHz, DMSO d6) δ 9.11 (m, 2H), 7.41 (m, 4H), 7.30 (m, 1H), 7.09 (m, 1H), 6.99 (m, 1H), 6.00 (s, 1H), 3.45 (m, 2H), 3.20 (m, 6H), 2.91 (m, 6H), 2.10 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=448.4 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 1HCl, 1.25H2O
  • Theory: % C, 64.02; % H, 7.26; % N, 8.30.
  • Found: % C, 64.03; % H, 7.21; % N, 8.23.
  • Example 3N
  • 3N was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4 k.
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (m, 2H), 7.44 (m, 5H), 7.09 (m, 2H), 5.99 (s, 1H), 3.41 (m, 2H), 3.36 (m, 4H), 3.21 (m, 6H), 2.10 (m, 4H), 1.78 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=474.5 (M+H)+
  • Elemental analysis:
  • C29H35N3O3, 1HCl, 1.25H2O
  • Theory: % C, 65.40; % H, 7.29; % N, 7.89.
  • Found: % C, 65.48; % H, 7.08; % N, 7.90.
  • Example 3O
  • 3O was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4l.
  • 1H NMR (400 MHz, DMSO d6) δ 9.03 (brs, 2H), 7.44 (m, 5H), 7.13 (m, 2H), 6.01 (s, 1H), 4.96 (m, 1H), 4.24 (m, 1H), 3.44 (m, 6H), 3.22 (m, 6H), 2.09 (m, 4H), 1.86 (m, 1H), 1.75 (m, 1H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=490.3 (M+H)+
  • Example 3P
  • 3P was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4m.
  • 1H NMR (400 MHz, DMSO d6) δ 9.25 (m, 2H), 7.44 (m, 5H), 7.10 (m, 2H), 6.00 (s, 1H), 4.93 (m, 1H), 4.24 (m, 1H), 3.45 (m, 6H), 3.21 (m, 6H), 2.11 (m, 4H), 1.88 (m, 1H), 1.76 (m, 1H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=490.5 (M+H)+
  • Elemental analysis:
  • C29H35N3O4, 1HCl, 1.5H2O
  • Theory: % C, 62.98; % H, 7.11; % N, 7.60.
  • Found: % C, 62.79; % H, 6.98; % N, 7.58.
  • Example 3Q
  • 3Q was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4n.
  • 1H NMR (400 MHz, DMSO d6) δ 9.25 (m, 2H), 7.40 (m, 5H), 7.09 (m, 1H), 6.99 (m, 1H), 6.01 (s, 1H), 4.10 (m, 2H), 3.45 (m, 2H), 3.25 (m, 6H), 2.11 (m, 6H), 2.51 (m, 2H), 1.19 (m, 9H), 0.80 (m, 3H)
  • Mass Spectral Analysis m/z=502.5 (M+H)+
  • Elemental analysis:
  • C31H39N3O3, 1HCl, 2H2O
  • Theory: % C, 64.85; % H, 7.72; % N, 7.32.
  • Found: % C, 64.54; % H, 7.37; % N, 7.35.
  • Example 3R
  • 3R was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 1.12.
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (m, 2H), 7.41 (m, 4H), 7.29 (m, 1H), 7.08 (m, 1H), 6.89 (m, 1H), 5.98 (s, 1H), 3.41 (m, 2H), 3.22 (m, 10H), 2.10 (m, 4H), 1.02 (m, 12H)
  • Mass Spectral Analysis m/z=476.5 (M+H)+
  • Elemental analysis:
  • C29H37N3O3, 1HCl, 1.25H2O
  • Theory: % C, 65.15; % H, 7.64; % N, 7.86.
  • Found: % C, 64.85; % H, 7.26; % N, 7.79.
  • Example 3S
  • 3S was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4o.
  • 1H NMR (400 MHz, DMSO d6) δ 8.67 (m, 1H), 8.55 (m, 1H), 7.43 (m, 4H), 7.22 (m, 1H), 7.09 (m, 1H), 6.82 (m, 1H), 6.01 (s, 1H), 3.66 (m, 2H), 3.44 (m, 2H), 3.23 (m, 6H), 2.10 (m, 2H), 1.98 (m, 2H), 1.16 (m, 18H)
  • Mass Spectral Analysis m/z=504.4 (M+H)+
  • Example 3T
  • 3T was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4p.
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (m, 1.3H), 7.45 (s, 4H), 7.32 (d, 1H), 7.10 (d, 1H), 7.00 (s, 1H), 6.00 (s, 1H), 4.10 (m, 4H), 3.35-3.60 (m, 8H), 3.20 (m, 4H), 2.10 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=490.1 (M+H)+
  • Example 3U
  • 3U was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.4b was replaced by 3.4q.
  • 1H NMR (400 MHz, DMSO d6) δ 9.23 (brs, 2H), 7.44 (m, 4H), 7.30 (m, 1H), 7.12 (m, 1H), 6.96 (m, 1H), 6.01 (s, 1H), 3.40 (m, 6H), 3.22 (m, 6H), 2.11 (m, 4H), 1.56 (m, 2H), 1.43 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=488.4 (M+H)+
  • Elemental analysis:
  • C30H37N3O3, 1HCl, 1.75H2O
  • Theory: % C, 64.85; % H, 7.53; % N, 7.56.
  • Found: % C, 64.99; % H, 7.37; % N, 7.46.
  • Example 3V
  • 3V was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a (X═CH) was replaced by 3.3b (X═N).
  • Step 3.5: 3.4b was replaced by 3.4a.
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (brm, 2H), 8.63 (d, 1H), 7.92 (m, 2H), 7.83 (dd, 1H), 7.64 (d, 1H), 7.53 (d, 1H), 7.25 (brs, 1H), 7.12 (d, 1H), 6.16 (s, 1H), 3.48 (q, 2H), 3.31 (q, 2H), 3.22 (brm, 4H), 2.10 (brm, 4H), 1.18 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=421.3 (M+H)+
  • Elemental analysis:
  • C24H28N4O3, 1.6HCl, 1.4H2O
  • Theory: % C, 57.19; % H, 6.48; % N, 11.12; % Cl, 11.25.
  • Found: % C, 57.14; % H, 6.41; % N, 10.98; % Cl, 11.00.
  • Example 3W
  • 3W was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.3a was replaced by 3.3b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (brm, 2H), 8.63 (d, 1H), 8.36 (m, 1H), 7.93 (dd, 1H), 7.79 (dd, 1H), 7.64 (d, 1H), 7.49 (d, 1H), 7.13 (d, 1H), 6.16 (s, 1H), 3.48 (q, 2H), 3.25 (brm, 6H), 2.71 (d, 3H), 2.10 (m, 4H), 1.18 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=435.3 (M+H)+
  • Elemental analysis:
  • C25H30N4O3, 1.8HCl, 2H2O
  • Theory: % C, 56.00; % H, 6.73; % N, 10.45; % Cl, 11.90.
  • Found: % C, 56.16; % H, 6.72; % N, 10.47; % Cl, 12.23.
  • Example 3X
  • 3X was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a was replaced by 3.3b.
  • Step 3.5: 3.4b was replaced by 3.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.23 (brm, 2H), 8.63 (d, 1H), 8.40 (t, 1H), 7.93 (dd, 1H), 7.81 (dd, 1H), 7.64 (d, 1H), 7.49 (d, 1H), 7.13 (d, 1H), 6.16 (s, 1H), 3.48 (q, 2H), 3.25 (brm, 8H), 2.10 (brm, 4H), 1.18 (t, 3H), 1.12 (t, 3H), 1.05 (t, 3H)
  • Mass Spectral Analysis m/z=449.3 (M+H)+
  • Elemental analysis:
  • C26H32N4O3, 2HCl, 1.5H2O
  • Theory: % C, 56.93; % H, 6.80; % N, 10.21; % Cl, 12.93.
  • Found: % C, 56.64; % H, 6.86; % N, 10.13; % Cl, 12.59.
  • Example 3Y
  • 3Y was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a was replaced by 3.3b.
  • Step 3.5: 3.4b was replaced by 3.4j.
  • 1H NMR (400 MHz, DMSO d6) δ 9.06 (brs, 2H), 8.62 (d, 1H), 7.92 (dd, 1H), 7.63 (d, 1H), 7.36 (dd, 1H), 7.11 (d, 1H), 6.98 (d, 1H), 6.16 (s, 1H), 3.47 (q, 2H), 3.25 (brm, 6H), 2.91 (s, 6H), 2.10 (brm, 4H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=449.3 (M+H)+
  • Elemental analysis:
  • C26H32N4O3, 1.75HCl, 1.25H2O
  • Theory: % C, 58.38; % H, 6.83; % N, 10.47; % Cl, 11.60.
  • Found: % C, 58.37; % H, 6.94; % N, 10.21; % Cl, 11.35.
  • Example 3Z
  • 3Z was obtained according to a procedure similar to the one described for 3AC, with the following exception:
  • Step 3.8: 3.6d was replaced by 3.6a; tetrakis(triphenylphosphine)palladium(0) was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (brm, 2H), 9.01 (s, 1H), 8.73 (d, 1H), 8.47 (d, 1H), 7.87 (m, 1H), 7.76 (dd, 1H), 7.53 (d, 2H), 7.44 (d, 2H), 7.38 (d, 1H), 7.28 (d, 1H), 6.07 (s, 1H), 3.44 (m, 2H), 3.23 (brm, 6H), 2.11 (brm, 4H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=454.0 (M+H)+
  • Example 3AA
  • 3AA was obtained according to a procedure similar to the one described for 3AC, with the following exception:
  • Step 3.8: 3.6d was replaced by 3.6b; tetrakis(triphenylphosphine)palladium(0) was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.84 (brm, 2H), 7.58 (dd, 1H), 7.46 (m, 5H), 7.27 (d, 1H), 7.18 (d, 1H), 7.12 (d, 1H), 7.06 (m, 1H), 6.04 (s, 1H), 3.46 (m, 2H), 3.23 (brm, 6H), 2.13 (m, 2H), 2.01 (m, 2H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=459.3 (M+H)+
  • Elemental analysis:
  • C28H30N2O2S, 1HCl, 0.5H2O
  • Theory: % C, 66.71; % H, 6.40; % N, 5.56; % Cl, 7.03.
  • Found: % C, 66.76; % H, 6.27; % N, 5.50; % Cl, 7.34.
  • Example 3AB
  • 3AB was obtained according to a procedure similar to the one described for 3AC, with the following exception:
  • Step 3.8: 3.6d was replaced by 3.6c; tetrakis(triphenylphosphine)palladium(0) was used.
  • 1H NMR (400 MHz, DMSO-d6) δ 9.39 (b, 1H), 9.32 (b, 1H), 8.83 (d, 2H), 8.16 (d, 2H), 7.98 (d, 1H), 7.49 (m, 3H), 7.46 (d, 2H), 7.34 (d, 1H), 6.14 (s, 1H), 3.3-3.7 (m, 8H), 2.12 (m, 4H), 1.05-1.2 (b, 6H)
  • Mass Spectral Analysis m/z=454.4 (M+H)+
  • Elemental analysis:
  • C29H33Cl2N3O2, 2HCl, 2.75H2O
  • Theory: % C, 60.47; % H, 6.74; % N, 7.29.
  • Found: % C, 60.35; % H, 6.46; % N, 7.32.
  • Example 3AC Preparation of 3.7a
  • To a solution of 3.1a (1.50 g, 2.40 mmol, 1.0 eq) in dimethoxyethane (DME) (20 mL) was added sequentially a 2N aqueous solution of sodium carbonate (3.6 mL, 7.20 mmol, 3.0 eq), lithium chloride (0.305 g, 7.20 mmol, 3.0 eq), 3.6d (0.357 g, 2.88 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (0.277 g, 0.24 mmol, 0.10 eq). The mixture was heated at 120° C. for 16 h. After this time, only starting material 3.1a was observed by LC/MS. Therefore, additional quantities of 3.6d (0.10 g, 0.81 mmol, 0.34 eq), tetrakis(triphenylphosphine)palladium(0) (0.10 g, 0.087 mmol, 0.036 eq) and [1,1′-bis(diphenylphosphino)ferrocene palladium (II) chloride, dichloromethane complex] (0.50 g, 0.68 mmol, 0.28 eq) were added to the reaction mixture, which was heated at 120° C. for 5 h. The crude mixture was cooled to room temperature, dissolved in ethyl acetate and the mixture was washed with water. The organic extract was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity), and the product was used without further purification. Yield: 20%
  • Mass Spectral Analysis m/z=555.5 (M+H)+
  • Preparation of 3AC
  • To a solution of 3.7a (0.3 g, purity: 90%, 0.489 mmol, 1.0 eq) in methylene chloride (10 mL) was added an excess of a 1.0M solution of anhydrous hydrochloric acid in diethyl ether (10 mL). The mixture was stirred for 16 h at room temperature, concentrated under reduced pressure and purified by column chromatography (eluent: methylene chloride/methanol mixtures of increasing polarity). Yield: 90%
  • 1H NMR (400 MHz, DMSO d6) δ 9.26 (brs, 2H), 9.13 (s, 1H), 8.99 (s, 2H), 7.72 (d, 1H), 7.53 (d, 2H), 7.44 (d, 2H), 7.34 (s, 1H), 7.25 (d, 1H), 6.07 (s, 1H), 3.44 (brs, 2H), 3.23 (brm, 6H), 2.12 (brm, 4H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=455.4 (M+H)+
  • Elemental analysis:
  • C28H30N4O2, 2HCl, 2.75H2O
  • Theory: % C, 58.28; % H, 6.55; % N, 9.71.
  • Found: % C, 58.53; % H, 6.27; % N, 9.74.
  • Example 4A Preparation of 4.2
  • To a suspension of 1A (21.9 g, 52.45 mmol, 1.0 eq) in tetrahydrofuran (200 mL) at 0° C. was added triethylamine (18.3 mL, 131 mmol, 2.5 eq), followed by trifluoroacetic anhydride (4.1) (8.75 ml, 63 mmol, 1.2 eq) dropwise. The reaction mixture was slowly warmed up to and stirred at room temperature for 10 h. Ethyl acetate (500 mL) was added and the organic layer was washed with a 1M aqueous solution of hydrochloric acid (5×100 mL) and brine, dried over sodium sulfate and filtered. The crude product was concentrated under reduced pressure and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 93%
  • 1H NMR (400 MHz, CDCl3) δ 7.42 (m, 2H), 7.36 (m, 2H), 7.22 (m, 1H), 7.02 (m, 1H), 6.96 (m, 1H), 6.90 (m, 1H), 5.54 (s, 1H), 4.39 (m, 1H), 3.87 (m, 1H), 3.71 (m, 1H), 3.58 (m, 2H), 3.35 (m, 3H), 2.22 (m, 2H), 1.74 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=473.3 (M+H)+
  • Preparation of 4.4
  • To a solution of 4.2 (4.0 g, 8.47 mmol, 1.0 eq) in dry dichloroethane (100 mL) was added sulfur trioxide N,N-dimethylformamide complex (4.3) (1.98 g, 12.9 mmol, 1.5 eq) portionwise. The reaction mixture was heated under reflux for 10 h and then cooled down to 0-10° C. at which point oxalyl chloride (1.2 mL, 13.55 mmol, 1.6 eq) was added drop wise. The reaction mixture was then stirred at 70° C. for another 3 h. The reaction was quenched with ice/water (100 mL). Dichloromethane (100 mL) was added and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 79%
  • 1H NMR (400 MHz, CDCl3) δ 7.90 (dd, 1H), 7.72 (d, 1H), 7.49 (m, 2H), 7.36 (m, 2H), 7.13 (d, 1H), 5.68 (s, 1H), 4.44 (m, 1H), 3.92 (m, 1H), 3.70 (m, 1H), 3.58 (m, 2H), 3.35 (m, 3H), 2.25 (m, 2H), 1.83 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=571.2 (M+H)+
  • Preparation of 4.6a
  • To a solution of 4.4 (0.7 g, 1.22 mmol, 1.0 eq) in dry dichloromethane (30 mL) at 0° C. was added triethylamine (0.85 mL, 6.10 mmol, 5.0 eq) and methylamine (3.4b) hydrochloride salt (0.25 g, 3.66 mmol, 3.0 eq) in one portion. The reaction mixture was slowly warmed up to room temperature and stirred at room temperature for 10 h. Water (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 86%
  • 1H NMR (400 MHz, CDCl3) δ 7.73 (dd, 1H), 7.53 (d, 1H), 7.45 (m, 2H), 7.35 (m, 2H), 7.07 (d, 1H), 5.63 (s, 1H), 4.42 (m, 1H), 4.29 (q, 1H), 3.90 (m, 1H), 3.69 (m, 1H), 3.58 (m, 2H), 3.35 (m, 3H), 2.63 (d, 3H), 2.22 (m, 2H), 1.79 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=566.2 (M+H)+
  • Preparation of 4A
  • To a solution of 4.6a (0.63 g, 1.11 mmol, 1.0 eq) in a mixture of methanol (20 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.92 g, 6.66 mmol, 6.0 eq) portionwise. The reaction mixture was warmed up to room temperature and stirred at room temperature for 10 h. Brine (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added a 2.0M solution of hydrogen chloride in diethyl ether (1.11 mL, 2.22 mmol, 2 eq) drop wise. The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under reduced pressure. Yield: 85%
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (m, 2H), 7.66 (dd, 1H), 7.49-7.37 (m, 6H), 7.25 (d, 1H), 6.10 (s, 1H), 3.45 (m, 2H), 3.22 (m, 6H), 2.36 (d, 3H), 2.01 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=470.2 (M+H)+
  • Elemental analysis:
  • C25H31N3O4S, 1HCl, 1.5H2O
  • Theory: % C, 56.33; % H, 6.62; % N, 7.88.
  • Found: % C, 56.06; % H, 6.50; % N, 8.18.
  • Example 4B
  • 4B was obtained according to a procedure similar to the one described for 4A, with the following exception:
  • Step 4.3: 3.4b was replaced by 3.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (brs, 1H), 7.67 (dd, 1H), 7.46 (m, 4H), 7.39 (d, 1H), 7.23 (d, 1H), 6.10 (s, 1H), 3.52-3.15 (m, 9H), 2.71 (m, 2H), 2.08 (m, 4H), 1.42 (m, 6H), 0.94 (t, 3H)
  • Mass Spectral Analysis m/z=484.3 (M+H)+
  • Elemental analysis:
  • C26H33N3O4S, 1HCl, 1.25H2O
  • Theory: % C, 57.55; % H, 6.78; % N, 7.74.
  • Found: % C, 57.61; % H, 6.75; % N, 7.60.
  • Example 4C
  • 4C was obtained according to a procedure similar to the one described for 4A, with the following exception:
  • Step 4.3: 3.4b was replaced by 3.4d.
  • 1H NMR (400 MHz, DMSO d6) δ 8.85 (m, 2H), 7.67 (dd, 1H), 7.51 (t, 1H), 7.45 (m, 3H), 7.39 (d, 1H), 7.23 (d, 1H), 6.10 (s, 1H), 3.45 (m, 2H), 3.24 (m, 7H), 2.63 (m, 2H), 2.08 (m, 4H), 1.34 (m, 2H), 1.12 (m, 6H), 0.77 (t, 3H)
  • Mass Spectral Analysis m/z=498.3 (M+H)+
  • Elemental analysis:
  • C27H35N3O4S, 1HCl, 1H2O
  • Theory: % C, 58.74; % H, 6.94; % N, 7.61.
  • Found: % C, 58.82; % H, 6.78; % N, 7.56.
  • Example 4D
  • 4D was obtained according to a procedure similar to the one described for 4A, with the following exception:
  • Step 4.3: 3.4b was replaced by 3.4 g.
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (m, 2H), 7.68 (m, 2H), 7.45 (m, 3H), 7.40 (d, 1H), 7.22 (d, 1H), 6.09 (s, 1H), 3.45 (m, 2H), 3.24 (m, 7H), 2.59 (t, 2H), 2.07 (m, 4H), 1.12 (m, 6H), 0.75 (m, 1H), 0.32 (m, 2H), 0.04 (m, 2H)
  • Mass Spectral Analysis m/z=510.3 (M+H)+
  • Elemental analysis:
  • C28H33N3O4S, 1HCl, 1H2O
  • Theory: % C, 59.61; % H, 6.79; % N, 7.45.
  • Found: % C, 59.55; % H, 6.75; % N, 7.40.
  • Example 4E
  • 4E was obtained according to a procedure similar to the one described for 4A, with the following exception:
  • Step 4.3: 3.4b was replaced by 3.4 h.
  • 1H NMR (400 MHz, DMSO d6) δ 8.79 (m, 2H), 7.69 (dd, 1H), 7.54 (d, 1H), 7.44 (m, 4H), 7.22 (d, 1H), 6.10 (s, 1H), 3.51-3.09 (m, 10H), 2.07 (m, 4H), 1.12 (m, 6H), 0.92 (d, 6H)
  • Mass Spectral Analysis m/z=498.3 (M+H)+
  • Elemental analysis:
  • C27H35N3O4S, 1HCl, 1.4H2O
  • Theory: % C, 57.98; % H, 6.99; % N, 7.51. Found: % C, 57.99; % H, 7.04; % N, 7.38.
  • Example 4F
  • 4F was obtained according to a procedure similar to the one described for 4A, with the following exception:
  • Step 4.3: 3.4b was replaced by 3.4j.
  • 1H NMR (400 MHz, DMSO d6) δ 9.11 (m, 2H), 7.64 (dd, 1H), 7.46 (m, 4H), 7.29 (d, 1H), 7.24 (d, 1H), 6.13 (s, 1H), 3.45 (m, 2H), 3.23 (m, 6H), 2.56 (s, 6H), 2.11 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=484.1 (M+H)+
  • Elemental analysis:
  • C26H33N3O4S, 1HCl, 2.75H2O
  • Theory: % C, 54.82; % H, 6.99; % N, 7.38.
  • Found: % C, 54.66; % H, 6.89; % N, 7.30.
  • Example 4G
  • 4G was obtained according to a procedure similar to the one described for 4A, with the following exception:
  • Step 4.3: 3.4b was replaced by 4.5.
  • 1H NMR (400 MHz, DMSO d6) δ 8.85 (m, 2H), 7.83 (d, 1H), 7.69 (dd, 1H), 7.45 (m, 3H), 7.41 (d, 1H), 7.25 (d, 1H), 6.11 (s, 1H), 3.45 (m, 2H), 3.25 (m, 7H), 2.09 (m, 5H), 1.12 (m, 6H), 0.45 (m, 2H), 0.34 (m, 2H)
  • Mass Spectral Analysis m/z=496.2 (M+H)+
  • Elemental analysis:
  • C27H33N3O4S, 1HCl, 0.75H2O
  • Theory: % C, 59.44; % H, 6.56; % N, 7.70.
  • Found: % C, 59.37; % H, 6.46; % N, 7.60.
  • Example 4H Preparation of 4H
  • To a solution of 4.4 (1.5 g, 2.82 mmol) in acetonitrile (20 mL) was added a concentrated aqueous solution of ammonium hydroxide (28-35%, 20 mL). The reaction mixture was heated under reflux for 10 h. Brine (100 mL) was added and the aqueous phase was adjusted to pH=10 with a 1M aqueous solution of sodium hydroxide. Chloroform (150 mL) was added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil (0.32 g, 0.70 mmol, 1.0 eq) in dichloromethane/methanol was added drop wise a 2.0M solution of hydrogen chloride in diethyl ether (0.7 mL, 1.4 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum.
  • Yield: 80%
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (m, 1.5H), 7.71 (dd, 1H), 7.45 (m, 5H), 7.27 (s, 2H), 7.22 (d, 1H), 6.09 (s, 1H), 3.46 (m, 2H), 3.23 (m, 6H), 2.07 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=456.0 (M+H)+
  • Elemental analysis:
  • C24H29N3O4S, 1HCl, 2H2O
  • Theory: % C, 54.59; % H, 6.49; % N, 7.96.
  • Found: % C, 54.50; % H, 6.49; % N, 7.82.
  • Example 4I Preparation of 4.8
  • To a suspension of 4H (1.12 g, 2.45 mmol, 1.0 eq) in a mixture of dichloromethane (50 mL) and methanol (5 mL) at 0° C. was added sequentially triethylamine (0.85 mL, 6.12 mmol, 2.5 eq), and di-tert-butyl dicarbonate 4.7 (0.80 g, 3.67 mmol, 1.5 eq) portion wise. The reaction mixture was slowly warmed to room temperature and stirred at room temperature for 10 h. The solvents were removed under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 92%
  • 1H NMR (400 MHz, CDCl3) δ 7.75 (dd, 1H), 7.57 (d, 1H), 7.43 (m, 2H), 7.35 (m, 2H), 7.03 (d, 1H), 5.65 (s, 1H), 4.83 (s, 2H), 3.89 (m, 2H), 3.57 (m, 2H), 3.32 (m, 4H), 2.04 (m, 2H), 1.71 (m, 2H), 1.47 (s, 9H), 1.21 (m, 6H)
  • Mass Spectral Analysis m/z=556.3 (M+H)+
  • Preparation of 4.10
  • To a solution of 4.8 (1.25 g, 2.25 mmol, 1.0 eq) in dichloromethane (40 mL) was added triethylamine (0.94 mL, 6.75 mmol, 3.0 eq), and acetic anhydride (4.9) (0.64 mL, 6.75 mmol, 3.0 eq) drop wise. The mixture was stirred at room temperature for 10 h. Dichloromethane (100 mL) and water (100 mL) were added to the reaction mixture and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 70%
  • Mass Spectral Analysis m/z=598.3 (M+H)+
  • Preparation of 4I
  • To a solution of 4.10 (0.16 g, 0.27 mmol, 1.0 eq) in dichloromethane (5 mL) was added iodotrimethylsilane (0.06 mL, 0.43 mmol, 1.6 eq) dropwise. The mixture was stirred at room temperature for 30 min. The mixture was diluted in chloroform (100 mL) and methanol (5 mL), washed with a 20% aqueous solution of sodium thiosulfate (2×30 mL) and a 1M aqueous solution of sodium carbonate (2×30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 7.73 (dd, 1H), 7.51 (d, 1H), 7.45 (s, 4H), 7.17 (d, 1H), 6.01 (s, 1H), 3.45 (brs, 2H), 3.38-3.15 (m, 7H), 2.07 (m, 4H), 1.79 (s, 3H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=498.3 (M+H)+
  • Example 5A Preparation of 5.2
  • To a solution of 4.4 (1.4 g, 2.45 mmol, 1.0 eq) in a mixture tetrahydrofuran (5 mL) and dichloromethane (1 mL) at 0° C. was added a 1.0 M solution of hydrazine (5.1) in tetrahydrofuran (24.5 mL, 24.5 mmol, 10.0 eq) in one portion. The reaction mixture was stirred at 0° C. for 30 min Water (50 mL) and chloroform (100 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 70%
  • 1H NMR (400 MHz, CDCl3) δ 7.78 (dd, 1H), 7.59 (d, 1H), 7.46 (d, 2H), 7.35 (d, 2H), 7.10 (d, 1H), 5.64 (s, 1H), 4.42 (m, 1H), 3.91 (m, 1H), 3.69 (m, 1H), 3.57 (m, 2H), 3.35 (m, 4H), 2.23 (m, 2H), 1.80 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=567.4 (M+H)+
  • Preparation of 5.3
  • To a suspension of 5.2 (0.9 g, 1.59 mmol, 1.0 eq) in ethanol (10 mL) was added sodium acetate (0.87 g, 10.8 mmol, 6.65 eq) and iodomethane (2.8c) (0.54 mL, 8.85 mmol, 5.45 eq). The mixture was heated under reflux for 10 h. Water (100 mL) and dichloromethane (100 mL) were added and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 74%
  • 1H NMR (400 MHz, CDCl3) δ 7.81 (dd, 1H), 7.64 (d, 1H), 7.46 (d, 2H), 7.35 (d, 2H), 7.11 (d, 1H), 5.64 (s, 1H), 4.42 (m, 1H), 3.91 (m, 1H), 3.69 (m, 1H), 3.57 (m, 2H), 3.35 (m, 3H), 3.00 (s, 3H), 2.23 (m, 2H), 1.80 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=551.2 (M+H)+
  • Preparation of 5A
  • To a solution of 5.3 (0.65 g, 1.18 mmol, 1.0 eq) in a mixture of methanol (20 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.98 g, 7.08 mmol, 6.0 eq) portion wise. The mixture was warmed up to and stirred at room temperature for 10 h. Brine (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added dropwise a 2.0M solution of hydrogen chloride in diethyl ether (1.18 mL, 2.36 mmol, 2.0 eq). The mixture was then stirred at room temperature for 1 h, concentrated under reduced pressure, and dried under vacuum. Yield: 88%
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (m, 2H), 7.83 (dd, 1H), 7.47 (m, 5H), 7.30 (d, 1H), 6.12 (s, 1H), 3.63-3.10 (m, 11H), 2.10 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=455.2 (M+H)+
  • Elemental analysis: C25H30N2O4S, 1HCl, 1.33H2O
  • Theory: % C, 58.30; % H, 6.59; % N, 5.44. Found: % C, 58.35; % H, 6.56; % N, 5.37.
  • Example 6A Preparation of 6.2
  • To a cold (0° C.) solution of 4.2 (0.23 g, 0.48 mmol, 1.0 eq) in dry acetonitrile (3 mL) under nitrogen was added nitronium tetrafluoroborate complex (6.1) (78.5 mg, 0.576 mmol, 1.2 eq) in one portion with rapid stiffing. The reaction mixture was kept at 0° C. for 1 h and then quenched with ice/water (1:1) (15 mL). Dichloromethane (50 mL) was added and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×30 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 38%
  • 1H NMR (400 MHz, CDCl3) δ 8.14 (dd, 1H), 7.97 (d, 1H), 7.48 (m, 2H), 7.36 (m, 2H), 7.06 (d, 1H), 5.66 (s, 1H), 4.43 (m, 1H), 3.92 (m, 1H), 3.70 (m, 1H), 3.58 (m, 2H), 3.36 (m, 3H), 2.23 (m, 2H), 1.82 (m, 2H), 1.23 (m, 6H)
  • Mass Spectral Analysis m/z=518.3 (M+H)+
  • Preparation of 6A
  • To a solution of 6.2 (0.2 g, 0.386 mmol, 1.0 eq) in a mixture of methanol (15 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.32 g, 2.32 mmol, 6.0 eq) portionwise. The mixture was warmed up to room temperature and stirred at room temperature for 10 h. Brine (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×30 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined and concentrated under reduced pressure. The product was dissolved in chloroform (100 mL), washed with a 1M aqueous solution of sodium carbonate (2×30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added dropwise a 1.0M solution of hydrogen chloride in diethyl ether (0.8 mL, 0.8 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum.
  • Yield: 50%
  • 1H NMR (400 MHz, DMSO d6) δ 9.01 (m, 2H), 8.19 (dd, 1H), 7.79 (d, 1H), 7.49 (m, 4H), 7.29 (d, 1H), 6.19 (s, 1H), 3.56-3.14 (m, 8H), 2.11 (m, 4H), 1.13 (m, 6H)
  • Mass Spectral Analysis m/z=422.3 (M+H)+
  • Example 6B Preparation of 6.4
  • To a cold (0° C.) solution of 6.2 (1.92 g, 3.71 mmol, 1.0 eq) in ethanol (50 mL) was added tin (II) chloride dihydrate (6.3) (2.51 g, 11.13 mmol, 3.0 eq) in one portion. The reaction mixture was heated under reflux for 10 h and then concentrated under reduced pressure to give the crude product, which was used for the next step without purification.
  • Mass Spectral Analysis m/z=488.2 (M+H)+
  • Preparation of 6B
  • To a suspension of 6.4 (1.3 g, crude, as of 0.91 mmol, 1.0 eq) in a mixture of methanol (30 mL) and water (10 mL) at 0° C. was added potassium carbonate (0.75 g, 5.46 mmol, 6.0 eq) portion wise. The reaction mixture was warmed up to room temperature and stirred at room temperature for 10 h. Brine (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×30 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined, concentrated under reduced pressure, and dried under vacuum.
  • Yield: 27% over two steps
  • 1H NMR (400 MHz, DMSO d6) δ 9.98 (brs, 2.5H), 9.11 (m, 2H), 7.44 (m, 4H), 7.23 (dd, 1H), 7.15 (d, 1H), 7.00 (d, 1H), 6.06 (s, 1H), 3.78-3.10 (m, 8H), 2.06 (m, 4H), 1.12 (m, 6H) Mass Spectral Analysis m/z=392.2 (M+H)+
  • Example 6C Preparation of 6.6a
  • To a suspension of 6.4 (1.5 g, crude, as of 1.05 mmol, 1.0 eq) in dichloroethane (50 mL) at 0° C. was added pyridine (0.42 g, 5.25 mmol, 5 eq) followed by drop wise addition of ethylsulfonyl chloride (6.5a) (0.30 mL, 3.15 mmol, 3.0 eq) dropwise. The mixture was stirred at 0° C. for another 2 h. A 1M aqueous solution of hydrochloric acid (100 mL) and chloroform (100 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 90%
  • Mass Spectral Analysis m/z=580.3 (M+H)+
  • Preparation of 6C
  • To a solution of 6.6a (0.55 g, 0.9 mmol, 1.0 eq) in a mixture of methanol (20 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.78 g, 5.4 mmol, 6.0 eq) portionwise. The mixture was warmed up to room temperature and stirred at room temperature for 10 h. Brine (100 mL) and chloroform (100 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added drop wise a 1.0M solution of hydrogen chloride in diethyl ether (1.8 mL, 1.8 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 80%
  • 1H NMR (400 MHz, DMSO d6) δ 9.49 (s, 1H), 8.91 (m, 2H), 7.43 (m, 4H), 7.11 (dd, 1H), 7.02 (d, 1H), 6.93 (d, 1H), 6.00 (s, 1H), 3.45 (brs, 2H), 3.21 (m, 6H), 2.97 (q, 2H), 2.03 (m, 4H), 1.20-1.00 (m, 9H)
  • Mass Spectral Analysis m/z=484.2 (M+H)+
  • Elemental analysis:
  • C26H33N3O4S, 1HCl, 1.25H2O
  • Theory: % C, 57.55; % H, 6.78; % N, 7.74. Found: % C, 57.52; % H, 6.67; % N, 7.73.
  • Example 6D
  • 6D was obtained according to a procedure similar to the one described for 6C, with the following exception:
  • Step 6.5: 6.5a was replaced by 6.5b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.48 (s, 1H), 8.66 (brm, 1H), 7.43 (s, 4H), 7.12 (dd, 1H), 7.01 (d, 1H), 6.95 (d, 1H), 6.00 (s, 1H), 3.46 (brs, 4H), 3.23 (brm, 4H), 3.12 (m, 1H), 2.06 (m, 2H), 1.95 (m, 2H), 1.20 (d, 6H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=498.2 (M+H)+
  • Example 6E Preparation of 6.8
  • To a suspension of 6.4 (1.0 g, crude, as of 0.58 mmol, 1.0 eq) in dichloroethane (30 mL) at 0° C. was added pyridine (0.23 mL, 2.9 mmol, 5.0 eq) followed by a drop wise addition of acetyl chloride (6.7) (0.16 mL, 2.32 mmol, 4.0 eq). The reaction mixture was slowly warmed up to room temperature and stirred at room temperature for 10 h. A 1M aqueous solution of hydrochloric acid (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixture of increasing polarity). Yield: 88%
  • Mass Spectral Analysis m/z=530.2 (M+H)+
  • Preparation of 6E
  • To a solution of 6.8 (0.27 g, 0.5 mmol, 1.0 eq) in a mixture of methanol (20 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.42 g, 3.0 mmol, 6.0 eq) portion wise. The reaction mixture was warmed up to room temperature and stirred at room temperature for 10 h. Brine (100 mL) and chloroform (100 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×30 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was first purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity) and then repurified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined and concentrated under reduced pressure. The product was dissolved in chloroform (100 mL) and washed with a 1M solution of sodium carbonate (2×30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added dropwise 1.0M hydrogen chloride in diethyl ether (1.0 mL, 1.0 mmol, 2 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under reduced pressure. Yield: 73%
  • 1H NMR (400 MHz, DMSO d6) δ 9.34 (s, 1H), 8.80 (brs, 2H), 7.68 (d, 1H), 7.42 (s, 4H), 6.90 (t, 1H), 6.77 (d, 1H), 5.95 (s, 1H), 3.45 (brs, 2H), 3.25 (m, 6H), 2.15 (s, 3H), 2.04 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=434.2 (M+H)+
  • Elemental analysis:
  • C26H31N3O3, 1HCl, 1.7H2O
  • Theory: % C, 62.38; % H, 7.13; % N, 8.39.
  • Found: % C, 62.26; % H, 6.81; % N, 8.29.
  • Example 7A Preparation of 7.2
  • To a solution of 3.1a (3 g, 4.80 mmol, 1.0 eq), sodium tert-butoxide (0.55 g, 5.67 mmol, 1.18 eq), tris(dibenzylideneacetone)dipalladium(0) (0.22 g, 0.24 mmol, 0.05 eq) and 1,1′-bis(diphenylphosphino)ferrocene (dppf) (0.39 g, 0.70 mmol, 0.145 eq) in anhydrous toluene (48 mL) was added 7.1 (0.95 mL, 5.67 mmol, 1.18 eq) at room temperature. The solution was stirred at 80° C. overnight and then cooled to room temperature. The mixture was diluted with ethyl acetate and vacuum filtered through a plug of celite. The filtrate was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 33%
  • Mass Spectral Analysis m/z=656.6 (M+H)+
  • Preparation of 7.3
  • To a solution of 7.2 (1.00 g, 1.52 mmol, 1.0 eq) in anhydrous methanol (5 mL) at room temperature under nitrogen was added hydroxylamine hydrochloride (0.21 g, 2.97 mmol, 1.95 eq) and sodium acetate (0.64 g, 7.78 mmol, 5.1 eq). The mixture was stirred overnight at room temperature. The mixture was then diluted with ethyl acetate, washed with a saturated aqueous solution of sodium bicarbonate and brine, dried over sodium sulfate and filtered. The organics were concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 99%
  • Mass Spectral Analysis m/z=492.5 (M+H)+
  • Preparation of 7.5
  • To a solution of 7.3 (0.75 g, 1.53 mmol, 1.0 eq) and triethylamine (1.06 mL, 7.63 mmol, 5.0 eq) in dichloromethane (10 mL) at 0° C. under nitrogen was added drop wise 7.4 (0.35 mL, 4.58 mmol, 3.0 eq). The mixture was stirred overnight at room temperature. An aqueous solution of sodium bicarbonate was added and the mixture was stirred for 20 min. The phases were separated and the organic phase was washed with an aqueous solution of sodium bicarbonate, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 83%
  • Mass Spectral Analysis m/z=648.5 (M+H)+
  • Preparation of 7.6
  • To a solution of 7.5 (0.82 g, 1.27 mmol, 1.0 eq) in tetrahydrofuran (5 mL) and methanol (5 mL) was added a 1N aqueous solution of sodium hydroxide (5 mL, 5 mmol, 4.0 eq). The mixture was stirred at room temperature for 3 h under nitrogen. The mixture was then neutralized with a 1N aqueous solution of hydrochloric acid (50 mL). The mixture was extracted with ethyl acetate and the organic layer was further washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 40%
  • 1H NMR (400 MHz, DMSO d6) δ 9.35 (m, 1H), 7.41 (s, 4H), 7.09 (m, 1H), 6.97 (d, 1H), 6.91 (d, 1H), 5.92 (s, 1H), 3.72 (m, 2H), 3.44 (m, 2H), 3.23 (m, 4H), 2.87 (s, 3H), 1.86 (m, 2H), 1.71 (m, 2H), 1.42 (s, 9H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=570.4 (M+H)+
  • Preparation of 7A
  • A 2.0M solution of hydrochloric acid in diethyl ether (1.4 mL, 2.78 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 7.6 (0.29 g, 0.51 mmol, 1.0 eq) in anhydrous dichloromethane (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. Diethyl ether (100 mL) was added to the solution and the resulting precipitate was collected by filtration and washed with diethyl ether. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 25%
  • 1H NMR (400 MHz, DMSO d6) δ 9.42 (s, 1H), 8.85 (m, 2H), 7.43 (m, 4H), 7.12 (m, 1H), 7.05 (m, 1H), 6.93 (m, 1H), 6.00 (s, 1H), 3.45 (m, 2H), 3.37 (m, 2H), 3.24 (m, 4H), 2.88 (s, 3H), 2.07 (m, 2H), 1.98 (m, 2H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=470.4 (M+H)+
  • Elemental analysis:
  • C25H31N3O4S, 1HCl, 2H2O
  • Theory: % C, 55.39; % H, 6.69; % N, 7.75.
  • Found: % C, 55.03; % H, 6.33; % N, 7.36.
  • Example 7B Preparation of 7.7
  • To a solution of 7.6 (0.5 g, 0.88 mmol, 1.0 eq) in dry tetrahydrofuran (20 mL) at 0° C. was added sodium hydride (60% dispersion in mineral oil, 70 mg, 1.76 mmol, 2.0 eq) in one portion. The reaction mixture was kept at 0° C. for 1 h and methyliodide (2.8c) (0.08 mL, 1.1 mmol, 1.3 eq) was added drop wise. The mixture was kept at 0° C. for another 30 min, warmed up to room temperature, and then heated at 80° C. for 10 h. Water (50 mL) and chloroform (100 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 83%
  • 1H NMR (400 MHz, CDCl3) δ 7.43 (m, 2H), 7.36 (m, 2H), 7.19 (dd, 1H), 7.01 (d, 1H), 6.95 (d, 1H), 5.61 (s, 1H), 3.87 (brs, 2H), 3.57 (brs, 2H), 3.32 (m, 4H), 3.21 (s, 3H), 2.81 (s 3H), 2.05 (m, 2H), 1.68 (m, 2H), 1.48 (s, 9H), 1.20 (m, 6H)
  • Mass Spectral Analysis m/z=584.3 (M+H)+
  • Preparation of 7B
  • To a cold (0° C.) solution of 7.7 (0.43 g, 0.73 mmol, 1.0 eq) in anhydrous dichloromethane (20 mL) was added drop wise a 1.0 M solution of hydrogen chloride in diethyl ether (4.38 mL, 4.38 mmol, 6.0 eq). The reaction mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure. The crude product was purified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined and concentrated under reduced pressure. The product was dissolved in chloroform (100 mL) and washed with a 1M solution of sodium carbonate (2×30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added dropwise 1.0M hydrogen chloride in diethyl ether (1.46 mL, 1.46 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 8.79 (m, 2H), 7.44 (m, 4H), 7.34 (dd, 1H), 7.10 (d, 1H), 7.00 (d, 1H), 6.03 (s, 1H), 3.23 (m, 8H), 3.14 (s, 3H), 2.89 (s, 3H), 2.04 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=484.2 (M+H)+
  • Elemental analysis:
  • C26H33N3O4S, 1HCl, 1.3H2O
  • Theory: % C, 57.46; % H, 6.79; % N, 7.73.
  • Found: % C, 57.46; % H, 6.86; % N, 7.80.
  • Example 7C Preparation of 7.8
  • To a suspension of 6.4 (2 g, crude, as of 1.4 mmol, 1.0 eq) in dichloromethane (50 mL) at 0° C. was added triethylamine (0.98 mL, 7.0 mmol, 5 eq) followed by drop wise addition of methylsulfonyl chloride (7.4) (0.33 mL, 4.2 mmol, 3.0 eq). The reaction mixture was stirred at 0° C. for 1 h. A 1M aqueous solution of hydrochloric acid (100 mL) and chloroform (100 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product, which was used for the next step without purification.
  • Mass Spectral Analysis m/z=644.2 (M+H)+
  • Preparation of the Mixture of 7A & 7C
  • To a suspension of 7.8 (1.57 g, crude, as of 1.4 mmol, 1.0 eq) in a mixture of methanol (20 mL), tetrahydrofuran (20 mL) and water (20 mL) was added lithium hydroxide hydrate (0.98 mL, 7.0 mmol, 5.0 eq). The reaction mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure to give the crude product as a mixture of 7A and 7C, which was carried over for the next step without purification.
  • Mass Spectral Analysis m/z=470.2 (M+H)+ (7A)
  • Mass Spectral Analysis m/z=484.2 (M+H)+ (7C)
  • Preparation of 7C
  • To a suspension of the mixture of 7A and 7C (2.2 g, crude, as of 1.4 mmol, 1.0 eq) in dry dichloroethane (50 mL) at 0° C. was added pyridine (0.34 mL, 4.2 mmol, 3 eq) followed by di-tert-butyl dicarbonate (4.7) (0.46 g, 2.1 mmol, 1.5 eq) portion wise. The reaction mixture was slowly warmed up to room temperature and stirred at room temperature for 10 h. Water (50 mL) and chloroform (100 mL) were added. The two phases were separated and the aqueous phase was further extracted with chloroform (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity to obtain 7.6 as pure compound; eluent: dichloromethane/methanol mixture of increasing polarity to obtain crude 7C). Yield: 62% for 7.6 over three steps.
  • The crude 7C (100 mg) was further purified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined and concentrated under reduced pressure. The product was dissolved in chloroform (100 mL) and washed with a 1M aqueous solution of sodium carbonate (2×30 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added drop wise a 1.0M solution of hydrogen chloride in diethyl ether (0.41 mL, 0.41 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum.
  • 1H NMR (400 MHz, DMSO d6) δ 10.47 (m, 1H), 9.435 & 9.422 (2s, 1H), 7.51-6.92 (m, 7H), 6.31 & 5.90 (2s, 1H,), 3.50-3.17 (m, 8H), 2.88 & 2.87 (2s, 3H,), 2.82 (d, 3H), 2.12 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=484.2 (M+H)+
  • Elemental analysis:
  • C26H33N3O4S, 1HCl, 0.9H2O
  • Theory: % C, 58.23; % H, 6.73; % N, 7.84.
  • Found: % C, 58.02; % H, 6.68; % N, 8.20.
  • Example 8A
  • 8A was obtained according to a procedure similar to the one described for 2A, with the following exception:
  • Step 2.1: 2.1 was replaced by 8.1 (see also step 8.1).
  • 1H NMR (400 MHz, DMSO d6) δ 9.16 (s, 1H), 8.92 (brs, 1H), 8.73 (brs, 1H), 7.40 (s, 4H), 6.78 (m, 2H), 6.43 (dd, 1H), 5.86 (s, 1H), 3.43 (brm, 4H), 3.20 (brm, 4H), 2.09 (m, 2H), 1.93 (m, 2H), 1.11 (brd, 6H)
  • Mass Spectral Analysis m/z=393.4 (M+H)+
  • Elemental analysis:
  • C24H28N2O3, 1HCl, 0.33H2O
  • Theory: % C, 66.27; % H, 6.87; % N, 6.44.
  • Found: % C, 66.24; % H, 6.77; % N, 6.44.
  • Example 8B
  • 8B was obtained according to a procedure similar to the one described for 2A, with the following exceptions:
  • Step 2.1: 2.1 was replaced by 8.1 (see also step 8.1).
  • Step 2.4: 1.6 was replaced by 1.7 (see also step 8.4).
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (brm, 1H), 8.99 (brm, 1H), 8.57 (d, 1H), 7.88 (dd, 1H), 7.59 (d, 1H), 6.84 (m, 1H), 6.78 (t, 1H), 6.40 (dd, 1H), 6.00 (s, 1H), 3.47 (q, 2H), 3.40 (m, 2H), 3.29 (q, 2H), 3.19 (m, 2H), 2.10 (m, 2H), 1.97 (m, 2H), 1.17 (t, 3H), 1.10 (t, 3H)
  • Mass Spectral Analysis m/z=394.2 (M+H)+
  • Elemental analysis:
  • C24H27N3O3, 2HCl, 0.67H2O
  • Theory: % C, 57.74; % H, 6.39; % N, 8.78; % Cl, 14.82.
  • Found: % C, 57.70; % H, 6.28; % N, 8.73; % Cl, 14.47.
  • Example 8C
  • 8C was obtained according to a procedure similar to the one described for 2C, with the following exception:
  • Step 2.1: 2.1 was replaced by 8.1 (see also step 8.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (brm, 2H), 7.42 (s, 4H), 7.00 (d, 1H), 6.86 (t, 1H), 6.58 (d, 1H), 5.97 (s, 1H), 3.90 (d, 2H), 3.44 (m, 2H), 3.23 (brm, 6H), 2.09 (m, 2H), 1.98 (m, 2H), 1.26 (m, 1H), 1.12 (brd, 6H), 0.59 (m, 2H), 0.37 (m, 2H)
  • Mass Spectral Analysis m/z=447.3 (M+H)+
  • Elemental analysis:
  • C28H34N2O3, 1HCl, 1.5H2O
  • Theory: % C, 65.93; % H, 7.51; % N, 5.49.
  • Found: % C, 65.64; % H, 7.29; % N, 5.41.
  • Example 8D
  • 8D was obtained according to a procedure similar to the one described for 2C, with the following exceptions:
  • Step 2.1: 2.1 was replaced by 8.1 (see also step 8.1).
  • Step 2.7: 2.8a was replaced by 2.8c (method 2A was used) (see also step 8.7).
  • 1H NMR (400 MHz, DMSO d6) δ 8.78 (brs, 2H), 7.41 (s, 4H), 7.04 (d, 1H), 6.90 (t, 1H), 6.58 (d, 1H), 5.97 (s, 1H), 3.83 (s, 3H), 3.44 (brs, 2H), 3.20 (brm, 6H), 2.08 (m, 2H), 1.97 (m, 2H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=407.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 1H2O
  • Theory: % C, 65.14; % H, 7.22; % N, 6.08.
  • Found: % C, 65.22; % H, 6.85; % N, 6.02.
  • Example 8E
  • 8E was obtained according to a procedure similar to the one described for 2C, with the following exceptions:
  • Step 2.1: 2.1 was replaced by 8.1 (see also step 8.1).
  • Step 2.4: 1.6 was replaced by 1.7 (see also step 8.4).
  • 1H NMR (400 MHz, DMSO d6) δ 8.94 (brm, 2H), 8.59 (d, 1H), 7.88 (dd, 1H), 7.60 (d, 1H), 7.03 (d, 1H), 6.88 (t, 1H), 6.56 (d, 1H), 6.11 (s, 1H), 3.91 (d, 2H), 3.47 (q, 2H), 3.29 (m, 4H), 3.17 (m, 2H), 2.10 (m, 2H), 2.01 (m, 2H), 1.26 (m, 1H), 1.17 (t, 3H), 1.11 (t, 3H), 0.59 (m, 2H), 0.37 (m, 2H)
  • Mass Spectral Analysis m/z=448.3 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 1.2HCl, 0.8H2O
  • Theory: % C, 64.12; % H, 7.14; % N, 8.31; % Cl, 8.41.
  • Found: % C, 64.09; % H, 7.20; % N, 8.18; % Cl, 8.15.
  • Example 8F
  • 8F was obtained according to a procedure similar to the one described for 2C, with the following exceptions:
  • Step 2.1: 2.1 was replaced by 8.1 (see also step 8.1).
  • Step 2.4: 1.6 was replaced by 1.7 (see also step 8.4).
  • Step 2.7: 2.8a was replaced by 2.8c (see also step 8.7).
  • 1H NMR (400 MHz, DMSO d6) δ 8.96 (brm, 2H), 8.59 (d, 1H), 7.88 (dd, 1H), 7.60 (d, 1H), 7.06 (d, 1H), 6.92 (t, 1H), 6.56 (d, 1H), 6.12 (s, 1H), 3.84 (S, 3H), 3.47 (q, 2H), 3.28 (m, 4H), 3.14 (m, 2H), 2.09 (m, 2H), 2.02 (m, 2H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=408.4 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 2HCl, 1.5H2O
  • Theory: % C, 56.81; % H, 6.75; % N, 8.28; % Cl, 13.97.
  • Found: % C, 56.80; % H, 6.48; % N, 8.24; % Cl, 13.89.
  • Example 9A
  • 9A was obtained according to a procedure similar to the one described for 2C, with the following exception:
  • Step 2.1: 2.1 was replaced by 9.1 (see also step 9.1).
  • 1H NMR (400 MHz, DMSO d6) δ 9.68 (brd, 2H), 7.41 (d, 2H), 7.35 (d, 2H), 6.92 (d, 1H), 6.43 (s, 1H), 6.37 (d, 1H), 5.44 (s, 1H), 3.80 (d, 2H), 3.56 (brs, 2H), 3.40 (brs, 4H), 3.30 (brs, 2H), 2.30 (m, 2H), 2.19 (m, 2H), 1.27 (m, 4H), 1.17 (brs, 3H), 0.66 (m, 2H), 0.36 (m, 2H)
  • Mass Spectral Analysis m/z=447.3 (M+H)+
  • Elemental analysis:
  • C28H34N2O3, 1.0HCl, 1.3H2O
  • Theory: % C, 66.40; % H, 7.48; % N, 5.53.
  • Found: % C, 66.28; % H, 7.48; % N, 5.48.
  • Example 9B Preparation of 9.5
  • 9.5 was obtained according to a procedure similar to the one described for 2.7a except 2.1 was replaced by 9.1 in step 2.1 (see also step 9.1).
  • Preparation of 9.8
  • To a solution of 9.5 (1.00 g, 2.02 mmol, 1.0 eq) in dimethylformamide (10 mL) was added sequentially cesium carbonate (3.30 g, 10.1 mmol, 5.0 eq) and methyl chlorodifluoroacetate (9.7) (1.47 g, 10.1 mmol, 5.0 eq.). The reaction mixture was heated at 90° C. for 48 h, poured into water (100 mL) and extracted with ethyl acetate. The organic extracts were washed with a 1N aqueous solution of sodium hydroxide and brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane:ethyl acetate mixtures of increasing polarity). Yield: 79%
  • 1H NMR (400 MHz, CDCl3) δ 7.41 (d, 2H), 7.36 (d, 2H), 6.98 (d, 1H), 6.73 (d, 1H), 6.61 (dd, 1H), 6.52 (ts, 1H, J=73.8 Hz), 5.54 (s, 1H), 3.86 (brs, 2H), 3.57 (brm, 2H), 3.32 (brm, 4H), 2.03 (d, 2H), 1.68 (m, 2H), 1.47 (s, 9H) 1.20 (brd, 6H)
  • Mass Spectral Analysis m/z=543.4 (M+H)+
  • Preparation of 9B
  • To a solution of 9.8 (860 mg, 1.58 mmol, 1.0 eq) in anhydrous methanol (15 mL) was added drop wise a 4.0M solution of hydrochloric acid in dioxane (4.0 mL, 15.8 mmol, 10.0 eq). The mixture was stirred at ambient temperature for 16 h and the solvent was evaporated under vacuum. The crude oil was purified by reverse phase HPLC chromatography (eluent: acetonitrile/water (0.1% trifluoroacetic acid) mixtures of decreasing polarity). The solvent was evaporated under vacuum and a 1N solution of HCl in diethyl ether (25 mL) was added. The resulting solid was filtered and washed with diethyl ether. Yield: 23%
  • 1H NMR (400 MHz, CDCl3) δ 7.42 (d, 2H), 7.35 (d, 2H), 7.02 (d, 1H), 6.75 (m, 1H), 6.66 (dd, 1H), 6.54 (ts, 1H, J=73.4 Hz), 5.59 (s, 1H), 3.57 (brs, 2H), 3.41 (brd, 4H), 3.31 (brs, 2H), 2.26 (m, 4H), 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=443.4 (M+H)+
  • Elemental analysis:
  • C28H34N2O3, 1.0HCl, 1.2H2O
  • Theory: % C, 59.99; % H, 6.32; % N, 5.60.
  • Found: % C, 60.01; % H, 6.25; % N, 5.54.
  • Example 10A
  • 10A was obtained from 9.5 according to a procedure similar to the one described for 3A, with the following exception:
  • Step 3.1: 2.7a was replaced by 9.5 (see also step 10.1).
  • 1H NMR (400 MHz, DMSO d6) δ 9.80 (brs, 1H), 7.60 (s, 1H), 7.58 (d, 1H), 7.42 (d, 2H), 7.36 (d, 2H), 7.09 (d, 1H), 5.75 (s, 1H), 3.91 (s, 3H), 3.61 (brs, 2H), 3.40 (m, 4H), 3.30 (brs, 2H), 2.27 (m, 4H), 1.20 (brd, 6H)
  • Mass Spectral Analysis m/z=435.3 (M+H)+
  • Elemental analysis:
  • C26H30N2O4, 1HCl, 1.1H2O
  • Theory: % C, 63.63; % H, 6.82; % N, 5.71.
  • Found: % C, 63.64; % H, 6.75; % N, 5.72.
  • Example 10B
  • 10B was obtained according to a procedure similar to the one described for 3B, with the following exception:
  • Step 3.1: 2.7a was replaced by 9.5 (see also step 10.1).
  • 1H NMR (400 MHz, DMSO-d6) δ 13.10 (brs, 1H), 9.10 (brm, 2H), 7.57 (d, 1H), 7.52 (dd, 1H), 7.44 (s, 4H), 7.12 (d, 1H), 6.09 (s, 1H), 3.45 (brs, 2H), 3.35 (brm, 2H), 3.23 (brm, 4H), 2.08 (m, 4H), 1.10 (brd, 6H)
  • Mass Spectral Analysis m/z=421.3 (M+H)+
  • Example 10C
  • 10C was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a was replaced by 10.3 and 3.4b was replaced by 3.4a (see also step 10.5).
  • 1H NMR (400 MHz, CDCl3) δ 9.50 (brd, 2H), 7.64 (brm, 2H), 7.32 (brm, 5H), 7.00 (brs, 2H), 5.68 (s, 1H), 3.50 (brm, 4H), 3.27 (brm, 4H), 2.62 (brs, 2H), 2.19 (brs, 2H), 1.17 (brd, 6H)
  • Mass Spectral Analysis m/z=420.3 (M+H)+
  • Example 10D Preparation of 10.2
  • Compound 10.2 was obtained according to a procedure similar to the one described for 3.2a except 2.7a was replaced by 9.5 in step 3.1 (see also step 10.1).
  • Preparation of 10.4
  • To a solution of a 2N solution of methylamine (3.4b) in methanol (10.0 mL, 20.0 mmol, 11.0 eq) was added portionwise at room temperature 10.2 (1.00 g, 1.86 mmol) in a sealed tube. The mixture was heated at 60° C. for 20 h to form a homogeneous solution. The mixture was poured into water (25 mL), extracted with methylene chloride, washed with brine, dried over sodium sulfate, filtered and evaporated solvent to an off-white solid. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 80%
  • 1H NMR (400 MHz, CDCl3) δ 7.53 (s, 1H), 7.47 (s, 1H), 7.45 (d, 2H), 7.23 (d, 1H), 7.04 (d, 1H), 6.20 (brs, 1H), 5.64 (s, 1H), 3.88 (brs, 2H), 3.57 (brm, 2H), 3.33 (brm, 4H), 3.00 (d, 3H), 2.03 (d, 2H), 1.68 (brm, 2H), 1.45 (s, 9H) 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=534.4 (M+H)+
  • Preparation of 10D
  • To a solution of 10.4a (790 mg, 1.48 mmol, 1.0 eq) in anhydrous methanol (20 mL) was added drop wise a 4M solution of hydrochloric acid in dioxane (3.7 mL, 14.8 mmol, 10.0 eq). The mixture was stirred at ambient temperature for 16 h and the solvent evaporated under vacuum to a white solid. The white solid was triturated in diethyl ether (50 mL). The resulting solid was collected by filtration and washed with diethyl ether. Yield: 85%
  • 1H NMR (400 MHz, CDCl3) δ 7.43 (m, 3H), 7.34 (m, 3H), 7.05 (d, 1H), 6.90 (brd, 1H), 5.69, (s, 1H), 3.57 (brm, 2H), 3.35 (brm, 6H), 3.00 (d, 3H), 2.20 (brs, 4H), 1.19 (brd, 6H)
  • Mass Spectral Analysis m/z=434.3 (M+H)+
  • Elemental analysis:
  • C26H31N3O3, 1.0HCl, 1.5H2O
  • Theory: % C, 62.83; % H, 7.10; % N, 8.45.
  • Found: % C, 62.74; % H, 6.95; % N, 8.29.
  • Example 10E
  • 10E was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a was replaced by 10.3 and 3.4b was replaced by 3.4c (see also step 10.5) (method 10A was used).
  • 1H NMR (400 MHz, CDCl3) δ 9.68 (brs, 2H), 7.43 (m, 3H), 7.34 (m, 3H), 7.06 (d, 1H), 6.61 (brs, 1H), 5.68 (s, 1H), 3.57 (brs, 2H), 3.50 (brm, 2H), 3.40 (brs, 2H), 3.32 (brs, 2H), 2.25 (brs, 4H), 1.28 (brm, 6H), 1.15 (brs, 3H)
  • Mass Spectral Analysis m/z=448.3 (M+H)+
  • Example 10F
  • 10F was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a was replaced by 10.3 and 3.4b was replaced by 3.4j (see also step 10.5) and TBTU was replaced by HATU (method 10B was used).
  • 1H NMR (400 MHz, DMSO d6) δ 9.77 (brm, 2H), 7.42 (d, 2H), 7.36 (d, 2H), 7.08 (d, 1H), 7.03 (s, 1H), 6.97 (d, 1H), 5.66 (s, 1H), 3.59 (brs, 2H), 3.40 (brs, 4H), 3.32 (brs, 2H), 3.12 (s, 3H), 3.04 (s, 3H), 2.28 (m, 4H), 1.20 (brd, 6H)
  • Mass Spectral Analysis m/z=448.3 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 1HCl, 1.7H2O
  • Theory: % C, 63.01; % H, 7.32; % N, 8.16.
  • Found: % C, 63.06; % H, 7.18; % N, 8.09.
  • Example 10G
  • 10G was obtained according to a procedure similar to the one described for 3E, with the following exceptions:
  • Step 3.5: 3.3a was replaced by 10.3 and 3.4b was replaced by 1.12 (see also step 10.5) (method 10A was used).
  • 1H NMR (400 MHz, DMSO d6) δ 9.73 (brs, 2H), 7.43 (d, 2H), 7.36 (d, 2H), 7.07 (d, 1H), 6.98 (s, 1H), 6.92 (d, 1H), 5.67 (s, 1H), 3.56 (brs, 4H), 3.40 (brs, 4H), 3.31 (brs, 4H), 2.26 (brs, 4H), 1.22 (brd, 12H)
  • Mass Spectral Analysis m/z=476.2 (M+H)+
  • Elemental analysis:
  • C29H37N3O3, 1HCl, 1.7H2O
  • Theory: % C, 64.18; % H, 7.69; % N, 7.74.
  • Found: % C, 64.08; % H, 7.45; % N, 7.60.
  • Example 10H
  • 10H was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.3a was replaced by 10.3 and 3.4b was replaced by 3.4k (see also step 10.5) (method 10A was used).
  • 1H NMR (400 MHz, DMSO d6) δ 9.77 (brs, 2H), 7.43 (d, 2H), 7.37 (d, 2H), 7.12 (s, 1H), 7.09 (s, 2H), 5.68 (s, 1H), 3.64 (m, 2H), 3.60 (brm, 2H), 3.47 (m, 2H), 3.40 (brm, 4H), 3.30 (brs, 2H), 2.30 (brs, 4H), 2.00 (m, 2H), 1.93 (m, 2H), 1.24 (brd, 6H)
  • Mass Spectral Analysis m/z=474.3 (M+H)+
  • Elemental analysis:
  • C29H35N3O3, 1HCl, 0.7H2O
  • Theory: % C, 66.64; % H, 7.21; % N, 8.04.
  • Found: % C, 66.56; % H, 7.07; % N, 7.91.
  • Example 10I
  • 10I was obtained according to a procedure similar to the one described for 3E, with the following exception:
  • Step 3.5: 3.3a was replaced by 10.3 and 3.4b was replaced by 3.4c (see also step 10.5) (method 10A was used).
  • 1H NMR (400 MHz, CDCl3) δ 9.70 (brs, 2H), 7.44 (d, 2H), 7.35 (d, 2H), 7.09 (d, 1H), 7.02 (s, 1H), 6.96 (dd, 1H), 5.68 (s, 1H), 3.73 (brm, 6H), 3.58 (brs, 4H), 3.41 (brm, 4H), 3.31 (brs, 2H), 2.28 (m, 4H), 1.21 (m, 6H)
  • Mass Spectral Analysis m/z=490.2 (M+H)+
  • Example 10J Preparation of 10.5
  • To a slurry of LiBH4 (82.0 mg, 3.75 mmol, 2.0 eq.) in tetrahydrofuran (20 mL) cooled to 0° C. under a nitrogen atmosphere was added drop wise a solution of 10.2 (1.00 g, 1.87 mmol, 1.0 eq) in tetrahydrofuran (10 mL). The reaction mixture was warmed to room temperature and stirred for 16 h at room temperature. The reaction mixture was quenched with water (0.54 mL, 8 eq.), extracted with ethyl acetate, washed with brine, dried over sodium sulfate and filtered. The solvent was removed under vacuum and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 49%
  • 1H NMR (400 MHz, CDCl3) δ 7.40 (d, 2H), 7.36 (d, 2H), 6.98 (m, 2H), 6.85 (d, 1H), 5.56 (s, 1H), 4.65 (s, 2H), 3.87 (brs, 2H), 3.57 (brs, 2H), 3.32 (brm, 4H), 2.05 (d, 2H), 1.91 (brt, 1H), 1.66 (m, 2H), 1.48 (s, 9H) 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=507.3 (M+H)+
  • Preparation of 10J
  • To a solution of 10.5 (460 mg, 0.91 mmol, 1.0 eq) in anhydrous methanol (30 mL) was added drop wise a 4M solution of hydrochloric acid in dioxane (2.3 mL, 9.1 mmol, 10.0 eq). The mixture was stirred at room temperature for 16 h and the solvent was evaporated under vacuum. The residue was triturated in ethyl ether (50 mL); the solid was collected by filtration and washed with diethyl ether. The crude product was purified by column chromatography (eluent: methylene chloride/methanol mixtures of increasing polarity). Yield: 46%
  • 1H NMR (400 MHz, CDCl3) δ 9.62 (brs, 2H), 7.38 (brd, 4H), 7.00 (m, 2H), 6.90 (brd, 1H), 5.60, (brs, 1H), 4.66 (brs, 2H), 3.58 (brm, 2H), 3.40 (brm, 4H), 3.31 (brm, 2H), 2.50 (brs, 1H), 2.25 (brs, 4H), 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Elemental analysis:
  • C26H31N3O3, 1HCl, 0.7H2O
  • Theory: % C, 65.91; % H, 7.17; % N, 6.15.
  • Found: % C, 65.93; % H, 6.99; % N, 6.08.
  • Example 11A Preparation of 11.2
  • 2′,6′-hydroxyacetophenone (11.1) (200.0 g, 1.31 mol, 1.0 eq) was added portion wise at room temperature to pyrrolidine (220 mL, 2.0 eq) followed by portion wise addition of 1-Boc-4-piperidone (1.2) (262.0 g, 1.31 mo, 1.0 eq). Anhydrous methanol (100 mL) was then added and the red slurry heated to reflux to dissolve all solids. On dissolution the reaction was cooled to room temperature overnight with stirring to form a solid mass. This solid mass was dissolved in ethyl acetate, washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide and brine, dried over sodium sulfate and filtered. The solvent was evaporated under vacuum. A mixture of hexane and diethyl ether (80:20) (400 mL) was added to the mixture and the resulting precipitate was collected by filtration, washed with hexane and used for the next step without further purification.
  • Yield: 74%.
  • 1H NMR (400 MHz, CDCl3) δ 11.61 (s, 1H), 7.37 (t, 1H), 6.49 (d, 1H), 6.44 (d, 1H), 3.89 (brs, 2H), 3.20 (brm, 2H), 2.73 (s, 2H), 2.02 (d, 2H), 1.64 (m, 2H), 1.46 (s, 9H)
  • Mass Spectral Analysis m/z=334.0 (M+H)+
  • Preparation of 11.4
  • To a solution of 11.2 (140.0 g, 0.420 mol, 1.0 eq) in dichloromethane (700 mL) at ambient temperature under nitrogen was added drop wise diisopropylethylamine (294.0 mL, 1.68 mol, 4.0 eq). To this solution was added drop wise chloro(methoxy)methane (11.3) (100.0 g, 1.26 mol, 3.0 eq). The mixture was heated to reflux for 16 h, cooled to room temperature and the solvent was removed under vacuum to afford a brown oil. This oil was dissolved in ethyl acetate (700 mL) and washed with a 1N aqueous solution of hydrochloric acid, an aqueous saturated solution of sodium bicarbonate and brine. The organic extracts were dried over sodium sulfate, filtered and the solvent was removed under vacuum to afford a brown oil. Diethyl ether (400 mL) was added and the resulting white precipitate was filtered and used for the next step without further purification. Yield: 83%
  • 1H NMR (400 MHz, CDCl3) δ 7.36 (t, 1H), 6.74 (d, 1H), 6.65 (d, 1H), 5.27 (s, 2H), 3.86 (brs, 2H), 3.52 (s, 3H), 3.22 (m, 2H), 2.69 (s, 2H), 2.02 (d, 2H), 1.60 (m, 2H), 1.46 (s, 9H)
  • Mass Spectral Analysis m/z=378.2 (M+H)+
  • Preparation of 11.5
  • To a solution of 11.4 (131.2 g, 0.348 mol) in tetrahydrofuran (600 mL) at −78° C. under nitrogen atmosphere was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (420.0 mL, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (149.4 g, 0.418 mol, 1.2 eq) in tetrahydrofuran (200 mL) was added drop wise. The mixture was warmed slowly to room temperature and stiffing was continued for a further 12 h at room temperature. The mixture was then poured into ice water and the two phases were separated. The organic phase was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide and brine, dried over sodium sulfate and filtered. The solvent was removed under vacuum and the tan oily residue was used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 6.98 (t, 1H), 6.62 (d, 1H), 6.39 (d, 1H), 5.24 (s, 1H), 5.03 (s, 2H), 3.62 (brs, 2H), 3.30 (s, 3H), 3.07 (m, 2H), 1.84 (d, 2H), 1.46 (m, 2H), 1.26 (s, 9H)
  • Mass Spectral Analysis m/z=510.0 (M+H)+
  • Preparation of 11.6a
  • To a solution of 11.5 (100 g, 196 mmol, 1.0 eq) in dimethoxyethane (DME) (600 mL) was added sequentially a 2N aqueous solution of sodium carbonate (294 mL, 588 mmol, 3.0 eq), lithium chloride (25.0 g, 588 mmol, 3.0 eq), 4-(N,N-diethylaminocarbonyl)phenylboronic acid) (1.6) (36.9 g, 166 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (4.54 g, 3.92 mmol, 0.02 eq). The mixture was refluxed for 10 h under nitrogen. The mixture was then cooled to room temperature, filtered through a celite pad and the filtercake was washed with DME (100 mL) and water (750 mL). The aqueous mixture was extracted with ethyl acetate. The organic layer was further washed with brine and dried over sodium sulfate. The crude product was purified by chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 62%
  • 1H NMR (400 MHz, CDCl3) δ 7.21 (d, 2H), 7.17 (d, 2H), 7.05 (t, 1H), 6.60 (m, 2H), 5.45 (s, 1H), 4.58 (s, 2H), 3.71 (brs, 2H), 3.45 (brm, 2H), 3.22 (brm, 4H), 3.06 (s, 3H), 1.90 (d, 2H), 1.56 (m, 2H), 1.38 (s, 9H), 1.09 (brd, 6H)
  • Mass Spectral Analysis m/z=537.4 (M+H)+
  • Preparation of 11A
  • To a solution of 11.6a (25.0 g, 46.6 mmol, 1.0 eq) in anhydrous methanol (250 mL) was added drop wise a 4M solution of hydrochloric acid in dioxane (58.2 mL, 233 mmol, 5.0 eq). The mixture was stirred at room temperature for 16 h and the solvent was evaporated under vacuum to afford a brown oil. Methanol (20 mL) followed by diethyl ether (300 mL) was added to the brown oil and the resulting precipitate was collected by filtration and washed with diethyl ether. The solid was used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 9.55 (s, 1H), 9.07 (brs, 2H), 7.27 (m, 4H), 7.06 (t, 1H), 6.52 (d, 1H), 6.47 (d, 1H), 5.76 (s, 1H), 3.42 (brm, 2H), 3.35 (s, 4H), 3.19, (brm, 6H), 2.03 (m, 4H), 1.11 (brm, 6H)
  • Mass Spectral Analysis m/z=393.0 (M+H)+
  • Elemental analysis:
  • C24H28N2O3, 1HCl, 0.67H2O
  • Theory: % C, 65.37; % H, 6.93; % N, 6.35.
  • Found: % C, 65.41; % H, 6.98; % N, 6.31.
  • Example 11B
  • 11B was obtained according to a procedure similar to the one described for 11A, with the following exception:
  • Step 11.4: 1.6 was replaced by 1.7.
  • 1H NMR (400 MHz, DMSO d6) δ 9.67 (brs, 1H), 9.23 (brd, 2H), 8.50 (s, 1H), 7.79 (d, 1H), 7.52 (d, 1H), 7.09 (t, 1H), 6.57 (d, 1H), 6.50 (d, 1H), 5.93 (s, 1H), 3.43 (q, 2H), 3.26 (q, 2H), 3.21 (m, 2H), 3.14 (m, 2H), 2.05 (m, 4H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=394.3 (M+H)+
  • Elemental analysis:
  • C23H27N3O3, 2HCl, 1.5H2O
  • Theory: % C, 55.99; % H, 6.54; % N, 8.52.
  • Found: % C, 56.11; % H, 6.54; % N, 8.53.
  • Example 11C Preparation of 11.7a
  • To a slurry of 11A (10.0 g, 23.3 mmol, 1.0 eq) in tetrahydrofuran (200 mL) under a nitrogen atmosphere was added triethylamine (9.75 mL, 69.9 mmol, 3.0 eq). The reaction mixture was cooled to 0° C. A solution of di-tert-butyl dicarbonate (4.7) (4.58 g, 21.0 mmol, 0.9 eq) in tetrahydrofuran (50 mL) was added drop wise to the reaction mixture which was stirred for 3 h at room temperature. The solvent was evaporated under vacuum and the residue was dissolved in ethyl acetate (500 mL), washed with water and brine, and dried over sodium sulfate and filtered. The solvent was evaporated under vacuum. The residue was sonicated and triturated in a mixture ethyl acetate/methanol 95:5 (75 mL). The solid was collected by filtration and washed with ethyl acetate. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 9.49 (s, 1H), 7.31 (s, 4H), 7.08 (t, 1H), 6.54 (d, 1H), 6.47 (d, 1H), 5.77 (s, 1H), 3.70 (m, 2H), 3.48 (brm, 2H), 3.30 (brm, 4H), 1.87 (d, 2H), 1.74 (m, 2H), 1.47 (s, 9H) 1.16 (brs, 6H)
  • Mass Spectral Analysis m/z=493.4 (M+H)+
  • Preparation of 11.9a
  • To a solution of 11.7a (1.00 g, 2.02 mmol, 1.0 eq) in dichloromethane (4 mL) under a nitrogen atmosphere was added sequentially cyclopropylmethanol (2.8e) (189 mg, 2.63 mmol, 1.3 eq) and triphenylphosphine (690 mg, 2.63 mmol, 1.3 eq). The reaction mixture was stirred for 5 min at room temperature and a solution of diethylazodicarboxylate (460 mg, 2.63 mmol, 1.3 eq) was added drop wise. The reaction was stirred an additional 30 min at room temperature and the solvent was evaporated under vacuum. The crude product was purified by chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 42%
  • 1H NMR (400 MHz, CDCl3) δ 7.31 (d, 2H), 7.27 (d, 2H), 7.13 (t, 1H), 6.64 (d, 1H), 6.42 (d, 1H), 5.50 (s, 1H), 3.78 (brd, 2H), 3.54 (brm, 2H), 3.49 (d, 2H), 3.35 (brt, 4H), 2.02 (d, 2H), 1.69 (m, 2H), 1.47 (s, 9H) 1.26 (brd, 6H), 0.53 (m, 1H), 0.29 (m, 2H), −0.07 (m, 2H)
  • Mass Spectral Analysis m/z=547.5 (M+H)+
  • Preparation of 11C
  • To a solution of 11.9a (460 mg, 0.84 mmol, 1.0 eq) in anhydrous methanol (15 mL) was added dropwise a 4M solution of hydrochloric acid in dioxane (2.0 mL, 8.4 mmol, 10.0 eq). The mixture was stirred at room temperature for 16 h and the solvent was evaporated under vacuum. The residue was triturated in diethyl ether (50 mL). The resulting solid was collected by filtration and washed with diethyl ether.
  • Yield: 97%
  • 1H NMR (400 MHz, CDCl3) δ 9.67 (brs, 2H), 7.32 (d, 2H), 7.26 (d, 2H), 7.16 (t, 1H), 6.64 (d, 1H), 6.46 (d, 1H), 5.50 (s, 1H), 3.54 (brm, 2H), 3.49 (d, 2H), 3.36 (brm, 6H), 2.28 (d, 2H), 2.18 (m, 2H), 1.19 (brd, 6H), 0.53 (m, 1H), 0.30 (m, 2H), −0.07 (m, 2H)
  • Mass Spectral Analysis m/z=447.4 (M+H)+
  • Elemental analysis:
  • C28H34N2O3, 1.0HCl, 0.7H2O
  • Theory: % C, 67.73; % H, 7.41; % N, 5.64.
  • Found: % C, 67.73; % H, 7.24; % N, 5.59.
  • Example 11D
  • 11D was obtained according to a procedure similar to the one described for 11C, with the following exceptions:
  • Step 11.4: 1.6 was replaced by 1.7.
  • 1H NMR (400 MHz, CDCl3) δ 9.67 (brs, 1H), 8.44 (m, 1H), 7.61 (dd, 1H), 7.55 (d, 1H), 7.19 (t, 1H), 6.64 (d, 1H), 6.43 (d, 1H), 5.55 (s, 1H), 3.56 (q, 2H), 3.50 (d, 2H), 3.46 (q, 2H), 3.38 (m, 4H), 2.29 (m, 2H), 2.21 (m, 2H), 1.28 (t, 3H), 1.17 (t, 3H), 0.54 (m, 1H), 0.33 (m, 2H), −0.05 (m, 2H)
  • Mass Spectral Analysis m/z=448.4 (M+H)+
  • Example 11E Preparation of 11.9b
  • To a solution of 11.7a (1.00 g, 2.02 mmol, 1.0 eq) in acetone (20 mL) was added sequentially potassium carbonate (1.70 g, 12.1 mmol, 6.0 eq) and bromocyclobutane (11.8) (1.66 g, 12.1 mmol, 6.0 eq). The reaction mixture was refluxed for 90 h, poured into water (100 mL) and extracted with ethyl acetate. The organic extracts were washed with a 1N aqueous solution of sodium hydroxide and brine, dried over sodium sulfate and filtered. The solvent was evaporated and the crude product was first purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity) and then repurified by reverse phase HPLC chromatography (eluent: acetonitrile/water (0.1% trifluoroacetic acid) mixtures of decreasing polarity). Yield: 18%
  • 1H NMR (400 MHz, CDCl3) δ 7.31 (d, 2H), 7.27 (d, 2H), 7.11 (t, 1H), 6.64 (d, 1H), 6.26 (d, 1H), 4.36 (m, 1H), 5.50 (s, 1H), 3.79 (brd, 2H), 3.54 (brm, 2H), 3.48 (d, 2H), 3.34 (brm, 4H), 2.12 (m, 2H), 2.02 (d, 2H), 1.67 (m, 2H), 1.55 (m, 2H), 1.47 (s, 9H) 1.19 (brd, 6H)
  • Mass Spectral Analysis m/z=547.5 (M+H)+
  • Preparation of 11E
  • To a solution of 11.9b (200 mg, 0.37 mmol, 1.0 eq) in anhydrous methanol (25 mL) was added drop wise a 2M solution of hydrochloric acid in diethyl ether (0.73 mL, 1.44 mmol, 4.0 eq). The mixture was stirred at room temperature for 16 h and the solvent was evaporated under vacuum. The residue was triturated in diethyl ether (50 mL). The solid was collected by filtration and washed with diethyl ether.
  • Yield: 96%
  • 1H NMR (400 MHz, DMSO d6) δ 9.14 (brs, 2H), 7.29 (d, 2H), 7.24 (d, 2H), 7.19 (t, 1H), 6.68 (d, 1H), 6.42 (d, 1H), 5.79 (s, 1H), 4.43 (m, 1H), 3.40 (brm, 4H), 3.35 (brs, 4H), 3.17 (brm, 4H), 2.10 (m, 2H), 2.03 (m, 2H), 1.45 (m, 2H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=447.3 (M+H)+
  • Example 11F
  • 11F was obtained according to a procedure similar to the one described for 11C, with the following exceptions:
  • Step 11.4: 1.6 was replaced by 1.7.
  • Step 11.7: 2.8e was replaced by 11.10.
  • 1H NMR (400 MHz, CDCl3) δ 9.71 (brd, 2H), 8.40 (s, 1H), 7.56 (m, 2H), 7.18 (t, 1H), 6.62 (d, 1H), 6.48 (d, 1H), 5.50 (s, 1H), 4.50 (m, 1H), 3.58 (m, 2H), 3.48 (m, 2H), 3.38 (brs, 4H), 2.30 (d, 2H), 2.22 (brs, 2H), 1.64 (m, 2H), 1.36 (m, 2H), 1.30 (m, 5H), 1.19 (m, 5H)
  • Mass Spectral Analysis m/z=462.4 (M+H)+
  • Example 12A Preparation of 12.1
  • To a solution of compound 11.2 (3.33 g, 10 mmol) in anhydrous methylene chloride (100 mL) was added sequentially triethylamine (3.48 mL, 25 mmol, 2.5 eq), 4-dimethylaminopyridine (122 mg, 1 mmol, 0.1 eq) and N-phenyltrifluoromethanesulfonimide
  • (1.4) (4.48 g, 12.5 mmol, 1.25 eq). The reaction mixture was stirred at room temperature for 24 h, washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate and filtered. The solvent was evaporated under vacuum and the residue was purified by column chromatography (eluent: hexane/ethyl acetate, 3:1). Yield: 92.5%
  • 1H NMR (400 MHz, DMSO d6) δ 7.52 (t, 1H), 7.09 (d, 1H), 6.88 (d, 1H), 3.90 (m, 2H), 3.21 (m, 2H), 2.80 (s, 2H), 2.03 (m, 2H), 1.63 (m, 2H), 1.48 (s, 9H)
  • Preparation of 12.3
  • To a solution of 12.1 (5.4 g, 11.6 mmol) in tetrahydrofuran (100 mL) at room temperature was added tetrakis(triphenylphosphine)palladium(0) (670 mg, 0.58 mmol, 0.05 eq) followed by drop wise addition of a 2.0 M solution of methylzinc chloride (12.2a) in tetrahydrofuran (10 mL, 20 mmol, 1.72 eq). The mixture was stirred at room temperature for 2 days. The reaction mixture was then quenched with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic layer was washed with brine and dried over sodium sulfate. The solvent was evaporated under vacuum and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 4:1). Yield: 80.6%
  • 1H NMR (400 MHz, CDCl3) δ 7.30 (t, 1H), 6.86 (d, 1H), 6.80 (d, 1H), 3.88 (m, 2H), 2.70 (s, 2H), 2.60 (s, 3H), 2.00 (m, 2H), 1.60 (m, 2H), 1.45 (s, 9H)
  • Preparation of 12.4
  • To a solution of 12.3 (2.8 g, 8.46 mmol) in anhydrous tetrahydrofuran (80 mL) at −78° C. under nitrogen was added drop wise a 1.0 M solution of LiHMDS in tetrahydrofuran (11 mL, 11 mmol, 1.1 eq). The reaction mixture was stirred for 45 min at −78° C. A solution of N-phenyltrifluoromethanesulfonimide (1.4) (3.95 g, 11 mmol, 1.1 eq) in tetrahydrofuran (15 mL) was added drop wise to the reaction mixture. The mixture was warmed slowly to room temperature and stiffing was continued for a further 3 h at room temperature. The mixture was then poured into ice water and extracted with a mixture of hexane and diethyl ether (1:1). The organic layer was washed with water and brine, and dried over sodium sulfate and filtered. The organics were concentrated under vacuum and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 6:1). Yield: 61.3%
  • 1H NMR (400 MHz, CDCl3) δ 7.11 (t, 1H), 6.80 (m, 2H), 3.82 (m, 2H), 3.29 (m, 2H), 2.50 (s, 3H), 2.03 (m, 2H), 1.68 (m, 2H), 1.48 (s, 9H)
  • Preparation of 12.5
  • To a solution of 12.4 (848 mg, 1.83 mmol) in dimethoxyethane (DME) (16 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (3.1 mL, 6.2 mmol, 3.4 eq), lithium chloride (259 mg, 6.1 mmol, 3.3 eq), 4-(N,N-diethylaminocarbonyl)phenylboronic acid (1.6) (486 mg, 2.2 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (64 mg, 0.055 mmol, 0.03 eq). The mixture was refluxed overnight under nitrogen. The mixture was then cooled to room temperature and water (20 mL) was added. The mixture was extracted with ethyl acetate. The organic layer was further washed with brine, dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1). Yield: 96.9%
  • 1H NMR (400 MHz, CDCl3) δ 7.36 (d, 2H), 7.26 (d, 2H), 7.10 (t, 1H), 6.86 (d, 1H), 6.70 (d, 1H), 5.60 (s, 1H), 3.80 (m, 2H), 3.55 (m, 2H), 3.30 (m, 4H), 2.00 (m, 2H), 1.74 (s, 3H), 1.65 (m, 2H), 1.49 (s, 9H), 1.20 (m, 6H)
  • Preparation of 12A
  • To a solution of 12.5 (860 mg, 1.76 mmol) in methylene chloride (10 mL) was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (30 mL). The mixture was stirred at room temperature for 24 h and diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 97.8%
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (m, 2H), 7.38 (d, 2H), 7.29 (d, 2H), 7.18 (t, 1H), 6.93 (d, 1H), 6.80 (d, 1H), 5.95 (s, 1H), 3.45 (m, 2H), 3.20 (m, 6H), 2.00 (m, 4H), 1.70 (s, 3H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Elemental analysis:
  • C24H28N2O2, 1HCl, 1/2H2O
  • Theory: % C, 68.87; % H, 7.40; % N, 6.43.
  • Found: % C, 68.99; % H, 7.33; % N, 6.39.
  • Example 12B Preparation of 12.6
  • To a solution of 12.1 (14.4 g, 31 mmol) in N,N-dimethylformamide was added sequentially methanol (50 mL), triethylamine (7 mL, 50 mmol, 1.6 eq), 1,3-bis(diphenylphosphino)propane (dppp) (1.04 g, 2.5 mmol, 0.08 eq) and palladium (II) acetate (565 mg, 2.5 mmol, 0.08 eq). The carbon monoxide was then bubbled through the reaction solution while the mixture was heated to 65-70° C. for 3.5 h. The reaction mixture was cooled to room temperature, diluted with diethyl ether and washed with water and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 4:1). Yield: 87.9%
  • 1H NMR (400 MHz, CDCl3) δ 7.50 (t, 1H), 7.10 (d, 1H), 6.99 (d, 1H), 3.94 (s, 3H), 3.90 (m, 2H), 3.21 (m, 2H), 2.73 (s, 2H), 2.05 (m, 2H), 1.63 (m, 2H), 1.48 (s, 9H)
  • Preparation of 12.11
  • To a solution of 12.6 (13.2 g, 35.2 mmol) in anhydrous tetrahydrofuran (300 mL) at −78° C. was added drop wise a 1.0 M solution of LiHMDS in tetrahydrofuran (42 mL, 42 mmol, 1.2 eq) under nitrogen. The reaction mixture was stirred for 45 min at −78° C. A solution of N-phenyltrifluoromethanesulfonimide (1.4) (15.1 g, 42 mmol, 1.2 eq) in tetrahydrofuran (60 mL) was added drop wise to the reaction mixture. The mixture was warmed slowly to room temperature and stirred for 3 h. The mixture was then poured into ice water and extracted with a mixture of hexane and diethyl ether (1:1). The organic layer was washed with water and brine, dried over sodium sulfate and filtered. The organics were concentrated under vacuum and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 4:1). Yield: 90.2%
  • 1H NMR (400 MHz, CDCl3) δ 7.32 (d, 1H), 7.26 (t, 1H), 7.10 (d, 1H), 5.70 (s, 1H), 3.90 (s, 3H), 3.83 (m, 2H), 3.30 (m, 2H), 2.10 (m, 2H), 1.77 (m, 2H), 1.48 (s, 9H)
  • Preparation of 12.12
  • To a solution of 12.11 (16 g, 31.6 mmol) in dimethoxyethane (DME) (260 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (53 mL, 106 mmol, 3.4 eq), lithium chloride (4.5 mg, 106 mmol, 3.4 eq.), 4-(N,N-diethylaminocarbonyl)phenylboronic acid (1.6) (8.4 g, 38 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (1.1 g, 0.95 mmol, 0.03 eq). The mixture was refluxed overnight under nitrogen and then cooled to room temperature. Water (300 mL) was added to the mixture and the crude product was extracted with ethyl acetate. The organic layer was further washed with brine, dried over sodium sulfate and filtered. The organics were concentrated under vacuum and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1).
  • Yield: 98.5%
  • 1H NMR (400 MHz, CDCl3) δ 7.33 (d, 2H), 7.25 (m, 4H), 7.15 (d, 1H), 5.72 (s, 1H), 3.85 (m, 2H), 3.53 (m, 2H), 3.32 (m, 4H), 3.10 (s, 3H), 2.06 (m, 2H), 1.76 (m, 2H), 1.50 (s, 9H), 1.20 (m, 6H)
  • Preparation of 12.13
  • To a suspension of potassium tert-butoxide (9 g, 80 mmol, 8.0 eq) in diethyl ether (200 mL) was added drop wise water (0.72 mL, 40 mmol, 4.0 eq) at 0° C. The slurry was stirred for 30 min. To this mixture was added 12.12 (5.34 g, 10 mmol). The ice-bath was removed and the reaction mixture was stirred at room temperature overnight and quenched by addition of ice water. The aqueous layer was separated, acidified to pH 2-3 with a 1N aqueous solution of hydrochloric acid and extracted with methylene chloride. The organic layers were combined, dried over sodium sulfate and concentrated under vacuum. The crude product was used for the next step without further purification. Yield: 86.9%
  • 1H NMR (400 MHz, DMSO d6) δ 12.55 (brs, 1H), 7.23 (m, 7H), 5.98 (s, 1H), 3.68 (m, 2H), 3.42-3.20 (m, 6H), 1.80 (m, 4H), 1.42 (s, 9H), 1.10 (m, 6H)
  • Preparation of 12B
  • To a solution of 12.13 (300 mg, 0.58 mmol) in methylene chloride (4 mL) was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (15 mL). The mixture was stirred at room temperature for 24 h and diluted with diethyl ether. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 95%
  • 1H NMR (400 MHz, DMSO d6) δ 12.61 (brs, 1H), 8.69 (m, 6H), 7.38-7.25 (m, 7H), 6.06 (s, 1H), 3.41 (m, 2H), 3.25 (m, 6H), 2.06 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=421.3 (M+H)+
  • Example 12C Preparation of 12.14a
  • To a solution of 12.13 (780 mg, 1.5 mmol) in acetonitrile (50 mL) was added sequentially diisopropylethylamine (1.75 mL, 10 mmol, 6.7 eq), a 0.5 M solution of ammonia (12.15) in dioxane (30 mL, 15 mmol, 10 eq) and TBTU (580 mg, 1.8 mmol, 1.2 eq). The reaction mixture was stirred at room temperature for 3 days and then concentrated under vacuum. The residue was dissolved in ethyl acetate and washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by column chromatography (eluent: hexane/acetone, 1:1). Yield: 60.4%
  • 1H NMR (400 MHz, DMSO d6) δ 7.51 (s, 1H), 7.29 (t, 1H), 7.22 (s, 4H), 7.10 (d, 1H), 7.05 (d, 1H), 6.97 (s, 1H), 5.90 (s, 1H), 3.63 (m, 2H), 3.41 (m, 2H), 3.32 (m, 2H), 3.20 (m, 2H), 1.80 (m, 4H), 1.42 (s, 9H), 1.10 (m, 6H)
  • Preparation of 12C
  • To a solution of 12.14a (420 mg, 0.81 mmol) in methylene chloride (6 mL) was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (20 mL). The mixture was stirred at room temperature for 2 days and diluted with diethyl ether. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 87.5%
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (m, 2H), 7.54 (s, 1H), 7.32-7.10 (m, 7H), 6.88 (s, 1H), 5.98 (s, 1H), 3.42 (m, 2H), 3.20 (m, 6H), 2.10 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=420.3 (M+H)+
  • Example 12D
  • 12D was obtained according to a procedure similar to the one described for 12C, with the following exception:
  • Step 12.16: 12.15 was replaced by 3.4b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.19 (m, 2H), 7.83 (m, 1H), 7.30-7.20 (m, 6H), 7.00 (d, 1H), 5.96 (s, 1H), 3.41 (m, 2H), 3.20 (m, 6H), 2.11 (m, 4H), 2.06 (d, 3H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=434.3 (M+H)+
  • Example 12E
  • 12E was obtained according to a procedure similar to the one described for 12C, with the following exception:
  • Step 12.16: 12.15 was replaced by 3.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.18 (m, 2H), 7.90 (t, 1H), 7.30-7.20 (m, 6H), 7.00 (d, 1H), 5.96 (s, 1H), 3.40 (m, 2H), 3.20 (m, 6H), 2.50 (m, 2H), 2.10 (m, 4H), 1.10 (m, 6H), 0.78 (t, 3H)
  • Mass Spectral Analysis m/z=448.4 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 5/4H2O
  • Theory: % C, 68.99; % H, 7.61; % N, 8.94.
  • Found: % C, 69.27; % H, 7.43; % N, 8.93.
  • Example 12F
  • 12 F was obtained according to a procedure similar to the one described for 12C, with the following exception:
  • Step 12.16: 12.15 was replaced by 3.4d.
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (m, 2H), 7.91 (t, 1H), 7.31 (m, 1H), 7.20 (m, 5H), 7.00 (m, 1H), 5.96 (s, 1H), 3.45 (m, 4H), 3.20 (m, 6H), 2.40 (m, 2H), 2.08 (m, 4H), 1.10 (m, 6H), 0.70 (t, 3H)
  • Mass Spectral Analysis m/z=462.4 (M+H)+
  • Elemental analysis:
  • C28H35N3O3, 1HCl, 7/3H2O
  • Theory: % C, 62.27; % H, 7.59; % N, 7.78.
  • Found: % C, 62.37; % H, 7.23; % N, 7.74.
  • Example 12G Preparation of 12.7
  • To a solution of 12.6 (2.25 g, 6 mmol) in a mixed solvent of methanol (40 mL), tetrahydrofuran (40 mL) and water (40 mL) was added lithium hydroxide (1.52 g, 36.2 mmol, 6.0 eq) in one portion. The reaction mixture was stirred at room temperature overnight. The mixture was concentrated under vacuum and extracted with diethyl ether. The aqueous phase was acidified to pH 2-3 using a 1 N aqueous solution of hydrochloric acid. The acidified solution was extracted with methylene chloride. The organics were combined, dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was used in the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 12.93 (brs, 1H), 7.59 (t, 1H), 7.15 (d, 1H), 6.97 (d, 1H), 3.71 (m, 2H), 3.12 (m, 2H), 1.90 (m, 2H), 1.65 (m, 2H), 1.40 (s, 9H)
  • Preparation of 12.8
  • To a solution of 12.7 (1.63 g, 4.5 mmol) in acetonitrile (100 mL) was added sequentially diisopropylethylamine (5.23, 30 mmol, 6.7 eq), dimethylamine (3.4j) hydrochloride (1.14 g, 14 mmol, 3.0 eq) and TBTU (1.74 g, 5.4 mmol, 1.2 eq). The reaction mixture was stirred at room temperature for 3 days and then concentrated under vacuum. The residue was dissolved in ethyl acetate and washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by column chromatography (eluent: hexane/acetone, 2:1). Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 7.50 (t, 1H), 7.00 (d, 1H), 6.85 (d, 1H), 3.89 (m, 2H), 3.22 (m, 2H), 3.14 (s, 3H), 2.74 (s, 3H), 2.03 (m, 2H), 1.62 (m, 2H), 1.48 (s, 6H)
  • Preparation of 12.9
  • To a solution of 12.8 (950 mg, 2.45 mmol) in anhydrous tetrahydrofuran (20 mL) at −78° C. under nitrogen was added drop wise a 1.0 M solution of LiHMDS in tetrahydrofuran (3.2 mL, 3.2 mmol, 1.3 eq). The reaction mixture was stirred for 45 min at −78° C. A solution of N-phenyltrifluoromethanesulfonimide (1.4) (1.15 g, 3.2 mmol, 1.3 eq) in tetrahydrofuran (8 mL) was added drop wise to the reaction mixture. The mixture was warmed slowly to room temperature and stirring was continued for an additional 2.5 h at room temperature. The mixture was then poured into ice water and extracted with a mixture of hexane and diethyl ether (1:1). The organic layer was washed with water and brine, and dried over sodium sulfate and filtered. The organic extracts were concentrated under vacuum and the crude product was purified by column chromatography (eluent: methylene chloride/ethyl acetate, 3:1). Yield: 78.6%
  • 1H NMR (400 MHz, CDCl3) δ 7.28 (t, 1H), 6.96 (d, 1H), 6.83 (d, 1H), 5.65 (s, 1H), 3.80 (m, 2H), 3.38 (m, 1H), 3.20 (m, 1H), 3.10 (s, 3H), 2.92 (s, 3H), 2.09 (m, 2H), 1.70 (m, 2H), 1.48 (s, 9H)
  • Preparation of 12.10
  • To a solution of 12.9 (950 mg, 1.83 mmol) in dimethoxyethane (DME) (16 mL) was added sequentially a 2N aqueous solution of sodium carbonate (3.1 mL, 6.2 mmol, 3.4 eq), lithium chloride (259 mg, 6.1 mmol, 3.3 eq.), 4-(N,N-diethylaminocarbonyl)phenylboronic acid (1.6) (486 mg, 2.2 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (64 mg, 0.055 mmol, 0.03 eq). The mixture was refluxed overnight under nitrogen and then cooled to room temperature. To this mixture was added water (20 mL) and the crude product was extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and filtered. The organics were concentrated under vacuum and the crude product was purified by column chromatography (eluent: hexane/acetone, 2:1). Yield: 88%
  • 1H NMR (400 MHz, CDCl3) δ 7.35 (d, 2H), 7.25 (m, 3H), 7.05 (d, 1H), 6.91 (d, 1H), 5.62 (s, 1H), 3.86 (m, 2H), 3.55 (m, 2H), 3.30 (m, 4H), 2.69 (s, 3H), 2.30 (s, 3H), 2.10 (m, 1H), 1.98 (m, 1H), 1.70 (m, 2H), 1.49 (s, 6H), 1.20 (m, 6H)
  • Preparation of 12G
  • To a solution of 12.10 (840 mg, 1.54 mmol) in methylene chloride (10 mL) was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (30 mL). The mixture was stirred at room temperature for 2 days and diluted with diethyl ether. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 9.28 (m, 2H), 7.35-7.19 (m, 6H), 6.90 (d, 1H), 5.96 (s, 1H), 3.43 (m, 2H), 3.22 (m, 6H), 2.66 (s, 3H), 2.18 (s, 3H), 2.18 (s, 3H), 2.09 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=448.4 (M+H)+
  • Example 12H
  • 12H was obtained according to a procedure similar to the one described for 12A, with the following exception:
  • Step 12.4: 1.6 was replaced by 1.7.
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (m, 2H), 8.48 (s, 1H), 7.73 (d, 1H), 7.58 (d, 1H), 7.20 (t, 1H), 6.98 (d, 1H), 6.82 (d, 1H), 6.10 (s, 1H), 3.42-3.12 (m, 8H), 2.02 (m, 4H), 1.70 (s, 3H), 1.18 (t, 3H), 1.10 (t, 3H)
  • Mass Spectral Analysis m/z=392.4 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 7/5HCl, 7/5H2O
  • Theory: % C, 61.60; % H, 7.15; % N, 8.98; % Cl, 10.61.
  • Found: % C, 61.70; % H, 6.78; % N, 8.86; % Cl, 10.73.
  • Example 12I
  • 12I was obtained according to a procedure similar to the one described for 12A, with the following exception:
  • Step 12.2: 12.2a was replaced by 12.2b.
  • 1H NMR (400 MHz, DMSO d6) δ 8.89 (brs, 2H), 7.12 (d, 2H), 7.04 (d, 2H), 6.95 (t, 1H), 6.71 (d, 1H), 6.58 (d, 1H), 5.66 (s, 1H), 3.20 (brs, 2H), 2.92 (brm, 6H), 1.75 (brm, 6H), 0.86 (brm, 8H), 0.22 (t, 3H)
  • Mass Spectral Analysis m/z=419.4 (M+H)+
  • Elemental analysis:
  • C27H34N2O2, 1HCl, 1H2O
  • Theory: % C, 68.55; % H, 7.88; % N, 5.92.
  • Found: % C, 68.42; % H, 7.73; % N, 5.92.
  • Example 12J
  • 12J was obtained according to a procedure similar to the one described for 12A, with the following exception:
  • Step 12.2: 12.2a was replaced by 12.2c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (brs, 1.5H), 7.54 (d, 2H), 7.47 (d, 2H), 7.38 (t, 1H), 7.13 (d, 1H), 7.02 (d, 1H), 6.09 (s, 1H), 3.62 (brs, 2H), 3.36 (brm, 5H), 2.18 (brm, 6H), 1.30 (brm, 8H), 1.00 (m, 2H), 0.81 (t, 3H)
  • Mass Spectral Analysis m/z=433.4 (M+H)+
  • Elemental analysis:
  • C28H36N2O2, 1HCl, 2H2O
  • Theory: % C, 66.58; % H, 8.18; % N, 5.55.
  • Found: % C, 66.82; % H, 7.88; % N, 5.59.
  • Example 12K
  • 12K was obtained according to a procedure similar to the one described for 12A, with the following exceptions:
  • Step 12.2: 12.2a was replaced by 12.2b.
  • Step 12.4: 1.6 was replaced by 1.7 and Method 12A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.73 (brs, 1H), 9.61 (brs, 1H), 8.47 (s, 1H), 7.65 (m, 2H), 7.20 (m, 1H), 6.90 (d, 1H), 6.82 (d, 1H), 5.66 (s, 1H), 3.59 (q, 2H), 3.41 (brm, 6H), 2.24 (brs, 4H), 2.01 (brm, 2H), 1.25 (brm, 8H), 0.54 (t, 3H)
  • Mass Spectral Analysis m/z=420.4 (M+H)+
  • Example 12L
  • 12L was obtained according to a procedure similar to the one described for 12A, with the following exceptions:
  • Step 12.2: 12.2a was replaced by 12.2c.
  • Step 12.4: 1.6 was replaced by 1.7 and Method 12A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.86 (brd, 1.5H), 8.43 (d, 1H), 7.66 (dd, 1H), 7.48 (d, 1H), 7.16 (t, 1H), 6.91 (d, 1H), 6.79 (d, 1H), 5.98 (s, 1H), 3.40 (q, 2H), 3.12 (brm, 5H), 1.94 (brm, 6H), 1.10 (m, 5H), 1.01 (t, 3H), 0.76 (m, 2H), 0.56 (t, 3H)
  • Mass Spectral Analysis m/z=434.3 (M+H)+
  • Example 13A Preparation of 13.2
  • To a solution of 1.5a (7.80 g, 17.35 mmol, 1.0 eq) in dimethoxyethane (75 mL) was added sequentially a 2N aqueous solution of sodium carbonate (26.03 mL, 52.06 mmol, 3.0 eq), lithium chloride (2.21 g, 52.06 mmol, 3.0 eq), 13.1 (3.44 g, 19.09 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.40 g, 0.35 mmol, 0.02 eq). The mixture was refluxed overnight under nitrogen. The mixture was then cooled to room temperature and water (250 mL) was added. The mixture was extracted with ethyl acetate. The organic layer was further washed with brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 64%
  • 1H NMR (400 MHz, DMSO d6) δ 8.02 (d, 2H), 7.49 (d, 2H), 7.23 (m, 1H), 6.99 (d, 1H), 6.92 (m, 2H), 5.92 (s, 1H), 3.88 (s, 3H), 3.70 (m, 2H), 3.27 (m, 2H), 1.89 (m, 2H), 1.71 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=436.0 (M+H)+
  • Preparation of 13.3
  • A solution of 13.2 (4.71 g, 10.81 mmol, 1.0 eq) in tetrahydrofuran (30 mL) at 0° C. under nitrogen was added drop wise to a solution of lithium hydroxide monohydrate (0.54 g, 12.98 mmol, 1.2 eq) in water (30 mL). The mixture was stirred overnight at room temperature. The mixture was then concentrated under reduced pressure and redissolved in water. The mixture was then acidified to pH 2 using concentrated hydrochloric acid. The resulting precipitate was collected by filtration and the crude product was used for the next step without further purification.
  • Yield: 98%
  • 1H NMR (400 MHz, DMSO d6) δ 13.03 (br s, 1H), 8.01 (d, 2H), 7.47 (d, 2H), 7.23 (m, 1H), 6.98 (d, 1H), 6.92 (m, 2H), 5.91 (s, 1H), 3.70 (m, 2H), 3.28 (m, 2H), 1.86 (m, 2H), 1.72 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=420.1 (M−H)
  • Preparation of 13A
  • Trifluoroacetic acid (0.15 mL, 1.96 mmol, 5.5 eq) was added drop wise to a cold (0° C.) solution of 13.3 (0.15 g, 0.36 mmol, 1.0 eq) in anhydrous dichloromethane (5 mL). The mixture was warmed to room temperature and stirred overnight at room temperature. The mixture was then concentrated under reduced pressure. The crude product was triturated with diethyl ether. The resulting precipitate was collected by filtration. Yield: 87%
  • 1H NMR (400 MHz, DMSO d6) δ 13.05 (brs, 1H), 8.67 (m, 2H), 8.02 (d, 2H), 7.49 (d, 2H), 7.27 (m, 1H), 7.05 (d, 1H), 6.96 (m, 2H), 5.98 (s, 1H), 3.26 (m, 4H), 2.08 (m, 2H), 1.97 (m, 2H)
  • Mass Spectral Analysis m/z=322.1 (M+H)+
  • Elemental analysis:
  • C20H19NO3, CF3CO2H, 0.2H2O
  • Theory: % C, 60.19; % H, 4.68; % N, 3.19.
  • Found: % C, 60.18; % H, 4.61; % N, 3.24.
  • Example 13B Preparation of 13.5a
  • O-Benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (150.8 mg, 0.47 mmol, 1.1 eq) was added to a cooled (0° C.) solution of 13.3 (180.0 mg, 0.43 mmol, 1.0 eq), 3.4a (50.3 mg, 0.94 mmol, 2.2 eq), and N,N-diisopropylethylamine (0.25 mL, 0.94 mmol, 2.2 eq) in acetonitrile (5 mL). The solution was stirred overnight at room temperature and then concentrated under reduced pressure. Ethyl acetate (10 mL) and a saturated aqueous solution of sodium bicarbonate (10 mL) were added to the crude product and the mixture was stirred for 20 min at room temperature. The phases were separated and the organic phase was washed with a saturated aqueous solution of sodium bicarbonate, brine, dried over sodium sulfate and filtered. The organics were concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 10%
  • Mass Spectral Analysis m/z=421.2 (M+H)+
  • Preparation of 13B
  • A 2.0M solution of hydrochloric acid in diethyl ether (0.12 mL, 0.24 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 13.5a (18 mg, 0.04 mmol, 1.0 eq) in anhydrous methanol (5 mL). The mixture was stirred overnight at room temperature and then concentrated under reduced pressure. The crude product was triturated with ethyl acetate. The resulting precipitate was collected by filtration.
  • Yield: 70%
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (m, 2H), 8.06 (m, 1H), 7.95 (m, 2H), 7.46 (m, 3H), 7.27 (m, 1H), 7.06 (m, 1H), 6.96 (m, 2H), 5.95 (s, 1H), 3.24 (m, 4H), 2.08 (m, 4H)
  • Mass Spectral Analysis m/z=321.1 (M+H)+
  • Example 13C
  • 13C was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.05 (m, 2H), 8.55 (m, 1H), 7.92 (m, 2H), 7.41 (m, 2H), 7.26 (m, 1H), 7.06 (m, 1H), 6.95 (m, 2H), 5.95 (s, 1H), 3.20 (m, 4H), 2.81 (m, 3H), 2.08 (m, 4H)
  • Mass Spectral Analysis m/z=335.2 (M+H)+
  • Example 13D
  • 13D was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 8.50 (m, 1H), 7.90 (d, 2H), 7.40 (d, 2H), 7.20 (m, 1H), 6.90 (m, 3H), 5.85 (s, 1H), 3.30 (m, 2H), 2.90 (m, 2H), 2.70 (m, 2H), 1.85-1.70 (m, 4H), 1.10 (t, 3H)
  • Mass Spectral Analysis m/z=349.2 (M+H)+
  • Elemental analysis:
  • C22H24N2O2, 0.25 (CH3)2CO3, 0.25H2O
  • Theory: % C, 70.89; % H, 7.32; % N, 7.27.
  • Found: % C, 71.13; % H, 7.04; % N, 7.07.
  • Example 13E
  • 13E was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4e.
  • 1H NMR (400 MHz, CDCl3) δ 9.75 (brs, 1H), 9.31 (brs, 1H), 7.81 (d, 2H), 7.39 (d, 2H), 7.21 (m, 1H), 6.98 (m, 2H), 6.90 (m, 1H), 6.25 (m, 1H), 5.56 (s, 1H), 3.46 (m, 2H), 3.33 (m, 4H), 2.30 (m, 2H), 2.12 (m, 2H), 1.94 (m, 1H), 1.04 (d, 6H)
  • Mass Spectral Analysis m/z=377.2 (M+H)+
  • Example 13F
  • 13F was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4j.
  • 1H NMR (400 MHz, DMSO d6) δ 9.08 (m, 2H), 7.42 (m, 4H), 7.24 (m, 1H), 7.00 (m, 3H), 5.91 (s, 1H), 3.25 (m, 4H), 2.96 (m, 6H), 2.07 (m, 4H)
  • Mass Spectral Analysis m/z=349.1 (M+H)+
  • Example 13G
  • 13G was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4 k.
  • 1H NMR (400 MHz, DMSO d6) δ 8.91 (m, 2H), 7.58 (d, 2H), 7.41 (d, 2H), 7.25 (m, 1H), 7.00 (m, 3H), 5.92 (s, 1H), 3.49 (m, 2H), 3.41 (m, 2H), 3.24 (m, 4H), 2.09 (m, 2H), 2.00 (m, 2H), 1.84 (m, 4H)
  • Mass Spectral Analysis m/z=375.1 (M+H)+
  • Example 13H
  • 13H was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4o.
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (m, 2H), 7.39 (dd, 4H), 7.24 (m, 1H), 6.95 (m, 3H), 5.91 (s, 1H), 3.66 (brs, 2H), 3.22 (m, 4H), 2.10 (m, 4H), 1.30 (m, 12H)
  • Mass Spectral Analysis m/z=405.3 (M+H)+
  • Elemental analysis:
  • C26H32N2O2, 1HCl, 0.5H2O
  • Theory: % C, 69.39; % H, 7.62; % N, 6.22.
  • Found: % C, 69.31; % H, 7.64; % N, 6.19.
  • Example 13I
  • 13I was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4p.
  • 1H NMR (400 MHz, DMSO d6) δ 8.91 (m, 2H), 7.46 (m, 4H), 7.26 (m, 1H), 7.01 (m, 3H), 5.94 (s, 1H), 3.61 (m, 6H), 3.35 (m, 2H), 3.21 (m, 4H), 2.09 (m, 2H), 1.98 (m, 2H)
  • Mass Spectral Analysis m/z=391.1 (M+H)+
  • Example 13J
  • 13J was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 3.4q.
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (m, 2H), 7.44 (m, 4H), 7.26 (m, 1H), 7.00 (m, 3H), 5.91 (s, 1H), 3.59 (m, 2H), 3.21 (m, 6H), 2.09 (m, 2H), 1.99 (m, 2H), 1.55 (m, 6H)
  • Mass Spectral Analysis m/z=389.1 (M+H)+
  • Example 13K
  • 13K was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4a.
  • 1H NMR (400 MHz, DMSO d6) δ 8.75 (m, 2H), 7.49 (m, 2H), 7.41 (m, 2H), 7.26 (m, 1H), 7.05 (m, 1H), 6.97 (m, 2H), 5.95 (s, 1H), 4.00 (brm, 4H), 3.23 (m, 4H), 2.10 (m, 2H), 1.97 (m, 2H), 1.64 (m, 2H), 1.15 (brm, 6H)
  • Mass Spectral Analysis m/z=403.3 (M+H)+
  • Elemental analysis:
  • C26H30N2O2, 1HCl, 0.3H2O
  • Theory: % C, 70.27; % H, 7.17; % N, 6.30.
  • Found: % C, 70.02; % H, 7.04; % N, 6.27.
  • Example 13L
  • 13L was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4b.
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (m, 2H), 7.70 (d, 2H), 7.50 (d, 2H), 7.40 (m, 1H), 7.30 (m, 4H), 7.00 (m, 3H), 5.95 (s, 1H), 4.90 (s, 2H), 4.80 (s, 2H), 3.30 (brm, 4H), 2.05 (m, 4H)
  • Mass Spectral Analysis m/z=423.1 (M+H)+
  • Elemental analysis:
  • C28H26N2O2, 1HCl, 1H2O
  • Theory: % C, 70.50; % H, 6.13; % N, 5.87.
  • Found: % C, 70.58; % H, 5.95; % N, 5.89.
  • Example 13M
  • 13M was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (m, 1H), 7.40 (m, 4H), 7.25 (m, 1H), 7.00 (m, 3H), 5.90 (s, 1H), 3.55-3.05 (m, 8H), 2.05 (m, 4H), 1.60 (m, 2H), 1.10 (m, 1H), 0.90 (m, 2H), 0.65 (m, 1H), 0.40 (m, 2H), 0.15 (m, 1H), 0.10 (m, 1H)
  • Mass Spectral Analysis m/z=417.2 (M+H)+
  • Elemental analysis:
  • C27H32N2O2, 1HCl, 0.4H2O
  • Theory: % C, 70.46; % H, 7.40; % N, 6.09.
  • Found: % C, 70.54; % H, 7.30; % N, 6.15.
  • Example 13N
  • 13N was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4d.
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (m, 2H), 7.40 (brm, 10H), 7.00 (m, 3H), 5.94 (s, 1H), 4.70 (m, 1H), 4.52 (m, 1H), 3.21 (m, 4H), 2.88 (m, 3H), 2.02 (m, 4H)
  • Mass Spectral Analysis m/z=425.2 (M+H)+
  • Elemental analysis:
  • C28H28N2O2, 1HCl, 0.6H2O
  • Theory: % C, 71.28; % H, 6.45; % N, 5.94.
  • Found: % C, 71.13; % H, 6.51; % N, 5.97.
  • Example 13O
  • 13O was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4e.
  • 1H NMR (400 MHz, DMSO d6) δ 8.65 (m, 2H), 7.45 (m, 4H), 7.26 (m, 1H), 7.00 (m, 3H), 5.95 (s, 1H), 4.36 (m, 2H), 4.11 (m, 2H), 3.88 (m, 2H), 3.60 (m, 2H), 3.00 (m, 2H), 2.65 (m, 1H), 2.09 (m, 2H), 1.99 (m, 4H), 1.52 (m, 2H), 1.19 (m, 3H)
  • Mass Spectral Analysis m/z=461.2 (M+H)+
  • Example 13P
  • 13P was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4f.
  • 1H NMR (400 MHz, DMSO d6) δ 8.60 (m, 2H), 7.47 (m, 4H), 7.25 (m, 1H), 7.00 (m, 3H), 5.95 (s, 1H), 4.18 (m, 2H), 3.80 (brs, 4H), 3.24 (m, 2H), 3.00 (s, 3H), 2.10 (m, 2H), 1.94 (m, 2H), 1.20 (m, 3H)
  • Mass Spectral Analysis m/z=421.2 (M+H)+
  • Example 13Q
  • 13Q was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4 g.
  • 1H NMR (400 MHz, DMSO d6) δ 10.32 (brs, 1H), 8.80 (m, 2H), 7.54 (m, 2H), 7.46 (m, 2H), 7.27 (m, 1H), 7.00 (m, 3H), 5.92 (s, 1H), 4.54 (brs, 2H), 3.84 (brs, 2H), 3.45 (m, 2H), 3.24 (m, 4H), 3.12 (m, 2H), 2.83 (s, 3H), 2.10 (m, 2H), 1.97 (m, 2H)
  • Mass Spectral Analysis m/z=404.3 (M+H)+
  • Example 13R
  • 13R was obtained according to a procedure similar to the one described for 13B, with the following exception:
  • Step 13.6: 3.4a was replaced by 13.4h.
  • 1H NMR (400 MHz, DMSO d6) δ 9.55 (m, 1H), 8.95 (m, 1H), 7.55 (m, 5H), 7.30 (brm, 10H), 7.04 (m, 1H), 6.95 (m, 2H), 5.93 (s, 1H), 4.62 (s, 2H), 4.46 (s, 2H), 3.20 (m, 4H), 2.02 (m, 4H)
  • Mass Spectral Analysis m/z=501.2 (M+H)+
  • Example 13S Preparation of 13S
  • A 2N aqueous solution of sodium hydroxide (1.0 mL, 2 mmol, 9.2 eq) was added to a solution of 13O (0.10 g, 0.22 mmol, 1.0 eq) in tetrahydrofuran (5 mL) and anhydrous absolute ethanol (1 mL). The mixture was stirred for 10 h at room temperature and acidified to pH 6 using a 2N aqueous solution of hydrochloric acid. The mixture was concentrated under reduced pressure. The crude product was dissolved in dichloromethane. The mixture was filtered and the filtrate was concentrated under reduced pressure. Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 7.43 (m, 4H), 7.25 (m, 1H), 7.01 (m, 2H), 6.94 (m, 1H), 5.93 (s, 1H), 4.33 (br s, 2H), 3.65-2.90 (m, 9H), 1.91 (m, 6H), 1.52 (m, 2H)
  • Mass Spectral Analysis m/z=433.1 (M+H)+
  • Example 14A Preparation of 14.2
  • To a solution of 1.5a (5.00 g, 11.12 mmol, 1.0 eq) in dimethoxyethane (17 mL) was added sequentially a 2N aqueous solution of sodium carbonate (16.69 mL, 33.37 mmol, 3.0 eq), lithium chloride (1.41 g, 33.37 mmol, 3.0 eq), 14.1 (1.80 g, 12.24 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.26 g, 0.22 mmol, 0.02 eq). The mixture was refluxed for 10 h under nitrogen. The mixture was then cooled to room temperature and a 1N aqueous solution of sodium hydroxide was added. The mixture was extracted with dichloromethane. The organic layer was further washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was triturated with diethyl ether. The resulting solid was collected bu filtration. Yield: 78%
  • 1H NMR (400 MHz, DMSO d6) δ 7.90 (d, 2H), 7.50 (d, 2H), 7.20 (m, 1H), 7.00 (m, 1H), 6.90 (m, 2H), 5.95 (s, 1H), 3.70 (m, 2H), 3.25 (m, 2H), 1.85 (m, 2H), 1.70 (m, 2H), 1.40 (s, 9H)
  • Mass Spectral Analysis m/z=403.1 (M+H)+
  • Preparation of 14.4
  • A mixture of 14.2 (3.49 g, 8.67 mmol, 1.0 eq), 14.3 (1.13 g, 17.34 mmol, 2.0 eq) and zinc bromide (0.98 g, 4.34 mmol, 0.5 eq) in isopropanol (70 mL) and water (50 mL) was refluxed for 3 days. The reaction mixture was then cooled to 0° C. and acidified to pH 1 using a 3N aqueous solution of hydrochloric acid. The mixture was extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. Diethyl ether (30 mL) was added. The resulting precipitate was collected by filtration and washed with diethyl ether. The crude compound was used for the next step without further purification. Yield: 89%
  • 1H NMR (400 MHz, DMSO d6) δ 8.10 (d, 2H), 7.55 (d, 2H), 7.20 (m, 1H), 7.00 (m, 2H), 6.90 (m, 1H), 5.90 (s, 1H), 3.70 (m, 2H), 3.30 (m, 2H), 1.90 (m, 2H), 1.70 (m, 2H), 1.40 (s, 9H)
  • Mass Spectral Analysis m/z=446.0 (M+H)+
  • Preparation of 14A
  • A 2.0M solution of hydrochloric acid in diethyl ether (21.3 mL, 42.58 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 14.4 (3.71 g, 7.74 mmol, 1.0 eq) in anhydrous dichloromethane (25 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. Diethyl ether (100 mL) was added to the solution. The resulting precipitate was collected by filtration and washed with diethyl ether. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 20%
  • 1H NMR (400 MHz, DMSO d6) δ 9.08 (brs, 2H), 8.16 (d, 2H), 7.61 (d, 2H), 7.28 (m, 1H), 7.02 (m, 3H), 6.02 (s, 1H), 3.59 (brs, 1H), 3.24 (m, 4H), 2.06 (m, 4H)
  • Mass Spectral Analysis m/z=346.1 (M+H)+
  • Elemental analysis:
  • C20H19N5O, 1HCl, 0.5H2O
  • Theory: % C, 61.46; % H, 5.42; % N, 17.92.
  • Found: % C, 61.52; % H, 5.23; % N, 17.63.
  • Example 14B Preparation of 14.5 and 14.6
  • Methyl iodide (2.8c) (0.35 mL, 0.0056 mol, 5.0 eq) was added drop wise to a solution of 14.4 (0.500 g, 0.0011 mol, 1.0 eq) and triethylamine (0.80 mL, 0.0056 mol, 5.0 eq) in anhydrous dimethylformamide (5 mL) and the mixture was stirred at room temperature for 3 days. The mixture was poured into water (50 mL) and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield 14.5 (major regioisomer): 65%; Mass Spectral Analysis m/z=460.1 (M+H)+
  • Yield 14.6 (minor regioisomer): 17%; Mass Spectral Analysis m/z=460.2 (M+H)+
  • Preparation of 14B
  • A 2.0M anhydrous solution of hydrochloric acid in diethyl ether (10 mL) was added drop wise to a cold (0° C.) solution of 14.5 (0.330 g, 0.00071 mol, 1.0 eq) in anhydrous dichloromethane (10 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 h at room temperature. The mixture was concentrated under reduced pressure and diethyl ether was added to the residue. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 90%
  • 1H NMR (400 MHz, DMSO d6) δ 8.80 (m, 1H), 8.10 (d, 2H), 7.55 (d, 2H), 7.25 (t, 1H), 6.90-7.10 (m, 3H), 6.00 (s, 1H), 4.45 (s, 3H), 3.15-3.40 (m, 4H), 1.95-2.15 (m, 4H)
  • Mass Spectral Analysis m/z=360.1 (M+H)+
  • Example 14C Preparation of 14C
  • A 2.0M anhydrous solution of hydrochloric acid in diethyl ether (5 mL) was added drop wise to a cold (0° C.) solution of 14.6 (0.090 g, 0.00019 mol, 1.0 eq) in anhydrous dichloromethane (10 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. The mixture was concentrated under reduced pressure and diethyl ether was added to the residue. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 88%
  • 1H NMR (400 MHz, DMSO d6) δ 8.80 (m, 1.5H), 7.90 (d, 2H), 7.60 (d, 2H), 7.25 (t, 1H), 6.90-7.10 (m, 3H), 6.00 (s, 1H), 4.20 (s, 3H), 3.20 (m, 4H), 1.95-2.15 (m, 4H)
  • Mass Spectral Analysis m/z=360.2 (M+H)+
  • Example 15A
  • 15A was obtained according to a procedure similar to the one described for 15C, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1a.
  • 1H NMR (400 MHz, DMSO d6) δ 8.87 (brm, 1H), 8.16 (d, 2H), 7.59 (d, 2H), 7.29 (m, 1H), 7.06 (m, 2H), 6.97 (m, 1H), 6.02 (s, 1H), 5.96 (s, 2H), 3.77 (s, 3H), 3.23 (brm, 4H), 2.11 (brm, 2H), 2.00 (brm, 2H)
  • Mass Spectral Analysis m/z=418.1 (M+H)+
  • Example 15B
  • 15B was obtained according to a procedure similar to the one described for 15C, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1b.
  • 1H NMR (400 MHz, DMSO d6) δ 8.75 (m, 1H), 8.15 (d, 2H), 7.57 (d, 2H), 7.25 (t, 1H), 7.00 (m, 3H), 6.00 (s, 1H), 5.00 (t, 2H), 3.60 (s, 3H), 3.10-3.40 (m, 6H), 1.95-2.18 (m, 4H)
  • Mass Spectral Analysis m/z=432.2 (M+H)+
  • Example 15C Preparation of 15.2a and 15.3a
  • Ethyl bromobutyrate (15.1c) (0.40 mL, 0.0028 mol, 2.5 eq) was added drop wise to a solution of 14.4 (0.500 g, 0.0011 mol, 1.0 eq) and triethylamine (0.40 mL, 0.0028 mol, 2.5 eq) in anhydrous N,N-dimethylformamide and the mixture was stirred at room temperature for 3 days. The mixture was poured into water (50 mL) and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield 15.2a (major regioisomer): 82%; Mass Spectral Analysis m/z=560.2 (M+H)+
  • (15.2a) 1H NMR (400 MHz, DMSO d6) δ 8.10 (d, 2H), 7.50 (d, 2H), 7.20 (m, 1H), 7.00 (m, 2H), 6.90 (m, 1H), 5.90 (s, 1H), 4.70 (t, 2H), 4.00 (q, 2H), 3.70 (m, 2H), 3.30 (m, 2H), 2.40 (m, 2H), 2.10 (m, 2H), 1.90 (m, 2H), 1.70 (m, 2H), 1.40 (s, 9H), 1.15 (t, 3H)
  • Yield 15.3a (minor regioisomer): 6%; Mass Spectral Analysis m/z=560.2 (M+H)+
  • (15.3a) 1H NMR (400 MHz, DMSO d6) δ 7.90 (d, 2H), 7.60 (d, 2H), 7.20 (m, 1H), 7.00 (m, 2H), 6.90 (m, 1H), 5.95 (s, 1H), 4.55 (t, 2H), 4.00 (q, 2H), 3.70 (m, 2H), 3.30 (m, 2H), 2.40 (m, 2H), 2.10 (m, 2H), 1.90 (m, 2H), 1.70 (m, 2H), 1.40 (s, 9H), 1.10 (t, 3H)
  • Preparation of 15C
  • A 2.0M anhydrous solution of hydrochloric acid in diethyl ether (10 mL) was added drop wise to a cold (0° C.) solution of 15.2a (0.520 g, 0.00092 mol, 1.0 eq) in anhydrous dichloromethane (10 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. An additional amount of a 2.0M anhydrous solution of hydrochloric acid in diethyl ether (10 mL) was added to the mixture, which was stirred for an additional 6 h at room temperature. The mixture was concentrated under reduced pressure and diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 70%
  • 1H NMR (400 MHz, DMSO d6) δ 8.80 (m, 1H), 8.15 (d, 2H), 7.60 (d, 2H), 7.25 (m, 1H), 7.00 (m, 3H), 6.00 (s, 1H), 4.80 (t, 2H), 4.00 (q, 2H), 3.35 (m, 2H), 3.20 (m, 2H), 2.40 (m, 2H), 2.20 (m, 2H), 2.10 (m, 2H), 1.95 (m, 2H), 1.15 (t, 3H)
  • Mass Spectral Analysis m/z=460.2 (M+H)+
  • Example 15D
  • 15D was obtained according to a procedure similar to the one described for 15C, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1d.
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (brm, 1.5H), 8.14 (d, 2H), 7.57 (d, 2H), 7.28 (t, 1H), 7.04 (m, 2H), 6.96 (m, 1H), 6.00 (s, 1H), 4.78 (t, 2H), 4.04 (q, 2H), 3.22 (brm, 4H), 2.37 (t, 2H), 2.11 (brm, 2H), 2.01 (brm, 4H), 1.57 (m, 2H), 1.16 (t, 3H)
  • Mass Spectral Analysis m/z=474.2 (M+H)+
  • Example 15E
  • 15E was obtained according to a procedure similar to the one described for 15C, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1e.
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (brm, 1.5H), 8.14 (d, 2H), 7.57 (d, 2H), 7.28 (t, 1H), 7.05 (m, 2H), 6.96 (m, 1H), 6.00 (s, 1H), 4.76 (t, 2H), 4.02 (q, 2H), 3.22 (brm, 4H), 2.29 (t, 2H), 2.10 (brm, 2H), 2.00 (brm, 4H), 1.57 (m, 2H), 1.30 (m, 2H), 1.14 (t, 3H)
  • Mass Spectral Analysis m/z=488.2 (M+H)+
  • Example 15F
  • 15F was obtained according to a procedure similar to the one described for 15H, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1a.
  • 1H NMR (400 MHz, DMSO d6) δ 8.86 (brm, 1H), 7.84 (d, 2H), 7.62 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.99 (m, 2H), 6.03 (s, 1H), 5.71 (s, 2H), 3.70 (s, 3H), 3.23 (m, 4H), 2.11 (brm, 2H), 2.00 (brm, 2H)
  • Mass Spectral Analysis m/z=418.2 (M+H)+
  • Example 15G
  • 15G was obtained according to a procedure similar to the one described for 15H, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1b.
  • 1H NMR (400 MHz, DMSO d6) δ 8.78 (brm, 1H), 7.91 (d, 2H), 7.64 (d, 2H), 7.29 (m, 1H), 7.05 (m, 2H), 6.98 (m, 1H), 6.04 (s, 1H), 4.71 (t, 2H), 3.56 (s, 3H), 3.23 (m, 4H), 3.11 (t, 2H), 2.12 (brm, 2H), 2.00 (brm, 2H)
  • Mass Spectral Analysis m/z=432.1 (M+H)+
  • Example 15H Preparation of 15H
  • A 2.0M anhydrous solution of hydrochloric acid in diethyl ether (10 mL) was added drop wise to a cold (0° C.) solution of 15.3a (0.030 g, 0.000053 mol, 1.0 eq) in anhydrous dichloromethane (10 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. An additional amount of a 2.0M anhydrous solution of hydrochloric acid in diethyl ether (10 mL) was added to the mixture, which was stirred for an additional 6 h at room temperature. The mixture was concentrated under reduced pressure and diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 57%
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (m, 1.5H), 7.90 (d, 2H), 7.62 (d, 2H), 7.30 (m, 1H), 7.05 (m, 2H), 6.95 (m, 1H), 6.00 (s, 1H), 4.60 (t, 2H), 4.00 (q, 2H), 3.25 (m, 4H), 2.40 (m, 2H), 2.10 (m, 6H), 1.15 (t, 3H)
  • Mass Spectral Analysis m/z=460.2 (M+H)+
  • Example 15I
  • 15I was obtained according to a procedure similar to the one described for 15H, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1d.
  • 1H NMR (400 MHz, DMSO d6) δ 8.96 (brm, 1.5H), 7.89 (d, 2H), 7.63 (d, 2H), 7.29 (t, 1H), 7.06 (m, 2H), 6.97 (m, 1H), 6.03 (s, 1H), 4.55 (t, 2H), 4.01 (q, 2H), 3.22 (brm, 4H), 2.29 (t, 2H), 2.12 (brm, 2H), 2.02 (brm, 2H), 1.85 (m, 2H), 1.49 (m, 2H), 1.13 (t, 3H)
  • Mass Spectral Analysis m/z=474.3 (M+H)+
  • Example 15J
  • 15J was obtained according to a procedure similar to the one described for 15H, with the following exception:
  • Step 15.1: 15.1c was replaced by 15.1e.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brm, 1H), 7.87 (d, 2H), 7.62 (d, 2H), 7.29 (t, 1H), 7.05 (m, 2H), 6.97 (m, 1H), 6.03 (s, 1H), 4.52 (t, 2H), 4.01 (q, 2H), 3.23 (brm, 4H), 2.22 (t, 2H), 2.11 (brm, 2H), 2.02 (brm, 2H), 1.83 (m, 2H), 1.47 (m, 2H), 1.23 (m, 2H), 1.14 (t, 3H)
  • Mass Spectral Analysis m/z=488.3 (M+H)+
  • Example 15K
  • 15K was obtained according to a procedure similar to the one described for 15L, with the following exception:
  • Step 15.6: 15C was replaced by 15A.
  • 1H NMR (400 MHz, DMSO d6) δ 8.18 (d, 2H), 7.60 (d, 2H), 7.29 (t, 1H), 7.06 (t, 2H), 6.97 (m, 1H), 6.02 (s, 1H), 5.80 (s, 2H), 3.27 (brm, 4H), 2.13 (brm, 2H), 2.00 (brm, 2H)
  • Mass Spectral Analysis m/z=404.1 (M+H)+
  • Example 15L Preparation of 15L
  • A 2N aqueous solution of sodium hydroxide (1.8 mL, 0.0036 mol, 5.5 eq) was added to a solution of 15C (0.300 g, 0.00060 mol, 1.0 eq) in tetrahydrofuran (10 mL) and absolute ethanol (1 mL). The mixture was stirred for 10 h at room temperature and acidified to pH 6 using a 2N aqueous solution of hydrochloric acid. The mixture was concentrated under reduced pressure and diethyl ether was added. The mixture was then stirred for 1 h at room temperature. The resulting precipitate was collected by filtration and washed several times with water and diethyl ether. Yield: 98%
  • 1H NMR (400 MHz, DMSO d6+CF3CO2D) δ 8.80 (m, 1H), 8.20 (m, 2H), 7.70 (m, 2H), 7.30 (m, 1H), 7.00 (m, 3H), 6.00 (s, 1H), 4.80 (m, 2H), 3.30 (m, 4H), 2.60-1.95 (m, 8H)
  • Mass Spectral Analysis m/z=432.1 (M+H)+
  • Example 15M
  • 15M was obtained according to a procedure similar to the one described for 15L, with the following exception:
  • Step 15.6: 15C was replaced by 15D.
  • 1H NMR (400 MHz, DMSO d6) δ 8.76 (brm 1H), 8.16 (d, 2H), 7.58 (d, 2H), 7.29 (t, 1H), 7.06 (t, 2H), 6.97 (m, 1H), 6.00 (s, 1H), 4.78 (t, 2H), 3.24 (m, 4H), 2.31 (t, 2H), 2.13 (brm, 2H), 2.01 (brm, 4H), 1.56 (m, 2H)
  • Mass Spectral Analysis m/z=446.2 (M+H)+
  • Example 15N
  • 15N was obtained according to a procedure similar to the one described for 15L, with the following exception:
  • Step 15.6: 15C was replaced by 15E.
  • 1H NMR (400 MHz, DMSO d6) δ 8.62 (brm, 1.5H), 8.15 (d, 2H), 7.57 (d, 2H), 7.28 (m, 1H), 7.05 (m, 2H), 6.97 (m, 1H), 6.00 (s, 1H), 4.76 (t, 2H), 3.25 (brm, 4H), 2.21 (t, 2H), 2.11 (brm, 2H), 1.98 (brm, 4H), 1.55 (m, 2H), 1.31 (m, 2H)
  • Mass Spectral Analysis m/z=460.2 (M+H)+
  • Example 16A
  • 16A was obtained according to a procedure similar to the one described for 14A, with the following exception:
  • Step 14.1: 14.1 was replaced by 16.1 (see also step 16.1).
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (brs, 2H), 8.12 (t, 2H), 7.70 (t, 1H), 7.60 (t, 1H), 7.25 (t, 1H), 7.00 (m, 3H), 6.00 (s, 1H), 3.30 (m, 4H), 2.05 (m, 4H)
  • Mass Spectral Analysis m/z=346.1 (M+H)+
  • Example 16B
  • 16B was obtained according to a procedure similar to the one described for 14B, with the following exception:
  • Step 14.1: 14.1 was replaced by 16.1 (see also step 16.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.66 (brm, 2H), 8.11 (m, 1H), 8.01 (m, 1H), 7.66 (t, 1H), 7.54 (m, 1H), 7.28 (m, 1H), 7.06 (d, 1H), 6.97 (m, 2H), 6.00 (s, 1H), 4.43 (s, 3H), 3.23 (brm, 4H), 2.12 (brm, 2H), 2.00 (brm, 2H)
  • Mass Spectral Analysis m/z=360.1 (M+H)+
  • Example 16C
  • 16C was obtained according to a procedure similar to the one described for 14C, with the following exception:
  • Step 14.1: 14.1 was replaced by 16.1 (see also step 16.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.73 (brm, 2H), 7.91 (m, 1H), 7.83 (t, 1H), 7.72 (t, 1H), 7.03 (m, 1H), 7.28 (m, 1H), 7.05 (m, 2H), 6.96 (m, 1H), 6.02 (s, 1H), 4.20 (s, 3H), 3.23 (brm, 4H), 2.11 (brm, 2H), 1.99 (brm, 2H)
  • Mass Spectral Analysis m/z=360.1 (M+H)+
  • Example 17A
  • 17A was obtained according to a procedure similar to the one described for 15A, with the following exception:
  • Step 15.1: 14.4 was replaced by 16.3 (see also step 17.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brs, 1.5H), 8.13 (m, 1H), 8.03 (t, 1H), 7.68 (t, 1H), 7.56 (m, 1H), 7.28 (m, 1H), 7.07 (m, 1H), 6.97 (m, 2H), 6.01 (s, 1H), 5.94 (s, 2H), 3.75 (s, 3H), 3.22 (brm, 4H), 2.12 (brm, 2H), 2.02 (brm, 2H)
  • Mass Spectral Analysis m/z=418.1 (M+H)+
  • Example 17B
  • 17B was obtained according to a procedure similar to the one described for 15C, with the following exception:
  • Step 15.1: 14.4 was replaced by 16.3 (see also step 17.1).
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (brs, 2H), 8.11 (m, 1H), 8.01 (t, 1H), 7.66 (t, 1H), 7.54 (m, 1H), 7.28 (m, 1H), 7.07 (dd, 1H), 6.96 (m, 2H), 5.99 (s, 1H), 4.79 (t, 2H), 4.03 (q, 2H), 3.22 (brm, 4H), 2.42 (t, 2H), 2.21 (m, 2H), 2.09 (brm, 4H), 1.16 (t, 3H)
  • Mass Spectral Analysis m/z=460.2 (M+H)+
  • Example 17C
  • 17C was obtained according to a procedure similar to the one described for 15F, with the following exceptions:
  • Step 15.1: 14.4 was replaced by 16.3 (see also step 17.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.95 (brs, 2H), 7.80 (m, 1H), 7.69 (m, 3H), 7.28 (m, 1H), 7.06 (d, 1H), 6.97 (m, 2H), 5.99 (s, 1H), 5.70 (s, 2H), 3.64 (s, 3H), 3.23 (brm, 4H), 2.10 (brm, 2H), 2.01 (brm, 2H)
  • Mass Spectral Analysis m/z=418.1 (M+H)+
  • Example 17D
  • 17D was obtained according to a procedure similar to the one described for 15C, with the following exceptions:
  • Step 15.1: 14.4 was replaced by 16.3 (see also step 17.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.37 (dt, 1H), 8.30 (t, 1H), 7.81 (t, 1H), 7.71 (dt, 1H), 7.44 (m, 1H), 7.22 (m, 2H), 7.10 (m, 1H), 5.98 (s, 1H), 5.47 (t, 2H), 4.22 (brs, 2H), 4.15 (t, 2H), 4.02-3.46 (brm, 10H), 2.48 (brm, 2H), 2.22 (brm, 2H)
  • Mass Spectral Analysis m/z=459.2 (M+H)+
  • Example 17E
  • 17E was obtained according to a procedure similar to the one described for 15K, with the following exceptions:
  • Step 15.1: 14.4 was replaced by 16.3 (see also step 17.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.87 (brm, 2H), 8.13 (dt, 1H), 8.03 (t, 1H), 7.68 (t, 1H), 7.56 (m, 1H), 7.28 (m, 1H), 7.07 (d, 1H), 6.98 (m, 2H), 6.01 (s, 1H), 5.77 (s, 2H), 3.24 (brm, 4H), 2.12 (brm, 2H), 2.02 (brm, 2H)
  • Mass Spectral Analysis m/z=404.1 (M+H)+
  • Example 17F
  • 17F was obtained according to a procedure similar to the one described for 15L, with the following exception:
  • Step 15.1: 14.4 was replaced by 16.3 (see also step 17.1).
  • 1H NMR (400 MHz, DMSO d6) δ 8.11 (dt, 1H), 8.01 (m, 1H), 7.66 (t, 1H), 7.54 (dt, 1H), 7.28 (m, 1H), 7.07 (d, 1H), 6.97 (m, 2H), 5.99 (s, 1H), 4.78 (t, 2H), 3.21 (brm, 4H), 2.34 (t, 2H), 2.18 (m, 2H), 2.10 (brm, 4H)
  • Mass Spectral Analysis m/z=432.1 (M+H)+
  • Example 18A Preparation of 18.2
  • A mixture of 13.5a (0.300 g, 0.00071 mole, 1.0 eq), and the Lawesson's reagent (18.1) (0.288 g, 0.00071 mole, 1 eq) in toluene (10 mL) was refluxed for 6 h. The mixture was cooled to room temperature, poured onto a saturated aqueous solution of sodium bicarbonate (50 mL) and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. Diethyl ether was added to the mixture, which was stirred at room temperature for 1 h. The resulting precipitate was collected by filtration, washed with diethyl ether and used for the next step without further purification. Yield: 64%
  • Mass Spectral Analysis m/z=434.93 (M−H)
  • Preparation of 18.4a
  • A mixture of 18.2 (1 g, 0.0022 mole, 1.0 eq) and 1-bromopinacolone (18.3a) (0.30 mL, 0.0022 mole, 1.0 eq) in N,N-dimethylformamide (5 mL) was stirred at room temperature for 48 h. The mixture was poured into a saturated aqueous solution of sodium bicarbonate and extracted with ethyl acetate. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 55%
  • 1H NMR (400 MHz, DMSO d6) δ 8.00 (d, 2H), 7.45 (d, 2H), 7.35 (s, 1H), 7.20 (t, 1H), 7.00 (d, 2H), 6.90 (t, 1H), 5.90 (s, 1H), 3.70 (m, 2H), 3.30 (m, 2H), 1.90 (m, 2H), 1.70 (m, 2H), 1.30 (s, 9H), 1.35 (s, 9H)
  • Mass Spectral Analysis m/z=517.2 (M+H)+
  • Preparation of 18A
  • To a cold (0° C.) solution of 18.4a (0.600 g, 0.0011 mole, 1.0 eq) in anhydrous dichloromethane (20 mL) was added drop wise a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (5.8 mL, 0.0011 mole, 10.0 eq). The mixture was warmed slowly to room temperature and stirring was continued for 12 h. The mixture was concentrated under reduced pressure. Diethyl ether was then added to the mixture, which was stirred for 1 h at room temperature. The precipitate was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 80%
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (s, 2H), 8.00 (d, 2H), 7.50 (d, 2H), 7.40 (s, 1H), 7.25 (t, 1H), 7.00 (m, 3H), 6.00 (s, 1H), 3.20 (m, 4H), 2.00 (m, 4H), 1.30 (s, 9H)
  • Mass Spectral Analysis m/z=417.3 (M+H)+
  • Example 18B
  • 18B was obtained according to a procedure similar to the one described for 18A, with the following exception:
  • Step 18.3: 18.3a was replaced by 18.3b.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brs, 2H), 8.24 (s, 1H), 8.10 (m, 4H), 7.52 (m, 4H), 7.40 (m, 1H), 7.29 (m, 1H), 7.06 (t, 2H), 6.97 (m, 1H), 6.00 (s, 1H), 3.22 (brm, 4H), 2.07 (brm, 4H)
  • Mass Spectral Analysis m/z=437.1 (M+H)+
  • Example 18C Preparation of 18.6
  • A mixture of 14.2 (1 g, 0.00248 mole, 1.0 eq), hydroxylamine hydrochloride (18.5) (0.260 g, 0.0037 mole, 1.5 eq.) and triethylamine (0.70 mL, 0.0049 mole, 2.0 eq) in absolute ethanol (15 mL) was refluxed for 6 h. The mixture was cooled to room temperature and poured onto water. The resulting precipitate was collected by filtration, washed with water, dried under high vacuum and used for the next step without further purification. Yield: 75%
  • Mass Spectral Analysis m/z=436.2 (M+H)+
  • Preparation of 18.7
  • Acetyl chloride (6.7) (0.07 mL, 0.00097 mol, 2.0 eq) was added drop wise to a refluxing solution of 18.6 (0.212 g, 0.00048 mole, 1.0 eq) in pyridine (2 mL). The mixture was heated to reflux for 3 h. The mixture was cooled to room temperature, poured onto a saturated aqueous solution of sodium bicarbonate and extracted with ethyl acetate. The organic phase was washed with a 1N aqueous solution of hydrochloric acid and brine, dried over sodium sulfate and filtered. The organics were concentrated under reduced pressure and the crude product was purified by flash column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 35%
  • 1H NMR (400 MHz, CDCl3) δ 8.10 (d, 2H), 7.45 (d, 2H), 7.20 (m, 1H), 7.00 (m, 1H), 6.95 (m, 1H), 6.85 (m, 1H), 5.60 (s, 1H), 3.90 (m, 2H), 3.35 (m, 2H), 2.65 (s, 3H), 2.05 (d, 2H), 1.70 (m, 2H), 1.55 (s, 4H), 1.40 (s, 5H)
  • Mass Spectral Analysis m/z=460.1 (M+H)+
  • Preparation of 18C
  • To a cold (0° C.) solution of 18.7 (0.300 g, 0.00065 mole, 1.0 eq) in anhydrous dichloromethane (20 mL) was added drop wise a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (3.2 mL, 0.0065 mole, 10.0 eq). The mixture was warmed slowly to room temperature and stirring was continued for 12 h. The mixture was concentrated under reduced pressure. Diethyl ether was then added to the mixture, which was stirred for 1 h at room temperature. The precipitate was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (m, 2H), 8.10 (m, 2H), 7.60 (m, 2H), 7.30 (m, 1H), 7.05 (m, 3H), 6.00 (s, 1H), 3.30 (m, 4H), 2.45-2.80 (m, 3H), 2.10 (m, 4H)
  • Mass Spectral Analysis m/z=360.3 (M+H)+
  • Example 19A Preparation of 19.2
  • To a solution of 19.1 (29.75 g, 127.5 mmol, 1.2 eq) in dry methanol (200 mL) was added pyrrolidine (17.6 mL, 212.6 mmol, 2.0 eq) followed by 2′-hydroxyacetophenone (1.1a) (12.8 mL, 106.3 mmol, 1.0 eq). The mixture was heated under reflux for 10 h. The volatiles were removed under reduced pressure and the residue was dissolved in ethyl acetate (500 mL), washed with a 1M aqueous solution of hydrochloric acid (3×200 mL), a 1M aqueous solution of sodium hydroxide (3×200 mL) and brine. The organics were dried over sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was used in the next step without further purification.
  • 1H NMR (400 MHz, CDCl3) δ 7.86 (dd, 1H), 7.50 (m, 1H), 7.42-7.29 (m, 5H), 7.00 (m, 2H), 5.14 (s, 2H), 3.97 (brs, 2H), 3.29 (brs, 2H), 2.71 (s, 2H), 2.04 (m, 2H), 1.61 (m, 2H)
  • Mass Spectral Analysis m/z=352.1 (M+H)+
  • Preparation of 19.3
  • Under nitrogen, to an oven-dried two-necked 1L flask charged with a solution of 19.2 (45.4 g, as of 106.3 mmol, 1.0 eq) in dry tetrahydrofuran (350 mL) at −78° C. was added a solution of 1.0M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (127.6 mL, 127.6 mmol, 1.2 eq) over a 45 min time period. The reaction mixture was kept at −78° C. for 1 h and a solution of N-phenylbis(trifluoromethanesulfonamide) (1.4) (45.57 g, 127.6 mmol, 1.2 eq) in tetrahydrofuran (150 mL) was added over a 45 min time period. The reaction mixture was kept at −78° C. for 1 h, then slowly warmed up to room temperature and stirred for an additional 10 h at room temperature. Ice water (300 mL) was added to quench the reaction and the product was extracted with diethyl ether (500 mL). The organic phase was then washed with a 1M aqueous solution of hydrochloric acid (3×150 mL), a 1M aqueous solution of sodium hydroxide (3×150 mL), and brine, dried over sodium sulfate and filtered. The organics were concentrated under reduced pressure to give the crude product, which was used for the next step without further purification.
  • Mass Spectral Analysis m/z=484.0 (M+H)+
  • Preparation of 19.4
  • To a solution of 1.14 (53.58 g, 212.6 mmol, 2.0 eq) in N,N-dimethylformamide (200 mL) at 0° C. was added potassium acetate (31.3 g, 318.9 mmol, 3.0 eq), 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (2.33 g, 3.19 mmol, 0.03 eq). The reaction mixture was heated to 80° C. at which point a solution of 19.3 (60 g, crude, as of 106.3 mmol, 1.0 eq) in N,N-dimethylformamide (100 mL) was added to the reaction mixture over a 30 min time period. The reaction mixture was then stirred at 80° C. for 10 h. Diethyl ether (500 mL) and water (300 mL) were added and the two phases were separated. The organic phase was washed with a 1M aqueous solution of hydrochloric acid (2×150 mL) and brine, dried over sodium sulfate and filtered. The organics were concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 75% over three steps
  • 1H NMR (400 MHz, CDCl3) δ 7.71 (dd, 1H), 7.43-7.28 (m, 5H), 7.11 (m, 1H), 6.90 (m, 1H), 6.82 (dd, 1H), 6.27 (s, 1H), 5.14 (s, 2H), 3.94 (brs, 2H), 3.34 (brs, 2H), 1.96 (m, 2H), 1.61 (m, 2H), 1.33 (s, 12H)
  • Mass Spectral Analysis m/z=462.2 (M+H)+
  • Preparation of 19.6
  • To a solution of tert-butyl 4-bromophenylcarbamate (19.5) (20.7 g, 76 mmol, 1.04 eq) in dimethoxyethane (200 mL) was added sequentially a 2M aqueous solution of sodium carbonate (109.5 mL, 210 mmol, 3.0 eq), lithium chloride (9.28 g, 210 mmol, 3.0 eq), tetrakis(triphenylphosphine)palladium(0) (1.69 g, 1.46 mmol, 0.02 eq), and 19.4 (33.7 g, 73 mmol, 1.0 eq) under nitrogen. The reaction mixture was heated under reflux for 10 h. Water (500 mL) and diethyl ether (300 mL) were added and the two phases were separated. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting foamy solids were soaked with hexane and the fine powders were collected by filtration. Yield: 91%
  • 1H NMR (400 MHz, CDCl3) δ 7.43-7.30 (m, 7H), 7.28-7.23 (m, 2H), 7.17 (m, 1H), 7.02 (m, 1H), 6.92 (m, 1H), 6.85 (m, 1H), 6.53 (s, 1H), 5.50 (s, 1H), 5.15 (s, 2H), 3.96 (brs, 2H), 3.40 (brs, 2H), 2.06 (m, 2H), 1.67 (m, 2H), 1.53 (s, 9H)
  • Mass Spectral Analysis m/z=527.4 (M+H)+
  • Preparation of 19.7
  • To a cold (0° C.) solution of 19.6 (35.5 g, 67 mmol, 1.0 eq) in anhydrous dichloromethane (150 mL) was slowly added a 2.0M solution of hydrogen chloride in diethyl ether (167.5 mL, 335 mmol, 5.0 eq). The reaction mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure. The resulting foamy solids were soaked in diethyl ether and the fine powders were collected by filtration. This crude product was used for the next steps without further purification.
  • Mass Spectral Analysis m/z=427.3 (M+H)+
  • Preparation of 19.9a
  • To a suspension of 19.7 (1.28 g, crude, as of 3 mmol, 1.0 eq) in dry dichloromethane (80 mL) at 0° C. was slowly added triethylamine (2.1 mL, 15 mmol, 5.0 eq) followed by drop wise addition of isobutyryl chloride (19.8a) (0.48 mL, 4.5 mmol, 1.5 eq). The mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. Dichloromethane (100 mL) was added and the mixture was washed with a 1N aqueous solution of hydrochloric acid (3×50 mL), a saturated aqueous solution of sodium bicarbonate (2×50 mL) and brine, dried over sodium sulfate and filtered. The crude product was concentrated under reduced pressure and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 81% over two steps
  • 1H NMR (400 MHz, CDCl3) δ 7.57 (d, 2H), 7.40-7.27 (m, 8H), 7.17 (m, 1H), 7.01 (d, 1H), 6.93 (d, 1H), 6.85 (m, 1H), 5.50 (s, 1H), 5.15 (s, 2H), 3.96 (brs, 2H), 3.41 (brs, 2H), 2.53 (m, 1H), 2.06 (m, 2H), 1.67 (m, 2H), 1.28 (d, 6H)
  • Mass Spectral Analysis m/z=467.3 (M+H)+
  • Preparation of 19A
  • To a stirred solution of 19.9a (1.2 g, 2.44 mmol, 1.0 eq) in dry dichloromethane (20 mL) was added iodotrimethylsilane (0.66 mL, 4.89 mmol, 2.0 eq) drop wise. After stirring at room temperature for 1 h, the mixture was concentrated to dryness under reduced pressure. A 1N aqueous solution of hydrochloric acid (300 mL) and diethyl ether (200 mL) were added to the residue. The resulting solid was collected by filtration, washed with diethyl ether, and dried under vacuum.
  • Yield: 92%
  • 1H NMR (400 MHz, DMSO d6) δ 10.02 (s, 1H), 8.98 (brs, 2H), 7.70 (d, 2H), 7.36-7.22 (m, 3H), 7.02 (m, 2H), 6.94 (m, 1H), 5.82 (s, 1H), 3.21 (m, 4H), 2.63 (m, 1H), 2.03 (m, 4H), 1.11 (d, 6H)
  • Mass Spectral Analysis m/z=363.4 (M+H)+
  • Example 19B
  • 19B was obtained according to a procedure similar to the one described for 19A, with the following exception:
  • Step 19.6: 19.8a was replaced by 19.8b.
  • 1H NMR (400 MHz, DMSO d6) δ 10.04 (s, 1H), 8.90 (m, 2H), 7.71 (m, 2H), 7.29 (m, 2H), 7.25 (m, 1H), 7.03 (m, 2H), 6.94 (m, 1H), 5.82 (s, 1H), 3.44-3.11 (m, 4H), 2.25 (m, 1H), 2.02 (m, 4H), 1.51 (m, 4H), 0.86 (t, 6H)
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Example 19C Preparation of 19.10
  • To a solution of 19.7 (4.63 g, crude, as of 10 mmol, 1.0 eq) in dry pyridine (10 mL) at 0° C. was slowly added isopropylsulfonyl chloride (6.5b) (1.68 mL, 15 mmol, 1.5 eq). The reaction mixture was stirred at room temperature for 10 h. Pyridine was removed under reduced pressure and the residue was dissolved in ethyl acetate (200 mL). The solution was washed with a 1M aqueous solution of hydrochloric acid (5×50 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 55% over two steps
  • 1H NMR (400 MHz, CDCl3) δ 7.43-7.16 (m, 10H), 6.99 (dd, 1H), 6.94 (dd, 1H), 6.86 (m, 1H), 6.60 (s, 1H), 5.51 (s, 1H), 5.15 (s, 2H), 3.96 (brs, 2H), 3.49-3.30 (m, 3H), 2.06 (m, 2H), 1.67 (m, 2H), 1.43 (d, 6H)
  • Mass Spectral Analysis m/z=533.3 (M+H)+
  • Preparation of 19C
  • To a stirred solution of 19.9a (1.37 g, 2.57 mmol, 1.0 eq) in dry dichloromethane (20 mL) was added iodotrimethylsilane (0.70 mL, 5.14 mmol, 2.0 eq) dropwise. The mixture was stirred at room temperature for 1 h and then concentrated under reduced pressure. To the residue was added a 1M aqueous solution of hydrochloric acid (300 mL) and diethyl ether (200 mL). The resulting solid was collected by filtration and washed with diethyl ether. The crude compound was further purified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined, concentrated under reduced pressure, and dried under vacuum. Yield: 66%
  • 1H NMR (400 MHz, DMSO d6) δ 9.93 (brs, 1H), 8.67 (brs, 2H), 7.36-7.22 (m, 5H), 7.05-6.91 (m, 3H), 5.83 (s, 1H), 3.32-3.14 (m, 5H), 2.06 (m, 2H), 1.93 (m, 2H), 1.26 (d, 6H)
  • Mass Spectral Analysis m/z=399.3 (M+H)+
  • Example 19D Preparation of 19.12
  • To a solution of 19.7 (1.28 g, crude, as of 2.67 mmol, 1.0 eq) in dry pyridine (15 mL) at 0° C. was slowly added ethyl isocyanate (19.11) (0.33 mL, 4.15 mmol, 1.5 eq). The reaction mixture was stirred at room temperature for 10 h. Pyridine was removed under reduced pressure and the residue was partitioned between water (100 mL) and dichlorometnane (200 mL). The organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 78% over two steps
  • 1H NMR (400 MHz, CDCl3) δ 7.44-7.12 (m, 10H), 7.05-6.79 (m, 4H), 5.45 (s, 1H), 5.16 (m, 3H), 3.95 (brs, 2H), 3.50-3.26 (m, 4H), 2.04 (m, 2H), 1.65 (m, 2H), 1.16 (t, 3H)
  • Mass Spectral Analysis m/z=498.4 (M+H)+
  • Preparation of 19D
  • To a stirred solution of 19.12 (1.03 g, 2.09 mmol, 1.0 eq) in dry dichloromethane (20 mL) was added iodotrimethylsilane (0.57 mL, 4.18 mmol, 2.0 eq) drop wise. The reaction mixture was stirred at room temperature for 1 h and then concentrated under reduced pressure. The residue was suspended in methanol (50 mL) and stirred for another 1 h at room temperature. The resulting solid was collected by filtration and washed with methanol. The solid was further washed with a 1M aqueous solution of sodium hydroxide (3×10 mL) and water (2×10 mL), and then dried under vacuum. Yield: 60%
  • 1H NMR (400 MHz, DMSO d6) δ 8.54 (s, 1H), 7.44 (d, 2H), 7.18 (m, 3H), 6.98 (m, 1H), 6.91 (m, 1H), 6.86 (m, 1H), 6.13 (t, 1H), 5.72 (s, 1H), 3.11 (m, 2H), 2.89 (m, 2H), 2.74 (m, 2H), 1.77 (m, 2H), 1.67 (m, 2H), 1.06 (t, 3H)
  • Mass Spectral Analysis m/z=364.4 (M+H)+
  • Example 20A Preparation of 20A
  • Triethylamine (0.37 mL, 2.66 mmol, 2.2 eq) was added to a solution of 1A (0.50 g, 1.21 mmol, 1.0 eq) in anhydrous tetrahydrofuran (4 mL). Anhydrous methanol (4 mL) was then added followed by 20.1a (0.20 mL, 2.42 mmol, 2.0 eq). Sodium cyanoborohydride (0.09 g, 1.45 mmol, 1.2 eq) was added to the reaction mixture which was stirred for 30 min at room temperature under nitrogen. The mixture was concentrated under reduced pressure. Dichloromethane (30 mL) and water (10 mL) were added and the suspension was stirred at room temperature for 10 min. The phases were separated. The organic phase was further washed with water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added drop wise a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (5 mL). The mixture was then stirred for 1 h at room temperature and concentrated under reduced pressure. Diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 65%
  • 1H NMR (400 MHz, DMSO d6) δ 10.63 (brs, 0.25H), 10.50 (brs, 0.75H), 7.42 (m, 4H), 7.28 (m, 1H), 7.08 (d, 1H), 6.98 (m, 2H), 6.27 (s, 0.25H), 5.85 (s, 0.75H), 3.37 (brm, 8H), 2.82 (s, 3H), 2.11 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=391.2 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.9H2O
  • Theory: % C, 67.75; % H, 7.46; % N, 6.32.
  • Found: % C, 67.89; % H, 7.32; % N, 6.26.
  • Example 20B
  • 20B was obtained according to a procedure similar to the one described for 20A, with the following exception:
  • Step 20.1: 1A was replaced by 11A.
  • 1H NMR (400 MHz, DMSO d6) δ 10.42 (brs, 1H), 9.47 (s, 1H), 7.30 (m, 4H), 7.08 (t, 1H), 6.60 (d, 1H), 6.46 (d, 1H), 5.68 (s, 1H), 3.40 (m, 4H), 3.30 (s, 3H), 3.20 (m, 2H), 2.81 (s, 2H), 2.15 (m, 2H), 2.05 (m, 2H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=407.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 0.5H2O
  • Theory: % C, 66.43; % H, 7.14; % N, 6.20.
  • Found: % C, 66.53; % H, 7.06; % N, 6.24.
  • Example 20C
  • 20C was obtained according to a procedure similar to the one described for 20A, with the following exception:
  • Step 20.1: 1A was replaced by 11B.
  • 1H NMR (400 MHz, DMSO d6) δ 10.79 (brs, 1H), 9.74 (d, 1H), 8.41 (s, 1H), 7.69 (dd, 1H), 7.45 (d, 1H), 7.09 (t, 1H), 6.62 (d, 1H), 6.49 (d, 2H), 5.81 (s, 1H), 3.42 (m, 4H), 3.30 (m, 4H), 2.79 (d, 3H), 2.12 (m, 4H), 1.16 (m, 3H), 1.08 (m, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Example 20D
  • 20D was obtained according to a procedure similar to the one described for 20A, with the following exception:
  • Step 20.1: 1A was replaced by 3D.
  • 1H NMR (400 MHz, DMSO d6) δ 11.00 (m, 0.25H), 10.85 (m, 0.75H), 7.80 (m, 2H), 7.54 (m, 1H), 7.40 (m, 4H), 7.22 (m, 1H), 7.10 (m, 0.75H), 7.02 (m, 0.25H), 6.32 (s, 0.25H), 5.91 (s, 0.75H), 3.33 (m, 10H), 2.80 (m, 2H), 2.20 (m, 3H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=434.4 (M+H)+
  • Elemental analysis:
  • C26H31N3O3, 1HCl, 1H2O
  • Theory: % C, 63.99; % H, 7.02; % N, 8.61.
  • Found: % C, 64.11; % H, 6.70; % N, 8.49.
  • Example 20E
  • 20E was obtained according to a procedure similar to the one described for 20A, with the following exception:
  • Step 20.1: 1A was replaced by 3E.
  • 1H NMR (400 MHz, DMSO d6) δ 10.84 (m, 1H), 8.31 (m, 1H), 7.78 (m, 1H), 7.52 (m, 1H), 7.42 (m, 3H), 7.10 (m, 1H), 5.90 (s, 1H), 3.46 (m, 2H), 3.31 (m, 10H), 2.82 (m, 2H), 2.72 (m, 2H), 2.12 (m, 3H), 1.16 (m, 6H)
  • Mass Spectral Analysis m/z=448.5 (M+H)+
  • Elemental analysis:
  • C27H33N3O3, 1HCl, 1H2O
  • Theory: % C, 64.59; % H, 7.23; % N, 8.37.
  • Found: % C, 64.77; % H, 7.27; % N, 8.40.
  • Example 20F
  • 20F was obtained according to a procedure similar to the one described for 20A, with the following exception:
  • Step 20.1: 1A was replaced by 3F.
  • 1H NMR (400 MHz, DMSO d6) δ 10.80 (brs, 1H), 8.35 (m, 1H), 7.78 (m, 1H), 7.50 (m, 1H), 7.40 (m, 3H), 7.09 (m, 1H), 5.93 (s, 1H), 3.41 (m, 2H), 3.20 (m, 10H), 2.72 (m, 2H), 2.10 (m, 3H), 1.10 (m, 9H)
  • Mass Spectral Analysis m/z=462.5 (M+H)+
  • Elemental analysis:
  • C28H35N3O3, 1HCl, 1H2O
  • Theory: % C, 65.17; % H, 7.42; % N, 8.14.
  • Found: % C, 65.28; % H, 7.37; % N, 8.21.
  • Example 20G
  • 20G was obtained according to a procedure similar to the one described for 20A, with the following exception:
  • Step 20.1: 1A was replaced by 3V.
  • 1H NMR (400 MHz, CDCl3) δ 8.57 (s, 1H), 7.70 (m, 2H), 7.66 (d, 1H), 7.38 (s, 1H), 7.02 (d, 1H), 5.70 (s, 1H), 3.61 (m, 2H), 3.46 (m, 2H), 2.62 (m, 2H), 2.52 (m, 2H), 2.12 (m, 2H), 2.78 (m, 2H), 1.30 (t, 3H), 1.23 (t, 3H)
  • Mass Spectral Analysis m/z=435.4 (M+H)+
  • Example 20H
  • 20H was obtained according to a procedure similar to the one described for 20L, with the following exception:
  • Step 20.1: 21A was replaced by 4H and 20.1d was replaced by 20.1a.
  • 1H NMR (400 MHz, DMSO d6) δ 10.44-10.12 (m, 1H), 7.74 (dd, 0.7H), 7.67 (dd, 0.7H), 7.45 (m, 5H), 7.27 (m, 3H), 6.38 (s, 0.3H), 6.00 (s, 0.7H), 3.53-3.16 (m, 8H), 2.84 (m, 3H), 2.35-2.03 (m, 4H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=470.3 (M+H)+
  • Elemental analysis:
  • C25H31N3O4S, 1HCl, 1H2O
  • Theory: % C, 57.30; % H, 6.54; % N, 8.02.
  • Found: % C, 57.46; % H, 6.44; % N, 7.96.
  • Example 20I
  • 20I was obtained according to a procedure similar to the one described for 20L, with the following exception:
  • Step 20.1: 20.1d was replaced by 20.1a.
  • 1H NMR (400 MHz, DMSO d6) δ 10.62 (brs, 1H), 7.41 (m, 4H), 7.24 (m, 1H), 6.97 (m, 2H), 6.93 (m, 1H), 5.92 & 5.86 (2s, 1H, rotamer), 3.55-2.92 (m, 8H), 2.80 & 2.77 (d, 3H), 2.56-1.76 (m, 6H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=405.4 (M+H)+
  • Example 20J
  • 20J was obtained according to a procedure similar to the one described for 20L, with the following exception:
  • Step 20.1: 20.1d was replaced by 20.1b.
  • 1H NMR (400 MHz, DMSO d6) δ 10.72 (m, 1H), 7.41 (m, 4H), 7.24 (m, 1H), 6.95 (m, 3H), 5.91 & 5.84 (2s, 1H, rotamer), 3.56-2.94 (m, 10H), 2.57-1.77 (m, 6H), 1.27 (m, 3H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=419.4 (M+H)+
  • Example 20K
  • 20K was obtained according to a procedure similar to the one described for 20L, with the following exception:
  • Step 20.1: 20.1d was replaced by 20.1c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.99 (m, 1H), 7.41 (m, 4H), 7.25 (m, 1H), 6.95 (m, 3H), 5.88 & 5.86 (2s, 1H rotamer), 3.70-2.93 (m, 10H), 2.57-1.76 (m, 7H), 1.12 (m, 6H), 0.99 (m, 6H)
  • Mass Spectral Analysis m/z=447.5 (M+H)+
  • Example 20L Preparation of 20L
  • To a stirred solution of cyclopropanecarbaldehyde (20.1d) (0.22 mL, 3.0 mmol, 2.0 eq) in dry dichloromethane (25 mL) was added sequentially 21A (0.64 g, 1.5 mmol, 1.0 eq), acetic acid (0.10 mL, 1.8 mmol, 1.2 eq), and sodium cyanoborohydride (0.14 g, 2.25 mmol, 1.5 eq). The reaction mixture was stirred at room temperature for 10 h. Water (40 mL) was added and the aqueous layer was basified to pH=10 with a 1M aqueous solution of sodium hydroxide. The two phases were separated and the aqueous phase was saturated with sodium chloride and extracted with dichloromethane (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in dichloromethane was added dropwise a 2.0M solution of hydrogen chloride in diethyl ether (1.0 mL, 2.0 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum.
  • Yield: 65%
  • 1H NMR (400 MHz, DMSO d6) δ 10.66 (brs, 1H), 7.41 (m, 4H), 7.25 (m, 1H), 7.03-6.89 (m, 3H), 5.91 & 5.86 (2s, 1H, rotomer), 3.80-2.95 (m, 10H), 2.44-1.78 (m, 6H), 1.12 (m, 7H), 0.64 (m, 2H), 0.42 (m, 2H)
  • Mass Spectral Analysis m/z=445.4 (M+H)+
  • Example 20M Preparation of 20M
  • Triethylamine (0.98 mL, 7.00 mmol, 3.3 eq) was added to a solution of 1A (0.80 g, 2.12 mmol, 1.0 eq) in anhydrous dichloromethane (5 mL). Compound 2.8a (0.68 mL, 7.00 mmol, 3.3 eq) was then added to the reaction mixture, which was stirred overnight at room temperature under nitrogen. The mixture was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). To a solution of the purified product in dichloromethane (5 mL) was added at 0° C. a 2.0 M solution of hydrochloric acid in diethyl ether (3.2 mL, 1.16 mmol, 5.5 eq). Diethyl ether was added to the mixture. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 46%
  • 1H NMR (400 MHz, DMSO d6) δ 10.83 (m, 0.25H), 10.71 (m, 0.75H), 7.45 (m, 4H), 7.28 (m, 1H), 7.08 (m, 1H), 7.00 (m, 2H), 6.24 (s, 0.25H), 5.85 (s, 0.75H), 3.47 (m, 5H), 3.25 (m, 4H), 3.06 (m, 2H), 2.18 (m, 4H), 1.12 (m, 6H), 0.65 (m, 2H), 0.43 (m, 2H)
  • Mass Spectral Analysis m/z=431.0 (M+H)+
  • Example 20N
  • 20N was obtained according to a procedure similar to the one described for 20M, with the following exception:
  • Step 20.1: 2.8a was replaced by 20.2a.
  • 1H NMR (400 MHz, DMSO d6) δ 10.10 (m, 1H), 7.43 (m, 4H), 7.28 (m, 1H), 7.09 (m, 1H), 6.98 (m, 2H), 6.28 (s, 0.25H), 5.85 (s, 0.75H), 3.35 (brm, 10H), 2.15 (m, 4H), 1.28 (m, 3H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=405.0 (M+H)+
  • Example 20O
  • 20O was obtained according to a procedure similar to the one described for 20M, with the following exception:
  • Step 20.1: 2.8a was replaced by 20.2b.
  • 1H NMR (400 MHz, DMSO d6) δ 10.18 (m, 1H), 7.45 (m, 4H), 7.29 (m, 1H), 7.09 (m, 1H), 6.98 (m, 2H), 6.25 (m, 0.25H), 5.84 (m, 0.75H), 3.41 (m, 4H), 3.21 (m, 4H), 3.09 (m, 2H), 2.16 (m, 4H), 1.75 (m, 2H), 1.11 (m, 6H), 0.91 (m, 3H)
  • Mass Spectral Analysis m/z=419.1 (M+H)+
  • Example 20P
  • 20P was obtained according to a procedure similar to the one described for 20M, with the following exception:
  • Step 20.1: 2.8a was replaced by 20.2c.
  • 1H NMR (400 MHz, CDCl3) δ 7.35 (m, 9H), 7.17 (m, 1H), 6.98 (dd, 1H), 6.94 (dd, 1H), 6.84 (m, 1H), 5.61 (s, 1H), 3.58 (brs, 4H), 3.32 (brs, 2H), 2.60 (brm, 4H), 2.08 (brm, 2H), 1.81 (brm, 2H), 1.20 (brd, 6H)
  • Mass Spectral Analysis m/z=467.3 (M+H)+
  • Example 20Q
  • 20Q was obtained according to a procedure similar to the one described for 20M, with the following exception:
  • Step 20.1: 2.8a was replaced by 20.2d.
  • 1H NMR (400 MHz, DMSO d6) δ 10.95 (brs, 0.5H) 7.44 (m, 4H), 7.33 (m, 6H), 7.04 (d, 1H), 6.99 (m, 2H), 6.24 (s, 0.3H), 5.87 (s, 0.7H), 3.40 (brm, 10H), 3.12 (m, 2H), 2.18 (brm, 4H), 1.13 (brd, 6H)
  • Mass Spectral Analysis m/z=481.3 (M+H)+
  • Example 20R
  • 20R was obtained according to a procedure similar to the one described for 20M, with the following exception:
  • Step 20.1: 2.8a was replaced by 20.2e.
  • 1H NMR (400 MHz, DMSO d6) δ 10.70 (brm, 0.50H), 7.43 (m, 4H), 7.28 (m, 6H), 7.08 (d, 1H), 6.97 (m, 2H), 6.36 (s, 0.3H), 5.83 (s, 0.7H), 3.44 (m, 4H), 3.18 (brm, 6H), 2.67 (t, 2H), 2.12 (brm, 6H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=495.3 (M+H)+
  • Example 21A Preparation of 21.2
  • To a stirred solution of N-boc 4-piperidone (1.2) (2.0 g, 10 mmol, 1.0 eq) in dry diethyl ether (15 mL) at −25° C. was simultaneously but independently added ethyl diazoacetate (21.1) (1.35 mL, 13 mmol, 1.3 eq) and boron trifluoride diethyl ether complex (1.33 mL, 10.5 mmol, 1.05 eq) under nitrogen atmosphere over a 20 min time period. The reaction mixture was stirred for another 1 h at −25° C. A 1M aqueous solution of potassium carbonate was added drop wise to the stirred reaction mixture until gaseous evolution ceased. The two phases were separated and the organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • Preparation of 21.3
  • A mixture of the crude 21.2 (3 g, as of 10 mmol) in a 4M aqueous hydrochloric acid solution (50 mL) was heated under reflux for 6 h. Water was removed under reduced pressure and the resulting solid was washed with diethyl ether and dried under vacuum. Yield: 90% over two steps
  • 1H NMR (400 MHz, DMSO d6) δ 9.41 (brs, 2H), 3.30 (m, 2H), 3.21 (m, 2H), 2.77 (m, 2H), 2.62 (m, 2H), 1.94 (m, 2H)
  • Preparation of 21.4
  • To a suspension of 21.3 (4.98 g, 33.3 mmol, 1.0 eq) in dry dichloromethane (100 mL) at 0° C. was slowly added triethylamine (11 mL, 79.92 mmol, 2.4 eq) followed by a solution of di-tert-butyl-dicarbonate (4.7) (8.72 g, 39.96 mmol, 1.2 eq) in dichloromethane (30 mL) over a 20 min time period. The reaction mixture was stirred at room temperature for 10 h and washed with a 1M aqueous solution of hydrochloric acid (3×100 mL), brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was used for next step without further purification.
  • 1H NMR (400 MHz, CDCl3) δ 3.58 (m, 4H), 2.65 (m, 4H), 1.78 (m, 2H), 1.45 (s, 9H)
  • Preparation of 21.5
  • To a solution of 21.4 (2.56 g, 12 mmol, 1.0 eq) in dry methanol (30 mL) was added pyrrolidine (2 mL, 24 mmol, 2.0 eq) followed by 2′-hydroxyacetophenone (1.1a) (1.44 mL, 12 mmol, 1.0 eq). The mixture was heated under reflux for 10 h. The volatiles were removed under reduced pressure and the residue was dissolved in ethyl acetate (200 mL) and washed with a 1M aqueous solution of hydrochloric acid (3×50 mL), a 1M aqueous solution of sodium hydroxide (3×50 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 72% over two steps
  • 1H NMR (400 MHz, CDCl3) δ 7.85 (dd, 1H), 7.49 (m, 1H), 6.99 (m, 2H), 3.78-3.49 (m, 2H), 3.32 (m, 2H), 2.83-2.63 (m, 2H), 2.19 (m, 2H), 2.00-1.55 (m, 4H), 1.47 (s, 9H)
  • Mass Spectral Analysis m/z=331.9 (M+H)+
  • Preparation of 21.6
  • To an oven-dried two-neck 250 mL flask charged with a solution of 21.5 (2.86 g, 8.6 mmol, 1.0 eq) in dry tetrahydrofuran (40 mL) at −78° C. under nitrogen was added a 1.0M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (10.3 mL, 10.3 mmol, 1.2 eq) over a 10 min time period. The mixture was kept at −78° C. for 1 h and a solution of N-phenylbis(trifluoromethanesulfonamide) (1.4) (3.68 g, 10.3 mmol, 1.2 eq) in tetrahydrofuran (20 mL) was added to the mixture over a 10 min time period. The mixture was kept at −78° C. for another 1 h, then slowly warmed to room temperature and stirred for an additional 10 h at room temperature. Ice water (50 mL) was added to quench the reaction and the product was extracted with diethyl ether (200 mL). The organic phase was then washed with a 1N aqueous solution of hydrochloric acid (3×50 mL), a 1N aqueous solution of sodium hydroxide (3×50 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 85%
  • 1H NMR (400 MHz, CDCl3) δ 7.30-7.23 (m, 2H), 6.97 (m, 1H), 6.89 (m, 1H), 5.60 (s, 1H), 3.80-3.53 (m, 2H), 3.36-3.24 (m, 2H), 2.30-2.06 (m, 3H), 1.90-1.64 (m, 3H), 1.47 (s, 9H)
  • Preparation of 21.7
  • To a solution of 21.6 (3.38 g, 7.3 mmol, 1.0 eq) in dimethoxyethane (50 mL) was added sequentially a 2M aqueous solution of sodium carbonate (11 mL, 22 mmol, 3.0 eq), lithium chloride (0.93 g, 22 mmol, 3.0 eq), tetrakis(triphenylphosphine)palladium(0) (0.17 g, 0.15 mmol, 0.02 eq), and 4-N,N-diethylphenylboronic acid (1.6) (1.77 g, 8.02 mmol, 1.1 eq) under a nitrogen atmosphere. The reaction mixture was heated under reflux for 10 h and then cooled to room temperature. Water (200 mL) and diethyl ether (300 mL) were added and the two phases were separated. The organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 81%
  • 1H NMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 7.18 (m, 1H), 6.99 (d, 1H), 6.92 (d, 1H), 6.85 (m, 1H), 5.60 (s, 1H), 3.86-3.50 (m, 4H), 3.42-3.24 (m, 4H), 2.27-1.68 (m, 6H), 1.48 (s, 9H), 1.21 (m, 6H)
  • Mass Spectral Analysis m/z=491.0 (M+H)+
  • Preparation of 21A
  • To a cold (0° C.) solution of 21.7 (1.15 g, 2.34 mmol, 1.0 eq) in anhydrous dichloromethane (20 mL) was added dropwise a 4.0M solution of hydrogen chloride in dioxane (3.51 mL, 14.04 mmol, 6.0 eq). The mixture was stirred at room temperature for 10 h and concentrated under reduced pressure. The resulting foamy solids were soaked in diethyl ether. The resulting fine powder was collected by filtration and washed with diethyl ether. Yield: 98%
  • 1H NMR (400 MHz, CDCl3) δ 9.76 (m, 2H), 7.41 (m, 2H), 7.36 (m, 2H), 7.20 (m, 1H), 7.00 (dd, 1H), 6.97 (dd, 1H), 6.88 (m, 1H), 5.63 (s, 1H), 3.68-3.23 (m, 8H), 2.50-2.23 (m, 4H), 2.02-1.82 (m, 2H), 1.35-1.07 (m, 6H)
  • Mass Spectral Analysis m/z=391.2 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl
  • Theory: % C, 70.32; % H, 7.32; % N, 6.56.
  • Found: % C, 70.14; % H, 7.23; % N, 6.55.
  • Example 21B Preparation of 21.7a & 21.7b
  • The racemic compound 21.7 (15 g) was resolved by chiral HPLC to provide
  • 21.7a (6.7 g) and 21.7b (6.0 g) as pure enantiomers.
  • Chiral separation conditions:
      • Column: Chiralcel OJ, 4.6×250 mm
      • Flow: 1.0 mL/min
      • Temperature: room temperature
      • Detection: 335 nm
      • Mobile Phase Methanol
  • 21.7a: 1H NMR (400 MHz, CDCl3) δ 7.38 (m, 4H), 7.18 (m, 1H), 6.99 (dd, 1H), 6.92 (dd, 1H), 6.85 (m, 1H), 5.60 (s, 1H), 3.84-3.49 (m, 4H), 3.31 (m, 4H), 2.25-1.65 (m, 6H), 1.48 (s, 9H), 1.21 (m, 6H)
  • Mass Spectral Analysis m/z=491.3 (M+H)+
  • [α]D 25=−1.04 (c. 1.14 mg/mL, MeOH)
  • Chiral purity: ee=99%; tR=4.6 min
  • 21.7b: 1H NMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 7.18 (m, 1H), 6.99 (dd, 1H), 6.92 (dd, 1H), 6.85 (m, 1H), 5.60 (s, 1H), 3.85-3.48 (m, 4H), 3.31 (m, 4H), 2.25-1.66 (m, 6H), 1.48 (s, 9H), 1.21 (m, 6H)
  • Mass Spectral Analysis m/z=491.3 (M+H)+
  • [α]D 25=+1.07 (c. 1.16 mg/mL, MeOH)
  • Chiral purity: ee=99%; tR=5.2 min
  • Preparation of 21B
  • To a cold (0° C.) solution of 21.7a (1.3 g, 2.65 mmol, 1.0 eq) in anhydrous dichloromethane (20 mL) was added drop wise a 4.0M solution of hydrogen chloride in dioxane (3.31 mL, 13.25 mmol, 5.0 eq). The reaction mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure. The foamy solids were soaked in diethyl ether and the resulting fine powder was collected by filtration and washed with diethyl ether. Yield: 87%
  • 1H NMR (400 MHz, DMSO d6) δ 8.97 (brs, 2H), 7.41 (m, 4H), 7.24 (m, 1H), 7.00-6.89 (m, 3H), 5.89 (s, 1H), 3.54-3.01 (m, 8H), 2.30-2.10 (m, 3H), 2.03-1.88 (m, 2H), 1.78 (m, 1H), 1.23-0.99 (m, 6H)
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 1/6H2O
  • Theory: % C, 69.83; % H, 7.35; % N, 6.51.
  • Found: % C, 69.84; % H, 7.27; % N, 6.46.
  • [α]D 25=+1.80 (c. 10.0 mg/mL, MeOH)
  • Example 21C Preparation of 21C
  • To a cold (0° C.) solution of 21.7b (1.3 g, 2.65 mmol, 1.0 eq) in anhydrous dichloromethane (20 mL) was added drop wise a 4.0 M solution of hydrogen chloride in dioxane (3.31 mL, 13.25 mmol, 5.0 eq). The reaction mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure. The foamy solids were soaked in diethyl ether and the resulting fine powder was collected by filtration and washed with diethyl ether. Yield: 89%
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (brs, 2H), 7.41 (m, 4H), 7.24 (m, 1H), 7.02-6.89 (m, 3H), 5.89 (s, 1H), 3.52-3.02 (m, 8H), 2.35-2.10 (m, 3H), 2.04-1.88 (m, 2H), 1.78 (m, 1H), 1.23-0.99 (m, 6H)
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 1/6H2O
  • Theory: % C, 69.83; % H, 7.35; % N, 6.51.
  • Found: % C, 69.84; % H, 7.32; % N, 6.47.
  • [α]D 25=−1.81 (c. 10.25 mg/mL, MeOH)
  • Example 21D Preparation of 21D
  • To a stirred solution of 21B (0.47 g, 1.1 mmol, 1.0 eq) in methanol (20 mL) was added palladium [90 mg, 10 wt. % (dry basis) on activated carbon, 20% wt. eq]. The reaction mixture was stirred under hydrogen atmosphere using a hydrogen balloon at room temperature for 10 h. The palladium on activated carbon was filtered off on a celite pad and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol/ammonium hydroxide mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in dichloromethane was added dropwise a 2.0M solution of hydrogen chloride in diethyl ether (1.1 mL, 2.2 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 89%
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (brs, 2H), 7.30 (m, 4H), 7.12 (m, 1H), 6.86 (m, 1H), 6.78 (m, 1H), 6.62 (m, 1H), 4.20 (m, 1H), 3.50-2.96 (m, 8H), 2.29-1.66 (m, 8H), 1.10 (brm, 6H)
  • Mass Spectral Analysis m/z=393.3 (M+H)+
  • Example 21E Preparation of 21E
  • To a stirred solution of 21C (0.49 g, 1.14 mmol, 1.0 eq) in methanol (20 mL) was added palladium [98 mg, 10 wt. % (dry basis) on activated carbon, 20% wt. eq]. The reaction mixture was stirred under hydrogen using a hydrogen balloon at room temperature for 10 h. The palladium on activated carbon was filtered off on a celite pad and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol/ammonium hydroxide mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in dichloromethane was added dropwise a 2.0M solution of hydrogen chloride in diethyl ether (1.14 mL, 2.28 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum.
  • Yield: 93%
  • 1H NMR (400 MHz, DMSO d6) δ 8.80 (brs, 2H), 7.29 (m, 4H), 7.12 (m, 1H), 6.85 (m, 1H), 6.77 (m, 1H), 6.62 (m, 1H), 4.20 (m, 1H), 3.52-2.96 (m, 8H), 2.22-1.66 (m, 8H), 1.10 (brm, 6H)
  • Mass Spectral Analysis m/z=393.3 (M+H)+
  • Example 21F Preparation of 21.9
  • To a stirred solution of 21A (1.93 g, 4.52 mmol, 1.0 eq) in dry dichloromethane (30 mL) at 0° C. was added triethylamine (1.51 mL, 10.85 mmol, 2.4 eq) followed by drop wise addition of benzyl chloroformate (21.8) (0.76 mL, 5.42 mmol, 1.2 eq). The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between diethyl ether (200 mL) and water (100 mL). The organic layer was washed with a 1N aqueous solution of hydrochloric acid (3×50 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give the crude product, which was used for the next step without further purification.
  • Mass Spectral Analysis m/z=525.0 (M+H)+
  • Preparation of 21.10
  • To a solution of 21.9 (0.9 g, crude, as of 1.71 mmol, 1.0 eq) in dry dichloroethane (10 mL) was added sulfur trioxide N,N-dimethylformamide complex (4.3) (315 mg, 2.06 mmol, 1.2 eq) portion wise. The reaction mixture was heated at 75° C. for 10 h and then cooled down to 0-10° C. at which point oxalyl chloride (0.2 mL, 2.22 mmol, 1.3 eq) was added drop wise. The mixture was then stirred at 65° C. for another 3 h and then quenched with ice water (50 mL) at room temperature. Dichloromethane (100 mL) was added and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product, which was used for next step without further purification.
  • Mass Spectral Analysis m/z=622.9 (M+H)+
  • Preparation of 21.11
  • To a solution of 21.10 (0.9 g, crude, as of 1.4 mmol, 1.0 eq) in dry dichloromethane (50 mL) at 0° C. was slowly added triethylamine (0.4 mL, 2.8 mmol, 2.0 eq) and a 2.0M solution of ethylamine (3.4c) in tetrahydrofuran (7 mL, 14 mmol, 10.0 eq) drop wise. The mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. Water (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 34% over three steps
  • 1H NMR (400 MHz, CDCl3) δ 7.68 (m, 1H), 7.50 (m, 1H), 7.43 (m, 2H), 7.40-7.30 (m, 7H), 6.98 (d, 1H), 5.66 & 5.44 (2s, 1H), 5.18 & 5.16 (2s, 2H), 4.21 (t, 1H), 3.89-3.23 (m, 8H), 2.97 (m, 2H), 2.32-1.66 (m, 6H), 1.35-1.05 (m, 9H)
  • Mass Spectral Analysis m/z=631.95 (M+H)+
  • Preparation of 21F
  • To a solution of 21.11 (0.35 g, 0.55 mmol, 1.0 eq) in dichloromethane (10 mL) was added iodotrimethylsilane (0.15 mL, 1.1 mmol, 2.0 eq) drop wise. The mixture was stirred at room temperature for 2 h. The mixture was diluted with chloroform 9100 mL) and methanol (5 mL). The solution was washed with a 20% aqueous solution of sodium thiosulfate (2×30 mL), with a 1M aqueous solution of sodium carbonate (2×30 mL), dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by preparative liquid chromatography (mobile phase: acetonitrile/water/trifluoroacetic acid). The desired fractions were combined and concentrated under reduced pressure. The product was dissolved in dichloromethane (50 mL); the organic phase was washed with a 1N aqueous solution of sodium hydroxide (2×20 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added dropwise a 1.0M solution of hydrogen chloride in diethyl ether (1.1 mL, 1.1 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 56%
  • 1H NMR (400 MHz, DMSO d6) δ 9.03 (brs, 2H), 7.65 (dd, 1H), 7.54-7.36 (m, 6H), 7.16 (d, 1H), 6.04 (s, 1H), 3.54-3.02 (m, 8H), 2.71 (m, 2H), 2.37-2.13 (m, 3H), 2.06-1.72 (m, 3H), 1.22-1.03 (m, 6H), 0.94 (t, 3H)
  • Mass Spectral Analysis m/z=498.5 (M+H)+
  • Elemental analysis:
  • C27H35N3O4S, 1HCl, 0.33H2O
  • Theory: % C, 60.04; % H, 6.84; % N, 7.78.
  • Found: % C, 59.93; % H, 6.81; % N, 7.80.
  • Example 22A Preparation of 22.1
  • To a suspension of 21B (4.06 g, 9.5 mmol, 1.0 eq) in tetrahydrofuran (50 mL) at 0° C. was added triethylamine (3.3 mL, 23.75 mmol, 2.5 eq) followed by drop wise addition of trifluoroacetic anhydride (4.1) (1.6 ml, 11.4 mmol, 1.2 eq). The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. Ethyl acetate (200 mL) was added to the reaction mixture and the organic layer was washed with a 1M aqueous solution of hydrochloric acid (3×50 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give the crude product, which was used for the next step without further purification.
  • Mass Spectral Analysis m/z=487.2 (M+H)+
  • Preparation of 22.2
  • To a solution of 22.1 (5.0 g, as of 9.5 mmol, 1.0 eq) in dry dichloroethane (100 mL) was added sulfur trioxide N,N-dimethylformamide complex (4.3) (2.18 g, 14.25 mmol, 1.5 eq) portion wise. The mixture was heated under reflux for 10 h and then cooled to 0-10° C. at which point oxalyl chloride (1.33 mL, 15.2 mmol, 1.6 eq) was added drop wise. The mixture was then stirred at 70° C. for another 3 h and quenched with ice water (1:1) (150 mL) at room temperature. Dichloromethane (100 mL) was added to the reaction mixture and the two phases were separated. The aqueous phase was further extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 84% over two steps
  • 1H NMR (400 MHz, CDCl3) δ 7.88 (m, 1H), 7.70 (m, 1H), 7.48 (m, 2H), 7.35 (m, 2H), 7.08 (d, 1H), 5.716 & 5.706 (2s, 1H), 4.03-3.26 (m, 8H), 2.49-2.21 (m, 3H), 2.03-1.72 (m, 3H), 1.33-1.11 (m, 6H)
  • Mass Spectral Analysis m/z=585.2 (M+H)+
  • Preparation of 22.3a
  • To a solution of 22.2 (0.6 g, 1.02 mmol, 1.0 eq) in dry dichloromethane (30 mL) at 0° C. was added triethylamine (0.71 mL, 5.10 mmol, 5.0 eq) and methylamine (3.4b) hydrochloride salt (0.21 g, 3.06 mmol, 3.0 eq) in one portion. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. Water (50 mL) and dichloromethane (50 mL) were added to the mixture and the two phases were separated. The aqueous phase was further extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 89%
  • 1H NMR (400 MHz, CDCl3) δ 7.71 (dd, 1H), 7.51 (t, 1H), 7.45 (m, 2H), 7.34 (m, 2H), 7.02 (d, 1H), 5.665 & 5.657 (2s, 1H), 4.29 (m, 1H), 4.02-3.25 (m, 8H), 2.63 (d, 3H), 2.47-2.19 (m, 3H), 1.99-1.68 (m, 3H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=580.3 (M+H)+
  • Preparation of 22A
  • To a solution of 22.3a (0.53 g, 0.91 mmol, 1.0 eq) in a mixture of methanol (20 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.75 g, 5.46 mmol, 6.0 eq) portion wise. The reaction mixture was warmed to room temperature and stirred for 10 h at room temperature. Brine (50 mL) and chloroform (50 mL) were added to the reaction mixture and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added drop wise a 2.0M solution of hydrogen chloride in diethyl ether (0.91 mL, 1.82 mmol, 2.0 eq). The mixture was stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 82%
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (brs, 2H), 7.64 (dd, 1H), 7.49-7.34 (m, 6H), 7.17 (d, 1H), 6.04 (s, 1H), 3.45 (m, 2H), 3.31-3.15 (m, 5H), 3.09 (m, 1H), 2.35 (d, 3H), 2.28 (m, 2H), 2.18 (m, 1H), 1.99 (m, 2H), 1.80 (m, 1H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=484.2 (M+H)+
  • Elemental analysis:
  • C26H33N3O4S, 1HCl, 1.2H2O
  • Theory: % C, 57.65; % H, 6.77; % N, 7.76.
  • Found: % C, 57.69; % H, 6.62; % N, 7.71.
  • [α]D 25=−4.18 (c. 9.4 mg/mL, MeOH)
  • Example 22B
  • 22B was obtained according to a procedure similar to the one described for 22A, with the following exception:
  • Step 22.3: 3.4b was replaced by 3.4c.
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (brs, 1H), 7.65 (dd, 1H), 7.44 (m, 5H), 7.37 (d, 1H), 7.16 (d, 1H), 6.04 (s, 1H), 3.45 (m, 2H), 3.32-3.05 (m, 6H), 2.71 (m, 2H), 2.35-1.75 (m, 6H), 1.12 (m, 6H), 0.94 (t, 3H)
  • Mass Spectral Analysis m/z=498.3 (M+H)+
  • Elemental analysis:
  • C27H35N3O4S, 1HCl, 1.1H2O
  • Theory: % C, 58.54; % H, 6.95; % N, 7.59.
  • Found: % C, 58.55; % H, 6.82; % N, 7.55.
  • [α]D 25=−5.10 (c=9.25 mg/ml, MeOH)
  • Example 22C
  • 22C was obtained according to a procedure similar to the one described for 22A, with the following exception:
  • Step 22.3: 3.4b was replaced by 3.4d.
  • 1H NMR (400 MHz, DMSO d6) δ 9.05 (brs, 2H), 7.65 (dd, 1H), 7.56 (t, 1H), 7.43 (m, 4H), 7.37 (d, 1H), 7.16 (d, 1H), 6.04 (s, 1H), 3.53-3.04 (m, 8H), 2.63 (m, 2H), 2.35-1.75 (m, 6H), 1.33 (m, 2H), 1.12 (m, 6H), 0.77 (t, 3H)
  • Mass Spectral Analysis m/z=512.4 (M+H)+
  • Elemental analysis:
  • C28H37N3O4S, 1HCl, 0.5H2O
  • Theory: % C, 60.36; % H, 7.06; % N, 7.54.
  • Found: % C, 60.28; % H, 7.10; % N, 7.53.
  • [α]D 25=−5.95 (c=9.55 mg/ml, MeOH)
  • Example 22D
  • 22D was obtained according to a procedure similar to the one described for 22A, with the following exception:
  • Step 22.3: 3.4b was replaced by 3.4 g.
  • 1H NMR (400 MHz, DMSO d6) δ 9.0 (brs, 2H), 7.66 (m, 2H), 7.42 (m, 5H), 7.16 (d, 1H), 6.04 (s, 1H), 3.45 (m, 2H), 3.22 (m, 6H), 2.59 (m, 2H), 2.35-1.75 (m, 6H), 1.12 (m, 6H), 0.75 (m, 1H), 0.32 (m, 2H), 0.03 (m, 2H)
  • Mass Spectral Analysis m/z=524.3 (M+H)+
  • Elemental analysis:
  • C29H37N3O4S, 1HCl, 0.66H2O
  • Theory: % C, 60.88; % H, 6.93; % N, 7.34.
  • Found: % C, 60.92; % H, 6.96; % N, 7.37.
  • [α]D 25=−5.89 (c=9.35 mg/ml, MeOH)
  • Example 22E Preparation of 22.4
  • To a solution of 22.2 (0.86 g, 1.47 mmol, 1.0 eq) in tetrahydrofuran (5 mL) at 0° C. was added a 1.0M solution of hydrazine in tetrahydrofuran (5.1) (15 mL, 15 mmol, 15.0 eq) in one portion. The reaction mixture was stirred at 0° C. for 30 min Water (50 mL) and dichloromethane (100 mL) were added and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 72%
  • Mass Spectral Analysis m/z=581.2 (M+H)+
  • Preparation of 22.5
  • To a suspension of 22.4 (0.62 g, 1.06 mmol, 1.0 eq) in ethanol (10 mL) was added sodium acetate (0.58 g, 7.1 mmol, 6.7 eq) and iodomethane (2.8c) (0.37 mL, 5.8 mmol, 5.5 eq). The reaction mixture was heated under reflux for 10 h. Water (100 mL) and dichloromethane (100 mL) were added and the two phases were separated. The aqueous phase was extracted with dichloromethane (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 78%
  • 1H NMR (400 MHz, CDCl3) δ 7.78 (m, 1H), 7.61 (t, 1H), 7.45 (m, 2H), 7.35 (m, 2H), 7.06 (d, 1H), 5.685 & 5.675 (2s, 1H), 4.01-3.42 (m, 6H), 3.33 (brs, 2H), 3.00 (s, 3H), 2.46-2.22 (m, 3H), 2.00-1.69 (m, 3H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=565.3 (M+H)+
  • Preparation of 22E
  • To a solution of 22.5 (0.45 g, 0.8 mmol, 1.0 eq) in a mixture of methanol (20 mL) and water (5 mL) at 0° C. was added potassium carbonate (0.86 g, 4.8 mmol, 6.0 eq) portion wise. The reaction mixture was warmed to room temperature and stirred for 10 h at room temperature. Brine (50 mL) and chloroform (50 mL) were added and the two phases were separated. The aqueous phase was extracted with chloroform (3×50 mL) and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added dropwise a 2.0M solution of hydrogen chloride in diethyl ether (0.8 mL, 1.6 mmol, 2.0 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 86%
  • 1H NMR (400 MHz, DMSO d6) δ 9.01 (brs, 2H), 7.80 (dd, 1H), 7.46 (m, 5H), 7.22 (d, 1H), 6.06 (s, 1H), 3.45 (m, 2H), 3.32-3.03 (m, 9H), 2.29 (m, 2H), 2.18 (m, 1H), 1.99 (m, 2H), 1.81 (m, 1H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=469.2 (M+H)+
  • Elemental analysis:
  • C26H32N2O4S, 1HCl
  • Theory: % C, 61.83; % H, 6.59; % N, 5.55.
  • Found: % C, 61.82; % H, 6.60; % N, 5.51.
  • [α]D 25=−4.50 (c. 10.3 mg/mL, MeOH)
  • Example 23A
  • 23A was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: Method 1B was used and 1.2 was replaced by 23.1a (see also step 23.1).
  • Step 1.3: Method 1C was used (see also step 23.3).
  • Step 1.4: Method 1E was used (see also step 23.4).
  • 1H NMR (400 MHz, CDCl3) δ 10.20 (m, 2H), 7.40 (m, 4H), 7.22 (m, 1H), 7.04 (m, 2H), 6.91 (m, 1H), 5.66 (s, 1H), 3.85-3.50 (m, 5H), 3.31 (m, 3H), 2.60 (m, 1H), 2.13 (m, 1H), 1.27 (m, 3H), 1.16 (m, 3H)
  • Mass Spectral Analysis m/z=363.2 (M+H)+
  • Example 23B
  • 23B was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: Method 1B was used and 1.2 was replaced by 23.1b (see also step 23.1).
  • Step 1.3: Method 1C was used (see also step 23.3).
  • Step 1.4: Method 1E was used (see also step 23.4).
  • 1H NMR (400 MHz, CDCl3) δ 10.33 (m, 1H), 9.21 (m, 1H), 7.39 (m, 5H), 7.21 (m, 1H), 6.98 (m, 1H), 6.87 (m, 1H), 5.50 (s, 1H), 3.55 (m, 4H), 3.34 (m, 2H), 2.93 (m, 2H), 2.44 (m, 1H), 2.33 (m, 1H), 1.83 (m, 1H), 1.70 (m, 1H), 1.26 (m, 3H), 1.16 (m, 3H)
  • Mass Spectral Analysis m/z=377.0 (M+H)+
  • Example 23C
  • 23C was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.1: Method 1B was used and 1.2 was replaced by 23.5 (see also step 23.5).
  • Step 1.3: Method 1C was used (see also step 23.7).
  • Step 1.4: Method 1E was used (see also step 23.8).
  • 1H NMR (400 MHz, DMSO d6) δ 9.28 (brm, 2H), 7.43 (d, 2H), 7.35 (d, 2H), 7.27 (m, 1H), 7.01 (d, 1H), 6.97 (m, 2H), 5.57 (s, 1H), 4.01 (brs, 2H), 3.44 (brs, 2H), 3.22 (brs, 2H), 2.36 (m, 2H), 2.27 (m, 4H), 2.04 (m, 2H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=403.2 (M+H)+
  • Example 24A Preparation of 24.2
  • To a solution of 24.1 (9.37 g, 60 mmol, 1.0 eq) in dry methanol (100 mL) was added pyrrolidine (10 mL, 120 mmol, 2.0 eq) followed by 2′-hydroxyacetophenone (1.1a) (7.22 mL, 60 mmol, 1.0 eq). The reaction mixture was heated under reflux for 10 h. The volatiles were removed under reduced pressure and the residue was dissolved in ethyl acetate (200 mL). The mixture was washed with a 1M aqueous solution of hydrochloric acid (3×50 mL), with a 1M aqueous solution of sodium hydroxide (3×50 mL) and brine. The organic extracts were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.86 (dd, 1H), 7.48 (m, 1H), 6.98 (m, 2H), 3.96 (m, 4H), 2.71 (s, 2H), 2.12 (m, 2H), 1.99 (m, 2H), 1.74 (m, 2H), 1.61 (m, 2H)
  • Preparation of 24.3
  • To an oven-dried two-neck 500 mL flask charged with a solution of 24.2 (16.46 g, 60 mmol, 1.0 eq) in dry tetrahydrofuran (100 mL) at −78° C. under nitrogen was added a 1.0M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (72 mL, 72 mmol, 1.2 eq) over a 30 min time period. The mixture was kept at −78° C. for 1 h and a solution of N-phenylbis(trifluoromethanesulfonamide) (1.4) (25.72 g, 72 mmol, 1.2 eq) in tetrahydrofuran (100 mL) was added to the mixture over a 30 min time period. The reaction mixture was kept at −78° C. for 1 h, and was slowly warmed to room temperature and stirred for 10 h at room temperature. Ice water (100 mL) was added to quench the reaction and the product was extracted with diethyl ether (200 mL). The organic phase was then washed with a 1M aqueous solution of hydrochloric acid (3×100 mL), with a 1M aqueous solution of sodium hydroxide (3×100 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 90%
  • 1H NMR (400 MHz, CDCl3) δ 7.34-7.19 (m, 2H), 6.97 (m, 1H), 6.89 (m, 1H), 5.60 (s, 1H), 4.03-3.91 (m, 4H), 2.20 (m, 2H), 2.09-1.97 (m, 2H), 1.81 (m, 2H), 1.62 (m, 2H)
  • Preparation of 24.4
  • To a solution of 24.3 (22 g, 54.14 mmol, 1.0 eq) in dimethoxyethane (200 mL) under nitrogen was added sequentially a 2M aqueous solution of sodium carbonate (81.2 mL, 162.42 mmol, 3.0 eq), lithium chloride (6.88 g, 162.42 mmol, 3.0 eq), tetrakis(triphenylphosphine)palladium(0) (1.25 g, 1.08 mmol, 0.02 eq), and 4-N,N-diethylphenylboronic acid (1.6) (13.16 g, 59.55 mmol, 1.1 eq). The reaction mixture was heated under reflux for 10 h. Water (200 mL) and diethyl ether (300 mL) were added and the two phases were separated. The aqueous phase was further extracted with diethyl ether (2×100 mL) and the combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 95%
  • 1H NMR (400 MHz, CDCl3) δ 7.38 (m, 4H), 7.18 (m, 1H), 6.99 (m, 1H), 6.93 (m, 1H), 6.85 (m, 1H), 5.62 (s, 1H), 3.99 (m, 4H), 3.57 (brs, 2H), 3.32 (brs, 2H), 2.24-2.02 (m, 4H), 1.80 (m, 2H), 1.65 (m, 2H), 1.21 (m, 6H)
  • Preparation of 24A
  • To a cold (0° C.) solution of 24.4 (22.32 g, 51.48 mmol, 1.0 eq) in tetrahydrofuran (200 mL) was added a 1.0M aqueous solution of hydrochloric acid (155 mL, 155 mmol, 3.0 eq). The mixture was stirred at room temperature for 10 h and then concentrated under reduced pressure. The resulting solid was collected by filtration, washed with hexane/ethyl acetate mixture (20:1), and dried under vacuum.
  • Yield: 85%
  • 1H NMR (400 MHz, CDCl3) δ 7.40 (m, 4H), 7.23 (m, 1H), 7.04 (d, 1HO, 7.00 (d, 1H), 6.91 (m, 1H), 5.62 (s, 1H), 3.57 (brs, 2H), 3.32 (brs, 2H), 2.87 (m, 2H), 2.50 (m, 2H), 2.33 (m, 2H), 1.94 (m, 2H), 1.21 (m, 6H)
  • Mass Spectral Analysis m/z=390.2 (M+H)+
  • Example 24B/Example 24C Preparation of 24B/24C
  • To a solution of 24A (0.51 g, 1.3 mmol, 1.0 eq) in dry tetrafydrofuran (30 mL) at 0° C. was added sodium borohydride (50 mg, 1.3 mmol, 1.0 eq) in one portion under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 1 h. Water (50 mL) and diethyl ether (100 mL) were added and the two phases were separated. The aqueous phase was further extracted with diethyl ether (2×50 mL) and the combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated to give the mixture of two isomers. The crude product was purified by preparative liquid chromatography affording 24B and 24C.
  • (24B) 1H NMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 7.18 (m, 1H), 6.97 (m, 2H), 6.85 (m, 1H), 5.55 (s, 1H), 3.73 (m, 1H), 3.58 (brs, 2H), 3.33 (brs, 2H), 2.51 (brs, 4H), 2.21 (m, 2H), 1.52 (m, 2H), 1.22 (brd, 6H)
  • Mass Spectral Analysis m/z=392.2 (M+H)+
  • (24C) 1H NMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 7.18 (m, 1H), 7.01-6.81 (m, 3H), 5.73 & 5.55 (2s, 1H), 4.07 & 3.74 (2m, 1H), 3.59 (brs, 2H), 3.34 (brs, 2H), 3.16 (brs, 4H), 2.31-1.89 (m, 2H), 1.68-1.46 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=392.2 (M+H)+
  • Example 24D/Example 24E Preparation of 24D/24E
  • To a stirred solution of 24A (0.63 mL, 1.62 mmol, 2.0 eq) in dry dichloromethane (20 mL) was added sequentially n-propylamine (3.4d) (0.16 g, 1.94 mmol, 1.2 eq), acetic acid (0.11 mL, 1.94 mmol, 1.2 eq), and sodium cyanoborohydride (0.153 g, 2.43 mmol, 1.5 eq). The reaction mixture was stirred at room temperature for 10 h. Water (40 mL) was added and the aqueous layer was basified to pH=10 with a 1M aqueous solution of sodium hydroxide. The two phases were separated and the aqueous phase was saturated with sodium chloride and extracted with dichloromethane (3×50 mL). The combined organic extracts were dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude mixture, which was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity).
  • (24D) 1H NMR (400 MHz, CDCl3) δ 7.38 (m, 4H), 7.17 (m, 1H), 6.99 (dd, 1H), 6.90 (dd, 1H), 6.84 (m, 1H), 5.91 (s, 1H), 3.57 (brs, 2H), 3.31 (brs, 2H), 2.75 (brs, 1H), 2.65 (t, 2H), 2.11 (m, 2H), 1.98 (m, 2H), 1.82-1.46 (m, 7H), 1.21 (m, 6H), 0.95 (t, 3H)
  • Mass Spectral Analysis m/z=433.2 (M+H)+
  • (24E) 1H NMR (400 MHz, CDCl3) δ 7.38 (m, 4H), 7.16 (m, 1H), 6.98 (dd, 1H), 6.93 (dd, 1H), 6.83 (m, 1H), 5.54 (s, 1H), 3.57 (brs, 2H), 3.31 (brs, 2H), 2.64 (t, 2H), 2.53 (m, 1H), 2.20 (m, 2H), 1.83-1.42 (m, 7H), 1.21 (m, 6H), 0.94 (t, 3H)
  • Mass Spectral Analysis m/z=433.2 (M+H)+
  • Example 24F
  • 24F was obtained according to a procedure similar to the one described for 24D, with the following exception:
  • Step 24.6: 3.4d was replaced by 3.4j.
  • 1H NMR (400 MHz, CDCl3) δ 7.38 (m, 4H), 7.17 (m, 1H), 6.96 (m, 2H), 6.84 (m, 1H), 5.54 (s, 1H), 3.57 (m, 2H), 3.32 (m, 2H), 2.35 (s, 6H), 2.25 (m, 3H), 1.79 (m, 4H), 1.46 (m, 2H), 1.26 (m, 3H), 1.16 (m, 3H)
  • Mass Spectral Analysis m/z=419.2 (M+H)+
  • Example 24G
  • 24G was obtained according to a procedure similar to the one described for 24E, with the following exception:
  • Step 24.6: 3.4d was replaced by 3.4j.
  • 1H NMR (400 MHz, CDCl3) δ 7.40 (m, 4H), 7.18 (m, 1H), 7.00 (m, 1H), 6.91 (m, 1H), 6.85 (m, 1H), 5.89 (s, 1H), 3.57 (m, 2H), 3.32 (m, 2H), 2.51 (m, 7H), 2.20 (m, 2H), 2.06 (m, 2H), 1.76 (m, 4H), 1.26 (m, 3H), 1.16 (m, 3H)
  • Mass Spectral Analysis m/z=419.2 (M+H)+
  • Example 25A
  • 25A was obtained according to a procedure similar to the one described for compound 1.8a with the following exception:
  • Step 1.1: 1.2 was replaced by 25.1 (see also step 25.1).
  • 1H NMR (400 MHz, DMSO d6) δ 7.42 (d, 2H), 7.38 (d, 2H), 7.19 (m, 1H), 6.97 (m, 2H), 6.86 (m, 1H), 5.62 (s, 1H), 3.96 (m, 2H), 3.79 (m, 2H), 3.57 (brs, 2H), 3.32 (brs, 2H), 2.03 (d, 2H), 1.84 (m, 2H), 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=378.2 (M+H)+
  • Example 26A Preparation of 26.2
  • To a solution of 1.5a (2.08 g, 4.63 mmol, 1 eq) in dry tetrahydrofuran (40 mL) was added tetrakis(triphenylphosphine)palladium(0) (0.535 g, 0.463 mmol, 0.1 eq), followed by 4-cyanobenzylzinc bromide (26.1) (0.5M solution in tetrahydrofuran, 23.16 mL, 11.58 mmol, 2.5 eq) drop wise under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 10 h. A saturated aqueous solution of ammonium chloride (40 mL) was added to quench the reaction and diethyl ether (50 mL) was added to partition the two phases. The aqueous phase was extracted with diethyl ether (3×50 mL) and the combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixture of increasing polarity). Yield: 62%
  • 1H NMR (400 MHz, CDCl3) δ 7.59 (d, 2H), 7.34 (d, 2H), 7.14 (m, 1H), 7.00 (dd, 1H), 6.88 (dd, 1H), 6.82 (m, 1H), 5.28 (s, 1H), 3.95-3.75 (m, 4H), 3.28 (m, 2H), 1.99 (m, 2H), 1.59 (m, 2H), 1.46 (s, 9H)
  • Mass Spectral Analysis m/z=417 (M+H)+
  • Preparation of 26.3a & 26.3b
  • A mixture of 26.2 (1.2 g, 2.88 mmol) in concentrated hydrochloric acid (30 mL) was heated under reflux for 10 h and then concentrated under reduced pressure to give the crude mixture of 26.3a and 26.3b. A 80 mg quantity of the mixture was purified by preparative liquid chromatography. The remaining mixture (26.3a/26.3b) was used for the next step without further purification.
  • 26.3a: 1H NMR (400 MHz, DMSO-d6) δ 12.87 (s, b, 1H), 8.58 (m, 2H), 7.86 (m, 2H), 7.41 (m, 2H), 7.21-7.12 (m, 2H), 6.92 (dd, 1H), 6.86 (m, 1H), 5.70 (s, 1H), 3.85 (s, 2H), 3.19 (m, 4H), 2.06 (m, 2H), 1.86 (m, 2H)
  • Mass Spectral Analysis m/z=336.2 (M+H)+
  • 26.3b: 1H NMR (400 MHz, DMSO-d6) δ 13.00 (s, b, 1H), 8.68 (m, 1H), 8.29 (m, 1H), 7.97 (m, 2H), 7.84 (dd, 1H), 7.50 (m, 2H), 7.41 (s, 1H), 7.27 (m, 1H), 7.03-6.94 (m, 2H), 3.19-3.00 (m, 4H), 2.82 (s, 2H), 1.91 (m, 2H), 1.63 (m, 2H)
  • Mass Spectral Analysis m/z=336.2 (M+H)+
  • Preparation of 26.4a & 26.4b
  • To a solution of the mixture of 26.3a and 26.3b (1 g, 2.69 mmol) in methanol (50 mL) was slowly added a 4.0M solution of hydrogen chloride in dioxane (20 mL). The reaction mixture was stirred at room temperature for 10 h and concentrated under reduced pressure. The residue was dissolved in ethyl acetate (100 mL), washed with a 1M aqueous solution of sodium carbonate (4×50 mL), brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude mixture of 26.4a and 26.4b. A small amount (150 mg) of the crude mixture was purified by column chromatography (eluent: hexane/ethyl acetate mixture of increasing polarity) and repurified preparative liquid chromatography. The remaining mixture (26.4a/26.4b) was used for the next step without further purification. Yield: 90%
  • 26.4a: 1H NMR (400 MHz, CDCl3) δ 9.05 (s, b, 1H), 8.72 (s, b, 1H), 7.98 (d, 2H), 7.29 (d, 2H), 7.17 (m, 1H), 7.11 (m, 1H), 6.93-6.85 (m, 2H), 5.29 (s, 1H), 3.91 (s, 3H), 3.80 (s, 2H), 3.37 (m, 4H), 2.24 (m, 2H), 1.95 (m, 2H)
  • Mass Spectral Analysis m/z=350.2 (M+H)+
  • 26.4b: 1H NMR (400 MHz, CDCl3) δ 9.42 (s, b, 1H), 8.95 (s, b, 1H), 8.05 (d, 2H), 7.66 (d, 1H), 7.40-7.22 (m, 4H), 7.00 (m, 1H), 6.92 (d, 1H), 3.94 (s, 3H), 3.25 (m, 4H), 2.78 (s, 2H), 2.04 (m, 2H), 1.75 (m, 2H)
  • Mass Spectral Analysis m/z=350.2 (M+H)+
  • Preparation of 26.5a & 26.5b
  • To a solution of the mixture of 26.4a and 26.4b (0.5 g, 1.5 mmol, 1 eq) in dry dichloromethane (30 mL) at 0° C. was slowly added triethylamine (0.42 mL, 3 mmol, 2 eq) and a solution of di-tert-butyl-dicarbonate 4.7 (0.38 g, 1.74 mmol, 1.2 eq) in dichloromethane (10 mL) drop wise. The reaction mixture was slowly warmed up to room temperature and stirred at room temperature for 10 h. Dichloromethane (50 mL) was added and the mixture was washed with a 1N aqueous solution of hydrochloric acid (3×50 mL), brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude mixture of 26.5a and 26.5b, which was used for the next step without purification.
  • Preparation of 26.6a & 26.6b
  • To a solution of the mixture of 26.5a and 26.5b (0.57 g, 1.26 mmol, 1 eq) in a mixture methanol (15 mL), tetrahydrofuran (15 mL) and water (15 mL) was added lithium hydroxide monohydrate (0.21 g, 5 mmol, 4 eq) in one portion. The reaction mixture was stirred at room temperature for 10 h. The volatiles were removed under reduced pressure and the remaining aqueous solution was acidified to pH=3 with a 1N aqueous solution of hydrochloric acid while stirring. The mixture was stirred for 1 h at room temperature and left at room temperature for 10 h. The resulting solid was collected by filtration, washed with water, and dried under vacuum to give the mixture of 26.6a and 26.6b, which was used for the next step without further purification.
  • Preparation of 26.7a & 26.7b
  • To a stirred solution of the mixture of 26.6a and 26.6b (0.49 g, 1.12 mmol, 1 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (0.46 mL, 2.69 mmol, 2.4 eq), diethylamine 1.12 (0.24 g, 3.36 mmol, 3 eq) at room temperature. The mixture was stirred for 10 min at room temperature. The mixture was cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (0.43 g, 1.34 mmol, 1.2 eq) was added portion wise. The reaction mixture was slowly warmed up to room temperature and stirred at room temperature for an additional 10 h. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (100 mL) and a 1M aqueous solution of sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), a 1M aqueous solution of hydrochloric acid (3×50 mL), brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude mixture of 26.7a and 26.7b. The crude mixture was purified by column chromatography (eluent: hexane/ethyl acetate mixture of increasing polarity). A small amount (85 mg) of the purified mixture was separated by preparative liquid chromatography. The remaining mixture (26.7a/26.7b) was used for the next step without further purification. Yield: 81% over three steps
  • 26.7a: 1H NMR (400 MHz, CDCl3) δ 7.33-7.24 (m, 4H), 7.15-7.07 (m, 2H), 6.89-6.80 (m, 2H), 5.25 (s, 1H), 3.84 (m, 2H), 3.74 (s, 2H), 3.55 (m, 2H), 3.28 (m, 4H), 1.98 (m, 2H), 1.57 (m, 2H), 1.46 (s, 9H), 1.18 (m, 6H)
  • Mass Spectral Analysis m/z=491.1 (M+H)+
  • 26.7b: 1H NMR (400 MHz, CDCl3) δ 7.63 (dd, 1H), 7.39 (m, 2H), 7.31 (m, 2H), 7.22 (m, 1H), 7.17 (s, 1H), 6.95 (m, 1H), 6.90 (dd, 1H), 3.81 (m, 2H), 3.58 (m, 2H), 3.34 (m, 2H), 3.17 (m, 2H), 2.71 (s, 2H), 1.82 (m, 2H), 1.43 (s, 9H), 1.38 (m, 2H), 1.22 (m, 6H)
  • Mass Spectral Analysis m/z=491.1 (M+H)+
  • Preparation of 26A
  • To a cold (0° C.) stirred solution of the mixture of 26.7a and 26.7b (0.36 g, 0.73 mmol, 1 eq) in dry dichloromethane (20 mL) was added dropwise a 4.0 M solution of hydrogen chloride in dioxane (1.8 mL, 7.2 mmol, 10 eq). The mixture was stirred at room temperature for 10 h and concentrated under reduced pressure to give the crude mixture of 26A and 26.8. The crude mixture was purified by preparative liquid chromatography. Yield: 85%
  • 26A: 1H NMR (400 MHz, CDCl3) δ 9.35 (s, b, 1H), 9.00 (s, b, 1H), 7.30 (m, 4H), 7.14 (m, 2H), 6.87 (m, 2H), 5.28 (s, 1H), 3.76 (s, 2H), 3.55 (m, 2H), 3.24 (m, 6H), 2.11 (m, 2H), 1.93 (m, 2H), 1.20 (m, 6H)
  • Mass Spectral Analysis m/z=391.0 (M+H)+
  • 26.8: 1H NMR (400 MHz, CDCl3) δ 9.12 (s, b, 1H), 8.71 (s, b, 1H), 7.65 (d, 1H), 7.39 (d, 2H), 7.31 (d, 2H), 7.28-7.19 (m, 2H), 7.00 (m, 1H), 6.92 (d, 1H), 3.59 (m, 2H), 3.29 (m, 6H), 2.78 (s, 2H), 2.05 (m, 2H), 1.78 (m, 2H), 1.23 (m, 6H)
  • Mass Spectral Analysis m/z=391.0 (M+H)+
  • Example 26B Preparation of 26B
  • To a stirred solution of 26.8 (0.12 g, 0.26 mmol, 1 eq) in methanol (10 mL) was added palladium [24 mg, 10 wt. % (dry basis) on activated carbon, 20% wt. eq]. The reaction mixture was stirred under hydrogen atmosphere using a hydrogen balloon at room temperature for 10 h. The palladium on activated carbon was filtered off on a celite pad and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol/ammonium hydroxide mixture of increasing polarity). The desired fractions were combined and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in dichloromethane was added dropwise a 2.0M solution of hydrogen chloride in diethyl ether (0.26 mL, 0.52 mmol, 2 eq). The mixture was then stirred for 1 h at room temperature, concentrated under reduced pressure, and dried under vacuum. Yield: 88%
  • 1H NMR (400 MHz, CDCl3) δ 9.41 (s, b, 1H), 8.95 (s, b, 1H), 7.40 (m, 1H), 7.33 (m, 2H), 7.25-7.14 (m, 3H), 6.97 (m, 1H), 6.86 (m, 1H), 3.62-3.04 (m, 10H), 2.63 (m, 1H), 2.03-1.49 (m, 6H), 1.20 (m, 6H)
  • Mass Spectral Analysis m/z=393.0 (M+H)+
  • Example 27A Preparation of 27A
  • A solution of 1A (0.66 g, 1.75 mmol, 1.0 eq) in anhydrous methanol (13 mL) was hydrogenated at atmospheric pressure in the presence of palladium hydroxide [Pd(OH)2: Pearlman's catalyst] (0.120 g, 0.09 mmol, 0.05 eq) for 10 h. The mixture was then filtered through celite. The filtrate was concentrated and was hydrogenated at atmospheric pressure in the presence of palladium hydroxide (0.120 g) for an additional 10 h. The mixture was filtered through celite and the filtrate was concentrated to dryness under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane was added drop wise a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (5 mL). The mixture was then stirred for 1 h at room temperature and concentrated under reduced pressure. Diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether and ethyl acetate. Yield: 63%
  • 1H NMR (400 MHz, DMSO d6) δ 9.15 (m, 2H), 7.30 (m, 4H), 7.10 (m, 1H), 6.90 (m, 1H), 6.75 (m, 1H), 6.60 (m, 1H), 4.20 (m, 1H), 3.40 (m, 3H), 3.20 (m, 4H), 3.00 (m, 1H), 2.15 (m, 1H), 1.95 (m, 5H), 1.05 (m, 6H)
  • Mass Spectral Analysis m/z=379.1 (M+H)+
  • Elemental analysis:
  • C24H30N2O2, 1HCl, 0.75H2O
  • Theory: % C, 67.28; % H, 7.65; % N, 6.54.
  • Found: % C, 67.32; % H, 7.63; % N, 6.37.
  • Example 27B Preparation of 27B
  • 27A (racemic mixture) (10 g, 24.10 mmol, 1.0 eq) was resolved using Chiral HPLC method:
  • Column: Chiralpak AD-H, 4.6×250 mm, 5p, Chiral Technologies PN#19325
  • Column temperature: room temperature
  • Detection: UV photo diode array, 200 to 300 nm, extract at 275 nm
  • Injection volume: 40 μL of 2 mg/mL sample in EtOH:MeOH (80:20)
  • Flow: 1 mL/minute
  • Mobile Phase: 85% Solution A, 15% Solution B
  • Solution A: 0.1% Di-isopropylethylamine in Hexane (HPLC Grade)
  • Solution B: 80% Ethanol, 20% Methanol (both HPLC Grade)
  • Note: Methanol is miscible in Hexane only if first dissolved in Ethanol. Solution B should be pre-mixed
  • Run time: 25 min
  • HPLC: Waters Alliance 2695 (system dwell volume is ˜350 μL)
  • Detector: Waters 996 (resolution: 4.8 nm, scan rate: 1 Hz)
  • Yield: 40%
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (m, 2H), 7.28 (m, 4H), 7.14 (m, 1H), 6.90 (d, 1H), 6.80 (m, 1H), 6.63 (d, 1H), 4.25 (m, 1H), 3.42 (m, 3H), 3.24 (m, 4H), 2.97 (m, 1H), 2.20 (m, 1H), 1.97 (m, 5H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=379.4 (M+H)+
  • Chiral HPLC Method: tR=8.64 min (ee=97%)
  • Elemental analysis:
  • C24H30N2O2, 1HCl, 0.25H2O
  • Theory: % C, 68.72; % H, 7.57; % N, 6.68.
  • Found: % C, 68.87; % H, 7.52; % N, 6.68.
  • [α]D 25=+58.40 (c. 0.01, MeOH)
  • Determination of absolute configuration of Example 27b Preparation of 27.3
  • Compound 27.2 (0.45 g, 1.78 mmol, 1.1 eq) was added at 0° C. to a solution of 27B (0.67 g, 1.61 mmol, 1 eq) and triethylamine (0.74 mL, 5.33 mmol, 3.3 eq) in dichloromethane (6 mL). The reaction was warmed to room temperature and stirred overnight at room temperature. The mixture was washed with a saturated aqueous solution of sodium hydrogenocarbonate and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 64%
  • 1H NMR (400 MHz, DMSO d6) δ 7.30 (m, 4H), 7.11 (t, 1H), 6.90 (d, 1H), 6.77 (t, 1H), 6.61 (d, 1H), 4.23 (m, 1H), 3.39 (br m, 9H), 2.93 (d, 1H), 2.37 (m, 2H), 2.24 (m, 1H), 2.06 (m, 2H), 1.93 (m, 6H), 1.53 (m, 1H), 1.41 (m, 1H), 1.10 (m, 6H), 1.03 (s, 3H), 0.83 (s, 3H)
  • Mass Spectral Analysis m/z=593.4 (M+H)+
  • Elemental analysis:
  • C33H44N2O5S, 0.25H2O
  • Theory: % C, 68.37; % H, 7.51; % N, 4.69.
  • Theory: % C, 68.38; % H, 7.50; % N, 4.55.
  • X-Ray Crystallography Data:
  • Single crystals were grown as needles by dissolving 27.3 (10 mg, 0.017 mmol, 1 eq) in isopropanol (1 mL) and letting sit still at room temperature for 72 h. Crystal data and structure refinement for 27.3:
  • Identification code: ptut001
  • Empirical formula: C34H44N2O5S
  • Formula weight: 592.77
  • Temperature: 120(2) K
  • Wavelength: 0.71073 A
  • Crystal system, space group: Monoclinic, P2(1)
  • Unit cell dimensions:
  • a=15.135(2) A, alpha=90 deg
  • b=6.1924(10) A, beta=91.802(2) deg
  • c=16.602(3) A, gamma=90 deg
  • Volume: 1555.2(4) A3
  • Z, Calculated density: 2, 1.266 Mg/m3
  • Absorption coefficient: 0.148 mm−1
  • F(000): 636
  • Crystal size: 0.30×0.08×0.04 mm
  • Theta range for data collection: 1.79 to 27.79 deg
  • Limiting indices: −18<=h<=19, −7<=k<=7, −20<=l<=21
  • Reflections collected/unique: 12166/6251 [R(int)=0.0168]
  • Completeness to theta=27.79: 91.9%
  • Absorption correction: Semi-empirical from equivalents
  • Max. and min transmission: 0.9941 and 0.9569
  • Refinement method: Full-matrix least-squares on F2
  • Data/restraints/parameters: 6251/1/383
  • Goodness-of-fit on F2: 1.040
  • Final R indices [I>2sigma(I)]: R1=0.0392, wR2=0.1030
  • R indices (all data): R1=0.0401, wR2=0.1041
  • Absolute structure parameter: −0.03(6)
  • Largest diff. peak and hole: 0.365 and −0.200 e.A−3
  • Example 27C Preparation of 27C
  • 27A (racemic mixture) (10 g, 24.10 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiralpak AD-H, 4.6×250 mm, 5p, Chiral Technologies PN#19325
  • Column temperature: room temperature
  • Detection: UV photo diode array, 200 to 300 nm, extract at 275 nm
  • Injection volume: 40 μL of 2 mg/mL sample in EtOH:MeOH (80:20)
  • Flow: 1 mL/minute
  • Mobile phase: 85% Solution A, 15% Solution B
  • Solution A: 0.1% Di-isopropylethylamine in Hexane (HPLC Grade)
  • Solution B: 80% Ethanol, 20% Methanol (both HPLC Grade)
  • Run time: 25 min
  • HPLC: Waters Alliance 2695 (system dwell volume is ˜350 μL.)
  • Detector: Waters 996 (Resolution: 4.8 nm, Scan Rate: 1 Hz)
  • Yield: 40%
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (m, 2H), 7.28 (m, 4H), 7.14 (m, 1H), 6.90 (d, 1H), 6.79 (m, 1H), 6.63 (d, 1H), 4.25 (m, 1H), 3.44 (m, 3H), 3.24 (m, 4H), 2.96 (m, 1H), 2.18 (m, 1H), 1.97 (m, 5H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=379.4 (M+H)+
  • Chiral HPLC Method: tR=11.914 min. (ee=100%)
  • Elemental analysis:
  • C24H30N2O2, 1HCl, 0.25H2O
  • Theory: % C, 68.72; % H, 7.57; % N, 6.68.
  • Found: % C, 68.79; % H, 7.55; % N, 6.68.
  • [α]D 25=−63.59 (c. 0.01, MeOH)
  • Example 27D
  • 27D was obtained according to a procedure similar to the one described for 27A, with the following exception:
  • Step 27.3: Method 27A was used and 1A was replaced by 1D.
  • 1H NMR (400 MHz, DMSO d6) δ 9.05 (m, 2H), 7.31 (q, 4H), 6.98 (m, 2H), 6.36 (dd, 1H), 6.47 (dd, 1H), 3.51-3.33 (m, 2H), 3.29-3.11 (m, 5H), 2.96 (m, 1H), 2.19 (m, 1H), 2.05-1.82 (m, 5H), 1.20-1.00 (m, 6H)
  • Mass Spectral Analysis m/z=397.3 (M+H)+
  • Example 27E
  • 27E was obtained from 27D by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 8.82 (m, 2H), 7.31 (m, 4H), 6.97 (m, 2H), 6.37 (m, 1H), 4.27 (m, 1H), 3.42 (m, 2H), 3.23 (m, 5H), 2.97 (m, 1H), 2.20 (m, 1H), 1.94 (m, 5H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=397.4 (M+H)+
  • Elemental analysis:
  • C24H29FN2O2, 1HCl, 0.33H2O
  • Theory: % C, 65.71; % H, 7.09; % N, 6.36.
  • Found: % C, 65.68; % H, 7.07; % N, 6.41.
  • [α]D 25=+65.32 (c=9.85 mg/mL, MeOH)
  • Example 27F
  • 27F was obtained from 27D by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (m, 2H), 7.32 (m, 4H), 6.98 (m, 2H), 6.37 (m, 1H), 4.27 (m, 1H), 3.42 (m, 2H), 3.24 (m, 5H), 2.97 (m, 1H), 2.20 (m, 1H), 1.95 (m, 5H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=397.3 (M+H)+
  • Elemental analysis:
  • C24H29FN2O2, 1HCl, 0.2H2O
  • Theory: % C, 66.03; % H, 7.02; % N, 6.42.
  • Found: % C, 66.07; % H, 6.99; % N, 6.34.
  • [α]D 25=−65.36 (c=9.75 mg/mL, MeOH)
  • Example 27G
  • 27G was obtained according to a procedure similar to the one described for 27A, with the following exception:
  • Step 27.3: Method 27A was used and 1A was replaced by 2C.
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (brs, 1H), 8.97 (brs, 1H), 7.32 (d, 2H), 7.27 (d, 2H), 6.84 (d, 1H), 6.73 (dd, 1H), 6.12 (d, 1H), 4.21 (m, 1H), 3.55 (m, 3H), 3.42 (brs, 1H), 3.20 (brm, 5H), 2.94 (m, 1H), 2.16 (m, 1H), 1.92 (m, 5H), 1.09 (m, 7H), 0.46 (m, 2H), 0.18 (m, 2H)
  • Mass Spectral Analysis m/z=449.3 (M+H)+
  • Elemental analysis:
  • C28H36N2O3, 1HCl, 1H2O
  • Theory: % C, 66.85; % H, 7.81; % N, 5.57; % Cl, 7.05.
  • Found: % C, 67.02; % H, 7.51; % N, 5.54; % Cl, 7.25.
  • Example 27H
  • 27H was obtained according to a procedure similar to the one described for 27A, with the following exception:
  • Step 27.3: Method 27A was used and 1A was replaced by 1N.
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (m, 1.5H), 8.53 (d, 1H), 7.70 (dd, 1H), 7.52 (d, 1H), 7.16 (m, 1H), 6.93 (dd, 1H), 6.82 (m, 1H), 6.63 (d, 1H), 4.36 (dd, 1H), 3.45 (q, 2H), 3.33-3.15 (m, 5H), 2.98 (m, 1H), 2.22 (m, 1H), 2.07-1.85 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=380.2 (M+H)+
  • Example 27I
  • 27I was obtained from 2711 by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 8.89 (m, 2H), 8.52 (d, 1H), 7.68 (dd, 1H), 7.51 (d, 1H), 7.16 (m, 1H), 6.94 (m, 1H), 6.82 (m, 1H), 6.62 (m, 1H), 4.35 (m, 1H), 3.44 (q, 2H), 3.26 (m, 5H), 2.98 (m, 1H), 2.23 (m, 1H), 1.95 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=380.2 (M+H)+
  • Elemental analysis:
  • C23H29N3O2, 1.3HCl, 1.4H2O
  • Theory: % C, 61.10; % H, 7.38; % N, 9.29; % Cl, 10.19.
  • Found: % C, 61.01; % H, 7.35; % N, 9.21; % Cl, 10.41.
  • [α]D 25=+44.59 (c=9.65 mg/mL, MeOH)
  • Example 27J
  • 27J was obtained from 2711 by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 9.08 (m, 2H), 8.53 (d, 1H), 7.70 (dd, 1H), 7.52 (d, 1H), 7.16 (m, 1H), 6.93 (m, 1H), 6.82 (m, 1H), 6.63 (m, 1H), 4.36 (m, 1H), 3.45 (q, 2H), 3.25 (m, 5H), 2.97 (m, 1H), 2.22 (m, 1H), 1.97 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=380.2 (M+H)+
  • Elemental analysis:
  • C23H29N3O2, 2HCl, 1.75H2O
  • Theory: % C, 57.08; % H, 7.19; % N, 8.68; % Cl, 14.65.
  • Found: % C, 56.92; % H, 7.15; % N, 8.58; % Cl, 15.02.
  • [α]D 25=−35.54 (c=10.3 mg/ml, MeOH)
  • Example 27K
  • 27K was obtained according to a procedure similar to the one described for 27A, with the following exception:
  • Step 27.3: Method 27A was used and 1A was replaced by 1O.
  • 1H NMR (400 MHz, DMSO d6) δ 9.17-8.85 (m, 2H), 8.53 (d, 1H), 7.70 (dd, 1H), 7.52 (d, 1H), 7.06-6.94 (m, 2H), 6.41 (dd, 1H), 4.37 (dd, 1H), 3.49-3.35 (m, 2H), 3.32-3.14 (m, 5H), 2.97 (m, 1H), 2.23 (m, 1H), 2.05-1.82 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=398.3 (M+H)+
  • Example 27L
  • 27L was obtained from 27K by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 9.15 (m, 2H), 8.54 (d, 1H), 7.72 (dd, 1H), 7.54 (d, 1H), 7.00 (m, 2H), 6.42 (dd, 1H), 4.38 (m, 1H), 3.45 (q, 2H), 3.25 (m, 5H), 2.96 (m, 1H), 2.22 (m, 1H), 1.96 (m, 5H), 1.15 (t, 2H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=398.3 (M+H)+
  • Elemental analysis:
  • C23H28FN3O2, 2HCl, 1.75H2O
  • Theory: % C, 55.04; % H, 6.73; % Cl, 14.13; % N, 8.37.
  • Found: % C, 54.85; % H, 6.53; % Cl, 14.28; % N, 8.45.
  • [α]D 25=+41.88 (c=10.2 mg/mL, MeOH)
  • Example 27M
  • 27M was obtained from 27K by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 9.14 (m, 2H), 8.54 (d, 1H), 7.79 (dd, 1H), 7.54 (d, 1H), 7.00 (m, 2H), 6.42 (dd, 1H), 4.38 (m, 1H), 3.45 (q, 2H), 3.25 (m, 5H), 2.96 (m, 1H), 2.23 (m, 1H), 1.96 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=398.3 (M+H)+
  • Elemental analysis:
  • C23H28FN3O2, 2HCl, 1.75H2O
  • Theory: % C, 55.04; % H, 6.73; % N, 8.37; % Cl, 14.13.
  • Found: % C, 54.85; % H, 6.66; % N, 8.37; % Cl, 14.31.
  • [α]D 25=−40.91 (c=10.25 mg/mL, MeOH)
  • Example 27N
  • 27N was obtained according to a procedure similar to the one described for 27A, with the following exception:
  • Step 27.3: 1A was replaced by 1S.
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Example 27O
  • 27O was obtained from 27N by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brs, 1H), 8.75 (brs, 1H), 8.50 (d, 1H), 7.65 (dd, 1H), 7.50 (d, 1H), 6.74 (s, 1H), 6.37 (s, 1H), 4.26 (m, 1H), 3.45 (q, 2H), 3.24 (m, 5H), 2.94 (m, 1H), 2.18 (m, 1H), 2.14 (s, 3H), 1.99 (s, 3H), 1.90 (m, 5H), 1.15 (t, 3H), 1.08 (t, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C25H33N3O2, 1.25HCl, 1.63H2O
  • Theory: % C, 62.25; % H, 7.84; % N, 8.70; % Cl, 9.19.
  • Found: % C, 62.52; % H, 7.64; % N, 8.30; % Cl, 8.80.
  • Example 27P
  • 27P was obtained from 27N by chiral HPLC chromatography
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (brs, 1H), 8.82 (brs, 1H), 8.50 (d, 1H), 7.65 (dd, 1H), 7.50 (d, 1H), 6.74 (s, 1H), 6.37 (s, 1H), 4.26 (m, 1H), 3.45 (q, 2H), 3.24 (m, 5H), 2.94 (m, 1H), 2.18 (m, 1H), 2.13 (s, 3H), 1.99 (s, 3H), 1.88 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C25H33N3O2, 1.2HCl, 1.6H2O
  • Theory: % C, 62.54; % H, 7.85; % N, 8.75; % Cl, 8.86.
  • Found: % C, 62.61; % H, 7.73; % N, 8.44; % Cl, 8.52.
  • Example 27Q Preparation of 27.6
  • A solution of 2.7a (15.00 g, 30.45 mmol, 1 eq) in anhydrous dichloromethane (50 mL) and anhydrous methanol (100 mL) was hydrogenated at 1 atm, in the presence of palladium, 10 weight % (dry basis) on activated carbon, wet, Degussa type E101 NE/W (3.24 g, 1.52 mmol, 0.05 eq) for 10 h. The mixture was then filtered through celite and the filtrate was concentrated to dryness under reduced pressure. The product was used without further purification. Yield: 99%
  • Mass Spectral Analysis m/z=495.4 (M+H)+
  • Preparation of 27Q
  • A 4.0M solution of hydrochloric acid in dioxane (41.9 mL, 167.46 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 27.6 (15.06 g, 30.45 mmol, 1 eq) in anhydrous methanol (50 mL). The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. The mixture was concentrated under reduced pressure. Diethyl ether (100 mL) was added to the solution. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 85%
  • 1H NMR (400 MHz, DMSO d6) δ 9.03 (m, 1H), 8.90 (m, 1H), 8.80 (s, 1H), 7.28 (m, 4H), 6.71 (d, 1H), 6.53 (m, 1H), 6.05 (d, 1H), 4.16 (m, 1H), 3.43 (m, 3H), 3.21 (m, 5H), 2.92 (m, 1H), 2.11 (m, 1H), 1.98 (m, 1H), 1.90 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=395.4 (M+H)+
  • Elemental analysis:
  • C24H30N2O2, 1HCl, 0.75H2O
  • Theory: % C, 64.85; % H, 7.37, % N, 6.30.
  • Found: % C, 65.12; % H, 7.43, % N, 6.18.
  • Example 27R Preparation of 27R
  • 27R was obtained from 27Q by chiral HPLC chromatography 27Q (racemic mixture) (10 g, 23.20 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiralpak AD-H, 4.4×250 mm
  • Column temperature: 25° C.
  • Detection: UV at 230 nm
  • Flow: 2.0 mL/minute
  • Mobile phase: 80% carbon dioxide, 20% ethanol, 0.1% ethane sulfonic acid
  • Run time: 24 min
  • The relevant fractions were combined and concentrated under reduced pressure. An aqueous 1N solution of sodium hydroxide was added to the resulting oil until the solution was basic using pH paper. The aqueous mixture was extracted with dichloromethane. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous methanol was added drop wise a 4M solution of anhydrous hydrochloric acid in dioxane (5.5 eq). The mixture was then stirred for 1 hour at room temperature and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 30%
  • 1H NMR (400 MHz, DMSO d6) δ 9.19 (m, 1H), 9.05 (m, 1H), 7.31 (m, 4H), 6.73 (d, 1H), 6.54 (m, 1H), 6.05 (d, 1H), 4.16 (m, 1H), 3.42 (br s, 2H), 3.17 (br m, 6H), 2.91 (m, 1H), 2.11 (m, 1H), 1.98 (m, 1H), 1.90 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=395.1 (M+H)+
  • Chiral HPLC purity: tR=9.932 min (ee=>99%)
  • [α]D 24.2=+21.49 (c. 0.01, MeOH)
  • Example 27S Preparation of 27S
  • 27S was obtained from 27Q by chiral HPLC chromatography 27Q (racemic mixture) (10 g, 23.20 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiralpak AD-H, 4.4×250 mm
  • Column Temperature: 25° C.
  • Detection: UV at 230 nm
  • Flow: 2.0 mL/minute
  • Mobile Phase: 80% carbon dioxide, 20% ethanol, 0.1% ethane sulfonic acid
  • Run Time: 24 min
  • The relevant fractions were combined and concentrated under reduced pressure. An aqueous 1N solution of sodium hydroxide was added to the resulting oil until the solution was basic using pH paper. The aqueous mixture was extracted with dichloromethane. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous methanol was added drop wise a 4M solution of anhydrous hydrochloric acid in dioxane (5.5 eq). The mixture was then stirred for 1 h at room temperature and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 18%
  • 1H NMR (400 MHz, DMSO d6) δ 9.03 (m, 1H), 8.87 (m, 1H), 8.80 (s, 1H), 7.31 (m, 4H), 6.71 (d, 1H), 6.55 (d, 1H), 6.05 (m, 1H), 4.18 (m, 1H), 3.36 (m, 2H), 3.18 (m, 5H), 2.93 (m, 1H), 2.11 (m, 1H), 1.98 (m, 1H), 1.87 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=395.1 (M+H)+
  • Chiral HPLC prity: tR=13.371 min (ee=98.1%)
  • [α]D 24.2=−25.96 (c. 0.01, MeOH)
  • Example 27T Preparation of 27.1
  • A solution of 11.6a (15.00 g, 27.95 mmol, 1 eq) in anhydrous methanol (100 mL) was hydrogenated at 70 psi in the presence of palladium hydroxide [Pd(OH)2: Pearlman's catalyst] (1.96 g, 1.40 mmol, 0.05 eq) for 10 h. The mixture was filtered through celite. The filtrate was concentrated under reduced pressure and was hydrogenated at 70 psi in the presence of palladium hydroxide (1.96 g) for an additional 10 h. The mixture was filtered through celite and the filtrate was concentrated to dryness under reduced pressure. The crude product was used without further purification. Yield: 84%
  • 1H NMR (400 MHz, DMSO d6) δ 7.23 (d, 2H), 7.11 (m, 3H), 6.60 (d, 1H), 6.52 (d, 1H), 4.85 (d, 1H), 4.74 (d, 1H), 4.16 (m, 1H), 3.61 (m, 2H), 3.30 (br m, 6H), 2.83 (s, 3H), 2.24 (m, 1H), 1.75 (m, 2H), 1.64 (m, 1H), 1.52 (m, 2H), 1.39 (s, 9H), 1.06 (m, 6H)
  • Mass Spectral Analysis m/z=539.5 (M+H)+
  • Preparation of 27T
  • To a cold (0° C.) solution of 27.1 (2.00 g, 3.71 mmol, 1.0 eq) in anhydrous methanol (40 mL) was added drop wise a 4M solution of anhydrous hydrochloric acid in dioxane (9.3 mL, 37.20 mmol, 10.0 eq). The mixture was then stirred for 10 h at room temperature and concentrated under reduced pressure. Diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether.
  • Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.30 (br s, 1H), 9.03 (br s, 1H), 8.96 (br s, 1H), 7.21 (d, 2H), 7.14 (d, 2H), 6.99 (t, 1H), 6.43 (d, 1H), 6.35 (d, 1H), 4.15 (m, 1H), 3.87 (br s, 3H), 3.39 (m, 2H), 3.15 (m, 5H), 2.90 (m, 1H), 2.25 (m, 1H), 1.83 (br m, 5H), 1.09 (m, 6H)
  • Mass Spectral Analysis m/z=395.3 (M+H)+
  • Example 27U Preparation of 27.4
  • Compound 27.1 (racemic mixture) (10 g, 18.56 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiralpak AD-H, 4.4×250 mm
  • Column temperature: 25° C.
  • Detection: UV at 280 nm
  • Flow: 2.0 mL/minute
  • Mobile phase: 75% carbon dioxide, 25% isopropanol
  • Run time: 10 minutes.
  • The relevant fractions were combined and concentrated under reduced pressure. The crude product was used without further purification. Yield: 79%
  • 1H NMR (400 MHz, DMSO d6) δ 7.21 (d, 2H), 7.11 (m, 3H), 6.60 (d, 1H), 6.55 (d, 1H), 4.83 (d, 1H), 4.74 (d, 1H), 4.16 (m, 1H), 3.62 (m, 2H), 3.15 (br m, 6H), 2.83 (s, 3H), 2.24 (m, 1H), 1.75 (m, 2H), 1.61 (m, 1H), 1.50 (m, 2H), 1.39 (s, 9H), 1.06 (m, 6H)
  • Mass Spectral Analysis m/z=539.1 (M+H)+
  • Chiral HPLC purity: tR=4.728 min (ee=>99%)
  • [α]D 24.1=−32.97 (c. 0.01, MeOH)
  • Preparation of 27U
  • To a cold (0° C.) solution of 27.4 (1.00 g, 1.86 mmol, 1 eq) in anhydrous methanol was added drop wise a 4M solution of anhydrous hydrochloric acid in dioxane (2.5 mL, 10.21 mmol, 5.5 eq). The mixture was stirred for 10 hours at room temperature and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 88%
  • 1H NMR (400 MHz, DMSO d6) δ 9.30 (s, 1H), 9.00 (m, 2H), 7.21 (d, 2H), 7.14 (d, 2H), 6.99 (t, 1H), 6.41 (d, 1H), 6.35 (d, 1H), 4.15 (m, 1H), 3.42 (br s, 5H), 3.12 (m, 2H), 2.90 (m, 1H), 2.24 (m, 1H), 1.83 (m, 4H), 1.72 (m, 1H), 1.09 (m, 6H)
  • Mass Spectral Analysis m/z=395.1 (M+H)+
  • [α]D 24.2=+3.24 (c. 0.01, MeOH)
  • Example 27V Preparation of 27.5
  • 27.1 (racemic mixture) (10 g, 18.56 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiralpak AD-H, 4.4×250 mm
  • Column temperature: 25° C.
  • Detection: UV at 280 nm
  • Flow: 2.0 mL/minute
  • Mobile phase: 75% carbon dioxide, 25% isopropanol
  • Run time: 10 minutes.
  • The relevant fractions were combined and concentrated under reduced pressure. The crude product was used without further purification.
  • Yield: 83%
  • 1H NMR (400 MHz, DMSO d6) δ 7.23 (d, 2H), 7.11 (m, 3H), 6.58 (d, 1H), 6.54 (d, 1H), 4.85 (d, 1H), 4.73 (d, 1H), 4.16 (m, 1H), 3.63 (m, 2H), 3.16 (br m, 6H), 2.83 (s, 3H), 2.24 (m, 1H), 1.75 (m, 2H), 1.61 (m, 1H), 1.52 (m, 2H), 1.39 (s, 9H), 1.05 (m, 6H)
  • Mass Spectral Analysis m/z=539.1 (M+H)+
  • Chiral HPLC Method: tR=5.943 min (ee=98.7%)
  • [α]D 24.0=+29.88 (c. 0.01, MeOH)
  • Preparation of 27V
  • To a cold (0° C.) solution of 27.5 (1.00 g, 1.86 mmol, 1 eq) in anhydrous methanol was added drop wise a 4M solution of anhydrous hydrochloric acid in dioxane (2.5 mL, 10.21 mmol, 5.5 eq). The mixture was then stirred for 10 h at room temperature and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity).
  • Yield: 92%
  • 1H NMR (400 MHz, DMSO d6) δ 9.32 (s, 1H), 9.09 (br s, 2H), 7.21 (d, 2H), 7.12 (d, 2H), 6.99 (t, 1H), 6.41 (d, 1H), 6.38 (d, 1H), 4.16 (m, 1H), 3.36 (m, 5H), 3.13 (br m, 2H), 2.90 (m, 1H), 2.24 (m, 1H), 1.81 (br m, 5H), 1.09 (m, 6H)
  • Mass Spectral Analysis m/z=395.1 (M+H)+
  • [α]D24.36.35 (c. 0.01, MeOH)
  • Example 27W
  • 27W was obtained according to a procedure similar to the one described for 27A, with the following exception:
  • Step 27.3: 1A was replaced by 1E.
  • 1H NMR (400 MHz, CDCl3) δ 7.34 (d, 2H), 7.18 (d, 2H), 6.96 (d, 1H), 6.78 (d, 1H), 6.54 (s, 1H), 4.06 (m, 1H), 3.72 (q, 1H), 3.55 (brm, 3H), 3.28 (brm, 3H), 3.17 (m, 1H), 3.03 (m, 1H), 2.14 (m, 5H), 1.97 (m, 2H), 1.49 (t, 1H), 1.20 (brd, 6H)
  • Mass Spectral Analysis m/z=393.4 (M+H)+
  • Example 28A Preparation of 28.2
  • To a solution of benzyl 4-oxopiperidine-1-carboxylate (19.1) (37.26 g, 160 mmol) in toluene (450 mL) were added ethyl cyanoacetate (28.1) (18.8 g, 166 mmol, 1.04 eq), acetic acid (2 mL) and ammonium acetate (1.24 g, 16 mmol, 0.1 eq). The reaction mixture was refluxed for 2 h with azeotropic removal of water formed during the reaction using a Dean-Stark trap. Additional ethyl cyanoacetate (10 g, 88.4 mmol, 0.55 eq), acetic acid (2 mL) and ammonium acetate (1.24 g, 6 mmol, 0.0375 eq) was added to the reaction mixture, which was then refluxed for 1.5 h. Additional ethyl cyanoacetate (10 g, 88.4 mmol, 0.55 eq), acetic acid (2 mL) and ammonium acetate (1.24 g, 6 mmol, 0.0375 eq) were added, and refluxed for an additional 1 h. The reaction mixture was cooled to room temperature and washed with a saturated aqueous solution of sodium bicarbonate, and dried over sodium sulfate. The mixture was filtered and the filtrate was concentrated under vacuum. To the residue was added hexane (300 mL) and ethyl acetate (20 mL). The mixture was kept at room temperature overnight. The solid was collected by filtration, washed with hexane and dried under vacuum. Yield: 87.7%
  • 1H NMR (400 MHz, CDCl3) δ 7.35 (m, 5H), 5.19 (s, 2H), 4.30 (q, 2H), 3.70 (m, 2H), 3.63 (m, 2H), 3.18 (m, 2H), 2.80 (m, 2H), 1.39 (t, 3H)
  • Preparation of 28.4a
  • To a suspension of copper (I) cyanide (17.3 g, 193.2 mmol, 2.0 eq) in anhydrous tetrahydrofuran (400 mL) was added drop wise a 2.0 M solution of benzylmagnesium chloride (28.3a) (192 mL, 384 mmol, 4.0 eq) in tetrahydrofuran under a nitrogen atmosphere at 0° C. After the reaction mixture was stirred at room temperature for 2 h, a solution of compound 28.2 (31.5 g, 96 mmol) in tetrahydrofuran (100 mL) was added dropwise at −30° C. After the addition, the reaction mixture was stirred at room temperature overnight, and then quenched with a saturated aqueous solution of ammonium chloride, and filtered. The filtrate was extracted by diethyl ether and the combined organic extracts were dried over sodium sulfate. The organics were concentrated under reduced pressure and the residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 4:1:1).
  • Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.35-7.20 (m, 10H), 5.11 (s, 2H), 4.25 (q, 2H), 3.72-3.50 (m, 5H), 3.06 (d, 1H), 2.91 (d, 1H), 1.90-1.65 (m, 4H), 1.32 (t, 3H)
  • Preparation of 28.6a
  • Concentrated sulfuric acid (210 mL) was added slowly to 28.4a (38 g, 90.5 mmol) at 0° C. The mixture was warmed to room temperature, stirred for 30 min at room temperature, and then heated at 90° C. overnight. The reaction mixture was cooled in an ice bath and carefully basified to pH=9-10 with a 6 N aqueous solution of sodium hydroxide. The mixture was extracted with methylene chloride, and the organic extracts were combined, dried over sodium sulfate and concentrated under vacuum. The residue was dissolved in methylene chloride (500 mL). To this solution was added triethylamine (30 mL, 215.6 mmol, 2.4 eq) followed by drop wise addition of benzyl chloroformate (21.8) (16 mL, 106.5 mmol, 1.2 eq) at 0° C. The reaction mixture was stirred at 0° C. for 1 h and then washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was dried over sodium sulfate and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 4:1:1). Yield: 41.2%
  • 1H NMR (400 MHz, CDCl3) δ 8.00 (d, 1H), 7.50 (t, 1H), 7.33-7.23 (m, 7H), 5.11 (s, 2H), 2.98 (s, 2H), 2.62 (s, 2H), 1.50 (m, 4H)
  • Preparation of 28.7a
  • A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (3.6 mL, 3.6 mmol, 1.2 eq) was added at −78° C. to a solution of 28.6a (1.047 g, 3.0 mmol) in tetrahydrofuran (30 mL). After 45 min, a solution of 1.4 (1.3 g, 3.6 mmol, 1.2 eq) in tetrahydrofuran (8 mL) was added drop wise to the reaction mixture. The reaction mixture was then warmed to room temperature and stirred for 2.5 h, quenched by addition of water (40 mL), and extracted with a mixture of hexane and diethyl ether (1:1). The organic extracts were combined and washed with water, brine and dried over sodium sulfate. Evaporation of the solvent gave the crude product, which was used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.35-7.18 (m, 9H), 5.98 (s, 1H), 5.11 (s, 2H), 3.70 (m, 2H), 3.40 (m, 2H), 2.83 (s, 2H), 1.66-1.56 (m, 4H)
  • Preparation of 28.8a
  • To the solution of crude 28.7a (3 mmol) in dimethoxyethane (25 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (5 mL, 10 mmol, 3.3 eq), lithium chloride (424 mg, 10 mmol, 3.3 eq), 4-(N,N-diethylaminocarbonyl)phenylboronic acid (796 mg, 3.6 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (104 mg, 0.09 mmol, 0.03 eq). The reaction mixture was refluxed overnight, cooled to room temperature, diluted with water (30 mL) and extracted with diethyl ether. The combined organic extracts were dried over sodium sulfate and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 2:1:1).
  • Yield: 91.9%
  • 1H NMR (400 MHz, CDCl3) δ 7.36-7.12 (m, 12H), 7.00 (d, 1H), 6.00 (s, 1H), 5.13 (s, 2H), 3.70 (m, 2H), 3.58 (m, 2H), 3.45 (m, 2H), 3.30 (m, 2H), 2.82 (s, 2H), 1.65-1.52 (m, 4H), 1.21 (m, 6H)
  • Preparation of 28A
  • Iodotrimethylsilane (0.29 mL, 2 mmol, 2 eq) was added to a solution of 28.8a (508 mg, 1 mmol) in anhydrous methylene chloride (10 mL) under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h and quenched with a 1N aqueous solution of hydrochloric acid (30 mL) and extracted with diethyl ether. The aqueous phase was basified to pH=9-10 with a 3N aqueous solution of sodium hydroxide, and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate and concentrated under vacuum. The residue was dissolved in methylene chloride (3 mL) and diluted with diethyl ether (15 mL). To this solution was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (1.5 mL, 3 mmol, 3.0 eq) and the reaction was stirred at room temperature for 30 min. The solid was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 92.7%
  • 1H NMR (400 MHz, CDCl3) δ 8.90 (m, 2H), 7.40-7.20 (m, 7H), 6.97 (d, 1H), 6.20 (s, 1H), 3.42 (m, 2H), 3.20 (m, 6H), 2.82 (s, 2H), 1.70 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=375.1 (M+H)+
  • Example 28B Preparation of 28.4b
  • Compound 28.4b was prepared as described for 28.4a except 28.3a was replaced by 23.8b.
  • Preparation of 28.9
  • To a solution of compound 28.4b (29 g, 64.4 mmol) in dimethylsulfoxide (200 mL) was added sodium chloride (1.5 g, 25.6 mmol, 0.4 eq) and water (3.0 mL, 167 mmol, 2.6 eq). The reaction mixture was heated at 160° C. for 2 h and then cooled to room temperature. Water (600 mL) was added to the mixture and the crude product was extracted with diethyl ether. The organic extracts were combined, washed with water and brine, dried over sodium sulfate, and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 4:1:1). Yield: 94.8%
  • 1H NMR (400 MHz, CDCl3) δ 7.35 (m, 5H), 7.08 (d, 2H), 6.83 (d, 2H), 5.12 (s, 2H), 3.80 (s, 3H), 3.68 (m, 2H), 3.40 (m, 2H), 2.74 (s, 2H), 2.21 (s, 2H), 1.60-1.52 (m, 4H)
  • Preparation of 28.10
  • To a solution of compound 28.9 (7.56 g, 20 mmol) in methanol (200 mL) was added concentrated sulfuric acid (40 mL). The mixture was heated at reflux for 2 days. The reaction mixture was cooled to 0° C., basified to pH=9 by slow addition of a 6 N aqueous solution of sodium hydroxide, and then concentrated under vacuum to remove the methanol. The mixture was extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under vacuum. The residue was dissolved in methylene chloride (80 mL) and cooled to 0° C. To this solution was added triethylamine (9.6 mL, 69 mmol, 3.5 eq) and followed by drop wise addition of benzyl chloroformate (21.8) (6.4 mL, 95%, 42.7 mmol, 2.1 eq). The reaction mixture was stirred at 0° C. for 1 h, washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 4:1:1). Yield: 94.8%
  • 1H NMR (400 MHz, CDCl3) δ 7.38 (m, 5H), 7.10 (d, 2H), 6.80 (d, 2H), 5.12 (s, 2H), 3.80 (s, 3H), 3.75 (m, 2H), 3.70 (s, 3H), 3.32 (m, 2H), 2.73 (s, 2H), 2.30 (s, 2H), 1.50 (m, 4H)
  • Preparation of 28.11
  • Compound 28.10 (2.06 g, 5 mmol) was dissolved in mixture of methanol (40 mL), tetrahydrofuran (40 mL) and water (40 mL). To this solution was added lithium hydroxide (1.52 g, 36 mmol, 7.2 eq) in one portion. The reaction mixture was stirred at room temperature overnight, concentrated under vacuum, acidified with a 3 N aqueous solution of hydrochloric acid and extracted with methylene chloride. The combined organic extracts were dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was used fro the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ12.22 (brs, 1H), 7.33 (m, 5H), 7.10 (d, 2H), 6.86 (d, 2H), 5.06 (s, 2H), 3.73 (s, 3H), 3.60 (m, 2H), 3.32 (m, 2H), 2.69 (s, 2H), 2.17 (s, 2H), 1.45-1.35 (m, 4H)
  • Preparation of 28.6b
  • To a solution of 28.11 (1.98 g, 5 mmol) in anhydrous methylene chloride (10 mL) was added a 2.0 M solution of oxalyl chloride in methylene chloride (20 mL, 40 mmol, 8.0 eq) followed by 2 drops of anhydrous N,N-dimethylformamide. The reaction mixture was stirred at room temperature for 4 h and then concentrated under vacuum. The resulting acyl chloride was dissolved in anhydrous methylene chloride (100 mL) and aluminum chloride (1.35 g, 10 mmol, 2.0 eq) was added in one portion. The reaction mixture was stirred at room temperature overnight and then quenched with water (60 mL) followed by addition of concentrated ammonium hydroxide to basify the aqueous layer. The organic layer was separated and the aqueous layer was further extracted with methylene chloride. The combined organic extracts were dried over sodium sulfate, filtered and concentrated under vacuum. The residue was then dissolved in methylene chloride (60 mL) and cooled to 0° C. To this solution was added triethylamine (3.0 mL, 21.6 mmol, 4.3 eq) followed by benzyl chloroformate (21.8) (2.0 mL, 13.3 mmol, 2.7 eq). The reaction mixture was stirred at 0° C. for 1 h and then washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 4:1:1).
  • Yield: 89.7%
  • 1H NMR (400 MHz, CDCl3) δ 7.48 (d, 1H), 7.35 (m, 5H), 7.16 (d, 1H), 7.10 (dd, 1H), 5.11 (s, 2H), 3.81 (s, 3H), 3.50 (m, 4H), 2.90 (s, 2H), 2.60 (s, 2H), 1.50 (m, 4H)
  • Preparation of 28B
  • 28B was obtained from 28.6b according to a procedure similar to the one described for 28A.
  • 1H NMR (DMSO d6) δ 8.90 (m, 2H), 7.48 (d, 2H), 7.40 (d, 2H), 7.26 (d, 1H), 6.85 (dd, 1H), 6.45 (d, 1H), 6.20 (s, 1H), 3.64 (s, 3H), 3.42 (m, 4H), 3.18 (m, 4H), 2.78 (s, 2H), 1.70 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=405.1 (M+H)+
  • Example 28C Preparation of 28C
  • Compound 28.8a (800 mg, 1.58 mmol) was dissolved in a mixture of methylene chloride (5 mL) and methanol (50 mL), and the reaction mixture was hydrogenated in the presence of 10% Pd/C (240 mg) using a hydrogen balloon. After 2 days at room temperature, the reaction mixture was filtered through celite and the filtrate was concentrated under vacuum. The residue was dissolved in methylene chloride (10 ml) and added 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (2 mL, 4 mmol, 2.5 eq). The mixture was stirred for 1 h at room temperature and then concentrated under vacuum. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (brs, 2H), 7.28-7.03 (m, 7H), 6.66 (d, 1H), 4.10 (m, 1H), 3.40 (m, 2H), 3.20-3.08 (m, 6H), 2.85 (d, 1H), 2.78 (d, 1H), 2.10 (m, 1H), 1.60 (m, 5H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=377.1 (M+H)+
  • Example 28D
  • 28D was obtained according to a procedure similar to the one described for 28C, with the following exception:
  • Step 28.12: 28.8a was replaced by 28.8b.
  • 1H NMR (400 MHz, DMSO d6) δ 8.77 (m, 2H), 7.28 (m, 4H), 7.89 (d, 1H), 6.75 (dd, 1H), 6.16 (d, 1H), 4.09 (m, 1H), 3.55 (s, 3H), 3.49-3.00 (m, 8H), 2.73 (m, 2H), 2.10 (m, 1H), 1.59 (m, 5H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=407.3 (M+H)+
  • Example 28E
  • 28E was obtained according to a procedure similar to the one described for 28A, with the following exception:
  • Step 28.10:1.6 was replaced by 1.7 (see also step 28.13).
  • 1H NMR (400 MHz, DMSO d6) δ 8.91 (m, 2H), 8.61 (s, 1H), 7.89 (d, 1H), 760 (d, 1H), 7.31-7.20 (m, 3H), 6.90 (d, 1H), 6.33 (s, 1H), 3.45-3.15 (m, 8H), 2.83 (s, 2H), 1.70 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=376.4 (M+H)+
  • Elemental analysis:
  • C24H29N30, 4/3HCl, 1H2O
  • Theory: % C, 65.20; % H, 7.37; % N, 9.50; % C, 10.69.
  • Found: % C, 64.94; % H, 7.06; % N, 9.36; % Cl, 10.56.
  • Example 29A Preparation of 29.2
  • To a solution of crude compound 28.7a (12 mmol) in anhydrous tetrahydrofuran (200 mL) at room temperature was added a 0.5 M solution of 4-(ethoxycarbonyl)phenylzinc iodide (29.1) in tetrahydrofuran (60 mL, 30 mmol, 2.5 eq) followed by tetrakis(triphenylphosphine)palladium(0) (833 mg, 0.72 mmol, 0.06 eq). The reaction mixture was heated at 40° C. for 2 days and then cooled to room temperature. The reaction was quenched by addition of a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic extracts were combined, dried over sodium sulfate and filtered. The organic extracts were concentrated under reduced pressure and the residue was purified by column chromatography (eluent: hexane/ethyl acetate, 5:1). Yield: 86.6%
  • 1H NMR (400 MHz, CDCl3) δ 8.05 (d, 2H), 7.40-7.10 (m, 10H), 6.96 (d, 1H), 6.00 (s, 1H), 5.13 (s, 2H), 4.40 (q, 2H), 3.70 (m, 2H), 3.48 (m, 2H), 2.82 (s, 2H), 1.66-1.53 (m, 6H), 1.40 (t, 3H)
  • Preparation of 29.3
  • Lithium hydroxide (3.36 g, 80 mmol, 8.0 eq) was added to a solution of 29.2 (4.81 g, 10 mmol) in a mixture of methanol (100 mL), tetrahydrofuran (100 mL) and water (100 mL). The reaction mixture was stirred at room temperature overnight, concentrated under vacuum and acidified to pH=1-2 with a 3N aqueous solution of hydrochloric acid. The acidified solution was extracted with methylene chloride and the organic extracts were combined, dried over sodium sulfate, filtered and concentrated under vacuum. The crude product was used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 13.00 (brs, 1H), 7.99 (d, 2H), 7.48 (d, 2H), 7.38-7.15 (m, 8H), 6.91 (d, 1H), 6.18 (s, 1H), 5.10 (s, 2H), 3.60-3.46 (m, 4H), 2.82 (s, 2H), 1.53 (m, 2H), 1.42 (m, 2H)
  • Preparation of 29.5a
  • To a solution of 29.3 (680 mg, 1.5 mmol, 1.0 eq)) in methylene chloride (40 mL) was added isopropylamine (3.4h) (0.26 mL, 3 mmol, 2.0 eq) followed by triethylamine (0.84 ml, 6 mmol, 4.0 eq) and the Mukaiyama acylating reagent (2-chloro-1-methylpyridinium iodide) (461 mg, 1.8 mmol, 1.2 eq). The reaction mixture was stirred at room temperature overnight, washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate, and filtered. The organic extracts were concentrated under reduced pressure and the residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 2:1:1).
  • Yield: 95.8%
  • 1H NMR (400 MHz, CDCl3) δ 7.78 (d, 2H), 7.40-7.10 (m, 10H), 6.94 (d, 1H), 6.00 (s, 1H), 5.95 (d, 1H), 5.12 (s, 2H), 4.31 (m, 1H), 3.70 (m, 2H), 3.46 (m, 2H), 2.81 (s, 2H), 1.62-1.52 (m, 6H), 1.30 (d, 6H)
  • Preparation of 29A
  • Iodotrimethylsilane (0.37 mL, 2.6 mmol, 2.0 eq) was added to a solution 29.5 (620 mg, 1.26 mmol) in anhydrous methylene chloride (20 mL) under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h, quenched with a 1N aqueous solution of hydrochloric acid (40 mL), and the mixture was extracted with diethyl ether. The aqueous phase was basified to pH=9-10 with a 3N aqueous solution of sodium hydroxide and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under vacuum. The residue was dissolved in methylene chloride (4 mL) and diluted with diethyl ether (20 mL). To this solution was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (2.0 mL, 4 mmol, 3.2 eq) and the mixture was stirred at room temperature for 30 min. The resulting precipitate was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 8.90 (brd, 2H), 8.29 (d, 1H), 7.90 (d, 2H), 7.43 (d, 2H), 7.31-7.16 (m, 3H), 6.90 (d, 1H), 6.18 (s, 1H), 4.11 (m, 1H), 3.16 (m, 4H), 2.86 (s, 2H), 1.70 (m, 4H), 1.20 (d, 6H)
  • Mass Spectral Analysis m/z=361.0 (M+H)+
  • Example 29B
  • 29B was obtained according to a procedure similar to the one described for 29A, with the following exception:
  • Step 29.3: 3.4 h was replaced by 29.4.
  • 1H NMR (400 MHz, DMSO d6) δ 8.89 (m, 2H), 8.10 (d, 1H), 7.92 (d, 2H), 7.45 (d, 2H), 7.31 (d, 1H), 7.25 (t, 1H), 7.20 (t, 1H), 6.90 (d, 1H), 6.18 (s, 1H), 3.80 (m, 1H), 3.20 (m, 4H), 2.88 (s, 2H), 1.60 (m, 8H), 0.90 (t, 6H)
  • Mass Spectral Analysis m/z=389.1 (M+H)+
  • Example 29C Preparation of 29.7
  • To a solution of the carboxylic acid 29.3 (1.82 g, 4 mmol) in a mixture of dioxane (18 mL) and tert-butyl alcohol (18 mL) was added triethylamine (0.78 mL, 5.6 mmol, 1.4 eq) and diphenylphosphoryl azide (29.6) (1.12 mL, 5.2 mmol, 1.3 eq). The reaction mixture was refluxed overnight and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 5:1:1) to afford the desired crude carbamate 29.7, which was used for the next step without further purification. Yield: 33.4%
  • Preparation of 29.8
  • To a solution of the crude carbamate 29.7 (700 mg) in methylene chloride (15 mL) was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (15 mL, 30 mmol). The reaction mixture was stirred at room temperature overnight and then diethyl ether was added to the reaction mixture, which was stirred for an additional 2 h at room temperature. The resulting precipitate was collected by filtration and used for the next step without further purification. Yield: 57%
  • 1H NMR (400 MHz, DMSO d6) δ 10.15 (brs, 3H), 7.40-7.15 (12H), 6.89 (d, 1H), 6.10 (s, 1H), 5.10 (s, 2H), 3.59 (m, 2H), 3.46 (m, 2H), 2.81 (s, 2H), 1.54 (m, 2H), 1.41 (m, 2H)
  • Preparation of 29.10
  • Triethylamine (0.42 mL, 3 mmol) was added to a suspension of 29.8 (300 mg, 0.65 mmol) in methylene chloride (20 mL) at 0° C. followed by drop wise addition of propionyl chloride (29.9) (0.12 mL, 1.3 mmol, 2.0 eq). The reaction mixture was stirred at room temperature for 6 h and washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 2:1:1). Yield: 89.5%
  • 1H NMR (400 MHz, CDCl3) δ 7.54 (d, 2H), 7.38-7.10 (m, 11H), 7.00 (d, 1H), 5.95 (s, 1H), 5.12 (s, 2H), 3.70 (m, 2H), 3.44 (m, 2H), 2.80 (s, 2H), 2.42 (q, 2H), 1.60 (m, 2H), 1.50 (m, 2H), 1.28 (t, 3H)
  • Preparation of 29C
  • Iodotrimethylsilane (0.21 mL, 1.47 mmol, 2.0 eq) was added to a solution of compound 29.10 (220 mg, 0.46 mmol) in anhydrous methylene chloride (8 mL) under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h and quenched with a 1 N aqueous solution of hydrochloric acid (15 mL). The crude product was extracted with diethyl ether. The aqueous layer was basified to pH=9-10 with a 3M aqueous solution of sodium hydroxide and the mixture was extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate and concentrated under vacuum. The residue was dissolved in methylene chloride (3 mL) and diluted with diethyl ether (10 mL). To this solution was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (0.7 mL, 1.4 mmol, 3.0 eq) and the mixture was stirred at room temperature for 30 min. The solid was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 83.9%
  • 1H NMR (400 MHz, DMSO d6) δ 10.05 (s, 1H), 8.94 (brd, 2H), 7.66 (d, 2H), 7.30-7.20 (m, 5H), 6.96 (d, 1H), 6.08 (s, 1H), 3.15 (m, 4H), 2.82 (s, 2H), 2.34 (q, 2H), 1.68 (m, 4H), 1.10 (t, 3H)
  • Mass Spectral Analysis m/z=347.0 (M+H)+
  • Example 29D Preparation of 29.11
  • Methanesulfonyl chloride (7.4) (0.051 mL, 0.66 mmol, 2.0 eq) was added to a solution of 29.8 (150 mg, 0.326 mmol) in pyridine (6 mL) at 0° C. The reaction mixture was stirred at room temperature overnight, diluted with methylene chloride (40 mL) and washed with a 1N aqueous solution of hydrochloric acid and brine. The organic layer was dried over sodium sulfate and concentrated under vacuum. The residue was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1).
  • Yield: 97.7%
  • 1H NMR (400 MHz, CDCl3) δ 7.38-7.13 (m, 12H), 6.99 (d, 1H), 6.50 (s, 1H), 5.96 (s, 1H), 5.12 (s, 2H), 3.70 (m, 2H), 3.46 (m, 2H), 3.08 (s, 3H), 2.81 (s, 2H), 1.62-1.52 (m, 4H)
  • Preparation of 29D
  • Iodotrimethylsilane (0.14 mL, 0.98 mmol, 3.5 eq) was added to a solution of 29.11 (140 mg, 0.28 mmol) in anhydrous methylene chloride (6 mL) under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h and quenched with a 1N aqueous solution of hydrochloric acid (10 mL). The crude product was extracted with diethyl ether. The aqueous layer was basified to pH=9-10 with a 3N aqueous solution of sodium hydroxide and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under vacuum. The residue was dissolved in methylene chloride (3 mL) and diluted with diethyl ether (10 mL). To this solution was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (0.42 mL, 0.84 mmol, 3.0 eq) and the mixture was stirred at room temperature for 30 min. The solid was collected by filtration, washed with diethyl ether and dried under vacuum. Yield: 90.5%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.88 (s, 1H), 8.91 (brd, 2H), 7.35-7.18 (m, 7H), 6.96 (d, 1H), 6.09 (s, 1H), 3.12 (m, 4H), 3.02 (s, 3H), 2.82 (s, 2H), 1.68 (m, 4H)
  • Mass Spectral Analysis m/z=368.9 (M+H)+
  • Example 30A Preparation of 30.3
  • A mixture of 30.1 (10.2 g, 0.050 mol, 1.0 eq) and 30.2 (25 g, 0.075 mol, 1.5 eq) in toluene (100 mL) under nitrogen was refluxed for 2 h. The mixture was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1). Yield: 92%
  • 1H NMR (400 MHz, CDCl3) δ 7.42 (s, 5H), 5.78 (brs, 1H), 3.83 (brs, 2H), 3.70 (s, 3H), 3.49 (brs, 2H), 3.02 (brm, 2H), 2.37 (brm, 2H)
  • Mass Spectral Analysis m/z=259.9 (M+H)+
  • Preparation of 30.5
  • A solution of 30.3 (5.0 g, 19.3 mmol, 1.0 eq), 30.4 (16.39 g, 149 mmol, 7.7 eq), and triethylamine (3.90 g, 38.6 mmol, 2.0 eq) in tetrahydrofuran (100 mL) was refluxed for 12 h. The mixture was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 60:40). Yield: 98%
  • 1H NMR (400 MHz, CDCl3) δ 7.56 (m, 2H), 7.37 (m, 8H), 4.40 (brs, 1H), 3.72 (s, 3H), 3.58 (brm, 3H), 2.56 (s, 2H), 1.76 (brm, 4H)
  • Mass Spectral Analysis m/z=369.9 (M+H)+
  • Preparation of 30.6
  • A solution of 30.5 (10.0 g, 27.07 mmol, 1.0 eq) and concentrated sulfuric acid (50 mL) was stirred at room temperature for 18 h. The mixture was poured onto ice water (1:1) (200 mL) and the crude product was extracted with ethyl acetate. The combined organic extracts were dried over magnesium sulfate, concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 70:30). Yield: 22%
  • 1H NMR (400 MHz, CDCl3) δ 8.08 (dd, 1H), 7.40 (m, 7H), 7.20 (m, 1H), 4.47 (brs, 1H), 3.44 (brm, 3H), 2.97 (brd, 2H), 1.92 (brm, 4H)
  • Mass Spectral Analysis m/z=337.9 (M+H)+
  • Preparation of 30.7
  • To a solution of 30.6 (1.2 g, 3.56 mmol, 1.0 eq) in acetic acid (5 mL) was added at room temperature a 30% aqueous solution of hydrogen peroxide (2 mL). The solution was heated at 90° C. for 2 h and then cooled to room temperature. The mixture was concentrated to ⅓ of its volume under reduced pressure. Water was added and the crude product was extracted with methylene chloride. The combined organic extracts were then washed with a saturated sodium thiosulfate solution, brine, dried over magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1). Yield: 84%
  • 1H NMR (400 MHz, CDCl3) δ 8.10 (m, 2H), 7.87 (m, 1H), 7.77 (m, 1H), 7.41 (m, 5H), 4.34 (brs, 1H), 3.90 (brm, 1H), 3.50 (brm, 4H), 2.36 (brs, 2H), 1.80 (brm, 2H)
  • Mass Spectral Analysis m/z=369.8 (M+H)+
  • Preparation of 30.8
  • A mixture of 30.7 (1.1 g, 2.98 mmol, 1.0 eq) and a 6N aqueous solution of hydrochloric acid (5 mL) in ethanol (20 mL) was heated at 90° C. for 12 h. The mixture was concentrated under reduced pressure and used for the next step without further purification. Yield: 100%; Mass Spectral Analysis m/z=265.8 (M+H)+
  • Preparation of 30.9
  • To a solution of 30.8 (0.9 g, 2.98 mmol, 1.0 eq) in tetrahydrofuran (10 mL) at 0° C. was added triethylamine (1.2 g, 11.92 mmol, 4.0 eq) and 4.7 (0.78 g, 3.58 mmol, 1.2 eq). The mixture was stirred at 0° C. for 1 h and at room temperature for 1 h. Water (20 mL) was added and the crude mixture was extracted with ethyl acetate. The combined organics were washed with water, brine, dried over magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: eluent: hexane/ethyl acetate, 1:1).
  • Yield: 79%
  • 1H NMR (400 MHz, CDCl3) δ 8.09 (m, 2H), 7.86 (m, 1H), 7.76 (m, 1H), 3.97 (brs, 2H), 3.39 (s, 2H), 3.20 (brm, 2H), 2.29 (m, 2H), 1.76 (brm, 2H), 1.46 (s, 9H)
  • Preparation of 30.10
  • To a solution of 30.9 (0.84 g, 2.30 mmol, 1.0 eq) in tetrahydrofuran (10 mL) at −78° C. under a nitrogen atmosphere was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (2.76 mL, 2.76 mmol, 1.2 eq). The mixture was stirred for 45 min at −78° C. A solution of 1.4 (0.986 g, 2.76 mmol, 1.2 eq) in tetrahydrofuran (3 mL) was added drop wise to the reaction mixture. The mixture was stirred for 3 h at 0° C. and at room temperature for 16 h. The mixture was poured into ice water (20 mL) and the crude product was extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over magnesium sulfate and filtered. The crude product was purified by column chromatography (eluent: 85/15 hexane/ethyl acetate mixture). Yield: 52%
  • 1H NMR (400 MHz, CDCl3) δ 8.09 (dd, 1H), 7.76 (m, 1H), 7.69 (m, 1H), 7.61 (d, 1H), 6.36 (s, 1H), 4.17 (brs, 2H), 3.06 (brs, 2H), 2.24 (m, 2H), 1.82 (m, 2H), 1.47 (s, 9H)
  • Preparation of 30.11
  • To a solution of 30.10 (0.15 g, 0.30 mmol, 1.0 eq) in dimethoxyethane (DME) (30 mL) was added sequentially a 2N aqueous solution of sodium carbonate (0.45 mL, 0.90 mmol, 3.0 eq), lithium chloride (0.038 g, 0.90 mmol, 3.0 eq), 1.6 (0.106 g, 0.33 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.007 g, 0.006 mmol, 0.02 eq). The mixture was refluxed for 16 h under a nitrogen atmosphere. The mixture was then cooled to room temperature and ice water (20 mL) was added. The mixture was extracted with ethyl acetate. The combined organic extracts were further washed with water, brine, dried over magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 70:30). Yield: 86%
  • 1H NMR (400 MHz, CDCl3) δ 8.09 (m, 1H), 7.56 (m, 2H), 7.44 (d, 2H), 7.38 (d, 2H), 7.15 (m, 1H), 6.22 (s, 1H), 4.16 (brs, 2H), 3.58 (brs, 2H), 3.30 (brs, 2H), 3.14 (brs, 2H), 2.23 (m, 2H), 1.88 (m, 2H), 1.47 (s, 9H), 1.23 (brd, 6H)
  • Mass Spectral Analysis m/z=525.9 (M+H)+
  • Preparation of 30A
  • To a solution of 30.11 (0.440 g, 0.84 mmol, 1.0 eq) in anhydrous methylene chloride (20 mL) was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (8.0 mL, 16 mmol, 19 eq). The mixture was stirred for 48 h at room temperature. The mixture was concentrated under reduced pressure and treated with diethyl ether. The resulting precipitate was collected by filtration. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 9.37 (brm, 1H), 8.80 (brm, 1H), 8.05 (d, 1H), 7.73 (m, 2H), 7.53 (d, 2H), 7.44 (d, 2H), 7.21 (d, 1H), 6.58 (s, 1H), 3.36 (brm, 8H), 2.26 (brm, 2H), 1.95 (brd, 2H), 1.13 (brd, 6H)
  • Mass Spectral Analysis m/z=425.3 (M+H)+
  • Example 31A Preparation of 13.2a
  • To a solution of 1.5a (7.80 g, 17.35 mmol, 1.0 eq) in dimethoxyethane (75 mL) was added sequentially a 2N aqueous solution of sodium carbonate (26.03 mL, 52.06 mmol, 3.0 eq), lithium chloride (2.21 g, 52.06 mmol, 3.0 eq), 13.1 (3.44 g, 19.09 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.40 g, 0.35 mmol, 0.02 eq). The mixture was refluxed overnight under nitrogen. The mixture was then cooled to room temperature and water (250 mL) was added. The mixture was extracted with ethyl acetate. The organic layer was further washed with brine and dried over sodium sulfate. The mixture was filtered and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 64%
  • 1H NMR (400 MHz, DMSO d6) δ 8.02 (d, 2H), 7.49 (d, 2H), 7.23 (m, 1H), 6.99 (d, 1H), 6.92 (m, 2H), 5.92 (s, 1H), 3.88 (s, 3H), 3.70 (m, 2H), 3.27 (m, 2H), 1.89 (m, 2H), 1.71 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=436.0 (M+H)+
  • Preparation of 31A
  • 31A was obtained according to a procedure similar to the one described for 1A, with the following exceptions:
  • Step 1.4: method 1E was used; 1.8a was replaced by 13.2a (see also step 31.2).
  • 1H NMR (DMSO d6) δ 8.81 (m, 2H), 8.00 (m, 2H), 7.45 (m, 2H), 7.24 (m, 1H), 7.03 (m, 1H), 6.91 (m, 2H), 5.99 (s, 1H), 3.90 (s, 3H), 3.22 (m, 4H), 2.06 (m, 2H), 1.98 (m,
  • 2H),
  • Mass Spectral Analysis m/z=336.0 (M+H)+
  • Elemental analysis:
  • C21H21NO3, 1HCl, 0.2H2O
  • Theory: % C, 67.18; % H, 6.01; % N, 3.73.
  • Found: % C, 67.32; % H, 5.98; % N, 3.77.
  • Example 31B
  • 31B was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 14.1.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (m, 2H), 7.94 (d, 2H), 7.59 (d, 2H), 7.29 (m, 1H), 7.06 (m, 1H), 6.94 (m, 2H), 6.02 (s, 1H), 3.22 (m, 4H), 2.05 (m, 4H)
  • Mass Spectral Analysis m/z=303.1 (M+H)+
  • Elemental analysis:
  • C20H18N2O, 1HCl, 0.8H2O
  • Theory: % C, 68.00; % H, 5.88; % N, 7.93.
  • Found: % C, 67.89; % H, 5.59; % N, 7.79.
  • Example 31C
  • 31C was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 16.1.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (brs, 1H), 7.90 (s, 2H), 7.65 (m, 2H), 7.25 (t, 1H), 7.10 (d, 1H), 6.00 (s, 1H), 3.20 (m, 4H), 2.00 (m, 4H)
  • Mass Spectral Analysis m/z=303.1 (M+H)+
  • Example 31D
  • 31D was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1a.
  • Step 31.2: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.18 (m, 2H), 7.51 (m, 1H), 7.41 (m, 2H), 7.26 (m, 2H), 7.05 (m, 1H), 6.94 (m, 2H), 5.92 (s, 1H), 3.46 (m, 2H), 3.20 (m, 6H), 2.06 (m, 4H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=377.4 (M+H)+
  • Example 31E
  • 31E was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1b.
  • Step 31.2: Method 1F was used.
  • 1H NMR (DMSO d6) δ 8.95 (m, 2H), 8.00 (d, 2H), 7.65 (d, 2H), 7.25 (m, 1H), 7.05 (m, 2H), 6.95 (m, 1H), 6.00 (s, 1H), 3.30 (s, 3H), 3.20 (m, 4H), 2.10 (m, 4H); Mass Spectral Analysis m/z=356.1 (M+H)+ tR=1.54 minutes.
  • Mass Spectral Analysis m/z=356.1 (M+H)+
  • Example 31F
  • 31F was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1c.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.60 (m, 2H), 7.41 (m, 4H), 7.26 (m, 1H), 7.03 (m, 1H), 6.95 (m, 2H), 5.89 (s, 1H), 4.11 (s, 2H), 3.23 (m, 4H), 2.09 (m, 2H), 1.94 (m, 2H)
  • Mass Spectral Analysis m/z=317.0 (M+H)+
  • Example 31G
  • 31G was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1d.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.16 (brs, 2H), 7.30 (d, 2H), 7.24 (m, 1H), 7.02 (m, 4H), 6.93 (m, 1H), 5.80 (s, 1H), 3.80 (s, 3H), 3.20 (brm, 4H), 2.03 (brm, 4H)
  • Mass Spectral Analysis m/z=308.0 (M+H)+
  • Example 31H
  • 31H was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1e.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (m, 2H), 7.26 (m, 5H), 6.98 (m, 3H), 5.82 (s, 1H), 3.21 (m, 4H), 2.35 (s, 3H), 2.03 (m, 4H)
  • Mass Spectral Analysis m/z=292.1 (M+H)+
  • Example 31I
  • 31I was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1f.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, CDCl3) δ 9.76 (m, 1H), 9.29 (m, 1H), 7.69 (m, 1H), 7.46 (m, 1H), 7.27 (brm, 4H), 6.96 (m, 2H), 5.64 (m, 1H), 3.44 (m, 2H), 3.30 (m, 2H), 2.29 (m, 2H), 2.11 (m, 2H)
  • Mass Spectral Analysis m/z=346.1 (M+H)+
  • Example 31J
  • 31J was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1 g.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (brs, 1.5H), 7.44 (m, 3H), 7.36 (m, 2H), 7.25 (m, 1H), 7.04 (d, 1H), 6.95 (m, 2H), 5.87 (s, 1H), 3.22 (brm, 4H), 2.09 (brm, 2H), 1.97 (brm, 2H)
  • Mass Spectral Analysis m/z=278.1 (M+H)+
  • Example 31K
  • 31K was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1h.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.66 (brs, 1H), 8.96 (brs, 2H), 7.50 (brm, 1H), 7.18 (brm, 3H), 6.97 (brm, 3H), 6.82 (brm, 1H), 5.67 (s, 1H), 3.18 (brm, 4H) 2.02 (brm, 4H) Mass Spectral Analysis m/z=294.0 (M+H)+
  • Example 31L
  • 31L was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1i.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brs, 2H), 7.37 (t, 1H), 7.25 (t, 1H), 6.97 (brm, 6H), 5.89 (s, 1H), 3.79 (s, 3H), 3.21 (brm, 4H), 2.03 (brm, 4H)
  • Mass Spectral Analysis m/z=308.0 (M+H)+
  • Example 31M
  • 31M was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1j.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.60 (s, 1H), 9.05 (brs, 2H), 7.24 (m, 2H), 7.02 (m, 2H), 6.94 (m, 1H), 6.82 (d, 1H), 6.76 (m, 2H), 5.82 (s, 1H), 3.20 (brm, 4H), 2.03 (brm, 4H)
  • Mass Spectral Analysis m/z=294.0 (M+H)+
  • Example 31N
  • 31N was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1k.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (brm, 1.5H), 8.20 (s, 1H), 8.05 (s, 2H), 7.29 (m, 1H), 7.08 (d, 1H), 6.97 (t, 1H), 6.90 (dd, 1H), 6.16 (s, 1H), 3.23 (brm, 4H), 2.08 (brm, 4H)
  • Mass Spectral Analysis m/z=414.1 (M+H)+
  • Example 31O
  • 31O was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1l.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (brs, 2H), 7.42 (m, 1H), 7.07 (brm, 5H), 6.83 (t, 1H), 6.60 (d, 1H), 5.73 (s, 1H), 3.65 (s, 3H), 3.18 (brm, 4H), 2.08 (brm, 2H), 1.96 (brm, 2H)
  • Mass Spectral Analysis m/z=308.0 (M+H)+
  • Example 31P
  • 31P was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1m.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.46 (s, 1H), 9.02 (brs, 2H), 7.22 (t, 1H), 7.16 (t, 1H), 7.10 (d, 1H), 6.93 (m, 2H), 6.84 (m, 2H), 6.70 (d, 1H), 5.71 (s, 1H), 3.20 (brm, 4H), 2.11 (brm, 2H), 1.97 (brm, 2H)
  • Mass Spectral Analysis m/z=294.0 (M+H)+
  • Example 31Q
  • 31Q was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1n.
  • Step 31.2: Method 1E was used.
  • 1H NMR (400 MHz, CDCl3) δ 9.75 (m, 2H), 7.85 (m, 1H), 7.78 (m, 1H), 7.49 (m, 1H), 7.37 (m, 3H), 7.28 (m, 1H), 6.99 (m, 2H), 5.88 (s, 1H), 3.42 (m, 4H), 2.27 (m, 4H)
  • Mass Spectral Analysis m/z=333.9 (M+H)+
  • Example 31R
  • 31R was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1o.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (m, 2H), 7.66 (m, 3H), 7.34 (m, 4H), 7.10 (m, 2H), 6.48 (m, 1H), 3.23 (m, 4H), 2.09 (m, 4H)
  • Mass Spectral Analysis m/z=318.1 (M+H)+
  • Example 31S
  • 31S was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1p.
  • Step 31.2: Method 31A was used.
  • 1H NMR (400 MHz, CDCl3) δ 9.81 (brs, 1H), 9.40 (brs, 1H), 8.76 (brs, 2H), 7.98 (d, 1H), 7.67 (brs, 1H), 7.29 (m, 1H), 7.01 (d, 1H), 6.95 (t, 1H), 6.91 (d, 1H), 5.70 (s, 1H), 3.43 (m, 2H), 3.34 (m, 2H), 2.29 (m, 2H), 2.15 (m, 2H)
  • Mass Spectral Analysis m/z=279.1 (M+H)+
  • Example 31T
  • 31T was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1q.
  • Step 31.2: Method 1E was used.
  • 1HNMR (400 MHz, CDCl3) δ 9.71 (m, 2H), 7.44-7.21 (m, 3H), 7.11 (m, 2H), 6.96 (m, 2H), 5.75 (s, 1H), 3.39 (m, 4H), 2.24 (m, 4H)
  • Mass Spectral Analysis m/z=283.9 (M+H)+
  • Example 31U
  • 31U was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1r.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (brm, 1.5H), 7.66 (m, 1H), 7.62 (m, 1H), 7.26 (m, 1H), 7.20 (m, 2H), 7.03 (d, 1H), 6.97 (t, 1H), 5.96 (s, 1H), 3.20 (brm, 4H), 2.07 (brm, 2H), 1.98 (brm, 2H)
  • Mass Spectral Analysis m/z=284.1 (M+H)+
  • Example 31V
  • 31V was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1s.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, CDCl3) δ 9.71 (brs, 1H), 9.29 (brs, 1H), 7.52 (m, 3H), 6.99 (m, 2H), 6.59 (m, 1H), 6.49 (m, 1H), 5.95 (s, 1H), 3.42 (m, 2H), 3.32 (m, 2H), 2.25 (m, 2H), 2.10 (m, 2H)
  • Mass Spectral Analysis m/z=268.1 (M+H)+
  • Example 31W
  • 31W was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1t.
  • Step 31.2: Method 1F was used.
  • 1H NMR (400 MHz, DMSO d6) δ 9.34 (brm, 1.5H), 8.12 (d, 1H), 7.60 (m, 6H), 7.42 (t, 1H), 7.32 (t, 1H), 7.22 (t, 1H), 7.02 (d, 1H), 6.89 (m, 2H), 6.81 (d, 1H), 5.98 (s, 1H), 3.41 (brs, 2H), 2.20 (brm, 6H)
  • Mass Spectral Analysis m/z=457.1 (M+H)+
  • Example 31X
  • 31X was obtained according to a procedure similar to the one described for 31A, with the following exceptions:
  • Step 31.1: 13.1 was replaced by 31.1u.
  • Step 31.2: Method 1E was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (m, 2H), 8.03 (d, 1H), 7.42 (d, 1H), 7.32 (m, 2H), 7.05 (m, 2H), 6.25 (s, 1H), 3.22 (m, 4H), 2.03 (m, 4H)
  • Mass Spectral Analysis m/z=308.8 (M+H)+
  • Example 31Y Preparation of 31Y
  • A solution of 16.2 (0.200 g, 0.0046 mol, 1.0 eq) in tetrahydrofuran (50 mL) was added drop wise to a cold (0° C.) suspension of lithium aluminum hydride (1.05 g, 0.027 mol, 6.0 eq) in tetrahydrofuran (50 mL). The mixture was allowed to warm to room temperature and was refluxed for 12 h under a nitrogen atmosphere. The reaction was cooled to room temperature and quenched by careful addition of water (3 mL). The mixture was stirred for 1 h at room temperature and filtered through celite. The celite was further rinsed with hot ethyl acetate. Evaporation of the filtrate afforded an oil which was dissolved in diethyl ether (20 mL). A 2.0M solution of hydrochloric acid in anhydrous diethyl ether (6.9 mL, 0.0138 mol, 3.0 eq) was added to the mixture. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 70%
  • 1H NMR (400 MHz, DMSO d6) δ 8.60 (m, 1H), 8.40 (m, 2H), 7.50 (m, 3H), 7.35 (m, 1H), 7.25 (m, 1H), 6.90-7.10 (m, 3H), 5.80 (s, 1H), 4.10 (m, 2H), 3.30 (m, 7H), 2.10 (m, 4H)
  • Mass Spectral Analysis m/z=321.1 (M+H)+
  • Example 31Z Preparation of 31Z
  • Acetyl chloride (0.14 mL, 0.0019 mol, 1.5 eq) was added drop wise to a cold solution of 31Y (dihydrochloric acid salt) (0.500 g, 0.0012 mol, 1.0 eq) and triethylamine (0.90 mL, 0.006 mol, 5.0 eq) in dichloromethane (10 mL). The mixture was allowed to warm to room temperature and stiffing was continued for 12 h at room temperature. The mixture was poured into water and ethyl acetate (30 mL) was added. The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and evaporated. The crude product was purified by column chromatography (eluent: dichloromethane/methanol, mixtures of increasing polarity). The purified compound was dissolved in diethyl ether (20 mL). A 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.8 mL, 0.0036 mol, 3.0 eq) was added to the mixture. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 31% Mass Spectral Analysis m/z=363.1 (M+H)+
  • 1H NMR (400 MHz, DMSO d6) δ 10.70 (m, 1H), 8.35 (m, 1H), 7.35 (m, 1H), 7.20-7.30 (m, 3H), 7.05 (m, 1H), 6.90 (m, 3H), 5.75 (s, 1H), 4.20 (s, 2H), 3.30 (m, 4H), 2.80 (s, 3H), 2.15 (m, 4H), 1.85 (s, 3H)
  • Example 31AA Preparation of 31AA
  • Methane sulfonyl chloride (0.15 mL, 0.0019 mol, 1.5 eq) was added drop wise to a cold solution of 31Y (dihydrochloric acid salt) (0.500 g, 0.0012 mol, 1.0 eq) and triethylamine (0.90 mL, 0.006 mol, 5.0 eq) in dichloromethane (10 mL). The mixture was allowed to warm to room temperature and stiffing was continued for 12 h at room temperature. The mixture was poured into water and ethyl acetate (30 mL) was added. The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and evaporated. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). The purified compound was dissolved in diethyl ether (20 mL). A 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.8 mL, 0.0036 mol, 3.0 eq) was added to the mixture. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 30%
  • 1H NMR (400 MHz, DMSO d6) δ 10.90 (m, 1H), 7.40 (m, 2H), 7.35 (m, 1H), 7.30 (m, 2H), 7.10 (m, 1H), 7.00 (m, 2H), 5.75 (s, 1H), 4.20 (d, 2H), 3.30 (m, 4H), 2.90 (s, 3H), 2.80 (s, 3H), 2.10 (m, 4H)
  • Mass Spectral Analysis m/z=399.1 (M+H)+
  • Example 32A Preparation of 32.1
  • To a solution of Bis(pinacolato)diboron 1.14 (14.7 g, 57.8 mmol, 2.0 eq) in N,N-dimethylformamide (200 mL) at room temperature under a nitrogen atmosphere was added 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (710 mg, 0.867 mmol, 0.03 eq) followed by addition of potassium acetate (8.58 g, 86.7 mmol, 3.0 eq.) The mixture was heated to 80° C. followed by drop wise addition of a solution of the enol triflate 1.5a (13.0 g, 28.9 mmol, 1.0 eq) in N,N-dimethylformamide (100 mL). After the addition was complete, the reaction mixture was heated at 80° C. for an additional 16 h. The solvent was evaporated under vacuum and the residue was added to a 1N aqueous solution of hydrochloric acid. The aqueous residue was extracted with ethyl acetate. The organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to afford a brown semisolid. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 96.0%
  • 1H NMR (400 MHz, CDCl3) δ 7.71 (d, 1H), 7.11 (t, 1H), 6.90 (t, 1H), 6.83 (d, 1H), 6.28 (s, 1H), 3.84 (brs, 2H), 3.27 (brm, 2H), 1.96 (d, 2H), 1.60 (m, 2H), 1.34 (s, 9H), 1.26 (s, 12H)
  • Mass Spectral Analysis m/z=428.0 (M+H)+
  • Preparation of 32.2a
  • To a solution of 4-bromophenylacetic acid (32.4) (3.21 g, 15 mmol) in methylene chloride (300 mL) was added diethylamine (1.12) (3.2 mL, 30 mmol, 2.0 eq) followed by triethylamine (8.4 ml, 60 mmol, 4.0 eq) and the Mukaiyama acylating reagent (2-chloro-1-methylpyridinium iodide) (4.61 mg, 18 mmol, 1.2 eq). The reaction mixture was stirred at room temperature overnight and the mixture was washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 2:1:1). Yield: 89.2%
  • 1H NMR (400 MHz, CDCl3) δ 7.43 (d, 2H), 7.15 (d, 2H), 3.63 (s, 2H), 3.40 (q, 2H), 3.30 (q, 2H), 1.10 (t, 3H)
  • Preparation of 32.3a
  • To a solution of 32.1 (2.14 g, 5 mmol) in dimethoxyethane (DME) (40 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (8 mL, 16 mmol, 3.2 eq), lithium chloride (679 mg, 16 mmol, 3.2 eq.), 32.2a (1.62 mg, 6 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (174 mg, 0.15 mmol, 0.03 eq). The mixture was refluxed overnight under a nitrogen atmosphere. The mixture was then cooled to room temperature and water (50 mL) was added. The mixture was extracted with ethyl acetate. The organic layer was further washed with brine, dried over sodium sulfate and concentrated under vacuum. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1). Yield: 61%
  • 1H NMR (400 MHz, CDCl3) δ 7.29 (s, 4H), 7.18 (t, 1H), 7.03 (d, 1H), 6.95 (d, 1H), 5.86 (t, 1H), 5.53 (s, 1H), 3.86 (m, 2H), 3.72 (s, 2H), 3.39 (m, 6H), 2.05 (m, 2H), 1.68 (m, 2H), 1.49 (s, 9H), 1.16 (m, 6H)
  • Preparation of 32A
  • To a solution of 32.3a (1.4 g, 3.38 mmol) in methylene chloride (15 mL) was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (50 mL). The mixture was stirred at room temperature for 24 h and diluted by addition of diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether. Yield: 92%
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (m, 2H), 7.20 (s, 4H), 7.24 (m, 1H), 7.00 (m, 3H), 5.83 (s, 1H), 3.40-3.20 (m, 8H), 2.03 (m, 4H), 1.08 (m, 6H)
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Example 32B
  • 32B was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2b and Method 1C was used.
  • Note: 32.2b was obtained according to a procedure similar to the one described for 32.2e (see 32E) except 13.4b was replaced by 1.12 in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.02 (brs, 2H), 8.88 (s, 2H), 8.57 (s, 2H), 7.23 (s, 1H), 7.05 (s, 1H), 6.91 (s, 2H), 6.00 (s, 1H), 3.32 (s, 4H), 3.12 (brs, 4H), 2.08 (m, 4H), 1.02 (brd, 6H)
  • Mass Spectral Analysis m/z=454.0 (M+H)+
  • Elemental analysis:
  • C23H28N2O3S, 1HCl, 1/3H2O
  • Theory: % C, 60.71; % H, 6.57; % N, 6.16.
  • Found: % C, 60.64; % H, 6.36; % N, 6.16.
  • Example 32C
  • 32C was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2c and Method 1D was used.
  • Note: 32.2c was obtained according to a procedure similar to the one described for 32.2e (see 32E) except 13.4b was replaced by 3.4c in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (brs, 2H), 7.86 (d, 2H), 7.68 (t, 1H), 7.60 (d, 2H), 7.28 (m, 1H), 7.06 (d, 1H), 6.96 (d, 2H), 6.01 (s, 1H), 3.21 (brm, 4H), 2.81 (m, 2H), 2.10 (brm, 2H), 2.01 (brm, 2H), 1.00 (t, 3H)
  • Mass Spectral Analysis m/z=385.3 (M+H)+
  • Elemental analysis:
  • C21H24N2O3S, 1HCl, 0.25H2O
  • Theory: % C, 59.28; % H, 6.04; % N, 6.58.
  • Found: % C, 59.06; % H, 5.92; % N, 6.44.
  • Example 32D
  • 32D was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2d.
  • Note: 32.2d was obtained according to a procedure similar to the one described for 32.2e (see 32E) except 13.4b was replaced by 32.6 in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.13 (brs, 2H), 7.90 (d, 2H), 7.64 (s, 1H), 7.56 (d, 2H), 7.27 (m, 1H), 7.06 (d, 1H), 6.95 (m, 2H), 6.01 (s, 1H), 3.22 (brm, 4H), 2.07 (brm, 4H), 1.12 (s, 9H)
  • Mass Spectral Analysis m/z=413.3 (M+H)+
  • Example 32E Preparation of 32.2e
  • 13.4b (7.33 mL, 64.58 mmol, 3.3 eq) was added at room temperature to a solution of 32.5 (5 g, 19.57 mmol, 1 eq) in tetrahydrofuran (20 mL). The reaction was stirred at room temperature overnight. The mixture was concentrated under reduced pressure and dichloromethane was added. The mixture was washed with water, a saturated aqueous solution of sodium bicarbonate and brine, and then dried over sodium sulfate and filtered. The organic extracts were concentrated under reduced pressure and the crude product was used for the next step without further purification. Yield: 40%
  • 1H NMR (400 MHz, DMSO d6) δ 7.82 (s, 4H), 7.25 (s, 4H), 4.58 (s, 4H)
  • Mass Spectral Analysis m/z=337.9 (M+H)+
  • Preparation of 32E
  • 32E was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2e.
  • 1H NMR (400 MHz, DMSO d6) δ 9.06 (brs, 2H), 7.94 (d, 2H), 7.60 (d, 2H), 7.26 (m, 5H), 7.04 (d, 1H), 6.90 (m, 2H), 5.97 (s, 1H), 4.62 (s, 4H), 3.19 (brm, 4H), 2.03 (brm, 4H)
  • Mass Spectral Analysis m/z=459.3 (M+H)+
  • Example 32F
  • 32F was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2f.
  • Note: 32.2f was obtained according to a procedure similar to the one described for 32.2e except 13.4b was replaced by 3.4e in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (brs, 2H), 7.86 (d, 2H), 7.72 (t, 1H), 7.59 (d, 2H), 7.28 (m, 1H), 7.06 (d, 1H), 6.95 (d, 2H), 6.01 (s, 1H), 3.22 brm, 4H), 2.57 (t, 2H), 2.10 (brm, 2H), 2.02 (brm, 2H), 1.65 (m, 1H), 0.83 (d, 6H)
  • Mass Spectral Analysis m/z=413.3 (M+H)+
  • Elemental analysis:
  • C23H28N2O3S, 1HCl, 0.5H2O
  • Theory: % C, 60.31; % H, 6.60; % N, 6.12.
  • Found: % C, 60.67; % H, 6.33; % N, 6.10.
  • Example 32G
  • 32G was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2 g and Method 1D was used.
  • 32.2 g was obtained according to a procedure similar to the one described for 32.2e except 13.4b was replaced by 3.4 h in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.16 (brs, 2H), 7.87 (d, 2H), 7.70 (d, 1H), 7.59 (d, 2H), 7.28 (m, 1H), 7.06 (d, 1H), 6.95 (m, 2H), 6.01 (s, 1H), 3.24 (brm, 5H), 2.07 (brm, 4H), 0.98 (d, 6H)
  • Mass Spectral Analysis m/z=399.4 (M+H)+
  • Elemental analysis:
  • C22H26N2O3S, 1HCl
  • Theory: % C, 60.75; % H, 6.26; % N, 6.44.
  • Found: % C, 60.58; % H, 6.29; % N, 6.36.
  • Example 32H
  • 32H was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2h.
  • 32.2h was obtained according to a procedure similar to the one described for 32.2e except 13.4b was replaced by 3.4o in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.09 (brs, 2H), 7.89 (d, 2H), 7.58 (d, 2H), 7.28 (m, 1H), 7.06 (d, 1H), 6.94 (m, 2H), 6.02 (s, 1H), 3.76 (m, 2H), 3.22 (brm, 4H), 2.05 (brm, 4H), 1.20 (d, 12H)
  • Mass Spectral Analysis m/z=441.4 (M+H)+
  • Example 32I
  • 32I was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2i.
  • 32.2i was obtained according to a procedure similar to the one described for 32.2e except 13.4b was replaced by 13.4c in step 32.8.
  • 1H NMR (400 MHz, DMSO d6) δ 9.03 (brs, 2H), 7.66 (d, 2H), 7.38 (d, 2H), 7.08 (m, 1H), 6.86 (d, 1H), 6.74 (m, 2H), 5.81 (s, 1H), 3.00 (brm, 6H), 2.82 (d, 2H), 1.87 (brm, 4H), 1.37 (m, 2H), 0.71 (m, 1H), 0.65 (t, 3H), 0.27 (m, 2H), 0.01 (m, 2H)
  • Mass Spectral Analysis m/z=453.3 (M+H)+
  • Example 32J Preparation of 32J
  • Trifluoroacetic acid (5 mL, 64.90 mmol, 10.0 eq) was added drop wise to 32.3b (3.83 g, 7.47 mmol, 1.0 eq) at 0° C. The mixture was warmed to room temperature and stirring was continued for an additional 10 h at room temperature. The mixture was concentrated under reduced pressure. A saturated solution of sodium bicarbonate (50 mL) was added to the mixture, which was then extracted with dichloromethane. The organic phase was separated, washed with brine, dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. To a cold (0° C.) solution of the resulting oil in anhydrous dichloromethane (35 mL) was added drop wise a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (17 mL, 35.70 mmol, 5.5 eq). The mixture was then stirred for 1 h at room temperature and concentrated under reduced pressure. Diethyl ether was added. The resulting precipitate was collected by filtration and washed with diethyl ether. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 10%
  • 1H NMR (400 MHz, DMSO d6) δ 9.08 (m, 2H), 7.90 (m, 2H), 7.56 (m, 2H), 7.46 (m, 2H), 7.28 (m, 1H), 7.07 (m, 1H), 6.94 (m, 2H), 5.98 (s, 1H), 3.46 (m, 2H), 3.17 (m, 2H), 2.05 (m, 4H)
  • Mass Spectral Analysis m/z=357.4 (M+H)+
  • Elemental analysis:
  • C19H20N2O3S, 1HCl, 1H2O
  • Theory: % C, 55.54; % H, 5.64; % N, 6.82.
  • Found: % C, 55.30; % H, 5.28; % N, 6.55.
  • Example 32K Preparation of 32.9a
  • Triethylamine (0.96 mL, 6.88 mmol, 1.3 eq) was added to a solution of 20.2a (0.40 mL, 5.29 mmol, 1.0 eq) and 32.7 (1.0 g, 5.29 mmol, 1.0 eq) in acetonitrile (60 mL). The solution was refluxed for 1 h and then concentrated under reduced pressure. Methylene chloride was added and the organic mixture was washed with water. The organic mixture was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 93%
  • 1HNMR (400 MHz, CDCl3) δ 7.40 (d, 2H), 7.18 (d, 2H), 2.92 (q, 2H), 1.31 (t, 3H)
  • Preparation of 32.2j
  • To a solution of 32.9a (1.07 g, 4.93 mmol, 1.0 eq) in acetic acid (7 mL) was added a 30% aqueous solution of hydrogen peroxide (3 mL). The mixture was heated at 90° C. for 2 h. The mixture was cooled to room temperature. Water was added and the mixture was extracted with methylene chloride. The organic mixture was then washed with a saturated aqueous sodium thiosulfate solution and brine. The organic mixture was dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 92%
  • 1HNMR (400 MHz, CDCl3) δ 7.78 (d, 2H), 7.72 (d, 2H), 3.11 (q, 2H), 1.28 (t, 3H)
  • Preparation of 32K
  • 32K was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2j and Method 1D was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.86 (brs, 1H), 7.96 (d, 2H), 7.66 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.96 (d, 2H), 6.04 (s, 1H), 3.37 (m, 2H), 3.22 (m, 4H), 2.10 (m, 2H), 2.00 (m, 2H), 1.13 (t, 3H)
  • Mass Spectral Analysis m/z 370.2 (M+H)+
  • Elemental analysis:
  • C21H23NO3S, 1HCl, 0.33H2O
  • Theory: % C, 61.23; % H, 6.04; % N, 3.40; % S, 7.78.
  • Found: % C, 61.15; % H, 5.92; % N, 3.39; % S, 7.68.
  • Example 32L
  • 32L was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2k and Method 12A was used.
  • Note: 32.2k was obtained according to a procedure similar to the one described for
  • 32.2j except 20.2a was replaced by 20.2b in step 32.6.
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (brs, 1H), 7.96 (d, 2H), 7.66 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.96 (d, 2H), 6.04 (s, 1H), 3.31 (m, 2H), 3.22 (m, 4H), 2.10 (m, 2H), 2.00 (m, 2H), 1.58 (m, 2H), 0.94 (t, 3H)
  • Mass Spectral Analysis m/z=384.2 (M+H)+
  • Elemental analysis:
  • C22H25NO3S, 1HCl, 0.5H2O
  • Theory: % C, 61.60; % H, 6.34; % N, 3.27; % S, 7.47.
  • Found: % C, 61.88; % H, 6.28; % N, 3.36; % S, 7.36.
  • Example 32M
  • 32M was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.21 and Method 12A was used.
  • Note: 32.21 was obtained according to a procedure similar to the one described for 32.2j except 20.2a was replaced by 2.8a in step 32.6.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brs, 1H), 7.97 (d, 2H), 7.65 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.95 (m, 2H), 6.04 (s, 1H), 3.32 (m, 2H), 3.22 (m, 4H), 2.10 (m, 2H), 2.01 (m, 2H), 0.87 (m, 1H), 0.47 (m, 2H), 0.13 (m, 2H)
  • Mass Spectral Analysis m/z=396.2 (M+H)+
  • Elemental analysis:
  • C23H25NO3S, 1HCl
  • Theory: % C, 63.95; % H, 6.07; % N, 3.24; % S, 7.42.
  • Found: % C, 63.94; % H, 6.03; % N, 3.32; % S, 7.32.
  • Example 32N
  • 32N was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2m and Method 12A was used.
  • Note: 32.2m was obtained according to a procedure similar to the one described for 32.2j except 20.2a was replaced by 32.8a in step 32.6.
  • 1H NMR (400 MHz, DMSO d6) δ 8.91 (brs, 1H), 7.98 (d, 2H), 7.66 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.96 (m, 2H), 6.04 (s, 1H), 3.32 (m, 2H), 3.22 (m, 4H), 2.10 (m, 2H), 2.02 (m, 2H), 1.62 (m, 1H), 1.46 (m, 2H), 0.84 (d, 6H)
  • Mass Spectral Analysis m/z=412.2 (M+H)+
  • Elemental analysis:
  • C24H29NO3S, 1HCl, 0.33H2O
  • Theory: % C, 63.49; % H, 6.81; % N, 3.08.
  • Found: % C, 63.45; % H, 6.71; % N, 3.39.
  • Example 32O
  • 32O was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2n and Method 12A was used.
  • Note: 32.2n was obtained according to a procedure similar to the one described for 32.2p (see 32Q) except 32.8d was replaced by 32.8b in step 32.6.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brm, 1H), 7.98 (d, 2H), 7.64 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.94 (m, 2H), 6.02 (s, 1H), 3.32 (m, 2H), 3.22 (m, 4H), 2.10 (m, 2H), 2.01 (m, 2H), 1.10 (s, 9H)
  • Mass Spectral Analysis m/z=412.2 (M+H)+
  • Elemental analysis:
  • C24H29NO3S, 1HCl, 0.33H2O
  • Theory: % C, 63.49; % H, 6.81; % N, 3.08; % S, 7.06.
  • Found: % C, 63.49; % H, 6.70; % N, 3.25; % S, 6.78.
  • Example 32P
  • 32P was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2o and Method 12A was used.
  • Note: 32.2o was obtained according to a procedure similar to the one described for 32.2p (see 32Q) except 32.8d was replaced by 32.8c in step 32.6.
  • 1H NMR (400 MHz, DMSO d6) δ 8.82 (brs, 2H), 7.93 (d, 2H), 7.66 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.96 (m, 2H), 6.05 (s, 1H), 3.47 (m, 1H), 3.23 (m, 4H), 2.10 (m, 2H), 2.00 (m, 2H), 1.19 (d, 6H)
  • Mass Spectral Analysis m/z=384.2 (M+H)+
  • Elemental analysis:
  • C22H25NO3S, 1HCl
  • Theory: % C, 62.92; % H, 6.24; % N, 3.34; % S, 7.63.
  • Found: % C, 63.18; % H, 6.26; % N, 3.46; % S, 7.54.
  • Example 32Q Preparation of 32.9b
  • To a suspension of sodium hydride (0.33 g, 13.75 mmol, 1.3 eq) in N,N-dimethylformamide (10 mL) at 0° C. under nitrogen was added drop wise a solution of 32.7 (2.0 g, 10.58 mmol, 1.0 eq) in N,N-dimethylformamide (5 mL). The mixture was stirred for 10 min at 0° C. and 32.8d (1.48 mL, 10.58 mmol, 1.0 eq) was added drop wise. The mixture was allowed to warm to room temperature and stirring continued for a further 16 h at room temperature. The reaction was carefully quenched with water and the mixture was extracted with diethyl ether. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • Yield: 87%
  • 1HNMR (400 MHz, CDCl3) δ 7.38 (d, 2H), 7.18 (d, 2H), 2.87 (d, 2H), 1.45 (m, 5H), 0.88 (t, 6H)
  • Preparation of 32.2p
  • To a solution of 32.9b (2.53 g, 9.26 mmol, 1.0 eq) in acetic acid (14 mL) was added a 30% aqueous solution of hydrogen peroxide (6 mL). The mixture was heated at 90° C. for 2 h. The mixture was cooled to room temperature. Water was added and the crude product was extracted with methylene chloride. The organic mixture was washed with a saturated aqueous sodium thiosulfate solution and brine. The mixture was dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 80%
  • 1HNMR (400 MHz, CDCl3) δ 7.78 (d, 2H), 7.71 (d, 2H), 3.00 (d, 2H), 1.88 (m, 1H), 1.46 (m, 4H), 0.82 (t, 6H)
  • Preparation of 32Q
  • 32Q was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2p and Method 12A was used.
  • (32Q) 1H NMR (400 MHz, DMSO d6) δ 8.97 (brs, 2H), 7.99 (d, 2H), 7.65 (d, 2H), 7.29 (m, 1H), 7.07 (d, 1H), 6.94 (m, 2H), 6.03 (s, 1H), 3.23 (m, 6H), 2.10 (m, 2H), 2.02 (m, 2H), 1.73 (m, 1H), 1.40 (m, 4H), 0.77 (t, 6H)
  • Mass Spectral Analysis m/z=426.2 (M+H)+
  • Elemental analysis:
  • C25H31NO3S, 1HCl, 0.33H2O
  • Theory: % C, 64.15; % H, 7.03; % N, 2.99; % S, 6.85.
  • Found: % C, 64.26; % H, 6.91; % N, 3.20; % S, 6.35.
  • Example 32R Preparation of 32.2q
  • To a solution 4-bromo-N-methylaniline of (32.10) (0.74 g, 4 mmol, 1.0 eq) in dry dichloromethane (50 mL) at 0° C. was slowly added triethylamine (2.23 mL, 8 mmol, 2.0 eq). The mixture was stirred for 10 min at room temperature and 19.8a (0.63 mL, 6 mmol, 1.5 eq) was added drop wise to the reaction mixture. The reaction mixture was slowly warmed to room temperature and was stirred for 10 h at room temperature. Dichloromethane (100 mL) was added to the mixture which was washed with a 1M aqueous solution of hydrochloric acid (3×50 mL), a saturated aqueous sodium bicarbonate (2×50 mL) and brine. The organic extracts were dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product, which was used for next step without purification.
  • 1H NMR (400 MHz, CDCl3) δ 7.56 (m, 2H), 7.08 (m, 2H), 3.23 (s, 3H), 2.49 (m, 1H), 1.02 (d, 6H)
  • Mass Spectral Analysis m/z=256.15 (M+H)+
  • Preparation of 32R
  • 32R was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2q and Method 1D was used.
  • 1H NMR (400 MHz, DMSO d6) δ 8.91 (brs, 2H), 7.43 (m, 4H), 7.27 (m, 1H), 7.01 (m, 3H), 5.96 (s, 1H), 3.40-3.14 (m, 8H), 2.04 (m, 4H), 0.96 (m, 6H)
  • Mass Spectral Analysis m/z=377.3 (M+H)+
  • Elemental analysis:
  • C24H28N2O3, 1HCl, 2/3H2O
  • Theory: % C, 67.83; % H, 7.19; % N, 6.59.
  • Found: % C, 67.78; % H, 7.19; % N, 6.50.
  • Example 32S
  • 32S was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2r and Method 1D was used.
  • Note: 32.2r was obtained according to a procedure similar to the one described for 32.2q except 19.8a was replaced by 19.8b in step 32.9.
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (brs, 2H), 7.47 (m, 2H), 7.33 (m, 2H), 7.27 (m, 1H), 7.00 (m, 3H), 5.96 (s, 1H), 3.40-3.12 (m, 7H), 2.25-1.94 (m, 5H), 1.48 (m, 2H), 1.30 (m, 2H), 0.76 (m, 6H)
  • Mass Spectral Analysis m/z=405.4 (M+H)+
  • Elemental analysis:
  • C26H32N2O2, 1HCl, 1/5H2O
  • Theory: % C, 70.24; % H, 7.57; % N, 6.30.
  • Found: % C, 70.20; % H, 7.50; % N, 6.19.
  • Example 32T
  • 32T was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2s and Method 1D was used.
  • Note: 32.2s was obtained according to a procedure similar to the one described for 32.2q except 19.8a was replaced by 32.11a in step 32.9.
  • 1H NMR (400 MHz, DMSO d6) δ 8.95 (brs, 2H), 7.44 (m, 2H), 7.37 (m, 2H), 7.27 (m, 1H), 7.00 (m, 3H), 5.96 (s, 1H), 3.21 (m, 7H), 2.03 (m, 7H), 0.81 (m, 6H)
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.1H2O
  • Theory: % C, 70.03; % H, 7.33; % N, 6.53.
  • Found: % C, 69.97; % H, 7.33; % N, 6.57.
  • Example 32U
  • 32U was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2t and Method 1D was used.
  • Note: 32.2t was obtained according to a procedure similar to the one described for 32.2q except 19.8a was replaced by 6.7 in step 32.9.
  • 1H NMR (400 MHz, DMSO d6) δ 8.95 (m, 2H), 7.42 (m, 4H), 7.26 (m, 1H), 7.00 (m, 3H), 5.93 (s, 1H), 3.20 (m, 7H), 2.04 (m, 4H), 1.83 (s, 3H)
  • Mass Spectral Analysis m/z=349.2 (M+H)+
  • Elemental analysis:
  • C22H24N2O2, 1HCl, 1.4H2O
  • Theory: % C, 64.43; % H, 6.83; % N, 6.83.
  • Found: % C, 64.49; % H, 6.87; % N, 6.89.
  • Example 32V
  • 32V was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2u and Method 1D was used.
  • Note: 32.2u was obtained according to a procedure similar to the one described for 32.2q except 19.8a was replaced by 32.11b in step 32.9.
  • 1H NMR (400 MHz, DMSO d6) δ 8.95 (m, 2H), 7.42 (m, 4H), 7.26 (m, 1H), 7.05 (m, 1H), 6.96 (m, 2H), 5.94 (s, 1H), 3.20 (m, 7H), 2.05 (m, 6H), 1.38 (m, 3H), 0.74 (m, 6H)
  • Mass Spectral Analysis m/z=405.3 (M+H)+
  • Elemental analysis:
  • C26H32N2O2, 1HCl, 1.5H2O
  • Theory: % C, 66.72; % H, 7.75; % N, 5.99.
  • Found: % C, 66.57; % H, 7.67; % N, 5.93.
  • Example 32W
  • 32W was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2v and Method 1D was used.
  • Note: 32.2v is commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 8.91 (brs, 2H), 7.74 (m, 2H), 7.37 (m, 2H), 7.25 (m, 1H), 7.02 (m, 2H), 6.94 (m, 1H), 5.86 (s, 1H), 3.87 (t, 2H), 3.20 (m, 4H), 2.52 (t, 2H), 2.08 (m, 4H), 1.99 (m, 2H)
  • Mass Spectral Analysis m/z=361.2 (M+H)+
  • Elemental analysis:
  • C23H24N2O2, 1HCl, 0.5H2O
  • Theory: % C, 68.06; % H, 6.46; % N, 6.90. Found: % C, 68.10; % H, 6.42; % N, 6.96.
  • Example 32X
  • 32X was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2w and Method 1D was used.
  • Note: 32.2w is commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 8.82 (brs, 2H), 8.07 (d, 1H), 7.24 (m, 2H), 7.14 (d, 1H), 7.02 (m, 2H), 6.94 (m, 1H), 5.82 (s, 1H), 4.13 (t, 2H), 3.19 (m, 6H), 2.18 (s, 3H), 2.06 (m, 2H), 1.96 (m, 2H)
  • Mass Spectral Analysis m/z=361.3 (M+H)+
  • Elemental analysis:
  • C23H24N2O2, 1HCl, 0.4H2O
  • Theory: % C, 68.36; % H, 6.44; % N, 6.93.
  • Found: % C, 68.41; % H, 6.23; % N, 6.93.
  • Example 32Y
  • 32Y was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2× and Method 1D was used.
  • Note: 32.2x was obtained according to a procedure similar to the one described for 32.2q except 19.8a was replaced by 32.11c in step 32.9.
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (brs, 2H), 7.41 (m, 4H), 7.26 (m, 1H), 7.00 (m, 3H), 5.94 (s, 1H), 3.20 (m, 7H), 2.05 (m, 6H), 1.49 (m, 2H), 3.79 (m, 3H)
  • Mass Spectral Analysis m/z=377.4 (M+H)+
  • Elemental analysis:
  • C24H28N2O2, 1HCl, 1.1H2O
  • Theory: % C, 66.61; % H, 7.27; % N, 6.47.
  • Found: % C, 66.51; % H, 7.20; % N, 6.39.
  • Example 32Z
  • 32Z was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 32.2y and Method 1D was used.
  • Note: 32.2y was obtained according to a procedure similar to the one described for 32.2q except 19.8a was replaced by 32.11d in step 32.9.
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (brs, 2H), 7.41 (m, 4H), 7.26 (m, 1H), 7.00 (m, 3H), 5.94 (s, 1H), 3.20 (m, 7H), 2.05 (m, 6H), 1.46 (m, 2H), 1.18 (m, 2H), 3.79 (m, 3H)
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.9H2O
  • Theory: % C, 67.75; % H, 7.46; % N, 6.32.
  • Found: % C, 67.71; % H, 7.45; % N, 6.30.
  • Example 33A
  • 33A was obtained according to a procedure similar to the one described for 32A, with the following exception:
  • Step 32.2: 32.2a was replaced by 33.1a (see also step 33.2).
  • Note: 33.1a was commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 7.98 (d, 1H), 7.89 (dd, 1H), 7.84 (d, 1H), 7.29 (m, 1H), 7.01 (m, 2H), 6.42 (s, 1H), 3.07 (m, 4H), 1.95 (m, 4H)
  • Mass Spectral Analysis m/z=284.9 (M+H)+
  • Example 33B
  • 33B was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1b and Method 33A was used (see also step 33.2).
  • Note: 33.1b was commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 9.19 (m, 3H), 8.86 (m, 2H), 7.29 (m, 1H), 7.07 (m, 1H), 6.97 (m, 2H), 6.15 (s, 1H), 3.22 (m, 4H), 2.08 (m, 4H)
  • Mass Spectral Analysis m/z=279.9 (M+H)+
  • Example 33C
  • 33C was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1c and Method 33A was used (see also step 33.2).
  • Note: 33.1c is commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 7.73 (m, 1H), 7.21 (m, 1H), 6.90 (m, 5H), 5.94 & 5.88 (2s, 1H rotamer), 3.6-2.7 (m, 7H), 1.91 (m, 4H)
  • Mass Spectral Analysis m/z=282.0 (M+H)+
  • Example 33D
  • 33D was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1d and Method 33A was used (see also step 33.2).
  • Note: 33.1d is commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 8.87 (m, 2H), 7.80 (s, 2H), 7.56 (m, 1H), 7.32 (m, 2H), 7.26 (m, 1H), 7.15 (m, 2H), 6.18 (s, 1H), 3.30-3.07 (m, 4H), 2.03 (m, 4H)
  • Mass Spectral Analysis m/z=362.9 (M+H)+
  • Example 33E
  • 33E was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1e and Method 33A was used (see also step 33.2).
  • Note: 33.1e is commercially available.
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (brs, 2H), 8.80 (s, 1H), 8.15 (m, 1H), 8.08 (m, 1H), 7.30 (m, 1H), 7.07 (m, 1H), 6.96 (m, 2H), 6.17 (s, 1H), 3.23 (m, 4H), 2.08 (m, 4H)
  • Mass Spectral Analysis m/z=303.9 (M+H)+
  • Example 33F Preparation of 33.1f
  • To a stirred solution of 33.3 (3 g, 14.85 mmol, 1.0 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (6.2 mL, 35.64 mmol, 2.4 eq) and diethylamine (1.12) (3.1 mL, 29.70 mmol, 2 eq) at room temperature. The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (5.72 g, 17.82 mmol, 1.2 eq) was added portion wise. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (200 mL) and a 1M aqueous solution of sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), a 1M aqueous solution of hydrochloric acid (3×50 mL) and brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 8.72 (d, 1H), 8.55 (d, 1H), 7.87 (m, 1H), 3.56 (q, 2H), 3.27 (q, 2H), 1.26 (t, 3H), 1.16 (t, 3H)
  • Mass Spectral Analysis m/z=256.81 (M+H)+
  • Preparation of 33F
  • 33F was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1f and Method 33A was used (see also step 33.2).
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (brs, 2H), 8.65 (m, 2H), 7.80 (m, 1H), 7.29 (m, 1H), 7.07 (m, 1H), 6.96 (m, 2H), 6.09 (s, 1H), 3.52-3.10 (m, 8H), 2.05 (m, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=378.3 (M+H)+
  • Example 33G Preparation of 33.1g
  • To a stirred solution of 33.4 (3 g, 14.85 mmol, 1.0 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (6.2 mL, 35.64 mmol, 2.4 eq) and diethylamine (1.12) (3.1 mL, 29.70 mmol, 2 eq) at room temperature. The mixture was stirred for 10 min, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (5.72 g, 17.82 mmol, 1.2 eq) was added portion wise. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (200 mL) and a 1M aqueous solution of sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), a 1M aqueous solution of hydrochloric acid (3×50 mL) and brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.64 (d, 1H), 7.59 (dd, 1H), 7.52 (dd, 1H), 3.54 (q, 2H), 3.38 (q, 2H), 1.25 (m, 6H)
  • Mass Spectral Analysis m/z=256.7 (M+H)+
  • Preparation of 33G
  • 33G was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1g and Method 33A was used (see also step 33.2).
  • (33G) 1H NMR (400 MHz, DMSO d6) δ 9.01 (m, 2H), 8.01 (m, 1H), 7.59 (m, 2H), 7.26 (m, 1H), 7.13 (m, 1H), 7.04 (m, 1H), 6.93 (m, 1H), 6.11 (s, 1H), 3.51-3.11 (m, 8H), 2.05 (m, 4H), 1.15 (t, 3H), 1.06 (t, 3H)
  • Mass Spectral Analysis m/z=378.2 (M+H)+
  • Example 33H
  • 33H was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced with 33.1h and Method 1D was used (see also step 33.2).
  • Note: 33.1h was obtained according to a procedure similar to the one described for 1.13 (see 1N) except 1.12 was replaced by 3.4j in step 1.8 (see also step 33.9).
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (brs, 1H), 8.61 (d, 1H), 7.91 (dd, 1H), 7.64 (d, 1H), 7.29 (m, 1H), 7.06 (d, 1H), 6.97 (m, 2H), 6.09 (s, 1H), 3.23 (m, 4H), 3.04 (s, 3H), 2.99 (s, 3H), 2.11 (m, 2H), 2.02 (m, 2H)
  • Mass Spectral Analysis m/z=350.2 (M+H)+
  • Elemental analysis:
  • C21H23N3O2, 1.35HCl, 0.8H2O
  • Theory: % C, 61.06; % H, 6.33; % N, 10.17; % Cl, 11.59.
  • Found: % C, 60.72; % H, 6.23; % N, 10.05; % Cl, 11.26.
  • Example 33I
  • 33I was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced with 33.1i and Method 1D was used (see also step 33.2).
  • Note: 33.1i was obtained according to a procedure similar to the one described for 1.13 (see 1N) except 1.12 was replaced by 3.4c in step 1.8 (see also step 33.9).
  • 1H NMR (400 MHz, DMSO d6) δ 8.87 (m, 2H), 8.62 (d, 1H), 8.11 (d, 1H), 7.99 (dd, 1H), 7.30 (m, 1H), 7.08 (d, 1H), 6.96 (m, 2H), 6.10 (s, 1H), 3.35 (m, 2H), 3.24 (m, 4H), 2.11 (m, 2H), 2.02 (m, 2H), 1.14 (t, 3H)
  • Mass Spectral Analysis m/z=350.2 (M+H)+
  • Elemental analysis:
  • C21H23N3O2, 1.4HCl, 1.8H2O
  • Theory: % C, 58.26; % H, 6.52; % N, 9.71; % Cl, 11.47.
  • Found: % C, 58.26; % H, 6.23; % N, 9.59; % Cl, 11.83.
  • Example 33J
  • 33J was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced with 33.1j and Method 1D was used (see also step 33.2).
  • Note: 33.1j was obtained according to a procedure similar to the one described for 1.13 (see 1N) except 1.12 was replaced by 3.4b in step 1.8 (see also step 33.9).
  • 1H NMR (400 MHz, DMSO d6) δ 8.94 (brs, 1H), 8.83 (m, 1H), 8.62 (d, 1H), 8.11 (d, 1H), 7.98 (dd, 1H), 7.30 (m, 1H), 7.08 (d, 1H), 6.96 (m, 2H), 6.10 (s, 1H), 3.22 (m, 4H), 2.84 (d, 3H), 2.11 (m, 2H), 2.02 (m, 2H)
  • Mass Spectral Analysis m/z=336.2 (M+H)+
  • Elemental analysis:
  • C20H21N3O2, 1.1HCl, 0.8H2O
  • Theory: % C, 61.61; % H, 6.13; % N, 10.78; % Cl, 10.00.
  • Found: % C, 61.84; % H, 5.90; % N, 10.75; % Cl, 10.01.
  • Example 33K Preparation of 33.6
  • To a mixture of a 2.5M solution of n-butyl lithium in hexanes (0.84 mL, 2.1 mmol, 1.05 eq) and toluene (4 mL) at −78° C. was added a solution of 33.5 (0.57 g, 2.0 mmol, 1.0 eq) in toluene (2 mL). The reaction was stirred for 1 h at −78° C. The reaction was quenched with freshly crushed dry ice. The mixture was warmed slowly to room temperature and was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure and the resulting solid was treated with acetic acid. The solid was collected by filtration, dried under vacuum and used for the next step without further purification. Yield: 62%
  • 1H NMR (400 MHz, CD3OD) δ 8.90 (s, 2H)
  • Preparation of 33.7
  • To a solution of 33.6 (0.055 g, 0.27 mmol, 1.0 eq) in methylene chloride (5 mL) was added oxalyl chloride (0.050 mL, 0.58 mmol, 2.1 eq). The mixture was refluxed for 1 h and concentrated under reduced pressure. The crude acyl chloride was used for the next step without further purification.
  • Preparation of 33.1k
  • To a solution of 33.7 (0.060 g, 0.27 mmol, 1.0 eq) in tetrahydrofuran (2.5 mL) was added 1.12 (0.11 mL, 1.06 mmol, 4.0 eq). The mixture was stirred for 16 h and then diluted with ethyl acetate. The organic mixture was washed with water, with a saturated aqueous solution of sodium bicarbonate, a 1N aqueous solution of hydrochloric acid and brine. The organic mixture was dried over sodium sulfate, filtered, concentrated under reduced pressure and the crude product was used for the next step without further purification. Note: the product was isolated with a 17% impurity corresponding to N,N-diethyl-2-iodopyrimidine-5-carboxamide.
  • Yield: 86%
  • 1H NMR (400 MHz, CDCl3) δ 8.82 (s, 2H), 3.56 (q, 2H), 3.20 (q, 2H), 1.28 (t, 3H), 1.18 (t, 3H)
  • Preparation of 33K
  • 33K was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 33.1k and Method 12A (see was used (see also step 33.2).
  • 1H NMR (400 MHz, DMSO d6) δ 8.81 (m, 2H), 7.18 (m, 1H), 6.92 (m, 2H), 6.85 (m, 1H), 6.06 (s, 0.8H), 6.04 (s, 0.2H), 3.41 (q, 2H), 3.06 (q, 2H), 2.86 (m, 2H), 2.76 (m, 2H), 1.73 (brm, 4H), 1.10 (t, 3H), 1.00 (t, 3H)
  • Mass Spectral Analysis m/z=379.3 (M+H)+
  • Example 33L Preparation of 33L
  • To a solution of 33.2a (0.27 g, 0.67 mmol, 1 eq) in dry dichloromethane (15 mL) was added dropwise a 4.0M solution of hydrogen chloride in dioxane (1.34 mL, 5.35 mmol, 8 eq). The reaction mixture was stirred at room temperature for 10 h and concentrated under reduced pressure. The crude mixture (containing a mixture of 33E and 33L) was purified by column chromatography (eluent: dichloromethane/methanol/ammonium hydroxide mixture of increasing polarity), affording the 33L in a pure form.
  • 1H NMR (400 MHz, DMSO d6) δ 8.59 (d, 1H), 8.17 (s, 1H), 8.09 (d, 1H), 7.95 (dd, 1H), 7.71 (s, 1H), 7.23 (m, 1H), 6.97 (d, 1H), 6.91 (m, 2H), 6.02 (s, 1H), 2.91 (m, 2H), 2.77 (m, 2H), 1.82 (m, 2H), 1.73 (m, 2H)
  • Mass Spectral Analysis m/z=321.9
  • Example 34A Preparation of 34.1a
  • To a stirred solution of 34.3 (2.5 g, 12.38 mmol, 1.0 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (4.74 mL, 27.24 mmol, 2.2 eq) and diethylamine (1.12) (2.56 mL, 24.76 mmol, 2.0 eq) at room temperature. The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (4.37 g, 13.62 mmol, 1.1 eq) was added portion wise to the reaction mixture. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (200 mL) and 1M aqueous sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), with a 1M aqueous solution of hydrochloric acid (3×50 mL) and brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 78%
  • 1H NMR (400 MHz, CDCl3) δ 8.41 (m, 1H), 7.59 (m, 1H), 7.55 (m, 1H), 3.55 (q, 2H), 3.27 (q, 2H), 1.25 (t, 3H), 1.15 (t, 3H)
  • Mass Spectral Analysis m/z=257.04 (M+H)+
  • Preparation of 34A
  • 34A was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1a (see also step 34.2).
  • 1H NMR (400 MHz, DMSO d6) δ 8.94 (brm, 2H), 8.64 (s, 1H), 7.92 (dd, 1H), 7.65 (d, 1H), 7.29 (m, 2H), 7.05 (d, 1H), 6.96 (t, 1H), 6.22 (s, 1H), 3.48 (m, 2H), 3.24 (brm, 6H), 2.05 (brm, 4H), 1.14 (brd, 6H)
  • Mass Spectral Analysis m/z=378.4 (M+H)+
  • Elemental analysis:
  • C23H27N3O2, 1HCl, 1.3H2O
  • Theory: % C, 63.16; % H, 7.05; % N, 9.61.
  • Found: % C, 63.05; % H, 6.75; % N, 9.50.
  • Example 34B
  • 34B was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1b (see also step 34.2).
  • Note: 34.1b was obtained according to a procedure similar to the one described for 34.1a except 1.12 was replaced by 3.4o in step 34.4.
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (brs, 2H), 8.59 (d, 1H), 7.85 (dd, 1H), 7.64 (d, 1H), 7.28 (m, 2H), 7.05 (d, 1H), 6.96 (t, 1H), 6.21 (s, 1H), 3.67 (m, 2H), 3.22 (brm, 4H), 2.06 (brm, 4H), 1.45 (brs, 6H), 1.15 (brs, 6H)
  • Mass Spectral Analysis m/z=406.4 (M+H)+
  • Elemental analysis:
  • C25H31N3O2, 1.5HCl, 0.66H2O
  • Theory: % C, 63.59; % H, 7.22; % N, 8.90; % Cl, 11.26.
  • Found: % C, 63.68; % H, 7.21; % N, 8.99; % Cl, 11.28.
  • Example 34C Preparation of 34.1c
  • To a stirred solution of 34.4 (2.1 g, 10 mmol, 1.0 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (4.2 mL, 24 mmol, 2.4 eq) and diethylamine (1.12) (2.1 mL, 20 mmol, 2 eq) at room temperature. The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (3.85 g, 12 mmol, 1.2 eq) was added portion wise. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (200 mL) and a 1M aqueous solution of sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), with a 1N aqueous solution of hydrochloric acid (3×50 mL) and brine, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was used for next step without further purification.
  • Mass Spectral Analysis m/z=262.1 (M+H)+
  • Preparation of 34C
  • 34C was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1c (see also step 34.2).
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (brs, 2H), 7.41 (d, 1H), 7.37 (d, 1H), 7.31 (t, 1H), 7.22 (d, 1H), 7.07 (d, 1H), 7.02 (t, 1H), 6.12 (s, 1H), 3.50 (brm, 4H), 3.21 (brm, 4H0, 2.03 (brm, 4H), 1.18 (brt, 6H)
  • Mass Spectral Analysis m/z=383.3 (M+H)+
  • Elemental analysis:
  • C22H26N2O2S, 1HCl
  • Theory: % C, 63.07; % H, 6.50; % N, 6.69.
  • Found: % C, 63.03; % H, 6.52; % N, 6.61.
  • Example 34D
  • 34D was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1d (see also step 34.2).
  • Note: 34.1d was obtained according to a procedure similar to the one described for 34.1c except 1.12 was replaced by 3.4o in step 34.5.
  • 1H NMR (400 MHz, DMSO d6) δ 8.93 (brs, 2H), 7.38 (d, 1H), 7.31 (t, 1H), 7.26 (d, 1H), 7.19 (d, 1H), 7.07 (d, 1H), 7.02 (t, 1H), 6.10 (s, 1H), 3.97 (brs, 2H), 3.21 (brm, 4H), 2.07 (brm, 2H), 1.97 (brm, 2H), 1.31 (brd, 12H)
  • Mass Spectral Analysis m/z=411.4 (M+H)+
  • Elemental analysis: C24H30N2O2S, 1HCl,
  • Theory: % C, 64.48; % H, 6.99; % N, 6.27.
  • Found: % C, 64.25; % H, 7.01; % N, 6.22.
  • Example 34E Preparation of 34.1e
  • To a stirred solution of 34.5 (4.58 g, 17.5 mmol, 1.0 eq) in dichloromethane (100 mL) at 0° C. was slowly added triethylamine (7.32 mL, 52.5 mmol, 3 eq) followed by drop wise addition of diethylamine (1.12) (3.64 mL, 35.0 mmol, 2.0 eq). The reaction mixture was kept at 0° C. for 30 min. and then stirred at room temperature for 3 h. The mixture was washed with a 1N aqueous solution of hydrochloric acid (3×50 mL) and brine. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.30 (d, 1H), 7.05 (d, 1H), 3.24 (q, 4H), 1.19 (t, 6H)
  • Mass Spectral Analysis m/z=297.92 (M+H)+
  • Preparation of 34E
  • 34E was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1e (see also step 34.2).
  • (34E) 1H NMR (400 MHz, DMSO d6) δ 8.98 (brs, 2H), 7.68 (d, 1H), 7.34 (brm, 3H), 7.06 (m, 2H), 6.23 (s, 1H), 3.22 (brm, 8H), 2.03 (brm, 4H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=419.2 (M+H)+
  • Example 34F Preparation of 34.1f
  • To a stirred solution of 34.6 (2 g, 10.47 mmol, 1.0 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (4 mL, 23.03 mmol, 2.2 eq) and diethylamine (1.12) (2.1 mL, 20.94 mmol, 2.0 eq) at room temperature. The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (3.7 g, 11.52 mmol, 1.1 eq) was added portion wise. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (200 mL) and a 1M aqueous solution of sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), a 1M aqueous solution of hydrochloric acid (3×50 mL) and brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 91%
  • 1H NMR (400 MHz, CDCl3) δ 6.99 (d, 1H), 6.41 (d, 1H), 3.54 (brs, 4H), 1.26 (brs, 6H)
  • Mass Spectral Analysis m/z=246.0 (M+H)+
  • Preparation of 34F
  • 34F was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1f (see also step 34.2).
  • 1H NMR (400 MHz, DMSO d6) δ 9.05 (brs, 2H), 7.52 (d, 1H), 7.32 (t, 1H), 7.07 (brm, 3H), 6.91 (d, 1H), 6.26 (s, 1H), 3.50 (brs, 4H), 3.20 (brm, 4H), 2.05 (brm, 4H), 1.17 (brs, 6H)
  • Mass Spectral Analysis m/z=367.3 (M+H)+
  • Example 34G
  • 34G was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1 g (see also step 34.2).
  • Note: 34.1 g was obtained according to a procedure similar to the one described for 34.1f except 1.12 was replaced by 3.4o in step 34.8.
  • 1H NMR (400 MHz, DMSO d6) δ 8.89 (brs, 2H), 7.52 (d, 1H), 7.32 (t, 1H), 7.07 (m, 2H), 6.92 (d, 1H), 6.87 (d, 1H), 6.24 (s, 1H), 4.02 (brs, 2H), 3.20 (brm, 4H), 2.03 (brm, 4H), 1.31 (brs, 12H),
  • Mass Spectral Analysis m/z=395.5 (M+H)+
  • Example 34H Preparation of 34.1h
  • To a stirred solution of 34.7 (2.1 g, 10 mmol, 1.0 eq) in acetonitrile (20 mL) was slowly added diisopropylethylamine (4.2 mL, 24 mmol, 2.4 eq) and diethylamine (1.12) (2.1 mL, 20 mmol, 2 eq) at room temperature. The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (3.85 g, 12 mmol, 1.2 eq) was added portion wise. The reaction mixture was slowly warmed to room temperature and stirred for 10 h at room temperature. The volatiles were removed under reduced pressure and the residue was partitioned between ethyl acetate (200 mL) and a 1M aqueous solution of sodium bicarbonate (100 mL). The organic phase was washed with a 1M aqueous solution of sodium bicarbonate (2×50 mL), a 1M aqueous solution of hydrochloric acid (3×50 mL) and brine. The organic phase was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 87%
  • Mass Spectral Analysis m/z=262.15 (M+H)+
  • Preparation of 34H
  • 34H was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1h (see also step 34.2).
  • 1H NMR (400 MHz, DMSO d6) δ 9.01 (brs, 2H), 7.80 (s, 1H), 7.41 (s, 1H), 7.27 (t, 1H), 7.19 (d, 1H), 7.04 (d, 1H), 6.99 (t, 1H), 6.04 (s, 1H), 3.48 (brm, 4H), 3.21 (brm, 4H), 2.02 (brm, 4H), 1.16 (brt, 6H)
  • Mass Spectral Analysis m/z=383.4 (M+H)+
  • Example 34I
  • 34I was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1i (see also step 34.2).
  • 34.1i was obtained according to a procedure similar to the one described for 34.1h except 1.12 was replaced by 3.4o in step 34.7.
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (brs, 2H), 7.73 (d, 1H), 7.27 (m, 2H), 7.21 (dd, 1H), 7.04 (d, 1H), 6.99 (t, 1H), 6.04 (s, 1H), 3.90 (brs, 2H), 3.21 (brm, 4H), 2.07 (brm, 2H), 1.98 (brm, 2H), 1.30 (brd, 12H)
  • Mass Spectral Analysis m/z=411.4 (M+H)+
  • Example 34J
  • 34J was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1j (see also step 34.2).
  • Note: 34.1j was obtained according to a procedure similar to the one described for 34.1k (see 34K) except 34.8b was replaced by 34.8a in step 34.9.
  • 1H NMR (400 MHz, DMSO d6) δ 8.85 (brs, 2H), 7.43 (t, 1H), 7.35 (d, 1H), 7.27 (m, 2H), 7.04 (m, 2H), 6.97 (m, 1H), 6.03 (s, 1H), 3.48 (q, 2H), 3.22 (brm, 6H), 2.04 (brm, 4H), 1.16 (t, 3H), 1.04 (t, 3H)
  • Mass Spectral Analysis m/z=395.0 (M+H)+
  • Elemental analysis:
  • C24H27FN2O2, 1HCl, 0.25H2O
  • Theory: % C, 66.20; % H, 6.60; % N, 6.43.
  • Found: % C, 65.97; % H, 6.48; % N, 6.21.
  • Example 34K Preparation of 34.1k
  • To a stirred solution of 34.8b (5.0 g, 22.83 mmol, 1.0 eq) in acetonitrile (50 mL) was added N,N-diisopropylethylamine (8.35 mL, 47.94 mmol, 2.1 eq), 1.12 (2.6 mL, 25.11 mmol, 1.1 eq) and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (8.06 g, 25.11 mmol, 1.1 eq). The reaction mixture was stirred at room temperature for 16 h. The mixture was concentrated under reduced pressure and the residue was dissolved in ethyl acetate. The mixture was washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 91%
  • 1H NMR (400 MHz, CDCl3) δ 7.30 (m, 2H), 7.03 (m, 1H), 3.53 (q, 2H), 3.24 (q, 2H), 1.27 (t, 3H), 1.13 (t, 3H)
  • Preparation of 34K
  • 34K was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1k (see also step 34.2).
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (brs, 2H), 7.29 (m, 3H), 7.13 (s, 1H), 7.05 (d, 1H), 6.98 (m, 2H), 6.01 (s, 1H), 3.43 (brm, 2H), 3.23 (brm, 6H), 2.04 (brm, 4H), 1.10 (brd, 6H)
  • Mass Spectral Analysis m/z=395.0 (M+H)+
  • Elemental analysis:
  • C24H27FN2O2, 1HCl, 0.25H2O
  • Theory: % C, 66.20; % H, 6.60; % N, 6.43.
  • Found: % C, 66.17; % H, 6.57; % N, 6.32.
  • Example 34L
  • 34L was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.11 (see also step 34.2).
  • Note: 34.11 was obtained according to a procedure similar to the one described for 34.1k except 34.8b was replaced by 34.8c in step 34.9.
  • 1H NMR (400 MHz, CDCl3) δ 9.76 (brs, 1H), 9.63 (brs, 1H), 7.20 (m, 4H), 7.05 (dd, 1H), 6.93 (m, 2H), 5.60 (s, 1H), 3.76 (brs, 2H), 3.42 (brm, 4H), 3.18 (q, 2H), 2.32 (s, 3H), 2.21 (brm, 4H), 1.28 (t, 3H), 1.08 (t, 3H)
  • Mass Spectral Analysis m/z=391.0 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl
  • Theory: % C, 70.32; % H, 7.32; % N, 6.56. Found: % C, 69.92; % H, 7.27; % N, 6.49.
  • Example 34M
  • 34M was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1m (see also step 34.2).
  • Note: 34.1m was obtained according to a procedure similar to the one described for 34.1k except 34.8b was replaced by 34.8d in step 34.9.
  • 1H NMR (400 MHz, CDCl3) δ 9.78 (brs, 1H), 9.62 (brs, 1H), 7.22 (m, 3H), 7.13 (d, 1H), 6.92 (d, 1H), 6.84 (t, 1H), 6.63 (dd, 1H), 5.48 (s, 1H), 3.42 (brm, 8H), 2.36 (brm, 2H), 2.21 (m, 2H), 2.13 (s, 3H), 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=391.0 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl
  • Theory: % C, 70.32; % H, 7.32; % N, 6.56.
  • Found: % C, 70.01; % H, 7.30; % N, 6.57.
  • Example 34N
  • 34N was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1n (see also step 34.2).
  • Note: 34.1n was obtained according to a procedure similar to the one described for 34.1k except 34.8b was replaced by 34.8e in step 34.9.
  • 1H NMR (400 MHz, CDCl3) δ 9.78 (brs, 1H), 9.68 (brs, 1H), 7.28 (m, 1H), 7.03 (dd, 1H), 6.95 (m, 4H), 5.64 (s, 1H), 3.62 (q, 2H), 3.41 (brm, 4H), 3.28 (q, 2H), 2.26 (brm, 4H), 1.28 (t, 3H), 1.05 (t, 3H)
  • Mass Spectral Analysis m/z=413.0 (M+H)+
  • Elemental analysis:
  • C24H26F2N2O2, 1HCl, 0.25H2O
  • Theory: % C, 63.57; % H, 6.11; % N, 6.18.
  • Found: % C, 63.54; % H, 6.09; % N, 6.20.
  • Example 34O
  • 34O was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1o (see also step 34.2).
  • Note: 34.1o was obtained according to a procedure similar to the one described for 34.1k except 34.8b was replaced by 34.8f in step 34.9.
  • 1H NMR (400 MHz, CDCl3) δ 9.78 (brs, 1H), 9.66 (brs, 1h), 7.38 (s, 1H), 7.33 (d, 1H), 7.25 (m, 2H), 7.02 (d, 1H), 6.95 (m, 2H), 5.63 (s, 1H), 3.81 (brs, 1H), 3.42 (brm 5H), 3.21 (brm, 2H), 2.26 (brm, 4H), 1.28 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=411.0 (M+H)+
  • Elemental analysis:
  • C24H27ClN2O2, 1HCl
  • Theory: % C, 64.43; % H, 6.31; % N, 6.26.
  • Found: % C, 64.34; % H, 6.35; % N, 6.28.
  • Example 34P
  • 34P was obtained according to a procedure similar to the one described for 32A, with the following exceptions:
  • Step 32.2: 32.2a was replaced by 34.1p (see also step 34.2).
  • Note: 34.1p was obtained according to a procedure similar to the one described for 34.1k except 34.8b was replaced by 34.9 in step 34.9 (see also step 34.10).
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (brs, 2H), 7.47 (m, 2H), 7.34 (m, 1H), 7.27 (m, 1H), 7.20 (m, 1H), 6.98 (m, 1H), 6.87 (m, 1H), 6.76 (m, 1H), 5.69 (s, 1H), 3.29 (m, 2H), 3.18 (m, 4H), 3.01 (m, 2H), 2.04 (m, 2H), 1.93 (m, 2H), 0.96 (m, 6H)
  • Mass Spectral Analysis m/z=377.4 (M+H)+
  • Example 35A Preparation of 35.2
  • To a solution of 35.1 (41.44 g, 0.3 mol, 1.0 eq) in ammonium hydroxide (105 mL, 30% solution in H2O) was added drop wise a solution of I2 (61.23 g, 0.24 mol, 0.8 eq) and KI (47.71 g, 0.287 mol, 0.96 eq) in water (300 mL) over a 20 min period. The mixture was stirred at room temperature for 1 h, and the mixture was concentrated under reduced pressure to half of its volume. The pH was adjusted to 3-4 with a 6N aqueous solution of hydrochloric acid. The white solid was collected by filtration and washed by a small amount of water. The solid was re-crystallized from water/EtOH (2:1), and dried under high vacuum. Yield: 22%
  • 1H NMR (400 MHz, DMSO-d6) δ 12.96 (b, 1H), 10.70 (s, 1H), 7.80 (d, 1H), 7.42 (s, 1H), 7.12 (d, 1H)
  • Preparation of 35.3
  • To an acidic methanolic solution, which was prepared by drop wise addition of acetyl chloride (0.5 mL) to anhydrous methanol (75 mL) was added 35.2 (20.0 g, 75.8 mmol). The mixture was heated to reflux for 18 h. The reaction mixture was allowed to cool to room temperature, and was concentrated under reduced pressure. The residue was diluted in ethyl acetate (100 mL), washed by water (100 mL), brine (100 mL), dried over Na2SO4. The solution was filtered and the filtrate was concentrated under reduced pressure. The crude product was dried under vacuum.
  • Yield: 92%
  • 1H NMR (400 MHz, DMSO-d6) δ10.79 (s, 1H), 7.85 (d, 1H), 7.46 (s, 1H), 7.15 (d, 1H), 3.84 (s, 3H)
  • Preparation of 35.4
  • A mixture of 35.3 (2.0 g, 7.19 mmol, 1.0 eq), 2.8c (4.08 g, 28.8 mmol, 4.0 eq) and potassium carbonate (9.94 g, 71.9 mmol, 10.0 eq) in acetone (100 mL) was refluxed for 16 h. The reaction was cooled to room temperature and the solid was collected by filtration. The volume of the filtrate was reduced to 15 mL and this solution was taken on to the next step without further purification.
  • Preparation of 35.5
  • To a solution of 35.4 (2.10 g, 7.19 mmol, 1.0 eq) in acetone (15 mL) was added lithium hydroxide (1.2 g, 28.8 mmol, 4.0 eq) and a 1:1 tetrahydrofuran/water solution (30 mL). The mixture was stirred at room temperature for 16 h. The mixture was reduced to half of its volume under reduced pressure and was acidified with a a 6N aqueous solution of hydrochloric acid (5 mL). The crude mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over magnesium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • 1H NMR (400 MHz, CDCl3) δ 7.91 (d, 1H), 7.49 (d, 1H), 7.45 (dd, 1H), 3.96 (s, 3H)
  • Preparation of 35.6
  • To a mixture of 35.5 (2.0 g, 7.19 mmol, 1.0 eq) and O-Benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (2.54 g, 7.91 mmol, 1.1 eq) in acetonitrile (75 mL) at 0° C. was added 1.12 (0.58 g, 7.91 mmol, 1.1 eq) and N,N-diisopropylethylamine (1.95 g, 15.1 mmol, 2.1 eq). The mixture was warmed to room temperature, stirred for 16 h at room temperature and concentrated under reduced pressure. The crude mixture was dissolved in ethyl acetate. The mixture was washed with a saturated aqueous solution of sodium bicarbonate, dried over magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixture, 60:40). Yield: 96%
  • 1H NMR (400 MHz, CDCl3) δ 7.78 (d, 1H), 6.84 (d, 1H), 6.70 (dd, 1H), 3.90 (s, 3H), 3.54 (brs, 2H), 3.26 (brs, 2H), 1.19 (brd, 6H)
  • Mass Spectral Analysis m/z=334.1 (M+H)+
  • Preparation of 35.9
  • To a solution of 35.6 (1.34 g, 4.02 mmol, 1.0 eq) in dimethoxyethane (DME) (20 mL) was added sequentially a 2N aqueous solution of sodium carbonate (6.03 mL, 12.06 mmol, 3.0 eq), lithium chloride (0.511 g, 12.06 mmol, 3.0 eq), 32.1 (2.06 g, 4.83 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (0.232 g, 0.20 mmol, 0.05 eq). The Suzuki coupling reaction was conducted under microwave conditions (A. 25° C. to 170° C. for 10 min; B. 170° C. for 7 min). The crude mixture was dissolved in ethyl acetate, washed with water, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 74%
  • 1H NMR (400 MHz, CDCl3) δ 7.18 (d, 1H), 7.13 (m, 1H), 6.98 (m, 2H), 6.90 (d, 1H), 6.79 (m, 1H), 6.70 (dd, 1H), 5.53 (s, 1H), 3.84 (brs, 2H), 3.72 (s, 3H), 3.56 (brs, 2H), 3.33 (brs, 4H), 2.07 (brm, 2H), 1.67 (brm, 2H), 1.47 (s, 9H), 1.22 (brd, 6H)
  • Mass Spectral Analysis m/z=507.3 (M+H)+
  • Preparation of 35A
  • Compound 35.9 (1.50 g, 2.96 mmol, 1.0 eq) was dissolved in a 4.0M anhydrous solution of hydrochloric acid in dioxane (15 mL, 60 mmol, 20 eq) and the mixture was stirred at room temperature for 16 h. The mixture was concentrated under reduced pressure. The residue was dissolved in a minimum amount (until complete dissolution of the product) of methylene chloride, and ethyl acetate was added until the solution became cloudy. The mixture was stirred for 2 h at room temperature. The resulting precipitate was collected by filtration. Yield: 77%
  • 1H NMR (400 MHz, CDCl3) δ 9.75 (brs, 1H), 9.58 (brs, 1H), 7.16 (m, 2H), 6.98 (m, 2H), 6.90 (d, 1H), 6.83 (m, 1H), 6.72 (dd, 1H), 5.56 (s, 1H), 3.72 (s, 3H), 3.50 (brm, 8H), 2.35 (brm, 2H), 2.16 (brm, 2H), 1.23 (brd, 6H)
  • Mass Spectral Analysis m/z=407.0 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 0.5H2O
  • Theory: % C, 66.43; % H, 7.14; % N, 6.20.
  • Found: % C, 66.28; % H, 7.10; % N, 5.94.
  • Example 35B Preparation of 35.7
  • To a solution of 35.6 (1.10 g, 3.30 mmol, 1.0 eq) in methylene chloride (30 mL) at 0° C. was added a 1.0M solution of boron tribromide in methylene chloride (5.0 mL, 5.0 mmol, 1.5 eq). The reaction was warmed to room temperature and stirred for 16 h at room temperature. A saturated aqueous solution of sodium bicarbonate was added to the mixture and the crude product was extracted with methylene chloride. The combined organic extracts were dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 87%
  • 1H NMR (400 MHz, CDCl3) δ 8.28 (brs, 1H), 7.64 (d, 1H), 6.95 (d, 1H), 6.56 (dd, 1H), 3.54 (q, 2H), 3.25 (q, 2H), 1.24 (t, 3H), 1.10 (t, 3H)
  • Mass Spectral Analysis m/z=320.0 (M+H)+
  • Preparation of 35.8
  • To a solution of 35.7 (0.90 g, 2.82 mmol, 1.0 eq) and N,N-diisopropylethylamine (2.91 g, 22.6 mmol, 8.0 eq) in methylene chloride (25 mL) at 0° C. under nitrogen was added drop wise 11.3 (0.86 mL, 11.3 mmol, 4.0 eq). The mixture was warmed to room temperature and stirred for 48 h at room temperature. The mixture was concentrated under reduced pressure, dissolved in ethyl acetate and the solution was washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • Mass Spectral Analysis m/z=364.1 (M+H)+
  • Preparation of 35.10
  • To a solution of 35.8 (1.02 g, 2.82 mmol, 1.0 eq) in dimethoxyethane (DME) (20 mL) was added sequentially a 2N aqueous solution of sodium carbonate (4.23 mL, 8.46 mmol, 3.0 eq), lithium chloride (0.359 g, 8.46 mmol, 3.0 eq), 32.1 (1.44 g, 3.38 mmol, 1.2 eq) and palladium on carbon (10%, 50% water) (0.038 g, 0.007 mmol, 0.0025 eq). The reaction was conducted under microwave conditions (A. 25° C. to 170° C. for 10 min; B. 170° C. for 7 min). The mixture was dissolved in ethyl acetate, washed with water, dried over sodium sulfate. The mixture was filtered and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 50%
  • 1H NMR (400 MHz, CDCl3) δ 7.21 (m, 2H), 7.13 (m, 1H), 7.06 (dd, 1H), 6.90 (d, 1H), 6.76 (m, 2H), 5.53 (s, 1H), 5.04 (s, 2H), 3.87 (brs, 2H), 3.55 (brs, 2H), 3.34 (brs, 4H), 3.30 (s, 3H), 2.08 (brm, 2H), 1.67 (brm, 2H), 1.48 (s, 9H), 1.24 (brm, 6H)
  • Mass Spectral Analysis m/z=537.3 (M+H)+
  • Preparation of 35B
  • To a solution of 35.10 (0.647 g, 1.21 mmole, 1 eq) in methanol (3 mL) was added an excess of a 4.0M solution of anhydrous hydrochloric acid in dioxane (20 mL). The mixture was stirred at room temperature for 16 h. The mixture was concentrated under reduced pressure and treated with a mixture of methylene chloride (15 mL) and ethyl acetate (25 mL). The resulting precipitate was collected by filtration and dried under vacuum. Yield: 77%
  • 1H NMR (400 MHz, DMSO d6) δ 9.75 (s, 1H), 8.84 (brm, 2H), 7.16 (m, 2H), 6.96 (d, 1H), 6.84 (m, 3H), 6.72 (d, 1H), 5.78 (s, 1H), 3.42 (brs, 2H), 3.22 (brs, 6H), 2.10 (brm, 2H), 1.96 (brm, 2H), 1.12 (brs, 6H)
  • Mass Spectral Analysis m/z=393.3 (M+H)+
  • Example 36A Preparation of 36.3
  • To a mixture of copper (II) bromide (8.8 g, 39.4 mmol, 1.2 eq) in acetonitrile (50 mL) under a nitrogen atmosphere was added 36.2 (5.1 g, 49.5 mmol, 1.5 eq). The mixture was cooled to 0° C. and 36.1 (5.0 g, 32.6 mmol, 1.0 eq) was added in small portions. Additional amount of acetonitrile (25 mL) was added to the mixture, which was stirred at 0° C. for 2 h. The mixture was poured onto a 20% aqueous solution of hydrochloric acid (200 mL) and extracted with diethyl ether. The combined organic extracts were washed with a 20% aqueous solution of hydrochloric acid, dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The residue was dissolved in diethyl ether. The mixture was extracted with a 15% aqueous solution of sodium hydroxide. The aqueous portion was washed with diethyl ether, acidified to pH 1 with a 6N aqueous solution of hydrochloric acid and the mixture was extracted with diethyl ether. The combined organic extracts were washed with brine, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The residue was treated with chloroform and the resulting precipitate was collected by filtration. The product was used for the next step without further purification.
  • Mass Spectral Analysis m/z=215.1 (M−H)
  • Preparation of 36.4
  • To a mixture of 1.12 (0.85 g, 11.58 mmol, 2.5 eq), O-Benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (1.93 g, 6.02 mmol, 1.3 eq) and N,N-diisopropylethylamine (1.25 g, 9.72 mmol, 2.1 eq) in acetonitrile (50 mL) at 0° C. was added drop wise a solution of 36.3 (1.0 g, 4.63 mmol, 1.0 eq) in acetonitrile (10 mL). The mixture was warmed to room temperature and stirred for 48 h at room temperature. An additional portion of TBTU (1.04 g, 3.24 mmol, 0.7 eq) was added to the mixture which was heated at 60° C. for 5 h. The mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate. The mixture was washed by water, brine, dried over magnesium sulfate and filtrate. The solution was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 63%
  • 1H NMR (400 MHz, CDCl3) δ 10.08 (s, 1H), 7.17 (d, 1H), 7.12 (d, 1H), 6.98 (dd, 1H), 3.50 (q, 4H), 1.27 (t, 6H)
  • Mass Spectral Analysis m/z=270.1 (M−H)
  • Preparation of 36.5
  • To a solution of 36.4 (0.30 g, 1.11 mmol, 1.0 eq) in dimethoxyethane (DME) (10 mL) was added sequentially a 2N aqueous solution of sodium carbonate (1.66 mL, 3.32 mmol, 3.0 eq), lithium chloride (0.141 g, 3.32 mmol, 3.0 eq), 32.1 (0.57 g, 1.33 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (0.128 g, 0.11 mmol, 0.1 eq). The reaction was conducted under microwave conditions (A. 25° C. to 170° C. for 10 min; B. 170° C. for 10 min). The crude mixture was dissolved in ethyl acetate. The mixture was washed with a 0.5N aqueous solution of hydrochloric acid, brine, and dried over magnesium sulfate. The mixture was filtered and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 37%
  • 1H NMR (400 MHz, CDCl3) δ 9.94 (s, 1H), 7.29 (d, 1H), 7.18 (m, 1H), 7.06 (dd, 1H), 7.00 (d, 1H), 6.94 (d, 1H), 6.85 (m, 2H), 5.59 (s, 1H), 3.85 (brs, 2H), 3.55 (q, 4H), 3.34 (brs, 2H), 2.04 (brm, 2H), 1.66 (m, 2H), 1.48 (s, 9H), 1.30 (t, 6H)
  • Mass Spectral Analysis m/z=493.2 (M+H)+
  • Preparation of 36A
  • To a solution of 36.5 (0.20 g, 0.406 mmol, 1.0 eq) in methylene chloride (2 mL) was added a 1.0M solution of anhydrous hydrochloric acid in diethyl ether (10 mL, 10 mmol, 25 eq). The mixture was stirred at room temperature for 16 h. The mixture was concentrated under reduced pressure and treated with diethyl ether. The resulting precipitate was collected by filtration. By LC/MS some starting material remained; therefore, so the precipitate was treated with an excess of a 4.0M solution of anhydrous hydrochloric acid in dioxane. This mixture was stirred at room temperature for 16 h. The mixture was concentrated under reduced pressure and the crude product was purified by column chromatography (eluent: methylene chloride/methanol mixtures of increasing polarity). Yield: 66%
  • 1H NMR (400 MHz, DMSO d6) δ 9.91 (brs, 1H), 9.08 (brs, 2H), 7.26 (m, 1H), 7.13 (d, 1H), 7.04 (m, 2H), 6.95 (m, 1H), 6.84 (m, 2H), 5.87 (s, 1H), 3.66 (brs, 4H), 3.20 (brm, 4H), 2.05 (brm, 4H), 1.08 (brd, 6H)
  • Mass Spectral Analysis m/z=393.4 (M+H)+
  • Elemental analysis:
  • C24H28N2O3, 1HCl, 1.5H2O
  • Theory: % C, 63.22; % H, 7.07; % N, 6.14.
  • Found: % C, 63.45; % H, 6.88; % N, 6.09.
  • Example 36B Preparation of 36.8
  • To a solution of 36.6 (13.0 mL, 89.41 mmol, 1.0 eq) and triethylamine (13.71 mL, 98.35 mmol, 1.1 eq) in methylene chloride (100 mL) at 0° C. under a nitrogen atmosphere was added drop wise ethyl chloroformate (9.40 mL, 98.35 mmol, 1.1 eq). The mixture was warmed to room temperature and stirred for 1 h at room temperature. Water and methylene chloride were added to the reaction mixture and the layers were separated. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.22 (t, 1H), 6.76 (m, 3H), 4.66 (brs, 1H), 4.11 (q, 2H), 3.80 (s, 3H), 3.43 (m, 2H), 2.78 (m, 2H), 1.23 (t, 3H)
  • Mass Spectral Analysis m/z=224.1 (M+H)+
  • Preparation of 36.9
  • A mixture of 36.8 (20 g, 89.58 mmol, 1.0 eq) and polyphosphoric acid (90 g) was heated at 120° C. under a nitrogen atmosphere for 1.5 h. The mixture was cooled to room temperature. Water (200 mL) was added to the mixture which was extracted with ethyl acetate. The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: ethyl acetate). Polyphosphoric acid was still present in the purified sample; therefore the residue was dissolved in ethyl acetate and the solution was washed with a saturated aqueous solution of sodium bicarbonate. The mixture was dried over sodium sulfate, filtered and concentrated under reduced pressure. Ethyl acetate (15 mL) was added to the mixture. The resulting precipitate was collected by filtration and was used for the next step without further purification.
  • Yield: 30%
  • 1H NMR (400 MHz, CDCl3) δ 8.02 (d, 1H), 6.86 (dd, 1H), 6.71 (d, 1H), 6.22 (brs, 1H), 3.85 (s, 3H), 3.55 (m, 2H), 2.97 (t, 2H)
  • Mass Spectral Analysis m/z=178.1 (M+H)+
  • Preparation of 36.11
  • To a suspension of NaH (0.81 g, 33.86 mmol, 6.0 eq) in tetrahydrofuran (30 mL) under a nitrogen atmosphere was added drop wise a solution of 36.9 (1.0 g, 5.64 mmol, 1.0 eq) in tetrahydrofuran (15 mL). To this mixture was added drop wise 36.10 (2.28 mL, 28.22 mmol, 5.0 eq) and stirring was continued for 16 h at room temperature. A thick precipitate formed; therefore additional amount of tetrahydrofuran (15 mL) and 36.10 (1.0 mL, 12.39 mmol, 2.2 eq) were added and stirring was continued for an additional 24 h at room temperature. The reaction was quenched by addition of a 1N aqueous solution of hydrochloric acid followed by ethyl acetate and water. The layers were separated. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 83%.
  • 1H NMR (400 MHz, CDCl3) δ 8.03 (d, 1H), 6.84 (dd, 1H), 6.65 (d, 1H), 3.84 (s, 3H), 3.61 (q, 2H), 3.53 (t, 2H), 2.95 (t, 2H), 1.21 (t, 3H)
  • Mass Spectral Analysis m/z=206.1 (M+H)+
  • Preparation of 36.12
  • To a solution of 36.11 (0.96 g, 4.68 mmol, 1.0 eq) in anhydrous methylene chloride (30 mL) at −78° C. under a nitrogen atmosphere was added drop wise a 1.0M solution of boron tribromide in methylene chloride (9.35 mL, 9.35 mmol, 2.0 eq). The reaction was warmed to room temperature and stirred for 16 h at room temperature. The mixture was cooled in an ice bath, quenched with methanol (10 mL) and concentrated under reduced pressure. The crude mixture was dissolved in ethyl acetate and the solution was washed with a 1N aqueous solution of hydrochloric acid and then brine. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude solid was triturated in a ethyl acetate/hexane (1:1). The precipitate was collected by filtration. Yield: 74%.
  • 1H NMR (400 MHz, CDCl3) δ 7.89 (d, 1H), 6.82 (dd, 1H), 6.68 (d, 1H), 3.63 (q, 2H), 3.54 (t, 2H), 2.91 (t, 2H), 1.22 (t, 3H)
  • Mass Spectral Analysis m/z=192.1 (M+H)+
  • Preparation of 36.14
  • To a solution of 36.12 (0.38 g, 1.99 mmol, 1.0 eq) and pyridine (0.32 mL, 3.98 mmol, 2.0 eq) in methylene chloride (10 mL) at 0° C. under a nitrogen atmosphere was added 36.13 (0.40 mL, 2.38 mmol, 1.2 eq). The reaction was warmed to room temperature and stirred for 2 h at room temperature. Methylene chloride was added to the mixture which was washed with a 1N aqueous solution of hydrochloric acid, and with a 1N aqueous solution of sodium hydroxide. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1).
  • Yield: 45%.
  • 1H NMR (400 MHz, CDCl3) δ 8.18 (d, 1H), 7.23 (dd, 1H), 7.11 (d, 1H), 3.62 (m, 4H), 3.04 (t, 2H), 1.23 (t, 3H)
  • Mass Spectral Analysis m/z=324.1 (M+H)+
  • Preparation of 36.15
  • To a solution of 36.14 (0.100 g, 0.309 mmol, 1.0 eq) in N,N-dimethylformamide (5 mL) under a nitrogen atmosphere was added 32.1(0.145 g, 0.340 mmol, 1.1 eq), potassium acetate (0.091 g, 0.928 mmol, 3.0 eq) and [1,1′-bis(diphenylphosphino)ferrocene]palladium(II), dichloromethane complex (0.005 g, 0.006 mmol, 0.02 eq). The reaction was stirred at 65° C. for 16 h. The mixture was cooled to room temperature. Water was added and the mixture was extracted with ethyl acetate. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 45%.
  • 1H NMR (400 MHz, CDCl3) δ 8.11 (d, 1H), 7.31 (dd, 1H), 7.19 (m, 1H), 7.15 (s, 1H), 6.96 (m, 2H), 6.86 (m, 1H), 5.58 (s, 1H), 3.86 (brm, 2H), 3.65 (q, 2H), 3.59 (t, 2H), 3.34 (m, 2H), 3.01 (t, 2H), 2.05 (m, 2H), 1.67 (m, 2H), 1.48 (s, 9H), 1.26 (t, 3H)
  • Mass Spectral Analysis m/z=475.3 (M+H)+
  • Preparation of 36B
  • To a solution of 36.15 (0.150 g, 0.316 mmol, 1.0 eq) in anhydrous methylene chloride (5 mL) at 0° C. under a nitrogen atmosphere was added a 1.0M solution of anhydrous hydrochloric acid in diethyl ether (1.26 mL, 1.26 mmol, 4.0 eq). The reaction was warmed to room temperature and stirred for 4 days at room temperature. Diethyl ether was added (5 mL) and the resulting precipitate was collected by filtration.
  • Yield: 27%.
  • 1H NMR (400 MHz, DMSO d6) δ 8.80 (brs, 2H), 7.92 (d, 1H), 7.29 (m, 3H), 7.05 (d, 1H), 6.97 (m, 2H), 5.94 (s, 1H), 3.54 (m, 4H), 3.23 (brm, 4H), 3.00 (t, 2H), 2.08 (brm, 2H), 1.97 (brm, 2H), 1.13 (t, 3H)
  • Mass Spectral Analysis m/z=375.3 (M+H)+
  • Elemental analysis:
  • C24H26N2O2, 1HCl, 1H2O
  • Theory: % C, 67.20; % H, 6.81; % N, 6.53.
  • Found: % C, 67.52; % H, 6.46; % N, 6.54.
  • Example 37A Preparation of 37.2 and 37.3
  • To a solution of 37.1 (5.0 g, 24.60 mmol, 1.0 eq) and 1.1a (2.56 mL, 24.60 mmol, 1.0 eq) in methanol (100 mL) was added pyrrolidine (5.53 mL, 66.90 mmol, 2.72 eq). The mixture was refluxed for 16 h. The mixture was concentrated under reduced pressure, dissolved in ethyl acetate and the mixture was washed with a 1N aqueous solution of sodium hydroxide and brine. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude mixture was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity) to give a mixture of 37.2/37.3 (1/1.7). Yield: 80% (37.2) 1H NMR (400 MHz, CDCl3) δ 7.82 (dd, 1H), 7.47 (m, 1H), 7.28 (m, 5H), 6.96 (m, 2H), 3.50 (q, 2H), 2.76 (q, 2H), 2.64 (brm, 1H), 2.40 (brm, 1H), 2.18 (brm, 2H), 2.00 (brm, 1H), 1.82 (brm, 1H), 1.70 (brm, 1H), 1.07 (brd, 3H)
  • Mass Spectral Analysis m/z=322.3 (M+H)+
  • (37.3) 1H NMR (400 MHz, CDCl3) δ 7.84 (dd, 1H), 7.48 (m, 1H), 7.29 (m, 5H), 6.98 (m, 2H), 3.51 (m, 2H), 3.15 (d, 1H), 2.65 (m, 1H), 2.55 (m, 1H), 2.34 (m, 2H), 2.24 (m, 1H), 2.15 (m, 1H), 1.91 (m, 1H), 1.56 (m, 1H), 1.02 (d, 3H)
  • Mass Spectral Analysis m/z=322.3 (M+H)+
  • Preparation of 37.4
  • To a solution of 37.2 (2.30 g, 7.16 mmol, 1.0 eq) in methanol (25 mL) was added 10% Pd/C (0.50 g). The mixture was shaken for 6 h under 55 psi of hydrogen. The mixture was filtered through celite, and concentrated under reduced pressure. The crude product was used for the next step without further purification.
  • Yield: 99%
  • 1H NMR (400 MHz, CDCl3) δ 7.83 (dd, 1H), 7.48 (m, 1H), 6.97 (m, 2H), 3.18 (dd, 1H), 3.02 (m, 1H), 2.77 (m, 2H), 2.55 (m, 1H), 2.06 (m, 1H), 1.80 (brm, 3H), 1.06 (d, 3H)
  • Mass Spectral Analysis m/z=232.3 (M+H)+
  • Preparation of 37.5
  • To a solution of 37.4 (1.65 g, 7.13 mmol, 1.0 eq) in tetrahydrofuran (50 mL) was added triethylamine (2.98 mL, 21.40 mmol, 3.0 eq) and 4.7 (1.87 g, 8.56 mmol, 1.2 eq). The mixture was stirred for 2 h at room temperature. Water (100 mL) was added and the crude mixture was extracted with ethyl acetate and washed with brine. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate, 70/30). Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.85 (dd, 1H), 7.50 (m, 1H), 6.99 (m, 2H), 3.80 (brs, 1H), 3.56 (brm, 2H), 3.30 (brs, 1H), 2.73 (m, 2H), 2.12 (brs, 1H), 1.82 (brm, 2H), 1.46 (s, 9H), 1.03 (d, 3H)
  • Mass Spectral Analysis m/z=332.3 (M+H)+
  • Preparation of 37.6
  • To a solution of 37.5 (2.70 g, 8.15 mmol, 1.0 eq) in tetrahydrofuran (20 mL) at −78° C. under a nitrogen atmosphere was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (9.78 mL, 9.78 mmol, 1.2 eq). The mixture was stirred for 45 min at −78° C. A solution of 1.4 (3.49 g, 9.78 mmol, 1.2 eq) in tetrahydrofuran (10 mL) was added drop wise to the mixture, which was warmed slowly to room temperature and stirred for 16 h at room temperature. The mixture was then poured into ice water. A 1N aqueous solution of hydrochloric was added and the crude mixture was extracted with ethyl acetate. The organic extracts were washed with a 1N aqueous solution of sodium hydroxide, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield:
  • 62%
  • 1H NMR (400 MHz, DMSO d6) δ 7.31 (m, 1H), 7.15 (m, 1H), 6.95 (m, 1H), 6.85 (m, 1H), 6.25 (s, 0.6H), 5.83 (s, 0.4H), 3.54 (brs, 2H), 3.19 (brm, 2H), 1.96 (brm, 2H), 1.55 (brm, 1H), 1.33 (s, 9H), 0.83 (d, 3H)
  • Mass Spectral Analysis m/z=464.2 (M+H)+
  • Preparation of 37.7
  • To a solution of 37.6 (1.17 g, 2.52 mmol, 1.0 eq) in dioxane (20 mL) was added sequentially 1.6 (0.61 g, 2.78 mmol, 1.1 eq), potassium phosphate (0.80 g, 3.79 mmol, 1.5 eq) and potassium bromide (0.33 g, 2.78 mmol, 1.1 eq). The mixture was placed under vacuum, flushed with nitrogen and then the process was repeated. Tetrakis(triphenylphosphine)palladium(0) (0.146 g, 0.13 mmol, 0.05 eq) was added and the mixture was heated at 100° C. for 16 h under a nitrogen atmosphere. The mixture was cooled to room temperature, dissolved in ethyl acetate and the mixture was washed with water. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 53%
  • 1H NMR (400 MHz, CDCl3) δ 7.42 (d, 2H), 7.37 (d, 2H), 7.18 (m, 1H), 6.99 (d, 1H), 6.92 (d, 1H), 6.84 (m, 1H), 5.70 (s, 1H), 3.65 (brm, 5H), 3.32 (brs, 3H), 2.15 (brs, 1H), 2.04 (m, 1H), 1.77 (brs, 1H), 1.48 (s, 9H), 1.22 (brd, 6H), 1.02 (d, 3H)
  • Mass Spectral Analysis m/z=491.5 (M+H)+
  • Preparation of 37 A
  • To a solution of 37.7 (0.65 g, 1.33 mmol, 1.0 eq) in anhydrous methylene chloride (10 mL) at 0° C. under a nitrogen atmosphere was added a 1.0M solution of anhydrous hydrochloric acid in diethyl ether (5.31 mL, 5.31 mmol, 4.0 eq). The mixture was warmed to room temperature and stirred for 5 days at room temperature. The mixture was concentrated under reduced pressure and dissolved in methylene chloride (5 mL). Diethyl ether (10 mL) was added drop wise to the mixture which was stirred forlh at room temperature. The resulting precipitate was collected by filtration and dried under vacuum. Yield: 82%
  • 1H NMR (400 MHz, DMSO d6) δ 9.46 (brm, 1.5H), 7.71 (d, 2H), 7.67 (d, 2H), 7.48 (m, 1H), 7.21 (m, 2H), 7.15 (m, 1H), 6.44 (s, 1H), 3.70 (brs, 2H), 3.42 (brm, 6H), 2.52 (brm, 1H), 2.44 (brm, 1H), 2.13 (brm, 1H), 1.36 (brd, 6H), 1.22 (d, 3H)
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.25H2O
  • Theory: % C, 69.59; % H, 7.36; % N, 6.49.
  • Found: % C, 69.29; % H, 7.28; % N, 6.40.
  • Example 37B
  • 37B was obtained according to a procedure similar to the one described for 37A, with the following exceptions:
  • Step 37.2: 37.2 was replaced by 37.3 (see also step 37.5).
  • 1H NMR (400 MHz, DMSO d6) δ 9.40 (brm, 1.5H), 7.66 (s, 4H) 7.48 (m, 1H), 7.27 (d, 1H), 7.21 (m, 1H), 7.15 (m, 1H), 6.03 (s, 1H), 3.69 (brs, 2H), 3.43 (brm, 4H), 3.24 (brm, 2H), 2.47 (brm, 1H), 2.35 (brm, 1H), 2.08 (brm, 1H), 1.37 (brd, 6H), 1.20 (d, 3H)
  • Mass Spectral Analysis m/z=391.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.25H2O
  • Theory: % C, 69.59; % H, 7.36; % N, 6.49.
  • Found: % C, 69.69; % H, 7.18; % N, 6.49.
  • Example 7D Preparation of 7.10a
  • 3.4k (0.10 mL, 1.20 mmol, 1.2 eq) was added drop wise at room temperature to a 1 neck 100 mL round bottom flask which was flame dried under N2 and contained a solution of 3.1a (625.0 mg, 1.00 mmol, 1 eq), tris(dibenzylideneacetone)dipalladium(0) (9.2 mg, 0.010 mmol, 0.01 eq), 7.9 (6.0 mg, 0.020 mmol, 0.02 eq) and potassium phosphate (297.3 mg, 1.40 mmol, 1.4 eq) in ethylene glycol dimethyl ether (5 mL). The solution was heated to 80° C. for 48 hours and then diluted with diethyl ether (90 mL) and filtered through a plug of celite. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 37%
  • 1H NMR (400 MHz, CDCl3) δ 7.41 (m, 4H), 7.16 (d, 1H), 6.85 (d, 1H), 6.44 (m, 1H), 5.86 (s, 1H), 3.71 (m, 2H), 3.44 (m, 2H), 3.21 (m, 4H), 3.05 (m, 4H), 1.87 (m, 4H), 1.66 (m, 2H), 1.41 (s, 9H), 1.37 (m, 2H), 1.11 (m, 6H)
  • Mass Spectral Analysis m/z=546.44 (M+H)+
  • Preparation of 7D
  • A 2M solution of hydrochloric acid in diethyl ether (1.8 mL, 3.53 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 7.10a (0.35 g, 0.64 mmol, 1 eq) in anhydrous dichloromethane (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 64 hours at room temperature. The solution was concentrated in vacuo. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity).
  • Yield: 20%
  • 1H NMR (400 MHz, DMSO d6) δ 9.10 (m, 2H), 7.44 (M, 4H), 7.04 (m, 1H), 6.00 (s, 1H), 4.30 (br s, 5H), 3.44 (br s, 3H), 3.22 (m, 8H), 1.99 (m, 7H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=446.40 (M+H)+
  • Example 7E Preparation of 7.10b
  • 3.4p (167.4 mg, 1.92 mmol, 1.2 eq) was added drop wise at room temperature to a 1 neck 100 mL round bottom flask which was flame dried under N2 and contained a solution of 3.1a (1.00 g, 1.60 mmol, 1 eq), tris(dibenzylideneacetone)dipalladium(0) (14.7 mg, 0.016 mmol, 0.01 eq), 7.9 (9.6 mg, 0.032 mmol, 0.02 eq) and potassium phosphate (475.7 mg, 2.24 mmol, 1.4 eq) in ethylene glycol dimethyl ether (10 mL). The solution was heated to 80° C. for 72 hours and then diluted with diethyl ether (90 mL) and filtered through a 1 inch plug of celite. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 30%
  • Mass Spectral Analysis m/z=561.76 (M+H)+
  • Preparation of 7E
  • A 2M solution of hydrochloric acid in diethyl ether (1.3 mL, 2.55 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 7.10b (0.26 g, 0.46 mmol, 1 eq) in anhydrous dichloromethane (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 20 hours at room temperature. The solution was concentrated in vacuo. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity).
  • Yield: 10%
  • 1H NMR (400 MHz, DMSO d6) δ 9.00 (m, 2H), 7.45 (m, 4H), 7.04 (m, 2H), 6.74 (m, 1H), 5.98 (s, 1H), 4.00 (br s, 5H), 3.74 (br s, 4H), 3.45 (br s, 2H), 3.22 (m, 4H), 3.03 (m, 2H), 2.02 (m, 4H), 1.15 (m, 6H)
  • Mass Spectral Analysis m/z=462.44 (M+H)+
  • Example 11G Preparation of 11.11
  • To a suspension of 11.2 (5.0 g, 15.00 mmol, 1.0 eq) and cesium carbonate (24.4 g, 75.00 mmol, 5.0 eq) in N,N-dimethylformamide (50 mL) under nitrogen was added 9.7 (7.91 mL, 75.00 mmol, 5.0 eq). Evolution of white smoke was observed. The reaction was heated at 90° C. for 16 h and then cooled to room temperature. Water was added and the product was extracted three times with diethyl ether. The combined organics were then washed three times with a 1N aqueous solution of sodium hydroxide to remove any unreacted starting material. The organic layer was concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 26%
  • 1HNMR (400 MHz, CDCl3) δ 7.44 (t, 1H), 6.91 (m, 1H), 6.80 (m, 1H), 6.57 (t, 1H, J=75, CF2H), 3.87 (brs, 2H), 3.21 (brt, 2H), 2.72 (s, 2H), 2.00 (brd, 2H), 1.62 (m, 2H), 1.46 (s, 9H)
  • Mass Spectral Analysis m/z=384.4 (M+H)+
  • Preparation of 11.12
  • To a solution of 11.11 (1.47 g, 3.83 mmol, 1.0 eq) in tetrahydrofuran (30 mL) at −78° C. under nitrogen was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (4.60 mL, 4.60 mmol, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (1.64 g, 4.60 mmol, 1.2 eq) in tetrahydrofuran (15 mL) was added drop wise to the mixture, which was warmed slowly to room temperature. Stirring was continued for a further 5 h at room temperature. Ice was added to the reaction and the mixture was stirred for 15 min Ethyl acetate and a 1N aqueous solution of sodium hydroxide were added and the layers were separated. The organics were washed again with a 1N aqueous solution of sodium hydroxide and concentrated. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 56%
  • 1HNMR (400 MHz, CDCl3) δ 7.24 (t, 1H), 6.82 (d, 2H), 6.57 (t, 1H, J=72, CF2H), 5.59 (s, 1H), 3.84 (brs, 2H), 3.26 (brs, 2H), 2.06 (brd, 2H), 1.69 (m, 2H), 1.46 (s, 9H)
  • Preparation of 11.13a (X═CH)
  • To a solution of 11.12 (0.50 g, 0.97 mmol, 1.0 eq) in dioxane (15 mL) was added 1.6 (0.24 g, 1.07 mmol, 1.1 eq), potassium phosphate (0.31 g, 1.46 mmol, 1.5 eq), potassium bromide (0.13 g, 1.07 mmol, 1.1 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (0.040 g, 0.049 mmol, 0.05 eq). The mixture was heated at 100° C. for 24 h and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The organics were washed with brine, concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 67%
  • 1HNMR (400 MHz, CDCl3) δ 7.33 (d, 2H), 7.23 (m, 3H), 6.90 (d, 1H), 6.67 (d, 1H), 6.00 (t, 1H, J=74, CF2H), 5.61 (s, 1H), 3.82 (brs, 2H), 3.55 (brs, 2H) 3.28 (brs, 4H), 2.01 (brd, 2H), 1.69 (m, 2H), 1.47 (s, 9H), 1.26 (brs, 3H), 1.11 (brs, 3H)
  • Mass Spectral Analysis m/z=543.8 (M+H)+
  • Preparation of 11G
  • To a solution of 11.13a (X═CH) (0.35 g, 0.645 mmol, 1.0 eq) in anhydrous methylene chloride (7 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.29 mL, 2.58 mmol, 4.0 eq). The reaction was warmed to room temperature and stirred for a further 16 h at room temperature. The reaction was concentrated and the resulting foam was sonicated in diethyl ether (10 mL) for 5 min, stirred for 1 h at room temperature, and the solids were collected by vacuum filtration. Yield: 87%.
  • 1H NMR (400 MHz, DMSO d6) δ 8.97 (brs, 2H), 7.35 (t, 1H), 7.28 (m, 4H), 7.02 (d, 1H), 6.81 (t, 1H, J=74, CF2H), 6.77 (d, 1H), 5.97 (s, 1H), 3.40 (brm, 2H), 3.20 (brm, 6H), 2.04 (brm, 4H), 1.14 (brs, 3H), 1.04 (brs, 3H)
  • Mass Spectral Analysis m/z=443.3 (M+H)+
  • Elemental analysis:
  • C25H28F2N2O3, 1HCl
  • Theory: % C, 62.69; % H, 6.10; % N, 5.85.
  • Found: % C, 62.39; % H, 6.01; % N, 5.77.
  • Example 11H Preparation of 11.13b (X═N)
  • To a solution of 11.12 (0.50 g, 0.97 mmol, 1.0 eq) in dioxane (15 mL) was added 1.7 (0.32 g, 1.07 mmol, 1.1 eq), potassium phosphate (0.31 g, 1.46 mmol, 1.5 eq), potassium bromide (0.13 g, 1.07 mmol, 1.1 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (0.040 g, 0.049 mmol, 0.05 eq). The mixture was heated at 100° C. for 24 h and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The organics were washed with brine, concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 77%
  • 1HNMR (400 MHz, CDCl3) δ 8.44 (s, 1H), 7.61 (dd, 1H), 7.54 (d, 1H), 7.22 (t, 1H), 6.91 (d, 1H), 6.67 (d, 1H), 6.06 (t, 1H, J=74, CF2H), 5.64 (s, 1H), 3.85 (brs, 2H), 3.58 (q, 2H) 3.40 (q, 2H), 3.30 (brs, 2H), 2.03 (brd, 2H), 1.72 (m, 2H), 1.48 (s, 9H), 1.28 (t, 3H), 1.15 (t, 3H)
  • Mass Spectral Analysis m/z=544.8 (M+H)+
  • Preparation of 11H
  • To a solution of 11.13b (X═N) (0.40 g, 0.736 mmol, 1.0 eq) in anhydrous methylene chloride (7 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (2.21 mL, 4.41 mmol, 6.0 eq). The reaction was warmed to room temperature and stirred for a further 16 h at room temperature. The reaction was concentrated and the resulting foam was sonicated in diethyl ether (10 mL) for 5 min, stirred for 1 h at room temperature, and the solids were collected by vacuum filtration. Yield: 84%.
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (brs, 2H), 8.45 (s, 1H), 7.75 (dd, 1H), 7.49 (d, 1H), 7.37 (t, 1H), 7.04 (d, 1H), 6.78 (d, 1H), 6.90 (t, 1H, J=74, CF2H), 6.12 (s, 1H), 3.45 (m, 2H), 3.21 (brm, 6H), 2.06 (brm, 4H), 1.16 (t, 3H), 1.06 (t, 3H)
  • Mass Spectral Analysis m/z=444.3 (M+H)+
  • Elemental analysis:
  • C24H27F2N3O3, 1.25HCl, 1H2O
  • Theory: % C, 56.85; % H, 6.01; % N, 8.29; % Cl, 8.74.
  • Found: % C, 56.93; % H, 6.01; % N, 8.23; % Cl, 8.84.
  • Example 11I Preparation of 11.14
  • To a solution of 32.2b (2.05 g, 7.02 mmol, 1.0 eq) in dioxane (30 mL) under nitrogen was added 1.14 (2.14 g, 8.42 mmol, 1.2 eq), potassium acetate (2.07 g, 21.05 mmol, 3.0 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (0.12 g, 0.14 mmol, 0.02 eq). The mixture was heated at 70° C. for 16 h and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The aqueous phase was washed with ethyl acetate and the organics were combined and concentrated. The crude product was purified by column chromatography (eluent: ethyl acetate/hexane=3:7). Yield: 87%
  • 1HNMR (400 MHz, CDCl3) δ 7.92 (d, 2H), 7.79 (d, 2H), 3.24 (q, 4H), 1.36 (s, 12H), 1.11 (t, 6H)
  • Mass Spectral Analysis m/z=340.3 (M+H)+
  • Preparation of 11.15
  • To a solution of 11.5 (0.70 g, 1.37 mmol, 1.0 eq) in dioxane (15 mL) was added 11.14 (0.513 g, 1.51 mmol, 1.1 eq), potassium phosphate (0.437 g, 2.06 mmol, 1.5 eq), potassium bromide (0.18 g, 1.51 mmol, 1.1 eq) and tetrakis(triphenylphosphine) palladium(0) (0.079 g, 0.069 mmol, 0.05 eq). The mixture was heated at 100° C. for 16 h and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The aqueous layer was washed with ethyl acetate and the organics were combined, concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 47%
  • 1HNMR (400 MHz, CDCl3) δ 7.74 (d, 2H), 7.36 (d, 2H), 7.16 (t, 1H), 6.70 (m, 2H), 5.56 (s, 1H), 4.65 (s, 2H), 3.81 (brs, 2H), 3.32 (brs, 2H), 3.25 (q, 4H), 3.13 (s, 3H), 2.02 (brd, 2H), 1.68 (m, 2H), 1.48 (s, 9H), 1.15 (t, 6H)
  • Mass Spectral Analysis m/z=573.4 (M+H)+
  • Preparation of 11I
  • To a solution of 11.15 (0.365 g, 0.637 mmol, 1.0 eq) in methanol (20 mL) under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (3.20 mL, 6.37 mmol, 10.0 eq). The reaction was stirred for 16 h at room temperature. The reaction was concentrated and the resulting solid was stirred in a methylene chloride (2 mL)/diethyl ether (15 mL) mixture for 30 min at room temperature. The solids were then collected by vacuum filtration. Yield: 79%
  • 1HNMR (400 MHz, DMSO d6) δ 9.60 (s, 1H), 8.83 (brs, 2H), 7.71 (d, 2H), 7.43 (d, 2H), 7.08 (t, 1H), 6.55 (d, 1H), 6.45 (d, 1H), 5.84 (s, 1H), 3.17 (brm, 8H), 2.00 (brm, 4H), 1.05 (t, 6H)
  • Mass Spectral Analysis m/z=429.3 (M+H)+
  • Elemental analysis:
  • C23H28N2O4S, 1HCl, 0.5H2O
  • Theory: % C, 58.28; % H, 6.38; % N, 5.91.
  • Found: % C, 58.29; % H, 6.20; % N, 5.78.
  • Example 22F Preparation of 22.7
  • To a suspension of 22.4 (0.8 g, 80% pure by LC, as of 1.37 mmol, 1 eq.) in ethanol (15 mL) was added sodium acetate (0.77 g, 9.38 mmol, 6.8 eq.) and iodoethane 22.6 (0.62 mL, 7.68 mmol, 5.6 eq.). The reaction mixture was heated under reflux for 10 h. Water (50 mL) was added to the reaction mixture and the organics were extracted with dichloromethane (3×75 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 89%
  • 1H NMR (400 MHz, CDCl3) δ 7.75 (m, 1H), 7.57 (m, 1H), 7.45 (m, 2H), 7.34 (m, 2H), 7.06 (d, 1H), 5.67 (d, 1H), 4.02-3.25 (m, 8H), 3.05 (q, 2H), 2.47-2.25 (m, 3H), 2.00-1.69 (m, 3H), 1.32-1.12 (m, 9H)
  • Mass Spectral Analysis m/z=579.26 (M+H)+
  • Preparation of 22F
  • To a solution of 22.7 (0.57 g, 0.98 mmol, 1 eq.) in a mixture methanol (30 mL) and water (10 mL) at 0° C. was added potassium carbonate (0.81 g, 5.88 mmol, 6 eq.) in one portion. The reaction mixture was slowly warmed up to room temperature and stirring was continued for 10 h at room temperature. The methanol was removed under reduced pressure and the organics were extracted with dichloromethane (3×50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was dissolved in anhydrous dichloromethane (10 mL). To this solution was added a 2M anhydrous solution of hydrogen chloride in diethyl ether (2 mL, 4 mmol, 4 eq.) dropwise at 0° C. The mixture was stirred for 1 h at room temperature and then concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). Yield: 63%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.19 (m, 2H), 7.75 (dd, 1H), 7.45 (m, 4H), 7.39 (d, 1H), 7.23 (d, 1H), 6.07 (s, 1H), 3.53-3.40 (m, 2H), 3.31-3.04 (m, 8H), 2.31 (m, 2H), 2.18 (m, 1H), 2.01 (m, 2H), 1.82 (m, 1H), 1.22-1.02 (m, 9H)
  • Mass Spectral Analysis m/z=483.2 (M+H)+
  • Elemental analysis:
  • C27H34N2O4S, 1HCl, 1.5H2O
  • Theory: % C, 59.38; % H, 7.01; % N, 5.13.
  • Found: % C, 59.26; % H, 6.64; % N, 5.15.
  • [α]D 25=−3.85 (c=10.25 mg/mL, MeOH)
  • Example 33M
  • To a solution of 32.1 (1.0 g, 2.34 mmol, 1.0 eq) in dioxane (40 mL) under nitrogen was added 33.1l (0.51 g, 2.57 mmol, 1.1 eq), potassium phosphate (0.75 g, 3.51 mmol, 1.5 eq), potassium bromide (0.31 g, 2.57 mmol, 1.1 eq) and tetrakis(triphenylphosphine) palladium(0) (0.14 g, 0.12 mmol, 0.05 eq). The mixture was heated at 100° C. for 16 h. Additional quantity of tetrakis(triphenylphosphine) palladium(0) (0.10 g, 0.087 mmol, 0.04 eq) was added to the reaction mixture, which was heated at 100° C. for an additional 24 h and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The organics were concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 65%
  • 1HNMR (400 MHz, CDCl3) δ 7.99 (d, 2H), 7.44 (d, 2H), 7.20 (m, 1H), 6.96 (m, 2H), 6.86 (m, 1H), 5.60 (s, 1H), 3.88 (brs, 2H), 3.34 (brs, 2H), 2.64 (s, 3H), 2.06 (brd, 2H), 1.68 (m, 2H), 1.48 (s, 9H)
  • Mass Spectral Analysis m/z=420.2 (M+H)+
  • Preparation of 33M
  • To a solution of the Boc derivative, obtained previously, (0.63 g, 1.50 mmol, 1.0 eq) in methylene chloride (30 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (3.00 mL, 6.00 mmol, 4.0 eq). The reaction mixture was stirred for 48 h at room temperature and the resulting precipitate was collected by vacuum filtration. A saturated aqueous solution of sodium bicarbonate was added to a suspension of the resulting solid in ethyl acetate. The organic layer was separated and concentrated under vacuum. The crude product was further purified by column chromatography [eluent: methanol/(methylene chloride/ammonium hydroxide=99:1) mixtures of increasing polarity]. Yield: 61%
  • 1HNMR (400 MHz, DMSO d6) δ 8.02 (d, 2H), 7.49 (d, 2H), 7.22 (m, 1H), 6.96 (d, 1H), 6.89 (m, 2H), 5.91 (s, 1H), 2.91 (m, 2H), 2.76 (m, 2H), 2.62 (s, 3H), 1.82 (m, 2H), 1.71 (m, 2H)
  • Mass Spectral Analysis m/z=320.1 (M+H)+
  • Elemental analysis:
  • C21H21NO2, 0.6H2O
  • Theory: % C, 76.38; % H, 6.78; % N, 4.24.
  • Found: % C, 76.33; % H, 6.73; % N, 4.33.
  • Example 33N
  • To a solution of 32.1 (1.0 g, 2.34 mmol, 1.0 eq) in dioxane (40 mL) under nitrogen was added 33.1m (0.67 g, 2.57 mmol, 1.1 eq), potassium phosphate (0.75 g, 3.51 mmol, 1.5 eq), potassium bromide (0.31 g, 2.57 mmol, 1.1 eq) and tetrakis(triphenylphosphine) palladium(0) (0.14 g, 0.12 mmol, 0.05 eq). The mixture was heated at 100° C. for 16 h. Additional quantity of tetrakis(triphenylphosphine) palladium(0) (0.10 g, 0.087 mmol, 0.04 eq) was added and the reaction was heated at 100° C. for an additional 24 h and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The organics were concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 64%
  • 1HNMR (400 MHz, CDCl3) δ 7.84 (m, 4H), 7.61 (m, 1H), 7.49 (m, 4H), 7.21 (m, 1H), 7.02 (dd, 1H), 6.96 (dd, 1H), 6.88 (m, 1H), 5.64 (s, 1H), 3.88 (brs, 2H), 3.35 (brs, 2H), 2.08 (brd, 2H), 1.70 (m, 2H), 1.49 (s, 9H)
  • Mass Spectral Analysis m/z=482.2 (M+H)+
  • Preparation of 33N
  • To a solution of the Boc derivative obtained previously (0.715 g, 1.48 mmol, 1.0 eq) in methylene chloride (30 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (2.97 mL, 5.94 mmol, 4.0 eq). The reaction mixture was stirred for 48 h at room temperature and diluted with diethyl ether (5 mL). The precipitate was sonicated and collected by vacuum filtration. Yield: 95% 1HNMR (400 MHz, DMSO d6) δ 8.75 (brs, 2H), 7.80 (m, 4H), 7.71 (m, 1H), 7.59 (m, 4H), 7.28 (m, 1H), 7.05 (m, 2H), 6.97 (m, 1H), 6.02 (s, 1H), 3.20 (brm, 4H), 2.06 (brm, 4H)
  • Mass Spectral Analysis m/z=382.4 (M+H)+
  • Elemental analysis:
  • C26H23NO2, 1HCl, 0.5H2O
  • Theory: % C, 73.14; % H, 5.90; % N, 3.28.
  • Found: % C, 72.95; % H, 5.75; % N, 3.32.
  • Examples 38A, 38B, 38C, 38D Preparation of 38.2
  • To a mixture of 19.1 (29.8 g, 127.7 mmol, 1 eq.) and 38.1 (18.4 g, 127.7 mmol, 1 eq.) was added pyridine (12.5 mL) followed by 10 drops of piperidine. The mixture was stirred at 45° C. for 1 h and then stood at room temperature for 10 h. The resulting solids were washed with methanol and then dried in vacuo. Yield: 82%
  • 1H NMR (400 MHz, CDCl3) δ 7.41-7.30 (m, 5H), 5.17 (s, 2H), 3.70 (m, 4H), 3.18 (m, 4H), 1.75 (s, 6H)
  • Mass Spectral Analysis m/z=360.22 (M+H)+
  • Preparation of 38.4
  • To a suspension of copper(I) iodide (0.636 g, 3.34 mmol, 0.04 eq.) in dry tetrahydrofuran (200 mL) at −10° C. was added 38.3 (500 mL, 0.25M in THF, 125 mmol, 1.5 eq.) over a 50 min time period. The mixture was kept at −10° C. for another 15 min and then solid 38.2 (30 g, 83.48 mmol, 1 eq.) was added in 10 portions waiting for the exotherm subside between additions. The reaction mixture was kept at −10° C. for another 2 h and then slowly added to a mixture of 13% ammonium hydroxide/saturated ammonium chloride/water (1/1/1, 200 mL) and ethyl acetate (300 mL). The mixture was stirred for 15 min and then the two layers were separated. The organic layer was washed with 13% ammonium hydroxide/saturated ammonium chloride/water (1/1/1, 2×200 mL), brine (3×200 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. Diethyl ether (800 mL) was added to the crude mixture and the suspension was stirred at room temperature for 10 h. The resulting fine powders were collected by filtration, washed with diethyl ether (3×50 mL) and dried in vacuo. Yield: 100%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.37-7.25 (m, 5H), 7.05-6.92 (m, 4H), 4.99 (s, 2H), 3.70 (m, 2H), 2.94 (m, 2H), 2.91-2.66 (m, 4H), 1.45 (s, 6H), 0.84 (m, 2H)
  • Mass Spectral Analysis m/z=468.25 (M−H)
  • Preparation of 38.5
  • A mixture of 38.4 (41 g, 83.48 mmol, 1 eq.) in N,N-dimethylformamide/water (1:1, 400 mL) was stirred at 120° C. for 10 h. The reaction mixture was cooled to room temperature and was acidified with 1N aqueous hydrochloric acid until pH 1-2. The mixture was stirred at room temperature for 15 min and the resulting solids were collected by filtration, washed with water (2×50 mL), and then dried in vacuum oven at 70° C. Yield: 95%
  • 1H NMR (400 MHz, CDCl3) δ 7.41-7.28 (m, 5H), 7.24 (m, 2H), 7.11 (m, 2H), 5.05 (s, 2H), 3.69-3.56 (m, 2H), 3.41-3.20 (m, 2H), 2.76 (s, 2H), 2.17 (s, 2H), 1.53-1.32 (m, 4H)
  • Mass Spectral Analysis m/z=384.35 (M−H)
  • Preparation of 38.6
  • To a stirred solution of 38.5 (20 g, 51.9 mmol, 1 eq.) in dry dichloromethane (300 mL) was added oxalyl chloride (27.2 mL, 311.4 mmol, 6 eq.) in one portion followed by 5 drops of N,N-dimethylformamide. The reaction mixture was stirred at room temperature for 1 h and then concentrated under reduced pressure. The crude mixture was further dried in vacuo for 4 h and then dissolved in dry dichloromethane (600 mL), to which was added anhydrous aluminum chloride (13.84 g, 103.8 mmol, 2 eq.) in one portion. The reaction mixture was stirred at room temperature for 10 h. The reaction was quenched with water at 0° C. and the mixture was basified with concentrated ammonium hydroxide until pH 8-9. The two phases were separated and the aqueous phase was extracted with dichloromethane (3×100 mL). The combined organics were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude material was dissolved in dichloromethane (300 mL), to which were added triethylamine (21.7 mL, 155.7 mmol, 3 eq.) and 4.7 (13.6 g, 62.28 mmol, 1.2 eq.) portionwise at 0° C. The reaction mixture was slowly warmed up to and stirred at room temperature for 10 h. Dichloromethane was removed under reduced pressure. The crude product was dissolved in ethyl acetate (500 mL) and washed with 0.5 N aqueous hydrochloric acid (2×100 mL), water (2×200 mL), brine (200 mL), and dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixture of increasing polarity). Yield: 62%
  • 1H NMR (400 MHz, CDCl3) δ 7.67 (dd, 1H), 7.25-7.17 (M, 2H), 3.50-3.35 (m, 4H), 2.92 (s, 2H), 2.62 (s, 2H), 1.50 (m, 4H), 1.45 (s, 9H)
  • Mass Spectral Analysis m/z=334.23 (M+H)+
  • Preparation of 38.7
  • To a stirred solution of 38.6 (19.3 g, 57.88 mmol, 1 eq.) in dry tetrahydrofuran (250 mL) at −78° C. under nitrogen was added as solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (1.0M, 69.46 mL, 69.46 mmol, 1.2 eq.) over a 20 min time period. The mixture was kept at −78° C. for 1 h and then N-phenylbis(trifluoromethanesulfonamide) 1.4 (24.81 g, 69.46 mmol, 1.2 eq.) in tetrahydrofuran (100 mL) was added to the mixture over a 20 min time period. The mixture was kept at −78° C. for another hour, then slowly warmed up to and stirred room temperature for 10 h. Tetrahydrofuran was removed under reduced pressure. The crude product was dissolved in diethyl ether (500 mL) and washed with water (2×150 mL), 0.5N aqueous hydrochloric acid (2×100 mL), 1N aqueous sodium hydroxide (3×100 mL), brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixture of increasing polarity). Yield: 91%
  • 1H NMR (400 MHz, CDCl3) δ 7.15 (dd, 1H), 7.07 (dd, 1H), 6.98 (m, 1H), 6.05 (s, 1H), 3.65-3.53 (m, 2H), 3.36-3.27 (m, 2H), 2.79 (s, 2H), 1.69-1.60 (m, 2H), 1.56-1.48 (m, 2H), 1.46 (s, 9H)
  • Mass Spectral Analysis m/z=466.20 (M+H)+
  • Preparation of 38.8
  • To a solution of aqueous potassium carbonate (2M solution, 6.7 mL, 13.4 mmol, 3 eq.) was added dioxane (45 mL), 1.7 (1.63 g, 5.36 mmol, 1.2 eq.), and 38.7 (2.08 g, 4.46 mmol, 1 eq.) successively. The reaction flask was purged with nitrogen and 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (163 mg, 0.22 mmol, 0.05 eq.) was added to the mixture. The mixture was stirred at room temperature for 30 min Water (200 mL) and ethyl acetate (300 mL) were added and the two phases were separated. The aqueous phase was extracted with ethyl acetate (100 mL) and the combined organics were washed with brine (2×100 mL) and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 94%
  • 1H NMR (400 MHz, CDCl3) δ 8.54 (dd, 1H), 7.73 (m, 1H), 7.64 (dd, 1H), 7.17 (dd, 1H), 6.90 (m, 1H), 6.65 (dd, 1H), 6.10 (s, 1H), 3.60 (q, 4H), 3.51-3.35 (m, 4H), 2.80 (s, 2H), 1.68-1.49 (m, 4H), 1.47 (s, 9H), 1.33-1.18 (m, 6H)
  • Mass Spectral Analysis m/z=494.46 (M+H)+
  • Preparation of 38A
  • To a solution of 38.8 (4.5 g, 9 mmol, 1 eq.) in dichloromethane/methanol (5:1, 60 mL) was slowly added a 2M solution of hydrogen chloride in diethyl ether (2.0M, 22.5 mL, 45 mmol, 5 eq.). The mixture was stirred at room temperature for 10 h. The organic solvents were removed under reduced pressure and the crude product was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). Yield: 81%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.15-8.92 (m, 2H), 8.64 (d, 1H), 7.91 (dd, 1H), 7.60 (dd, 1H), 7.38 (dd, 1H), 7.11 (m, 1H), 6.68 (dd, 1H), 6.44 (s, 1H), 3.47 (q, 2H), 3.32 (q, 2H), 3.27-3.08 (m, 4H), 2.83 (s, 2H), 1.79-1.62 (m, 4H), 1.17 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=394.3 (M+H)+
  • Elemental analysis:
  • C24H28FN30, 1.4HCl, 0.8H2O
  • Theory: % C, 62.81; % H, 6.81; % Cl, 10.81; % N, 9.16.
  • Found: % C, 62.61; % H, 6.67; % Cl, 10.96; % N, 9.04.
  • Preparation of 38B
  • To a stirred solution of 38A (150 mg, 0.35 mmol, 1 eq.) in methanol (10 mL) was added palladium [30 mg, 10 wt. % (dry basis) on activated carbon, 20% wt. eq.]. The reaction mixture was stirred under a hydrogen atmosphere using a hydrogen balloon at room temperature for 10 h. The palladium on activated carbon was filtered off on a celite pad and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). Yield: 46%
  • 1H NMR (400 MHz, DMSO-d6) δ 8.88-8.78 (m, 2H), 8.51 (d, 1H), 7.67 (dd, 1H), 7.51 (d, 1H), 7.73 (m, 1H), 7.01 (m, 1H), 6.40 (dd, 1H), 4.22 (m, 1H), 3.44 (q, 2H), 3.28 (q, 2H), 3.18-3.02 (m, 4H), 2.85 (d, 1H), 2.75 (d, 1H), 2.16 (m, 1H), 1.75-1.46 (m, 5H), 1.15 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=396.4 (M+H)+
  • Preparation of 38.9
  • To a solution of 38.7 (11.88 g, 25.5 mmol, 1 eq.) in N,N-dimethylformamide (125 mL) at 0° C. was added potassium acetate (7.51 g, 76.5 mmol, 3 eq.), bis(pinacolato)diboron 1.14 (7.77 g, 30.6 mmol, 1.2 eq.), 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (560 mg, 0.76 mmol, 0.03 eq.) successively. The reaction mixture was stirred at 100° C. for 10 h. The reaction mixture was cooled to room temperature, diethyl ether (300 mL) and water (300 mL) were added and the mixture was stirred for another 30 minute at room temperature. The two phases were separated and the organic phase was washed with water (2×150 mL), brine (200 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 60%
  • 1H NMR (400 MHz, CDCl3) δ 7.54 (dd, 1H), 7.01 (dd, 1H), 6.84-6.77 (m, 2H), 3.51-3.37 (m, 4H), 2.65 (s, 2H), 1.61-1.38 (m, 13H), 1.34 (s, 12H)
  • Mass Spectral Analysis m/z=444.38 (M+H)+
  • Preparation of 38.10
  • To a solution of an aqueous solution of potassium carbonate (2M solution, 16.2 mL, 48.6 mmol, 3 eq.) was added dioxane (110 mL), 35.8 (3.926 g, 10.8 mmol, 1 eq.), and 38.9 (5.6 g, 12.6 mmol, 1.17 eq.) successively. The reaction flask was purged with nitrogen and 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (403 mg, 0.95 mmol, 0.05 eq.) was added to the mixture. The mixture was stirred at room temperature for 1 h and then heated at 55° C. for 10 h. Water (200 mL) and ethyl acetate (300 mL) were added and the mixture was stirred for another 10 min at room temperature. The two phases were separated and the organic phase was washed with brine (200 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 81%
  • 1H NMR (400 MHz, CDCl3) δ 7.22-7.16 (m, 2H), 7.13-7.07 (m, 2H), 6.82 (m, 1H), 6.48 (dd, 1H), 6.01 (s, 1H), 5.04 (s, 2H), 3.66-3.50 (m, 4H), 3.43-3.30 (m, 4H), 3.28 (s, 3H), 2.79 (s, 2H), 1.70-1.49 (m, 4H), 1.46 (s, 9H), 1.32-1.13 (m, 6H)
  • Mass Spectral Analysis m/z=553.51 (M+H)+
  • Preparation of 38C
  • To a stirred solution of 38.10 (1 g, 1.8 mmol, 1 eq.) in methanol (50 mL) was slowly added a 4M anhydrous solution of hydrogen chloride in dioxane (4.5 mL, 18 mmol, 10 eq.). The reaction mixture was stirred at room temperature for 10 h. The organic solvents were removed under reduced pressure and the crude material was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). Yield: 80%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.85-8.66 (m, 2H), 7.29 (m, 1H), 7.19 (d, 1H), 7.01 (m, 1H), 6.87 (d, 1H), 6.83 (dd, 1H), 6.39 (dd, 1H), 6.12 (s, 1H), 3.51-3.07 (m, 8H), 2.82 (s, 2H), 1.79-1.57 (m, 4H), 1.20-1.04 (m, 6H)
  • Mass Spectral Analysis m/z=409.7 (M+H)+
  • Elemental analysis:
  • C25H29FN2O2, 1HCl, 1.2H2O
  • Theory: % C, 64.35; % H, 7.00; % N, 6.00.
  • Found: % C, 64.34; % H, 6.92; % N, 6.04.
  • Preparation of 38.11
  • To a stirred solution of 38.10 (1.8 g, 3.25 mmol, 1 eq.) in methanol (32 mL) was added palladium [360 mg, 10 wt. % (dry basis) on activated carbon, 20% wt. eq.]. The reaction mixture was stirred under a hydrogen atmosphere using a hydrogen balloon at room temperature for 10 h. The palladium on activated carbon was filtered off on a celite pad and the filtrate was concentrated under reduced pressure and dried in vacuo. Yield: 91%
  • 1H NMR (400 MHz, CDCl3) δ 7.14 (d, 1H), 7.04 (m, 2H), 6.96 (dd, 1H), 6.80 (m, 1H), 6.51 (m, 1H), 5.13 (s, 2H), 4.46 (m, 1H), 3.60-3.25 (m, 11H), 2.78 (d, 1H), 2.65 (d, 1H), 2.04 (m, 1H), 1.68-1.38 (m, 14H), 1.30-1.10 (m, 6H)
  • Mass Spectral Analysis m/z=555.53 (M+H)+
  • Preparation of 38D
  • To a solution of 38.11 (0.30 g, 0.54 mmol, 1 eq.) in methanol (20 mL) was slowly added a 4M anhydrous solution of hydrogen chloride in dioxane (1.35 mL, 5.4 mmol, 10 eq.). The reaction mixture was stirred at room temperature for 10 h. The organic solvents were removed under reduced pressure and the crude material was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). Yield: 79%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.84 (s, 1H), 8.92-8.73 (m, 2H), 7.17 (m, 1H), 7.03-6.91 (m, 2H), 6.86 (d, 1H), 6.73 (dd, 1H), 6.41 (dd, 1H), 4.37 (m, 1H), 3.58-2.97 (m, 8H), 2.86 (d, 1H), 2.68 (d, 1H), 1.99 (m, 1H), 1.80-1.49 (m, 5H), 1.17-1.02 (m, 6H)
  • Mass Spectral Analysis m/z=411.76 (M+H)+
  • Example 39A Preparation of 39.1
  • To a suspension of copper (I) iodide (550 mg, 2.88 mmol, 0.036 eq) in anhydrous tetrahydrofuran (600 mL) was added dropwise a 2.0 M solution of benzylmagnesium chloride (28.3a) (100 mL, 200 mmol, 2.5 eq) in tetrahydrofuran under a nitrogen atmosphere at −10° C. After the reaction mixture was stirred at −10° C. for 30 min, solid 38.2 (28.72 g, 80 mmol, 1.0 eq) was added in ten portions over a 1 h period. After the addition was complete, the reaction mixture was stirred between −10° C. and −0° C. for 3 h, and then quenched by a mixture of concentrated ammonium hydroxide/saturated aqueous ammonium chloride/water (1:2:3, 400 mL). The mixture was extracted with ethyl acetate, and the combined organic layers were washed with a mixture of concentrated ammonium hydroxide/saturated aqueous ammonium chloride/water (1:2:3) and brine, dried over sodium sulfate and concentrated in vacuo. To the residue was added diethyl ether and the mixture was stirred overnight at room temperature. The solid was collected by filtration, washed with diethyl ether and dried in vacuo. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 7.30-7.00 (m, 10H), 4.96 (s, 2H), 3.70 (m, 2H), 2.98 (m, 2H), 2.80 (s+m, 4H), 1.49 (s, 6H), 0.83 (m, 2H).
  • Mass Spectral Analysis m/z=450.36 (M−Na)+
  • Preparation of 39.2
  • Compound 39.1 (39 g, 82.5 mmol) was dissolved in a mixture of N,N dimethylformamide (200 mL) and water (200 mL) and heated at ˜135° C. for 2 days, then cooled to room temperature. To the reaction mixture was added a 1 N aqueous sodium hydroxide (125 mL) and water (500 mL), the resulting mixture was washed with diethyl ether, acidified with a 6 N aqueous solution of hydrochloric acid and extracted with diethyl ether. The combined organic extracts were washed with water and brine, dried over sodium sulfate and concentrated in vacuo. Yield: 92.9%.
  • 1H NMR (400 MHz, DMSO-d6) δ 12.25 (brs, 1H), 7.36-7.20 (m, 10H), 5.08 (s, 2H), 3.60 (m, 2H), 3.33 (m, 2H), 2.79 (s, 2H), 2.19 (s, 2H), 1.50-1.40 (m, 4H).
  • Preparation of 28.6a
  • To a solution of 39.2 (1.1 g, 3 mmol, 1 eq) in anhydrous methylene chloride (20 mL) was added oxalyl chloride (1.6 mL, 18.3 mmol, 6.1 eq) in one portion followed by 2 drops of anhydrous N,N dimethylformamide. The reaction mixture was stirred at room temperature for 4 h and then concentrated in vacuo. The resulting acyl chloride was dissolved in anhydrous methylene chloride (60 mL) and aluminum chloride (804 mg, 6 mmol, 2 eq) was added in one portion. The reaction mixture was stirred at room temperature overnight and then quenched by water (40 mL) followed by addition of concentrated ammonium hydroxide to basify the aqueous layer. The organic layer was separated and the aqueous layer was extracted with methylene chloride. The combined organic layers were dried over sodium sulfate and concentrated in vacuo. The residue was then dissolved in methylene chloride (30 mL) and cooled to 0° C. To this solution was added triethylamine (1.3 mL, 9.34 mmol, 3.1 eq) followed by benzyl chloroformate (0.9 mL, 6.0 mmol, 2 eq). The reaction mixture was stirred at 0° C. for 1 h and then washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate and concentrated in vacuo. The residue was purified by column chromatography on silica gel (hexane-ethyl acetate-methylene chloride, 4:1:1), to yield the spiro ketone 28.6a. Yield: 95.5%.
  • 1H NMR (400 MHz, CDCl3) δ 8.0 (d, 1H), 7.50 (t, 1H), 7.33-7.23 (m, 7H), 5.11 (s, 2H), 2.98 (s, 2H), 2.62 (s, 2H), 1.50 (m, 4H).
  • Preparation of 28.7a
  • A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (3.6 mL, 3.6 mmol, 1.2 eq) was added at −78° C. to a solution of compound 28.6a (1.047 g, 3.0 mmol, 1 eq) in tetrahydrofuran (30 mL). After 45 minutes, a solution of N-phenyltrifluoromethanesulfonimide (1.4) (1.3 g, 3.6 mmol, 1.2 eq) in tetrahydrofuran (8 mL) was added dropwise to the reaction mixture. The reaction mixture was then warmed to room temperature and stirred for 2.5 h, quenched by addition of water (40 mL), and extracted with a mixture of hexane and diethyl ether (1:1). The organic extracts were combined and washed with water, brine and dried over sodium sulfate. Evaporation of the solvent provided 28.7a used for the next step without further purification. Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 7.35-7.18 (m, 9H), 5.98 (s, 1H), 5.11 (s, 2H), 3.70 (m, 2H), 3.40 (m, 2H), 2.83 (s, 2H), 1.66-1.56 (m, 4H).
  • Preparation of 28.12
  • To the solution of the enol triflate 28.7a (2.91 g, 6.04 mmol) in dimethoxyethane (60 mL) was added sequentially 2 N aqueous solution of sodium carbonate (10.4 mL, 20.8 mmol, 3.4 eq), lithium chloride (860 mg, 20.3 mmol, 3.4 eq), 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyridine-2-carboxylic acid diethylamide (1.7) (2.13 g, 7.0 mmol, 1.16 eq) and tetrakis(triphenylphosphine)palladium(0) (212 mg, 0.183 mmol, 0.03 eq). The reaction mixture was refluxed overnight, cooled to room temperature, diluted with water (60 mL) and extracted with diethyl ether. The combined organic extracts were dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1). Yield: 99%
  • 1H NMR (400 MHz, CDCl3) δ 8.53 (d, 1H), 7.76 (dd, 1H), 7.60 (d, 1H), 7.35-7.12 (m, 8H), 6.92 (d, 1H), 6.05 (s, 1H), 5.12 (s, 1H), 3.70 (m, 2H), 3.60 (m, 2H), 3.48 (m, 2H), 2.82 (s, 2H), 1.65-1.55 (m, 4H), 1.30 (t, 3H), 1.20 (t, 3H).
  • Preparation of 39A
  • Compound 28.12 (1.0 g, 2.7 mmol) was dissolved in methylene chloride (10 mL) and methanol (80 mL), and the mixture was hydrogenated in the presence of 10% Pd/C (300 mg) using a hydrogen balloon. After 2 days at room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride/methanol/conc. ammonia hydroxide, 10:1:1). Yield: 80%
  • 1H NMR (400 MHz, CDCl3) δ 8.42 (d, 1H), 7.52 (m, 2H), 7.12 (m, 2H), 7.05 (m, 1H), 6.70 (d, 1H), 4.10 (m, 1H), 3.56 (q, 2H), 3.42 (q, 2H), 3.10-2.50 (m, 6H), 2.10 (m, 1H), 1.60 (m, 5H), 1.28 (t, 3H), 1.20 (t, 3H).
  • Mass Spectral Analysis m/z=378.3 (M+H)+
  • Examples 39B, 39C Preparation of 39.3
  • To the solution of compound 39A (650 mg, 1.72 mmol) in methylene chloride (10 mL) was added triethylamine (0.34 mL, 2.4 mmol, 1.4 eq) followed by di-t-butyl dicarbonate (4.7) (450 mg, 2.06 mmol, 1.2 eq.). The reaction mixture was stirred at room temperature for 4 h and then concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate, 1:1).
  • Yield: 91.2%
  • 1H NMR (400 MHz, CDCl3) δ 8.41 (d, 1H), 7.52 (m, 2H), 7.12 (m, 2H), 7.05 (m, 1H), 6.70 (d, 1H), 4.10 (m, 1H), 3.58-3.33 (m, 8H), 2.90 (d, 1H), 2.72 (d, 1H), 2.09 (m, 1H), 1.65-1.52 (m, 5H), 1.47 (s, 9H), 1.29 (t, 3H), 1.20 (t, 3H).
  • Preparation of 39.4 & 39.5
  • Chiral separation of 39.3 (680 mg) gave the two enantiomers 39.4 and 39.5.
  • Column: Chiralpak ADH, 21×250 nm, 35° C.; SFC
  • Eluent: 35% MeOH/65% CO2; 50 mL/min, 200 bar
  • UV wavelength: 260 nm
  • Polarimeter: 670 nm
  • Sample: 80 mg/mL in MeOH, 2 mL injected
  • Positive polarimeter peak elutes first at about 6.5 minutes and the negative polarimeter peak elutessecond at about 9 minutes in 35% MeOH/CO2
  • 39.4: (−) enantiomer; ee>96% (268 mg)
  • 39.5: (+) enantiomer; ee>99% (295 mg)
  • Preparation of 39B
  • To the solution of pure enantiomer 39.4 (268 mg, 0.56 mmol) in methylene chloride (5 mL) was added 2.0 M hydrochloric acid in diethyl ether (10 mL, 20 mmol, 35.7 eq.) The mixture was stirred at ambient temperature for 24 h and the solvent was evaporated in vacuo. The resulting solid was triturated in diethyl ether, filtered and washed with diethyl ether. Yield: 86.2%.
  • 1H NMR (400 MHz, DMSO-d6) δ 8.90 (brs, 2H), 8.50 (s, 1H), 7.65 (d, 1H), 7.50 (d, 1H), 7.14 (m, 2H), 7.05 (m, 1H), 6.62 (d, 1H), 4.20 (m, 1H), 3.42 (q, 2H), 3.28 (q, 2H), 3.09 (m, 4H), 2.85 (d, 1H), 2.78 (d, 1H), 2.15 (m, 1H), 1.70-1.50 (m, 5H), 1.12 (t, 3H), 1.08 (t, 3H).
  • Mass Spectral Analysis m/z=378.83 (M+H)+
  • Elemental analysis:
  • C24H31N3O, 8/7HCl, 6/7H2O
  • Theory: % C, 66.32; % H, 7.85; % N, 9.67.
  • Found: % C, 66.33; % H, 7.72; % N, 9.53.
  • [α]25 D −69.1° (c=0.5, MeOH)
  • Preparation of 39C
  • To the solution of enantiomer 39.5 (295 mg, 0.62 mmol) in methylene chloride (5 mL) was added a 2.0 M solution of hydrochloric acid in diethyl ether (10 mL, 20 mmol, 32 eq.) The mixture was stirred at ambient temperature for 24 h and the solvent was evaporated in vacuo. The resulting solid was triturated in diethyl ether, filtered and washed with diethyl ether. Yield: 88.2%
  • 1H NMR (400 MHz, DMSO-d6) δ 8.90 (brs, 2H), 8.50 (s, 1H), 7.65 (d, 1H), 7.50 (d, 1H), 7.14 (m, 2H), 7.05 (m, 1H), 6.62 (d, 1H), 4.20 (m, 1H), 3.42 (q, 2H), 3.28 (q, 2H), 3.09 (m, 4H), 2.85 (d, 1H), 2.78 (d, 1H), 2.15 (m, 1H), 1.70-1.50 (m, 5H), 1.12 (t, 3H), 1.08 (t, 3H).
  • Mass Spectral Analysis m/z=378.83 (M+H)+
  • Elemental analysis:
  • C24H31N3O, 6/5HCl, 6/5H2O
  • Theory: % C, 65.09; % H, 7.87; % N, 9.49.
  • Found: % C, 65.03; % H, 7.68; % N, 9.34.
  • [α]25 D +70.2° (c=0.7, MeOH)
  • Examples 39D, 39E Preparation of 39.7
  • To the solution of the enol triflate 28.7a (2.91 g, 6.04 mmol) in dimethoxyethane (60 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (10.4 mL, 20.8 mmol, 3.4 eq), lithium chloride (860 mg, 20.3 mmol, 3.4 eq), 39.6 (2.792 g, 7.69 mmol, 1.27 eq) and tetrakis(triphenylphosphine)palladium(0) (212 mg, 0.183 mmol, 0.03 eq). The reaction mixture was refluxed overnight, cooled to room temperature, diluted with water (60 mL) and extracted with diethyl ether. The combined organic extracts were dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/acetone, 2:1).
  • Yield: 98.4%
  • 1H NMR (400 MHz, CDCl3) δ 7.38-7.05 (m, 11H), 6.72 (d, 1H), 5.95 (s, 1H), 5.12 (s, 2H), 5.00 (s, 2H), 3.69 (m, 2H), 3.53 (m, 2H), 3.45 (m, 2H), 3.32 (m, 2H), 3.25 (s, 3H), 2.80 (s, 2H), 1.66 (m, 2H), 1.55 (m, 2H), 1.26 (brs, 3H), 1.18 (brs, 3H).
  • Preparation of 39D
  • Iodotrimethylsilane (1.02 mL, 7.5 mmol, 2.84 eq) was added to the solution of compound 39.7 (1.5 g, 2.64 mmol) in anhydrous methylene chloride (30 mL) under nitrogen. The reaction mixture was stirred at room temperature for 2 h, quenched with a 1N aqueous solution of hydrochloric acid (40 mL) and washed with diethyl ether. The aqueous phase was basified to pH=9-10 by addition of a 3N aqueous solution of sodium hydroxide, and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate and concentrated in vacuo. The residue was dissolved in methylene chloride (10 mL) and methanol (20 mL). To this solution was added a 2.0 M anhydrous solution of hydrochloric acid in diethyl ether (30 mL, 60 mmol, 22.7 eq) and stirred at room temperature for 2 days. The reaction mixture was concentrated in vacuo and the residue was purified by column chromatography (eluent: methylene chloride/methanol, 5:1). Yield: 100%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.60 (s, 1H), 8.82 (brs, 2H), 7.24-7.10 (m, 4H), 6.86 (s, 1H), 6.80 (d, 1H), 6.70 (d, 1H), 6.00 (s, 1H), 3.40 (m, 2H), 3.23 (m, 2H), 3.12 (m, 4H), 2.82 (s, 2H), 1.66 (m, 4H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 4/5H2O
  • Theory: % C, 68.03; % H, 7.44; % N, 6.35.
  • Found: % C, 67.94; % H, 7.27; % N, 6.34.
  • Preparation of 39E
  • Compound 39.7 (1.7 g, 2.99 mmol) was dissolved in a mixed solvent of methylene chloride (15 mL) and methanol (120 mL), and the mixture was hydrogenated in the presence of 10% Pd/C (510 mg) using a hydrogen balloon. After 2 days of stirring at room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was dissolved in methylene chloride (10 mL) and methanol (20 mL). To this solution was added a 2.0 M anhydrous solution of hydrochloric acid in diethyl ether (30 mL, 60 mmol, 22.7 eq) and the mixture was stirred at room temperature for 2 days. The reaction mixture was concentrated in vacuo and the residue was dissolved in methylene chloride. The organic solution was washed with a saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate and concentrated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride/methanol, 5:1). Yield: 90.4%
  • 1H NMR (400 MHz, CDCl3) δ 7.10 (m, 2H), 7.03 (m, 1H), 6.94 (d, 1H), 6.82 (d, 1H), 6.76 (d, 1H), 6.70 (d, 1H), 4.70 (m, 1H), 3.50 (m, 2H), 3.32 (m, 2H), 2.98 (m, 5H), 2.62 (d, 1H), 2.11 (m, 1H), 1.78 (m, 1H), 1.55 (m, 2H), 1.40 (m, 2H), 1.21 (brs, 3H), 1.12 (brs, 3H).
  • Mass Spectral Analysis m/z=393.4 (M+H)+
  • Examples 39F, 39G Preparation of 39.8
  • To the solution of compound 39E (800 mg, 2.04 mmol) in methylene chloride (15 mL) was added triethylamine (0.42 mL, 3.0 mmol, 1.47 eq) followed by di-t-butyl dicarbonate (4.7) (446 mg, 2.04 mmol, 1.0 eq.). The reaction mixture was stirred at room temperature for 45 min and then concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate, 1:2).
  • Yield: 81%
  • 1H NMR (400 MHz, CDCl3) δ 7.98 (brs, 1H), 7.09 (m, 2H), 7.00 (m, 2H), 6.90 (d, 1H), 6.80 (d, 1H), 6.72 (d, 1H), 4.43 (m, 1H), 3.56 (m, 2H), 3.40 (m, 4H), 3.30 (m, 2H), 2.75 (d, 1H), 2.66 (d, 1H), 2.00 (m, 1H), 1.51-1.37 (m, 5H), 1.43 (s, 9H), 1.22 (m, 3H), 1.11 (m, 3H).
  • Preparation of 39.9 and 39.10
  • Chiral separation of 39.8 (800 mg) gave the two enantiomers 39.9 and 39.10.
  • Column: Chiralpak ADH, 21×250 nm, 35° C.; SFC
  • Eluent: 35% MeOH/65% CO2; 50 mL/min, 200 bar
  • UV wavelength: 260 nm
  • Polarimeter: 670 nm
  • Sample: 40 mg/mL in MeOH, 1.5 mL injected
  • Negative polarimeter peak elutes first at about 7.2 minutes and the positive polarimeter peak elutessecond at about 10.8 minutes in 35% MeOH/CO2
  • 39.9: (−) enantiomer; ee>99% (363 mg)
  • 39.10: (+) enantiomer; ee>99% (304 mg)
  • Preparation of 39F
  • To the solution of pure enantiomer 39.9 (305 mg, 0.62 mmol) in methylene chloride (5 mL) was added a 2.0 M anhydrous solution of hydrochloric acid in diethyl ether (10 mL, 20 mmol, 32.3 eq.) The mixture was stirred at ambient temperature for 24 h and the solvent was evaporated in vacuo. The resulting solid was triturated in diethyl ether, filtered and washed with diethyl ether. Yield: 90.5%.
  • 1H NMR (400 MHz, DMSO-d6) δ 9.80 (s, 1H), 8.80 (brs, 2H), 7.10-6.93 (m, 4H), 6.83 (s, 1H), 6.70 (m, 2H), 4.40 (m, 1H), 3.35-3.03 (m, 8H), 2.86 (d, 1H), 2.72 (d, 1H), 2.00 (m, 1H), 1.60 (m, 5H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=393.8 (M+H)+
  • Elemental analysis:
  • C25H32N2O2, 1HCl, 3/5H2O
  • Theory: % C, 68.27; % H, 7.84; % N, 6.37.
  • Found: % C, 68.17; % H, 7.72; % N, 6.41.
  • [α]25 D −59.6° (c=0.55, MeOH)
  • Preparation of 39G
  • To the solution of pure enantiomer 39.10 (280 mg, 0.57 mmol) in methylene chloride (5 mL) was added a 2.0 M anhydrous solution of hydrochloric acid in diethyl ether (10 mL, 20 mmol, 35 eq.) The mixture was stirred at ambient temperature for 24 h and the solvent was evaporated in vacuo. The resulting solid was triturated in diethyl ether, filtered and washed with diethyl ether. Yield: 90.3%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.80 (s, 1H), 8.80 (brs, 2H), 7.10-6.93 (m, 4H), 6.83 (s, 1H), 6.70 (m, 2H), 4.40 (m, 1H), 3.35-3.03 (m, 8H), 2.86 (d, 1H), 2.72 (d, 1H), 2.00 (m, 1H), 1.60 (m, 5H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=393.8 (M+H)+
  • Elemental analysis:
  • C24H31N3O, 1HCl, 3/5H2O
  • Theory: % C, 68.27; % H, 7.84; % N, 6.37.
  • Found: % C, 68.02; % H, 7.63; % N, 6.33.
  • [α]25 D +52.6° (c=0.85, MeOH)
  • Example 40A Preparation of 40.1
  • To a suspension of copper (I) iodide (553 mg, 2.90 mmol, 0.036 eq) in anhydrous tetrahydrofuran (50 mL) was added dropwise a 0.25 M solution of 4-methoxybenzylmagnesium chloride (28.3b) (640 mL, 160 mmol, 2.0 eq) in tetrahydrofuran under nitrogen atmosphere at −10° C. After the reaction mixture was stirred at −10° C. for 30 min, solid 38.2 (28.8 g, 80 mmol, 1.0 eq) was added to the mixture in ten portions over a 1 h period. After the addition, the reaction mixture was stirred between −10° C. and −0° C. for 3 h, and then quenched by a mixture of concentrated ammonium hydroxide/aqueous saturated ammonium chloride/water (1:2:3, 400 mL) The mixture was extracted with ethyl acetate, and the combined organic layers were washed with concentrated ammonium hydroxide/aqueous saturated ammonium chloride/water (1:2:3) and brine, dried over sodium sulfate and concentrated in vacuo. To the residue was added diethyl ether and the mixture was stirred overnight at room temperature. The solid was collected by filtration, washed with diethyl ether and dried in vacuo. Yield: 100%
  • 1H NMR (400 MHz, DMSO d6) δ 7.29 (m, 5H), 6.91 (δ, 2H), 6.73 (δ, 2H), 4.98 (s, 2H), 3.70 (s+m, 5H), 2.93 (m, 2H), 2.80 (m, 2H), 2.70 (s, 2H), 1.49 (s, 6H), 0.82 (m, 2H).
  • Mass Spectral Analysis m/z=480.40 (M−Na)+
  • Preparation of 28.11
  • Compound 40.1 (40.2 g, 79.92 mmol) was dissolved in a mixture of N,N dimethylformamide (200 mL) and water (200 mL) and the mixture was heated at ˜135° C. for 2 days, then cooled to room temperature. To the reaction mixture was added a 1N aqueous solution of sodium hydroxide (125 mL) and water (500 mL). The resulting mixture was washed with diethyl ether, acidified with a 6 N aqueous solution of hydrochloric acid and extracted with diethyl ether. The combined organic extracts were washed water and brine, dried over sodium sulfate and concentrated in vacuo. Yield: 100%.
  • 1H NMR (400 MHz, DMSO d6) δ 12.22 (brs, 1H), 7.33 (m, 5H), 7.10 (d, 2H), 6.86 (d, 2H), 5.06 (s, 2H), 3.73 (s, 3H), 3.60 (m, 2H), 3.32 (m, 2H), 2.69 (s, 2H), 2.17 (s, 2H), 1.45-1.35 (m, 4H).
  • Preparation of 28.6b
  • To a solution of 28.11 (1.98 g, 5 mmol) in anhydrous methylene chloride (10 mL) was added a 2.0 M solution of oxalyl chloride in methylene chloride (20 mL, 40 mmol, 8 eq) followed by 2 drops of anhydrous N,N-dimethylformamide. The reaction mixture was stirred at room temperature for 4 h and then concentrated in vacuo. The resulting acyl chloride was dissolved in anhydrous methylene chloride (100 mL) and aluminum chloride (1.35 g, 10 mmol, 2 eq) was added in one portion. The reaction mixture was stirred at room temperature overnight and then quenched with water (60 mL) followed by addition of concentrated ammonium hydroxide to basify the aqueous layer. The organic layer was separated and the aqueous layer was extracted with methylene chloride. The combined organic layers were dried over sodium sulfate and concentrated in vacuo. The residue was then dissolved in methylene chloride (60 mL) and cooled to 0° C. To this solution was added triethylamine (3.0 mL, 21.6 mmol, 4.3 eq) followed by benzyl chloroformate (2.0 mL, 13.3 mmol, 2.7 eq). The reaction mixture was stirred at 0° C. for 1 h and then washed with saturated aqueous solution of sodium bicarbonate, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/methylene chloride/ethyl acetate, 4:1:1). Yield: 89.7%
  • 1H NMR (400 MHz, CDCl3) δ 7.48 (d, 1H), 7.35 (m, 5H), 7.16 (d, 1H), 7.10 (dd, 1H), 5.11 (s, 2H), 3.81 (s, 3H), 3.50 (m, 4H), 2.90 (s, 2H), 2.60 (s, 2H), 1.50 (m, 4H).
  • Preparation of 28.7b
  • A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (100 mL, 100 mmol, 1.22 eq) was added at −78° C. to a solution of compound 28.6b (31 g, 81.8 mmol, 1 eq) in tetrahydrofuran (600 mL). After 45 minutes, a solution of N-phenyltrifluoromethanesulfonimide (1.4) (38 g, 106.4 mmol, 1.3 eq) in tetrahydrofuran (120 mL) was added dropwise. The reaction mixture was then warmed to room temperature, stirred for 4.5 h at room temperature, quenched by addition of water (500 mL), and extracted with a mixture of hexane and diethyl ether (1:1). The organic extracts were combined and washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate, 1:4). Yield: 98%.
  • 1H NMR (400 MHz, CDCl3) δ 7.33 (m, 5H), 7.09 (d, 1H), 6.90 (d, 1H), 6.80 (dd, 1H), 5.98 (s, 1H), 5.12 (s, 2H), 3.80 (s, 3H), 3.68 (m, 2H), 3.40 (m, 2H), 2.76 (s, 2H), 1.65 (m, 2H), 1.55 (m, 2H).
  • Preparation of 40.2
  • To the solution of the enol triflate 28.7b (4.0 g, 7.83 mmol) in dimethoxyethane (80 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (13.6 mL, 27.2 mmol, 3.47 eq), lithium chloride (1.12 g, 26.4 mmol, 3.37 eq), 1.7 (2.74 g, 9.0 mmol, 1.15 eq) and tetrakis(triphenylphosphine)palladium(0) (276 mg, 0.238 mmol, 0.03 eq). The reaction mixture was refluxed overnight, cooled to room temperature, diluted with water (120 mL) and extracted with diethyl ether. The combined organic extracts were dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate, 2:3). Yield: 100%
  • 1H NMR (400 MHz, CDCl3) δ 8.57 (d, 1H), 7.73 (dd, 1H), 7.60 (d, 1H), 7.32 (m, 5H), 7.11 (d, 1H), 6.76 (dd, 1H), 6.50 (d, 1H), 6.05 (s, 1H), 5.12 (s, 1H), 3.70 (m, 2H), 3.68-3.43 (m, 8H), 2.78 (s, 2H), 1.66 (m, 2H), 1.55 (m, 2H), 1.30 (t, 3H), 1.22 (t, 3H).
  • Preparation of 40A
  • Iodotrimethylsilane (0.86 mL, 6 mmol, 3 eq) was added to a solution of compound 40.2 (1.08 g, 2 mmol) in anhydrous methylene chloride (15 mL) under nitrogen. The reaction mixture was stirred at room temperature for 1.5 h and quenched with a 1N aqueous solution of hydrochloric acid (40 mL). The aqueous phase was washed with diethyl ether. The aqueous was basified to pH=9-10 by addition of a 3N aqueous solution of sodium hydroxide, and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate and concentrated in vacuo. The residue was dissolved in methylene chloride (10 mL) and diluted with diethyl ether (40 mL). To this solution was added a 2.0 M anhydrous solution of hydrochloric acid in diethyl ether (20 mL, 40 mmol, 20 eq) and the mixture was stirred at room temperature overnight. The precipitated solid was collected by filtration and washed with ether, dried in vacuo. Yield: 87%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.08 (brs, 1H), 8.96 (brs, 1H), 8.66 (d, 1H), 7.91 (dd, 1H), 7.60 (d, 1H), 7.27 (d, 1H), 6.84 (dd, 1H), 6.40 (d, 1H), 6.35 (s, 1H), 3.65 (s, 3H), 3.47 (q, 2H), 3.30-3.10 (m, 6H), 2.78 (s, 2H), 1.70 (m, 4H), 1.12 (m, 6H).
  • Mass Spectral Analysis m/z=406.3 (M+H)+
  • Elemental analysis:
  • C25H31N3O2, 3/2HCl, 3/4H2O
  • Theory: % C, 63.38; % H, 7.23; % N, 8.87; % Cl, 11.23.
  • Found: % C, 63.25; % H, 7.24; % N, 8.70; % Cl, 11.24.
  • Example 40B Preparation of 40B
  • Compound 40.2 (1.3 g, 2.41 mmol) was dissolved in methylene chloride (10 mL) and methanol (80 mL), and hydrogenated in the presence of 10% Pd/C (400 mg) using a hydrogen balloon. After 3 days at room temperature, the reaction mixture was filtered through celite and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride/methanol/concentrated ammonium hydroxide, 10:1:1). Yield: 92.7%
  • 1H NMR (400 MHz, CDCl3) δ 8.46 (d, 1H), 7.53 (m, 2H), 7.12 (m, 2H), 7.05 (d, 1H), 6.72 (dd, 1H), 6.23 (d, 1H), 4.10 (m, 1H), 3.63 (s, 3H), 3.58 (q, 2H), 3.44 (q, 2H), 2.90 (m, 5H), 2.60 (d, 1H), 2.08 (m, 1H), 1.52 (m, 5H), 1.28 (t, 3H), 1.20 (t, 3H).
  • Mass Spectral Analysis m/z=408.5 (M+H)+
  • Example 40C Preparation of 40.3
  • To a solution of the enol triflate 28.7b (2.05 g, 4 mmol) in dimethoxyethane (40 mL) was added sequentially 2 N aqueous solution of sodium carbonate (7.0 mL, 14 mmol, 3.5 eq), lithium chloride (580 mg, 13.7 mmol, 3.43 eq), 39.6 (1.96 g, 5.4 mmol, 1.35 eq) and tetrakis(triphenylphosphine)palladium(0) (142 mg, 0.123 mmol, 0.03 eq). The reaction mixture was refluxed overnight, cooled to room temperature, diluted with water (50 mL) and extracted with diethyl ether. The combined organic extracts were dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/acetone, 2:1). Yield: 96.6%
  • 1H NMR (400 MHz, CDCl3) δ 7.35 (m, 5H), 7.20 (m, 2H), 7.08 (m, 2H), 6.69 (dd, 1H), 6.35 (d, 1H), 5.95 (s, 1H), 5.12 (s, 2H), 5.00 (s, 2H), 3.70 (m, 2H), 3.68 (s, 3H), 3.54 (m, 2H), 3.44 (m, 2H), 3.30 (m, 2H), 3.27 (s, 3H), 2.79 (s, 2H), 1.68 (m, 2H), 1.52 (m, 2H), 1.28 (brs, 3H), 1.20 (brs, 3H).
  • Preparation of 40C
  • Iodotrimethylsilane (1.02 mL, 7.5 mmol, 2.84 eq) was added to a solution of compound 40.3 (1.5 g, 2.64 mmol) in anhydrous methylene chloride (30 mL) under nitrogen. The reaction mixture was stirred at room temperature for 2 h, quenched with 1N aqueous solution of hydrochloric acid (40 mL). The aqueous phase was washed with diethyl ether. The aqueous phase was basified o pH=9-10 by addition of a 3 N aqueous solution of sodium hydroxide, and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate and concentrated in vacuo. The residue was dissolved in methylene chloride (10 mL) and methanol (20 mL). To this solution was added a 2.0 M solution of anhydrous hydrochloric acid in diethyl ether (30 mL, 60 mmol, 22.7 eq) and stirred at room temperature for 2 days. The reaction mixture was concentrated in vacuo and the residue was purified by column chromatography (eluent: methylene chloride/methanol, 5:1). Yield: 94%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.63 (brs, 1H), 8.98 (brd, 2H), 7.14 (m, 2H), 6.89 (s, 1H), 6.80 (d, 1H), 6.73 (dd, 1H), 6.21 (d, 1H), 6.00 (s, 1H), 3.60 (s, 3H), 3.40 (m, 2H), 3.23 (m, 2H), 3.12 (m, 4H), 2.75 (s, 2H), 1.69 (m, 4H), 1.12 (m, 6H).
  • Mass Spectral Analysis m/z=421.3 (M+H)+
  • Elemental analysis:
  • C26H32N2O3, 1HCl, 1H2O
  • Theory: % C, 65.74; % H, 7.43; % N, 5.90.
  • Found: % C, 66.02; % H, 7.32; % N, 5.89.
  • Example 41A Preparation of 41.2
  • To a solution of 13.3 (0.50 g, 1.19 mmol, 1.0 eq) in acetonitrile (15 mL) under nitrogen was added N,N-diisopropylethylamine (0.50 mL, 2.85 mmol, 2.4 eq) and 41.1 (0.30 mL, 2.37 mmol, 2.0 eq). The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (0.46 g, 1.42 mmol, 1.2 eq) was slowly added to the reaction mixture which was warmed to room temperature and stiffing was continued for 16 h at room temperature. The reaction mixture was concentrated, dissolved in ethyl acetate and washed three times with a saturated aqueous solution of sodium bicarbonate, then brine. The organics were concentrated and purified by column chromatography (eluent: 50% hexane/ethyl acetate). Yield: 75% 1HNMR (400 MHz, CDCl3) δ 7.43 (d, 2H), 7.37 (d, 2H), 7.19 (m, 1H), 7.00 (dd, 1H), 6.94 (dd, 1H), 6.86 (m, 1H), 5.57 (s, 1H), 3.86 (brs, 2H), 3.69 (brs, 3H), 3.46-3.30 (brm, 8H), 2.05 (brd, 2H), 1.67 (m, 2H), 1.48 (s, 9H), 1.14 (brs, 3H)
  • Mass Spectral Analysis m/z=507.5 (M+H)+
  • Preparation of 41A
  • To a solution of 41.2 (0.45 g, 0.888 mmol, 1.0 eq) in methylene chloride (10 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.78 mL, 3.55 mmol, 4.0 eq). The reaction was warmed to room temperature, stirred for 48 h and concentrated to a foam, which was sonicated in 25 mL of a 4:1 hexane/diethyl ether solution. The resulting solid was collected by vacuum filtration. Yield: 71%
  • 1HNMR (400 MHz, DMSO d6) δ 8.96 (brs, 2H), 7.35 (s, 4H), 7.18 (m, 1H), 6.97 (d, 1H), 6.88 (m, 2H), 5.86 (s, 1H), 3.52-3.10 (brm, 13H), 1.96 (brm, 4H), 1.02 (brd, 3H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 0.50H2O
  • Theory: % C, 66.43; % H, 7.14; % N, 6.20.
  • Found: % C, 66.43; % H, 7.00; % N, 6.10.
  • Example 41B Preparation of 41.4
  • To a solution of 13.3 (0.50 g, 1.19 mmol, 1.0 eq) in acetonitrile (15 mL) under nitrogen was added N,N-diisopropylethylamine (0.50 mL, 2.85 mmol, 2.4 eq) and 41.3 (0.37 mL, 2.37 mmol, 2.0 eq). The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (0.46 g, 1.42 mmol, 1.2 eq) was slowly added to the reaction mixture, which was warmed to room temperature and stirred for 16 h at room temperature. The reaction was concentrated, dissolved in ethyl acetate and washed three times with a saturated aqueous solution of sodium bicarbonate, then brine. The organics were concentrated and purified by column chromatography (eluent: methanol/methylene chloride, 5:95). Yield: 77%
  • 1HNMR (400 MHz, CDCl3) δ 7.42 (d, 2H), 7.37 (d, 2H), 7.19 (m, 1H), 6.99 (brs, 1H), 6.94 (dd, 1H), 6.85 (m, 1H), 5.57 (s, 1H), 3.86 (brs, 2H), 3.61 (brs, 2H), 3.35 (brs, 4H), 2.62 (brs, 1H), 2.34 (brs, 4H), 2.06 (brm, 5H), 1.68 (m, 2H), 1.48 (s, 9H), 1.15 (brs, 3H)
  • Mass Spectral Analysis m/z=520.5 (M+H)+
  • Preparation of 41B
  • To a solution of 41.4 (0.47 g, 0.904 mmol, 1.0 eq) in methylene chloride (10 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (2.71 mL, 5.43 mmol, 6.0 eq). The reaction was warmed to room temperature, stirred for 48 h at room temperature and diluted with diethyl ether (10 mL). The precipitate was filtered and partitioned in a saturated aqueous solution of sodium bicarbonate and ethyl acetate. The organic layer was separated. The aqueous phase was further extracted with methylene chloride and all organics were combined and concentrated. Yield: 72%
  • 1HNMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 7.19 (m, 1H), 6.97 (m, 2H), 6.85 (t, 1H), 5.63 (s, 1H), 3.62 (brs, 2H), 3.36 (brs, 2H), 3.19 (m, 2H), 2.98 (m, 2H), 2.60 (brs, 3H), 2.33 (brs, 3H), 2.09 (brm, 4H), 1.81 (brm, 2H), 1.22 (brd, 3H)
  • Mass Spectral Analysis m/z=420.3 (M+H)+
  • Elemental analysis:
  • C26H33N3O2, 0.67H2O
  • Theory: % C, 72.36; % H, 8.02; % N, 9.74.
  • Found: % C, 72.02; % H, 7.80; % N, 9.55.
  • Example 41C Preparation of 41.7
  • To a solution of 41.6 (5.82 mL, 48.93 mmol, 1.0 eq) in diethyl ether (15 mL) at 0° C. under nitrogen was added drop wise a solution of 41.5 (5.0 g, 48.93 mmol, 1.0 eq) in diethyl ether (10 mL). The reaction was stirred at 0° C. for 30 min and then at room temperature for 2 h. The reaction was concentrated. The crude product was used for the next step without further purification. Yield: 99%
  • 1HNMR (400 MHz, CDCl3) δ 9.91 (brs, 1H), 3.48 (t, 2H), 2.87 (t, 2H), 2.64 (q, 2H), 1.70 (m, 2H), 1.11 (t, 3H)
  • Preparation of 41.8
  • To a solution of 13.3 (3.0 g, 7.12 mmol, 1.0 eq) in acetonitrile (60 mL) under nitrogen was added N,N-diisopropylethylamine (2.98 mL, 17.08 mmol, 2.4 eq) and 41.7 (2.82 g, 14.23 mmol, 2.0 eq). The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (2.74 g, 8.54 mmol, 1.2 eq) was slowly added. The reaction mixture was warmed to room temperature and stirred for 16 h at room temperature. The reaction was concentrated, dissolved in ethyl acetate and the solution was washed with a saturated aqueous solution of sodium bicarbonate, then brine. The organics were concentrated to a solid, which was triturated in a ethyl acetate/hexane solution (1:9, 50 mL). The resulting precipitate was collected by vacuum filtration.
  • Yield: 74%
  • 1HNMR (400 MHz, CDCl3) δ 8.42 (brs, 1H), 7.41 (s, 4H), 7.20 (m, 1H), 6.96 (m, 2H), 6.86 (m, 1H), 5.57 (s, 1H), 3.87 (brs, 2H), 3.65 (brt, 2H), 3.46-3.30 (brm, 6H), 2.06 (brd, 2H), 1.87 (brm, 2H), 1.67 (m, 2H), 1.48 (s, 9H), 1.19 (t, 3H)
  • Mass Spectral Analysis m/z=602.4 (M+H)+
  • Preparation of 41.9
  • To a suspension of 41.8 (1.0 g, 1.66 mmol, 1.0 eq) in methanol (30 mL) was added potassium carbonate (0.69 g, 4.99 mmol, 3.0 eq). The reaction was stirred for 48 h at room temperature and concentrated. Water was added and the product was extracted three times with ethyl acetate. The organics were combined and concentrated to a solid, which was triturated in hexane and collected by vacuum filtration. Yield: 75%
  • 1HNMR (400 MHz, DMSO d6) δ 7.47 (s, 4H), 7.29 (m, 1H), 7.05 (m, 2H), 6.97 (m, 1H), 5.95 (s, 1H), 3.79 (brd, 2H), 3.58-3.25 (brm, 8H), 1.94 (m, 2H), 1.78 (brm, 4H), 1.49 (s, 9H), 1.17 (brd, 3H)
  • Mass Spectral Analysis m/z=506.5 (M+H)+
  • Preparation of 41.11
  • To a solution of 41.9 (0.466 g, 0.922 mmol, 1.0 eq) in N,N dimethylformamide (10 mL) was added triethylamine (0.19 mL, 1.38 mmol, 1.5 eq) and 41.10 (0.22 g, 0.968 mmol, 1.05 eq). The reaction was stirred for 3.5 h at room temperature. Ethyl acetate and a saturated aqueous solution of sodium bicarbonate were added to the mixture. The layers were separated and the aqueous phase was further extracted with ethyl acetate. The organics were combined, concentrated and used for the next step without further purification. Yield: 100% (crude)
  • Mass Spectral Analysis m/z=691.8 (M+H)+
  • Preparation of 41.12
  • To a solution of 41.11 (0.64 g, 0.926 mmol, 1.0 eq) in N,N dimethylformamide (10 mL) was added potassium carbonate (0.196 g, 1.42 mmol, 1.5 eq) and methyl iodide (0.12 mL, 1.85 mmol, 2.0 eq). The reaction was stirred at room temperature for 16 h. Ethyl acetate and water were added to the mixture. The layers were separated and the organics were concentrated. The crude product was used for the next step without further purification. Yield: 96%
  • Mass Spectral Analysis m/z=705.7 (M+H)+
  • Preparation of 41.13
  • To a solution of 41.12 (0.626 g, 0.888 mmol, 1.0 eq) in N,N dimethylformamide (10 mL) under nitrogen was added potassium carbonate (0.307 g, 2.22 mmol, 2.5 eq) and benzenethiol (0.14 mL, 1.33 mmol, 1.5 eq). The reaction was stirred for 2 h at room temperature. Ethyl acetate and a saturated aqueous solution of sodium bicarbonate were added to the mixture. The layers were separated. The organics were concentrated and the crude product was purified by column chromatography [eluent: methanol/(methylene chloride/ammonium hydroxide, 99:1) mixtures of increasing polarity]. Yield: 68%
  • 1HNMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 7.19 (m, 1H), 6.96 (m, 2H), 6.87 (m, 1H), 5.57 (s, 1H), 3.87 (brs, 2H), 3.60 (brs, 2H), 3.34 (brs, 4H), 2.70 (brs, 1H), 2.50 (brs, 2H), 2.35 (brs, 1H), 2.06 (brd, 2H), 1.91 (brs, 1H), 1.68 (m, 4H), 1.48 (s, 9H), 1.21 (brd, 3H)
  • Mass Spectral Analysis m/z=520.5 (M+H)+
  • Preparation of 41C
  • To a solution of 41.13 (0.31 g, 0.60 mmol, 1.0 eq) in methylene chloride (10 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.80 mL, 3.58 mmol, 6.0 eq). The reaction mixture was warmed to room temperature, stirred for 48 h at room temperature and concentrated to a foam, which was triturated in diethyl ether (7 mL). The resulting solid was collected by vacuum filtration. Yield: 82%
  • 1HNMR (400 MHz, DMSO d6) δ 9.22 (brs, 2H), 8.93 (brs, 2H), 7.52 (m, 4H), 7.33 (m, 1H), 7.12 (d, 1H), 7.06 (m, 2H), 6.00 (s, 1H), 3.57 (brs, 2H), 3.26 (brm, 6H), 3.00 (brs, 2H), 2.62 (brs, 2H), 2.18-1.92 (brm, 7H), 1.15 (brm, 3H)
  • Mass Spectral Analysis m/z=420.3 (M+H)+
  • Elemental analysis:
  • C26H33N3O2, 2HCl, 0.50H2O
  • Theory: % C, 62.27; % H, 7.24; % N, 8.38; % Cl, 14.14.
  • Found: % C, 62.15; % H, 7.05; % N, 8.31; % Cl, 14.19.
  • Example 41D Preparation of 41.15
  • To a solution of 13.3 (1.00 g, 2.37 mmol, 1.0 eq) in acetonitrile (15 mL) under nitrogen was added N,N-diisopropylethylamine (1.45 mL, 8.30 mmol, 3.5 eq) and 41.14 (0.60 mL, 4.74 mmol, 2.0 eq). The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (0.91 g, 2.85 mmol, 1.2 eq) was slowly added to the reaction mixture, which was warmed to room temperature and stirred for 40 h at room temperature. Water was added and the product was extracted two times with ethyl acetate. The organics were concentrated and the crude product was purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity). Yield: 51%
  • 1HNMR (400 MHz, CDCl3) δ 8.32 (brt, 1H), 7.85 (d, 2H), 7.41 (d, 2H), 7.19 (m, 1H), 6.96 (m, 2H), 6.85 (m, 1H), 5.58 (s, 1H), 3.86 (brs, 2H), 3.61 (q, 2H), 3.34 (brs, 2H), 2.77 (t, 2H), 2.53 (s, 6H), 2.04 (brd, 2H), 1.94 (m, 2H), 1.67 (m, 2H), 1.47 (s, 9H)
  • Mass Spectral Analysis m/z=506.5 (M+H)+
  • Preparation of 41D
  • To a solution of 41.15 (0.090 g, 0.18 mmol, 1.0 eq) in methylene chloride (5 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (0.53 mL, 1.07 mmol, 6.0 eq). The reaction was warmed to room temperature and stirred for 16 h at room temperature. Additional amount of an anhydrous solution of hydrochloric acid in diethyl ether (0.50 mL) was added to the reaction which was stirred for an additional 16 h at room temperature. The precipitate was collected by vacuum filtration. Yield: 65%
  • 1HNMR (400 MHz, DMSO d6) δ 9.98 (brs, 1H), 8.94 (brs, 2H), 8.77 (t, 1H), 7.96 (d, 2H), 7.47 (d, 2H), 7.27 (m, 1H), 7.06 (d, 1H), 6.95 (d, 2H), 5.94 (s, 1H), 3.36 (m, 2H), 3.22 (brm, 4H), 3.09 (m, 2H), 2.76 (s, 6H), 2.12-1.89 (brm, 6H)
  • Mass Spectral Analysis m/z=406.4 (M+H)+
  • Example 41E Preparation of 41.17
  • To a solution of 41.6 (6.75 mL g, 56.72 mmol, 1.0 eq) in diethyl ether (15 mL) at 0° C. under nitrogen was added drop wise 41.16 (5.97 mL, 56.72 mmol, 1.0 eq). The reaction was stirred at 0° C. for 30 min and then at room temperature for 2 h. The reaction mixture was concentrated to a solid, which was triturated in hexanes. The suspension was stirred overnight at room temperature and the precipitate was collected by vacuum filtration. Yield: 90%
  • 1HNMR (400 MHz, CDCl3) δ 7.15 (brs, 1H), 3.41 (t, 2H), 2.83 (t, 2H), 2.66 (q, 2H), 1.11 (t, 3H)
  • Preparation of 41.18
  • To a solution of 13.3 (2.0 g, 4.74 mmol, 1.0 eq) in acetonitrile (50 mL) under nitrogen was added N,N-diisopropylethylamine (1.98 mL, 11.39 mmol, 2.4 eq) and 41.17 (1.75 g, 9.48 mmol, 2.0 eq). The mixture was stirred for 10 min at room temperature, cooled to 0° C. and O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (1.83 g, 5.69 mmol, 1.2 eq) was slowly added to the reaction mixture, which was warmed to room temperature and stirred for 16 h at room temperature. The reaction mixture was concentrated, dissolved in ethyl acetate and washed with a saturated aqueous solution of sodium bicarbonate, then brine. The organics were concentrated to a solid, which was triturated in a ethyl acetate/hexane solution (1:9; 50 mL). The precipitate was collected by vacuum filtration.
  • Yield: 72%
  • 1HNMR (400 MHz, CDCl3) δ 8.23 (brs, 1H), 7.41 (s, 4H), 7.20 (m, 1H), 6.96 (m, 2H), 6.86 (m, 1H), 5.58 (s, 1H), 3.97 (brs, 2H), 3.79 (brs, 2H), 3.66 (brs, 2H), 3.38 (brm, 4H), 2.05 (m, 2H), 1.68 (m, 2H), 1.48 (s, 9H), 1.22 (t, 3H)
  • Mass Spectral Analysis m/z=588.5 (M+H)+
  • Preparation of 41.19
  • To a suspension of 41.18 (1.0 g, 1.70 mmol, 1.0 eq) in methanol (30 mL) was added potassium carbonate (0.71 g, 5.11 mmol, 3.0 eq). The reaction was stirred for 48 h at room temperature and concentrated. Water was added to the mixture and the product was extracted with ethyl acetate. The organics were combined and concentrated to a solid, which was triturated in hexane and collected by vacuum filtration. Yield: 72%
  • 1HNMR (400 MHz, CDCl3) δ 7.82 (d, 1H), 7.40 (m, 2H), 7.19 (m, 1H), 6.95 (m, 2H), 6.85 (m, 2H), 5.58 (s, 1H), 3.86 (brs, 2H), 3.56 (q, 2H), 3.34 (brs, 2H), 2.89 (t, 1H), 2.70 (q, 1H), 2.05 (m, 2H), 1.68 (m, 2H), 1.48 (s, 11H), 1.13 (t, 3H)
  • Mass Spectral Analysis m/z=492.5 (M+H)+
  • Preparation of 41.20
  • To a solution of 41.19 (0.90 g, 1.83 mmol, 1.0 eq) and triethylamine (0.77 mL, 5.49 mmol, 3.0 eq) in tetrahydrofuran (20 mL) at 0° C. under nitrogen was added 4.7 (0.44 g, 2.01 mmol, 1.1 eq). The ice bath was removed and the reaction was stirred at room temperature for 30 min. Water was added and the product was extracted with ethyl acetate. The organics were combined, concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 67%
  • 1HNMR (400 MHz, CDCl3) δ 7.86 (d, 2H), 7.67 (s, 1H), 7.38 (d, 2H), 7.19 (m, 1H), 6.94 (d, 2H), 6.85 (m, 1H), 5.58 (s, 1H), 3.86 (brs, 2H), 3.61 (m, 2H), 3.53 (s, 2H), 3.30 (brm, 4H), 2.05 (m, 2H), 1.68 (m, 2H), 1.48 (s, 9H), 1.46 (s, 9H), 1.14 (t, 3H)
  • Mass Spectral Analysis m/z=592.6 (M+H)+
  • Preparation of 41.21
  • To a solution of 41.20 (0.70 g, 1.18 mmol, 1.0 eq) in tetrahydrofuran (30 mL) under nitrogen was added sodium hydride (0.085 g, 3.55 mmol, 3.0 eq) and the mixture was stirred for 10 min at room temperature. Methyl iodide (0.22 mL, 3.55 mmol, 3.0 eq) was added to the reaction mixture, which was stirred for 16 h at room temperature. The reaction mixture was carefully quenched with water and the product was extracted with ethyl acetate. The organics were concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 96%
  • 1HNMR (400 MHz, CDCl3) δ 7.40 (brm, 4H), 7.19 (m, 1H), 6.96 (m, 2H), 6.85 (m, 1H), 5.54 (s, 1H), 3.86 (brs, 2H), 3.70 (brs, 2H), 3.48 (m, 1H), 3.32 (brs, 4H), 3.15 (brs, 2H), 3.07 (brs, 2H), 2.05 (m, 2H), 1.67 (m, 2H), 1.48 (s, 9H), 1.41 (s, 9H), 1.14 (brt, 3H)
  • Mass Spectral Analysis m/z=606.7 (M+H)+
  • Preparation of 41E
  • To a solution of 41.21 (0.68 g, 1.12 mmol, 1.0 eq) in methylene chloride (10 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (6.74 mL, 13.47 mmol, 12.0 eq). The reaction mixture was warmed to room temperature and stirred for 16 h at room temperature. The precipitate was collected by vacuum filtration. Yield: 90%
  • 1HNMR (400 MHz, DMSO d6) δ 9.16 (brs, 2H), 8.97 (brs, 2H), 7.62 (m, 2H), 7.44 (d, 2H), 7.27 (m, 1H), 7.06 (d, 1H), 6.97 (m, 2H), 5.94 (s, 1H), 3.77 (brs, 2H), 3.34 (s, 3H), 3.20 (brm, 4H), 3.00 (brs, 4H), 2.06 (brm, 4H), 1.24 (brt, 3H)
  • Mass Spectral Analysis m/z=406.8 (M+H)+
  • Elemental analysis:
  • C25H31N3O2, 2HCl, 0.50H2O
  • Theory: % C, 61.60; % H, 7.03; % N, 8.62; % Cl, 14.55.
  • Found: % C, 61.45; % H, 6.78; % N, 8.64; % Cl, 14.78.
  • Example 42A Preparation of 42.1
  • To a solution of 21.6 (6.03 g, 13.0 mmol) and 1.7 (3.95 g, 13.0 mmol, 1 eq) in dimethoxyethane (DME) (125 mL) was added sequentially a 2N aqueous solution of sodium carbonate (19.5 mL, 39.0 mmol, 3 eq), lithium chloride (1.65 g, 39.0 mmol, 3 eq) and tetrakis(triphenylphosphine)palladium(0) (0.45 g, 0.39 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min and then purged with argon and heated under reflux for 23 h. The mixture was then cooled to room temperature and diluted with ethyl acetate (100 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 65%
  • 1H NMR (400 MHz, CDCl3) δ 8.50 (br.s, 1H), 7.71 (dd, 1H), 7.58 (dd, 1H), 7.15 (dt,
  • 1H), 6.87-6.80 (m, 3H), 5.62 (br.s, 1H), 3.85-3.51 (m, 4H), 3.50-3.20 (m, 4H), 2.29-2.04 (m, 2H), 2.00-1.80 (m, 2H), 1.47 (s, 9H), 1.29-1.14 (m, 6H)
  • Mass Spectral Analysis m/z=492.39 (M+H)+
  • Preparation of 42.2
  • 42.1 (1 g, 2.03 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=492.36 (M+H)+
  • Chiral HPLC Method: tR=6.611 min (ee>99%)
  • Preparation of 42A
  • Hydrochloric acid, 4M, in 1,4-dioxane (1.1 mL, 4.26 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.2 (0.38 g, 0.77 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 90%
  • 1H NMR (400 MHz, DMSO d6) δ 9.23 (br s, 2H), 8.56 (d, 1H), 7.89 (dd, 1H), 7.62 (d, 1H), 7.26 (m, 1H), 6.99 (d, 1H), 6.95 (m, 2H), 6.03 (s, 1H), 3.79 (br s, 2H), 3.46 (q, 2H), 3.29 (q, 2H), 3.21 (brs, 2H), 3.10 (br s, 1H), 2.26 (m, 2H), 2.17 (m, 1H), 1.95 (m, 2H), 1.79 (m, 1H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=392.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O2, 1.25HCl, 0.75H2O
  • Theory: % C, 63.97; % H, 7.10; % N, 9.33; % Cl, 9.83.
  • Found: % C, 63.78; % H, 7.04; % N, 9.17; % Cl, 9.81.
  • [α]D 25=−1.93 (c. 0.01, MeOH)
  • Example 42B Preparation of 42.3
  • 42.1 (1 g, 2.03 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=492.29 (M+H)+
  • Chiral HPLC Method: tR=8.399 min (ee>99%)
  • Preparation of 42B
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.1 mL, 4.35 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.3 (0.39 g, 0.79 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 90%
  • 1H NMR (400 MHz, DMSO d6) δ 9.18 (br s, 2H), 8.56 (d, 1H), 7.87 (dd, 1H), 7.62 (d, 1H), 7.26 (m, 1H), 7.01 (d, 1H), 6.95 (m, 2H), 6.03 (s, 1H), 3.61 (br s, 2H), 3.46 (q, 2H), 3.29 (q, 2H), 3.22 (br s, 2H), 3.10 (br s, 1H), 2.56 (m, 2H), 2.17 (m, 1H), 1.95 (m, 2H), 1.79 (m, 1H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=392.30 (M+H)+
  • Elemental analysis:
  • C24H29N3O2, 1.25HCl, 0.75H2O
  • Theory: % C, 63.97; % H, 7.10; % N, 9.33; % Cl, 9.83.
  • Found: % C, 64.04; % H, 7.03; % N, 9.18; % Cl, 9.43.
  • [α]D 25=+0.57 (c. 0.01, MeOH)
  • Example 42C Preparation of 42C
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.1 (0.49 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the suspension was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration.
  • Yield: 90%
  • 1HNMR (400 MHz, DMSO d6) δ 9.44 (brs, 2H), 8.58 (d, 1H), 7.90 (dd, 1H), 7.62 (d, 1H), 7.26 (m, 1H), 7.01 (d, 1H), 6.93 (m, 2H), 6.04 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 3.15 (brm, 4H), 2.28 (brm, 2H), 2.16 (m, 1H), 1.97 (m, 2H), 1.81 (brm, 1H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=392.4 (M+H)+
  • Example 42D Preparation of 42.4
  • A solution of 1.14 (5.58 g, 22.0 mmol), potassium acetate (5.89 g, 60.0 mmol, 3 eq) and [1,1′bis(diphenylphosphino)ferrocene]palladium(II) chloride Pd(dppf)Cl2 (0.44 g, 0.60 mmol, 0.03 eq) in anhydrous N,N-dimethylformamide (30 mL) was evacuated for 2 min and then purged with argon and heated to 85° C. A solution of 21.6 (9.27 g, 20.0 mmol) in anhydrous N,N-dimethylformamide (20 mL) was added to this reaction mixture and the resultant mixture was stirred at 85° C. under argon for 22 h. The mixture was then cooled to room temperature, N,N-dimethylformamide was removed under reduced pressure and o solution of the resultant residue in ethyl acetate (150 mL) was filtered through a plug of celite. The cake was further washed with ethyl acetate (50 mL), the organic layer was then washed with water (2×250 mL), brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 80%
  • 1H NMR (400 MHz, CDCl3), δ 7.69 (dd, 1H), 7.10 (dt, 1H), 6.88 (dt, 1H), 6.80 (d, 1H), 6.31 (s, 1H), 3.77-3.52 (m, 2H), 3.36-3.23 (m, 2H), 2.17-2.05 (m, 3H), 2.00 (m, 2H), 1.81-1.53 (m, 3H), 1.46 (s, 9H); 1.32 (s, 12H)
  • Preparation of 42.5
  • To a solution of 42.4 (7.94 g, 18.0 mmol, 1 eq) and 34.1a (4.62 g, 18.0 mmol) in dimethoxyethane (DME) (130 mL) was added sequentially a 2N aqueous solution of sodium carbonate (27 mL, 54.0 mmol, 3 eq), lithium chloride (2.29 g, 54.0 mmol, 3 eq), and tetrakis(triphenylphosphine)palladium(0) (0.62 g, 0.54 mmol, 0.03 eq). The mixture was evacuated and then purged with argon and heated under reflux for 17 h. The mixture was then cooled to room temperature and diluted with ethyl acetate (125 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 75%
  • 1H NMR (400 MHz, CDCl3) δ 8.65 (m, 1H), 7.70 (d, 1H), 7.41 (dd, 1H), 7.20-7.10 (m, 2H), 6.90-6.80 (m, 2H), 5.91 and 5.89 (s, 1H), 3.78-3.62 (m, 1H), 3.60-3.47 (m, 4H), 3.35-3.20 (m, 4H), 2.21-2.13 (m, 2H), 1.92-1.75 (m, 1H), 1.73-1.62 (m, 2H), 1.43 and 1.41 (s, 9H), 1.27-1.15 (m, 6H)
  • Mass Spectral Analysis m/z=492.37 (M+H)+
  • Preparation of 42.6
  • 42.5 (1 g, 2.03 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 50%
  • Mass Spectral Analysis m/z=492.84 (M+H)+
  • Chiral HPLC Method: tR=9.178 min (ee=97.62%)
  • Preparation of 42D
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.40 mL, 5.59 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.6 (0.50 g, 1.02 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the suspension was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration.
  • Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (br s, 2H), 8.65 (d, 1H), 7.91 (dd, 1H), 7.60 (d, 1H), 7.29 (d, 1H), 7.25 (t, 1H), 6.98 (d, 1H), 6.92 (t, 1H), 6.18 (s, 1H), 4.23 (br s, 2H), 3.47 (m, 2H), 3.24 (m, 4H), 3.10 (m, 1H), 2.26 (m, 2H), 2.15 (m, 1H), 1.99 (m, 2H), 1.78 (m, 1H), 1.18 (br s, 3H), 1.10 (br s, 3H)
  • Mass Spectral Analysis m/z=392.81 (M+H)+
  • Elemental analysis:
  • C24H29N3O2, 1HCl, 1.25H2O
  • Theory: % C, 63.99; % H, 7.27; % N, 9.33.
  • Found: % C, 63.90; % H, 6.98; % N, 9.14.
  • [α]D 25=−1.48 (c. 0.01, MeOH)
  • Example 42E Preparation of 42.7
  • 42.5 (1 g, 2.03 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 50%
  • Mass Spectral Analysis m/z=492.84 (M+H)+
  • Chiral HPLC Method: tR=12.364 min. (ee=96.90%)
  • Preparation of 42E
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.40 mL, 5.59 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.7 (0.50 g, 1.02 mmol, 1 eq) in anhydrous methanol (5 mL). The reaction mixture was warmed to room temperature and stiffing was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.27 (br s, 2H), 8.65 (d, 1H), 7.93 (dd, 1H), 7.61 (d, 1H), 7.28 (d, 1H), 7.25 (t, 1H), 6.98 (d, 1H), 6.92 (t, 1H), 6.18 (s, 1H), 4.40 (br s, 2H), 3.47 (m, 2H), 3.24 (m, 4H), 3.10 (m, 1H), 2.27 (m, 2H), 2.15 (m, 1H), 1.99 (m, 2H), 1.79 (m, 1H), 1.17 (br s, 3H), 1.10 (br s, 3H)
  • Mass Spectral Analysis m/z=392.80 (M+H)+
  • Elemental analysis:
  • C24H29N3O2, 1HCl, 1.25H2O
  • Theory: % C, 63.99; % H, 7.27; % N, 9.33.
  • Found: % C, 64.02; % H, 7.08; % N, 9.11.
  • [α]D 25=−2.83 (c. 0.01, MeOH)
  • Example 42F Preparation of 42F
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.5 (0.49 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.42 (brs, 2H), 8.69 (d, 1H), 7.99 (dd, 1H), 7.67 (d, 1H), 7.26 (m, 2H), 7.00 (d, 1H), 6.93 (t, 1H), 6.21 (s, 1H), 3.48 (brs, 2H), 3.18 (brm, 6H), 2.28 (brm, 2H), 2.16 (m, 1H), 1.98 (m, 2H), 1.78 (brm, 1H), 1.14 (brd, 6H)
  • Mass Spectral Analysis m/z=392.4 (M+H)+
  • Example 42G Preparation of 42.8
  • To a solution of 42.4 (4.41 g, 10.0 mmol) and 35.8 (3.45 g, 9.50 mmol, 0.95 eq) in dimethoxyethane (DME) (60 mL) was added sequentially a 2N aqueous solution of sodium carbonate (15 mL, 30.0 mmol, 3 eq), lithium chloride (1.27 g, 30.0 mmol, 3 eq), and tetrakis(triphenylphosphine)palladium(0) (0.58 g, 0.50 mmol, 0.03 eq). The mixture was evacuated and then purged with argon and heated under reflux for 20 h. The mixture was then cooled to room temperature and diluted with ethyl acetate (50 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate; the organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product was triturated with a 10:1 hexane/ether mixture and the resultant colorless crystalline precipitate was collected by vacuum filtration. The filtrate was collected, concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 42%
  • 1H NMR (300 MHz, CDCl3) δ 7.22-7.16 (m, 2H), 7.13-7.07 (m, 1H), 7.03 (dd, 1H), 6.89-6.85 (m, 1H), 6.79-6.69 (m, 2H), 5.58 (s, 1H), 5.05 (s, 2H), 3.74-3.48 (m, 4H), 3.40-3.25 (m, 4H), 3.32 (s, 3H), 2.29-2.08 (m, 2H), 1.97-1.67 (m, 4H), 1.48 (s, 9H), 1.32-1.12 (m, 6H)
  • Mass Spectral Analysis m/z=551.45 (M+H)+
  • Preparation of 42.9
  • 42.8 (1 g, 1.82 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Isopropanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 30%
  • Mass Spectral Analysis m/z=551.84 (M+H)+
  • Chiral HPLC Method: tR=9.796 min (ee=97.60%)
  • Preparation of 42G
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.83 mL, 3.30 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.9 (0.33 g, 0.60 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration.
  • Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.78 (s, 1H), 9.12 (br s, 2H), 7.13 (m, 2H), 6.89 (m, 2H), 6.83 (m, 2H), 6.69 (m, 1H), 5.73 (s, 1H), 3.42 (br s, 2H), 3.25 (br s, 5H), 3.09 (br s, 1H), 2.20 (m, 3H), 1.95 (m, 2H), 1.77 (m, 1H), 1.12 (br s, 6H)
  • Mass Spectral Analysis m/z=407.7 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl
  • Theory: % C, 67.78; % H, 7.05; % N, 6.32.
  • Found: % C, 67.50; % H, 6.93; % N, 6.18.
  • [α]D 25=+1.11 (c. 0.01, MeOH)
  • Example 42H Preparation of 42.10
  • 42.8 (1 g, 1.82 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Isopropanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 30%
  • Mass Spectral Analysis m/z=551.97 (M+H)+
  • Chiral HPLC Method: tR=15.281 min. (ee=98.30%)
  • Preparation of 42H
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.83 mL, 3.30 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.10 (0.33 g, 0.60 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.78 (s, 1H), 9.10 (br s, 2H), 7.13 (m, 2H), 6.89 (m, 2H), 6.83 (m, 2H), 6.69 (m, 1H), 5.73 (s, 1H), 3.42 (br s, 2H), 3.25 (br s, 5H), 3.09 (br s, 1H), 2.20 (m, 3H), 1.95 (m, 2H), 1.77 (m, 1H), 1.12 (br s, 6H)
  • Mass Spectral Analysis m/z=407.8 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 0.25H2O
  • Theory: % C, 67.10; % H, 7.10; % N, 6.26.
  • Found: % C, 67.13; % H, 7.04; % N, 6.19.
  • [α]D 25=−5.36 (c. 0.01, MeOH)
  • Example 42I Preparation of 42I
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 42.8 (0.55 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.81 (brs, 1H), 9.22 (brs, 2H), 7.14 (m, 2H), 6.90 (m, 2H), 6.82 (m, 2H), 6.69 (dd, 1H), 5.72 (s, 1H), 3.77 (brs, 3H), 3.42 (brs, 2H), 3.16 (brm, 3H), 2.20 (m, 3H), 1.95 (m, 2H), 1.76 (brm, 1H), 1.12 (brs, 6H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Example 43A Preparation of 43.1
  • To a round bottomed flask was added successively 11.1 (7.60 g, 50.0 mmol) followed by pyrrolidine (8.3 mL, 100.0 mmol, 2 eq), 21.4 (10.66 g, 50.0 mmol) and a minimum amount of methanol used to wash any remaining material. The resultant reaction was heated to 80° C. for 30 min to dissolve all solids. The mixture was then cooled to room temperature and diluted with ethyl acetate (50 mL). The mixture was washed with a 1N aqueous solution of hydrochloric acid, water, brine and dried over sodium sulfate. The crude material was triturated with hexanes and then left to stand at room temperature over 2 days. A pale yellow solid was formed which was filtered, washed with hexanes and collected. Yield: 65%
  • 1H NMR (400 MHz, CDCl3) δ 11.61 and 11.60 (s, 1H), 7.34 (t, 1H), 6.50-6.40 (m, 2H), 3.80-3.49 (m, 2H), 3.38-3.21 (m, 2H), 2.27-2.10 (m, 2H), 2.08-1.57 (m, 4H), 1.46 (s, 9H)
  • Preparation of 43.2
  • To a solution of 43.1 (41.69 g, 0.12 mol) and N,N-diisopropylethylamine (62.7 mL, 0.36 mol, 3 eq) in dichloromethane (200 mL) was added dropwise, 11.3 (27.5 mL, 0.36 mol, 3 eq) under argon. The resultant reaction mixture was heated under reflux for 16 h then allowed to cool to room temperature. The mixture was concentrated to remove the majority of dichloromethane then diluted with ethyl acetate (200 mL) and washed with a 2N aqueous solution of hydrochloric acid until the aqueous layer was acidic. The organic layer was washed with brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 68%
  • 1H NMR (400 MHz, CDCl3) δ 7.34 (t, 1H), 6.70 (d, 1H), 6.60 (d, 1H), 5.27 (s, 2H), 3.67-3.44 (m, 2H), 3.52 (s, 3H), 3.37-3.23 (m, 2H), 2.80-2.58 (m, 2H), 2.13-2.09 (m, 2H), 2.05-1.54 (m, 4H), 1.45 (s, 9H)
  • Preparation of 43.3
  • To a solution of 43.2 (32.11 g, 82.0 mmol) in tetrahydrofuran (275 mL) at −78° C. under argon was added drop wise, a 1.0M solution of LiHMDS in tetrahydrofuran (95 mL). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (33.94 g, 1.16 eq) in tetrahydrofuran (175 mL) was added drop wise to the reaction mixture. The mixture was warmed slowly to room temperature and stirring was continued for a further 12 h at room temperature. The mixture was then poured into ice water and the two phases were separated. The organic phase was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 87%.
  • 1H NMR (300 MHz, CDCl3) δ 7.14 (t, 1H), 6.77 (d, 1H), 6.55 (d, 1H), 5.48 (s, 1H), 5.21 (s, 2H), 3.78-3.43 (m, 2H), 3.49 (s, 3H), 3.33-3.20 (m, 2H), 2.27-2.13 (m, 2H), 2.11-1.95 (m, 1H), 1.88-1.57 (m, 3H), 1.48 (s, 9H)
  • Preparation of 43.4
  • To a solution of 43.3 (10.47 g, 20.0 mmol) in dimethoxyethane (DME) (175 mL) was added sequentially a 2N aqueous solution of sodium carbonate (30.0 mL, 60.0 mmol, 3 eq), lithium chloride (2.54 g, 60.0 mmol, 3 eq), 1.6 (4.20 g, 19.0 mmol, 0.95 eq) and tetrakis(triphenylphosphine)palladium(0) (0.69 g, 0.6 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min and then purged with argon and heated under reflux for 18 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (120 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate; the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 60%
  • 1H NMR (300 MHz, CDCl3) δ 7.35-7.24 (m, 4H), 7.13 (t, 1H), 6.65 (d, 2H), 5.55 (s, 1H), 4.67 (s, 2H), 3.81-3.45 (m, 4H), 3.37-3.20 (m, 4H), 3.18 (s, 3H), 2.22-2.10 (m, 2H), 1.97-1.64 (m, 4H), 1.49 and 1.48 (s, 9H), 1.30-1.04 (m, 6H)
  • Mass Spectral Analysis m/z=551.50 (M+H)+
  • Preparation of 43.5
  • 43.4 (1 g, 1.81 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=551.49 (M+H)+
  • Chiral HPLC Method: tR=5.305 min (ee>99%)
  • Preparation of 43A
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.0 mL, 4.09 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 43.5 (0.41 g, 0.74 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the reaction mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration.
  • Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.49 (s, 1H), 9.14 (br s, 2H), 7.25 (s, 4H), 7.04 (t, 1H), 6.49 (d, 1H), 6.42 (d, 1H), 5.70 (s, 1H), 3.39 (s, 2H), 3.21 (br s, 4H), 3.11 (br s, 2H), 2.18 (m, 2H), 2.10 (m, 1H), 1.92 (m, 2H), 1.75 (m, 1H), 1.10 (br s, 6H)
  • Mass Spectral Analysis m/z=407.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 1.75H2O
  • Theory: % C, 63.28; % H, 7.33; % N, 5.90.
  • Found: % C, 63.36; % H, 7.07; % N, 5.71.
  • [α]D 25=+0.53 (c. 0.01, MeOH)
  • Example 43B Preparation of 43.6
  • 43.4 (1 g, 1.81 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=551.43 (M+H)+
  • Chiral HPLC Method: tR=6.361 min (ee=98.52%)
  • Preparation of 43B
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.1 mL, 4.59 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 43.6 (0.46 g, 0.83 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.49 (s, 1H), 9.13 (br s, 2H), 7.25 (s, 4H), 7.04 (t, 1H), 6.49 (d, 1H), 6.42 (d, 1H), 5.70 (s, 1H), 3.39 (br s, 2H), 3.22 (br s, 4H), 3.11 (br s, 2H), 2.18 (m, 2H), 2.11 (m, 1H), 1.92 (m, 2H), 1.75 (m, 1H), 1.11 (br s, 6H)
  • Mass Spectral Analysis m/z=407.3 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 1.75H2O
  • Theory: % C, 63.28; % H, 7.33; % N, 5.90.
  • Found: % C, 63.13; % H, 7.14; % N, 5.81.
  • [α]D 25=−1.43 (c. 0.01, MeOH)
  • Example 43C Preparation of 43C
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 43.4 (0.55 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.49 (s, 1H), 9.13 (brs, 2H), 7.25 (s, 4H), 7.04 (t, 1H), 6.48 (d, 1H), 6.42 (d, 1H), 5.70 (s, 1H), 3.40 (brs, 2H), 3.16 (brm, 6H), 2.18 (brm, 2H), 2.10 (m, 1H), 1.90 (m, 2H), 1.75 (brm, 1H), 1.11 (brs, 6H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Example 43D Preparation of 43.7
  • To a solution of 43.3 (6.81 g, 13.0 mmol) and 1.7 (3.95 g, 13.0 mmol, 1 eq) in dimethoxyethane (DME) (125 mL) was added sequentially a 2N aqueous solution of sodium carbonate (19.5 mL, 39.0 mmol, 3 eq), lithium chloride (1.65 g, 39.0 mmol, 3 eq) and tetrakis(triphenylphosphine)palladium(0) (0.45 g, 0.39 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min and then purged with argon and heated under reflux for 20 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (100 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 59%
  • 1H NMR (300 MHz, CDCl3) δ 8.41 (br.s, 1H), 7.62 (dd, 1H), 7.57 (dd, 1H), 7.13 (t, 1H), 6.69 (d, 2H), 5.56 (br.s, 1H), 4.70 (s, 2H), 3.82-3.60 (m, 1H), 3.55 (q, 2H), 3.40 (q, 2H), 3.37-3.22 (m, 3H), 3.17 (s, 3H), 2.25-2.10 (m, 2H), 2.10-1.82 (m, 1H), 1.80-1.63 (m, 3H), 1.45 (s, 9H), 1.29 (t, 3H), 1.16 (t, 3H)
  • Mass Spectral Analysis m/z=552.50 (M+H)+
  • Preparation of 43.8
  • 43.7 (1 g, 1.81 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=552.47 (M+H)+
  • Chiral HPLC Method: tR=6.387 min (ee>99%)
  • Preparation of 43D
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.9 mL, 3.48 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 43.8 (0.35 g, 0.63 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 80%
  • 1HNMR (400 MHz, DMSO d6) δ 9.75 (brs, 1H), 9.37 (brs, 2H), 8.45 (d, 1H), 7.77 (dd, 1H), 7.53 (d, 1H), 7.06 (t, 1H), 6.49 (m, 2H), 5.87 (s, 1H), 3.46 (q, 2H), 3.29 (q, 2H), 3.15 (brm, 4H), 2.22 (brm, 2H), 2.11 (m, 1H), 1.94 (brm, 2H), 1.78 (brm, 1H), 1.17 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 2HCl, 1.75H2O
  • Theory: % C, 56.31; % H, 6.79; % N, 8.21; % Cl, 13.85.
  • Found: % C, 56.36; % H, 6.73; % N, 7.94; % Cl, 13.59.
  • [α]D 25=+1.76 (c. 0.01, MeOH)
  • Example 43E Preparation of 43.9
  • 43.7 (1 g, 1.81 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=552.42 (M+H)+
  • Chiral HPLC Method: tR=7.915 min (ee=98.36%)
  • Preparation of 43E
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.9 mL, 3.69 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 43.9 (0.37 g, 0.67 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 74%
  • 1HNMR (400 MHz, DMSO d6) δ 9.72 (brs, 1H), 9.32 (brs, 2H), 8.44 (d, 1H), 7.75 (dd, 1H), 7.51 (d, 1H), 7.06 (t, 1H), 6.48 (m, 2H), 5.86 (s, 1H), 3.46 (q, 2H), 3.29 (q, 2H), 3.15 (brm, 4H), 2.22 (brm, 2H), 2.12 (m, 1H), 1.93 (brm, 2H), 1.78 (brm, 1H), 1.16 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 2HCl, 2H2O
  • Theory: % C, 55.82; % H, 6.83; % N, 8.14; % Cl, 13.73.
  • Found: % C, 55.56; % H, 6.71; % N, 7.84; % Cl, 13.38.
  • [α]D 25=−1.42 (c. 0.01, MeOH)
  • Example 43F Preparation of 43F
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 43.7 (0.55 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.71 (brs, 1H), 9.31 (brs, 2H), 8.44 (d, 1H), 7.74 (dd, 1H), 7.51 (d, 1H), 7.06 (t, 1H), 6.48 (m, 2H), 5.86 (s, 1H), 3.46 (q, 2H), 3.29 (q, 2H), 3.15 (brm, 4H), 2.22 (brm, 2H), 2.11 (m, 1H), 1.93 (m, 2H), 1.78 (brm, 1H), 1.16 (t, 3H), 1.09 (t, 3H)
  • Mass Spectral Analysis m/z=408.4 (M+H)+
  • Example 44A Preparation of 44.1
  • To a roud bottomed flas was added successively 1.1d (13.87 g, 0.09 mol, 0.90 eq) followed by pyrrolidine (20.7 mL, 0.25 mol, 2.5 eq), 21.4 (21.33 g, 0.10 mol) and a minimum amount of methanol used to wash any remaining material. The resultant reaction mixture was heated to 80° C. for 30 min to dissolve all solids. The mixture was then cooled to room temperature and diluted with ethyl acetate (100 mL). The mixture was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 64%
  • 1H NMR (400 MHz, CDCl3) δ 7.47 (dd, 1H), 7.24-7.15 (m, 1H), 6.92 (dd, 1H), 3.77-3.48 (m, 2H), 3.37-3.23 (m, 2H), 2.82-2.62 (m, 2H), 2.22-2.11 (m, 2H), 2.02-1.57 (m, 4H), 1.47 (s, 9H)
  • Preparation of 44.2
  • To a solution of 44.1 (14.91 g, 42.7 mmol) in tetrahydrofuran (175 mL) at −78° C. under argon was added drop wise, a 1.0M solution of LiHMDS in tetrahydrofuran (49 mL). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (17.51 g, 49.0 mmol, 1.15 eq) in tetrahydrofuran (100 mL) was added drop wise to the reaction mixture. The mixture was warmed slowly to room temperature and stirring was continued for a further 15 h at room temperature. The mixture was then poured into ice water and the two phases were separated. The organic phase was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 100% (crude).
  • Preparation of 44.3
  • To a solution of 44.2 (6.50 g, 13.5 mmol) and 1.7 (3.95 g, 13.0 mmol, 1 eq) in dimethoxyethane (DME) (125 mL) was added sequentially a 2N aqueous solution of sodium carbonate (20.5 mL, 41.0 mmol, 3 eq), lithium chloride (1.72 g, 40.6 mmol, 3 eq) and tetrakis(triphenylphosphine)palladium(0) (0.47 g, 0.41 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 17 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (100 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 64%
  • 1H NMR (300 MHz, CDCl3) δ 8.52 (br.s, 1H), 7.72 (dd, 1H), 7.63 (dd, 1H), 6.61 (d, 1H), 5.69 (br.s, 1H), 3.84-3.52 (m, 4H), 3.43 (q, 2H), 3.38-3.25 (m, 2H), 2.24-2.00 (m, 2H), 1.82-1.65 (m, 4H), 1.47 (s, 9H), 1.29 (t, 3H), 1.19 (t, 3H)
  • Mass Spectral Analysis m/z=510.44 (M+H)+
  • Preparation of 44.4
  • 44.3 (1 g, 1.96 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=510.37 (M+H)+
  • Chiral HPLC Method: tR=7.430 min (ee>99%)
  • Preparation of 44A
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.1 mL, 4.32 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 44.4 (0.40 g, 0.78 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.32 (brs, 2H), 8.59 (d, 1H), 7.92 (dd, 1H), 7.62 (d, 1H), 7.11 (m, 1H), 7.05 (m, 1H), 6.74 (dd, 1H), 6.13 (s, 1H), 3.47 (q, 2H), 3.30 (q, 2H), 3.15 (brm, 4H), 2.27 (brm, 2H), 2.15 (m, 1H), 1.96 (brm, 2H), 1.80 (brm, 1H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=410.80 (M+H)+
  • Elemental analysis:
  • C24H28FN3O2, 1HCl, 1.5H2O
  • Theory: % C, 60.95; % H, 6.82; % N, 8.88.
  • Found: % C, 60.93; % H, 6.68; % N, 8.73.
  • [α]25=−2.77 (c. 0.01, MeOH)
  • Example 44B Preparation of 44.5
  • 44.3 (1 g, 1.96 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • Mass Spectral Analysis m/z=510.97 (M+H)+
  • Chiral HPLC Method: tR=11.689 min. (ee=98.2%)
  • Preparation of 44B
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.1 mL, 4.32 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 44.5 (0.40 g, 0.78 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 80%
  • 1HNMR (400 MHz, DMSO d6) δ 9.30 (brs, 2H), 8.59 (d, 1H), 7.92 (dd, 1H), 7.62 (d, 1H), 7.12 (m, 1H), 7.05 (m, 1H), 6.74 (dd, 1H), 6.13 (s, 1H), 3.47 (q, 2H), 3.30 (q, 2H), 3.15 (brm, 4H), 2.27 (brm, 2H), 2.15 (m, 1H), 1.96 (brm, 2H), 1.80 (brm, 1H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=410.7 (M+H)+
  • Elemental analysis:
  • C24H28FN3O2, 2HCl, 1H2O
  • Theory: % C, 57.60; % H, 6.45; % N, 8.40.
  • Found: % C, 57.68; % H, 6.32; % N, 8.17.
  • [α]D 25=−1.50 (c. 0.01, MeOH)
  • Example 44C Preparation of 44C
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 44.3 (0.51 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.36 (brs, 2H), 8.59 (d, 1H), 7.92 (dd, 1H), 7.62 (d, 1H), 7.12 (m, 1H), 7.05 (m, 1H), 6.74 (dd, 1H), 6.13 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 3.16 (brm, 4H), 2.27 (brm, 2H), 2.12 (m, 1H), 1.96 (m, 2H), 1.80 (brm, 1H), 1.18 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=410.41 (M+H)+
  • Example 44D Preparation of 44.6
  • A solution of 1.14 (3.56 g, 14.0 mmol), potassium acetate (4.42 g, 45.0 mmol, 3 eq) and Pd(dppf)Cl2 (0.33 g, 0.45 mmol, 0.03 eq) in anhydrous N,N dimethylformamide (25 mL) was evacuated for 2 min, purged with argon and heated to 85° C. A solution of 44.2 (7.22 g, 15.0 mmol) in anhydrous N,N dimethylformamide (15 mL) was added to this reaction mixture, which was stirred at 85° C. under argon for 18 h. The mixture was then cooled to room temperature. The N,N dimethylformamide was removed under reduced pressure. The resultant residue was dissolved in ethyl acetate (120 mL) and filtered through a plug of celite. The cake was further washed with ethyl acetate (35 mL), the organic layer was then washed with water, brine and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 64% (crude)
  • 1H NMR (400 MHz, CDCl3), δ 7.13-7.05 (m, 1H), 6.90-6.76 (m, 2H), 6.25 (br.s, 1H), 3.78-3.49 (m, 2H), 3.35-3.2 (m, 2H), 2.08-2.03 (m, 3H), 1.83-1.51 (m, 3H), 1.45 (s, 9H); 1.32 (s, 12H)
  • Preparation of 44.7
  • To a solution of 44.6 (4.37 g, 9.5 mmol, 1 eq) and 34.1a (2.44 g, 9.5 mmol) in dimethoxyethane (DME) (75 mL) was added sequentially a 2N aqueous solution of sodium carbonate (14.25 mL, 28.5 mmol, 3 eq), lithium chloride (1.21 g, 28.5 mmol, 3 eq), and tetrakis(triphenylphosphine)palladium(0) (0.35 g, 0.54 mmol, 0.03 eq). The mixture was evacuated, purged with argon and heated under reflux for 22 h. The mixture was then cooled to room temperature and diluted with ethyl acetate (80 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 70%
  • 1H NMR (400 MHz, CDCl3) δ 8.69 (br.s, 1H), 7.78 (dd, 1H), 7.45 (dd, 1H), 6.96 (dd, 1H), 6.92-6.85 (m, 2H), 5.98 and 5.97 (s, 1H), 3.82-3.53 (m, 4H), 3.38-3.25 (m, 4H), 2.25-2.03 (m, 2H), 1.96-1.65 (m, 4H), 1.47 and 1.45 (s, 9H), 1.30-1.15 (m, 6H)
  • Mass Spectral Analysis m/z=510.35 (M+H)+
  • Preparation of 44.8
  • 44.7 (1 g, 1.96 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 30%
  • Mass Spectral Analysis m/z=510.89 (M+H)+
  • Chiral HPLC Method: tR=8.818 min (ee=98.98%)
  • Preparation of 44D
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.8 mL, 3.24 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 44.8 (0.30 g, 0.59 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 95%
  • 1HNMR (400 MHz, DMSO d6) δ 9.35 (brd, 2H), 8.67 (d, 1H), 7.95 (dd, 1H), 7.69 (d, 1H), 7.21 (dd, 1H), 7.10 (m, 1H), 7.03 (m, 1H), 6.31 (s, 1H), 3.49 (brm, 2H), 3.24 (brm, 5H), 3.09 (brs, 1H), 2.28 (brm, 2H), 2.15 (m, 1H), 1.98 (m, 2H), 1.79 (brm, 1H), 1.14 (brd, 6H)
  • Mass Spectral Analysis m/z=410.8 (M+H)+
  • Elemental analysis:
  • C24H28FN3O2, 1HCl, 1.75H2O
  • Theory: % C, 60.37; % H, 6.86; % N, 8.80.
  • Found: % C, 60.32; % H, 6.61; % N, 8.56.
  • [α]D 25=−3.34 (c. 0.01, MeOH)
  • Example 44E Preparation of 44.9
  • 44.7 (1 g, 1.96 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 36%
  • Mass Spectral Analysis m/z=510.87 (M+H)+
  • 1HNMR (400 MHz, DMSO d6) δ 8.64 (m, 1H), 7.90 (m, 1H), 7.65 (d, 1H), 7.19 (m, 1H), 7.06 (m, 1H), 6.96 (m, 1H), 6.28 (s, 0.5H), 6.25 (s, 0.5H), 3.48 (brm, 4H), 3.28 (brm, 4H), 2.12-1.87 (brm, 4H), 1.72 (brm, 2H), 1.43 (s, 4.5H), 1.41 (s, 4.5H), 1.14 (brd, 6H)
  • Chiral HPLC Method: tR=11.120 min. (ee=98.17%)
  • Preparation of 44E
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.0 mL, 3.89 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 44.9 (0.36 g, 0.71 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.46 (brd, 2H), 8.70 (d, 1H), 8.00 (dd, 1H), 7.72 (d, 1H), 7.21 (dd, 1H), 7.11 (m, 1H), 7.04 (m, 1H), 6.34 (s, 1H), 3.57 (s, 2H), 3.48 (m, 2H), 3.25 (m, 4H), 3.10 (m, 1H), 2.28 (m, 2H), 2.15 (m, 1H), 1.98 (m, 2H), 1.79 (m, 1H), 1.18 (m, 3H), 1.11 (m, 3H)
  • Mass Spectral Analysis m/z=410.8 (M+H)+
  • [α]D 25=+1.87 (c. 0.01, MeOH)
  • Example 44F Preparation of 44F
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 44.7 (0.51 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.39 (brd, 2H), 8.68 (d, 1H), 7.96 (dd, 1H), 7.69 (d, 1H), 7.21 (dd, 1H), 7.10 (m, 1H), 7.03 (m, 1H), 6.32 (s, 1H), 3.48 (brs, 2H), 3.18 (brm, 6H), 2.28 (brm, 2H), 2.15 (m, 1H), 1.97 (m, 2H), 1.80 (brm, 1H), 1.14 (brd, 6H)
  • Mass Spectral Analysis m/z=410.4 (M+H)+
  • Example 45A Preparation of 45.1
  • To a solution of 44.6 (4.44 g, 9.70 mmol) and 35.8 (3.45 g, 9.50 mmol, 0.98 eq) in dimethoxyethane (DME) (60 mL) was added sequentially a 2N aqueous solution of sodium carbonate (15 mL, 30.0 mmol, 3 eq), lithium chloride (1.27 g, 30.0 mmol, 3 eq), and tetrakis(triphenylphosphine)palladium(0) (0.56 g, 0.48 mmol, 0.03 eq). The mixture was evacuated, purged with argon and heated under reflux for 20 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (50 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate; the organic layer was washed with brine and dried over sodium sulfate and concentrated. The crude product was triturated with a 10:1 hexane/ether mixture and the resultant colorless crystalline precipitate was filtered. The filtrate was collected, concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 64%
  • 1HNMR (400 MHz, DMSO d6) δ 7.26 (dd, 1H), 7.12 (s, 1H), 7.00 (m, 2H), 6.90 (m, 1H), 6.36 (dd, 1H), 5.85 (s, 0.5H), 5.83 (s, 0.5H), 5.14 (s, 1H), 5.13 (s, 1H), 3.44 (brm, 4H), 3.32-3.20 (brm, 7H), 2.10-1.82 (brm, 4H), 1.73 (brm, 2H), 1.42 (s, 4.5H), 1.40 (s, 4.5H), 1.13 (brd, 6H)
  • Mass Spectral Analysis m/z=569.43 (M+H)+
  • Preparation of 45.2
  • 45.1 (1 g, 1.76 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Isopropanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 25%
  • Mass Spectral Analysis m/z=569.78 (M+H)+
  • Chiral HPLC Method: tR=11.024 min. (ee=97.96%)
  • Preparation of 45A
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.6 mL, 2.42 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 45.2 (0.25 g, 0.44 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 80%
  • 1HNMR (400 MHz, DMSO d6) δ 9.92 (s, 1H), 9.19 (brs, 2H), 7.15 (d, 1H), 7.00 (m, 1H), 6.93 (m, 2H), 6.84 (d, 1H), 6.42 (dd, 1H), 5.84 (s, 1H), 3.42 (brs, 2H), 3.23 (brm, 5H), 3.08 (brs, 1H), 2.20 (brm, 3H), 1.95 (m, 2H), 1.77 (brm, 1H), 1.12 (brs, 6H)
  • Mass Spectral Analysis m/z=425.80 (M+H)+
  • Elemental analysis:
  • C25H29FN2O3, 1HCl, 0.5H2O
  • Theory: % C, 63.89; % H, 6.65; % N, 5.96.
  • Found: % C, 63.70; % H, 6.51; % N, 5.67.
  • [α]D 25=+1.87 (c. 0.01, MeOH)
  • Example 45B Preparation of 45.3
  • 45.1 (1 g, 1.76 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Isopropanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 25%
  • 1HNMR (400 MHz, DMSO d6) δ 7.26 (d, 1H), 7.13 (s, 1H), 7.04 (d, 1H), 6.98 (m, 1H), 6.90 (m, 1H), 6.37 (dd, 1H), 5.85 (s, 0.5H), 5.83 (s, 0.5H), 5.14 (s, 1H), 5.13 (s, 1H), 4.12 (q, 2H), 3.46 (brm, 4H), 3.30-3.16 (brm, 5H), 2.14-1.84 (brm, 4H), 1.73 (brm, 2H), 1.41 (s, 4.5H), 1.40 (s, 4.5H), 1.13 (brd, 6H)
  • Mass Spectral Analysis m/z=569.98 (M+H)+
  • Chiral HPLC Method: tR=18.406 min. (ee=98.27%)
  • Preparation of 45B
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.6 mL, 2.51 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 45.3 (0.26 g, 0.46 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.94 (br s, 1H), 9.23 (br s, 2H), 7.15 (d, 1H), 6.99 (m, 1H), 6.93 (m, 2H), 6.83 (d, 1H), 6.41 (dd, 1H), 5.83 (s, 1H), 3.42 (br s, 2H), 3.24 (m, 4H), 3.09 (m, 2H), 2.24 (m, 2H), 2.15 (m, 1H), 1.95 (m, 2H), 1.77 (m, 1H), 1.12 (br s, 6H)
  • Mass Spectral Analysis m/z=425.8 (M+H)+
  • [α]D 25=−3.05 (c. 0.01, MeOH)
  • Example 45C Preparation of 45C
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 45.1 (0.57 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1HNMR (400 MHz, DMSO d6) δ 9.92 (brs, 1H), 9.20 (brs, 2H), 7.14 (d, 1H), 6.97 (m, 2H), 6.91 (s, 1H), 6.84 (d, 1H), 6.42 (dd, 1H), 5.84 (s, 1H), 3.64 (brs, 3H), 3.42 (brs, 2H), 3.18 (brm, 3H), 2.20 (m, 3H), 1.95 (m, 2H), 1.76 (brm, 1H), 1.12 (brs, 6H)
  • Mass Spectral Analysis m/z=425.4 (M+H)+
  • Example 45D Preparation of 45.4
  • To a round bottomed flask was added successively 2.1 (15.21 g, 0.1 mol) followed by pyrrolidine (20.7 mL, 0.25 mol, 2.5 eq), 21.4 (21.33 g, 0.10 mol) and a minimum amount of methanol used to wash any remaining material. The resultant reaction was heated to 80° C. for 30 min to dissolve all solids. The mixture was then cooled to room temperature and diluted with ethyl acetate (100 mL). The mixture was washed with a 1N aqueous solution of hydrochloric acid, water, brine and dried over sodium sulfate. The crude material was used for the next step without further purification. Yield: 100% (crude)
  • Preparation of 45.5
  • To a cooled (0° C.) and stiffing solution of 45.4 (34.74 g, 0.10 mol) and imidazole (14.30 g, 0.21 mol, 2.1 eq) in anhydrous N,N-dimethylformamide (200 mL) was added drop wise, a solution of 2.3 (17.33 g, 0.12 mol, 1.2 eq) in N,N-dimethylformamide. The resultant mixture was allowed to warm to room temperature and stirring was continued for 16 h at room temperature. The N,N-dimethylformamide was removed and the residue diluted with ethyl acetate. The organic solution was washed with water, brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 51%
  • 1H NMR (400 MHz, CDCl3) δ 7.22 (d, 1H), 6.95 (dd, 1H), 6.80 (d, 1H), 3.72-3.44 (m, 2H), 3.33-3.21 (m, 2H), 2.75-2.58 (m, 2H), 2.20-2.07 (m, 2H), 2.02-1.52 (m, 4H), 1.43 (s, 9H), 0.94 (s, 9H), 0.15 (s, 6H)
  • Preparation of 45.6
  • To a solution of 45.5 (23.55 g, 51.0 mmol) in tetrahydrofuran (250 mL) at −78° C. under argon was added drop wise, a 1.0M solution of LiHMDS in tetrahydrofuran (59 mL). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (21.08 g, 59.0 mmol, 1.16 eq) in tetrahydrofuran (175 mL) was added drop wise to the mixture, which was warmed slowly to room temperature. Stirring was continued for a further 15 h at room temperature. The mixture was then poured into ice water and the two phases were separated. The organic phase was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 100% (crude)
  • 1H NMR (400 MHz, CDCl3) δ 7.13 (m, 1H), 6.75-6.68 (m, 2H), 5.58 (s, 1H), 3.78-3.50 (m, 2H), 3.37-3.20 (m, 2H), 2.27-2.04 (m, 3H), 1.97-1.62 (m, 3H), 1.47 (s, 9H), 0.98 (s, 9H), 0.19 (s, 6H)
  • Preparation of 45.7
  • To a solution of 45.6 (9.50 g, 16.0 mmol) and 1.7 (4.87 g, 16.0 mmol, 1 eq) in dimethoxyethane (DME) (165 mL) was added sequentially a 2N aqueous solution of sodium carbonate (24 mL, 48.0 mmol, 3 eq), lithium chloride (2.03 g, 48.0 mmol, 3 eq) and tetrakis(triphenylphosphine)palladium(0) (0.56 g, 0.48 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 23 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (150 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate. The organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 47%
  • 1H NMR (300 MHz, CDCl3) δ 8.46 (br.s, 1H), 7.77-7.72 (m, 1H), 7.51 (dd, 1H), 6.87-6.81 (m, 1H), 6.76 (dd, 1H), 6.35 (m, 1H), 5.62 and 5.61 (s, 1H), 3.76-3.55 (m, 2H), 3.51 (q, 2H), 3.35 (q, 2H), 3.31-3.19 (m, 2H), 2.19-2.02 (m, 2H), 1.86-1.55 (m, 4H), 1.44 and 1.43 (s, 9H), 1.26-1.17 (m, 3H), 1.14 (t, 3H)
  • Mass Spectral Analysis m/z=508.37 (M+H)+
  • Preparation of 45.8
  • 45.7 (1 g, 1.97 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 40%
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (s, 1H), 8.54 (d, 1H), 7.86 (m, 1H), 7.60 (dd, 1H), 6.77 (dd, 1H), 6.62 (dd, 1H), 6.34 (d, 1H), 5.96 (s, 0.5H), 5.92 (s, 0.5H), 3.47 (brm, 4H), 3.30 (brm, 4H), 2.08-1.84 (brm, 4H), 1.70 (brm, 2H), 1.42 (s, 4.5H), 1.41 (s, 4.5H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=508.39 (M+H)+
  • Chiral HPLC Method: tR=8.583 min (ee=97.58%)
  • Preparation of 45D
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.1 mL, 4.33 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 45.8 (0.40 g, 0.79 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 95%
  • 1H NMR (400 MHz, DMSO d6) δ 9.31 (brs, 2H), 8.56 (d, 1H), 7.89 (dd, 1H), 7.62 (d, 1H), 6.82 (d, 1H), 6.65 (dd, 1H), 6.36 (d, 1H), 6.00 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 3.15 (brm, 4H), 2.22 (brm, 2H), 2.14 (m, 1H), 1.94 (m, 2H), 1.78 (brm, 1H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=408.3 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 1.9HCl, 1.6H2O
  • Theory: % C, 57.01; % H, 6.80; % N, 8.31; % Cl, 13.32.
  • Found: % C, 57.24; % H, 6.82; % N, 8.24; % Cl, 13.40.
  • [α]D 25=+1.71 (c. 0.01, MeOH)
  • Example 45E Preparation of 45.9
  • 45.7 (1 g, 1.97 mmol, 1 eq) was resolved using Chiral HPLC method:
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Ethanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • Yield: 35%
  • 1H NMR (400 MHz, DMSO d6) δ 8.99 (s, 1H), 8.54 (d, 1H), 7.86 (m, 1H), 7.60 (dd, 1H), 6.77 (dd, 1H), 6.62 (dd, 1H), 6.34 (d, 1H), 5.96 (s, 0.5H), 5.92 (s, 0.5H), 3.47 (brm, 4H), 3.30 (brm, 4H), 2.08-1.84 (brm, 4H), 1.70 (brm, 2H), 1.42 (s, 4.5H), 1.41 (s, 4.5H), 1.17 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=508.34 (M+H)+
  • Chiral HPLC Method: tR=11.101 min. (ee>99%)
  • Preparation of 45E
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.0 mL, 3.79 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 45.9 (0.35 g, 0.69 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 95%
  • 1H NMR (400 MHz, DMSO d6) δ 9.31 (brs, 2H), 8.56 (d, 1H), 7.89 (dd, 1H), 7.62 (d, 1H), 6.83 (d, 1H), 6.66 (dd, 1H), 6.36 (d, 1H), 6.00 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 3.16 (brm, 4H), 2.22 (brm, 2H), 2.13 (m, 1H), 1.94 (m, 2H), 1.78 (brm, 1H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=408.8 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 1HCl, 2.25H2O
  • Theory: % C, 59.50; % H, 7.18; % N, 8.67.
  • Found: % C, 59.37; % H, 7.05; % N, 8.40.
  • [α]D 25=−2.98 (c. 0.01, MeOH)
  • Example 45F Preparation of 45F
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 45.7 (0.51 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.38 (brs, 2H), 8.56 (d, 1H), 7.90 (dd, 1H), 7.63 (d, 1H), 6.83 (d, 1H), 6.66 (dd, 1H), 6.37 (d, 1H), 6.00 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 3.15 (brm, 4H), 2.23 (brm, 2H), 2.13 (m, 1H), 1.94 (m, 2H), 1.78 (brm, 1H), 1.18 (t, 3H), 1.12 (t, 3H)
  • Mass Spectral Analysis m/z=408.4 (M+H)+
  • Examples 46A, 46B Preparation of 46.1
  • To a solution of 45.6 (8.91 g, 15.0 mmol) in dimethoxyethane (DME) (150 mL) was added sequentially a 2N aqueous solution of sodium carbonate (22.5 mL, 45.0 mmol, 3 eq), lithium chloride (1.91 g, 45.1 mmol, 3 eq), compound 45.6 (3.32 g, 15.0 mmol, 1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.52 g, 0.45 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 42 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (120 mL) and the mixture was filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 69%
  • 1H NMR (300 MHz, CDCl3) δ 7.37-7.29 (m, 4H), 6.75 (d, 1H), 6.64 (dd, 1H), 6.60-6.50 (m, 1H), 6.48 (d, 1H), 5.60 (s, 1H), 3.77-3.48 (m, 4H), 3.36-3.18 (m, 4H), 2.21-2.04 (m, 2H), 1.87-1.61 (m, 4H), 1.47 (s, 9H), 1.31-1.08 (m, 6H)
  • Preparation of 46.2
  • To a solution of 46.1 (3.20 g, 6.32 mmol, 1.0 eq) in methylene chloride (100 mL) was added N,N-diisopropylethylamine (4.40 mL, 25.56 mmol, 4.0 eq) and then 11.3 (1.54 g, 18.95 mmol, 3.0 eq) was added drop wise to the reaction mixture. The reaction mixture was stirred for 48 h at room temperature and then poured onto water (150 mL). The layers were separated and the aqueous was washed two times with methylene chloride (50 mL). The combined organics were washed with a 0.1N aqueous solution of hydrochloric acid, a saturated aqueous solution of sodium bicarbonate and brine. The organics were dried over sodium sulfate and concentrated. The crude product was purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity). Yield: 50%;
  • Mass Spectral Analysis m/z=551.2 (M+H)+
  • 1H NMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 6.87 (m, 2H), 6.68 (d, 1H), 5.63 (s, 1H), 5.03 (s, 2H), 3.84-3.55 (brm, 4H), 3.43 (s, 3H), 3.31 (brm, 4H), 2.16 (brm, 3H), 1.94-1.68 (brm, 3H), 1.49 (s, 4.5H), 1.47 (s, 4.5H), 1.21 (brd, 6H)
  • Preparation of 46.3 and 46.4
  • The racemic 46.2 was separated into pure enantiomers 46.3 and 46.4 by chiral chromatography.
  • Column: Chiral Technologies Chiralcel OD-H, 4.6×250 mm
  • Mobile Phase: 90% Hexane/0.2% Diisopropylethylamine 10% Isopropanol
  • Flow: 1.0 mL min
  • Detector: UV 275 nm
  • 46.3: 1HNMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 6.87 (m, 2H), 6.68 (d, 1H), 5.63 (s, 1H), 5.03 (s, 2H), 3.84-3.52 (brm, 4H), 3.43 (s, 3H), 3.31 (brm, 4H), 2.16 (brm, 3H), 1.93-1.68 (brm, 3H), 1.48 (s, 4.5H), 1.47 (s, 4.5H), 1.21 (brd, 6H)
  • 46.4: 1HNMR (400 MHz, CDCl3) δ 7.39 (m, 4H), 6.87 (m, 2H), 6.68 (d, 1H), 5.63 (s, 1H), 5.03 (s, 2H), 3.82-3.52 (brm, 4H), 3.43 (s, 3H), 3.31 (brm, 4H), 2.17 (brm, 3H), 1.92-1.68 (brm, 3H), 1.48 (s, 4.5H), 1.47 (s, 4.5H), 1.21 (brd, 6H)
  • Preparation of 46A
  • To a solution of 46.3 (0.83 g, 1.51 mmol, 1.0 eq) in methanol (40 mL) under nitrogen was added a 4.0M solution of anhydrous hydrochloric acid in dioxane (3.80 mL, 15.1 mmol, 10.0 eq). The reaction was stirred for 16 h at room temperature and concentrated. The crude product was purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity). The resulting solid was triturated in diethyl ether and collected by vacuum filtration. Yield: 93%
  • 1H NMR (400 MHz, DMSO d6) δ 9.19 (brs, 2H), 9.03 (s, 1H), 7.40 (q, 4H), 6.80 (d, 1H), 6.63 (dd, 1H), 6.40 (d, 1H), 5.86 (s, 1H), 3.45 (brs, 2H), 3.28-3.02 (brm, 6H), 2.20 (brm, 2H), 2.12 (m, 1H), 1.92 (brm, 2H), 1.76 (brm, 1H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 2H2O
  • Theory: % C, 62.69; % H, 7.36; % N, 5.85.
  • Found: % C, 62.37; % H, 7.20; % N, 5.83.
  • [α]D=+1.67 (c=10.5 mg/mL, MeOH, 22.6° C.)
  • Preparation of 46B
  • To a solution of 46.4 (0.86 g, 1.56 mmol, 1.0 eq) in methanol (40 mL) under nitrogen was added a 4.0M solution of anhydrous hydrochloric acid in dioxane (3.90 mL, 15.6 mmol, 10.0 eq). The reaction was stirred for 16 h at room temperature and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity). The resulting solid was triturated in diethyl ether and collected by vacuum filtration. Yield: 85%
  • 1H NMR (400 MHz, DMSO d6) δ 9.22 (brs, 2H), 9.03 (s, 1H), 7.40 (q, 4H), 6.80 (d, 1H), 6.63 (dd, 1H), 6.40 (d, 1H), 5.86 (s, 1H), 3.45 (brs, 2H), 3.30-3.02 (brm, 6H), 2.20 (brm, 2H), 2.12 (m, 1H), 1.92 (brm, 2H), 1.76 (brm, 1H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O3, 1HCl, 2.3H2O
  • Theory: % C, 61.99; % H, 7.41; % N, 5.78.
  • Found: % C, 62.08; % H, 7.38; % N, 5.86.
  • [α]D=−1.52 (c=10.0 mg/mL, MeOH, 22.6° C.)
  • Example 46C Preparation of compound 46.1
  • To a solution of 45.6 (8.91 g, 15.0 mmol) in dimethoxyethane (DME) (150 mL) was added sequentially a 2N aqueous solution of sodium carbonate (22.5 mL, 45.0 mmol, 3 eq), lithium chloride (1.91 g, 45.1 mmol, 3 eq) 1.6 (3.32 g, 15.0 mmol, 1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.52 g, 0.45 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 42 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (120 mL) and filtered through a 1 inch plug of celite. The cake was further washed with ethyl acetate, the organic layer was washed with water followed by brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 69%
  • 1H NMR (400 MHz, DMSO d6) δ 8.94 (s, 1H), 7.39 (s, 4H), 6.75 (d, 1H), 6.60 (dd, 1H), 6.38 (d, 1H), 5.82 (s, 0.5H), 5.79 (s, 0.5H), 3.46 (brm, 4H), 3.24 (brm, 4H), 2.08-1.82 (brm, 4H), 1.68 (brm, 2H), 1.42 (s, 4.5H), 1.40 (s, 4.5H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=507.59 (M+H)+
  • Preparation of 46C
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.38 mL, 5.50 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 46.1 (0.51 g, 1.00 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. Ethyl acetate was added to the resulting oil and the mixture was stirred for 1 hour at room temperature. The resulting solids were isolated by vacuum filtration. Yield: 99%
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (brs, 2H), 9.03 (brs, 1H), 7.40 (q, 4H), 6.80 (d, 1H), 6.63 (dd, 1H), 6.40 (d, 1H), 5.86 (s, 1H), 3.43 (brs, 2H), 3.16 (brm, 6H), 2.21 (brm, 2H), 2.12 (m, 1H), 1.92 (m, 2H), 1.76 (brm, 1H), 1.13 (brd, 6H)
  • Mass Spectral Analysis m/z=407.4 (M+H)+
  • Example 47A Preparation of 47.1
  • To a solution of 23.3a (2.71 g, 6.20 mmol, 1.0 eq) and compound 1.7 (1.89 g, 6.20 mmol, 1.0 eq) in dimethoxyethane (DME) (65 mL) was added sequentially a 2N aqueous solution of sodium carbonate (9.30 mL, 18.60 mmol, 3.0 eq), lithium chloride (0.79 g, 18.60 mmol, 3.0 eq) and tetrakis(triphenylphosphine)palladium(0) (0.23 g, 0.20 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min and purged with argon and heated under reflux for 19 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (100 mL) and filtered through a 1 inch plug of celite. The cake was washed with ethyl acetate. The organics were washed with water and brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 61%
  • 1H NMR (400 MHz, CDCl3) δ 8.56 (brs, 1H), 7.75 (dd, 1H), 7.64 (d, 1H), 7.25-7.17 (m, 1H), 6.96-6.85 (m, 3H), 5.70 (brs, 1H), 3.94-3.81 (m, 1H), 3.68-3.54 (m, 3H), 3.48-3.33 (m, 3H), 2.46-2.39 (m, 1H), 2.00-1.80 (m, 1H), 1.82-1.69 (m, 1H), 1.47 (s, 9H), 1.28 (t, 3H), 1.19 (t, 3H)
  • Preparation of 47A
  • To a solution of 47.1 (1.40 g, 3.02 mmol, 1.0 eq) in methylene chloride (40 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (9.06 mL, 18.12 mmol, 6.0 eq). The reaction was warmed to room temperature and stirred for 48 h at room temperature. Diethyl ether (15 mL) was added and stiffing was continued for 10 min at room temperature. The solids were collected by vacuum filtration. Yield: 92%
  • 1H NMR (400 MHz, DMSO d6) δ 9.92 (brs, 1H), 9.70 (brs, 1H), 8.61 (m, 1H), 7.92 (dd, 1H), 7.63 (dd, 1H), 7.30 (m, 1H), 6.99 (m, 3H), 6.13 (s, 1H), 3.57 (brm, 1H), 3.45 (brm, 4H), 3.31 (m, 3H), 2.42 (brm, 1H), 2.16 (m, 1H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=364.7 (M+H)+
  • Example 47B Preparation of 47.2
  • A solution of bis(pinacolato)diboron (1.14) (4.06 g, 16.0 mmol, 1.14 eq), potassium acetate (4.12 g, 42.0 mmol, 3.0 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (0.31 g, 0.42 mmol, 0.03 eq) in anhydrous N,N-dimethylformamide (35 mL) was evacuated for 2 min, purged with argon and heated to 85° C. A solution of 23.3a (6.10 g, 14.0 mmol, 1.0 eq) in anhydrous N,N-dimethylformamide (20 mL) was added to this reaction mixture, which was stirred at 85° C. under argon for 15 h. The mixture was then cooled to room temperature and concentrated. The residue was dissolved in ethyl acetate (120 mL) and filtered through a plug of celite. The cake was washed with ethyl acetate. The organics were washed with water and brine, and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 100% (crude)
  • Preparation of 47.3
  • To a solution of 47.2 (7.94 g, 19.2 mmol, 1.0 eq) and compound 35.8 (4.72 g, 13.0 mmol, 0.68 eq) in dimethoxyethane (DME) (110 mL) was added sequentially a 2N aqueous solution of sodium carbonate (19.5 mL, 39.0 mmol, 3.0 eq), lithium chloride (1.65 g, 39.0 mmol, 3.0 eq), and tetrakis(triphenylphosphine)palladium(0) (0.45 g, 0.39 mmol, 0.03 eq). The mixture was evacuated, purged with argon and heated under reflux for 17 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (60 mL) and filtered through a 1 inch plug of celite. The filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 28%
  • 1H NMR (300 MHz, CDCl3) δ 7.26-7.02 (m, 4H), 6.82-6.69 (m, 2H), 6.85 (d, H), 5.60 (s, 1H), 5.30 (s, 2H), 3.95-3.83 (m, 1H), 3.69-3.43 (m, 3H), 3.40-3.23 (m, 3H), 3.26 (s, 3H), 2.49-2.40 (m, 1H), 2.00-1.88 (m, 1H), 1.76-1.64 (brs, 1H), 1.47 (s, 9H), 1.31-1.10 (m, 6H)
  • Preparation of 47B
  • To a solution of 47.3 (2.16 g, 4.13 mmol, 1.0 eq) in methanol (35 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (12.4 mL, 24.80 mmol, 6.0 eq). The reaction mixture was warmed to room temperature and stirred for 48 h at room temperature. The reaction was concentrated to a solid and triturated in a mixture of methylene chloride (5 mL) and diethyl ether (25 mL). The solids were collected by vacuum filtration. Yield: 100%; Mass Spectral Analysis m/z=379.8 (M+H)+
  • 1H NMR (400 MHz, DMSO d6) δ 9.85 (brs, 1H), 9.65 (brs, 1H), 9.53 (brs, 1H), 7.19 (m, 2H), 6.87 (m, 4H), 6.74 (d, 1H), 5.84 (s, 1H), 3.57 (brm, 1H), 3.43 (brm, 4H), 3.25 (brm, 3H), 2.42 (brm, 1H), 2.12 (brm, 1H), 1.12 (brs, 6H)
  • Example 47C Preparation of 47.4
  • To a roud bottomed flask was added successively 2′,6′-dihydroxyacetophenone (11.1) (9.12 g, 60.0 mmol, 1.0 eq) followed by pyrrolidine (12.4 mL, 150.0 mmol, 2.5 eq), N-Boc-pyrrolidin-3-one (23.1a) (11.11 g, 60.0 mmol, 1.0 eq) and a minimum amount of methanol used to wash any remaining material. The reaction mixture was heated to 80° C. for 30 min to dissolve all solids. The mixture was then cooled to room temperature and diluted with ethyl acetate (80 mL). The mixture was washed with a 1N aqueous solution of hydrochloric acid, water and brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 52%
  • 1H NMR (400 MHz, CDCl3) δ 11.50 (s, 1H), 7.30 (dd, 1H), 6.52-6.32 (m, 2H), 3.82-3.40 (m, 3H), 3.38-3.20 (m, 1H), 3.00-2.60 (m, 2H), 2.34-2.20 (m, 1H), 1.98-1.74 (m, 1H), 1.43 (s, 9H)
  • Preparation of 47.5
  • To a solution of 47.4 (5.15 g, 16.1 mmol, 1.0 eq) and N,N-diisopropylethylamine (8.5 mL, 48.8 mmol, 3.0 eq) in methylene chloride (30 mL) was added drop wise compound 11.3 (3.7 mL, 48.7 mmol, 3.0 eq) under argon. The reaction mixture was heated under reflux for 15 h and then allowed to cool to room temperature. The mixture was concentrated, diluted with ethyl acetate (50 mL) and washed with a 2N aqueous solution of hydrochloric acid until the aqueous layer was acidic. The organic layer was washed with brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 52%
  • Preparation of 47.6
  • To a solution of 47.5 (3.07 g, 8.4 mmol, 1.0 eq) in tetrahydrofuran (25 mL) at −78° C. under argon was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (10.1 mL, 10.1 mmol, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of N-phenyltrifluoromethanesulfonimide (1.4) (3.32 g, 9.29 mmol, 1.11 eq) in tetrahydrofuran (20 mL) was added drop wise to the reaction mixture. The mixture was warmed slowly to room temperature and stirring was continued for a further 15 h at room temperature. The mixture was poured into ice water and the two phases were separated. The organic layer was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide and brine, and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 100% (crude)
  • Preparation of 47.7
  • To a solution of 47.6 (2.34 g, 4.7 mmol, 1.0 eq) in dimethoxyethane (DME) (50 mL) was added sequentially a 2N aqueous solution of sodium carbonate (6.75 mL, 13.5 mmol, 3.0 eq), lithium chloride (0.57 g, 13.5 mmol, 3.0 eq), compound 1.6 (1.00 g, 4.5 mmol, 0.96 eq) and tetrakis(triphenylphosphine) palladium(0) (0.16 g, 0.14 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 20 h. The mixture was cooled to room temperature, diluted with ethyl acetate (40 mL) and filtered through a 1 inch plug of celite. The cake was rinsed with ethyl acetate. The filtrate was washed with water and brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 51%
  • 1H NMR (300 MHz, CDCl3) δ 7.36-7.29 (m, 4H), 7.18-7.09 (m, 1H), 6.72-6.62 (m, 2H), 5.65 (s, 1H), 4.68 (s, 2H), 3.92-3.74 (m, 2H), 3.68-3.19 (m, 6H), 3.16 (s, 3H), 2.43-2.31 (m, 1H), 2.00-1.85 (m, 1H), 1.49 (s, 4.5H), 1.47 (s, 4.5H), 1.30-1.04 (m, 6H)
  • Preparation of 47C
  • To a solution of 47.7 (1.29 g, 2.47 mmol, 1.0 eq) in methanol (20 mL) under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (7.40 mL, 14.81 mmol, 6.0 eq). The reaction was stirred for 24 h at room temperature. The reaction was concentrated to an oil and stirred in a 1:1 mixture of methylene chloride and diethyl ether for 20 min. The solids that precipitated were collected by vacuum filtration. Yield: 73%
  • 1H NMR (400 MHz, DMSO d6) δ 9.62 (s, 1H), 9.55 (brs, 1H), 9.44 (brs, 1H), 7.27 (m, 4H), 7.08 (t, 1H), 6.49 (m, 2H), 5.86 (s, 1H), 3.56-3.16 (brm, 8H), 2.36 (m, 1H), 2.07 (m, 1H), 1.11 (brs, 6H)
  • Mass Spectral Analysis m/z=379.3 (M+H)+
  • Example 47D Preparation of 47.8
  • To a solution of 47.6 (2.34 g, 4.7 mmol, 1.0 eq) and compound 1.7 (1.37 g, 4.5 mmol, 0.96 eq) in dimethoxyethane (DME) (60 mL) was added sequentially a 2N aqueous solution of sodium carbonate (6.75 mL, 13.5 mmol, 3.0 eq), lithium chloride (0.57 g, 13.5 mmol, 3.0 eq) and tetrakis(triphenylphosphine)palladium(0) (0.16 g, 0.14 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 20 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (100 mL) and filtered through a 1 inch plug of celite. The cake was rinsed with ethyl acetate. The filtrate was washed with water, brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 54%
  • 1H NMR (400 MHz, CDCl3) δ 8.42 (brs, 1H), 7.57 (dd, 1H), 7.38 (d, 1H), 7.17-7.07 (m, 1H), 6.73-6.60 (m, 2H), 5.65 (brs, 1H), 4.69 (s, 2H), 3.91-3.74 (m, 1H), 3.65-3.49 (m, 4H), 3.48-3.28 (m, 3H), 3.14 (s, 3H), 2.43-2.30 (m, 1H), 1.99-1.85 (m, 1H), 1.49 (s, 4.5H), 1.47 (s, 4.5H), 1.21 (t, 3H), 1.14 (t, 3H)
  • Preparation of 47D
  • To a solution of 47.8 (1.36 g, 2.60 mmol, 1.0 eq) in methanol (20 mL) under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (7.79 mL, 15.58 mmol, 6.0 eq). The reaction was stirred for 24 h at room temperature. The reaction was concentrated to a foamy solid and dissolved in methylene chloride (5 mL). With stiffing, diethyl ether (20 mL) was added and the mixture was stirred for 10 min at room temperature. The resulting solid was collected by vacuum filtration. Yield: 87%
  • 1H NMR (400 MHz, DMSO d6) δ 9.79 (brs, 1H), 9.57 (brs, 1H), 9.46 (brs, 1H), 8.46 (dd, 1H), 7.71 (dd, 1H), 7.49 (dd, 1H), 7.11 (t, 1H), 6.50 (m, 2H), 5.98 (s, 1H), 3.58-3.25 (brm, 8H), 2.38 (m, 1H), 2.10 (m, 1H), 1.16 (t, 3H), 1.08 (t, 3H)
  • Mass Spectral Analysis m/z=380.4 (M+H)+
  • Example 47E Preparation of 47.9
  • To a round bottomed flask was added successively 2′,5′-dihydroxyacetophenone (2.1) (10.19 g, 67.0 mmol, 0.96 eq) followed by pyrrolidine (14.5 mL, 175 mmol, 2.5 eq), N-Boc-pyrrolidin-3-one (23.1a) (12.97 g, 70.0 mmol, 1.0 eq) and a minimum amount of methanol used to wash any remaining material. The resultant reaction was heated to 80° C. for 30 min to dissolve all solids. The mixture was then cooled to room temperature and diluted with ethyl acetate (65 mL). The mixture was washed with a 1N aqueous solution of hydrochloric acid, water and brine, and dried over sodium sulfate. The crude material was used without further purification. Yield: 100% (crude)
  • Preparation of 47.10
  • To a cooled (0° C.) and stiffing solution of 47.9 (19.55 g, 61.2 mmol, 1.0 eq) and imidazole (8.78 g, 129.0 mmol, 2.11 eq) in anhydrous N,N-dimethylformamide (110 mL) was added drop wise a solution of tert-butyldimethylsilyl chloride (2.3) (10.25 g, 68.0 mmol, 1.11 eq) under argon. The mixture was allowed to warm to room temperature and was stirred for 17 h at room temperature. The N,N-dimethylformamide was removed and the residue was diluted with ethyl acetate. The organic phase was washed with water and brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 65%
  • 1H NMR (300 MHz, CDCl3) δ 7.24 (d, 1H), 7.01-6.91 (m, 1H), 6.81 (d, 1H), 3.82-3.44 (m, 3H), 3.37-3.24 (m, 1H), 2.93-2.71 (m, 2H), 2.32-2.22 (m, 1H), 1.95-1.78 (m, 1H), 1.42 (s, 9H), 0.98 (s, 9H), 0.20 (s, 6H)
  • Preparation of 47.11
  • To a solution of 47.10 (17.34 g, 40.0 mmol, 1.0 eq) in tetrahydrofuran (150 mL) at −78° C. under argon was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (48 mL, 48 mmol, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of N-phenyltrifluoromethanesulfonimide (1.4) (15.72 g, 44.00 mmol, 1.1 eq) in tetrahydrofuran (100 mL) was added dropwise to the mixture. The mixture was warmed slowly to room temperature and stiffing was continued for a further 16 h at room temperature. The mixture was then poured into ice water and the layers were separated. The organics were washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine, and dried over sodium sulfate. The crude product was used for the next step without further purification.
  • Yield: 100% (crude)
  • Preparation of Compound 47.12
  • To a solution of 47.11 (9.56 g, 17.4 mmol, 1.0 eq) in dimethoxyethane (DME) (190 mL) was added sequentially a 2N aqueous solution of sodium carbonate (21.75 mL, 43.5 mmol, 3.0 eq), lithium chloride (1.84 g, 43.5 mmol, 3.0 eq), compound 1.6 (3.20 g, 14.5 mmol, 0.83 eq) and tetrakis(triphenylphosphine)palladium(0) (0.51 g, 0.44 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 42 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (200 mL) and filtered through a 1 inch plug of celite. The cake was rinsed with ethyl acetate. The organic layer was washed with water, brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 44%
  • 1H NMR (300 MHz, CDCl3) δ 7.39-7.31 (m, 4H), 6.95-6.66 (m, 3H), 6.49 (m, 1H), 5.67 (s, 1H), 3.94-3.80 (m, 1H), 3.66-3.50 (m, 4H), 3.41-3.27 (m, 3H), 2.47-2.37 (m, 1H), 2.02-1.85 (m, 1H), 1.47 (s, 4.5H), 1.45 (s, 4.5H), (s, 9H), 1.36-1.10 (m, 6H)
  • Preparation of 47E
  • To a solution of 47.12 (3.66 g, 7.65 mmol, 1.0 eq) in methanol (40 mL) under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (15.3 mL, 30.59 mmol, 4.0 eq). The reaction mixture was stirred for 24 h, concentrated to a foam and diethyl ether (20 mL) was added. The mixture was sonicated and methylene chloride (10 mL) was added with stirring. The mixture was stirred for 20 min and the solids were collected by vacuum filtration. Yield: 85%
  • 1H NMR (400 MHz, DMSO d6) δ 9.66 (brs, 1H), 9.51 (brs, 1H), 9.14 (brs, 1H), 7.43 (m, 4H), 6.81 (d, 1H), 6.66 (dd, 1H), 6.43 (d, 1H), 5.98 (s, 1H), 3.56-3.20 (brm, 8H), 2.38 (m, 1H), 2.08 (m, 1H), 1.11 (brd, 6H)
  • Mass Spectral Analysis m/z=379.8 (M+H)+
  • Example 47F Preparation of 47.13
  • To a solution of 47.11 (9.56 g, 17.4 mmol, 1.2 eq) and compound 1.7 (4.41 g, 14.5 mmol, 1.0 eq) in dimethoxyethane (DME) (165 mL) was added sequentially a 2N aqueous solution of sodium carbonate (21.75 mL, 43.5 mmol, 3.0 eq), lithium chloride (1.84 g, 43.5 mmol, 3.0 eq) and tetrakis(triphenylphosphine)palladium(0) (0.51 g, 0.44 mmol, 0.03 eq). The reaction mixture was evacuated for 2 min, purged with argon and heated under reflux for 19 h. The mixture was then cooled to room temperature, diluted with ethyl acetate (150 mL) and filtered through a 1 inch plug of celite, which was further rinsed with ethyl acetate. The organic layer was washed with water, brine, and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 33%
  • 1H NMR (300 MHz, CDCl3) δ 8.49 (brs, 1H), 7.77 (dd, 1H), 7.55-7.41 (m, 2H), 6.80-6.67 (m, 2H), 6.39 (brs, 1H), 5.70 (s, 1H), 3.90-3.78 (m, 1H), 3.64-3.50 (m, 4H), 3.43-3.27 (m, 3H), 2.44-2.34 (m, 1H), 1.95-1.83 (m, 1H), 1.48 (s, 4.5H), 1.46 (s, 4.5H), 1.25 (t, 3H), 1.17 (t, 3H)
  • Preparation of 47F
  • To a solution of 47.13 (2.65 g, 5.53 mmol, 1.0 eq) in methanol (30 mL) under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (16.6 mL, 33.15 mmol, 6.0 eq). The reaction was stirred for 24 h at room temperature, concentrated to a foam and diethyl ether (20 mL) was added. The mixture was sonicated and methylene chloride (10 mL) was added with stiffing. The mixture was stirred for 20 min and the solids were collected by vacuum filtration.
  • Yield: 81%
  • 1H NMR (400 MHz, DMSO d6) δ 9.88 (brs, 1H), 9.62 (brs, 1H), 8.59 (m, 1H), 7.92 (dd, 1H), 7.58 (m, 1H), 6.83 (d, 1H), 6.70 (dd, 1H), 6.40 (d, 1H), 6.11 (s, 1H), 3.56-3.26 (brm, 8H), 2.40 (m, 1H), 2.11 (m, 1H), 1.18 (t, 3H), 1.11 (t, 3H)
  • Mass Spectral Analysis m/z=380.7 (M+H)+
  • Example 48A Preparation of 48.1
  • To a solution of 1.5f (20.0 g, 41.71 mmol, 1.0 eq) in dioxane (300 mL) was added potassium carbonate (17.3 g, 125 mmol, 3.0 eq), water (50 mL), 14.1 (6.7 g, 45.88 mmol, 1.1 eq) and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (1.70 g, 2.09 mmol, 0.05 eq). The mixture was stirred at room temperature for 1 h. Water (500 mL) was added and product was extracted with ethyl acetate. The organics were combined, concentrated and the crude product was purified by column chromatography (eluent: ethyl acetate/hexane=3:7). Pure Fractions were concentrated to an oil, which was dissolved in diethyl ether (30 mL). Hexane was added to the stirred solution and the precipitated solid was collected by vacuum filtration. Yield: 76%
  • 1H NMR (400 MHz, CDCl3) δ 7.59 (d, 2H), 7.31 (d, 2H), 7.19 (t, 1H), 6.67 (dd, 1H), 6.47 (d, 1H), 5.54 (s, 1H), 3.80 (brs, 2H), 3.42 (s, 3H), 3.32 (brs, 2H), 2.02 (brd, 2H), 1.68 (m, 2H), 1.47 (s, 9H)
  • Mass Spectral Analysis m/z=433.5 (M+H)+
  • Preparation of 48.2
  • To a suspension of 48.1 (5.0 g, 11.56 mmol, 1.0 eq) in isopropanol (100 mL) was added 14.3 (1.50 g, 23.12 mmol, 2.0 eq), zinc bromide (1.30 g, 5.78 mmol, 0.50 eq) and water (50 mL). The reaction mixture was heated to reflux (105° C.). Additional amount of water (10 mL) and isopropanol (30 mL) were added to the reaction mixture, which was heated for 5 days at 105° C. The reaction mixture was then cooled to room temperature and then cooled in an ice/brine bath. A 3N aqueous solution of hydrochloric acid (10 mL) was slowly added to the mixture until the pH ˜1. The homogeneous mixture was stirred for 10 min at room temperature. Water (200 mL) and ethyl acetate were added to the mixture followed by diethyl ether (100 mL). The organics were concentrated to a solid. This solid was triturated in methanol (20 mL) and collected by vacuum filtration. The filtrate was concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity, then methanol/ethyl acetate mixtures of increasing polarity). The starting material (48.1, 1.28 g white solid) was isolated along with 983 mg of the desired product (48.2). Yield: 58%
  • 1H NMR (400 MHz, CDCl3) δ 8.01 (brd, 2H), 7.30 (m, 2H), 7.15 (t, 1H), 6.64 (d, 1H), 6.42 (d, 1H), 5.50 (s, 1H), 3.81 (brd, 2H), 3.33 (brs, 5H), 1.99 (m, 2H), 1.63 (m, 2H), 1.50 (s, 9H)
  • Mass Spectral Analysis m/z=474.9 (M−H)
  • Preparation of 48A
  • To a solution of 48.2 (0.434 g, 0.913 mmol, 1.0 eq) in methylene chloride (15 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (2.74 mL, 5.48 mmol, 6.0 eq). The reaction was warmed to room temperature, stirred for 3 days at room temperature and diluted with diethyl ether (10 mL). The solids were collected by vacuum filtration. Yield: 87%
  • 1H NMR (400 MHz, DMSO d6) δ 8.89 (brs, 2H), 8.02 (d, 2H), 7.42 (d, 2H), 7.28 (t, 1H), 6.75 (dd, 1H), 6.69 (d, 1H), 5.92 (s, 1H), 3.41 (s, 3H), 3.20 (brm, 4H), 2.02 (brm, 4H)
  • Mass Spectral Analysis m/z=376.3 (M+H)+
  • Elemental analysis:
  • C21H21N5O2, 1HCl, 1H2O
  • Theory: % C, 58.67; % H, 5.63; % N, 16.29.
  • Found: % C, 58.79; % H, 5.30; % N, 16.12.
  • Examples 48B, 48C Preparation of 48.3 and 48.4
  • To a solution of 48.2 (2.20 g, 4.63 mmol, 1.0 eq) in N,N-dimethylformamide (20 mL) under nitrogen was added potassium carbonate (3.20 g, 23.13 mmol, 5.0 eq) and 2.8c (0.87 mL, 13.88 mmol, 3.0 eq). The mixture was stirred for 4 days at room temperature. Water was added and product was extracted with ethyl acetate. The combined organics were washed with brine, concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). The major product was 48.3 (1.36 g) and the minor product was 48.4 (365 mg). Yield: 76% (combined yield)
  • 48.3: 1H NMR (400 MHz, DMSO d6) δ 7.99 (d, 2H), 7.34 (d, 2H), 7.23 (t, 1H), 6.66 (m, 2H), 5.83 (s, 1H), 4.43 (s, 3H), 3.64 (m, 2H), 3.39 (s, 3H), 3.27 (brs, 2H), 1.84 (m, 2H), 1.71 (m, 2H), 1.41 (s, 9H)
  • Mass Spectral Analysis m/z=490.4 (M+H)+
  • 48.4: 1H NMR (400 MHz, DMSO d6) δ 7.81 (d, 2H), 7.40 (d, 2H), 7.24 (t, 1H), 6.67 (m, 2H), 5.85 (s, 1H), 4.19 (s, 3H), 3.66 (m, 2H), 3.41 (s, 3H), 3.28 (brs, 2H), 1.84 (m, 2H), 1.72 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=490.6 (M+H)+
  • Preparation of 48B
  • To a solution of 48.3 (0.40 g, 0.817 mmol, 1.0 eq) in methylene chloride (10 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.63 mL, 3.27 mmol, 4.0 eq). The reaction was warmed to room temperature, stirred for 16 h at room temperature and diluted with diethyl ether (10 mL). The solids were collected by vacuum filtration. Yield: 84%
  • 1H NMR (400 MHz, DMSO d6) δ 8.88 (brs, 2H), 8.01 (d, 2H), 7.38 (d, 2H), 7.27 (t, 1H), 6.74 (d, 1H), 6.67 (d, 1H), 5.90 (s, 1H), 4.44 (s, 3H), 3.40 (s, 3H), 3.19 (m, 4H), 2.01 (m, 4H)
  • Mass Spectral Analysis m/z=390.5 (M+H)+
  • Elemental analysis:
  • C22H23N5O2, 1HCl
  • Theory: % C, 62.04; % H, 5.68; % N, 16.44.
  • Found: % C, 62.05; % H, 5.79; % N, 16.51.
  • Preparation of 48C
  • To a solution of 48.4 (0.36 g, 0.735 mmol, 1.0 eq) in methylene chloride (10 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (1.47 mL, 2.94 mmol, 4.0 eq). The reaction was stirred for 16 h at room temperature and diluted with diethyl ether (10 mL). The solids were collected by vacuum filtration. Yield: 80%
  • 1H NMR (400 MHz, DMSO d6) δ 8.96 (brs, 2H), 7.82 (d, 2H), 7.43 (d, 2H), 7.28 (t, 1H), 6.75 (d, 1H), 6.69 (d, 1H), 5.92 (s, 1H), 4.19 (s, 3H), 3.42 (s, 3H), 3.19 (m, 4H), 2.06 (m, 4H)
  • Mass Spectral Analysis m/z=390.4 (M+H)+
  • Example 48D Preparation of 48.5
  • To a mixture of 1.14 (2.91 g, 11.47 mmol, 1.1 eq), potassium carbonate (2.16 g, 15.64 mmol, 1.5 eq), dichlorobis(triphenylphosphine)palladium (II) (0.22 g, 0.313 mmol, 0.03 eq) and triphenylphosphine (0.17 g, 0.626 mmol, 0.06 eq) in dioxane (75 mL) was added 1.5f (5.0 g, 10.43 mmol, 1.0 eq). The reaction was heated at 50° C. for 16 h. Additional amount of dichlorobis(triphenylphosphine)palladium (II) (0.22 g) and triphenylphosphine (0.17 g) were added to the reaction mixture, which was heated for another 2 days at 50° C. The mixture was then cooled to room temperature. Water was added and the product was extracted with ethyl acetate. The organics were concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 64%
  • 1H NMR (400 MHz, CDCl3) δ 7.06 (t, 1H), 6.51 (dd, 1H), 6.45 (d, 1H), 5.82 (s, 1H), 3.81 (brs, 5H), 3.25 (brs, 2H), 1.93 (brd, 2H), 1.57 (m, 2H), 1.46 (s, 9H), 1.34 (s, 12H)
  • Mass Spectral Analysis m/z=458.4 (M+H)+
  • Preparation of 48.6
  • To a solution of 48.5 (1.96 g, 4.29 mmol, 1.0 eq) in dioxane (40 mL) was added potassium carbonate (1.78 g, 12.86 mmol, 3.0 eq), water (6 mL), 34.1c (1.24 g, 4.71 mmol, 1.1 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (0.17 g, 0.214 mmol, 0.05 eq). The reaction was stirred at room temperature for 16 h. Water was added and the product was extracted with ethyl acetate. The organics were concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 48%
  • 1H NMR (400 MHz, CDCl3) δ 7.23 (d, 1H), 7.18 (t, 1H), 6.82 (d, 1H), 6.64 (dd, 1H), 6.50 (d, 1H), 5.70 (s, 1H), 3.78 (brs, 2H), 3.56 (m, 7H), 3.32 (brs, 2H), 2.00 (m, 2H), 1.67 (m, 2H), 1.47 (s, 9H), 1.26 (t, 6H)
  • Mass Spectral Analysis m/z=513.4 (M+H)+
  • Preparation of 48D
  • To a solution of 48.6 (1.06 g, 2.07 mmol, 1.0 eq) in methylene chloride (15 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (4.14 mL, 8.27 mmol, 4.0 eq). The reaction mixture was warmed to room temperature and stirred for 16 h, diluted with diethyl ether (10 mL) and stirred for an additional 30 min at room temperature. The solids were collected by vacuum filtration. Yield: 86%
  • 1H NMR (400 MHz, DMSO d6) δ 9.02 (brs, 2H), 7.34 (m, 2H), 7.00 (d, 1H), 6.77 (m, 2H), 6.05 (s, 1H), 3.59 (s, 3H), 3.54 (brm, 4H), 3.23 (brm, 4H), 2.12-1.99 (brm 4H), 1.23 (brt, 6H)
  • Mass Spectral Analysis m/z=413.7 (M+H)+
  • Elemental analysis:
  • C23H28N2O3S, 1HCl, 1H2O
  • Theory: % C, 59.36; % H, 6.50; % N, 5.86.
  • Found: % C, 59.15; % H, 6.69; % N, 6.00.
  • Example 48E Preparation of 48.7
  • To a solution of 48.5 (2.0 g, 4.37 mmol, 1.0 eq) in dioxane (40 mL) was added potassium carbonate (1.81 g, 13.12 mmol, 3.0 eq), water (6 mL), 34.1d (1.40 g, 4.81 mmol, 1.1 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) (0.18 g, 0.22 mmol, 0.05 eq). The reaction mixture was stirred at room temperature for 24 h. Water was added and the product was extracted with ethyl acetate. The combined organics were concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 49%
  • 1H NMR (400 MHz, CDCl3) δ 7.17 (t, 1H), 7.07 (d, 1H), 6.79 (d, 1H), 6.63 (d, 1H), 6.50 (d, 1H), 5.70 (s, 1H), 4.00 (brs, 2H), 3.77 (brs, 2H), 3.57 (s, 3H), 3.32 (brs, 2H), 1.99 (m, 2H), 1.67 (m, 2H), 1.46 (s, 9H), 1.37 (brd, 12H)
  • Mass Spectral Analysis m/z=541.8 (M+H)+
  • Preparation of 48E
  • To a solution of 48.7 (1.16 g, 2.15 mmol, 1.0 eq) in methylene chloride (30 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (4.29 mL, 8.58 mmol, 4.0 eq). The reaction mixture was warmed to room temperature, stirred for 16 h and concentrated to a foam. Diethyl ether (15 mL) was added to the mixture, which was stirred for 1 h at room temperature. The solids were collected by vacuum filtration. Yield: 77%
  • 1H NMR (400 MHz, DMSO d6) δ 8.83 (brs, 2H), 7.14 (t, 1H), 6.96 (d, 1H), 6.78 (d, 1H), 6.57 (m, 2H), 5.83 (s, 1H), 3.82 (brs, 2H), 3.39 (s, 3H), 3.02 (brm, 4H), 1.94-1.80 (brm, 4H), 1.16 (brd, 6H)
  • Mass Spectral Analysis m/z=441.4 (M+H)+
  • Elemental analysis:
  • C25H32N2O3S, 1HCl, 1H2O
  • Theory: % C, 60.65; % H, 7.13; % N, 5.66.
  • Found: % C, 61.02; % H, 6.90; % N, 5.57.
  • Example 48F Preparation of 48.8
  • To a solution of 48.5 (1.50 g, 3.28 mmol, 1.0 eq) in dioxane (30 mL) was added potassium phosphate (1.04 g, 4.92 mmol, 1.5 eq), potassium bromide (0.43 g, 3.61 mmol, 1.1 eq), 34.1a (0.93 g, 3.61 mmol, 1.1 eq) and tetrakis(triphenylphosphine) palladium(0) (0.38 g, 0.328 mmol, 0.10 eq). The mixture was heated at 100° C. for 6 days and then cooled to room temperature. Ethyl acetate and water were added and the layers were separated. The organics were concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). The resulting solid was triturated in diethyl ether and filtered. The filtrate, which contained the product, was concentrated. Yield: 40%
  • 1H NMR (400 MHz, CDCl3) δ 8.60 (dd, 1H), 7.67 (m, 2H), 7.19 (m, 1H), 6.67 (dd, 1H), 6.48 (d, 1H), 5.87 (s, 1H), 3.82 (brs, 2H), 3.57 (brs, 2H), 3.44 (s, 3H), 3.32 (brs, 4H), 2.04 (brd, 2H), 1.72 (m, 2H), 1.47 (s, 9H), 1.22 (brd, 6H)
  • Mass Spectral Analysis m/z=508.5 (M+H)+
  • Preparation of 48F
  • To a solution of 48.8 (0.668 g, 1.32 mmol, 1.0 eq) in methylene chloride (15 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (3.95 mL, 7.89 mmol, 6.0 eq). The reaction mixture was warmed to room temperature, stirred for 16 h at room temperature and concentrated to a foam. Diethyl ether (15 mL) was added to the mixture and the solids were collected by vacuum filtration. Yield: 75%
  • 1H NMR (400 MHz, DMSO d6) δ 9.11 (brd, 2H), 8.54 (s, 1H), 7.90 (d, 1H), 7.43 (d, 1H), 7.21 (t, 1H), 6.68 (dd, 1H), 6.61 (d, 1H), 6.07 (s, 1H), 3.41 (brm, 2H), 3.32 (s, 3H), 3.13 (brm, 6H), 1.98 (brm, 4H), 1.07 (brd, 6H)
  • Mass Spectral Analysis m/z=408.4 (M+H)+
  • Elemental analysis:
  • C24H29N3O3, 2HCl, 1H2O
  • Theory: % C, 57.83; % H, 6.67; % N, 8.43; % Cl, 14.23.
  • Found: % C, 57.98; % H, 6.41; % N, 8.29; % Cl, 14.21.
  • Example 49A Preparation of 49.2
  • Pyrrolidine (2.09 mL, 25.09 mmol, 1.0 eq) was added at 0° C. to 1.2 (5.00 g, 25.09 mmol, 1.0 eq) and 49.1 (3.87 g, 25.09 mmol, 1.0 eq) in methanol (50 mL). The solution was stirred overnight at room temperature and then concentrated under reduced pressure. Ethyl acetate (100 mL) was added to the mixture. The organic mixture was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 40%
  • 1H NMR (400 MHz, CDCl3) δ 7.56 (m, 1H), 6.92 (d, 1H), 6.83 (m, 1H), 3.69 (m, 2H), 3.12 (m, 2H), 2.83 (s, 2H), 1.86 (d, 2H), 1.63 (m, 2H), 1.40 (s, 9H)
  • Mass Spectral Analysis m/z=336.14 (M+H)+
  • Preparation of 49.3
  • To a solution of 49.2 (3.00 g, 8.95 mmol, 1 eq) in tetrahydrofuran (10 mL) at −78° C. under nitrogen was added drop wise a 1M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (10.73 mL, 10.73 mmol, 1.2 eq). The mixture was stirred for 1 hour at −78° C. A solution of 1.4 (3.83 g, 10.73 mmol, 1.2 eq) in tetrahydrofuran (2 mL) was added drop wise to the mixture. The mixture was warmed slowly to room temperature and stirring was continued for 1 hour at room temperature. The reaction was then concentrated under reduced pressure. The crude product was used without further purification. Yield: 85%
  • 1H NMR (400 MHz, DMSO d6) δ 7.38 (m, 1H), 6.92 (m, 1H), 6.74 (m, 1H), 6.23 (s, 1H), 3.71 (m, 2H), 3.17 (br s, 2H), 1.89 (m, 2H), 1.76 (m, 2H), 1.41 (s, 9H)
  • Mass Spectral Analysis m/z=468.48 (M+H)+
  • Preparation of 49.4
  • To a solution of 49.3 (1.00 g, 2.14 mmol, 1.0 eq) in dimethoxyethane (5 mL) was added sequentially a 2N aqueous solution of sodium carbonate (3.2 mL, 6.42 mmol, 3 eq), lithium chloride (0.27 g, 6.42 mmol, 3 eq), 1.6 (0.52 g, 2.35 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.05 g, 0.04 mmol, 0.02 eq). The mixture was refluxed for 16 hours under nitrogen, then cooled to room temperature. Water (250 mL) was added to the mixture, which was extracted with ethyl acetate. The organic layer was further washed with brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 50%
  • Mass Spectral Analysis m/z=495.36 (M+H)+
  • Preparation of 49A
  • A 2M anhydrous solution of hydrochloric acid in diethyl ether (2.95 mL, 5.89 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 49.4 (0.53 g, 1.07 mmol, 1 eq) in anhydrous dichloromethane (6 mL). The mixture was warmed to room temperature and stiffing was continued for an additional 16 hours at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 97%
  • 1H NMR (400 MHz, DMSO d6) δ 9.15 (m, 2H), 7.35 (m, 5H), 6.99 (d, 1H), 6.82 (m, 1H), 6.03 (s, 1H), 3.44 (br s, 2H), 3.19 (m, 6H), 2.07 (m, 4H), 1.10 (m, 6H)
  • Mass Spectral Analysis m/z=395.3 (M+H)+
  • Elemental analysis:
  • C24H27FN2O2, 1HCl, 1H2O
  • Theory: % C, 64.21; % H, 6.74; % N, 6.24.
  • Found: % C, 64.27; % H, 6.30; % N, 6.28.
  • Example 49B Preparation of 49.5
  • 3.4k (270.8 mL, 3.24 mmol, 5.0 eq) was added at room temperature to a solution of 49.2 (100 mg, 0.65 mmol, 1.0 eq) in N,N-dimethylformamide (5 mL). The solution was stirred overnight at 80° C. and then concentrated under reduced pressure. The crude product was used for the next step without further purification. Yield: 90%
  • 1H NMR (400 MHz, CDCl3) δ 7.23 (t, 1H), 6.38 (d, 1H), 6.25 (d, 1H), 3.69 (m, 2H), 3.10 (m, 6H), 2.70 (s, 2H), 1.94 (m, 2H), 1.87 (m, 4H), 1.62 (m, 2H), 1.39 (s, 9H)
  • Mass Spectral Analysis m/z=387.25 (M+H)+
  • Preparation of 49.6
  • To a solution of 49.5 (7.50 g, 19.40 mmol, 1 eq) in tetrahydrofuran (20 mL) at −78° C. under nitrogen was added drop wise a 1M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (23.29 mL, 23.29 mmol, 1.2 eq). The mixture was stirred for 1 hour at −78° C. A solution of 1.4 (8.32 g, 23.29 mmol, 1.2 eq) in tetrahydrofuran (5 mL) was added drop wise to the mixture, which was warmed slowly to room temperature. Stirring was continued for 16 hours at room temperature. The reaction was then concentrated under reduced pressure and dissolved in ethyl acetate. The organic solution was washed with a 1N aqueous solution of hydrochloric acid, a 1N aqueous solution of sodium hydroxide, brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 50%
  • 1H NMR (400 MHz, DMSO d6) δ 7.15 (t, 1H), 6.56 (d, 1H), 6.50 (d, 1H), 5.91 (s, 1H), 3.61 (m, 2H), 3.21 (br s, 2H), 3.10 (m, 4H), 1.86 (m, 6H), 1.76 (m, 2H), 1.40 (s, 9H)
  • Mass Spectral Analysis m/z=519.30 (M+H)+
  • Preparation of 49.7
  • To a solution of 49.6 (1.00 g, 1.93 mmol, 1.0 eq) in dimethoxyethane (5 mL) was added sequentially a 2N aqueous solution of sodium carbonate (2.9 mL, 5.79 mmol, 3 eq), lithium chloride (0.25 g, 5.79 mmol, 3 eq), 1.6 (0.47 g, 2.12 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (0.05 g, 0.04 mmol, 0.02 eq). The mixture was refluxed for 16 hours under nitrogen, then cooled to room temperature. Water (250 mL) was added to the mixture, which was extracted with ethyl acetate. The organic layer was further washed with brine and dried over sodium sulfate. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 10%
  • Mass Spectral Analysis m/z=546.47 (M+H)+
  • Preparation of 49B
  • A 2M anhydrous solution of hydrochloric acid in diethyl ether (0.4 mL, 0.81 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 49.7 (0.08 g, 0.15 mmol, 1 eq) in anhydrous dichloromethane (4 mL). The mixture was warmed to room temperature and stiffing was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 97%
  • 1H NMR (400 MHz, DMSO d6) δ 9.01 (m, 2H), 7.26 (q, 4H), 7.15 (t, 1H), 6.55 (m, 2H), 5.89 (s, 1H), 3.54 (br s, 5H), 3.42 (br s, 2H), 3.18 (m, 6H), 2.76 (br s, 2H), 2.11 (m, 4H), 1.28 (br s, 2H), 1.08 (m, 6H)
  • Mass Spectral Analysis m/z=446.8 (M+H)+
  • Examples 49C, 49D Preparation of 49.8
  • To a solution of 1.5d (14.02 g, 30 mmol, 1 eq.) in N,N-dimethylformamide (150 mL) at 0° C. was added successively potassium acetate (8.83 g, 90 mmol, 3 eq.), bis(pinacolato)diboron 1.14 (9.14 g, 36 mmol, 1.2 eq.) and 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (658 mg, 0.9 mmol, 0.03 eq.). The reaction mixture was stirred at 100-110° C. for 10 h. The mixture was cooled to room temperature. Diethyl ether (300 mL) and water (300 mL) were added to the mixture, which was stirred for an additional 30 minutes at room temperature. The two phases were separated and the organic phase was washed with water (2×150 mL), brine (200 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 82%
  • 1H NMR (400 MHz, CDCl3) δ 7.49 (dd, 1H), 6.78 (m, 2H), 6.34 (s, 1H), 3.95-3.72 (m, 2H), 3.37-3.15 (m, 2H), 1.97-1.87 (m, 2H), 1.66-1.53 (m, 2H), 1.46 (s, 9H), 1.34 (s, 12H)
  • Mass Spectral Analysis m/z=446.31 (M+H)+
  • Preparation of 49.9
  • To a solution of a 2M aqueous solution of potassium carbonate (3.08 mL, 6.15 mmol, 3 eq.) was added dioxane (20 mL), 35.8 (0.745 g, 2.05 mmol, 1 eq.), and 49.8 (1.37 g, 3.075 mmol, 1.5 eq.) successively. The reaction flask was purged with nitrogen and 1,1′-bis(diphenylphosphino)ferrocene palladium(II) chloride complex with dichloromethane (75 mg, 0.1 mmol, 0.05 eq) was added to the mixture. The mixture was stirred at room temperature for 1 h and then heated at 55° C. for 10 h. The mixture was then cooled to room temperature. Water (50 mL) and ethyl acetate (150 mL) were added and the two phases were separated. The organic phase was washed with brine (100 mL) and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 94%
  • 1H NMR (400 MHz, CDCl3) δ 7.23-7.17 (m, 2H), 7.06 (dd, 1H), 6.88-6.78 (m, 2H), 6.47 (dd, 1H), 5.59 (s, 1H), 5.07 (s, 2H), 3.97-3.77 (m, 2H), 3.63-3.47 (m, 2H), 3.42-3.22 (m, 7H), 2.11-2.01 (m, 2H), 1.73-1.61 (m, 2H), 1.48 (s, 9H)
  • Mass Spectral Analysis m/z=555.47 (M+H)+
  • Preparation of 49C
  • To a solution of 49.9 (1.2 g, 2.16 mmol, 1 eq.) in methanol (50 mL) was slowly added a 4M anhydrous solution of hydrogen chloride in dioxane (3.2 mL, 12.8 mmol, 6 eq.). The reaction mixture was stirred at room temperature for 10 h. Additional amount of 4M anhydrous solution of hydrogen chloride in dioxane (2.16 mL, 8.64 mmol, 4 eq.) was slowly added to the mixture, which was stirred at room temperature for an additional 10 h. The mixture was concentrated under reduced pressure. The resulting foamy solids were soaked in diethyl ether to give the fine powders, which were collected by filtration, washed with ethyl acetate and diethyl ether. Yield: 90%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.87 (s, 1H), 9.00-8.75 (m, 2H), 7.18 (d, 1H), 7.07-6.97 (m, 2H), 6.91-6.82 (m, 2H), 6.44 (dd, 1H), 5.89 (s, 1H), 3.53-3.10 (m, 8H), 2.15-2.05 (m, 2H), 2.03-1.92 (m, 2H), 1.20-1.03 (m, 6H)
  • Mass Spectral Analysis m/z=411.7 (M+H)+
  • Elemental analysis:
  • C24H27FN2O3, 1HCl, 0.5H2O
  • Theory: % C, 63.22; % H, 6.41; % N, 6.14.
  • Found: % C, 63.32; % H, 6.34; % N, 6.13.
  • Preparation of 49D
  • To a stirred solution of 49C (0.20 g, 0.45 mmol, 1 eq.) in methanol (20 mL) was added palladium [40 mg, 10 wt. % (dry basis) on activated carbon, 20% wt. eq]. The reaction mixture was stirred under hydrogen atmosphere using a hydrogen balloon at room temperature for 10 h. The mixture was filtered through celite and the filtrate was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixture of increasing polarity). Yield: 80%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.87 (s, 1H), 8.85-8.55 (m, 2H), 7.08-6.89 (m, 3H), 6.84 (s, 1H), 6.75 (dd, 1H), 6.41 (dd, 1H), 4.47 (m, 1H), 3.50-2.91 (m, 8H), 2.15-1.74 (m, 6H), 1.20-1.11 (m, 6H)
  • Mass Spectral Analysis m/z=413.7 (M+H)+
  • Example 50A Preparation of 50.1
  • To a solution of 2.5 (15.00 g, 25.88 mmol, 1.0 eq) in dimethoxyethane (78 mL) was added sequentially a 2N aqueous solution of sodium carbonate (38.8 mL, 77.63 mmol, 3 eq), lithium chloride (3.29 g, 77.63 mmol, 3 eq), 31.1 g (3.47 g, 28.46 mmol, 1.1 eq) and palladium, 10 weight % (dry basis) on activated carbon, wet, (Degussa type E101) (0.28 g, 0.13 mmol, 0.005 eq). The mixture was refluxed for 6 days under nitrogen. The mixture was then cooled to room temperature and diluted with dichloromethane (350 mL). The mixture was then filtered through a celite plug. The filtrate was dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography (eluent: ethyl acetate/hexanes mixtures of increasing polarity). Yield: 49%
  • 1H NMR (400 MHz, DMSO d6) δ 8.98 (br s, 1H), 7.42 (m, 3H), 7.33 (m, 2H), 6.79 (d, 1H), 6.58 (m, 1H), 6.38 (m, 1H), 5.76 (s, 1H), 3.68 (m, 2H), 3.23 (m, 2H), 1.82 (m, 2H), 1.65 (m, 2H), 1.41 (s, 9H)
  • Mass Spectral Analysis m/z=394.46 (M+H)+
  • Preparation of 50.2
  • To a solution of 50.1 (4.30 g, 10.9 mmol, 1.0 eq) in dichloromethane (20 mL) was added sequentially triethylamine (1.83 mL, 13.1 mmol, 1.2 eq) and 1.4 (4.29 g, 12.0 mmol, 1.1 eq). The mixture was stirred at room temperature overnight under nitrogen. The mixture was then concentrated under reduced pressure. Ethyl acetate (800 mL) was added. The organic mixture was washed with a 1N aqueous solution of sodium hydroxide, water, brine and dried over sodium sulfate. The crude product was used for the next step without further purification. Yield: 87%
  • Mass Spectral Analysis m/z=525.93 (M+H)+
  • Preparation of 50.3
  • A solution of 50.2 (5.00 g, 9.51 mmol, 1.0 eq), triethylamine (2.9 mL, 20.93 mmol, 2.2 eq), palladium(II) acetate (0.21 g, 0.95 mmol, 0.1 eq) and 1,1′-bis(diphenylphosphino)ferrocene (1.05 g, 1.90 mmol, 0.2 eq) was stirred at 65° C. under a carbon monoxide atmosphere for 4 days. The mixture was then cooled to room temperature and diluted with ethyl acetate (350 mL). The mixture was then filtered through a celite plug. The filtrate was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: ethyl acetate/hexanes mixtures of increasing polarity). Yield: 49%
  • 1H NMR (400 MHz, DMSO d6) δ 7.82 (m, 1H), 7.57 (m, 1H), 7.47 (m, 3H), 7.35 (m, 2H), 7.07 (d, 1H), 5.90 (s, 1H), 3.74 (s, 3H), 3.34 (m, 4H), 1.91 (m, 2H), 1.75 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=436.07 (M+H)+
  • Preparation of 50A
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.6 mL, 6.31 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 50.3 (0.50 g, 1.15 mmol, 1 eq) in anhydrous methanol (5 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 10%
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (m, 2H), 7.87 (m, 1H), 7.58 (d, 1H), 7.48 (m, 3H), 7.38 (m, 2H), 7.17 (d, 1H), 5.97 (s, 1H), 3.75 (s, 3H), 3.24 (m, 4H), 2.08 (m, 4H)
  • Mass Spectral Analysis m/z=336.4 (M+H)+
  • Example 50B Preparation of 50.4
  • To a solution of 50.3 (0.90 g, 2.07 mmol, 1.0 eq) in tetrahydrofuran (3 mL) and methanol (3 mL) was added a solution of lithium hydroxide monohydrate (0.48 g, 11.37 mmol, 5.5 eq) dissolved in water (3 mL). The reaction was stirred at room temperature overnight. The mixture was concentrated under reduced pressure. Water (100 mL) was added and any undissolved material was removed by filtration. A 6N aqueous solution of hydrochloric acid was added dropwise to the filtrate until the solution was acidic (pH=2). The precipitate was collected by vacuum filtration. The crude product was further purified by column chromatography (eluent: ethyl acetate/hexanes mixtures of increasing polarity). Yield: 40%
  • Mass Spectral Analysis m/z=420.31 (M−H)
  • Preparation of 50B
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.11 mL, 4.44 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 50.4 (0.34 g, 0.81 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 10%
  • 1H NMR (400 MHz, DMSO d6) δ 12.76 (br s, 1H), 9.17 (br s, 2H), 7.83 (m, 1H), 7.58 (d, 1H), 7.48 (m, 3H), 7.37 (d, 2H), 7.13 (d, 1H), 5.95 (s, 1H), 3.23 (m, 4H), 2.08 (m, 4H)
  • Mass Spectral Analysis m/z=322.1 (M+H)+
  • Example 50C Preparation of 50.5
  • 0-Benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (0.10 g, 0.31 mmol, 1.1 eq) was added to a cooled (0° C.) solution of 50.4 (0.12 g, 0.28 mmol, 1.0 eq), 1.12 (0.034 g, 0.31 mmol, 1.1 eq), and N,N-diisopropylethylamine (0.11 mL, 0.63 mmol, 2.2 eq) dissolved in acetonitrile (5 mL). The solution was stirred for 2 hours at room temperature and then concentrated under reduced pressure. Ethyl acetate (10 mL) and a saturated aqueous solution of sodium hydrogenocarbonate (10 mL) were added to the crude product and the resulting mixture was stirred for 20 minutes at room temperature. The phases were separated and the organic phase was washed with a saturated aqueous solution of sodium hydrogenocarbonate, brine dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 55%
  • Mass Spectral Analysis m/z=477.43 (M+H)+
  • Preparation of 50C
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (0.17 mL, 0.69 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 50.5 (60 mg, 0.13 mmol, 1 eq) in anhydrous methanol (4 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 54%
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (br s, 2H), 7.45 (m, 3H), 7.36 (m, 2H), 7.27 (m, 1H), 7.09 (d, 1H), 6.89 (d, 1H), 5.93 (s, 1H), 3.23 (br s, 8H), 2.07 (m, 4H), 1.02 (br s, 6H)
  • Mass Spectral Analysis m/z=377.7 (M+H)+
  • Example 50D Preparation of 50D
  • A 4M anhydrous solution of hydrochloric acid in 1,4-dioxane (1.7 mL, 6.99 mmol, 5.5 eq) was added drop wise to a cooled (0° C.) solution of 50.1 (0.50 g, 1.27 mmol, 1 eq) in anhydrous methanol (3 mL). The mixture was warmed to room temperature and stirring was continued for an additional 16 hours at room temperature. The reaction mixture was concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity). Yield: 55%
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (s, 3H), 7.46 (m, 3H), 7.35 (m, 2H), 6.84 (d, 1H), 6.64 (d, 1H), 6.42 (s, 1H), 5.83 (s, 1H), 3.16 (m, 4H), 2.07 (m, 2H), 1.96 (m, 2H)
  • Mass Spectral Analysis m/z=294.1 (M+H)+
  • Elemental analysis:
  • C19H19NO2, 1HCl, 1H2O
  • Theory: % C, 65.61; % H, 6.38; % N, 4.03.
  • Found: % C, 65.89; % H, 6.29; % N, 3.95.
  • Examples 51A, 51B, 51C Preparation of 51.2
  • To a solution of 51.1 (2.0 g, 9.38 mmol, 1.0 eq) in methanol (50 mL) was added 1.1a (1.13 mL, 9.38 mmol, 1.0 eq) and pyrrolidine (2.11 mL, 25.51 mmol, 2.72 eq). The reaction mixture was refluxed for 16 h and then concentrated. The crude product was dissolved in ethyl acetate. The organic solution was washed with water, with a 1N aqueous solution of sodium hydroxide and then brine. The organics were concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 29%
  • 1H NMR (400 MHz, CDCl3) δ 7.88 (dd, 1H), 7.50 (m, 1H), 7.02 (t, 1H), 6.96 (d, 1H), 4.38 (brs, 1H), 3.96 (brd, 1H), 3.30 (brt, 1H), 2.66 (q, 2H), 2.05 (m, 2H), 1.65 (m, 2H), 1.46 (s, 9H), 1.28 (d, 3H)
  • Mass Spectral Analysis m/z=332.3 (M+H)+
  • Preparation of 51.3
  • To a solution of 51.2 (0.89 g, 2.69 mmol, 1.0 eq) in tetrahydrofuran (30 mL) at −78° C. under nitrogen was added drop wise a 1.0M solution of LiHMDS in tetrahydrofuran (3.22 mL, 3.22 mmol, 1.2 eq). The mixture was stirred for 1 h at −78° C. A solution of 1.4 (1.15 g, 3.22 mmol, 1.2 eq) in tetrahydrofuran (10 mL) was added drop wise to the mixture. The mixture was warmed slowly to room temperature and stirring was continued for 5 h at room temperature. Ice was added to the reaction mixture, which was stirred for 5 min Ethyl acetate and a 1N aqueous solution of sodium hydroxide were added to the mixture and the layers were separated. The organics were washed with a 1N aqueous solution of sodium hydroxide and then brine. The mixture was concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 78%
  • 1H NMR (400 MHz, CDCl3) δ 7.28 (m, 2H), 6.99 (t, 1H), 6.89 (d, 1H), 5.46 (s, 1H), 4.38 (brs, 1H), 3.97 (brd, 1H), 3.33 (brt, 1H), 2.18 (m, 1H), 2.06 (m, 1H), 1.72 (m, 1H), 1.62 (m, 1H), 1.47 (s, 9H), 1.34 (d, 3H)
  • Mass Spectral Analysis m/z=464.1 (M+H)+
  • Preparation of 51.4
  • To a solution of 51.3 (0.95 g, 2.05 mmol, 1.0 eq) in dioxane (25 mL) was added 1.6 (0.50 g, 2.25 mmol, 1.1 eq), potassium phosphate (0.65 g, 3.07 mmol, 1.5 eq), potassium bromide (0.27 g, 2.25 mmol, 1.1 eq) and tetrakis(triphenylphosphine) palladium(0) (0.12 g, 0.102 mmol, 0.05 eq). The mixture was heated at 100° C. for 16 h and then cooled to room temperature. Ethyl acetate and water were added to the mixture and the layers were separated. The organics were concentrated and the crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). The resulting solid was triturated in diethyl ether (15 mL) and collected by vacuum filtration. Yield: 75%
  • 1H NMR (400 MHz, CDCl3) δ 7.40 (d, 2H), 7.36 (d, 2H), 7.19 (m, 1H), 7.00 (dd, 1H), 6.93 (d, 1H), 6.85 (t, 1H), 5.46 (s, 1H), 4.39 (brs, 1H), 3.96 (brd, 1H), 3.57 (brs, 2H), 3.33 (brm, 3H), 2.14 (m, 1H), 2.04 (m, 1H), 1.72 (m, 1H), 1.63 (m, 1H), 1.48 (s, 9H), 1.39 (d, 3H), 1.21 (brd, 6H)
  • Mass Spectral Analysis m/z=491.2 (M+H)+
  • Preparation of 51.5 and 51.6
  • Chiral separation of 51.4 (720 mg) gave the two enantiomers 51.5 and 51.6.
  • Column: Chiralpak ADH, 21×250 nm, 35° C.; SFC
  • Eluent: 20% EtOH/80% CO2; 50 mL/min, 200 bar
  • UV wavelength: 260 nm
  • Polarimeter: 670 nm
  • Sample: 10 mg/mL in MeOH, 2.1 mL injected
  • Negative polarimeter peak elutes first at about 5 minutes and the positive polarimeter peak elutessecond at about 6.5 minutes in 20% EtOH/CO2
  • 51.5: (+) enantiomer; ee>99% (226 mg) 51.6: (−) enantiomer; ee>98% (238 mg)
  • Preparation of 51A
  • To a solution of 51.5 (0.226 g, 0.461 mmol, 1.0 eq) in methylene chloride (7 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (0.92 mL, 1.84 mmol, 4.0 eq). The reaction was warmed to room temperature and stirred for an additional 16 h at room temperature. The reaction was concentrated to a solid, which was triturated in diethyl ether (5 mL), and the solids were collected by vacuum filtration. Yield: 79%
  • 1H NMR (400 MHz, DMSO d6) δ 9.04 (brs, 1H), 8.62 (brs, 1H), 7.40 (d, 2H), 7.35 (d, 2H), 7.18 (m, 1H), 6.89 (m, 3H), 6.23 (s, 1H), 3.48 (brs, 2H), 3.37 (brs, 2H), 3.24 (brs, 1H), 3.14 (brs, 2H), 2.12 (brm, 2H), 1.86 (brm, 1H), 1.67 (brm, 1H), 1.19 (d, 3H), 1.04 (brd, 6H)
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.5H2O
  • Theory: % C, 68.87; % H, 7.40; % N, 6.43.
  • Found: % C, 69.01; % H, 7.36; % N, 6.39.
  • [α]D=−4.62 (c=8.25 mg/mL, MeOH)
  • Preparation of 51B
  • To a solution of 51.6 (0.238 g, 0.485 mmol, 1.0 eq) in methylene chloride (7 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (0.97 mL, 1.94 mmol, 4.0 eq). The reaction was warmed to room temperature and stirred for an additional 16 h at room temperature. The reaction was concentrated to a solid, which was triturated in diethyl ether (5 mL), and the solids were collected by vacuum filtration. Yield: 77%
  • 1H NMR (400 MHz, DMSO d6) δ 9.12 (brs, 1H), 8.72 (brs, 1H), 7.40 (d, 2H), 7.34 (d, 2H), 7.17 (m, 1H), 6.89 (m, 3H), 6.23 (s, 1H), 3.47 (brs, 2H), 3.36 (brs, 2H), 3.23 (brs, 1H), 3.14 (brs, 2H), 2.10 (brm, 2H), 1.87 (brm, 1H), 1.68 (t, 1H), 1.19 (d, 3H), 1.04 (brd, 6H)
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1HCl, 0.33H2O
  • Theory: % C, 69.35; % H, 7.37; % N, 6.47.
  • Found: % C, 69.44; % H, 7.37; % N, 6.46.
  • [α]D=+5.89 (c=9.60 mg/mL, MeOH)
  • Preparation of 51C
  • To a solution of 51.4 (0.75 g, 1.53 mmol, 1.0 eq) in methylene chloride (15 mL) at 0° C. under nitrogen was added a 2.0M solution of anhydrous hydrochloric acid in diethyl ether (3.06 mL, 6.11 mmol, 4.0 eq). The reaction was warmed to room temperature and stirred for an additional 16 h at room temperature. The reaction mixture was diluted with diethyl ether (20 mL) and the mixture was stirred for 15 min at room temperature. The solids were collected by vacuum filtration. Yield: 95%
  • 1H NMR (400 MHz, DMSO d6) δ 9.24 (brs, 1H), 8.87 (brs, 1H), 7.49 (d, 2H), 7.42 (d, 2H), 7.26 (m, 1H), 6.97 (m, 3H), 6.31 (s, 1H), 3.55 (brs, 1H), 3.45 (brs, 2H), 3.26 (brm, 4H), 2.19 (brm, 2H), 1.96 (brm, 1H), 1.77 (t, 1H), 1.28 (d, 3H), 1.12 (brd, 6H)
  • Mass Spectral Analysis m/z=391.4 (M+H)+
  • Example 52A Preparation of 52.1
  • 4.1 (1.78 mL, 12.60 mmol, 1.2 eq) was added dropwise to a cooled (0° C.) solution of Example 21C (4.10 g, 10.50 mmol, 1.0 eq) and triethylamine (4.39 mL, 31.50 mmol, 3.0 eq) in tetrahydrofuran (25 mL). The reaction was stirred overnight at room temperature. Dichloromethane (10 mL) was added to help solubilize the reaction. An additional portion of 4.1 (1.00 mL, 7.11 mmol, 0.68 eq) was added dropwise and stirring was continued for 3 hours. The reaction was diluted with an aqueous solution of 1N hydrochloric acid (200 mL) and extracted with dichloromethane (2×100 mL). The organic extracts were combined, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 85%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.39 (q, 4H), 7.23 (t, 1H), 6.93 (m, 3H), 5.87 (d, 1H), 3.51-3.78 (br m, 4H), 3.44 (br s, 2H), 3.23 (br s, 2H), 1.91-2.29 (br m, 4H), 1.80 (m, 2H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=487.45 (M+H)+
  • Preparation of 52.2
  • To a solution of 52.1 (4.22 g, 8.67 mmol, 1.0 eq) in 1,2-dichloroethane (20 mL) was added 4.3 (1.99 g, 13.01 mmol, 1.5 eq) portionwise. The reaction mixture was refluxed at 65° C. overnight. The reaction mixture was cooled to room temperature and then to 0° C. in an ice/brine bath. A 2M solution of oxalyl chloride in methylene chloride (6.94 mL, 13.88 mmol, 1.6 eq) was added dropwise to the reaction mixture. The reaction mixture was warmed to room temperature, then heated to 65° C. for 3 hours. The reaction was quenched by cooling to 0° C. and adding water (50 mL). The layers were separated and the aqueous was extracted with dichloromethane (2×100 mL). The organic extracts were combined, washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 58%
  • 1H NMR (400 MHz, DMSO d6) δ 7.44 (t, 3H), 7.36 (d, 2H), 7.25 (s, 1H), 6.87 (q, 1H), 5.88 (d, 1H), 3.51-3.78 (br m, 4H), 3.45 (br s, 2H), 3.25 (br s, 2H), 1.91-2.34 (br m, 4H), 1.79 (m, 2H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=585.44 (M+H)+
  • Preparation of 52.3
  • To a solution of 52.2 (2.50 g, 4.27 mmol, 1.0 eq) and tetrahydrofuran (10 mL) at 0° C. under an atmosphere of nitrogen was added 5.1 (0.88 mL, 28.20 mmol, 6.6 eq) dropwise. The reaction mixture was stirred at 0° C. for 30 minutes. The reaction mixture was concentrated under reduced pressure. Dichloromethane (50 mL) and water (50 mL) were added and the reaction mixture was stirred for 10 minutes. The phases were separated and the aqueous phase was extracted with dichloromethane (2×50 mL). The organic extracts were combined, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was used without further purification. Yield: 90%
  • 1H NMR (400 MHz, DMSO-d6) 8.23 (s, 1H), 7.65 (d, 1H), 7.44 (m, 5H), 7.11 (q, 1H), 6.01 (d, 1H), 4.06 (m, 2H), 3.52-3.80 (br m, 4H), 3.45 (br s, 2H), 3.23 (br s, 2H), 1.95-2.34 (br m, 4H), 1.85 (m, 2H), 1.13 (m, 6H)
  • Mass Spectral Analysis m/z=581.45 (M+H)+
  • Preparation of 52.4
  • To a solution of 52.3 (2.00 g, 3.44 mmol, 1.0 eq) in ethanol (10 mL) at room temperature under an atmosphere of nitrogen was added sodium acetate (1.89 g, 23.08 mmol, 6.7 eq) and 2.8c (1.18 mL, 18.94 mmol, 5.5 eq). The reaction mixture was refluxed at 90° C. overnight. The reaction mixture was concentrated under reduced pressure. The resulting oil was diluted with water (50 mL) then extracted with dichloromethane (3×50 mL). The organic extracts were combined, washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 82%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.80 (m, 1H), 7.45 (m, 5H), 7.15 (q, 1H), 6.04 (d, 1H), 3.53-3.81 (br m, 4H), 3.45 (br s, 2H), 3.23 (br s, 2H), 3.13 (s, 3H), 1.99-2.36 (br m, 4H), 1.84 (m, 2H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=565.51 (M+H)+
  • Preparation of 52A
  • To a solution of 52.4 (1.50 g, 2.66 mmol, 1.0 eq) in methanol (9 mL) and water (1 mL) at room temperature and under an atmosphere of nitrogen was added potassium carbonate (2.42 g, 17.53 mmol, 6.6 eq). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water and ethyl acetate. The phases were separated and the organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was used without further purification. Yield: 93%
  • 1H NMR (400 MHz, DMSO d6) δ 7.75 (m, 1H), 7.44 (m, 5H), 7.16 (d, 1H), 6.06 (d, 1H), 3.17-3.55 (br m, 8H), 2.72-2.96 (br m, 4H), 1.98-2.16 (br m, 4H), 1.83 (br s, 1H), 1.62 (br s, 1H), 1.12 (m, 6H)
  • Mass Spectral Analysis m/z=469.43 (M+H)+
  • Example 53A Preparation of 53A
  • To a solution of 48.2 (0.335 g, 0.704 mmol, 1.00 eq) in acetic acid (4 mL) was added a 48% aqueous solution of HBr (4 mL). The reaction mixture was refluxed at 110° C. for 8 hours and then cooled to room temperature. The reaction mixture was concentrated and a saturated aqueous solution of sodium bicarbonate was added until the mixture was basic. The product was extracted with 5% methanol/methylene chloride and purified by HPLC using the following conditions to give 40 mg of a light orange solid.
  • HPLC conditions:
  • Column: Waters Xterra Prep RP18 OBD Column, 19×150 mm
  • Detection: UV 210 nm
  • Flow: 15 mL/min
  • Mobile Phase A: 0.1% TFA in HPLC Water
  • Mobile Phase B: Acetonitrile
  • Gradient: Linear, 15% B to 90% B in 15 min
  • Yield: 12%
  • 1H NMR (400 MHz, DMSO) δ 9.58 (s, 1H), 8.62 (brs, 2H), 7.98 (d, 2H), 7.46 (d, 2H), 7.09 (t, 1H), 6.56 (dd, 1H), 6.46 (dd, 1H), 5.85 (s, 1H), 3.20 (brm, 4H), 2.08-1.92 (brm, 4H)
  • Mass Spectral Analysis m/z=360.8 (M−H)
  • Example 53B Preparation of 53B
  • To a solution of 48.3 (0.500 g, 1.02 mmol, 1.00 eq) in acetic acid (5 mL) was added a 48% aqueous solution HBr (5 mL). The reaction mixture was refluxed at 110° C. for 8 hours and then cooled to room temperature and stirred for an additional 48 h at room temperature. A precipitate formed and the reaction was concentrated. The solid was collected by filtration. The crude product was purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity, methanol contained 5% ammonium hydroxide). The resulting solid was triturated in a methylene chloride/diethyl ether mixture (5 mL/10 mL), filtered, and dried to give 168 mg of a white solid. Yield: 44%
  • 1H NMR (400 MHz, DMSO) δ 9.39 (brs, 1H), 7.97 (d, 2H), 7.38 (d, 2H), 7.02 (t, 1H), 6.46 (d, 1H), 6.39 (d, 1H), 5.74 (s, 1H), 4.43 (s, 3H), 2.86 (brm, 2H), 2.70 (brm, 2H), 1.70 (brm, 4H)
  • Mass Spectral Analysis m/z=376.8 (M+H)+
  • Example 53C Preparation of 53C
  • To a solution of 53D (0.300 g, 0.802 mmol, 1.00 eq) in anhydrous methanol (15 mL) was added concentrated hydrochloric acid (0.50 mL) and the reaction mixture was refluxed for 16 hours and then concentrated. The crude product was dissolved in methanol (2 mL) and then diethyl ether (10 mL) was added with stiffing. The resulting solid was filtered, rinsed with diethyl ether, and dried to give 272 mg of an off-white solid. Yield: 87%
  • 1H NMR (400 MHz, DMSO) δ 9.56 (s, 1H), 8.86 (brs, 2H), 7.89 (d, 2H), 7.37 (d, 2H), 7.08 (t, 1H), 6.55 (dd, 1H), 6.46 (dd, 1H), 5.82 (s, 1H), 3.86 (s, 3H), 3.18 (brm, 4H), 2.08-1.92 (brm, 4H)
  • Mass Spectral Analysis m/z=352.7 (M+H)+
  • C21H21NO4, 0.5H2O, 1HCl
  • Theory: % C, 63.56; % H, 5.84; % N, 3.53.
  • Found: % C, 63.39; % H, 5.72; % N, 3.53.
  • Example 53D Preparation of 53.2
  • To a solution of 11.5 (0.500 g, 0.981 mmol, 1.00 eq) in dioxane (8 mL) was added potassium carbonate (0.407 g, 2.94 mmol, 3.00 eq), water (2 mL), 53.1 (0.180 g, 1.08 mmol, 1.10 eq) and tetrakis(triphenylphosphine)palladium(0) (0.060 g, 0.05 mmol, 0.05 eq). The reaction mixture was stirred for 30 minutes at room temperature and no product was detected by LC/MS. Water (5 mL) was added and the reaction was homogeneous. The reaction mixture was heated at 50° C. for 20 hours. Additional quantities of tetrakis(triphenylphosphine)palladium(0) (0.060 g, 0.05 mmol, 0.05 eq) were added and the reaction mixture was heated at 80° C. for 3 hours. The reaction mixture was complete by LC/MS. The reaction mixture was cooled, acidified with a 1N aqueous solution of hydrochloric acid and product was extracted two times with ethyl acetate. The crude product was concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity, then methanol/ethyl acetate mixtures of increasing polarity). The product was dissolved in diethyl ether (3 mL) and hexane (10 mL) was added to precipitate the product, which was filtered and rinsed with hexane to give 400 mg of a pale orange solid. Yield: 85%
  • 1H NMR (400 MHz, DMSO) δ 7.89 (d, 2H), 7.32 (d, 2H), 7.19 (t, 1H), 6.70 (m, 2H), 5.83 (s, 1H), 4.71 (s, 2H), 3.65 (brm, 2H), 3.32 (s, 2H), 3.01 (s, 3H), 1.84-1.70 (brm, 4H), 1.41 (s, 9H)
  • Mass Spectral Analysis m/z=480.6 (M−H)
  • Preparation of 53D
  • A 2.0 M solution of hydrochloric acid in diethyl ether (1.66 mL, 3.32 mmol, 4.00 eq) was added dropwise to a solution of 53.2 (0.400 g, 0.83 mmol, 1.00 eq) in anhydrous methylene chloride (15 mL). The reaction mixture was stirred for 3 days. The precipitate was filtered and purified by HPLC using the following conditions to give 75 mg of an off-white solid.
  • HPLC conditions:
  • Column: Waters Xterra Prep RP18 OBD Column, 19×150 mm
  • Detection: UV 210 nm
  • Flow: 15 mL/min
  • Mobile Phase A: 0.2% Ammonium Hydroxide in HPLC Water
  • Mobile Phase B: Acetonitrile
  • Gradient: Linear, 15% B to 90% B in 15 min
  • Yield: 27%
  • 1H NMR (400 MHz, DMSO) δ 7.91 (d, 2H), 7.37 (d, 2H), 7.09 (t, 1H), 6.57 (dd, 1H), 6.48 (dd, 1H), 5.82 (s, 1H), 3.24 (brm, 4H), 2.08 (brm, 2H), 1.97 (brm, 2H)
  • Mass Spectral Analysis m/z=338.7 (M+H)+
  • C20H19NO4, 1H2O
  • Theory: % C, 67.59; % H, 5.96; % N, 3.94.
  • Found: % C, 67.57; % H, 5.92; % N, 4.00.
  • Example 53F Preparation of 53.4
  • To a solution of 53.2 (0.600 g, 1.25 mmol, 1.00 eq), N,N-diisopropylethylamine (0.65 mL, 3.75 mmol, 3.00 eq), and ethylamine hydrochloride (3.4c) (0.203 g, 2.50 mmol, 2.00 eq) in acetonitrile (15 mL) under nitrogen and cooled in an ice/water bath was added O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (0.480 g, 1.50 mmol, 1.20 eq). The ice bath was removed and the reaction was stirred at room temperature for 2 hours and then concentrated. The residue was dissolved in ethyl acetate and the resulting mixture was washed with water and brine. The organic extracts were concentrated and purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity, methanol contained 5% ammonium hydroxide) to give 520 mg of a pale yellow foam.
  • Yield: 82%
  • 1H NMR (400 MHz, DMSO) δ 8.46 (t, 1H), 7.80 (d, 2H), 7.27 (d, 2H), 7.19 (t, 1H), 6.71 (m, 2H), 5.80 (s, 1H), 4.70 (s, 2H), 3.65 (brm, 2H), 3.29 (brm, 4H), 3.02 (s, 3H), 1.83 (brm, 2H), 1.71 (brm, 2H), 1.41 (s, 9H), 1.13 (t, 3H)
  • Mass Spectral Analysis m/z=509.5 (M+H)+
  • Preparation of 53F
  • A 2.0 M solution of hydrochloric acid in diethyl ether (4.05 mL, 8.10 mmol, 8.00 eq) was added drop wise to a solution of 53.4 (0.515 g, 1.01 mmol, 1.00 eq) in anhydrous methanol (10 mL) cooled in an ice/water bath. The mixture was warmed to room temperature and stirring was continued for an additional 3.5 hours. Diethyl ether (10 mL) was added to the solution and the resulting precipitate was collected by filtration, washed with diethyl ether, and dried to give 275 mg of a white solid.
  • Yield: 68%
  • 1H NMR (400 MHz, DMSO) δ 9.50 (s, 1H), 8.92 (brs, 2H), 8.47 (t, 1H), 7.78 (d, 2H), 7.29 (d, 2H), 7.07 (t, 1H), 6.54 (dd, 1H), 6.46 (dd, 1H), 5.76 (s, 1H), 3.30-3.11 (brm, 6H), 2.00 (brm, 4H), 1.13 (t, 3H)
  • Mass Spectral Analysis m/z=365.8 (M+H)+
  • Elemental analysis:
  • C22H24N2O3, 1HCl
  • Theory: % C, 65.91; % H, 6.29; % N, 6.99.
  • Found: % C, 65.54; % H, 6.20; % N, 6.87.
  • Example 54A Preparation of 54.1
  • To a suspension of the crude HCl salt of ketone 21.3 (mother liquor of the recrystallization) (61 g, 400 mmol) in methylene chloride (1200 mL) at 0° C. was added triethylamine (223 mL, 1.6 mol, 4 eq) followed by dropwise addition of benzyl chloroformate (112.6 mL, 800 mmol, 2 eq). The reaction mixture was slowly warmed up to room temperature and stirred overnight, washed with 1 N aqueous solution of hydrochloric acid, 1 N aqueous solution of sodium hydroxide and brine. The organic layer was separated, dried over sodium sulfate, and concentrated in vacuo. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 19%
  • 1H NMR (400 MHz, CDCl3) δ 7.32 (m, 5H), 5.12 (s, 2H), 3.68 (m, 4H), 2.78 (s, 2H), 2.65 (m, 4H), 1.80 (m, 2H).
  • Preparation of 54.2
  • To a solution of Meldrum's acid (38.1) (11.07 g, 76.8 mmol) in methanol (190 mL) was added ammonium acetate (1.18 g, 15.36 mmol, 0.2 eq) followed by compound 54.1 in one portion. The reaction mixture was stirred at room temperature overnight, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity), to yield 26 g (90.6%) of the crude product contaminated with Meldrum's acid and 54.1, which was used without further purification for the next step.
  • Preparation of 54.3
  • To a suspension of CuI (270 mg, 1.4 mmol, 0.03 eq) in anhydrous tetrahydrofuran (200 mL) was added dropwise a 2.0 M solution of benzylmagnesium chloride (28.3a) (35.25 mL, 70.5 mmol, 1.5 eq) in anhydrous tetrahydrofuran under nitrogen atmosphere at −10° C. After the reaction mixture was stirred at −10° C. for 15 min, a solution of crude 54.2 (17.7 g, 47 mmol, 1 eq) in anhydrous tetrahydrofuran (100 mL) was added during a 30 min period. After the addition, the reaction mixture was stirred between −10° C. and −5° C. for 3 hours, and then quenched by a mixture of conc. NH4OH: sat. NH4Cl:H2O (200 mL, 1:2:3). The mixture was extracted with ethyl acetate, and the combined organic layers were washed with a mixture of conc. NH4OH: sat. NH4Cl:H2O (1:2:3) and brine, dried over sodium sulfate, and concentrated in vacuo. Diethyl ether was added to the residue and the mixture was stirred at room temperature overnight. The resulting solid was collected by filtration, washed with diethyl ether, and dried in vacuo. Yield: 60.6%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.31-7.08 (m, 10H), 5.02 (d, 1H), 4.90 (d, 1H), 3.50-2.85 (m, 7H), 2.70 (d, 1H), 1.53 (m, 2H), 1.43 (s, 6H), 1.0 (m, 1H), 0.75 (m, 1H).
  • Preparation of 54.4
  • Compound 54.3 (14 g, 28.72 mmol) was dissolved in a mixture of N,N-dimethylformamide (70 mL) and water (70 mL) and the reaction mixture was heated at ˜120° C. overnight, then cooled to room temperature. The reaction mixture was acidified by adding 1 N aqueous solution of hydrochloric acid to pH 2-3, and kept overnight at room temperature. The upper solvent was decanted, and the sticky residue at the bottom was dissolved in diethyl ether (400 mL). This solution was washed with water, brine, and dried over sodium sulfate. Evaporation of the solvent provided the crude product, which was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 86%
  • 1H NMR (400 MHz, CDCl3) δ 9.7 (brs, 1H), 7.35-7.20 (m, 10H), 5.12 (s, 2H), 3.50 (m, 4H), 2.80 (m, 2H), 2.29 (s, 2H), 1.82-1.50 (m, 6H).
  • Preparation of 54.5
  • To a solution of 54.4 (9.5 g, 24.9 mmol, 1 eq) in anhydrous methylene chloride (150 mL) was added oxalyl chloride (4.36 mL, 49.8 mmol, 2 eq) in one portion followed by 5 drops of anhydrous N,N-dimethylformamide. The reaction mixture was stirred at room temperature for 2 hours and then concentrated in vacuo. The resulting acyl chloride was dissolved in anhydrous methylene chloride (450 mL) and aluminum chloride (6.7 g, 49.8 mmol, 2 eq) was added in one portion. The reaction mixture was stirred at room temperature overnight and then quenched by water (400 mL) followed by addition of concentrated ammonium hydroxide to make the aqueous layer basic. The organic layer was separated and the aqueous layer was extracted with methylene chloride. The combined organic layers were dried over sodium sulfate, and concentrated in vacuo.
  • To the solution of the above crude product in methylene chloride (250 mL) was added triethylamine (6.94 mL, 49.8 mmol, 2 eq) followed by di-tert-butyl dicarbonate (4.7) (7.08 g, 32.37 mmol, 1.3 eq.). The reaction mixture was stirred at room temperature overnight, washed with 0.5 N aqueous solution of hydrochloric acid, brine, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 85%.
  • 1H NMR (400 MHz, CDCl3) δ 8.0 (m, 1H), 7.50 (t, 1H), 7.30 (m, 1H), 7.22 (m, 6H), 3.50-3.23 (m, 4H), 2.92 (d, 1H), 2.83 (d, 1H), 2.60 (d, 1H), 2.52 (d, 1H), 1.60 (m, 6H), 1.47 (s, 9H).
  • Preparation of 54.6
  • A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (25.4 mL, 25.4 mmol, 1.2 eq) was added at −78° C. to a solution of compound 54.5 (7 g, 21 mmol, 1 eq) in tetrahydrofuran (200 mL). After 1 hour, a solution of N-phenyltrifluoromethanesulfonimide (1.4) (9.1 g, 25.4 mmol, 1.2 eq) in tetrahydrofuran (80 mL) was added dropwise. The reaction mixture was then slowly warmed up to room temperature, stirred overnight at room temperature and concentrated in vacuo. The residue was dissolved in diethyl ether, washed with 0.5 N aqueous solution of hydrochloric acid, 1 N aqueous solution of sodium hydroxide, brine, dried over sodium sulfate, and concentrated in vacuo. Yield: 100%.
  • 1H NMR (400 MHz, CDCl3) δ 7.33 (m, 1H), 7.27 (m, 2H), 7.16 (m, 1H), 5.90 (s, 1H), 3.40 (m, 4H), 2.80 (m, 2H), 1.70 (m, 6H), 1.49 (s, 9H).
  • Preparation of 54.7
  • To a solution of 54.6 (1.48 g, 3.2 mmol, 1.0 eq) in dioxane (50 mL) was added sequentially a 2N aqueous solution of potassium carbonate (4.8 mL, 9.6 mmol, 3.0 eq), 1.6 (780 mg, 3.52 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (111 mg, 0.096 mmol, 0.03 eq). The mixture was stirred at room temperature for 2 hours under nitrogen. Ethyl acetate (200 mL) and water (100 mL) were added. The organic layer was separated, washed with water, brine, dried over sodium sulfate, and concentrated in vacuo. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 83%
  • 1H NMR (400 MHz, CDCl3) δ 7.40 (m, 4H), 7.18 (m, 1H), 7.11 (m, 1H), 7.0 (d, 1H), 5.90 (d, 1H), 3.54-3.30 (m, 8H), 2.78 (m, 2H), 1.82-1.62 (m, 6H), 1.50 (s, 9H), 1.23 (m, 3H), 1.12 (m, 3H).
  • Preparation of 54A
  • To the solution of 54.7 (1.3 g, 2.6 mmol) in methylene chloride (50 mL) was added a 2.0 M solution of hydrochloric acid in diethyl ether (7.8 mL, 15.6 mmol, 6 eq). The mixture was stirred at ambient temperature for 24 hours and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (eluent: methylene chloride/methanol mixtures of increasing polarity). Yield: 44.3%.
  • 1H NMR (400 MHz, DMSO-d6) δ 8.80 (brs, 2H), 7.40 (s, 4H), 7.20 (m, 3H), 6.90 (d, 1H), 6.0 (s, 1H), 3.43 (m, 2H), 3.20 (m, 4H), 3.10 (m, 2H), 2.79 (s, 2H), 1.83-1.62 (m, 6H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=389.98 (M+H)+
  • Example 54B Preparation of 54.8
  • To a solution of 54.6 (1.55 g, 3.3 mmol, 1.0 eq) in dioxane (50 mL) was added sequentially a 2N aqueous solution of potassium carbonate (5.0 mL, 10 mmol, 3.0 eq), 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyridine-2-carboxylic acid diethylamide (1.7) (1.1 g, 3.63 mmol, 1.1 eq) and 1,1′-bis(diphenylphosphin0)ferrocene palladium (II) chloride complex with methylene chloride (72 mg, 0.099 mmol, 0.03 eq). The mixture was stirred at room temperature for 45 min under nitrogen. Ethyl acetate (200 mL) and water (100 mL) were added. The organic layer was separated, washed with water, brine, dried over sodium sulfate, and concentrated in vacuo. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 81%
  • 1H NMR (400 MHz, CDCl3) δ 8.56 (δ, 1H), 7.78 (m, 1H), 7.60 (t, 1H), 7.18 (m, 2H), 7.11 (m, 1H), 6.92 (d, 1H), 5.93 (d, 1H), 3.58-3.30 (m, 8H), 2.80 (m, 2H), 1.82-1.62 (m, 6H), 1.49 (s, 9H), 1.22 (m, 6H).
  • Preparation of 54B
  • To the solution of 54.8 (1.31 g, 2.6 mmol) in methylene chloride (50 mL) was added a 2.0 M solution of hydrochloric acid in diethyl ether (9.1 mL, 18.2 mmol, 7 eq). The mixture was stirred at ambient temperature for 24 hours and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (eluent: methylene chloride/methanol mixtures of increasing polarity). Yield: 44.2%.
  • 1H NMR (400 MHz, DMSO-d6) δ 8.90 (brs, 2H), 8.55 (s, 1H), 7.83 (d, 1H), 7.59 (d, 1H), 7.26 (m, 2H), 7.16 (t, 1H), 6.88 (d, 1H), 6.13 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 3.20-3.10 (m, 4H), 2.80 (s, 2H), 1.86-1.62 (m, 6H), 1.16 (t, 3H), 1.10 (t, 3H).
  • Mass Spectral Analysis m/z=390.88 (M+H)+
  • Example 55A Preparation of 55.2
  • To a suspension of CuI (343 mg, 1.8 mmol, 0.036 eq) in anhydrous tetrahydrofuran (500 mL) was added dropwise a 0.25 M solution of 3,5-dimethoxybenzylmagnesium chloride (55.1) (500 mL, 125 mmol, 2.5 eq) in tetrahydrofuran under a nitrogen atmosphere at −10° C. After the reaction mixture was stirred at −10° C. for 30 min, 4-(2,2-dimethyl-4,6-dioxo-[1,3]dioxan-5-ylidene)-piperidine-1-carboxylic acid benzyl ester (38.2) (17.95 g, 50 mmol, 1.0 eq) was added in ten portions to the mixture in a 1 hour period. After the addition, the reaction mixture was stirred between −10° C. and 0° C. for 3 hours, and then quenched by a mixture of conc. NH4OH: sat. NH4Cl:H2O (300 mL, 1:2:3). The mixture was extracted with ethyl acetate, and the combined organic layers were washed with a mixture of conc. NH4OH: sat. NH4Cl:H2O (1:2:3), brine, dried over sodium sulfate, and concentrated in vacuo. To the residue was added diethyl ether and the mixture was stirred overnight. The solid was collected by filtration, washed with diethyl ether, and dried in vacuo. Yield: 100%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.32 (m, 5H), 6.23 (s, 1H), 6.20 (s, 2H), 5.0 (s, 2H), 3.70 (s+m, 8H), 2.98 (m, 2H), 2.81 (m, 2H), 2.70 (s, 2H), 1.46 (s, 6H), 0.90 (m, 2H).
  • Preparation of 55.3
  • Compound 55.2 (26 g, 48.8 mmol) was dissolved in a mixture of N,N-dimethylformamide (150 mL) and water (150 mL) and heated at ˜120° C. for 3 days, then cooled to room temperature. To the reaction mixture was added 1 N aqueous solution of sodium hydroxide (125 mL) and water (500 mL). The resulting mixture was extracted with diethyl ether. The aqueous phase was then acidified with 6 N aqueous hydrochloric acid, and extracted with diethyl ether. The combined organic extracts were washed with water and brine, dried over sodium sulfate, and concentrated in vacuo. Yield: 98.4%.
  • 1H NMR (400 MHz, CDCl3) δ 11.0 (brs, 1H), 7.35 (m, 5H), 5.10 (s, 2H), 3.75 (s+m, 8H), 3.32 (m, 2H), 2.78 (s, 2H), 2.38 (s, 2H), 1.59 (m, 4H).
  • Preparation of 55.4
  • To a solution of compound 55.3 (23.8 g, 55.73 mmol) in trifluoroacetic acid (250 mL) was added dropwise trifluoroacetic anhydride (93 mL, 669 mmol, 12 eq) under nitrogen atmosphere at room temperature. The reaction was stirred for 2 hours and concentrated in vacuo. The residue was dissolved in methylene chloride. The mixture was washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was separated, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (ethyl acetate-methylene chloride, 1:1), to yield the spiro ketone 55.4. Yield: 70.8%.
  • 1H NMR (400 MHz, CDCl3) δ 7.33 (m, 5H), 6.33 (s, 1H), 6.30 (s, 1H), 5.10 (s, 2H), 3.90 (s, 3H), 3.86 (s, 3H), 3.59 (m, 2H), 3.41 (m, 2H), 2.85 (s, 2H), 2.56 (s, 2H), 1.50 (m, 4H).
  • Preparation of 55.5
  • A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (28 mL, 28 mmol, 1.24 eq) was added at −78° C. to a solution of compound 55.4 (9.26 g, 22.64 mmol, 1 eq) in tetrahydrofuran (200 mL). After 45 minutes, a solution of N-phenyltrifluoromethanesulfonimide (1.4) (9.8 g, 27.4 mmol, 1.2 eq) in tetrahydrofuran (80 mL) was added dropwise. The reaction mixture was then warmed up to room temperature and stirred for 2 hours at room temperature, quenched by addition of water (200 mL), and extracted with a mixture of hexane and diethyl ether (1:1). The organic extracts were combined and washed with water, brine and dried over sodium sulfate. Evaporation of the solvent gave the crude compound, which was purified by column chromatography on silica gel (hexane-ethyl acetate, 2:1), to yield the enol triflate derivative 55.5. Yield: 83.3%
  • 1H NMR (400 MHz, CDCl3) δ 7.33 (m, 5H), 6.33 (m, 2H), 5.70 (s, 1H), 5.11 (s, 2H), 3.86 (s, 3H), 3.81 (s, 3H), 3.73 (m, 2H), 3.29 (m, 2H), 2.69 (s, 2H), 1.61 (m, 2H), 1.45 (m, 2H).
  • Preparation of 55.6
  • To a solution of the enol triflate derivative 55.5 (9.8 g, 18.12 mmol) in dimethoxyethane (150 mL) was added sequentially a 2 N aqueous solution of sodium carbonate (30 mL, 60 mmol, 3.3 eq), lithium chloride (2.6 g, 61.3 mmol, 3.4 eq), 4-(N,N-diethylaminocarbonyl)phenylboronic acid) 1.6 (4.81 g, 21.77 mmol, 1.2 eq) and tetrakis(triphenylphosphine)palladium(0) (630 mg, 0.55 mmol, 0.03 eq). The reaction mixture was refluxed overnight, cooled to room temperature, diluted with water (200 mL) and extracted with diethyl ether. The combined organic extracts were dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane-ethyl acetate, 1:2). Yield: 98.2%
  • 1H NMR (400 MHz, CDCl3) δ 7.36-7.22 (m, 10H), 6.40 (s, 1H), 6.30 (s, 1H), 5.89 (s, 1H), 5.11 (s, 2H), 3.81 (s, 3H), 3.78 (m, 2H), 3.53 (m, 2H), 3.39 (s, 3H), 3.30 (m, 4H), 2.68 (s, 2H), 1.62 (m, 2H), 1.43 (m, 2H), 1.22 (m, 3H), 1.10 (m, 3H).
  • Preparation of 55A
  • To a solution of 55.6 (3.41 g, 6 mmol) in methylene chloride (60 mL) was added dropwise a 1.0 M solution of boron tribromide in methylene chloride (60 mL, 60 mmol, 10 eq) under nitrogen atmosphere at −40° C. The reaction was slowly warmed to room temperature and stirred overnight at room temperature. A 1 N aqueous solution of hydrochloric acid was added to quench the reaction and the resulting mixture was extracted with diethyl ether. The aqueous layer was basified to pH 9 with a 3 N aqueous solution of sodium hydroxide, and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate, and concentrated in vacuo to yield 1.46 g (60%) of the crude product, which was further purified by column chromatography (eluent: methylene chloride-methanol-ammonium hydroxide, 20:1:1).
  • 1H NMR (400 MHz, DMSO-d6) δ 9.42 (s, 1H), 9.07 (s, 1H), 8.90 (br, 2H), 7.28 (d, 2H), 7.20 (d, 2H), 6.20 (s, 1H), 6.13 (s, 1H), 5.80 (s, 1H), 3.33-3.10 (m, 8H), 2.55 (s, 2H), 1.68 (m, 2H), 1.58 (m, 2H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=407.51 (M+H)+
  • Example 55B Preparation of 55.7
  • To the solution of crude 55A (1.46 g, 3.6 mmol) in methylene chloride (50 mL) was added triethylamine (2 mL, 14.4 mmol, 4 eq) followed by di-t-butyl dicarbonate (4.7) (787 mg, 3.6 mmol, 1.0 eq.). The reaction mixture was stirred at room temperature for 1 hour, and then concentrated in vacuo. The residue was purified by column chromatography (eluent: hexane-acetone, 3:2).
  • Yield: 42.8% for two steps.
  • 1H NMR (400 MHz, CDCl3) δ 8.30 (s, 1H), 7.39 (s, 4H), 6.35 (s, 1H), 5.80 (s, 1H), 5.66 (s, 1H), 5.02 (s, 1H), 3.60 (m, 4H), 3.30 (m, 4H), 2.60 (s, 2H), 2.00 (m, 1H), 1.60 (m, 2H), 1.43 (s+m, 11H), 1.28 (m, 3H), 1.12 (m, 3H).
  • Preparation of 55.8
  • To a solution of 55.7 (506 mg, 1 mmol) in dichloromethane was added added triethylamine (0.35 mL, 2.5 mmol, 2.5 eq) followed by N-phenyltrifluoromethanesulfonimide (1.4) (368 mg, 1.03 mmol, 1.03 eq). The reaction mixture was stirred at room temperature overnight and additional quantities of N-phenyltrifluoromethanesulfonimide (1.4) (90 g, 0.25 mmol, 0.25 eq) were added to the reaction mixture. The reaction mixture was stirred for an additional 24 hours at room temperature, washed with a saturated aqueous solution of sodium bicarbonate. The organic layer was separated, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (hexane-acetone-triethylamine, 3:1:0.1), to yield 55.8. Yield: 50.2%
  • 1H NMR (400 MHz, CDCl3) δ 7.40 (d, 2H), 7.37 (d, 2H), 6.73 (s, 1H), 6.70 (s, 1H), 6.06 (s, 1H), 3.69 (m, 2H), 3.55 (m, 2H), 3.28 (m, 2H), 2.73 (s, 2H), 1.60 (m, 2H), 1.48 (s+m, 11H), 1.28 (m, 3H), 1.12 (m, 3H).
  • Preparation of 55.9
  • To a mixture of 55.8 (383 mg, 0.6 mmol), palladium acetate (4.5 mg, 0.02 mmol), 1,3-bis(diphenylphosphine)propane (8 mg, 0.02 mmol) in anhydrous N,N-dimethylformamide (6 mL) was added under nitrogen atmosphere at 60° C. triethylsilane (0.40 mL, 2.5 mmol). The reaction mixture was stirred at 60° C. for 24 hours, cooled to room temperature, and diluted with diethyl ether. The mixture was washed successively with water, a saturated aqueous solution of sodium bicarbonate, brine and dried over sodium sulfate. Evaporation of the solvent provided the crude residue, which was purified by column chromatography on silica gel (ethyl acetate-methylene chloride 1:3), to yield 80 mg (30%) of the desired phenol 55.9, and 120 mg (33%) of the corresponding silyl ether.
  • Compound 55.9: 1H NMR (400 MHz, CDCl3) δ 7.42 (s, 4H), 7.10 (t, 1H), 6.80 (d, 1H), 6.72 (d, 1H), 6.0 (s, 1H), 5.0 (s, 1H), 3.64 (m, 2H), 3.52 (m, 2H), 3.28 (m, 4H), 2.71 (s, 2H), 1.60 (m, 2H), 1.45 (s+m, 11H), 1.23 (m, 3H), 1.12 (m, 3H).
  • Preparation of 55B
  • To the solution of 55.9 (78 mg, 0.16 mmol) in methylene chloride (4 mL) was added a 2.0 M hydrochloric acid in diethyl ether (10 mL, 20 mmol). The mixture was stirred at ambient temperature for 2 days. To the solution of the silyl ether of 55.9 (110 mg, 0.18 mmol) in methylene chloride (4 mL) was added 2.0 M hydrochloric acid in diethyl ether (10 mL, 20 mmol). The mixture was stirred at ambient temperature for 2 days. The two reaction mixtures were diluted with diethyl ether, combined and filtered. The solid was collected and dried in vacuo.
  • Yield: 75.3%.
  • 1H NMR (400 MHz, DMSO-d6) δ 9.20 (s, 1H), 8.90 (brs, 1H), 8.72 (brs, 1H), 7.30 (d, 2H), 7.21 (d, 2H), 7.06 (t, 1H), 6.75 (d, 1H), 6.69 (d, 1H), 6.03 (s, 1H), 3.50-3.10 (m, 8H), 2.66 (s, 2H), 1.68 (m, 2H), 1.53 (m, 2H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=391.53 (M+H)+
  • Example 55C
  • Iodotrimethylsilane (0.62 mL, 4.4 mmol, 2.9 eq) was added to the solution of compound 55.6 (852 mg, 1.5 mmol) in anhydrous methylene chloride (10 mL) under nitrogen. The reaction mixture was stirred at room temperature for 1 hour, quenched with 1 N aqueous solution of hydrochloric acid (20 mL), and extracted with diethyl ether. The aqueous phase was basified to pH 9-10 with a 3 N aqueous solution of sodium hydroxide, and extracted with methylene chloride. The organic extracts were combined, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (methylene chloride-methanol-ammonium hydroxide, 10:1:1). Yield: 60%.
  • 1H NMR (400 MHz, CDCl3) δ 7.30 (d, 2H), 7.25 (d, 2H), 7.21 (d, 2H), 6.43 (d, 1H), 6.30 (d, 1H), 5.93 (s, 1H), 3.80 (s, 3H), 3.53 (m, 2H), 3.38 (s, 3H), 2.90 (m, 4H), 2.68 (s, 2H), 1.60 (m, 2H), 1.41 (m, 2H), 1.22 (m, 3H), 1.12 (m, 3H).
  • Mass Spectral Analysis m/z=435.70 (M+H)+
  • Elemental analysis:
  • C27H34N2O3, 1H2O
  • Theory: % C, 71.65; % H, 8.02; % N, 6.19.
  • Found: % C, 71.32; % H, 7.80; % N, 6.11.
  • Example 56A
  • To a solution of 28.8b (3.23 g, 6 mmol) in methylene chloride (50 mL) was added dropwise a 1.0 M solution of boron tribromide in methylene chloride (60 mL, 60 mmol, 10 eq) under nitrogen atmosphere at −50° C. The reaction was kept between −50° C. and −10° C. for 1 hour, and then slowly warmed up to room temperature and stirred overnight at room temperature. A 1 N aqueous solution of hydrochloric acid was added to quench the reaction mixture, which was extracted with diethyl ether. The aqueous layer was basified to pH 9 with a 3 N aqueous solution of sodium hydroxide, and extracted with methylene chloride (small amount of methanol was added to increase the solubility). The organic extracts were combined, dried over sodium sulfate, and concentrated in vacuo. Yield: 98.3%.
  • 1H NMR (400 MHz, DMSO-d6) δ 9.10 (brs, 1H), 9.07 (s, 1H), 7.38 (d, 2H), 7.32 (d, 2H), 7.03 (d, 1H), 6.58 (dd, 1H), 6.35 (d, 1H), 6.0 (s, 1H), 3.40-3.25 (m, 4H), 2.72 (m, 4H), 2.63 (s, 2H), 1.42 (m, 2H), 1.35 (m, 2H), 1.10 (m, 6H).
  • Mass Spectral Analysis m/z=391.45 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1/4H2O
  • Theory: % C, 76.01; % H, 7.78; % N, 7.09.
  • Found: % C, 75.90; % H, 7.67; % N, 7.09.
  • Example 56B
  • Compound 56A (190 mg, 1 mmol) was dissolved in methanol (30 mL), and hydrogenated in the presence of 10% Pd/C (120 mg) using a hydrogen balloon. After 3 days at room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride-methanol-conc. ammonia hydroxide, 8:1:1). Yield: 94.4%
  • 1H NMR (400 MHz, CDCl3) δ 7.28 (d, 2H), 7.18 (d, 2H), 6.91 (d, 1H), 6.60 (dd, 1H), 6.19 (d, 1H), 4.4 (br, 1H), 3.97 (m, 1H), 3.50 (m, 2H), 3.25 (m, 2H), 2.80 (m, 2H), 2.75 (m, 2H), 2.54 (d, 1H), 2.0 (m, 1H), 1.55-1.43 (m, 4H), 1.21 (m, 3H), 1.11 (m, 3H).
  • Mass Spectral Analysis m/z=393.59 (M+H)+
  • Example 56C
  • To a solution of 40.3 (850 mg, 1.58 mmol) in methylene chloride (15 mL) was added dropwise a 1.0 M solution of boron tribromide in methylene chloride (11 mL, 11 mmol, 7 eq) under nitrogen atmosphere at −50° C. The reaction was kept between −50° C.-−10° C. for 1 hour, and then slowly warmed up to room temperature and stirred overnight at room temperature. A 1 N aqueous solution of hydrochloric acid was added to quench the reaction mixture, which was extracted with diethyl ether. The aqueous layer was basified to pH 9 with a 3 N aqueous solution of sodium hydroxide, and extracted with methylene chloride (small amount of methanol was added to increase the solubility). The organic extracts were combined, dried over sodium sulfate, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (methylene chloride-methanol-ammonium hydroxide, 6:1:1). Yield: 73%.
  • 1H NMR (400 MHz, DMSO-d6) δ 9.22 (brs, 1H), 8.51 (d, 1H), 7.83 (dd, 1H), 7.58 (d, 1H), 7.04 (d, 1H), 6.60 (dd, 1H), 6.30 (d, 1H), 6.12 (s, 1H), 3.48 (q, 2H), 3.30 (q, 2H), 2.77 (m, 4H), 2.69 (s, 2H), 1.50 (m, 2H), 1.32 (m, 2H), 1.18 (t, 3H), 1.11 (t, 3H).
  • Mass Spectral Analysis m/z=392.44 (M+H)+
  • Elemental analysis:
  • C25H30N2O2, 1/4H2O
  • Theory: % C, 71.44; % H, 7.58; % N, 10.41.
  • Found: % C, 71.44; % H, 7.40; % N, 10.38.
  • Example 56D
  • Compound 56C (190 mg, 1 mmol) was dissolved in methanol (20 mL) and hydrogenated in the presence of 10% Pd/C (120 mg) using a hydrogen balloon. After 3 days at room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride-methanol-conc. ammonia hydroxide, 8:1:1). Yield: 53.9%
  • 1H NMR (400 MHz, CDCl3) δ 8.40 (d, 1H), 7.52 (dd, 1H), 7.48 (d, 1H), 6.96 (d, 1H), 6.62 (dd, 1H), 6.12 (d, 1H), 4.50 (br, 1H), 4.01 (m, 1H), 3.53 (q, 2H), 3.40 (q, 2H), 2.88 (m, 2H), 2.80 (m, 2H), 2.60 (d, 1H), 2.0 (m, 1H), 1.50 (m, 4H), 1.25 (t, 3H), 1.18 (m, 3H).
  • Mass Spectral Analysis m/z=394.51 (M+H)+
  • Example 57D Preparation of 57.1
  • 4.1 (0.27 mL, 1.91 mmol, 1.2 eq) was added dropwise to a cooled 0° C. solution of 31J (0.50 g, 1.59 mmol, 1.0 eq) and triethylamine (0.67 mL, 4.78 mmol, 3.0 eq) in tetrahydrofuran (5 mL). The reaction mixture was stirred overnight at room temperature. Dichloromethane (1 mL) was added to help solubilizing the reaction mixture. An additional portion of 4.1 (1.00 mL, 7.11 mmol, 4.5 eq) was added dropwise. Stirring was continued for 3 hours. The reaction mixture was diluted with a 1N aqueous solution of hydrochloric acid (100 mL) and extracted with dichloromethane (2×100 mL). The organic extracts were combined, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 87%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.43 (m, 3H), 7.33 (m, 2H), 7.23 (t, 1H), 7.01 (d, 1H), 6.97 (m, 1H), 6.91 (m, 1H), 5.81 (s, 1H), 4.11 (m, 1H), 3.79 (m, 1H), 3.66 (m, 1H), 3.40 (m, 1H), 2.04 (m, 2H), 1.85 (m, 2H)
  • Preparation of 57.2
  • To a solution of 57.1 (0.50 g, 1.34 mmol, 1.0 eq) in 1,2-dichloroethane (5 mL) was added 4.3 (0.31 g, 2.01 mmol, 1.5 eq) portionwise. The reaction mixture was refluxed at 65° C. overnight. The reaction mixture was cooled to room temperature and then to 0° C. in an ice/brine bath. A 2M solution of oxalyl chloride in methylene chloride (1.07 mL, 2.14 mmol, 1.6 eq) was added dropwise to the reaction mixture. The reaction mixture was warmed to room temperature, then heated to 65° C. for 3 hours. The reaction mixture was cooled to 0° C. and quenched by addition of water (50 mL). The layers were separated and the aqueous was extracted with dichloromethane (2×100 mL). The organic extracts were combined, washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 74%
  • 1H NMR (400 MHz, DMSO d6) δ 7.46 (m, 4H), 7.31 (m, 2H), 7.24 (d, 1H), 6.95 (d, 1H), 5.81 (s, 1H), 4.10 (m, 1H), 3.75 (m, 1H), 3.66 (m, 1H), 3.40 (m, 1H), 2.04 (m, 2H), 1.85 (m, 2H)
  • Preparation of 57.4
  • To a solution of 57.2 (0.40 g, 0.85 mmol, 1.0 eq) in tetrahydrofuran (5 mL) at 0° C. under an atmosphere of nitrogen was added 5.1 (0.18 mL, 5.60 mmol, 6.6 eq) dropwise. The reaction mixture was stirred at 0° C. for 30 minutes. The reaction mixture was concentrated under reduced pressure. Dichloromethane (50 mL) and water (50 mL) were added and the reaction mixture was stirred for 10 minutes. The phases were separated and the aqueous phase was extracted with dichloromethane (2×50 mL). The organic extracts were combined, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity).
  • Yield: 67%; Mass Spectral Analysis m/z=466.43 (M−H)
  • 1H NMR (400 MHz, DMSO-d6) δ 8.22 (s, 1H), 7.65 (m, 1H), 7.47 (m, 4H), 7.39 (m, 2H), 7.20 (d, 1H), 5.94 (s, 1H), 4.13 (m, 1H), 3.80 (m, 1H), 3.68 (m, 1H), 3.41 (m, 1H), 3.34 (s, 2H), 2.07 (m, 2H), 1.91 (m, 2H)
  • Preparation of 57.5
  • To a solution of 57.4 (0.25 g, 0.54 mmol, 1.0 eq) in ethanol (5 mL) at room temperature under an atmosphere of nitrogen was added sodium acetate (0.29 g, 3.58 mmol, 6.7 eq) and 2.8c (0.18 mL, 2.94 mmol, 5.5 eq). The reaction mixture was refluxed at 90° C. overnight. The reaction mixture was concentrated under reduced pressure. The resulting oil was diluted with water (50 mL) then extracted with dichloromethane (3×50 mL). The organic extracts were combined, washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 73%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.81 (m, 1H), 7.48 (m, 4H), 7.38 (m, 2H), 7.26 (d, 1H), 5.97 (s, 1H), 4.14 (m, 1H), 3.79 (m, 1H), 3.69 (m, 1H), 3.41 (m, 1H), 3.13 (s, 3H), 2.08 (m, 2H), 1.93 (m, 2H)
  • Mass Spectral Analysis m/z=451.33 (M+H)+
  • Preparation of 57D
  • To a solution of 57.5 (0.15 g, 0.33 mmol; 1.0 eq) in methanol (4 mL) and water (1 mL) at room temperature and under an atmosphere of nitrogen was added potassium carbonate (0.30 g, 2.19 mmol, 6.6 eq). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water and ethyl acetate. The phases were separated and the organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure.
  • Yield: 94%
  • 1H NMR (400 MHz, DMSO d6) δ 7.76 (m, 1H), 7.47 (m, 4H), 7.37 (m, 2H), 7.20 (d, 1H), 5.96 (s, 1H), 3.12 (s, 3H), 3.03 (s, 1H), 2.93 (m, 2H), 2.83 (m, 2H), 1.85 (m, 2H), 1.78 (m, 2H)
  • Mass Spectral Analysis m/z=356.30 (M+H)+
  • Example 58A Preparation of 58.1a
  • To a solution of 2.5 (3.58 g, 6.18 mmol, 1.0 eq) in 1,4-dioxane (18 mL) at room temperature and under an atmosphere of nitrogen was added 2M solution of potassium carbonate in water (9.26 mL, 18.53 mmol, 3.0 eq), 3.6a (1.11 g, 6.79 mmol, 1.1 eq) and [1,1′-bis(diphenylphosphino)ferrocene] dichloropalladium(II) complex with dichloromethane (1:1) (0.25 g, 0.31 mmol, 0.05 eq). The reaction mixture was heated at 60° C. for 4 hours. The reaction mixture was cooled to room temperature then diluted with water (20 mL) and stirred for an additional 20 minutes at room temperature. The mixture was then extracted with diethyl ether (1×20 mL). The organic layer was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 54%
  • 1H NMR (400 MHz, DMSO-d6) δ 8.61 (d, 1H), 8.56 (s, 1H), 7.77 (m, 1H), 7.47 (m, 1H), 6.88 (d, 1H), 6.72 (m, 1H), 6.31 (d, 1H), 5.95 (s, 1H), 3.69 (m, 2H), 3.34 (m, 2H), 1.84 (m, 2H), 1.70 (m, 2H), 1.41 (s, 9H), 0.87 (s, 9H), 0.83 (s, 6H)
  • Mass Spectral Analysis m/z=509.56 (M+H)+
  • Preparation of 58A
  • To a solution of 58.1a (0.60 g, 1.18 mmol, 1.0 eq) in methanol (5 mL) at room temperature and under an atmosphere of nitrogen was added a 4M solution of hydrogen chloride in 1,4-dioxane (0.59 mL, 2.40 mmol, 2.0 eq). The reaction mixture was stirred for 2 days at room temperature. The reaction mixture was concentrated under reduced pressure to dryness. The resulting oil was purified by column chromatography (eluent: dichloromethane/methanol mixtures of increasing polarity, methanol contains 10% ammonium hydroxide). Yield: 25%
  • 1H NMR (400 MHz, DMSO d6) δ 9.38 (br s, 2H), 9.19 (br s, 1H), 8.74 (s, 2H), 8.04 (d, 1H), 7.70 (m, 1H), 6.90 (d, 1H), 6.68 (d, 1H), 6.37 (s, 1H), 6.06 (s, 1H), 4.05 (br s, 1H), 3.17 (m, 4H), 2.06 (m, 4H)
  • Mass Spectral Analysis m/z=295.32 (M+H)+
  • Example 58B
  • Example 58B was obtained according to a procedure similar to the one described for 58A, with the following exceptions:
  • Step 58.1: 3.6a was replaced by 31.1n.
  • Step 58.2: 58.1a was replaced by 58.1b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.20 (s, 1H), 9.14 (br s, 2H), 8.00 (m, 1H), 7.89 (m, 1H), 7.58 (s, 1H), 7.42 (m, 2H), 6.90 (m, 2H), 6.72 (m, 1H), 6.14 (s, 1H), 3.17 (m, 4H), 2.02 (m, 4H)
  • Mass Spectral Analysis m/z=350.33 (M+H)+
  • Example 58C Preparation of 58.2
  • To a solution of 2.5 (15.00 g, 25.87 mmol, 1.00 eq) in 1,4-dioxane (65 mL) at room temperature and under an atmosphere of nitrogen was added a solution of potassium carbonate (10.73 g, 77.63 mmol, 3.0 eq) in water (32 mL), phenylboronic acid (3.47 g, 28.46 mmol, 1.1 eq) and tetrakis(triphenylphosphine)palladium(0) (1.49 g, 1.29 mmol, 0.05 eq). The reaction mixture was stirred at 80° C. overnight. The reaction mixture was cooled to room temperature then diluted with water (20 mL) and stirred for an additional 20 minutes at room temperature. The mixture was then extracted with diethyl ether (1×20 mL). The organic layer was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 39%
  • Mass Spectral Analysis m/z=508.53 (M+H)+
  • Preparation of 58.3
  • To a solution of 58.2 (5.11 g, 10.06 mmol, 1.0 eq) in tetrahydrofuran (10 mL) at 0° C., under an atmosphere of nitrogen, was added dropwise a 1M solution of tetra-n-butylammonium fluoride in tetrahydrofuran (30.19 mL, 30.19 mmol, 3.0 eq). The reaction mixture was allowed to warm to room temperature and stirred overnight at room temperature. The reaction mixture was diluted with saturated sodium bicarbonate (50 mL) then extracted with ethyl acetate (2×50 mL). The organic layer was washed with a 1N aqueous solution of hydrogen chloride (50 mL) then brine (50 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The product was triturated with diethyl ether then isolated by vacuum filtration and dried under vacuum overnight. Yield: 89%
  • 1H NMR (400 MHz, DMSO d6) δ 8.92 (s, 1H), 7.43 (m, 3H), 7.34 (m, 2H), 6.78 (d, 1H), 6.58 (m, 1H), 6.40 (d, 1H), 5.77 (s, 1H), 3.71 (m, 2H), 3.23 (m, 2H), 1.82 (m, 2H), 1.65 (m, 2H), 1.41 (s, 9H)
  • Mass Spectral Analysis m/z=394.44 (M+H)+
  • Preparation of 58.5a
  • To a solution of 58.3 (0.50 g, 1.27 mmol, 1.0 eq) and potassium carbonate (0.58 g, 4.19 mmol, 3.3 eq) in N,N-dimethylformamide (5 mL) at 0° C. and under an atmosphere of nitrogen was added dropwise 2.8c (0.26 mL, 4.19 mmol, 3.3 eq). The reaction mixture was stirred for 3 days at 100° C. The reaction mixture was cooled to room temperature and partitioned between water (50 mL) and diethyl ether (50 mL). The phases were separated and the organic layer was dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 58%
  • 1H NMR (400 MHz, DMSO-d6) δ 7.43 (m, 3H), 7.35 (m, 2H), 6.92 (d, 1H), 6.81 (m, 1H), 6.46 (d, 1H), 5.84 (s, 1H), 3.71 (m, 2H), 3.61 (s, 3H), 3.24 (m, 2H), 1.83 (m, 2H), 1.68 (m, 2H), 1.41 (s, 9H)
  • Mass Spectral Analysis m/z=408.86 (M+H)+
  • Preparation of 58C
  • To a solution of 58.5a (0.30 g, 0.74 mmol, 1.0 eq) in methylene chloride (4 mL) at room temperature and under an atmosphere of nitrogen was added a 2M solution of hydrogen chloride in diethyl ether (2.02 mL, 4.04 mmol, 5.5 eq). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated under reduced pressure, then triturated with ethyl acetate. The product was isolated by vacuum filtration and dried under vacuum overnight. Yield: 69%
  • 1H NMR (400 MHz, DMSO d6) δ 9.17 (br s, 2H), 7.44 (m, 3H), 7.37 (m, 2H), 7.00 (d, 1H), 6.84 (m, 1H), 6.47 (d, 1H), 5.90 (s, 1H), 3.62 (s, 3H), 3.20 (m, 4H), 2.03 (m, 4H)
  • Mass Spectral Analysis m/z=308.28 (M+H)+
  • Example 58D
  • 58D was obtained according to a procedure similar to the one described for 58C, with the following exceptions:
  • Step 58.5: 2.8c was replaced by 58.4.
  • Step 58.6: 58.5a was replaced by 58.5b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.07 (br s, 2H), 7.45 (m, 3H), 7.37 (m, 2H), 6.97 (d, 1H), 6.82 (m, 1H), 6.45 (d, 1H), 5.89 (s, 1H), 3.64 (d, 2H), 3.20 (m, 4H), 2.01 (m, 4H), 1.10 (m, 1H), 0.50 (m, 2H), 0.22 (m, 2H)
  • Mass Spectral Analysis m/z=348.26 (M+H)+
  • Example 59A Preparation of 59.4a
  • To a solution of 59.2a (3.00 g, 14.6 mmol, 1.00 eq) in methanol (75 mL) was added triethylamine (2.24 mL, 16.1 mmol, 1.10 eq). The solution was cooled in an ice/water bath and 59.3 (1.92 mL, 16.1 mmol, 1.10 eq) was added slowly. The reaction mixture was stirred at 0° C. for 2.5 hours and then concentrated. The residue was dissolved in methylene chloride. The organic phase was washed with water, a saturated solution of sodium bicarbonate and brine. The organic extracts were concentrated under reduced pressure and dried to give 2.85 g of a white solid. Yield: 89%
  • 1H NMR (400 MHz, DMSO) δ 9.67 (brs, 1H), 3.59 (m, 4H)
  • Preparation of 59.6
  • To a solution of 59.5 (20.0 g, 0.110 mol, 1.00 eq) in N,N-dimethylformamide (100 mL) was added sodium azide (7.86 g, 0.121 mol, 1.10 eq) and ammonium chloride (6.46 g, 0.121 mol, 1.10 eq). The reaction mixture was heated at 125° C. for 20 hours, cooled to room temperature and then cooled in a brine/ice bath. A 1N solution of hydrochloric acid (50 mL) was slowly added to the reaction mixture. A thick precipitate formed and water (200 mL) was added to facilitate stirring. To reach pH 1, a 6N solution of hydrochloric acid (20 mL) was added carefully. The solids were filtered and dried to give 25 g of a off-white solid. Yield: 100%
  • 1H NMR (400 MHz, DMSO) δ 8.00 (d, 2H), 7.84 (d, 2H)
  • Mass Spectral Analysis m/z=223.5 (M−H)
  • Preparation of 59.7a
  • To a solution of 59.6 (2.33 g, 10.4 mmol, 1.00 eq) in N,N-dimethylformamide (50 mL) was added triethylamine (2.89 mL, 20.7 mmol, 2.00 eq). 59.4a (2.85 g, 13.0 mmol, 1.25 eq) was then added to the reaction mixture, which was stirred at room temperature for 16 hours. The reaction was not complete as evidenced by LC/MS, so it was heated at 50° C. for 24 hours. The reaction mixture was diluted with cold water and the product was extracted two times with ethyl acetate. The crude product was concentrated and purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity) and the resulting solid was triturated in hexanes, filtered, and dried to give 1.67 g of a white solid. Yield: 44%
  • 1H NMR (400 MHz, DMSO) δ 9.64 (t, 1H), 7.99 (d, 2H), 7.79 (d, 2H), 4.90 (m, 2H), 3.76 (m, 2H)
  • Mass Spectral Analysis m/z=362.2 (M−H)
  • Preparation of 59.8a
  • To a solution of 59.7a (1.00 g, 2.75 mmol, 1.00 eq), 32.1 (2.35 g, 5.49 mmol, 2.00 eq), and potassium carbonate (1.14, 8.24 mmol, 3.00 eq) in dioxane (25 mL) and water (5 mL) was added tetrakis(triphenylphosphine)palladium(0) (0.200 g, 0.10 mmol, 0.05 eq). The reaction mixture was stirred at room temperature for 16 hours, heated at 45° C. for 1 hour, and then diluted with water. Methylene chloride was added and the layers were separated. The aqueous layer was washed again with methylene chloride and the organic extracts were combined and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity) to give 1.52 g of a light yellow foam. Yield: 95%
  • 1H NMR (400 MHz, CDCl3) δ 8.15 (d, 2H), 7.49 (d, 2H), 7.20 (m, 1H), 7.08 (brs, 1H), 7.02 (dd, 1H), 6.96 (dd, 1H), 6.87 (m, 1H), 5.62 (s, 1H), 4.90 (m, 2H), 4.07 (m, 2H), 3.87 (m, 2H), 3.34 (m, 2H), 2.06 (m, 2H), 1.68 (m, 2H), 1.48 (s, 9H)
  • Mass Spectral Analysis m/z=583.6 (M−H)
  • Preparation of 59.9a
  • To a solution of 59.8a (1.50 g, 2.56 mmol, 1.00 eq) in methanol (40 mL) was added potassium carbonate (1.77 g, 12.8 mmol, 5.00 eq). The reaction mixture was heated at 50° C. for 24 hours and then at 60° C. for an additional 24 hours. The reaction mixture was concentrated under reduced pressure. The crude residue was purified by column chromatography (eluent: methanol/methylene chloride mixtures of increasing polarity, methanol containing 5% ammonium hydroxide) to give 1.11 g of a light orange foam. Yield: 88%
  • 1H NMR (400 MHz, DMSO) δ 8.13 (d, 2H), 7.54 (d, 2H), 7.23 (m, 1H), 7.00 (m, 2H), 6.92 (m, 1H), 5.93 (s, 1H), 4.69 (t, 2H), 3.73 (m, 2H), 3.33 (s, 2H), 3.13 (t, 2H), 1.89 (m, 2H), 1.73 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=489.5 (M+H)+
  • Preparation of 59A
  • To a solution of 59.9a (0.500 g, 1.02 mmol, 1.00 eq) in methylene chloride (15 mL) was added a 2.0 M solution of hydrochloric acid in diethyl ether (3.10 mL, 6.10 mmol, 6.00 eq). The reaction mixture was stirred at room temperature for 3 days. The resulting solid was filtered, rinsed with diethyl ether, and dried to give 458 mg of a pale orange solid. Yield: 96%
  • 1H NMR (400 MHz, DMSO) δ 9.05 (brs, 2H), 8.27 (brs, 3H), 8.17 (d, 2H), 7.60 (d, 2H), 7.29 (m, 1H), 7.07 (dd, 1H), 7.04 (dd, 1H), 6.97 (m, 1H), 6.00 (s, 1H), 5.05 (t, 2H), 3.51 (t, 2H), 3.22 (brm, 4H), 2.05 (brm, 4H)
  • Mass Spectral Analysis m/z=389.4 (M+H)+
  • Elemental analysis:
  • C22H24N6O, 2HCl, 1.5H2O
  • Theory: % C, 54.10; % H, 5.98; % N, 17.21; % Cl, 14.52.
  • Found: % C, 54.43; % H, 5.94; % N, 16.90; % Cl, 14.80.
  • Example 59B
  • 59B was obtained according to a procedure similar to the one described for 59A, with the following exceptions:
  • Step 59.2: 59.2a was replaced by 59.2b.
  • Step 59.4: 59.4a was replaced by 59.4b.
  • Step 59.5: 59.7a was replaced by 59.7b.
  • Step 59.6: 59.8a was replaced by 59.8b.
  • Step 59.7: 59.9a was replaced by 59.9b.
  • 1H NMR (400 MHz, DMSO d6) δ 9.21 (brs, 2H), 8.15 (m, 5H), 7.59 (d, 2H), 7.28 (m, 1H), 7.07 (m, 1H), 7.03 (dd, 1H), 6.96 (m, 1H), 5.99 (s, 1H), 4.91 (t, 2H), 3.22 (brm, 4H), 2.93 (brm, 2H), 2.32 (m, 2H), 2.06 (brm, 4H)
  • Mass Spectral Analysis m/z=403.4 (M+H)+
  • Elemental analysis:
  • C23H26N6O, 2HCl, 1H2O
  • Theory: % C, 55.99; % H, 6.13; % N, 17.03.
  • Found: % C, 56.01; % H, 6.23; % N, 16.93.
  • Example 59C Preparation of 59.2c
  • A solution of 59.1a (4.79 g, 46.4 mmol, 1.00 eq) in 48% aqueous hydrobromic acid (10 mL) was refluxed at 105° C. for 4 hours. The reaction was concentrated and dried to give 10.1 g of a sticky, tan solid. Yield: 88%
  • 1H NMR (400 MHz, DMSO) δ 7.76 (brs, 3H), 3.54 (t, 1H), 3.35 (m, 1H), 2.78 (brm, 2H), 1.82 (m, 1H), 1.60-1.30 (brm, 5H)
  • 59C was obtained according to a procedure similar to the one described for 59A, with the following exceptions:
  • Step 59.2: 59.2a was replaced by 59.2c.
  • Step 59.4: 59.4a was replaced by 59.4c.
  • Step 59.5: 59.7a was replaced by 59.7c.
  • Step 59.6: 59.8a was replaced by 59.8c.
  • Step 59.7: 59.9a was replaced by 59.9c.
  • 1H NMR (400 MHz, DMSO d6) δ 9.16 (brs, 2H), 8.14 (d, 2H), 7.90 (brs, 3H), 7.58 (d, 2H), 7.28 (m, 1H), 7.07 (m, 1H), 7.02 (m, 1H), 6.96 (m, 1H), 5.98 (s, 1H), 4.77 (t, 2H), 3.21 (brm, 4H), 2.77 (m, 2H), 2.06 (brm, 6H), 1.61 (m, 2H), 1.36 (m, 2H)
  • Mass Spectral Analysis m/z=431.5 (M+H)+
  • Elemental analysis:
  • C25H30N6O, 2HCl, 2/3H2O
  • Theory: % C, 58.25; % H, 6.52; % N, 16.30; % Cl, 13.76.
  • Found: % C, 58.01; % H, 6.45; % N, 16.24; % Cl, 14.10.
  • Example 59D Preparation of 59.2d
  • A solution of 59.1b (5.00 g, 42.7 mmol, 1.00 eq) in 48% aqueous hydrobromic acid (10 mL) was refluxed at 105° C. for 4 hours. The reaction was concentrated and dried to give 9.27 g of an orange/brown oil. Yield: 83%
  • 1H NMR (400 MHz, DMSO) δ 7.70 (brs, 3H), 3.38 (m, 3H), 2.77 (m, 2H), 1.53 (m, 2H), 1.35 (brm, 5H)
  • 59D was obtained according to a procedure similar to the one described for 59A, with the following exceptions:
  • Step 59.2: 59.2a was replaced by 59.2d.
  • Step 59.4: 59.4a was replaced by 59.4d.
  • Step 59.5: 59.7a was replaced by 59.7d.
  • Step 59.6: 59.8a was replaced by 59.8d.
  • Step 59.7: 59.9a was replaced by 59.9d.
  • 1H NMR (400 MHz, DMSO d6) δ 9.18 (brs, 2H), 8.14 (d, 2H), 7.91 (brs, 3H), 7.58 (d, 2H), 7.28 (m, 1H), 7.07 (dd, 1H), 7.03 (dd, 1H), 6.96 (m, 1H), 5.98 (s, 1H), 4.77 (t, 2H), 3.22 (brm, 4H), 2.75 (brs, 2H), 2.13-1.96 (brm, 6H), 1.55 (m, 2H), 1.34 (brm, 4H)
  • Mass Spectral Analysis m/z=445.5 (M+H)+
  • Elemental analysis:
  • C26H32N6O, 2HCl, 1.25H2O
  • Theory: % C, 57.83; % H, 6.81; % N, 15.56.
  • Found: % C, 57.99; % H, 6.83; % N, 15.63.
  • Example 59E Preparation of 59.10a
  • To a solution of 59.9a (0.400 g, 0.82 mmol, 1.00 eq) and triethylamine (0.342 mL, 2.46 mmol, 3.00 eq) in methylene chloride (20 mL) in an ice/water bath was added acetic anhydride (0.085 mL, 0.900 mmol, 1.10 eq). The reaction mixture was warmed to room temperature, stirred for an additional 6 hours at room temperature and diluted with a 1N solution of hydrochloric acid. The layers were separated and the aqueous phase was extracted with methylene chloride. The organic extracts were combined and concentrated under reduced pressure. The crude residue was purified by column chromatography (eluent: ethyl acetate/hexane mixtures of increasing polarity) to give 400 mg of a white foam. Yield: 91%
  • 1H NMR (400 MHz, DMSO) δ 8.14 (d, 2H), 8.07 (t, 1H), 7.55 (d, 2H), 7.24 (m, 1H), 7.00 (m, 2H), 6.92 (m, 1H), 5.93 (s, 1H), 4.78 (m, 2H), 3.73 (m, 2H), 3.63 (m, 2H), 3.31 (s, 2H), 1.89 (m, 2H), 1.77 (s, 3H), 1.72 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=531.4 (M+H)+
  • Preparation of 59E
  • To a solution of 59.10a (0.390 g, 0.735 mmol, 1.00 eq) in methylene chloride (10 mL) was added a 2.0 M solution of hydrochloric acid in diethyl ether (1.50 mL, 2.90 mmol, 4.00 eq). The reaction mixture was stirred at room temperature for 3 days. The solids were filtered, rinsed with methylene chloride and diethyl ether, and dried to give 335 mg of a white solid. Yield: 98%
  • 1H NMR (400 MHz, DMSO) δ 8.93 (brs, 2H), 8.15 (d, 2H), 8.09 (t, 1H), 7.58 (d, 2H), 7.28 (m, 1H), 7.05 (m, 2H), 6.96 (m, 1H), 6.00 (s, 1H), 4.78 (m, 2H), 3.63 (m, 2H), 3.22 (brm, 4H), 2.05 (brm, 4H), 1.77 (s, 3H)
  • Mass Spectral Analysis m/z=431.5 (M+H)+
  • Elemental analysis:
  • C24H26N6O2, 1HCl, 0.5H2O
  • Theory: % C, 60.56; % H, 5.93; % N, 17.66.
  • Found: % C, 60.39; % H, 5.81; % N, 17.53.
  • Example 59F
  • 59F was obtained according to a procedure similar to the one described for 59E, with the following exceptions:
  • Step 59.8: 59.9a was replaced by 59.9b.
  • Step 59.9: 59.10a was replaced by 59.10b.
  • 1H NMR (400 MHz, DMSO) δ 8.98 (brs, 2H), 8.14 (d, 2H), 8.01 (brt, 1H), 7.57 (d, 2H), 7.28 (m, 1H), 7.05 (m, 2H), 6.96 (m, 1H), 6.00 (s, 1H), 4.77 (t, 2H), 3.22 (brm, 4H), 3.13 (m, 2H), 2.16-1.97 (brm, 6H), 1.80 (s, 3H)
  • Mass Spectral Analysis m/z=445.5 (M+H)+
  • Elemental analysis:
  • C25H28N6O2, 1HCl, 1.5H2O
  • Theory: % C, 59.11; % H, 6.35; % N, 16.54.
  • Found: % C, 59.46; % H, 6.27; % N, 16.60.
  • Example 59G
  • 59G was obtained according to a procedure similar to the one described for 59E, with the following exceptions:
  • Step 59.8: 59.9a was replaced by 59.9c.
  • Step 59.9: 59.10a was replaced by 59.10c.
  • 1H NMR (400 MHz, DMSO) δ 8.88 (brs, 2H), 8.14 (d, 2H), 7.82 (brt, 1H), 7.58 (d, 2H), 7.28 (m, 1H), 7.07 (dd, 1H), 7.03 (dd, 1H), 6.96 (m, 1H), 5.99 (s, 1H), 4.75 (t, 2H), 3.22 (brm, 4H), 3.00 (q, 2H), 2.10 (m, 2H), 1.99 (brm, 4H), 1.76 (s, 3H), 1.43 (m, 2H), 1.29 (m, 2H)
  • Mass Spectral Analysis m/z=473.5 (M+H)+
  • Elemental analysis:
  • C27H32N6O2, 1HCl, 2/3H2O
  • Theory: % C, 62.24; % H, 6.64; % N, 16.13.
  • Found: % C, 61.92; % H, 6.65; % N, 15.91.
  • Example 59H
  • 59H was obtained according to a procedure similar to the one described for 59E, with the following exceptions:
  • Step 59.8: 59.9a was replaced by 59.9d.
  • Step 59.9: 59.10a was replaced by 59.10d.
  • 1H NMR (400 MHz, DMSO) δ 8.99 (brs, 2H), 8.14 (d, 2H), 7.80 (brt, 1H), 7.57 (d, 2H), 7.28 (m, 1H), 7.07 (dd, 1H), 7.03 (dd, 1H), 6.96 (m, 1H), 5.99 (s, 1H), 4.75 (t, 2H), 3.22 (brm, 4H), 2.99 (q, 2H), 2.12-1.94 (brm, 6H), 1.77 (s, 3H), 1.34 (brm, 6H)
  • Mass Spectral Analysis m/z=487.5 (M+H)+
  • Elemental analysis:
  • C28H34N6O2, 1HCl, 1H2O
  • Theory: % C, 62.15; % H, 6.89; % N, 15.53.
  • Found: % C, 62.27; % H, 6.83; % N, 15.48.
  • Example 59I Preparation of 59.11a
  • To a solution of 59.9a (0.250 g, 0.512 mmol, 1.00 eq) and triethylamine (0.214 mL, 1.54 mmol, 3.00 eq) in methylene chloride (20 mL) cooled in an ice/water bath was added methanesulfonyl chloride (7.4) (0.044 mL, 0.563 mmol, 1.10 eq). The reaction was stirred at 0° C. for 30 minutes and diluted with water. The layers were separated and the aqueous was extracted with methylene chloride. The organic extracts were combined and concentrated under reduced pressure. The crude residue was purified by column chromatography (eluent: ethyl acetate/hexane mixtures of increasing polarity) to give 164 mg of a white foam. Yield: 57%
  • 1H NMR (400 MHz, DMSO) δ 8.14 (d, 2H), 7.56 (d, 2H), 7.38 (brs, 1H), 7.23 (m, 1H), 7.00 (m, 2H), 6.92 (m, 1H), 5.93 (s, 1H), 4.85 (t, 2H), 3.73 (m, 2H), 3.61 (m, 2H), 3.32 (brs, 2H), 2.91 (s, 3H), 1.89 (m, 2H), 1.73 (m, 2H), 1.42 (s, 9H)
  • Mass Spectral Analysis m/z=565.6 (M−H)
  • Preparation of 59I
  • To a solution of 59.11a (0.155 g, 0.274 mmol, 1.00 eq) in methylene chloride (10 mL) was added a 2.0 M solution of hydrochloric acid in diethyl ether (0.55 mL, 1.10 mmol, 4.00 eq). The reaction was stirred at room temperature for 16 hours. The solids were filtered, rinsed with methylene chloride and diethyl ether, and dried to give 83 mg of a white solid. Yield: 58%
  • 1H NMR (400 MHz, DMSO) δ 9.04 (brs, 2H), 8.15 (d, 2H), 7.58 (d, 2H), 7.41 (t, 1H), 7.28 (m, 1H), 7.05 (m, 2H), 6.96 (m, 1H), 6.00 (s, 1H), 4.85 (t, 2H), 3.62 (m, 2H), 3.23 (brm, 4H), 2.92 (s, 3H), 2.07 (brm, 4H)
  • Mass Spectral Analysis m/z=467.3 (M+H)+
  • Elemental analysis:
  • C23H26N6O3S, 1HCl, 0.5H2O
  • Theory: % C, 53.95; % H, 5.51; % N, 16.41.
  • Found: % C, 54.00; % H, 5.39; % N, 16.10.
  • Example 59J
  • 59J was obtained according to a procedure similar to the one described for 59I with the following exceptions:
  • Step 59.10: 59.9a was replaced by 59.9b.
  • Step 59.11: 59.11a was replaced by 59.11b.
  • 1H NMR (400 MHz, DMSO) δ 8.90 (brs, 2H), 8.15 (d, 2H), 7.57 (d, 2H), 7.28 (m, 1H), 7.21 (t, 1H), 7.05 (m, 2H), 6.96 (m, 1H), 6.00 (s, 1H), 4.83 (t, 2H), 3.22 (brm, 4H), 3.06 (q, 2H), 2.92 (s, 3H), 2.20 (m, 2H), 2.11 (brm, 2H), 2.01 (brm, 2H)
  • Mass Spectral Analysis m/z=481.5 (M+H)+
  • Elemental analysis:
  • C24H28N6O3S, 1HCl, 0.5H2O
  • Theory: % C, 54.80; % H, 5.75; % N, 15.98.
  • Found: % C, 54.96; % H, 5.64; % N, 15.67.
  • Example 59K
  • 59K was obtained according to a procedure similar to the one described for 59I, with the following exceptions:
  • Step 59.10: 59.9a was replaced by 59.9c.
  • Step 59.11: 59.11a was replaced by 59.11c.
  • 1H NMR (400 MHz, DMSO) δ 8.13 (d, 2H), 7.54 (d, 2H), 7.22 (m, 1H), 6.98 (m, 3H), 6.90 (m, 1H), 5.92 (s, 1H), 4.76 (t, 2H), 2.92 (brm, 4H), 2.86 (s, 3H), 2.78 (m, 2H), 1.99 (m, 2H), 1.82 (m, 2H), 1.73 (m, 2H), 1.51 (m, 2H), 1.33 (m, 2H)
  • Mass Spectral Analysis m/z=509.5 (M+H)+
  • Elemental analysis:
  • C26H32N6O3S, 1.45H2O
  • Theory: % C, 58.40; % H, 6.58; % N, 15.71.
  • Found: % C, 58.79; % H, 6.58; % N, 15.31.
  • Example 59L
  • 59L was obtained according to a procedure similar to the one described for 59I, with the following exceptions:
  • Step 59.10: 59.9a was replaced by 59.9d.
  • Step 59.11: 59.11a was replaced by 59.11d.
  • 1H NMR (400 MHz, DMSO) δ 8.92 (brs, 2H), 8.14 (d, 2H), 7.57 (d, 2H), 7.28 (m, 1H), 7.07 (dd, 1H), 7.03 (dd, 1H), 6.96 (m, 2H), 5.99 (s, 1H), 4.76 (t, 2H), 3.22 (brm, 4H), 2.91 (q, 2H), 2.86 (s, 3H), 2.05 (brm, 6H), 1.39 (brm, 6H)
  • Mass Spectral Analysis m/z=523.6 (M+H)+
  • Elemental analysis:
  • C27H34N6O3S, 1HCl, 0.5H2O
  • Theory: % C, 57.08; % H, 6.39; % N, 14.79.
  • Found: % C, 57.36; % H, 6.34; % N, 14.81.
  • Example 60A Preparation of 60.2
  • To a solution of 60.1 (5.00 g, 56.1 mmol, 1.00 eq) and triethylamine (15.6 mL, 112 mmol, 2.00 eq) in methanol (100 mL) cooled in an ice/water bath was added 59.3 (7.34 mL, 61.7 mmol, 1.10 eq). The reaction mixture was stirred at 0° C. for 1.5 hours and then concentrated. The mixture was dissolved in methylene chloride and washed with a 0.5N solution of hydrochloric acid. The aqueous phase was extracted three times with 5% methanol/methylene chloride. The organic extracts were combined, concentrated, and dried to give 6.96 g of an orange oil. Yield: 67%
  • 1H NMR (400 MHz, CDCl3) δ 3.72 (t, 2H), 3.41 (q, 2H), 1.94 (s, 1H), 1.70 (m, 4H)
  • Preparation of 60.3
  • To a solution of 60.2 (3.69 g, 19.9 mmol, 1.00 eq) and triethylamine (5.56 mL, 39.9 mmol, 2.00 eq) in methylene chloride (100 mL) cooled in an ice/water bath was added 7.4 (2.31 mL, 29.9 mmol, 1.50 eq). The reaction mixture was warmed to room temperature, stirred for an additional 1 hour at room temperature and then diluted with water. The layers were separated and the aqueous phase was extracted with methylene chloride. The organic extracts were combined and washed with a 0.5M solution of hydrochloric acid. The organic extracts were concentrated and dried to give 4.96 g of an orange oil. Yield: 95%
  • 1H NMR (400 MHz, DMSO) δ 9.46 (brm, 1H), 4.21 (t, 2H), 3.22 (q, 2H), 3.16 (s, 3H), 1.62 (m, 4H)
  • Mass Spectral Analysis m/z=262.6 (M−H)−60A was obtained according to a procedure similar to the one described for 59A, with the following exceptions:
  • Step 60.3: 59.4a from Step 59.4 was replaced by 60.3.
  • Step 60.4: 59.7a from Step 59.5 was replaced by 60.4.
  • Step 60.5: 59.8a from Step 59.6 was replaced by 60.5.
  • Step 60.6: 59.9a from Step 59.7 was replaced by 60.6.
  • 1H NMR (400 MHz, DMSO d6) δ 9.22 (brs, 2H), 8.14 (d, 2H), 7.97 (brs, 3H), 7.57 (d, 2H), 7.28 (m, 1H), 7.08 (dd, 1H), 7.02 (dd, 1H), 6.96 (m, 1H), 5.99 (s, 1H), 4.82 (t, 2H), 3.21 (brm, 4H), 2.83 (t, 2H), 2.06 (brm, 6H), 1.61 (m, 2H)
  • Mass Spectral Analysis m/z=417.5 (M+H)+
  • Elemental analysis:
  • C24H28N6O, 2HCl, 3/2H2O
  • Theory: % C, 55.81; % H, 6.44; % N, 16.27; % Cl, 13.73.
  • Found: % C, 55.95; % H, 6.48; % N, 16.28; % Cl, 14.00.
  • Example 60B
  • 60B was obtained according to a procedure similar to the one described for 59E, with the following exceptions:
  • Step 60.7: 59.9a from Step 59.8 was replaced by 60.6.
  • Step 60.8: 59.10a from Step 59.9 was replaced by 60.7.
  • 1H NMR (400 MHz, DMSO d6) δ 9.09 (brs, 2H), 8.14 (d, 2H), 7.90 (brt, 1H), 7.57 (d, 2H), 7.28 (m, 1H), 7.07 (dd, 1H), 7.03 (dd, 1H), 6.96 (m, 1H), 5.99 (s, 1H), 4.78 (t, 2H), 3.22 (brm, 4H), 3.07 (q, 2H), 2.04 (brm, 6H), 1.78 (s, 3H), 1.42 (m, 2H)
  • Mass Spectral Analysis m/z=459.5 (M+H)+
  • Elemental analysis:
  • C26H30N6O, 1HCl, 3/2H2O
  • Theory: % C, 59.82; % H, 6.56; % N, 16.10.
  • Found: % C, 59.77; % H, 6.31; % N, 16.05.
  • Example 60C
  • 60C was obtained according to a procedure similar to the one described for 59I, with the following exceptions:
  • Step 60.9: 59.9a from Step 59.10 was replaced by 60.6.
  • Step 60.10: 59.11a from Step 59.11 was replaced by 60.8.
  • 1H NMR (400 MHz, DMSO d6) δ 8.13 (d, 2H), 7.54 (d, 2H), 7.23 (m, 1H), 7.01 (m, 3H), 6.91 (m, 1H), 5.93 (s, 1H), 4.78 (t, 2H), 2.97 (m, 4H), 2.88 (s, 3H), 2.84 (m, 2H), 2.03 (m, 2H), 1.86 (m, 2H), 1.76 (m, 2H), 1.50 (m, 2H)
  • Mass Spectral Analysis m/z=495.5 (M+H)+
  • Elemental analysis:
  • C25H30N6O3S, 1H2O
  • Theory: % C, 58.58; % H, 6.29; % N, 16.39.
  • Found: % C, 58.78; % H, 5.94; % N, 16.40.
  • Example 61A, 61B Preparation of 61.1
  • To a stirred solution of 49.9 (1.47 g, 2.65 mmol, 1 eq) in methanol (80 mL) was added 10% Pd/C (294 mg). The reaction mixture was stirred under a hydrogen atmosphere using a hydrogen balloon overnight. The reaction mixture was then filtered, the catalyst was washed with methanol, and the filtrate was concentrated in vacuo. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: ˜100%.
  • Preparation of 61.2a, 61.2b
  • Compound 61.1 (1.47 g, 2.64 mmol) was subjected to chiral separation to yield two enantiomeric pure isomers 61.2a (600 mg, 40.8%) and 61.2b (550 mg, 37.4%).
  • HPLC conditions:
  • Column: Chiralcel AD 2×15 cm
  • Flow: 20 mL/min, 75% A, 25% B
  • Detection: UV 254 nm
  • Mobile phase A: hexane
  • Mobile phase B: 60 EtOH/40 MeOH
  • 61.2a: 1H NMR (400 MHz, CDCl3) δ 7.16 (s, 1H), 7.08 (m, 1H), 6.99 (d, 1H), 6.80 (m, 2H), 6.46 (dd, 1H), 5.12 (brs, 2H), 4.58 (m, 1H), 3.89 (m, 2H), 3.53 (m, 2H), 3.38-3.10 (m, 7H), 1.98 (m, 3H), 1.78 (m, 1H), 1.67-1.52 (m, 2H), 1.48 (s, 9H), 1.27 (m, 3H), 1.18 (m, 3H).
  • [α]25 D −62.10° (c=1.34, MeOH)
  • 61.2b: 1H NMR (400 MHz, CDCl3) δ 7.16 (s, 1H), 7.08 (m, 1H), 6.99 (d, 1H), 6.80 (m, 2H), 6.46 (dd, 1H), 5.12 (brs, 2H), 4.58 (m, 1H), 3.89 (m, 2H), 3.53 (m, 2H), 3.38-3.10 (m, 7H), 1.98 (m, 3H), 1.78 (m, 1H), 1.67-1.52 (m, 2H), 1.48 (s, 9H), 1.27 (m, 3H), 1.18 (m, 3H).
  • [α]25 D +62.82° (c=1.19, MeOH)
  • Preparation of 61A
  • To a solution of pure enantiomer 61.2a (590 mg, 1.06 mmol) in methanol (30 mL) was added 4.0 M hydrochloric acid in dioxane (2.65 mL, 10.6 mmol, 10 eq). The mixture was stirred at ambient temperature for 24 hours, An additional amount of a 4.0 M hydrochloric acid in dioxane (1.0 mL, 4 mmol, 3.8 eq) was added to the reaction mixture, which was stirred for another 3 days at room temperature. The solvent was evaporated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride-methanol, 10:1). Yield: 77.5%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.90 (brs, 1H), 8.93 (brs, 1H), 8.78 (brs, 1H), 7.02-6.86 (m, 4H), 6.73 (d, 1H), 6.40 (dd, 1H), 4.49 (m, 1H), 3.39-3.18 (m, 7H), 2.98 (m, 1H), 2.05-1.82 (m, 6H), 1.10 (m, 6H).
  • [α]25 D −47.31° (c=0.96, MeOH)
  • Mass Spectral Analysis m/z=413.91 (M+H)+
  • Elemental analysis:
  • C24H29FN2O3, 1HCl, 9/10H2O
  • Theory: % C, 61.97; % H, 6.89; % N, 6.02.
  • Found: % C, 61.89; % H, 6.72; % N, 5.95.
  • Preparation of 61B
  • To a solution of pure enantiomer 61.2b (550 mg, 0.988 mmol) in methanol (30 mL) was added 4.0 M hydrochloric acid in dioxane (2.47 mL, 9.88 mmol, 10 eq). The mixture was stirred at ambient temperature for 24 hours. An additional amount of a 4.0 M hydrochloric acid in dioxane (1.0 mL, 4 mmol, 4 eq) was added to the reaction mixture, which was stirred for another 3 days at room temperature. The solvent was evaporated in vacuo. The residue was purified by column chromatography (eluent: methylene chloride-methanol, 10:1). Yield: 86%
  • 1H NMR (400 MHz, DMSO-d6) δ 9.94 (brs, 1H), 9.08 (brs, 1H), 8.90 (brs, 1H), 7.0-6.86 (m, 4H), 6.72 (d, 1H), 6.40 (dd, 1H), 4.49 (m, 1H), 3.39-3.16 (m, 7H), 2.98 (m, 1H), 2.03-1.82 (m, 6H), 1.10 (m, 6H).
  • [α]25 D +46.78° (c=1.17, MeOH)
  • Mass Spectral Analysis m/z=413.88 (M+H)+
  • Elemental analysis:
  • C24H29FN2O3, 1HCl, 13/10H2O
  • Theory: % C, 61.02; % H, 6.96; % N, 5.93.
  • Found: % C, 61.00; % H, 6.78; % N, 5.88.
  • Preparation of 61.4
  • O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium tetrafluoroborate (0.41 g, 1.28 mmol, 1.1 eq) was added at 0° C. to a solution of 21C (0.50 g, 1.28 mmol, 1.1 eq), 61.3 (0.29 g, 1.16 mmol, 1.0 eq) and diisopropylethylamine (0.46 mL, 2.56 mmol, 2.2 eq) in acetonitrile (4 mL). The reaction mixture was warmed to room temperature and stirred for an additional 2 days at room temperature. The reaction mixture was concentrated under reduced pressure, then re-dissolved in ethyl acetate. The solution was washed with a saturated aqueous solution of sodium hydrogen carbonate (50 mL) and brine (50 mL) then dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography (eluent: hexane/ethyl acetate mixtures of increasing polarity). Yield: 95%
  • 1H NMR (400 MHz, DMSO d6) δ 8.28 (s, 1H), 8.07 (d, 1H), 7.99 (m, 2H), 7.70 (m, 1H), 7.62 (m, 1H), 7.42 (m, 2H), 7.38 (d, 1H), 7.31 (d, 1H), 7.21 (m, 1H), 6.98 (m, 1H), 6.91 (m, 1H), 6.88 (m, 1H), 5.89 (d, 1H), 3.93 (m, 1H), 3.72 (m, 1H), 3.55 (m, 2H), 3.45 (m, 2H), 3.24 (m, 2H), 2.22 (m, 1H), 2.07 (m, 2H), 1.85 (m, 2H), 1.63 (m, 1H), 1.13 (m, 6H)
  • Mass Spectral Analysis m/z=642.13 (M+H)+
  • X-Ray Crystallography:
  • Single crystals were grown as plates by dissolving 61.4 (50 mg, 0.008 mmol, 1 eq) in an aqueous methanolic solution (MeOH/water=80:20) and letting sit still at room temperature for 9 months.
  • Crystal data and structure refinement for 61.4:
  • Empirical formula: C36 H35 Br N2 O3
  • Formula weight: 623.57
  • Temperature: 120(2) K
  • Wavelength: 0.71073 A
  • Crystal system, space group: Monoclinic, P2(1)
  • Unit cell dimensions:
  • a=7.435(3) A; alpha=90 deg.
  • b=14.851(6) A; beta=93.695(6) deg.
  • c=13.628(5) A; gamma=90 deg.
  • Volume: 1501.7(10) A3
  • Z, Calculated density: 2, 1.379 Mg/m3
  • Absorption coefficient: 1.408 mm−1
  • F(000): 648
  • Crystal size: 0.22×0.10×0.04 mm
  • Theta range for data collection: 2.03 to 28.27 deg.
  • Limiting indices: −9<=h<=9, −19<=k<=18, −17<=l<=17
  • Reflections collected/unique: 16930/6798 [R(int)=0.0287]
  • Completeness to theta=28.27: 94.1%
  • Absorption correction: Semi-empirical from equivalents
  • Max. and min transmission: 0.9458 and 0.7470
  • Refinement method: Full-matrix least-squares on F2
  • Data/restraints/parameters: 6798/1/381
  • Goodness-of-fit on F2-1.034
  • Final R indices [I>2sigma(I)]: R1=0.0340, wR2=0.0846
  • R indices (all data): R1=0.0354, wR2=0.0857
  • Absolute structure parameter: −0.002(5)
  • Largest diff. peak and hole: 0.792 and −0.236 e.A−3
  • Biological Methods
  • In Vitro Assays
  • The potencies of the compounds listed in Table 2 were determined by testing the ability of a range of concentrations of each compound to inhibit the binding of the non-selective opioid antagonist, [3H]diprenorphine, to the cloned human μ, κ, and δ opioid receptors, expressed in separate cell lines. IC50 values were obtained by nonlinear analysis of the data using GraphPad Prism version 3.00 for Windows (GraphPad Software, San Diego). Ki values were obtained by Cheng-Prusoff corrections of IC50 values.
  • Receptor Binding
  • The receptor binding method (DeHaven and DeHaven-Hudkins, 1998) was a modification of the method of Raynor et al. (1994). After dilution in buffer A and homogenization as before, membrane proteins (10-80 μg) in 250 μL were added to mixtures containing test compound and [3H]diprenorphine (0.5 to 1.0 nM, 40,000 to 50,000 dpm) in 250 μL of buffer A in 96-well deep-well polystyrene titer plates (Beckman). After incubation at room temperature for one hour, the samples were filtered through GF/B filters that had been presoaked in a solution of 0.5% (w/v) polyethylenimine and 0.1% (w/v) bovine serum albumin in water. The filters were rinsed 4 times with 1 mL of cold 50 mM Tris HCl, pH 7.8 and radioactivity remaining on the filters determined by scintillation spectroscopy. Nonspecific binding was determined by the minimum values of the titration curves and was confirmed by separate assay wells containing 10 μM naloxone. Ki values were determined by Cheng-Prusoff corrections of IC50 values derived from nonlinear regression fits of 12 point titration curves using GraphPad Prism® version 3.00 for Windows (GraphPad Software, San Diego, Calif.).
  • To determine the equilibrium dissociation constant for the inhibitors (Ki), radioligand bound (cpm) in the presence of various concentrations of test compounds was measured. The concentration to give half-maximal inhibition (EC50) of radioligand binding was determined from a best nonlinear regression fit to the following equation,
  • Y = Bottom + ( Top - Bottom ) 1 + 10 X - LogEC 50
  • where Y is the amount of radioligand bound at each concentration of test compound, Bottom is the calculated amount of radioligand bound in the presence of an infinite concentration of test compound, Top is the calculated amount of radioligand bound in the absence of test compound, X is the logarithm of the concentration of test compound, and LogEC50 is the log of the concentration of test compound where the amount of radioligand bound is half-way between Top and Bottom. The nonlinear regression fit was performed using the program Prism® (GraphPad Software, San Diego, Calif.). The Ki values were then determined from the EC50 values by the following equation,
  • K i = EC 50 1 + [ ligand ] K d
  • where [ligand] is the concentration of radioligand and Kd is the equilibrium dissociation constant for the radioligand.
  • Receptor-mediated [35S]GTPγS binding
  • The potency and efficacy of compounds at each of the receptors are assessed by modifications of the methods of Selley et al., 1997 and Traynor and Nahorski, 1995 using receptor-mediated [35S]GTPγS binding in the same membrane preparations used to measure receptor binding. Assays are carried out in 96-well FlashPlates® (Perkin Elmer Life Sciences, Inc, Boston, Mass.). Membranes prepared from CHO cells expressing the appropriate receptor (50-100 μg of protein) are added to assay mixtures containing agonist with or without antagonists, 100 μM [35S]GTPγS (approx. 100,000 dpm), 3.0 μM GDP, 75 mM NaCl, 15 mM MgCl2, 1.0 mM ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetracetic acid, 1.1 mM dithiothreitol, 10 μg/mL leupeptin, 10 μg/mL pepstatin A, 200 μg/mL bacitracin, and 0.5 μg/mL aprotinin in 50 mM Tris-HCl buffer, pH 7.8. After incubation at room temperature for one hour, the plates are sealed, centrifuged at 800×g in a swinging bucket rotor for 5 mM and bound radioactivity determined with a TopCount microplate scintillation counter (Packard Instrument Co., Meriden, Conn.).
  • EC50 values for agonists are determined from nonlinear regression fits of 8- or 12-point titration curves to the 4-parameter equation for a sigmoidal dose-response with a slope factor of 1.0 using GraphPad Prism® version 3.00 for Windows (GraphPad Software, San Diego, Calif.).
  • To determine IC50 values, the concentrations to give half-maximal inhibition of agonist-stimulated [35S]GTPγS binding, the amount of [35S]GTPγS bound in the presence of a fixed concentration of agonist and various concentrations of antagonist was measured. The fixed concentration of agonist was the EC80, the concentration to give 80% of the relative maximum stimulation of [35S]GTPγS binding. The agonists loperamide (100 nM), U50,488 (50 nM), and BW373U86 (2.0 nM) were used to stimulate [35S]GTPγS binding via the μ, δ, and κ opioid receptors, respectively. The IC50 value was determined from a best nonlinear regression fit of the data to the 4-parameter equation for a sigmoidal dose-response with a slope factor of 1.0 using GraphPad Prism® version 3.00 for Windows.
  • The potencies of the compounds were determined by testing the ability of a range of concentrations of each compound to inhibit the binding of the non-selective opioid antagonist, [3H]diprenorphine, to the cloned human μ, κ, and δ opioid receptors, expressed in separate cell lines. All the compounds tested (compounds included in Table 2) bind with affinity to the human cloned 8 opioid receptor less than 2 μM (Ki values). These compounds display high selectivity δ/κ and δ/μ (at least 10-fold). The potencies of the agonists were assessed by their abilities to stimulate [35S]GTPγS binding to membranes containing the cloned human 8 opioid receptors. All the compounds listed in Table 2 were shown to be agonists at the 8 opioid receptor.
  • As example, 33M (Table 2) binds to the delta, mu, and kappa opioid receptors with affinity (expressed as Ki value) of 9.7 nM, >1000 nM and >1000 nM, respectively. Furthermore, 33M displayed in vitro agonist activity (EC50=287 nM).
  • As further example, 43D (Table 2) binds to the delta, mu, and kappa opioid receptors with affinity (expressed as Ki value) of 2.5 nM, >1000 nM and >1000 nM, respectively. Furthermore, 43D displayed potent in vitro agonist activity (EC50=63 nM).
  • Example 61A (Table 3) and 61B (enantiomeric analog of Example 61A) bind to the 8 opioid receptor with affinity (expressed as Ki value) of 0.59 nM, and 75 nM, respectively. Furthermore, Example 61A displayed potent in vitro δ agonist activity (EC50=16.8 nM), whereas Example 61B displayed weaker in vitro δ agonist activity (EC50=1282 nM), when compared to Example 61A.
  • In Vivo Assay
  • Freunds Complete Adjuvant (FCA)-Induced Hyperalgesia
  • Rats were injected intraplantar with FCA and 24 h later treated with tested compounds administered orally. Paw Pressure Thresholds (PPT) was assessed 60 minutes after drug treatment. In this assay, 43D produced significant anti-hyperalgesic activity (193±47% antihyperalgesia) after oral administration (3 mg/kg dose).
  • The disclosures of each patent, patent application and publication cited or described in this document are hereby incorporated herein by reference, in their entirety.
  • Those skilled in the art will appreciate that numerous changes and modifications can be made to the preferred embodiments of the invention and that such changes and modifications can be made without departing from the spirit of the invention. It is, therefore, intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention.

Claims (56)

1. A pharmaceutical composition, comprising:
a pharmaceutically acceptable carrier; and a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00713
wherein:
D is:
Figure US20110218186A1-20110908-C00714
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
2. A pharmaceutical composition according to claim 1, wherein the compound of formula XXVIII has the following structure:
Figure US20110218186A1-20110908-C00715
3. A pharmaceutical composition according to claim 1, further comprising an opioid, an agent for the treatment of neuralgia/neuropathic pain, an agent for the treatment of depression, an agent for the treatment of incontinence, or an anti-Parkinson's agent.
4. A pharmaceutical composition according to claim 3, wherein said opioid is alfentanil, allylprodine, alphaprodine, anileridine, benzyl-morphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioaphetylbutyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacylmorphan, lofentanil, loperamide, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbuphine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, normorphine, norpinanone, opium, oxycodone, oxymorphone, papaveretum, pentazocine, phenadoxone, phenomorphan, phanazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propiram, propoxyphene, sulfentanil, tilidine, tramadol, a diastereoisomer thereof, a pharmaceutically acceptable salt thereof, a complex thereof, or a mixture thereof.
5. A pharmaceutical composition according to claim 3, wherein said agent for the treatment of neuralgia/neuropathic pain is a mild OTC analgesic, a narcotic analgesic, an anti-seizure medication or an anti-depressant.
6. A pharmaceutical composition according to claim 3, wherein said agent for the treatment of depression is a selective serotonin re-uptake inhibitor, a tricyclic compound, a monoamine oxidase inhibitor, or an antidepressant compound belonging to the heterocyclic class.
7. A pharmaceutical composition according to claim 3, wherein said agent for the treatment of urge incontinence is an anticholinergic agent, an antispasmodic medication, a tricyclic antidepressant, a calcium channel blocker or a beta agonist.
8. A pharmaceutical composition according to claim 3, further comprising:
an antibiotic, antiviral, antifungal, anti-inflammatory, anesthetic, or mixture thereof.
9. A method of binding opioid receptors in a patient in need thereof, comprising the step of:
administering to said patient an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00716
wherein:
D is:
Figure US20110218186A1-20110908-C00717
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
10. A method according to claim 9, wherein said compound binds δ opioid receptors.
11. A method according to claim 10, wherein said δ opioid receptors are located in the central nervous system.
12. A method according to claim 10, wherein said δ opioid receptors are located peripherally to the central nervous system.
13. A method according to claim 9, wherein said binding modulates the activity of said opioid receptors.
14. A method according to claim 13, wherein said binding agonizes the activity of said opioid receptors.
15. A method according to claim 12, wherein said compound does not substantially cross the blood-brain bather.
16. A method of preventing, inhibiting or treating a condition, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00718
wherein:
D is:
Figure US20110218186A1-20110908-C00719
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
said condition selected from any one of the group consisting of: pain, gastrointestinal dysfunction, a urogenital disorder, an immunomodulatory disorder, an inflammatory disorder, a mood disorder, anxiety, a respiratory function disorder, a stress-related disorder, attention deficit hyperactivity disorder, a sympathetic nervous system disorder, tussis, a motor disorder, stroke, cardiac arrhythmia, glaucoma, sexual dysfunction, and substance addiction.
17. A method of preventing, inhibiting or treating a condition, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00720
wherein:
D is:
Figure US20110218186A1-20110908-C00721
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof;
said condition selected from any one of the group consisting of: shock, brain edema, cerebral ischemia, cerebral deficits subsequent to cardiac bypass surgery and grafting, systemic lupus erythematosus, Hodgkin's disease, Sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts.
18. A method of improving organ and cell survival, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00722
wherein:
D is:
Figure US20110218186A1-20110908-C00723
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
19. A method of preventing, inhibiting or treating pain, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00724
wherein:
D is:
Figure US20110218186A1-20110908-C00725
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
20. A method of preventing, inhibiting or treating a urogenital disorder, comprising the step of:
administering to a patient in need thereof an effective amount a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00726
wherein:
D is:
Figure US20110218186A1-20110908-C00727
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
21. A method of preventing, inhibiting or treating an inflammatory disorder, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00728
wherein:
D is:
Figure US20110218186A1-20110908-C00729
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
22. A method of preventing, inhibiting or treating a mood disorder, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00730
wherein:
D is:
Figure US20110218186A1-20110908-C00731
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
23. A method of preventing, inhibiting or treating anxiety, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00732
wherein:
D is:
Figure US20110218186A1-20110908-C00733
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
24. A method of preventing, inhibiting or treating a motor dysfunction, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00734
wherein:
D is:
Figure US20110218186A1-20110908-C00735
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
25. A method according to claim 24, wherein the motor dysfunction is Parkinson's disease.
26. A method according to claim 24, wherein the motor dysfunction is tremors, Tourette's syndrome, or dyskinesia.
27. A method of preventing, inhibiting or treating cardiac arrhythmia, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00736
wherein:
D is:
Figure US20110218186A1-20110908-C00737
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
28. A method of preventing, inhibiting or treating sexual dysfunction, comprising the step of:
administering to a patient in need thereof an effective amount of a compound according to formula XXVIII:
Figure US20110218186A1-20110908-C00738
wherein:
D is:
Figure US20110218186A1-20110908-C00739
K is carboxy (—COOH), —C(═O)—O-alkyl, —S(═O)2—N(alkyl)(alkyl), heteroaryl, alkylheteroaryl, aminocarbonyl (—C(═O)—NH2), or N-alkylaminocarbonyl (—C(═O)—NH(alkyl));
R23, R24, and R26 are each independently H or alkyl;
p is 1;
A2 and B2 are each H, or together form a double bond; and
X2 is —CH2— or —O—;
or a stereoisomer, prodrug, pharmaceutically acceptable salt, hydrate, solvate, acid salt hydrate, or N-oxide thereof.
29. A method according to claim 19 wherein the pain is selected from the group consisting of acute pain and chronic pain.
30. A method according to claim 29 wherein the pain is acute pain.
31. A method according to claim 29 wherein the pain is chronic pain.
32. A method according to claim 19 wherein the pain is selected from the group consisting of nociceptive pain, inflammatory pain, visceral pain, somatic pain, neuralgia, neuropathic pain, AIDS pain, cancer pain, phantom pain, psychogenic pain, pain resulting from hyperalgesia, pain caused by rheumatoid arthritis, migraine and allodynia.
33. A method according to claim 32 wherein the pain is neuropathic pain.
34. A method according to claim 32 wherein the pain is migraine.
35. A method according to claim 9, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00740
36. A method according to claim 16, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00741
37. A method according to claim 17, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00742
38. A method according to claim 18, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00743
39. A method according to claim 19, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00744
40. A method according to claim 20, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00745
41. A method according to claim 21, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00746
42. A method according to claim 22, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00747
43. A method according to claim 23, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00748
44. A method according to claim 24, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00749
45. A method according to claim 25, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00750
46. A method according to claim 28, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00751
47. A method according to claim 29, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00752
48. A method according to claim 30, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00753
49. A method according to claim 31, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00754
50. A method according to claim 32, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00755
51. A method according to claim 33, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00756
52. A method according to claim 34, wherein said compound has the following structure:
Figure US20110218186A1-20110908-C00757
53. A pharmaceutical composition according to claim 1, wherein the compound of formula XXVIII is selected from the group consisting of:
Figure US20110218186A1-20110908-C00758
Figure US20110218186A1-20110908-C00759
Figure US20110218186A1-20110908-C00760
54. A pharmaceutical composition according to claim 53, wherein the compound of formula XXVIII is selected from the group consisting of:
Figure US20110218186A1-20110908-C00761
55. A method according to claim 9, wherein the compound of formula XXVIII is selected from the group consisting of:
Figure US20110218186A1-20110908-C00762
Figure US20110218186A1-20110908-C00763
Figure US20110218186A1-20110908-C00764
56. A method according to claim 55, wherein the compound of formula XXVIII is selected from the group consisting of:
Figure US20110218186A1-20110908-C00765
US13/097,940 2005-03-31 2011-04-29 Spirocyclic heterocyclic derivatives and methods of their use Abandoned US20110218186A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/097,940 US20110218186A1 (en) 2005-03-31 2011-04-29 Spirocyclic heterocyclic derivatives and methods of their use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US66717705P 2005-03-31 2005-03-31
US11/393,133 US7598261B2 (en) 2005-03-31 2006-03-30 Spirocyclic heterocyclic derivatives and methods of their use
US12/542,302 US8022060B2 (en) 2005-03-31 2009-08-17 Spirocyclic heterocyclic derivatives and methods of their use
US13/097,940 US20110218186A1 (en) 2005-03-31 2011-04-29 Spirocyclic heterocyclic derivatives and methods of their use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/542,302 Division US8022060B2 (en) 2005-03-31 2009-08-17 Spirocyclic heterocyclic derivatives and methods of their use

Publications (1)

Publication Number Publication Date
US20110218186A1 true US20110218186A1 (en) 2011-09-08

Family

ID=37054196

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/393,133 Expired - Fee Related US7598261B2 (en) 2005-03-31 2006-03-30 Spirocyclic heterocyclic derivatives and methods of their use
US12/542,302 Expired - Fee Related US8022060B2 (en) 2005-03-31 2009-08-17 Spirocyclic heterocyclic derivatives and methods of their use
US13/097,940 Abandoned US20110218186A1 (en) 2005-03-31 2011-04-29 Spirocyclic heterocyclic derivatives and methods of their use

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/393,133 Expired - Fee Related US7598261B2 (en) 2005-03-31 2006-03-30 Spirocyclic heterocyclic derivatives and methods of their use
US12/542,302 Expired - Fee Related US8022060B2 (en) 2005-03-31 2009-08-17 Spirocyclic heterocyclic derivatives and methods of their use

Country Status (11)

Country Link
US (3) US7598261B2 (en)
EP (1) EP1871761A4 (en)
JP (1) JP5118788B2 (en)
KR (1) KR20080005245A (en)
AU (1) AU2006230517B2 (en)
BR (1) BRPI0612189A2 (en)
CA (1) CA2603126A1 (en)
IL (1) IL186342A0 (en)
MX (1) MX2007011962A (en)
NZ (1) NZ562144A (en)
WO (1) WO2006105442A2 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0415165A (en) * 2003-10-01 2007-01-09 Adolor Corp spirocyclic heterocyclic compound, pharmaceutical composition, method to bind opioid receptors, methods to prevent or treat pain, gastrointestinal dysfunction, a urogenital tract disorder, an immunomodulatory disorder, an inflammatory disorder, a respiratory function disorder, anxiety, mood disorder, a stress-related disorder, sympathetic nervous system disorder, cough, and a motor disorder, method for treating traumatic injury to the central nervous system, methods for preventing or treating stroke, cardiac arrhythmia, glaucoma, and sexual dysfunction, methods for treating a condition selected from the group consisting of shock, cerebral edema, cerebral ischemia, cerebral deficit after cardiac surgery (bypass) and graft, systemic lupus erythematosus, hodgkin's disease, sjogren's disease, epilepsy, and rejection in organ transplants and skin grafts, and to treat substance dependence, method to improve organ and cell survival, and cardioprotection after myocardial infarction, methods to reduce the need for anesthesia, to produce or maintain an anesthetic state, radiolabeled derivative of a compound, isotopically labeled derivative of a compound, compound and method of diagnostic imaging
US7598261B2 (en) * 2005-03-31 2009-10-06 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US7576207B2 (en) * 2006-04-06 2009-08-18 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
WO2008033299A2 (en) * 2006-09-12 2008-03-20 Adolor Corporation Use of n-containing spirocompounds for the enhancement of cognitive function
US8258131B2 (en) * 2007-08-01 2012-09-04 Mitsubishi Tanabe Pharma Corporation Fused bicyclic compound
RU2010151951A (en) 2008-05-20 2012-06-27 Ньюроджесэкс, Инк. (Us) GENERAL PROCEDURES OF HEPATOPROTECTOR AND ACETAMINOPHEN
WO2011140425A1 (en) 2010-05-06 2011-11-10 Vertex Pharmaceuticals Incorporated Heterocyclic chromene-spirocyclic piperidine amides as modulators of ion channels
SG192248A1 (en) 2011-02-02 2013-09-30 Vertex Pharma Pyrrolopyrazine-spirocyclic piperidine amides as modulators of ion channels
CN103443105A (en) * 2011-02-18 2013-12-11 沃泰克斯药物股份有限公司 Chroman-spirocyclic piperidine amides as modulators of ion channels
CA2829803A1 (en) 2011-03-14 2012-09-20 Vertex Pharmaceuticals Incorporated Morpholine-spirocyclic piperidine amides as modulators of ion channels
GB201113538D0 (en) 2011-08-04 2011-09-21 Karobio Ab Novel estrogen receptor ligands
US9434693B2 (en) 2012-08-16 2016-09-06 Janssen Pharmaceutica Nv Substituted pyrazoles as N-type calcium channel blockers
EP3033337B1 (en) * 2013-08-16 2018-02-28 Janssen Pharmaceutica NV Substituted imidazoles as n-type calcium channel blockers
US9453002B2 (en) 2013-08-16 2016-09-27 Janssen Pharmaceutica Nv Substituted imidazoles as N-type calcium channel blockers
CN107602515A (en) * 2017-10-13 2018-01-19 南开大学 One kind 5A5B6CTricyclic spiro lactone derivatives and its production and use
WO2023283743A1 (en) * 2021-07-16 2023-01-19 Aphiotx Inc. Sulfamoyl benzene derivatives and uses thereof

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5106998A (en) * 1989-06-05 1992-04-21 Tokuyama Soda Kabushiki Kaisha Photochromic compound, composition and use thereof
US5132307A (en) * 1988-11-08 1992-07-21 Merck Patent Gesellschaft Mit Beschrankter Haftung Tetralin compounds
US5354863A (en) * 1992-01-21 1994-10-11 G. D. Searle & Co. Opioid agonist compounds
US5356915A (en) * 1992-10-19 1994-10-18 J. Uriach & Cia. S.A. Tetralones with pharmacological activity, compositions containing them
US5387587A (en) * 1986-12-23 1995-02-07 Merck Patent Gesellschaft Mit Beschrankter Haftung Chroman derivatives
US5403846A (en) * 1993-11-22 1995-04-04 Merck & Co., Inc. Spirocycles
US5628935A (en) * 1994-10-28 1997-05-13 Vision-Ease Lens, Inc. Photochromic spironaphthopyran compounds
US5656420A (en) * 1995-02-24 1997-08-12 University Of Kentucky Research Foundation Method for employing the delta opioid dadle to extend tissue survival time during ischemia
US5786378A (en) * 1996-09-25 1998-07-28 Gpi Nil Holdings, Inc. Heterocyclic thioesters
US6022895A (en) * 1997-03-14 2000-02-08 Gruenenthal Gmbh Substituted amino compounds and their use as substances having an analgesic effect
US6031115A (en) * 1993-05-14 2000-02-29 Smithkline Beecham Plc Process for preparing epoxides
US6200978B1 (en) * 1998-03-05 2001-03-13 Pfizer Inc. Compounds as delta opioid agonists
US6218424B1 (en) * 1996-09-25 2001-04-17 Gpi Nil Holdings, Inc. Heterocyclic ketone and thioester compounds and uses
US6319939B1 (en) * 1997-12-11 2001-11-20 Janssen Pharmaceutica N.V. Retinoic acid mimetic anlides
US6436959B1 (en) * 1998-12-23 2002-08-20 Ortho-Mcneil Pharmaceutical, Inc. 4-[aryl(piperidin-4-yl)]aminobenzamides
US20030069241A1 (en) * 2001-07-24 2003-04-10 Mchardy Stanton F. 1-Diphenylmethyl-pyrazole derivatives as opioid receptor ligands
US6596758B1 (en) * 1998-12-29 2003-07-22 Merck Patent Gmbh Benzopyrans and benzoxepines, pharmaceutical compositions comprising them and preparation process
US6645973B1 (en) * 1999-11-17 2003-11-11 Akzo Nobel Spiro(2h-1-benzopyran-2,4-piperidine)derivatives as glycine transport inhibitors
US20040082612A1 (en) * 2002-10-15 2004-04-29 Baxter Ellen W Benzyl substituted (piperidin-4-yl)aminobenzamido derivatives
US6790854B2 (en) * 2000-03-24 2004-09-14 Meiji Seika Kaisha, Ltd. Diphenylalkylamine derivatives useful as opioid receptor agonists
US20050159438A1 (en) * 2003-10-01 2005-07-21 Dolle Roland E. Spirocyclic heterocyclic derivatives and methods of their use
US7226933B2 (en) * 2001-05-18 2007-06-05 Astrazeneca Ab 4-(phenyl-(piperidin-4-yl)-amino)-benzamide derivatives and their use for the treatment of pain, anxiety or gastrointestinal disorders
US7598261B2 (en) * 2005-03-31 2009-10-06 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR8904728A (en) 1988-09-26 1990-05-01 Goodyear Tire & Rubber PROCESS FOR THE CATALYTED SYNTHESIS OF N-REPLACED ACRYLAMIDS
ES2071668T3 (en) 1988-12-13 1995-07-01 Beecham Group Plc BENZOPYRAN AND RELATED COMPOUNDS.
IT1230062B (en) * 1989-04-18 1991-09-27 Enichem Sintesi NEW SILANES CONTAINING AT LEAST TWO OXAZOLIDINE GROUPS, THEIR PREPARATION AND USE.
CA2026389A1 (en) 1989-09-29 1991-03-30 Hideo Nakai Chromene or thiochromene derivatives, process for preparing the same and intermediate therefor
JP2985322B2 (en) 1991-03-01 1999-11-29 東レ株式会社 Isoquinoline derivatives and their pharmaceutical uses
GB9202238D0 (en) 1992-02-03 1992-03-18 Wellcome Found Compounds
GB9203596D0 (en) 1992-02-20 1992-04-08 Smithkline Beecham Plc Novel process
GB9207400D0 (en) 1992-04-02 1992-05-13 Smithkline Beecham Plc Novel use
CA2100514C (en) 1992-07-29 2005-03-29 Johannes A. M. Hamersma 17-spiromethylene steroids
CA2154569A1 (en) 1993-01-28 1994-08-04 Jeffrey J. Hale Spiro-substituted azacycles as tachykinin receptor antagonists
JPH09505030A (en) 1993-08-06 1997-05-20 スミスクライン・ビーチャム・ソシエタ・ペル・アチオニ Hydroisoquinoline derivative
SE9401727D0 (en) 1994-05-18 1994-05-18 Astra Ab New compounds I
US5683628A (en) 1994-10-17 1997-11-04 Seiko Epson Corp. Photochromic composition and photochromic resin obtainable by using the same
AU4458996A (en) 1995-01-20 1996-08-07 Nippon Shinyaku Co. Ltd. Ethylamine derivatives and drugs
IT1307327B1 (en) 1995-09-12 2001-10-30 Smithkline Beecham Spa REPLACED HYDROISOKINOLINIC DERIVATIVES
SE9504661D0 (en) * 1995-12-22 1995-12-22 Astra Pharma Inc New compounds
JPH09301973A (en) 1996-05-09 1997-11-25 Kumiai Chem Ind Co Ltd Chromene derivative and herbicide containing the same as active ingredient
US6103722A (en) 1997-07-23 2000-08-15 The Medical College Of Wisconsin Research Foundation Inc. Ischemic preconditioning
GB9912413D0 (en) * 1999-05-28 1999-07-28 Pfizer Ltd Compounds useful in therapy
JP2005231995A (en) 1999-12-22 2005-09-02 Meiji Seika Kaisha Ltd SPIRO COMPOUND USEFUL AS OPIOID delta RECEPTOR AGONIST/ANTAGONIST
JP4275288B2 (en) 2000-03-31 2009-06-10 新明和工業株式会社 Hydraulic control device for dump vehicle
EP1278742B1 (en) 2000-04-27 2004-12-15 Basilea Pharmaceutica AG Chromenylmethylpyrimidinediamines as antibacterial agents
KR20020012346A (en) 2000-08-07 2002-02-16 유현식 Supported Catalyst for Producing a Syndiotactic Styrene Polymer with High Productivity and Reduced Reactor Fouling
WO2002048122A2 (en) 2000-12-14 2002-06-20 Ortho-Mcneil Pharmaceutical, Inc. Benzamidine derivatives
SE0101773D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
SE0101767D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
BR0209678A (en) 2001-05-18 2004-09-14 Astrazeneca Ab Compound, process for preparing and using same, pharmaceutical composition, and methods for treating pain and functional gastrointestinal disorders
SE0101771D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
SE0101768D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
SE0101770D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
SE0101766D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
SE0101765D0 (en) 2001-05-18 2001-05-18 Astrazeneca Ab Novel compounds
SE0103313D0 (en) 2001-10-03 2001-10-03 Astrazeneca Ab Novel compounds
HUP0600447A3 (en) 2001-10-15 2008-03-28 Janssen Pharmaceutica Nv Substituted 4-phenyl-4-[1h-imidazol-2-yl]-piperidine derivatives, pharmaceutical compositions containing them and their use as selective non-peptide delta opioid agonists
JP4892654B2 (en) 2001-10-29 2012-03-07 マウント クック ファーマ,インコーポレーテッド Method of treating depression with delta receptor agonist compounds
AU2003214800A1 (en) 2002-01-02 2003-07-24 Ardent Pharmaceuticals, Inc. Method of treating sexual dysfunctions with delta opioid receptor agonist compounds
MXPA05002983A (en) 2002-09-18 2005-06-22 Univ Missouri OPIATE ANALOGS SELECTIVE FOR THE delta-OPIOID RECEPTOR.
CA2501552A1 (en) 2002-10-15 2004-04-29 Janssen Pharmaceutica, N.V. Substituted aminoquinuclidine compounds
SE0203302D0 (en) 2002-11-07 2002-11-07 Astrazeneca Ab Novel Compounds
SE0203300D0 (en) 2002-11-07 2002-11-07 Astrazeneca Ab Novel Compounds
SE0203303D0 (en) 2002-11-07 2002-11-07 Astrazeneca Ab Novel Compounds
SE0203301D0 (en) 2002-11-07 2002-11-07 Astrazeneca Ab Novel Compounds
US7314880B2 (en) 2003-01-02 2008-01-01 Mount Cook Biosciences, Inc. Cardioprotective delta opioid receptor agonists and methods of using same
SE0300103D0 (en) 2003-01-16 2003-01-16 Astrazeneca Ab Diarylmethylidene piperidine derivatives, preparations thereof and uses thereof
SE0300104D0 (en) 2003-01-16 2003-01-16 Astrazeneca Ab Diarylmethylidene piperidine derivatives, preparations thereof and uses thereof
SE0300105D0 (en) 2003-01-16 2003-01-16 Astrazeneca Ab Diarylmethylidene piperdine derivatives, preparations thereof and uses thereof
WO2004082612A2 (en) 2003-03-14 2004-09-30 Optime Therapeutics Inc. Liposomal formulations and methods of use

Patent Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6153627A (en) * 1986-12-23 2000-11-28 Merck Patent Gesellschaft Mit Beschrankter Haftung Chroman derivatives
US6040308A (en) * 1986-12-23 2000-03-21 Merck Gesellschaft Mit Beschrankter Haftung Chroman derivatives
US5387587A (en) * 1986-12-23 1995-02-07 Merck Patent Gesellschaft Mit Beschrankter Haftung Chroman derivatives
US5132307A (en) * 1988-11-08 1992-07-21 Merck Patent Gesellschaft Mit Beschrankter Haftung Tetralin compounds
US5349065A (en) * 1989-06-05 1994-09-20 Tokuyama Soda Kabushiki Kaisha Photochromic compound
US5106998A (en) * 1989-06-05 1992-04-21 Tokuyama Soda Kabushiki Kaisha Photochromic compound, composition and use thereof
US5354863A (en) * 1992-01-21 1994-10-11 G. D. Searle & Co. Opioid agonist compounds
US5356915A (en) * 1992-10-19 1994-10-18 J. Uriach & Cia. S.A. Tetralones with pharmacological activity, compositions containing them
US6031115A (en) * 1993-05-14 2000-02-29 Smithkline Beecham Plc Process for preparing epoxides
US5403846A (en) * 1993-11-22 1995-04-04 Merck & Co., Inc. Spirocycles
US5705102A (en) * 1994-10-28 1998-01-06 Vision-Ease Lens, Inc. Photochromic spironaphthopyran compounds
US5628935A (en) * 1994-10-28 1997-05-13 Vision-Ease Lens, Inc. Photochromic spironaphthopyran compounds
US5656420A (en) * 1995-02-24 1997-08-12 University Of Kentucky Research Foundation Method for employing the delta opioid dadle to extend tissue survival time during ischemia
US5990131A (en) * 1996-09-25 1999-11-23 Gpi Nil Holdings Inc. Heterocyclic thioesters and ketones
US20020193420A1 (en) * 1996-09-25 2002-12-19 Gpi Nil Holdings, Inc. Heterocyclic ketone and thioester compounds and uses
US20040106652A1 (en) * 1996-09-25 2004-06-03 Gpi Nil Holdings, Inc. Heterocyclic ketone and thioester compounds and uses
US6218424B1 (en) * 1996-09-25 2001-04-17 Gpi Nil Holdings, Inc. Heterocyclic ketone and thioester compounds and uses
US5786378A (en) * 1996-09-25 1998-07-28 Gpi Nil Holdings, Inc. Heterocyclic thioesters
US20010056103A1 (en) * 1996-09-25 2001-12-27 Gpi Nil Holdings, Inc. Heterocyclic ketone and thioester compounds and uses
US6417209B2 (en) * 1996-09-25 2002-07-09 Gpi Nil Holdings, Inc. Heterocyclic ketone and thioester compounds and uses
US6022895A (en) * 1997-03-14 2000-02-08 Gruenenthal Gmbh Substituted amino compounds and their use as substances having an analgesic effect
US6319939B1 (en) * 1997-12-11 2001-11-20 Janssen Pharmaceutica N.V. Retinoic acid mimetic anlides
US20020115653A1 (en) * 1997-12-11 2002-08-22 Dominique Mabire Retinoic acid mimetic anilides
US6200978B1 (en) * 1998-03-05 2001-03-13 Pfizer Inc. Compounds as delta opioid agonists
US6436959B1 (en) * 1998-12-23 2002-08-20 Ortho-Mcneil Pharmaceutical, Inc. 4-[aryl(piperidin-4-yl)]aminobenzamides
US6596758B1 (en) * 1998-12-29 2003-07-22 Merck Patent Gmbh Benzopyrans and benzoxepines, pharmaceutical compositions comprising them and preparation process
US6645973B1 (en) * 1999-11-17 2003-11-11 Akzo Nobel Spiro(2h-1-benzopyran-2,4-piperidine)derivatives as glycine transport inhibitors
US6790854B2 (en) * 2000-03-24 2004-09-14 Meiji Seika Kaisha, Ltd. Diphenylalkylamine derivatives useful as opioid receptor agonists
US7226933B2 (en) * 2001-05-18 2007-06-05 Astrazeneca Ab 4-(phenyl-(piperidin-4-yl)-amino)-benzamide derivatives and their use for the treatment of pain, anxiety or gastrointestinal disorders
US20030069241A1 (en) * 2001-07-24 2003-04-10 Mchardy Stanton F. 1-Diphenylmethyl-pyrazole derivatives as opioid receptor ligands
US20040082612A1 (en) * 2002-10-15 2004-04-29 Baxter Ellen W Benzyl substituted (piperidin-4-yl)aminobenzamido derivatives
US20050159438A1 (en) * 2003-10-01 2005-07-21 Dolle Roland E. Spirocyclic heterocyclic derivatives and methods of their use
US7338962B2 (en) * 2003-10-01 2008-03-04 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US7638527B2 (en) * 2003-10-01 2009-12-29 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US20100280058A1 (en) * 2003-10-01 2010-11-04 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US7906646B2 (en) * 2003-10-01 2011-03-15 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US8071611B2 (en) * 2003-10-01 2011-12-06 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US7598261B2 (en) * 2005-03-31 2009-10-06 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use
US8022060B2 (en) * 2005-03-31 2011-09-20 Adolor Corporation Spirocyclic heterocyclic derivatives and methods of their use

Also Published As

Publication number Publication date
NZ562144A (en) 2011-04-29
JP5118788B2 (en) 2013-01-16
US7598261B2 (en) 2009-10-06
AU2006230517A1 (en) 2006-10-05
EP1871761A2 (en) 2008-01-02
WO2006105442A3 (en) 2007-08-02
EP1871761A4 (en) 2011-10-19
KR20080005245A (en) 2008-01-10
US8022060B2 (en) 2011-09-20
JP2008534613A (en) 2008-08-28
CA2603126A1 (en) 2006-10-05
BRPI0612189A2 (en) 2009-01-20
WO2006105442A2 (en) 2006-10-05
MX2007011962A (en) 2007-12-13
US20100029614A1 (en) 2010-02-04
US20060270695A1 (en) 2006-11-30
AU2006230517B2 (en) 2012-09-13
IL186342A0 (en) 2008-01-20

Similar Documents

Publication Publication Date Title
US8071611B2 (en) Spirocyclic heterocyclic derivatives and methods of their use
US8022060B2 (en) Spirocyclic heterocyclic derivatives and methods of their use
US7576207B2 (en) Spirocyclic heterocyclic derivatives and methods of their use
US20080119452A1 (en) Methods for enhancing cognitive function
CN101184749B (en) Spirocyclic heterocyclic derivatives and methods of their use
AU2012216319A1 (en) Spirocyclic heterocyclic derivatives and methods of their use
MXPA06003639A (en) Spirocyclic heterocyclic derivatives and methods of their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT, CAN

Free format text: SECURITY AGREEMENT;ASSIGNORS:CUBIST PHARMACEUTICALS, INC.;ADOLOR CORPORATION;CALIXA THERAPEUTICS, INC.;AND OTHERS;REEL/FRAME:029339/0669

Effective date: 20121120

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: CALIXA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT;REEL/FRAME:036180/0070

Effective date: 20150707

Owner name: ADOLOR CORPORATION, PENNSYLVANIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT;REEL/FRAME:036180/0070

Effective date: 20150707

Owner name: CUBIST PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT;REEL/FRAME:036180/0070

Effective date: 20150707