WO2011060113A1 - Sulfenamide compounds for phototherapy - Google Patents

Sulfenamide compounds for phototherapy Download PDF

Info

Publication number
WO2011060113A1
WO2011060113A1 PCT/US2010/056294 US2010056294W WO2011060113A1 WO 2011060113 A1 WO2011060113 A1 WO 2011060113A1 US 2010056294 W US2010056294 W US 2010056294W WO 2011060113 A1 WO2011060113 A1 WO 2011060113A1
Authority
WO
WIPO (PCT)
Prior art keywords
independently
compound
conr
aryl
cor
Prior art date
Application number
PCT/US2010/056294
Other languages
French (fr)
Inventor
Raghavan Rajagopalan
Original Assignee
Mallinckrodt Inc.
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 Mallinckrodt Inc. filed Critical Mallinckrodt Inc.
Publication of WO2011060113A1 publication Critical patent/WO2011060113A1/en

Links

Classifications

    • 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
    • C07D223/18Dibenzazepines; Hydrogenated dibenzazepines
    • C07D223/22Dibenz [b, f] azepines; Hydrogenated dibenz [b, f] azepines
    • C07D223/24Dibenz [b, f] azepines; Hydrogenated dibenz [b, f] azepines with hydrocarbon radicals, substituted by nitrogen atoms, attached to the ring nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles

Definitions

  • Optical agents currently play a central role in a large number of in vivo, in vitro and ex vivo clinical procedures including important diagnostic and therapeutic procedures.
  • Photodiagnostic and phototherapeutic agents include a class of molecules capable of absorbing, emitting, or scattering electromagnetic radiation applied to a biological material, particularly in the visible and near infrared regions of the electromagnetic spectrum. This property of optical agents is used in a range of biomedical applications for visualizing, imaging or otherwise characterizing biological materials and/or achieving a desired therapeutic outcome. Recent developments in targeted administration and delivery of optical agents, and advanced systems and methods for applying and detecting electromagnetic radiation in biological environments has considerably expanded the applicability and effectiveness of optical agents for clinical applications.
  • optical agents that absorb and/or emit in the visible and near- infrared (NIR) region of the electromagnetic spectrum include their use in biomedical imaging and visualization.
  • NIR near- infrared
  • compounds absorbing and/or emitting light in these regions of the electromagnetic spectrum currently are useful for optical tomography, optoacoustic tomography, optical coherence tomography, confocal scanning laser tomography, optical coherence
  • Biomedical images are generated, for example, by detecting electromagnetic radiation, nuclear radiation, acoustic waves, electrical fields, and/or magnetic fields transmitted, emitted and/or scattered by components of a biological sample.
  • Modulation of the energy or intensity of the applied radiation yields patterns of transmitted, scattered and/or emitted radiation, acoustic waves, electrical fields or magnetic fields that contain useful anatomical, physiological, and/or biochemical information.
  • a number of applications of biomedical imaging have matured into robust, widely used clinical techniques including planar projection and tomographic X-ray imaging, magnetic resonance imaging, ultrasound imaging, and gamma ray imaging.
  • Established optical imaging and visualization techniques are based on monitoring spatial variations in a variety of optical parameters including the intensities, polarization states, and frequencies of transmitted, reflected, and emitted electromagnetic radiation.
  • NIR visible and near infrared
  • NIR light 700 nm to 900 nm
  • electromagnetic radiation of this wavelength range is capable of substantial penetration (e.g. , up to four centimeters) in a range of biological media.
  • Optical imaging and visualization using optical agents has potential to provide a less invasive and safer imaging technology, as compared to X-ray, and other widely used nuclear medicine technologies.
  • Applications of optical imaging for diagnosis and monitoring of the onset, progression and treatment of various disease conditions, including cancer, are well established.
  • D. A. Benaron and D. K. Stevenson Optical time-of-flight and absorbance imaging of biologic media, Science, 1993, 259, pp. 1463-1466; R. F. Potter (Series Editor), Medical optical tomography: functional imaging and monitoring, SPIE Optical Engineering Press, Bellingham, 1993; G. J.
  • optical imaging agents are commonly used for enhancing signal-to-noise and resolution of optical images and extending these techniques to a wider range of biological settings and media.
  • use of optical imaging agents having specific molecular recognition and/or tissue targeting functionality has also been demonstrated as effective for identifying, differentiating and characterizing discrete components of a biological sample at the organ, tissue, cellular, and molecular levels.
  • optical agents have been developed as tracers for real time monitoring of physiological function in a patient, including fluorescence-based monitoring of renal function. (See International Patent Publication
  • Visudyne and Photofrin are two optical agents that have been developed for the treatment of macular degeneration of the eye and for ablation of several types of tumors, respectively.
  • Photodiagnostics and Phototherapy 2006, 3, 266-271 See, Zheng Huang "A Review of Progress in Clinical Photodynamic Therapy", Technol Cancer Res Treat. 2005 June; 4(3): 283-293; "Photodiagnosis And Photodynamic Therapy", Brown S, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment.
  • Phototherapy is carried out by administration and delivery of a photosensitizer to a therapeutic target tissue (e.g., tumor, lesion, organ, etc.) followed by photoactivation of the photosensitizer by exposure to applied electromagnetic radiation.
  • a therapeutic target tissue e.g., tumor, lesion, organ, etc.
  • Phototherapeutic procedures require photosensitizers that are relatively chemically inert, and become activated only upon irradiation with light of an appropriate wavelength.
  • Selective tissue injury can be induced with light when photosensitizers bind to the target tissues, either directly or through attachment to a bioactive carrier or targeting moiety.
  • Photosensitizers essentially operate via two different pathways, classified as Types 1 and 2. A primary distinction between these classes of
  • Type 1 process operates via direct energy or electron transfer from the photosensitizer to the cellular components thereby inducing cell death
  • Type 2 process involves first the conversion of singlet oxygen from the triplet oxygen found in the cellular environment followed by either direct reaction of singlet oxygen with the cellular components or further generating secondary reactive oxygen species (ROS) (e.g. peroxides, hydroxyl radical, etc.) which will induce cell death
  • ROS secondary reactive oxygen species
  • Type 1 agents may also interact with oxygen, if present, to produce ROS, but this is not a necessary requirement, and does not have to be not mediated via singlet oxygen.
  • the Type 1 mechanism proceeds via a multistep process involving activation of the photosensitizer by absorption of electromagnetic radiation followed by direct interaction of the activated photosensitizer, or reactive intermediates derived from the photosensitizer, with the target tissue, for example via energy transfer, electron transfer or reaction with reactive species (e.g., radicals, ions, nitrene, carbene etc.) resulting in tissue damage.
  • the Type 1 mechanism can be schematically represented by the following sequence of reactions:
  • the Type 2 mechanism proceeds via a multi-step process involving activation of the photosensitizer by absorption of electromagnetic radiation followed by energy transfer from the activated photosensitizer to oxygen molecules in the environment of the target tissue. This energy transfer process generates excited state oxygen ( 0 2 ) which subsequently interacts with the target tissue so as to cause tissue damage.
  • the Type 2 mechanism can be schematically represented by the following sequence of reactions: n i lOTA r kl lTI 7rn hv PHOTOSENSITIZER* 3 °2 ., _ . _ . . . PHOTOSENSITIZER ⁇ ( Exc ited State) ⁇ 2 (Singlet Oxygen)
  • Type 2 phototherapeutic agents based on the Type 2 (PDT) mechanism.
  • PDT Type 2
  • the Type 1 process does not require oxygen; and hence Type 1 photosensitizers are expected to be potentially more effective than Type 2 photosensitizers under hypoxic environments typically found in solid tumors.
  • the Type 1 mechanism involves two steps (photoexcitation and direct energy transfer), whereas the Type 2 mechanism involves three steps (photoexcitation, singlet oxygen generation, and energy transfer).
  • Type 1 photosensitizers by their very nature, are not expected to produce reactive oxygen species; rather, the reactive species produced by these photosensitizers will immediately react with the cellular component at the binding site and trigger cell death.
  • Type 2
  • Type 2 agents can potentially be catalytic, i.e., the Type 2 photosensitizer is regenerated once the energy transfer to the oxygen has taken place.
  • Type 1 process would generally be expected to require stoichiometric amounts of the photosensitizer in some clinical settings.
  • Table B1 provides a summary of the attributes of Type 1 and Type 2 phototherapeutic agents. Given these attributes, it is clear that development of safe and effective Type 1 phototherapeutic agents would be useful to complement the existing therapeutic approaches provided by Type 2 agents, and to enhance the therapeutic portfolio available for clinicians.
  • Light of any wavelength can be used. Requires red light for optimal performance. Does not require oxygen. Requires oxygen.
  • Intramolecular energy transfer to generate Intermolecular energy transfer to generate reactive species reactive oxygen species. No products in the market. Two products are in use.
  • optical agents for these applications preferably have strong absorption in the visible or NIR regions, and also exhibit low systemic toxicity, low mutagenicity, and rapid clearance from the blood stream. These optical agents must also be compatible with effective administration and delivery to the target tissue, for example by having reasonable solubilities and a low tendency for aggregation in solution.
  • optical agents for Type 1 and 2 phototherapy preferably provide large yields of singlet oxygen (Type 2) or other reactive species, such as free radicals or ions, capable of causing local tissue damage.
  • Type 1 and Type 2 photosensitizers typically undergo photoactivation followed by intersystem crossing to their lowest triplet excited state, and therefore, a relatively long triplet lifetime is usually beneficial for providing effective tissue damage.
  • Other useful properties of optical agents for these applications include chemical inertness and stability, insensitivity of optical properties to changes in pH, and compatibility with conjugation to ligands providing targeted delivery via molecular recognition functionality.
  • Multifunctional optical agents have also been developed for phototherapy that are capable of providing both imaging and visual functionality upon excitation at a first range of wavelengths and phototherapeutic functionality upon excitation at a second range of wavelength. (See, US Patent No. 7,235,685 and International Patent Publication WO
  • Optical agents for some phototherapeutic applications preferably exhibit a high degree of selectivity for the target tissue.
  • Selectivity provided by optical agents facilitates effective delivery to a target tissue of interest and provides a means of differentiating different tissue classes during therapy.
  • Selective tissue injury can be induced with light when photosensitizers bind to the target tissues either directly, as in the case of Photofrin, or through attachment to a bioactive carrier, or through in situ biochemical synthesis of the photosensitizer in localized area, as in the case of 2- aminolevulinic acid, which is an intermediate in the biosynthesis of porphyrin.
  • Previous studies have shown that certain dyes selectively localize in tumors and serve as a powerful probe for the detection and treatment of small cancers.
  • receptor-target mediated phototherapy agents provide a promising pathway for achieving site selective activation at various target tissues.
  • optical agents for biomedical applications are needed having enhanced specificity for important target tissue classes, such as tumors and other lesions.
  • optical agents are needed having enhanced optical, physical, chemical and pharmacokinetic properties for administration, delivery and excitation with electromagnetic radiation.
  • the invention relates generally to optical agents for biomedical applications, including phototherapy.
  • sulfenamide compounds having an acyclic S-N bond with at least one aromatic and/or heterocyclic aromatic group, including fused ring carbocyclic and/or heterocyclic aromatic groups, providing phototherapeutic agents, including Type 1 phototherapeutic agents.
  • Optical agents of the invention enable a versatile phototherapy platform for treatment of a range of pathological conditions, including the treatment of cancers, stenosis and inflammation.
  • the invention further provides preparations and formulations comprising the sulfenamide optical agents and related methods of making and using sulfenamide optical agents in an in vivo or ex vivo biomedical procedure.
  • the invention provides sulfenamide compounds for phototherapeutic methods having one or more photolabile acyclic S-N bonds capable of undergoing photoactivated cleavage to produce reactive species, such as radicals, ions, etc., that achieve a desired therapeutic effect, such as selective and/or localized tissue damage and/or cell death.
  • Optical agents of an aspect of the invention include compositions having one or more carbocyclic and/or heterocyclic aromatic groups, including fused ring aromatic groups, providing a chromophore directly or indirectly coupled to an acyclic S-N bond capable of undergoing photodissociation and/or photofragmentation upon exposure to electromagnetic radiation having wavelengths in the visible and/or near infrared regions of the electromagnetic spectrum.
  • Optical agents further include conjugates, such as sulfenamide bioconjugate compositions including at least one targeting ligand such as a polypeptide, protein, oligonucleotide, carbohydrate, antibody, or other biomolecule, or fragments or fusions thereof, capable of providing molecular recognition and/or tissue specific targeting functionality.
  • Optical agents further include multifunctional optical agents providing tandem imaging and phototherapy functionality, wherein the agent comprises a photosensitizer component having an acyclic S-N bond directly or indirectly linked to an optical dye component, such as a C 5 -C 30 aryl or C 5 -C 30 heteroaryl fluorophore, and optionally further comprising one or more targeting ligands.
  • a photosensitizer component having an acyclic S-N bond directly or indirectly linked to an optical dye component, such as a C 5 -C 30 aryl or C 5 -C 30 heteroaryl fluorophore, and optionally further comprising one or more targeting ligands.
  • the invention provides sulfenamide compounds having an acyclic S-N bond useful as optical agents for phototherapeutic methods, including Type 1 phototherapy.
  • the invention provides a compound for use in a phototherapy
  • each of L 1 , L 2 , and L 3 is independently CrC 10 alkylene, C 3 -Ci 0 cycloalkylene, C 2 -C 10 alkenylene, C 3 -C 10 cycloalkenylene, C 2 -Ci 0 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, ⁇ (CH 2 CH 2 0) b - or -(CHOH) a -;
  • each of W 1 , W 2 , and W 3 is independently a single bond, -(CH 2 ) n - -(HCCH) n -, -0-, -S-, -SO-, -S0 2 - -S0 3 - -OS0 2 -, -NR 1 -, -CO-, -COO-, -OCO-, -OCOO-, -CONR 2 -, - NR 3 CO- -OCONR 4 -, -NR 5 COO- -NR 16 CONR 17 -, -NR 18 CSNR 19 -, -0(CH 2 ) n - -S(CH 2 ) n - - NR 20 (CH 2 ) n -, -CO(CH 2 ) n - -COO(CH 2 ) n - -OCO(CH 2 ) n -, -OCOO(CH 2 ) n -, -
  • each of R 1 , R 2 , and R 3 is independently a hydrogen, -OCF 3 , C C 20 alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, C C 20 acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C C 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 4 °, -SOR 41 , - OSR 42 , -S0 2 OR 43 , -CH 2 (CH 2 OCH 2 ) b CH 2 OH, -P0 3 R 4 R 45 , -OR 46 , -SR 47 , -NR 8 R 49 , - NR 50 COR 51 , -CN, -CONR 52 R 53 , -COR 54 , -N0 2
  • R 2 , R 3 , W 2 , W 3 , and L 2 and L 3 if present, together with the atoms to which they are attached combine to form one to five substituted or unsubstituted carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings;
  • R 1 , R 2 , and R 3 is a C 5 -C 30 aryl or C 5 -C 30 heteroaryl, or wherein R 2 , R 3 , W 2 , W 3 , and L 2 and L 3 , if present, together with the atoms to which they are attached combine to form a C 5 -C 30 aryl or C 5 -C 30 heteroaryl;
  • each of a and b is independently an integer selected from the range of 1 to 100;
  • each n is independently an integer selected from the range of 1 to 10;
  • each of e, f, and g is independently 0 or 1 ; each of R - R 33 is independently hydrogen, C C 2 o alkyl, C 3 -C 2 o cycloalkyl, C 5 -C 20 heteroaryl, or C 5 -C 2 o aryl;
  • each of R 40 -R 55 and R 58 - R 61 is independently hydrogen, C 3 -C 10 cycloalkyl or d-C 10 alkyl; each of R 62 and R 63 is independently a side chain residue of a natural a-amino acid;
  • each FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a
  • glycopeptide a glycoprotein, a peptidomimetic, a drug, a steriod, a hormone, an aptamer, a receptor, a metal chelating agent, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
  • the composition of R , R 2 , R 3 , W 1 , W 2 , W 3 , L , L 2 , and/or L 3 is selected such that the compound undergoes cleavage of the central S-N bond upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, and optionally wavelengths selected over the range of 400 nanometers to 900 nanometers.
  • the invention provides a compound having formula (FX1), wherein at least one of R 1 , R 2 and R 3 is a C 5 -C 30 aryl or C 5 -C 30 heteroaryl group
  • benzene naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline.
  • the invention provides a compound having formula (FX1), wherein R 2 , R 3 , W 2 , W 3 , and L 2 and L 3 , if present, together with the atoms to which they are attached combine to form one to five, and optionally one to three, carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings substituted to include one or more electron donating group, electron with drawing group, targeting group (e.g., Bm) or fluorophore group (e.g., FL).
  • FX1 formula (FX1), wherein R 2 , R 3 , W 2 , W 3 , and L 2 and L 3 , if present, together with the atoms to which they are attached combine to form one to five, and optionally one to three, carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings substituted to include one or more electron donating group, electron with drawing group, targeting group (e.g., Bm
  • reference to embodiments wherein e, f, and/or g is equal to 0 refers to compounds where L 1 , L , and/or L 3 , respectively, is not present; and reference to embodiments wherein e, f, and/or g is equal to 1 refers to compounds where L 1 , L 2 , and/or L 3 , respectively, is present.
  • W 1 is directly linked to the central S-N group when e is equal to 0; and/or W 2 is directly linked to the central S-N group when f is equal to 0; and/or W 3 is directly linked to the central S-N group when g is equal to 0.
  • Embodiments wherein W 1 is a single bond and e is equal to 0 refer to compositions having R 1 directly linked to the central S-N group via a single bond.
  • Embodiments wherein W 2 is a single bond and f is equal to 0 refer to compositions having R 2 directly linked to the central S-N group via a single bond.
  • Embodiments wherein W 3 is a single bond and g is equal to 0 refer to compositions having R 3 directly linked to the central S-N group via a single bond. This convention is used throughout the present description, for example, in connection with other L and W groups, such as L 20 -L 127 and W 120 - W 127 .
  • a group corresponding to" an indicated species expressly includes a radical (including a monovalent, divalent and trivalent radical), for example an aromatic radical or heterocyclic aromatic radical, of the species or group of species provided in a covalently bonded configuration, optionally with one or more substituents, including but not limited to one or more electron donating groups, electron withdrawing groups, fluorophore groups, photosensitizer groups and/or targeting ligands.
  • a radical including a monovalent, divalent and trivalent radical
  • an aromatic radical or heterocyclic aromatic radical for example an aromatic radical or heterocyclic aromatic radical, of the species or group of species provided in a covalently bonded configuration, optionally with one or more substituents, including but not limited to one or more electron donating groups, electron withdrawing groups, fluorophore groups, photosensitizer groups and/or targeting ligands.
  • R 1 is C 5 -C 30 aryl or C 5 -C 30 heteroaryl
  • each of R 2 and R 3 is independently hydrogen, C ⁇ -C 2 o alkyl, C 3 -C 2 o cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, FL, or Bm.
  • R 2 is C 5 -C 30 aryl or C 5 -C 30 heteroaryl
  • each of R 1 and R 3 is independently hydrogen, C C 2 o alkyl, C 3 -C 2 o cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, FL, or Bm.
  • R 3 is C 5 -C 30 aryl or C 5 -C 30 heteroaryl, and each of R 1 and R 2 is independently hydrogen, C C 2 o alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, FL, or Bm.
  • the invention provides compounds useful as optical agents for phototherapeutic methods having formula (FX1 ), wherein R is C ⁇ C W alkyl, C 3 -C 10 cycloalkyl, C 5 -C 30 heteroaryl, or a C 5 -C 30 aryl, wherein optionally the C 5 -C 30 aryl or C 5 -C 30 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R 2 and R 3 is independently a hydrogen, C r C 2 o alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, a FL or Bm, wherein optionally the C 5 -C 30 aryl or C 5 -C 30 heteroaryl, includes an electron donating group, electron withdrawing group, or a targeting ligand (Bm) as a substituent.
  • R is C ⁇ C W alkyl, C 3 -C 10
  • the invention provides compounds useful as optical agents for phototherapeutic methods having formula (FX1), wherein R 2 is C C 10 alkyl, C 3 -Ci 0 cycloalkyl, C 5 -C 30 heteroaryl, or a C 5 -C 30 aryl, wherein optionally the C 5 -C 30 aryl or C 5 -C 30 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R 1 and R 3 is independently a hydrogen, C C 20 alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 heteroaryl, C 5 -C 30 aryl, a FL or Bm, wherein optionally the C 5 -C 30 aryl or C 5 -C 30 heteroaryl includes an electron donating group, electron withdrawing group, or a targeting ligand (Bm) as a substituent.
  • R 2 is C C 10 alkyl, C 3 -Ci 0 cyclo
  • the invention provides compounds useful as optical agents for phototherapeutic methods having formula (FX1 ), wherein R 3 is C C 0 alkyl, C 3 -C 10 cycloalkyl, C 5 -C 30 heteroaryl, or a C 5 -C 30 aryl, wherein optionally the C 5 -C 30 aryl or C 5 -C 30 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R 1 and R 2 is independently a hydrogen, Ci-C 2 o alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 heteroaryl, C 5 -C 30 aryl, a FL or Bm, wherein optionally the C 5 -C 30 aryl or C 5 -C 30 heteroaryl includes an electron donating group, electron withdrawing group, or a targeting ligand (Bm) as a substituent
  • R 3 is C C 0 alkyl, C 3 -C 10 cycl
  • the invention further provides compounds useful as optical agents for phototherapeutic methods having formula (FX1 ), wherein one or more of R 1 , R 2 and R 3 are independently one or more carbocylic and/or heterocyclic aromatic groups, including fused ring aromatic groups, providing a chromophore capable of excitation upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, and optionally in some embodiments wavelengths selected over the range of 400 nanometers to 900 nanometers.
  • the invention provides compounds for use in a phototherapeutic methods having formula (FX2), (FX3), (FX4), (FX5), (FX6), (FX7) or (FX8):
  • L , L 2 , L 3 , W 1 , W 2 , W 3 , R 1 , R 2 , R 3 , e, f and g are as described in connection with formula (FX1); and wherein each of Ar 1 , Ar 2 and Ar 3 is independently a C 5 -C 30 aryl or C 5 -C 30 heteroaryl group comprising one or more aromatic rings or heterocyclic aromatic rings, optionally having one or more electron donating groups and/or electron withdrawing groups provided as substituents.
  • the invention provide compounds being of formula (FX2) - (FX15), wherein each of Ar 1 , Ar 2 and Ar 3 is independently a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene,
  • benzothiophene dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline.
  • the invention provides compounds having formula (FX1 ) wherein one or more aromatic group (e.g., C 5 -C 30 aryl or C 5 -C 30 heteroaryl) is directly linked to the central S-N group.
  • one or more aromatic group e.g., C 5 -C 30 aryl or C 5 -C 30 heteroaryl
  • the expression directly linked refers to a configuration wherein an aromatic group is covalently bonded to either the S atom or the N atom of the central S-N group, for example, via a single bond.
  • the invention provides compounds for use in a phototherapeutic methods having formula (FX9),
  • the invention provides compounds for use in a phototherapeutic methods having any one of formula (FX2) - (FX15): wherein at least one of R 1 - R 3 , R 65 - R 67 and Ar 1 - Ar 3 is independently (or optionally each of Ar 1 and Ar 2 , Ar 1 and Ar 3 , Ar 2 and Ar 3 , or Ar 1 , Ar 2 and Ar 3 is): ; wherein each of R - R is independently a hydrogen, -OCF 3 , alkyl, C 3 -C 20 cycloalkyl,C 5 -C 2 o aryl, C 5 -C 2 o heteroaryl, C-i-C 2 o acyl, C 2 - C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C C 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2
  • the invention provides compounds for use in a phototherapeutic methods having any one of formula (FX2) - (FX1 5): wherein at least one of R 1 - R 3 , R 65 - R 67 and 1 - Ar 3 is independently (or optionally each of Ar 1 and Ar 2 , Ar 1 and Ar 3 , Ar 2 and Ar 3 , or Ar , Ar 2 - R is independently a hydrogen, -OCF 3 , C
  • the invention provides compounds for use in a phototherapeutic methods having any one of formula (FX2) - (FX1 5): wherein at least one of R - R 3 , R 65 - R 67 and Ar 1 - Ar 3 is independently (or optionally each of Ar 1 and Ar 2 , Ar 1 and Ar 3 , Ar 2 and Ar 3 , or Ar 1 , Ar 2 and Ar 3 is):
  • each of R - R is independently a hydrogen, -OCF 3 , C C 20 alkyl, C 3 -C 2 o cycloalkyl, C 5 -C 2 o aryl, C 5 -C 20 heteroaryl, C C 2 o acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C C 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 40 , -
  • R B - R are as described in the context of formula (FX1). In an embodiment, the invention
  • the invention provides compounds wherein at least one of R R , or at least one of R R , or at least one of R - R is independently an electron donating group; and wherein at least one of R - R , or at least one of R 75 - R 77 , or at least one of R 78 - R 85 is independently an electron withdrawing group.
  • the invention provides compounds wherein each of R 70 - R 85 is independently a hydrogen, -OR 46 , -SR 47 , -NR 48 R 49 , -NR 50 COR 51 , -CN, halo, -CO 2 R 40 ,-COR 54 , - N0 2 , -S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the invention provides compounds wherein at least one of R R , or at least one of R R , or at least one of R - R is independently an electron donating group; and wherein at least one of R - R , or at least one of R 75
  • R R is independently a
  • OR 4b -SR 4 , -NR R , -NFTOOR , -CN, halo, -C0 2 R 4U ,-COR S4 , -N0 2 , -S0 2 R t>b , or - S0 2 NR 58 R 59 .
  • the invention provides compounds wherein at least one of R R , or at least one of R /s - R , or at least one of R' a - R 8S , is independently a -OR 4b , -SR , - NR 48 R 49 , or -NR 50 COR 51 ; and at least one of R 70 - R 74 , or at least one of R 75 - R 77 , or at least one of R 78 - R 85 , is independently -CN, halo, -CO 2 R 40 ,-COR 54 , -N0 2 , -S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the invention provides compounds wherein at least one of R 70 - R 74 , or at least one of R 75 - R 77 , or at least one of R 78 - R 85 , is independently Bm. In an embodiment, the invention provides compounds, wherein each of R 70 - R 85 is hydrogen or Ci - C 6 alkyl, and optionally wherein each of R 70 - R 85 is hydrogen or C-i - C 3 alkyl, and optionally each of wherein R 70 - R 85 is hydrogen.
  • the invention provides compounds for use in phototherapy having formula (FX1 ), wherein R 2 , W 2 , R 3 , and W 2 , and L 2 and L 3 , if present, together with the atoms to which they are attached combine to form one or more alicyclic ring structures.
  • the invention provides a compound for use in a phototherapy procedure, the compound being of the
  • X is independently -CR 65 R 66 -, -NR 67 -, -0-, -S-, -SO-, or -S0 2 -;
  • each of Y and Y 2 is independently -CR 65 R 66 -;
  • each of R 65 , R 66 and R 67 is independently a hydrogen, -OCF 3 , C r C 2 o alkyl, C 3 -C 2 o cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, C C 20 acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C-
  • each of i and h is independently an integer selected from the range of 0 to 6, wherein at least one of i and h is greater than or equal to 1 ; wherein R 1 , W , L 1 , e, b, FL, Bm, R 40 - R 55 and R 58 - R 63 are as described in the context of formula (FX1).
  • i 0, Y 1 is not present and X is directly bonded to the central nitrogen of formula (FX16).
  • h is 0, Y 2 is not present and X is directly bonded to the central nitrogen of formula (FX16).
  • R 1 , R 65 , R 66 and R 67 is independently C 5 -C 30 aryl or C 5 -C 30 heteroaryl.
  • the invention provides a compound for use in a phototherapy procedure, the compound being of the formula (FX17): (FX17); or a pharmaceutically acceptable salt or ester thereof, wherein L 1 , W 1 , R 1 , i, h, e and X are as described in connection with formula (FX16).
  • the invention provides a compound for use in a phototherapeutic procedure having formula (FX16) or (FX17), wherein R is C 5 -C 30 aryl or C 5 -C 30 heteroaryl, optionally C 5 -C 2 o aryl or C 5 -C 30 heteroaryl, or C 5 -C 10 aryl or C 5 -C 30 heteroaryl.
  • the invention provides a compound for use in a phototherapeutic procedure having any one of formula (FX16) or (FX17), wherein each of X, Y 1 and Y 2 is independently -CR 65 R 66 -, wherein optionally each of R 65 and R 66 is a hydrogen.
  • the invention provides a compound for use in a phototherapeutic procedure having any one of formula (FX16) and (FX17), wherein X is -0-, and wherein each of Y and Y 2 is independently -CR 65 R 66 -, wherein optionally each of R 65 and R 66 is hydrogen.
  • the invention provides a compound for use in a phototherapeutic procedure having any one of formula (FX16) and (FX17), wherein X is -S-, and each of Y and Y 2 is independently -CR 65 R 66 -, wherein optionally each of R 65 and R 66 is hydrogen.
  • the invention provides a compound having formula (FX16) or (FX17) wherein the sum of i and h is 2 and, therefore, a carbocyclic or heterocyclic four-membered alicyclic ring incorporating the N from the central S-N group is formed; or wherein the sum of i and h is 3 and a carbocyclic or heterocyclic five-membered alicyclic ring incorporating the N from the central S-N group is formed; or wherein the sum of i and h is 4 and a carbocyclic or heterocyclic six-membered alicyclic ring incorporating the N from the central S-N group is formed; or wherein the sum of i and h is 5 and a carbocyclic or heterocyclic seven-membered alicyclic ring
  • the invention provides compounds having formula (FX16) and (FX17), wherein R is a C 5 -C 30 aryl or C 5 -C 30 heteroaryl, and R 65 , R 66 , and R 67 are independently hydrogen, C C 2 o alkyl, C 3 -C 2 o cycloalkyl, FL, or Bm.
  • the invention provides a compound having formula (FX16) or (FX17), wherein at least one of R 65 , R 66 , and R 67 is Bm.
  • the invention provides compounds having formula (FX16) and (FX17), wherein R 1 is a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene,
  • the invention provides compounds for phototherapy being of the formula (FX20): (FX20); or a pharmaceutically acceptable salt or ester thereof, wherein:
  • each of rings A, B, and C are independently or ; and each of R 70
  • - R 77 is independently a hydrogen, -OCF 3 , C C 2 o aikyl, C 3 -C 20 cycloalkyl, C 5 -C 2 o aryl, C 5 -C 20 heteroaryl, C C 20 acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, CTC 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 4 °, -SOR 41 , -OSR 42 , -S0 2 OR 43 , - CH 2 (CH 2 OCH 2 ) B CH 2 OH, -P0 3 R 44 R 45 , -OR 46 , -SR 47 , -N R 48 R 49 , -NR 50 COR 51 , -CN , -CONR 52 R 53 , -COR 54 , -N0 2 ,
  • the invention provides a compound being of formula (FX20) - (FX26), wherein at least one of R 70 - R 74 is and electron donating group and at least one of R 70 - R 74 is and electron withdrawing group. In an embodiment, the invention provides a compound being of formula (FX20) - (FX26), wherein at least one of R 75 - R 77 is and electron donating group and at least one of R 75 - R 77 is and electron withdrawing group.
  • the invention provides a compound being of formula (FX20) - (FX26), wherein each of R 70 - R 77 is independently a hydrogen, -N0 2 , -OCF 3 , C C 2 o alky!, C 3 -C 20 cycloalkyl, C 5 - C 2 o aryl, or C 5 -C 20 heteroaryl, and optionally wherein each of R 70 - R 77 is independently a hydrogen, -N0 2 , -OCF 3 , or Ci-C 6 alkyl, and optionally wherein each of R 70 - R 77 is independently a hydrogen, or C-i-C 6 alkyl.
  • the invention provides a compound being of formula (FX20) - (FX26), wherein at least one of R 70 - R 77 is -N0 2 .
  • the invention provides compounds being of formula (FX20) - (FX26), wherein each of R 70 - R 77 is independently a hydrogen,-OR 46 , -SR 47 , -NR 48 R 49 , -NR 50 COR 51 , - CN, halo, -CO 2 R 40 ,-COR 54 , -N0 2 , -S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R 70 - R 77 is independently a -OR 46 , -SR 47 , -NR 48 R 49 , -NR 50 COR 51 , -CN, halo, -CO 2 R 40 ,-COR 54 , -N0 2 , - S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R 70 - R 77 is independently a -OR 46 , -SR 47 , -NR 48 R 49 , or -NR 50 COR 51 ; and at least one of R 70 - R 77 is independently -CN, halo, -CO 2 R 0 ,-COR 54 , -N0 2 , - S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R 70 - R 77 is independently is independently Bm.
  • the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R 70 - R 77 is independently hydrogen or Ci - C 6 alkyl, and optionally wherein each of R 70 - R 77 is hydrogen or C - C 3 alkyl, and optionally each of wherein R 70 - R 77 is hydrogen.
  • the invention provides compounds for use in phototherapy having formula (FX1), wherein R 2 , W 2 , R 3 , and W 2 , and L 2 and L 3 , if present, together with the atoms to which they are attached combine to form a fused ring structure comprising one to five, and optionally one to three, alicyclic and/or aromatic rings, optionally substituted with one or more electron donating groups, electron withdrawing groups, chromophore groups and/or targeting ligands.
  • the invention provides class of photosensitizers having a S-N bond with a fused ring configuration, wherein the nitrogen atom of the S-N bond is a part of the fuse ring structure. Examples of compounds of this aspect include dibenzazepine derivatives.
  • the invention provides a compound for phototherapy being of the
  • Z is a single bond
  • -CR 128 CR 129 -
  • -CR 130 R 131 CR 132 R 133 -
  • -CR 134 R 135 0- -CR 136 R 137 S-
  • each of R 20 to R 4 is independently a hydrogen, -OCF 3 , C C 2 o alkyl, C 3 -C 2 o cycloalkyl, C5-C30 aryl, C 5 -C 30 heteroaryl, C C 20 acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C C 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 40 , -SOR 41 , -OSR 42 , - S0 2 OR 43 , -CH 2 (CH 2 OCH 2 ) b CH 2 OH , -P0 3 R R 45 , -OR 46 , -SR 47 , -NR 48 R 49 , -NR 50 COR 51 , -CN , - CON R 52 R 53 , -COR 54 , -NO2, -S0 2
  • each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
  • each of L 120 , L 121 , L 122 , L 123 , L 124 , L 25 , L 126 and L 127 is independently C C 10 alkylene, C 3 -C 0 cycloalkylene, C 2 -Ci 0 alkenylene, C 3 -C 0 cycloalkenylene, C 2 -Ci 0 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH 2 CH 2 0)b- or -(CHOH) a -;
  • each of W 120 , W 121 , W 122 , W 123 , W 24 , W 125 , W 26 and W 127 is independently a single bond, -(CH 2 ) n - -(HCCH) n -, -0-, -S-, -SO-, -SO2-, -SO3-, -OSO2-, -NR -, -CO-, -COO-, -OCO- , -OCOO-, -CONR 12 -, -NR 13 CO- -OCONR 14 -, -NR 15 COO- -NR 16 CONR 17 -, -NR 18 CSNR 19 -, -0(CH 2 ) n -, -S(CH 2 ) n -, -NR 20 (CH 2 ) n -, -CO(CH 2 ) n - -COO(CH 2 ) n -, -OCO(CH 2 ) n
  • the invention provides a compound for phototherapy of formula (FX27) wherein at least one of R 1 and R 120 to R 41 is hydrogen, optionally wherein all of R and R 120 to R 141 are hydrogen.
  • the invention provides a compound for phototherapy of formula (FX27) wherein at least one of R 1 and R 120 to R 141 is independently FL or Bm.
  • the invention provides a compound for phototherapy of formula
  • R 1 and R 120 to R 41 is hydrogen or C-, - C 6 alkyl, and optionally hydrogen or - C 3 alkyl, and optionally hydrogen.
  • reference to embodiments wherein o, p, q, r, s, t, u and/or v is equal to 0 refers to compounds where L 120 , L 121 , L 122 , L 123 , L 24 , L 125 , L 26 , and/or L 127 , respectively, is not present; and reference to embodiments wherein o, p, q, r, s, t, u and/or v is equal to 1 refers to compounds where L 120 , L 21 , L 122 , L 123 , L 124 , L 125 , L 126 , and/or L 127 , respectively, is present.
  • the invention provides a compound for phototherapy of formula (FX27) wherein at least one of e, o, p, q, r, s, t, u, and v is independently 0, optionally wherein all of e, o, p, q, r, s, t, u, and v are 0.
  • the invention provides a compound for phototherapy of formula (FX27) wherein at least one of e, o, p, q, r, s, t, u, and v is independently 1 , optionally wherein all of e, o, p, q, r, s, t, u, and v are 1.
  • the invention provides a compound for phototherapy of
  • the invention provides a compound for phototherapy of formula (FX27) wherein each of R 30 to R 133 is independently a hydrogen, -OCF 3 , C 2 -C 20 alkyl, C3-C20 cycloalkyl, C 5 -C 2 o aryi, C 5 -C 20 heteroaryl, C -C 2 o acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C-i-C 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 40 , -SOR 41 , - OSR 42 , -S0 2 OR 43 , -CH 2 (CH 2 OCH 2 ) b CH 2 OH, -P0 3 R 4 R 45 , -OR 46 , -SR 47 , -NR 48 R 49 , -
  • the invention provides a compound for phototherapy being of the formula (FX28):
  • the invention provides a compound for phototherapy being of the formula : (FX29), (FX30), (FX31), (FX32), (FX33), or (FX34):
  • the invention provides a compound for phototherapy being of the formula: (FX35), (FX36), (FX37), or (FX38):
  • the invention provides a compound for phototherapy being of the formula (FX39):
  • each of R 144 to R 148 is independently a hydrogen , -OCF 3 , C r C 20 aikyl, C 3 -C 20 cycloalkyl, C5-C20 aryl, C 5 -C 20 heteroaryl, C C 2 o acyl , C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C C 6 alkoxycarbonyl , halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 40 , -SOR 41 , -OSR 42 , - S0 2 OR 43 , -CH 2 (CH 2 OCH 2 ) b CH 2 OH, -P0 3 R 4 R 45 , -OR 46 , -SR 47 , -N R 48 R 49 , -NR 50 COR 51 , -CN , - CONR 52 R
  • the invention provides a compound for phototherapy being of the formula: (FX40), (FX41 ), (FX42), (FX43), (FX44), or (FX45):
  • the invention provides a compound for phototherapy being of the formula (FX46): (FX46), or a pharmaceutically acceptable salt or ester thereof.
  • the invention provides a compound being of formula (FX55):
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
  • the invention provides a compound being of formula (FX56):
  • each of L 20 , L 121 , L 122 , L 23 , L 124 , L 25 , L 126 and L 127 is independently C C 10 alkylene, C 3 -C 10 cycloalkylene, C 2 -C 10 alkenylene, C 3 -Ci 0 cycloalkenylene, C 2 -Ci 0 alkynylene, ethenylene, ethynyiene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH 2 CH 2 0) b - or -(CHOH) a -; each of W 20 , W 2 , W 122 , W 123 , W 124 , W 125 , W 126 and W 127 is independently a single bond, -(CH 2 )n
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
  • the invention provides a compound being of formula (FX57):
  • each of L 20 , L 2 , L 122 , L 123 , L 24 , L 125 , L 26 and L 127 is independently C C 10 alkylene, C 3 -C 10 cycloalkylene, C 2 -C 10 alkenylene, C 3 -Ci 0 cycloalkenylene, C 2 -C 10 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH 2 CH 2 0) b -, or -(CHOH) a -; each of W 120 , W 21 , W 22 , W 123 , W 24 , W 125 , W 26 and W 127 is independently a single bond, -(CH 2 )n- -(HCCH)n-, -0-
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
  • each of L 120 , L 121 , L 122 , L 23 , L 124 , L 125 , L 126 and L 27 is independently C C 10 alkylene, C 3 -C 10 cycloalkylene, C 2 -C 10 alkenylene, C 3 -C 10 cycloalkenylene, C 2 -C 10 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH 2 CH 2 0) b -, or -(CHOH) a -; each of W 120 , W 121 , W 122 , W 123 , W 24 , W 25 , W 26 and W 127 is independently a single bond, -(CH 2 )n- -(HCCH)n- -0-,
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
  • the invention provides a compound being of formula (FX59):
  • each of L 20 , L 2 , L 22 , L 123 , L 124 , L 125 , L 126 and L 127 is independently C C 10 alkylene, C 3 -C 0 cycloalkylene, C 2 -Ci 0 alkenylene, C 3 -C 0 cycloalkenylene, C 2 -C 10 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH 2 CH 2 0) b - or -(CHOH) a -; each of W 20 , W 12 , W 122 , W 123 , W 124 , W 125 , W 126 and W 127 is independently a single bond, -(
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 27 is an electron donating group and at least one of R 120 to R 127 is an electron withdrawing group.
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 127 is C C 6 alkyl, -OR 46 , -SR 47 , - NR 48 R 49 , or -NR 50 COR 51 ; and wherein at least one of R 120 to R 127 is -CN, -C0 2 R 40 , -S0 2 OR 43 , - CONR 52 R 53 , -COR 54 , -N0 2 , -SOR 41 , -S0 2 R 55 , -P0 3 R 44 R 45 , halo, C C 6 acyl, trihalomethyl, or - S0 2 NR 58 R 59 .
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 127 is -OR 46 , -SR 47 , -NR 48 R 49 , or -NR 50 COR 51 .
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 127 is -NR 48 R 49 , or -NR 50 COR 51 .
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 127 is -CN, - CO 2 R 40 ,-COR 54 , -N0 2 , -S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the invention provides a compound being of formula (FX27) - (FX59),wherein at least one of R 120 to R 127 is -C0 2 R 4 °, - COR -S0 2 NR R or -S0 2 R
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 127 is -NR 48 R 49 , and wherein at least one of R 120 to R 27 is -CO 2 R 40 ,-COR 54 , -S0 2 NR 58 R 59 or -S0 2 R 55 .
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 20 to R 127 and R 144 to R 148 is Bm. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 120 to R 127 and R 144 to R 148 is FL.
  • the invention provides a compound being of formula (FX27) - (FX59), wherein o is 0 and W 20 is a single bond; or wherein p is 0 and W 21 is a single bond; or wherein q is 0 and W 122 is a single bond; or wherein r is 0 and W 123 is a single bond; or wherein s is 0 and W 24 is a single bond; or wherein t is 0 and W 125 is a single bond; or wherein u is 0 and W 126 is a single bond; or wherein v is 0 and W 27 is a single bond.
  • the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of: L 20 with W 120 , L 121 with W 12 , L 122 with W1 22 , L 123 with W 123 , L 24 with W 24 , L 25 with W 25 , L 26 with W 126 , and L 127 with W 127 combine to form: -(CH 2 ) n - -0(CH 2 ) n - -CO(CH 2 ) n - -OCO(CH 2 ) n - -COO(CH 2 ) n - -OCOO(CH 2 ) n -, - N(R 20 )(CH 2 ) n -, -CON(R 22 )(CH 2 ) n -, -N(R 23 )CO(CH 2 ) n -, -OCONR 24 (CH 2 ) n -, -NR 25 COO(CH 2 ) n
  • the invention provides a compound being of formula (FX39) - (FX59), wherein at least one of R 144 to R 48 is -N0 2 , and optionally wherein at least two of R 144 to R 148 are -N0 2 .
  • the invention provides a compound being of formula (FX39) - (FX59), wherein at least one of R 44 to R 148 is -CH 3 .
  • the invention provides a compound being of formula (FX39) - (FX59), wherein at each of R 144 to R 48 is -N0 2 hydrogen, or C C 6 alkyl, optionally hydrogen, or C-
  • the invention provides a compound being of formula (FX39) - (FX59), wherein at least two of R 144 to R 148 are -N0 2 .
  • the present invention includes therapeutic agents for biomedical applications, including phototherapy, comprising purified stereoisomers (e.g., enantiomers and diastereomers), salts (including quarternary salts), and/or ionic forms (e.g., protonated and deprotonated forms) of the compounds of any of formula (FX1) - (FX59), and mixtures thereof.
  • purified stereoisomers e.g., enantiomers and diastereomers
  • salts including quarternary salts
  • ionic forms e.g., protonated and deprotonated forms
  • acidic functional groups and basic functional groups of the compounds of any of formula (FX1) - (FX59) may be in protonated or deprotonated states depending on the molecular environment (e.g., pH, ionic strength, composition, etc.), for example during synthesis, formulation and/or administration
  • the invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar - Ar 3 is independently a group comprising one or more aromatic rings or heterocyclic aromatic rings having one or more electron donating groups and electron withdrawing groups provided as substituents directly or indirectly linked to the aromatic rings or heterocyclic aromatic rings.
  • compounds of the present invention have any one of formula (FX1) - (FX59), wherein at least one of R - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is a C 5 -C 30 aryl or C 5 -C 30 heteroaryl group, optionally a C 5 -C 2 o aryl or C 5 -C 20 heteroaryl group, or C 5 -C 10 aryl or C 5 -C 10 heteroaryl group having at least one electron withdrawing group and at least one electron donating group as substituents.
  • formula (FX1) - (FX59) wherein at least one of R - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is a C 5 -C 30 aryl or C 5 -C 30 heteroaryl group, optionally a C 5 -C 2
  • the electron withdrawing group of the C 5 -C 30 aryl or C 5 -C 30 heteroaryl group is -CN, halo, -C0 2 R 40 , -COR 54 , -N0 2 , -S0 2 R 55 , or -S0 2 NR 58 R 59 .
  • the electron donating group of the C 5 -C 30 aryl or C 5 -C 3 o heteroaryl group is -OR 46 , -SR 47 , -NR 48 R 49 , or -NR 50 COR 51 .
  • the invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is independently a group comprising one or more aromatic rings or heterocyclic aromatic rings having one or more targeting ligands (e.g., Bm) provided as substituents directly or indirectly linked to the aromatic rings or heterocyclic aromatic rings, and optionally having one or more electron donating groups and electron withdrawing groups provided as substituents directly or indirectly linked to the aromatic rings or heterocyclic aromatic rings.
  • targeting ligands e.g., Bm
  • the invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 20 -R 127 , and Ar 1 - Ar 3 is independently a group corresponding to benzene, naphthalene, naphthoquinone,
  • naphthacenedione pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline, optionally having one or more electron donating groups, electron withdrawing groups, or targeting ligands provided as one or more substituents.
  • the invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 - R 127 , and Ar 1 - Ar 3 is independently a group corresponding to a cyanine, indocyanine,
  • the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 1 0 -R 127 , and Ar 1 - Ar 3 is independently benzene or a derivative thereof.
  • the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , 7 and Ar 1 - Ar 3 is independently a polycyclic aromatic group corresponding to naphthalene, diphenylmethane, fluorene, anthracene, phenanthrene, tetracene, naphthoquinone, anthraquinone, naphthacenedione, azulene, aza-azulene or anthracycline.
  • the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is independently a heterocyclic aromatic group having a six member nitrogen-containing ring corresponding to pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, purine, acridine, acridone, or phenanthridine.
  • the invention provides a compound for use in a
  • phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one R 1 - R 3 , R 65 _ R 67 ⁇ R 7o_ R 85 ⁇ R i2o_ R i27 ⁇ a nd Af i _ Af 3 js j nc j epenc i en tiy a heterocyclic aromatic group having a five member nitrogen-containing ring corresponding to pyrrole, pyrazole, indole, isoindole, imidazole, oxazole, thiazole, purine, benzimidazole, or carbazole.
  • the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is
  • heterocyclic aromatic group having a five or six member oxygen-containing ring corresponding to furan, oxazole, benzofuran, dibenzofuran, xanthene, xanthone, flavone, or coumarin.
  • the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is independently a heterocyclic aromatic group having a five member sulfur-containing ring corresponding to thiophene, thiazole, benzothiophene, or dibenzothiophene.
  • the invention includes compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , Ar -Ar 3 , or R 120 -R 127 is independently a dye (abbreviated as "FL"), such as a C 5 -C 30 aryl or C 5 -C 30 heteroaryl chromophore and/or C 5 -C 30 aryl or C 5 -C 30 heteroaryl fluorophore, that is excited upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, optionally selected over the range of 400 nanometers to 900 nanometers.
  • FL dye
  • the invention provides a compound having any one of formula (FX1 ) - (FX59) that functions as a photosensitizer upon exposure to electromagnetic radiation having a first distribution of wavelengths, and wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 1 0 -R 127 , and Ar 1 - Ar 3 is independently a fluorophore that is excited upon exposure to electromagnetic radiation having a second distribution of wavelengths that is different from the first distribution of wavelengths, for example, wherein the first and second distributions of wavelengths correspond to different absorption maxima and, optionally wherein the first and second distributions of wavelengths corresponding to absorption peaks that are not overlapping, or corresponding to absorption maxima in the visible or near IR regions of the spectrum that differ by 20 nanometers or more.
  • At least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , and Ar 1 - Ar 3 is independently a C 5 -C 30 aryl or C5-C30 heteroaryl fluorophore having one or more electron donating groups as substituents, having one or more electron withdrawing groups as substituents, or having both electron donating and electron withdrawing groups as substituents.
  • At least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 20 -R 127 , and Ar 1 - Ar 3 is independently a fluorophore group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an azaazulene, a triphenyl methane dye, an indole, a benzoindole, an in
  • Targeting ligands of the present invention may be indirectly or directly linked to, or non- covalently associated with, the central S-N group of formulas (FX1) - (FX59).
  • the invention includes, for example, compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , or R 20 -R 127 is independently a targeting ligand (abbreviated as "Bm" throughout this description).
  • the invention includes compounds wherein R is Bm and W 1 is -NR 13 CO- -CONR 12 -, -OCONR 14 -, -NR 15 COO- or -NR 16 CONR 17 - ; or R 2 is Bm and W 2 is -NR 3 CO- -CONR 12 -, -OCONR 14 -, -NR 15 COO- or -NR 6 CONR 17 -, or
  • R 3 is Bm and W 3 is -NR 13 CO- -, -CONR 12 - -, -OCONR 14 - -, -NR 5 COO- -, or -NR 16 CONR 17 - -
  • R 120 is Bm and W 20 is -NR 13 CO- -CONR 12 -, -OCONR 14 - -NR 15 COO- or - NR 6 CONR 17 -; or R 121 is Bm and W 12 is -NR 13 CO- -CONR 12 - -OCONR 14 -, -NR 5 COO- or - NR 16 CONR 17 -; or R 122 is Bm and W 122 is -NR 13 CO- -CONR 12 - -OCONR 14 -, -NR 15 COO- or - NR 16 CONR 17 -; or R 123 is Bm and W 123 is -NR 13 CO- -CONR 2 -, -OCONR 14 -,
  • the invention includes compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , or R 20 -R 127 is independently a polypeptide comprising 2 to 30 amino acid units.
  • the invention includes compounds of any one of formula (FX1) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , or R 120 -R 127 is independently an antibody or fragment thereof.
  • the invention includes compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R - R 3 , R 65 - R 67 , R 70 -R 85 , or R 120 -R 127 is independently a polynucleotide comprising 1 to . 50 nucleic acid units.
  • the invention includes compounds of any one of formula (FX1) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , or R 1 0 -R 127 is independently an aptamer.
  • the invention provides compounds of any one of formula (FX1) -
  • each of R - R 33 is independently hydrogen, a C1-C10 cycloalkyl, or a C C 10 alkyl, and optionally wherein each of R 11 - R 33 is hydrogen, a C C 10 cycloalkyl, or a C C 5 alkyl, and optionally wherein each of R 11 - R 33 is hydrogen.
  • the invention provides compounds of any one of formulas (FX1) - (FX59), wherein each of R 40 - R 55 and R 58 - R 6 is independently hydrogen, C C 0 cycloalkyl, or C C 5 alkyl, and optionally hydrogen, or C C 3 alkyl, and optionally hydrogen.
  • the invention provides compounds of any one of formula (FX1) - (FX59), wherein each of R 40 - R 55 and R 58 - R 61 is hydrogen. In an embodiment, the invention provides compounds of any one of formulas (FX1 ) - (FX59), wherein at least one of R 1 , R 2 , and R 3 is independently C 5 -C 2 o aryl or C 5 -C 20 heteroaryl, optionally C 5 -C 10 aryl or C 5 -C 10 heteroaryl.
  • the invention provides compounds useful as optical agents for phototherapeutic methods having any one of formula (FX1) - (FX59), wherein each of R 1 , R 2 , and R 3 is independently hydrogen, C C 2 o alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, - CH(R 62 )C0 2 H, -CH(R 63 )NH 2 , FL a dye, or a targeting Iigand (Bm); wherein at least one of R 1 , R 2 , and R 3 is independently C 5 -C 30 aryl or C 5 -C 30 heteroaryl, and optionally C 5 -C 20 aryl or C 5 -C 20 heteroaryl, or C 5 -C 10 aryl or C 5 -Ci 0 heteroaryl.
  • the invention provides compounds useful as optical agents for phototherapeutic methods having any one of formula (FX1) - (FX59), wherein each of R
  • R 1 is C C 10 alkyl, or C 3 -Ci 0 cycloalkyl, C 5 -C 30 aryl, or C 5 -C 30 heteroaryl wherein optionally the C 5 -C 30 aryl or C 5 -C 20 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent
  • each of R 2 , and R 3 is independently a hydrogen, Ci-C 20 alkyl, C 3 -C 20 cycloalkyl, C 5 -C 30 aryl, C 5 -C 20 heteroaryl, a FL or Bm, wherein optionally the C 5 -C 30 aryl or C 5 -C 20 heteroaryl includes an electron donating group, electron withdrawing group, or a targeting Iigand (Bm) as a substituent.
  • the invention provides compounds useful as optical agents for phototherapeutic methods having any one of formulas (FX1) - (FX59), wherein at least one of R 1 , R 2 , and R 3 is a FL, such as a fluorophore that is excited upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, preferably wavelengths selected over the range of 400 nanometers to 900 nanometers for some applications.
  • a FL such as a fluorophore that is excited upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, preferably wavelengths selected over the range of 400 nanometers to 900 nanometers for some applications.
  • L 1 , L 2 , L 3 and W 1 , W 2 , W 3 groups may be spacer and attaching groups, respectively, for providing an appropriate linkage between R 1 , R 2 , R 3 and the central S-N bond in the compounds of (FX1) - (FX59).
  • the invention provides compounds of any one of formulas (FX1) - (FX59), wherein any one of L , L 2 , and L 3 is independently a spacer moiety for establishing the steric environment between R , R 2 , and/or R 3 and the central S-N group providing useful optical, pharmacokinetic, or targeting properties.
  • the invention provides compounds of any one of formulas (FX1) - (FX59), wherein any one of W , W 2 , and W 3 is independently an attaching moiety for attaching R 1 , R 2 , and/or R 3 directly or indirectly to the central S-N group.
  • the invention provides compounds having formula (FX1) - (FX59), wherein at least one of e, f and g is 0, and optionally all of e, f and g are 0.
  • the invention provides compounds having formula (FX1) - (FX59), and related phototherapy methods, wherein e is 0, and/or f is 0, and/or g is 0.
  • the invention provides compounds useful as optical agents for phototherapeutic methods having any of formulas (FX1 ) - (FX59), wherein at least one of W 1 , W 2 , and W 3 is a single bond, and optionally all of W 1 , W 2 , and W 3 are single bonds.
  • the invention provides compounds having formula (FX1) - (FX59), wherein W 1 is a single bond, and/or W 2 is a single bond, and/or W 3 is a single bond.
  • the invention provides compounds having formula (FX1) - (FX59), and related phototherapy methods, wherein e is 0 and W is a single bond, and/or f is 0 and W 2 is a single bond, and/or g is 0 and W 3 is a single bond.
  • At least one of L - L 3 is independently -(CH 2 ) m - -(HCCH) m -, - (CHOH)m- , or -(CH 2 CH 2 0) m -, wherein each of m is independently an integer selected from the range of 1 to 10.
  • the invention provides compounds of any one of formulas (FX1) - (FX59), wherein at least one of W 1 - W 3 is independently a single bond, -0-, -CO-, - COO- -OCO-, -OCOO-, -NR 1 -, -CONR 12 -, -NR 13 CO- -NR 16 CONR 17 -, or -NR 8 CSNR 19 -.
  • the invention provides compounds of any one of formulas (FX1) - (FX59), wherein at least one of: L 1 with W , L 2 with W 2 , and L 3 with W 3 , combine to form: -(CH 2 ) n - > - 0(CH 2 )n- -CO(CH 2 )n- -OCO(CH 2 ) n -, -COO(CH 2 ) n -, -OCOO(CH 2 )n- -N(R 20 )(CH 2 ) n -, - CON(R 22 )(CH 2 ) n -, -N(R 23 )CO(CH 2 ) n -, -OCONR 24 (CH 2 ) n -, -NR 25 COO(CH 2 ) n -, - NR 26 CONR 7 (CH 2 )n-, or -NR 28 CSNR 29 (CH 2 ) n -.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an azo group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an diaza group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an oxaza group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an azide group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 20 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include a sulfenate group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R - R 3 , R 65 - R 67 , R 70 -R 85 , R 20 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include a thiadiazole group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 20 - R 27 , Ar 1 , Ar 2 , and Ar 3 do not include a cyanate group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an isocyanide group.
  • the invention provides a compound for phototherapy having any one of formula (FX1) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an isocyanate group.
  • the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include an isothiocyanate group.
  • the invention provides a compound for phototherapy having any one of formula (FX1) - (FX59), wherein R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 120 -R 127 , Ar 1 , Ar 2 , and Ar 3 do not include a thiocyanate group.
  • compounds of the invention may optionally include a
  • the invention provides a composition having any one of the formula (FX1 ) - (FX59), wherein at least one of R 1 - R 3 , R 65 - R 67 , R 70 -R 85 , R 20 -R 127 , or L - L 3 is a substituent comprising -(CH 2 OCH 2 ) m- wherein m is an integer selected from the range of 1 to 100.
  • Incorporation of a poly(ethylene glycol) component in some compositions of the invention provides pharmacokinetic, chemical, and/or physical properties useful for bioanalytical, diagnostic and/or phototherapeutic applications.
  • Poly(ethylene glycol) containing compounds of some embodiments of the present invention for example, provide enhanced biocompatibility, low toxicity and suppress immune responses upon administration.
  • Poly(ethylene glycol) containing compounds of some embodiments of the invention facilitate formulation, administration and/or delivery of the compounds, for example, by enhancing solubility.
  • the invention further provides a compound having any one of formula (FX1 ) - (FX59), or a pharmaceutical formulation thereof, for use in an optical imaging, diagnostic, and/or
  • the invention provides an optical agent comprising a pharmaceutically acceptable formulation, wherein at least one active ingredient of the formulation is a compound having any one of formula (FX1 ) - (FX59) provided in a therapeutically effective amount.
  • the invention includes, for example, formulations comprising a compound having any one of formula (FX1 ) - (FX59) and one or more pharmaceutically acceptable carriers or excipients.
  • the invention provides a pharmaceutically acceptable formulation for combination therapy comprising a compound having any one of formula (FX1) - (FX59) and one or more additional diagnostic and/or therapeutic agents, such as anti-cancer agents, anti-inflammatory agents, and/or imaging agents (e.g., optical and/or non-optical imaging agents).
  • additional diagnostic and/or therapeutic agents such as anti-cancer agents, anti-inflammatory agents, and/or imaging agents (e.g., optical and/or non-optical imaging agents).
  • the invention provides methods for a medical phototherapy procedure, such as a phototherapy procedure, wherein the method comprises: (i) administering (e.g., via intravenous or intraarterial injection, oral administration, topical administration, subcutaneous administration, etc.) to a subject a therapeutically or diagnostically effective amount of a compound having any one of formula (FX1 ) - (FX59) and (ii) exposing the administered compound to electromagnetic radiation.
  • the administrating step is carried out under conditions sufficient for contacting the compound with a target tissue or cell, wherein the compound selectively binds to or otherwise preferentially associates with the target tissue or cell.
  • the administered compound is exposed to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, optionally having wavelengths selected over the range of 400 nanometers to 900 nanometers.
  • exposing the administered compound to electromagnetic radiation cleaves a S-N bond of the compound.
  • exposing the administered compound to electromagnetic radiation generates a therapeutically effective amount of photoactivated compound.
  • exposing the administered compound to electromagnetic radiation generates a therapeutically effective amount of reactive species causing localized cell death, inactivation or injury.
  • the medical phototherapy procedure comprises administering, contacting or otherwise targeting the compound to or with a target tissue or cell of the subject, such as a tumor, lesion, site of inflammation, vasculature tissue, or organ.
  • methods of the invention further comprise exposing the administered compound at the target tissue to light having sufficient power, fluence, intensity and/or dose (net number of photons provided to the target tissue) to result in injury, inactivation and/or death to target cells or cells at the target tissue.
  • the medical phototherapy procedure comprises administering, contacting or otherwise targeting the administered compound to or with a target tissue or cell of the subject, such as a tumor, lesion, site of inflammation, vasculature tissue, or an organ.
  • a target tissue or cell of the subject such as a tumor, lesion, site of inflammation, vasculature tissue, or an organ.
  • the target tissue is a tissue type selected from the group consisting of breast, lung, throat, cervical, colon, kidney, stomach, ovarian, testicular, prostate, gastric, esophageal, uterine, endometrial, and pancreatic tissue.
  • exposing the administered compound to electromagnetic radiation generates fluorescence, wherein the medical phototherapy procedure further comprises detecting fluorescence from the administered compound.
  • exposing the administered compound to electromagnetic radiation generates a diagnostically effective amount of fluorescence, for example an amount of fluorescence allowing for optical detection, visualizing and/or imaging of the target tissue.
  • a method of the invention further comprises exposing the administered compound at the target tissue to electromagnetic radiation having sufficient power, fluence, intensity and/or dose (net number of photons provided to the target tissue) to provide optical detection, visualization and/or imaging of the target tissue.
  • a method of the invention further comprises generating an image of the fluorescence from the compound administered to the subject.
  • a method of the invention further comprises visualizing the fluorescence from the compound.
  • the electromagnetic radiation exposed to the compound of any one of formulas (FX1) - (FX59) does not have wavelengths in the X-ray region of the electromagnetic spectrum.
  • the electromagnetic radiation exposed to the compound of any one of formulas (FX1) - (FX59) does not have wavelengths in the ultraviolet region of the electromagnetic spectrum, for example, not including light having wavelengths equal to or less than 380 nanometers.
  • non-ionizing electromagnetic radiation is used in the present methods.
  • the term "non-ionizing electromagnetic radiation" as used herein refers to
  • Figures 1 A - 1 F provide schematic representations of reaction mechanisms for sulfenamide optical agents having an acyclic S-N bond, wherein photoactivation generates reactive species and cell death.
  • Figures 1A and 1 B provide schematic representations of reaction mechanisms for phototherapeutic agents comprising a sulfenamide compound having formula (FX1).
  • Figures 1 C and 1 D provide schematic representations of reaction mechanisms for phototherapeutic agents comprising a sulfenamide compound having formula (FX16).
  • Figures 1 E and 1 F provide schematic representations of reaction mechanisms for phototherapeutic agents comprising a sulfenamide compound having a fused ring chromophore group.
  • Figure 2 provides schemes for the general synthesis of sulfenamide optical agents having an acyclic S-N bond useful in phototherapeutic methods of the present invention.
  • Figures 3A and 3B provide schemes for the preparation of sulfenamide bioconjugate optical agents.
  • Figure 3A provides schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond and peptide targeting ligand useful in phototherapeutic methods of the invention.
  • Figure 3B provides schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond, peptide targeting ligand and phenyl group.
  • Figures 4A and 4B provide schemes for the preparation of sulfenamide optical agents having fused ring chromophore groups, and optionally a peptide targeting ligand.
  • Figure 5 provides cell viability results for control conditions (no photosensitizer) wherein cells were exposed to light in the presence of dimethyl sulfoxide.
  • Figure 6 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX18). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
  • Figure 7 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX49). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
  • Figure 8 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX47). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
  • Figure 9 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX48). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
  • Figure 10 provides general schemes for the syntheses of bioconjugates of sulfenamide optical agents having formula (FX1).
  • Figures 11 A and 11B illustrate examples of coupling reactions useful for synthesis of sulfenamide optical agents of the invention having specific targeting ligands.
  • Figure 12 provides general schemes for the syntheses of bioconjugates of sulfenamide optical agents having formula (FX49).
  • Figures 13A and 13B illustrate examples of coupling reactions useful for synthesis of sulfenamide optical agents of the invention having specific targeting ligands.
  • Figure 14 provides electron spin resonance spectra for the sulfenamide of formula (FX18) for test conditions before, during, and after the exposure of the sulfenamide to light.
  • the spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
  • Figure 15 provides electron spin resonance spectra for the sulfenamide of formula (FX47) for test conditions before, during, and after the exposure of the sulfenamide to light.
  • the spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
  • Figure 16 provides electron spin resonance spectra for the sulfenamide of formula (FX47) for test conditions before, during, and after the exposure of the sulfenamide to light.
  • the spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
  • Figure 17 provides electron spin resonance spectra for the sulfenamide of formula (FX48) for test conditions before, during, and after the exposure of the sulfenamide to light.
  • the spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
  • Figure 18 provides electron spin resonance spectra for the sulfenamide of formula (FX48) for test conditions before, during, and after the exposure of the sulfenamide to light.
  • the spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
  • Figure 19 provides electron spin resonance spectra for the sulfenamide of formula (FX49) for test conditions before, during, and after the exposure of the sulfenamide to light.
  • the spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
  • Figure 20 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX50). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
  • a composition or compound of the invention is isolated or purified.
  • an isolated or purified compound is at least partially isolated or purified as would be understood in the art.
  • the composition or compound of the invention has a chemical purity of 95%, optionally for some applications 99%, optionally for some applications 99.9%, optionally for some applications 99.99%, and optionally for some applications 99.999% pure.
  • Many of the molecules disclosed herein contain one or more ionizable groups, lonizable groups include groups from which a proton can be removed (e.g., -COOH) or added (e.g., amines) and groups which can be quaternized (e.g., amines).
  • the compounds of this invention can contain one or more chiral centers. Accordingly, this invention is intended to include racemic mixtures, diasteromers, enantiomers, tautomers and mixtures enriched in one or more stereoisomer.
  • the scope of the invention as described and claimed encompasses the racemic forms of the compounds as well as the individual enantiomers and non-racemic mixtures thereof.
  • group may refer to a functional group of a chemical compound.
  • Groups of the present compounds refer to an atom or a collection of atoms that are a part of the compound.
  • Groups of the present invention may be attached to other atoms of the compound via one or more covalent bonds.
  • Groups may also be characterized with respect to their valence state.
  • the present invention includes groups characterized as monovalent, divalent, trivalent, etc. valence states.
  • substituted refers to a compound wherein a hydrogen is replaced by another functional group.
  • alkylene and “alkylene group” are used synonymously and refer to a divalent group derived from an alkyl group as defined herein.
  • the invention includes compounds having one or more alkylene groups. Alkylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention may have substituted and/or unsubstituted C C 20 alkylene, C-i-C-io alkylene and C C 5 alkylene groups.
  • the invention includes compounds having one or more cycloalkylene groups. Cycloalkyi groups in some compounds function as attaching and/or spacer groups. Compounds of the invention may have substituted and/or unsubstituted C 3 -C 2 o cycloalkylene, C 3 -C 10 cycloalkylene and C 3 -C 5 cycloalkylene groups.
  • arylene and "arylene group” are used synonymously and refer to a divalent group derived from an aryl group as defined herein.
  • the invention includes compounds having one or more arylene groups.
  • an arylene is a divalent group derived from an aryl group by removal of hydrogen atoms from two intra-ring carbon atoms of an aromatic ring of the aryl group.
  • Arylene groups in some compounds function as attaching and/or spacer groups.
  • Arylene groups in some compounds function as chromophore, fluorophore, aromatic antenna, dye and/or imaging groups.
  • Compounds of the invention include substituted and/or unsubstituted C 3 -C 30 arylene, C 3 -C 20 arylene, C 3 -C 10 arylene and Ci-C 3 arylene groups.
  • heteroarylene and “heteroarylene group” are used synonymously and refer to a divalent group derived from a heteroaryl group as defined herein.
  • the invention includes compounds having one or more heteroarylene groups.
  • a heteroarylene is a divalent group derived from a heteroaryl group by removal of hydrogen atoms from two intra-ring carbon atoms or intra-ring nitrogen atoms of a heteroaromatic or aromatic ring of the heteroaryl group.
  • Heteroarylene groups in some compounds function as attaching and/or spacer groups.
  • Heteroarylene groups in some compounds function as chromophore, aromatic antenna, fluorophore, dye and/or imaging groups.
  • Compounds of the invention include substituted and/or unsubstituted C 3 -C 30 heteroarylene, C 3 -C 2 o heteroarylene, C C-i o heteroarylene and C 3 -C 5 heteroarylene groups.
  • alkenylene and “alkenylene group” are used synonymously and refer to a divalent group derived from an alkenyl group as defined herein.
  • the invention includes compounds having one or more alkenylene groups. Alkenylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention include substituted and/or unsubstituted C 2 -C 20 alkenylene, C 2 -Ci 0 alkenylene and C 2 -C 5 alkenylene groups.
  • cylcoalkenylene and “cylcoalkenylene group” are used synonymously and refer to a divalent group derived from a cylcoalkenyl group as defined herein.
  • the invention includes compounds having one or more cylcoalkenylene groups. Cycloalkenylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention include substituted and/or unsubstituted C 3 -C 20 cylcoalkenylene, C 3 -C-i 0 cylcoalkenylene and C 3 -C 5 cylcoalkenylene groups.
  • alkynylene and “alkynylene group” are used synonymously and refer to a divalent group derived from an alkynyl group as defined herein.
  • the invention includes compounds having one or more alkynylene groups. Alkynylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention include substituted and/or unsubstituted C 2 -C 2 o alkynylene, C 2 -C 10 alkynylene and C 2 -C 5 alkynylene groups.
  • halo refers to a halogen group such as a fluoro (-F), chloro (-CI), bromo (-Br), iodo (-I) or astato (-At).
  • heterocyclic refers to ring structures containing at least one other kind of atom, in addition to carbon, in the ring. Examples of such heteroatoms include nitrogen, oxygen and sulfur. Heterocyclic rings include heterocyclic alicyclic rings and heterocyclic aromatic rings.
  • heterocyclic rings include, but are not limited to, pyrrolidinyl, piperidyl, imidazolidinyl, tetrahydrofuryl, tetrahydrothienyl, furyl, thienyl, pyridyl, quinolyl, isoquinolyl, pyridazinyl, pyrazinyl, indolyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, pyridinyl, benzoxadiazolyl, benzothiadiazolyl, triazolyl and tetrazolyl groups. Atoms of heterocyclic rings can be bonded to a wide range of other atoms and functional groups, for example, provided as substituents.
  • carbocyclic refers to ring structures containing only carbon atoms in the ring. Carbon atoms of carbocyclic rings can be bonded to a wide range of other atoms and functional groups, for example, provided as substituents..
  • alicyclic ring refers to a ring, or plurality of fused rings, that is not an aromatic ring. Alicyclic rings include both carbocyclic and heterocyclic rings.
  • aromatic ring refers to a ring, or a plurality of fused rings, that includes at least one aromatic ring.
  • aromatic ring includes aromatic rings comprising carbon, hydrogen and heteroatoms.
  • Aromatic ring includes carbocyclic and heterocyclic aromatic rings.
  • Aromatic rings are components of aryl groups.
  • fused ring or “fused ring structure” refers to a plurality of alicyclic and/or aromatic rings provided in a fused ring configuration, such as fused rings that share at least two intra ring carbon atoms and/or heteroatoms.
  • alkoxyalkyl refers to a substituent of the formula alkyl-O-alkyl.
  • polyhydroxyalkyl refers to a substituent having from 2 to 12 carbon atoms and from 2 to 5 hydroxyl groups, such as the 2,3-dihydroxypropyi, 2,3,4- trihydroxybutyl or 2,3,4,5-tetrahydroxypentyl residue.
  • polyalkoxyalkyl refers to a substituent of the formula alkyl- (alkoxy)n-alkoxy wherein n is an integer from 1 to 10, preferably 1 to 4, and more preferably for some embodiments 1 to 3.
  • Amino acids include glycine, alanine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tryptophan, asparagine, glutamine, glycine, serine, threonine, serine, Threonine, asparagine, glutamine, tyrosine, cysteine, lysine, arginine, histidine, aspartic acid and glutamic acid.
  • reference to "a side chain residue of a natural a-amino acid” specifically includes the side chains of the above-referenced amino acids.
  • Aikyl groups include straight-chain, branched and cyclic alkyl groups.
  • AlkyI groups include those having from 1 to 30 carbon atoms.
  • Alkyl groups include small alkyl groups having 1 to 3 carbon atoms.
  • Alkyl groups include medium length alkyl groups having from 4-10 carbon atoms.
  • Alkyl groups include long alkyl groups having more than 10 carbon atoms, particularly those having 10-30 carbon atoms.
  • the term cycloalkyi specifically refers to an alky group having a ring structure such as ring structure comprising 3-30 carbon atoms, optionally 3-20 carbon atoms and optionally 2 - 10 carbon atoms, including an alkyl group having one or more rings.
  • Cycloalkyi groups include those having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring(s) and particularly those having a 3-, 4-, 5-, 6-, or 7-member ring(s).
  • the carbon rings in cycloalkyi groups can also carry alkyl groups.
  • Cycloalkyi groups can include bicyclic and tricycloalkyl groups.
  • Alkyl groups are optionally substituted.
  • Substituted alkyl groups include among others those which are substituted with aryl groups, which in turn can be optionally substituted.
  • alkyl groups include methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, n-butyl, s-butyl, t-butyl, cyclobutyl, n-pentyl, branched- pentyl, cyclopentyl, n-hexyl, branched hexyl, and cyclohexyl groups, all of which are optionally substituted.
  • Substituted alkyl groups include fully halogenated or semihalogenated alkyl groups, such as alkyl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms.
  • Substituted alkyl groups include fully fluorinated or semifluorinated alkyl groups, such as alkyl groups having one or more hydrogens replaced with one or more fluorine atoms.
  • An alkoxy group is an alkyl group that has been modified by linkage to oxygen and can be represented by the formula R-0 and can also be referred to as an alkyl ether group.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, butoxy and heptoxy.
  • Alkoxy groups include substituted alkoxy groups wherein the alky portion of the groups is substituted as provided herein in connection with the description of alkyl groups. As used herein MeO- refers to CH 3 0-
  • Alkenyl groups include straight-chain, branched and cyclic alkenyl groups. Alkenyl groups include those having 1 , 2 or more double bonds and those in which two or more of the double bonds are conjugated double bonds. Alkenyl groups include those having from 2 to 20 carbon atoms. Alkenyl groups include small alkenyl groups having 2 to 3 carbon atoms. Alkenyl groups include medium length alkenyl groups having from 4-10 carbon atoms. Alkenyl groups include long alkenyl groups having more than 10 carbon atoms, particularly those having 10-20 carbon atoms. Cycloalkenyl groups include those in which a double bond is in the ring or in an alkenyl group attached to a ring.
  • cycloalkenyl specifically refers to an alkenyl group having a ring structure, including an alkenyl group having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring(s) and particularly those having a 3-, 4-, 5-, 6- or 7-member ring(s).
  • cycloalkenylgroups can also carry alkyl groups.
  • Cycloalkenylgroups can include bicyclic and tricyclic alkenyl groups.
  • Alkenyl groups are optionally substituted.
  • Substituted alkenyl groups include among others those which are substituted with alkyl or aryl groups, which groups in turn can be optionally substituted.
  • alkenyl groups include ethenyl, prop-1 -enyl, prop-2-enyl, cycloprop-1 -enyl, but-1 -enyl, but-2-enyl, cyclobut-1 -enyl, cyclobut-2-enyl, pent-1 -enyl, pent-2-enyl, branched pentenyl, cyclopent-1 -enyl, hex-1 -enyl, branched hexenyl, cyclohexenyl, all of which are optionally substituted.
  • Substituted alkenyl groups include fully halogenated or semihalogenated alkenyl groups, such as alkenyl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms.
  • Substituted alkenyl groups include fully fluorinated or semifluorinated alkenyl groups, such as alkenyl groups having one or more hydrogen atoms replaced with one or more fluorine atoms.
  • Aryl groups include groups having one or more 5-, 6- or 7- member aromatic rings, including heterocyclic aromatic rings.
  • heteroaryl specifically refers to aryl groups having at least one 5-, 6- or 7- member heterocyclic aromatic rings.
  • Aryl groups can contain one or more fused aromatic rings, including one or more fused heteroaromatic rings, and/or a combination of one or more aromatic rings and one or more nonaromatic rings that may be fused or linked via covalent bonds.
  • Heterocyclic aromatic rings can include one or more N, O, or S atoms in the ring.
  • Heterocyclic aromatic rings can include those with one, two or three N atoms, those with one or two O atoms, and those with one or two S atoms, or combinations of one or two or three N, O or S atoms.
  • Aryl groups are optionally substituted.
  • Substituted aryl groups include among others those which are substituted with alkyl or alkenyl groups, which groups in turn can be optionally substituted.
  • aryl groups include phenyl, biphenyl groups, pyrrolidinyl, imidazolidinyl, tetrahydrofuryl, tetrahydrothienyl, furyl, thienyl, pyridyl, quinolyl, isoquinolyl, pyridazinyl, pyrazinyl, indolyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, pyridinyl, benzoxadiazolyl, benzothiadiazolyl, and naphthyl groups, all of which are optionally substituted.
  • Substituted aryl groups include fully halogenated or semihalogenated aryl groups, such as aryl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms.
  • Substituted aryl groups include fully fluorinated or semifluorinated aryl groups, such as aryl groups having one or more hydrogens replaced with one or more fluorine atoms.
  • Aryl groups include, but are not limited to, aromatic group-containing or heterocylic aromatic group-containing groups corresponding to any one of the following: benzene, naphthalene, naphthoquinone,
  • diphenylmethane fluorene, anthracene, anthraquinone, phenanthrene, tetracene, tetracenedione, pyridine, quinoline, isoquinoline, indoles, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furans, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene or anthracycline.
  • a group corresponding to the groups listed above expressly includes an aromatic or heterocyclic aromatic group, including monovalent, divalent and polyvalent groups, of the aromatic and heterocyclic aromatic groups listed herein are provided in a covalently bonded configuration in the compounds of the invention at any suitable point of attachment.
  • aryl groups contain between 5 and 30 carbon atoms.
  • aryl groups contain one aromatic or heteroaromatic six-membered ring and one or more additional five- or six-membered aromatic or heteroaromatic ring.
  • aryl groups contain between five and eighteen carbon atoms in the rings.
  • Aryl groups optionally have one or more aromatic rings or heterocyclic aromatic rings having one or more electron donating groups, electron withdrawing groups and/or targeting ligands provided as substituents.
  • Arylalkyl groups are alkyl groups substituted with one or more aryl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted.
  • Specific alkylaryl groups are phenyl-substituted alkyl groups, e.g., phenylmethyl groups.
  • Alkylaryl groups are alternatively described as aryl groups substituted with one or more alkyl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted.
  • Specific alkylaryl groups are alkyl-substituted phenyl groups such as methylphenyl.
  • Substituted arylalkyl groups include fully halogenated or semihalogenated arylalkyl groups, such as arylalkyl groups having one or more alkyl and/or aryl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms.
  • any of the groups described herein which contain one or more substituents do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible.
  • the compounds of this invention include all stereochemical isomers arising from the substitution of these compounds.
  • Optional substitution of alkyl groups includes substitution with one or more alkenyl groups, aryl groups or both, wherein the alkenyl groups or aryl groups are optionally substituted.
  • Optional substitution of alkenyl groups includes substitution with one or more alkyl groups, aryl groups, or both, wherein the alkyl groups or aryl groups are optionally substituted.
  • Optional substitution of aryl groups includes substitution of the aryl ring with one or more alkyl groups, alkenyl groups, or both, wherein the alkyl groups or alkenyl groups are optionally substituted.
  • Optional substituents for any alkyl, alkenyl and aryl group includes substitution with one or more of the following substituents, among others:
  • halogen including fluorine, chlorine, bromine or iodine
  • R is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted;
  • R is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted;
  • each R independently of each other R, is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted; and where R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
  • each R independently of each other R, is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted; and where R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
  • each R independently of each other R, is a hydrogen, or an alkyl group, or an acyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, phenyl or acetyl group, all of which are optionally substituted; and where R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
  • R is hydrogen or an alkyl group or an aryl group and more specifically where R is hydrogen, methyl, ethyl, propyl, butyl, or a phenyl group, which are optionally substituted;
  • R is an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group, all of which are optionally substituted;
  • each R independently of each other R, is a hydrogen, or an alkyl group, or an aryl group all of which are optionally substituted and wherein R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
  • R is H, an alkyl group, an aryl group, or an acyl group all of which are optionally substituted.
  • R can be an acyl yielding -OCOR" where R" is a hydrogen or an alkyl group or an aryl group and more specifically where R" is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted.
  • Specific substituted alkyl groups include haloalkyl groups, particularly trihalomethyl groups and specifically trifluoromethyl groups.
  • Specific substituted aryl groups include mono-, di-, tri, tetra- and pentahalo-substituted phenyl groups; mono-, di-, tri-, tetra-, penta-, hexa-, and hepta- halo-substituted naphthalene groups; 3- or 4-halo-substituted phenyl groups, 3- or 4-alkyl- substituted phenyl groups, 3- or 4-alkoxy-substituted phenyl groups, 3- or 4-RCO-substituted phenyl, 5- or 6-halo-substituted naphthalene groups.
  • substituted aryl groups include acetylphenyl groups, particularly 4-acetylphenyl groups; fluorophenyl groups, particularly 3-fluorophenyl and 4-fluorophenyl groups; chlorophenyl groups, particularly 3-chlorophenyl and 4- chlorophenyl groups; methylphenyl groups, particularly 4-methylphenyl groups; and
  • methoxyphenyl groups particularly 4-methoxyphenyl groups.
  • substituents it is understood that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible.
  • the compounds of this invention include all stereochemical isomers arising from the substitution of these compounds.
  • Pharmaceutically acceptable salts comprise pharmaceutically-acceptable anions and/or cations.
  • pharmaceutically acceptable salt can refer to acid addition salts or base addition salts of the compounds in the present disclosure.
  • a pharmaceutically acceptable salt is any salt which retains at least a portion of the activity of the parent compound and does not impart significant deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered.
  • Pharmaceutically acceptable salts include metal complexes and salts of both inorganic and organic acids.
  • Pharmaceutically acceptable salts include metal salts such as aluminum, calcium, iron, magnesium, manganese and complex salts.
  • Pharmaceutically acceptable salts include, but are not limited to, acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, -32-cilexetil, citric, edetic, edisylic, estolic, esyl, esylic, formic, fumaric, gluceptic, gluconic, glutamic, glycolic,
  • acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, -32-
  • glycolylarsanilic hexamic, hexylresorcjnoic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, lactobionic, maleic, malic, malonic, mandelic,
  • Pharmaceutically acceptable salts may be derived from amino acids, including but not limited to cysteine.
  • Pharmaceutically-acceptable cations include among others, alkali metal cations (e.g., Li + , Na + , K + ), alkaline earth metal cations (e.g., Ca 2+ , Mg 2+ ), non-toxic heavy metal cations and ammonium (NH 4 + ) and substituted ammonium (N(R') 4 + , where R' is hydrogen, alkyl, or substituted alkyl, i.e., including, methyl, ethyl, or hydroxyethyl, specifically, trimethyl ammonium, triethyl ammonium, and triethanol ammonium cations).
  • alkali metal cations e.g., Li + , Na + , K +
  • alkaline earth metal cations e.g., Ca 2+ , Mg 2+
  • NH 4 + non-toxic heavy metal cations and ammonium
  • N(R') 4 + substituted ammonium
  • R' is hydrogen,
  • Pharmaceutically- acceptable anions include among other halides (e.g., CI “ , Br “ ), sulfate, acetates (e.g., acetate, trifluoroacetate), ascorbates, aspartates, benzoates, citrates, and lactate.
  • the compounds of this invention can contain one or more chiral centers. Accordingly, this invention is intended to include racemic mixtures, diasteromers, enantiomers, tautomers and mixtures enriched in one or more stereoisomer.
  • the scope of the invention as described and claimed encompasses the racemic forms of the compounds as well as the individual enantiomers and non-racemic mixtures thereof.
  • inflammation generally refers to a biological response of tissues to harmful stimuli, such as pathogens, damaged cells, irritants, etc. Inflammation can be either acute or chronic. Acute inflammation is an initial response of the body to harmful stimuli and can be achieved by the increased movement of plasma and leukocytes from the blood into injured tissues. An inflammatory response can involve the local vascular system, the immune system, and/or various cells within the injured tissue. Prolonged inflammation, referred to as chronic inflammation, can lead to a progressive shift in the type of cells which are present at the site of inflammation can be characterized by simultaneous destruction and healing of the tissue from the inflammatory process.
  • amino acid comprises naturally occurring amino acids as well as non-naturally occurring amino acids, including amino acid analogs and derivatives.
  • amino acid analogs and derivatives include, for example, naturally occurring proteogenic L-amino acids; D-amino acids; chemically modified amino acids such as amino acid analogs and derivatives; naturally occurring non-proteogenic amino acids, and chemically synthesized compounds having properties known in the art to be characteristic of amino acids.
  • nucleic acid generally refers to a molecule or strand of DNA, RNA, or derivatives or analogs thereof including one or more nucleobases. Nucleobases comprise purine or pyrimidine bases typically found in DNA or RNA (e.g., adenine, guanine, thymine, cytosine, and/or uracil). The term “nucleic acid” also comprises oligonucleotides and polynucleotides. Nucleic acids may be single-stranded molecules, or they may be double-, triple- or quadruple-stranded molecules that may comprise one or more complementary strands of a particular molecule.
  • Nucleic acid includes artificial nucleic acids including peptide nucleic acids, morpholino nucleic acids, glycol nucleic acids and threose nucleic acids. Artificial nucleic acids may be capable of nucleic acid hybridization.
  • sequence means the linear order in which monomers occur in a polymer, the order of amino acids in a polypeptide or the order of nucleotides in a polynucleotide for example.
  • sequence means the linear order in which monomers occur in a polymer, the order of amino acids in a polypeptide or the order of nucleotides in a polynucleotide for example.
  • peptide and polypeptide are used synonymously in the present description, and refer to a class of compounds comprising of amino acid residues chemically bonded together by amide bonds (or peptide bonds), regardless of length, functionality, environment, or associated molecule(s).
  • Peptides and polypeptides are polymeric compounds comprising at least two amino acid residues or modified amino acid residues. Modifications can be naturally occurring or non- naturally occurring, such as modifications generated by chemical synthesis.
  • Modifications to amino acids in peptides include, but are not limited to, phosphorylation, glycosylation, lipidation, prenylation, sulfonation, hydroxylation, acetylation, methionine oxidation, alkylation, acylation, carbamylation, iodination and the addition of cofactors.
  • Peptides include proteins and further include compositions generated by degradation of proteins, for example by proteolyic digestion. Peptides and polypeptides can be generated by substantially complete digestion or by partial digestion of proteins.
  • Polypeptides comprising 2 to 100 amino acid units, optionally for some embodiments 2 to 50 amino acid units and, optionally for some embodiments 2 to 20 amino acid units can be used as polypeptide targeting ligands in the invention, for example, where the polypepetide preferentially binds to proteins, peptides or other biomolecules expressed, or otherwise generated by, a target tissue, such as a tumor, precancerous tissue, site of inflammation or other lesion.
  • the polypeptide is at least four amino acid residues in length and can range up to a full-length protein.
  • Protein refers to a class of compounds comprising one or more polypeptide chains and/or modified polypeptide chains. Proteins can be modified by naturally occurring processes such as post-translational modifications or co-translational modifications. Exemplary post- translational modifications or co-translational modifications include, but are not limited to, phosphorylation, glycosylation, lipidation, prenylation, sulfonation, hydroxylation, acetylation, methionine oxidation, the addition of cofactors, proteolysis, and assembly of proteins into macromolecular complexes. Modification of proteins can also include non-naturally occurring derivatives, analogues and functional mimetics generated by chemical synthesis. Exemplary derivatives include chemical modifications such as alkylation, acylation, carbamylation, iodination or any modification that derivatizes the protein.
  • polynucleotide and “oligonucleotide” are used interchangeably and refer to a class of compounds composed of nucleic acid residues chemically bonded together.
  • the invention provides optical agents having an oligonucleotide or polynucleotide targeting ligand which comprises a plurality of nucleic acid residues, such as DNA or RNA residues, and/or modified nucleic acid residues that preferentially binds to proteins, peptides or other biomolecules expressed, or otherwise generated by, a target tissue, such as a tumor, precancerous tissue, site of inflammation or other lesion.
  • Oligo- or polynucleotide targeting ligands include, for example, oligo- or poly-nucleotides comprising 2 to 100 nucleic acid units, optionally for some embodiments 2 to 50 nucleic acid units and, optionally for some embodiments 2 to 20 nucleic acid units, and optionally for some embodiments 2 to 10 nucleic acid units.
  • Polypeptide and oligonucleotide include a polymer of at least two nucleotides joined together by phosphodiester bonds and may consist of either ribonucleotides or
  • aptamer refers to an oligo- or poly-nucleotide or polypeptide that binds to, or otherwise selectively or preferentially associates with, a specific target molecule.
  • the invention provides optical agents having an aptamer targeting ligand that preferentially binds to proteins, peptides or other biomolecules expressed, or otherwise generated by, a target tissue, such as a tumor, precancerous tissue, site of inflammation or other lesion.
  • Peptidomimetic refers to a molecule having activity, including biological activity, that resembles that of a polypeptide or is substantially the same as a polypeptide.
  • Morphine for example, is a peptidomimetic of endorphin peptide.
  • a peptidomimetic is a small protein-like polymer designed to mimic the functionality of a peptide.
  • Peptidomimetics useful as targeting ligands for some compounds of the invention in the present invention include peptoids and ⁇ -peptides. The composition and biological activity of peptidomimetics and use of
  • attaching moiety refers to a component provided to attach any of R 1 , R 2 or R 3 directly or indirectly to central sulfenamide group in compounds of the invention.
  • L , L 2 , L 3 , W 1 , W 2 and W 3 in formulas (FX1 ) - (FX59) are an attaching moieties.
  • an “electron withdrawing group” refers to a chemical group that draws electrons or electron density from a center, such a C 5 -C 30 aryl or C 5 -C 30 heteroaryl of the sulfenamide compounds of the invention.
  • the electron withdrawing group(s) are independently selected from cyano (-CN), carbonyl (-CO), carboxylate (-C0 2 R a ), halo (-F, -CI, -Br, -I, -At), carbamate (-CONR R c ), acyl (-COR d ), nitro (-N0 2 ), sulfinyl (-SOR e ), sulfonyl (-S0 2 R f , -S0 2 OR 9 , and -POaR 1 ⁇ , wherein in the context of this description, R a - R' are independently selected to enhance biological and/or physiochemical properties of the optical agents of the invention.
  • R a - R 1 are independently selected from any one of a hydrogen atom, an anionic functional group (e.g., carboxylate, sulfonate, sulfate, phosphonate or phosphate) and a hydrophilic functional group (e.g., hydroxyl, carboxyl, sulfonyl, sulfonato or phosphonato).
  • R a - R 1 are independently selected from hydrogen, d.
  • the EWG(s) are independently selected from is -CN, halo, -C0 2 R A - COR B , -N0 2 , -S0 2 R°, or -S0 2 NR D R E , wherein each of R A - R E is independently H or d - C 10 alkyl.
  • an EWG is located at the terminus of a substituent arm of a C 5 -C 30 aryl or C 5 -C 30 heteroaryl of the sulfenamide compounds of formulas (FX1 ) - (FX59).
  • an “electron donating group” refers to a chemical group that releases electrons or electron density to a center, such as a C 5 -C 30 aryl or C 5 -C 30 heteroaryl of the sulfenamide compounds of the invention.
  • the electron donating group(s) are independently selected from C C 10 alkyl, C 5 -C 10 aryl, -(CH 2 ) z OH, -OR J , - SR K , -NR'RTM, -N(R N )COR°, and -P(R P ), wherein in the context of this description, R J - R P are independently selected to enhance biological and/or physiochemical properties of the optical agents of the invention and wherein z is selected from the range of 1 to 1 0.
  • R J - R P are independently selected from any one of a hydrogen atom, an anionic functional group (e.g., carboxylate, sulfonate, sulfate, phosphonate or phosphate) and a hydrophilic functional group (e.g., hydroxyl, carboxyl, sulfonyl, sulfonato or phosphonato).
  • an anionic functional group e.g., carboxylate, sulfonate, sulfate, phosphonate or phosphate
  • a hydrophilic functional group e.g., hydroxyl, carboxyl, sulfonyl, sulfonato or phosphonato.
  • an EDG is located at the terminus of a substituent arm of a C 5 -C 30 aryl or C 5 -C 30 heteroaryl of the sulfenamide compounds of formulas (FX1) - (FX59) of the invention.
  • two substituents, such as EDG and EWG substituents, on a compound of the invention can act in what is known as a "push-pull" arrangement.
  • the electron density of the compound or a portion thereof, such as an aryl or heteroaryl group is polarized due in part to the location of an EWG and EDG on the compound.
  • an EWG is positioned at a terminus of a substituent arm of the structure and an EDG is positioned at a terminus of a different substituent arm of the structure.
  • an EWG is positioned at one end of a ⁇ bond and an EDG is positioned at the other end of a ⁇ bond.
  • an EWG is positioned para- to an EDG in a six-membered ring structure.
  • an EWG is positioned trans- to an EDG in an alkylene structure.
  • compounds having the "push-pull" arrangement exhibit a shift in the optical absorbance and emission spectrum as compared to compounds not having the "push-pull" arrangement.
  • optical agent generally refers to compounds, compositions, preparations, and/or formulations that absorb, emit, or scatter electromagnetic radiation of wavelength generally in the range of 350 - 1300 nanometers, within a biologically relevant environment or condition.
  • optical agents of the invention when excited by electromagnetic radiation, undergo emission via fluorescence or phosphorescence pathways. These pathways are useful for diagnostic imaging, visualization, or organ function monitoring. Compounds belonging to this class are commonly referred to as “optical imaging agents” or “optical contrast agents.”
  • optical agents of the invention absorb electromagnetic radiation and undergo photochemical reactions such as photofragmentation of one or more photolabile bonds to generate reactive species such as nitrenes, carbene, free radicals, ions, excited species, etc.
  • photosensitizers refers to a phototherapeutic agent or a component thereof providing for photoactivation, for example, photoactivation resulting in generation of reactive species that locally kill, injure, inactivate or otherwise degrade cells (e.g., cancer cells, tumor cells, non-cancer cells, etc.).
  • Photosensitizers of some embodiments undergo photoactivation that initiates bond cleavage reactions, such as photolysis and/or nitrogen extrusion reactions, thereby generating reactive species capable of causing localized cell death or injury.
  • Optical agents include Type 1 and Type 2 phototherapeutic agents.
  • Optical agents include, but are not limited to, phototherapeutic agents (Type 1 and 2), photosensitizers, imaging agents, dyes, detectable agents, photosensitizer agents, photoactivators, and photoreactive agents; and conjugates, complexes, and derivatives thereof.
  • chromophore is a compound or functional group of a compound that results in absorption of electromagnetic radiation, preferably for some applications electromagnetic radiation having wavelengths in the UV (e.g. 200 nm to 350 nm) or visible (e.g. 350 nm to 750 nm) of the electromagnetic spectrum.
  • a fluorophore is a compound or functional group of a compound that results in absorption of electromagnetic radiation and subsequent fluorescence.
  • incorporation of a fluorophore results in compounds of the invention that absorb electromagnetic radiation and generate fluorescence having wavelengths in the UV (e.g. 200 nm to 350 nm) or visible (e.g. 350 nm to 750 nm) of the electromagnetic spectrum.
  • incorporation of a fluorophore results in compounds having an appreciable quantum yield for fluorescence, such as a quantum yield over the range of 0.001 to 1 , 0.01 to 1 , optionally 0.1 to 1 .
  • Optical agents of the present invention can contain fluorophores. Fluorophores can be functional groups in a molecule which absorb electromagnetic radiation of first specific
  • fluorophores emit energy in the visible (e.g. 350 nm to 750 nm) and NIR regions (e.g., 750 - 1300nm) of the electromagnetic spectrum.
  • the term "luminescence” refers to the emission of electromagnetic radiation from excited electronic states of atoms or molecules.
  • Luminescence generally refers to electromagnetic radiation emission, such as photoluminescence, chemiluminescence, and electrochemiluminescence, among others.
  • photoluminescence including fluorescence and phosphorescence
  • the excited electronic state is created by the absorption of electromagnetic radiation.
  • Luminescence detection involves detection of one or more properties of the
  • luminescence or associated luminescence process can include intensity, excitation and/or emission spectrum, polarization, lifetime, and energy transfer, among others. These properties can also include time-independent (steady-state) and/or time-dependent (time- resolved) properties of the luminescence.
  • Representative luminescence techniques include fluorescence intensity (FLINT), fluorescence polarization (FP), fluorescence resonance energy transfer (FRET), fluorescence lifetime (FLT), total internal reflection fluorescence (TIRF), fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), and bioluminescence resonance energy transfer (BRET), among others.
  • the wavelength of radiation be non-ionizing and be such that it excites the optical agent.
  • This excitation can cause a bond of the molecule to break and can lead to creation of one or more appropriate radical(s).
  • This excitation can also cause the molecule to emit part of the absorbed energy at a different wavelength.
  • emission can be detected using fluorometric techniques as described above.
  • One skilled in the art can readily determine the most appropriate treatment and optional detection technique based, at least in part, on the specific phototherapeutic agent(s) administered and/or the particular use (e.g., tissue to be treated).
  • Optical condition refers to one or more of the following: the fluorescence quantum yield, fluorescence intensity, fluorescence excitation wavelength, wavelength distribution or spectrum, emission wavelength, wavelength distribution or spectrum, Stokes shift, color, reflectance, phosphorescence, chemiluminescence, scattering, and/or other observable and/or measurable spectral property or phenomenon.
  • Phototherapy procedure refers to a therapeutic procedure involving administration of a phototherapeutic agent to a patient followed by subsequent excitation by exposure to applied electromagnetic radiation, such as electromagnetic radiation having wavelengths in the visible and/or near IR region of the electromagnetic spectrum. Such wavelengths can be in the range of 350 - 1300 nanometers, so as to generate a therapeutically effective amount of excited phototherapeutic agent.
  • Phototherapy includes, but is not limited to, photodynamic therapy.
  • phototherapy includes procedures involving administration of Type 1 and/or Type 2 phototherapeutic agents, optionally further including administration of one or more additional therapeutic agents.
  • a detectable optical signal may be, for example, an observable change in absorbance, reflectance, phosphorescence, chemiluminescence, scattering, or other spectral property.
  • tumor-specific agent refers to a compound or composition, such as an optical agent, that preferentially accumulates in a tumor at a higher level than normal tissue regardless of the particular mechanism of uptake in the tumors, for example, receptor mediated or enhanced permeability and retention (EPR).
  • Optical agents of the invention include tumor-specific agents, including tumor specific phototherapy agents, for example having a targeting ligand providing specificity in the administration, delivery and/or binding to tumor tissue.
  • targeting ligand refers to a chemical group and/or substituent having functionality for targeting a compound of any one of formula (FX1 ) - (FX59) to an anatomical and/or physiological site of a patient, such as a selected cell, tissue or organ.
  • a targeting ligand is characterized as a ligand that selectively or preferentially binds to a specific biological site(s) (e.g., enzymes, receptors, etc.) and/or biological surface(s) (e.g., membranes, fibrous networks, etc.).
  • the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is an amino acid, or a polypeptide comprising 2 to 30 amino acid units. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a mono- or polysaccharide comprising 1 to 50 carbohydrate units. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a mono-, oligo- or poly-nucleotide comprising 1 to 50 nucleic acid units.
  • the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a protein, an enzyme, a carbohydrate, a peptidomimetic, a glycomimetic, a glycopeptide, a glycoprotein, a lipid, an antibody (polyclonal or monoclonal), or fragment thereof.
  • the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is an aptamer.
  • the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a drug, a hormone, steroid or a receptor.
  • each occurrence of Bm in the compounds of (FX1 ) - (FX59) is
  • each occurrence of Bm in the compounds of (FX1 ) - (FX59) is independently integrin, selectin, vascular endothelial growth factor, fibrin, tissue plasminogen, thrombin, LDL, HDL, Sialyl LewisX or a mimic thereof, or an atherosclerotic plaque binding molecule.
  • biomolecule can be a targeting ligand (Bm).
  • Bm is a targeting ligand, optionally providing molecular recognition functionality.
  • the targeting ligand is a particular region of the compound that is recognized by, and binds to, a target site on an organ, tissue, tumor or cell.
  • Targeting ligands are often, but not always, associated with biomolecules or fragments thereof which include, but are not limited to, hormones, amino acids, peptides, peptidomimetics, proteins, nucleosides, nucleotides, nucleic acids, enzymes, carbohydrates, glycomimetics, lipids, albumins, mono- and polyclonal antibodies, receptors, inclusion compounds such as cyclodextrins, and receptor binding molecules.
  • Targeting ligands for use in the invention can also include synthetic polymers. Examples of synthetic polymers that are useful for targeting ligands include polyaminoacids, polyols, polyamines, polyacids,
  • oligonucleotides oligonucleotides, aborols, dendrimers, and aptamers.
  • useful targeting ligands can include integrin, selectin, vascular endothelial growth factor, fibrin, tissue plasminogen activator, thrombin, LDL, HDL, Sialyl LewisX and its mimics, and atherosclerotic plaque binding molecules.
  • targeting ligands include, but are not limited to: steroid hormones for the treatment of breast and prostate lesions; whole or fragmented somatostatin, bombesin, and neurotensin receptor binding molecules for the treatment of neuroendocrine tumors; whole or fragmented cholecystekinin receptor binding molecules for the treatment of lung cancer; whole or fragmented heat sensitive bacterioendotoxin (ST) receptor and carcinoembryonic antigen (CEA) binding molecules for the treatment of colorectal cancer; dihydroxyindolecarboxylic acid and other melanin producing biosynthetic intermediates for the treatment of melanoma; whole or fragmented integrin receptor and atherosclerotic plaque binding molecules for the treatment of vascular diseases; and whole or fragmented amyloid plaque binding molecules for the treatment of brain lesions.
  • ST heat sensitive bacterioendotoxin
  • CEA carcinoembryonic antigen
  • Bm if present, is selected from heat-sensitive bacterioendotoxin receptor binding peptide, carcinoembryonic antigen antibody (anti-CEA), bombesin receptor binding peptide, neurotensin receptor binding peptide, cholecystekinin receptor binding peptide, somastatin receptor binding peptide, ST receptor binding peptide, neurotensin receptor binding peptide, leukemia binding peptides, folate receptor binding agents, steroid receptor binding peptide, carbohydrate receptor binding peptide or estrogen.
  • Bm if present, is a ST enterotoxin or fragment thereof.
  • Bm if present, is selected from octreotide and octreotate peptides.
  • Bm if present, is a synthetic polymer.
  • synthetic polymers useful for some applications include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers. Examples of specific peptide targeting ligands are described in WO/2008/108941.
  • Target tissue refers to tissue of a subject to which an optical agent is administered or otherwise contacted, for example during a biomedical procedure such as an optical imaging, phototherapy, monitoring or visualization procedure.
  • Target tissues can be contacted with an optical agent of the invention under in vivo conditions in vitro conditions or ex vivo conditions.
  • Target tissues in some embodiments include cancerous tissue, cancer cells, precancerous tissue, a tumor, a lesion, a site of inflammation, or vasculature tissue.
  • a target tissue includes a melanoma cell, a breast lesion, a prostate lesion, a lung cancer cell, a colorectal cancer cell, an atherosclerotic plaque, a brain lesion, a blood vessel lesion, a lung lesion, a heart lesion, a throat lesion, an ear lesion, a rectal lesion, a bladder lesion, a stomach lesion, an intestinal lesion, an esophagus lesion, a liver lesion, a pancreatic lesion, and a solid tumor.
  • Target tissue in some embodiments refers to a selected organ of the subject or component thereof, such as lung, heart, brain, stomach, liver, kidneys, gallbladder, pancreas, intestines, rectum, skin, colon, prostate, ovaries, breast, bladder, blood vessel, throat, ear, or esophagus.
  • Methods of this invention comprise the step of administering an "effective amount" of the present diagnostic and therapeutic compositions, formulations and preparations containing the present compounds or compositions, to diagnose, image, monitor, evaluate, treat, reduce, alleviate, ameliorate or regulate a biological condition and/or disease state in a patient.
  • the term "effective amount,” as used herein, refers to the amount of the diagnostic and therapeutic formulation, that, when administered to the individual is effective to diagnose, image, monitor, evaluate, treat, reduce alleviate, ameliorate or regulate a biological condition and/or disease state.
  • an effective amount of a given composition or formulation will depend at least in part upon the mode of administration (e.g. intravenous, oral, topical administration), any carrier or vehicle employed, and the specific individual to whom the formulation is to be
  • an effective amount of a compound or composition of the invention is a therapeutically effective amount.
  • the phrase "therapeutically effective” qualifies the amount of compound or composition administered in the therapy. This amount achieves the goal of ameliorating, suppressing, eradicating, preventing, reducing the risk of, or delaying the onset of a targeted condition.
  • an effective amount of a compound or composition of the invention is a diagnostically effective amount.
  • the phrase "diagnostically effective" qualifies the amount of compound or composition administered in diagnosis, for example of a disease state or other pathological condition. The amount achieves the goal of being detectable while avoiding adverse side effects found with higher doses.
  • an active ingredient or other component is included in a therapeutically acceptable amount.
  • an active ingredient or other component is included in a diagnostically acceptable amount.
  • the compounds and pharmaceutically acceptable salts of the invention can be used as part of a combination.
  • the term "combination" means the administration of two or more compounds directed to a target condition.
  • the treatments of the combination generally can be co-administered in a simultaneous manner.
  • Two compounds can be coadministered as, for example: (a) a single formulation (e.g., a single capsule) having a fixed ratio of active ingredients; or (b) multiple, separate formulations (e.g., multiple capsules) for each compound.
  • the treatments of the combination can alternatively (or additionally) be administered at different times.
  • the invention encompasses administering optical agents useful in the invention to a patient or subject.
  • a "patient” or “subject”, used equivalently herein, refers to an animal.
  • an animal refers to a mammal, preferably a human.
  • the subject can either: (1 ) have a condition able to be monitored, diagnosed, prevented and/or treated by administration of an optical agent of the invention; or (2) is susceptible to a condition that is able to be monitored, diagnosed, prevented and/or treated by administering an optical agent of the invention.
  • diagnosis When used herein, the terms "diagnosis”, “diagnostic” and other root word derivatives are as understood in the art and are further intended to include a general monitoring, characterizing and/or identifying a state of health or disease. The term is meant to encompass the concept of prognosis.
  • diagnosis of cancer can include an initial determination and/or one or more subsequent assessments regardless of the outcome of a previous finding. The term does not necessarily imply a defined level of certainty regarding the prediction of a particular status or outcome.
  • administering means that a compound or formulation thereof of the invention, such as an optical agent, is provided to a patient or subject, for example in a
  • the invention includes methods for a biomedical procedure wherein a therapeutically or diagnostically effective amount of a compound having any one of formulas (FX1) - (FX59) is administered to a patient in need of treatment, for example to a patient undergoing treatment for a diagnosed diseased state including cancer and vascular diseases.
  • Administering can be carried out by a range of techniques known in the art including parenteral administration including intravenous, intraperitoneal or subcutaneous injection or infusion, oral administration, topical or transdermal absorption through the skin, or by inhalation, for example.
  • the chosen route of administration may depend on such factors as solubility of the compound or composition, location of targeted condition, and other factors which are within the knowledge of one having ordinary skill in the relevant art.
  • Topical administration includes the use of transdermal administration, such as transdermal patches or iontophoresis devices.
  • Parenteral administration includes subcutaneous injections, intravenous injections, intraarterial injections, intraorbital injections, intracapsular injections, intraspinal injections, intraperitoneal injections, intramuscular injections, intrasternal injections, and infusion.
  • Dosage forms suitable for parenteral administration include solutions, suspensions, dispersions, emulsions, and any other dosage form that can be administered parenterally.
  • controlled-release component refers to an agent that facilitates the controlled-release of a compound including, but not limited to, polymers, polymer matrices, gels, permeable membranes, liposomes, microspheres, or the like, or any combination thereof. Methods for producing compounds in combination with controlled-release components are known to those of skill in the art.
  • the term "pharmaceutically acceptable” means approved by a regulatory agency of an appropriate federal or state government; or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans; or does not impart significant deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered.
  • the groups and structures described herein as portions of the compounds of the invention may be defined as if they are separate valence-satisfied chemical structures. It is intended that when a group is described or shown as being a substituent of another group, that the group be viewed as having a valency to allow this binding to occur.
  • EXAMPLE 1 SULFENAMIDE COMPOUNDS FOR PHOTOTHERAPY
  • Type 1 phototherapeutic agents including compositions, preparations and formulations, and methods of using and making Type 1 phototherapeutic agents.
  • Type 1 phototherapeutic agents of the invention include compounds comprising one or more photolabile, acyclic S-N bond directly or indirectly linked to a C 5 -C 30 aryl comprising one or more aromatic and/or heterocyclic aromatic groups. Incorporation of a C 5 -C 30 aryl comprising aromatic and/or heterocyclic aromatic groups in some compounds provides a chromophore moiety capable of absorption of electromagnetic radiation, preferably for some applications electromagnetic radiation having wavelengths in the visible (e.g.
  • the C 5 -C 30 aryl group(s) of some compositions of the invention functions as an aromatic antenna group for coupling energy from incident
  • an aromatic antenna group comprising a C 5 -C 30 aryl having one or more aromatic and/or heterocyclic aromatic groups is useful in some embodiments for initiating cleavage of a photolabile, acyclic S-N bond upon absorption of electromagnetic radiation and subsequent internal energy transfer processes.
  • compounds of the present invention are photoactivated by exposure to visible or near infrared electromagnetic radiation, for example electromagnetic radiation having wavelengths ranging from 350 nm to 1 300 nm. Absorption of at least a portion of the applied electromagnetic radiation generates a therapeutically effective amount of photoactivated phototherapeutic agent, which is schematically represented in figures 1 A-1 D by the compound provided in brackets with an asterisk symbol (*). Activation of the phototherapeutic agent may occur via a single photon absorption process, a mulitphoton absorption process or a combination of a single photon absorption process and a mulitphoton absorption process.
  • the activated photosensitizer subsequently undergoes processes, such as internal energy transfer and/or bond cleavage processes, resulting in formation of reactive species capable of causing a desired therapeutic result.
  • Reactive species generated by the compounds of the invention may include free radicals, intramolecular diradicals, ions, electrons, electrophiles, nitrene, vibrationally excited species, and translationally excited species. As illustrated in Figures 1 B and 1 D, excitation of the photosensitizer in these embodiments causes cleavage of the S-N bond, thereby generating two free radical species.
  • excitation of a C 5 -C 30 aryl results in rapid intramolecular energy transfer to the photolabile S-N bond resulting in bond cleavage and radical formation.
  • the coupling between the C 5 -C 3 o aryl and the central S-N bond may be selected to enhance the intramolecular energy transfer process(es) and provide efficient photoinitated bond cleavage.
  • the central S-N group is directly coupled to one or more rings of an aromatic group or heterocyclic aromatic group of a C 5 -C 30 aryl.
  • the reactive species generated upon excitation of the photosensitizer collide, react with, or otherwise interact with cell components of a target organ or tissue class, thereby resulting in death, injury and/or damage to cells at the target tissue.
  • Figures 1 E and 1 F provide schematics showing mechanisms for generation of reactive species for a class of photosensitizers having a S-N bond with a fused ring configuration, wherein the nitrogen atom of the S-N bond is a part of the fuse ring structure.
  • excitation results in formation of a stable free radical having a fused ring structure and a stable thiyl radical .
  • the free radical having a fused ring structure may further undergo electron transfer to generate a cation and anion reactive species.
  • photoactivation results in formation of a moderately stable cation and a stable anion.
  • Type 1 phototherapeutic agents useful for certain phototherapy applications incorporate one or more C 5 -C 30 aryl or C 5 -C 30 heteroaryl groups, including aromatic groups, heterocyclic aromatic groups, polycyclic aromatic groups and polycyclic heterocyclic aromatic groups, that absorb strongly in the visible and/or NIR region of the electromagnetic spectrum.
  • C 5 -C 30 aryl or C5-C30 heteroaryl groups providing effective photoactivation by electromagnetic radiation having wavelengths selected over the range of 300 nm to 1300 nm include, but are not limited to, groups corresponding to azulenes, aza-azulenes, anthracenes, pyrazines, pyridazines, quinolines, quinoxalines, courmarins, phenoxazines, phenothiazines, rhodamines, and the like.
  • the invention further includes phototherapeutic agents having one or more C 5 -C 30 aryl or C 5 -C 30 heteroaryl groups comprising aromatic group(s) and heterocyclic aromatic group(s) that are functionalized by incorporation of heteroatom ring members and substituents on the ring structure(s) providing excitation wavelength selection and/or tunability.
  • the C 5 -C 30 aryl or C 5 -C 30 heteroaryl comprises one or more aromatic or heterocyclic aromatic groups independently having one or more electron donating and/or electron withdrawing groups provided as ring substituents for providing selected excitation characteristics, such as a selected absorption spectrum and/or strong absorption in the visible and/or NIR regions.
  • Some phototherapeutic agents of the present invention operate, at least in part, via the Type 2 process involving formation of excited state oxygen ( 1 0 2 ), and optionally contain a C 5 -C 30 aryl or C 5 -C 30 heteroaryl that is a group corresponding to a cyanine, indocyanine, phenothiazine, or phthalocyanine.
  • compositions in phototherapeutic methods are selected to provide optical properties supporting and enabling use of these compositions in phototherapeutic methods, such as providing one or more of the following: (i) large extinction coefficients; (ii) strong absorption in the visible and/or infrared regions of the electromagnetic spectrum (e.g., 350 to 1300 nanometers, preferably for some applications 350-900 nanometers); and (iii) a large quantum yield for the production of reactive species, such as free radicals or ions, capable of causing photoactivation initiated tissue damage. Selection of the composition of R 1 - R 3 , R 11 - R 33 , R 40 - R 55 , R 58 - R 63 ,
  • FX59 may also be based, at least in part, on a number of pharmacokinetic and physical properties supporting effective delivery and clearance of the optical agents of the present methods and compositions. Such factors may include solubility, toxicity, immune response,
  • PEG oly(ethylene glycol)
  • a phototherapeutic agent of the invention incorporates aromatic groups and/or heterocyclic aromatic groups that are derivatized by the addition of at least one electron withdrawing group and at least one electron donating group bonded directly or indirectly to a carbon atom of the ring structure.
  • one or more the electron withdrawing (EWG) and electron donating (EDG) group(s) are directly attached to the ring structure of the aromatic group.
  • EWG and EDG are indirectly attached to the to the ring structure of the aromatic group through an unsaturated spacer that is in conjugation with the double bonds of a C 5 -C 30 aryl or C 5 -C 3 o heteroaryl group.
  • Electron donating and withdrawing groups in these dye compositions may be positioned ortho, meta or para to each other with respect to the to the ring structure of the aromatic group.
  • two electron withdrawing groups are positioned para to each other on the ring structure of the aromatic group and two electron donating groups are positioned para to each other on the ring structure of the aromatic group.
  • electron withdrawing groups and electron donating groups are positioned so as to make the overall compound symmetrical.
  • Optical agents of the invention support a broad therapeutic platform useful for a variety of in vivo phototherapy procedures, for example for the treatment of cancer, stenosis, inflammation, infection and arthritis.
  • Optical agents of the invention are optionally multifunctional agents capable of providing a useful combination of photodiagnostic, phototherapeutic, molecular recognition and/or targeting functionality.
  • a dye component is incorporated into the phototherapeutic agent of the present compositions for imparting useful optical functionality, for example by functioning as an optical absorber, chromophore, and/or fluorophore. This functionality is useful for targeted administration and excitation of the therapeutic agent.
  • optical agents of the invention further comprise a targeting component, such as a targeting ligand.
  • an optical agent of the invention comprises a targeting ligand integrated with a photosensitizer component to access enhanced administration, delivery and photoactivation functionality for phototherapy therapy.
  • bioconjugates thereof having one or more targeting ligands covalently bonded to or noncovalently associated with the phototherapeutic agents of the present invention, thereby providing specificity for administering, targeting, delivery and/or localizing an optical agent to a specific biological environment, such as a target tissue such as a specific organ, tissue, cell type or tumor site.
  • Bm is a targeting ligand, optionally providing molecular recognition functionality.
  • the targeting ligand is a particular region of the compound that is recognized by, and binds to, the target site on the organ, tissue, tumor or cell.
  • Targeting ligands are often, but not always, associated with biomolecules or fragments thereof which include hormones, amino acids, peptides,
  • peptidomimetics proteins, nucleosides, nucleotides, nucleic acids, enzymes, carbohydrates, glycomimetics, lipids, albumins, mono- and polyclonal antibodies, receptors, inclusion compounds such as cyclodextrins, and receptor binding molecules.
  • biomolecules include steroid hormones for the treatment of breast and prostate lesions; somatostatin receptor binding molecules, bombesin receptor binding molecules, and neurotensin receptor binding molecules for the treatment of neuroendocrine tumors, cholecystekinin receptor binding molecules for the treatment of lung cancer; heat sensitive bacterioendotoxin (ST) receptor binding molecules and carcinoembryonic antigen (CEA) binding molecules for the treatment of colorectal cancer, dihydroxyindolecarboxylic acid and other melanin producing biosynthetic intermediates for melanoma, integrin receptor and atheroscleratic plaque binding molecules for the treatment of vascular diseases, amyloid plaque binding molecules for the treatment of brain lesions, cholecystokinin (CCK) receptor binding molecules, steroid receptor binding molecules,
  • Targeting ligands for use in the invention may also include synthetic polymers.
  • synthetic polymers include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers.
  • appropriate targeting ligands may include integrin, selectin, vascular endothelial growth factor, fibrin, tissue plasminogen activator, thrombin, LDL, HDL, Sialyl LewisX and its mimics, and atherosclerotic plaque binding molecules.
  • the inventive receptor-targeted phototherapeutic agents are expected to be effective in the treatment of various lesions.
  • a targeting ligand may contain all or part of a steroid hormone or a steroid receptor binding compound, and therefore target steroid hormone sensitive receptors.
  • the compound is administered, targets the desired site such as breast and/or prostate lesion, is photoactivated, and forms free radicals at this site thereby effecting cell injury, damage, or death at the desired target site.
  • the desired site such as breast and/or prostate lesion
  • the targeting group may be a compound that targets and binds to a somatostatin, bombesin, CCK, and/or neurotensin receptor binding molecule, or may be a carcinogenic embryonic antigen-binding compound that binds to a carcinogenic embryonic antigen.
  • These are then photoactivated for radical formation at, for example, lung cancer cells with CCK receptor binding molecules, colorectal cancer cells with ST receptor and carcinoembryonic antigen (CEA) binding molecules, melanoma cells with dihyroxyindolecarboxylic acid, vascular sites of atherosclerotic plaque with integrin receptor binding molecules, brain lesions with amyloid plaque binding molecules, and the like.
  • the optical agents of this example may contain additional functionalities that can be used to attach various types of biomolecules, synthetic polymers, and organized aggregates for selective delivery to various organs or tissues of interest.
  • synthetic polymers include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers.
  • the invention includes, but is not limited to, phototherapeutic agents comprising a photosensitizer - biomolecule conjugate which provide advantages over nonspecific
  • phototherapeutic agents or the conjugation of photosensitizers to very large biomolecules provide enhanced localization and rapid visualization of tumors which is beneficial for both diagnosis and therapy.
  • the agents are rapidly cleared from blood and non-target tissues so there is less concern for accumulation and for toxicity.
  • a variety of high purity compounds may be synthesized for combinatorial screening of new targets, e.g., to identify receptors or targeting agents, and for the ability to affect the pharmacokinetics of the conjugates by minor structural changes.
  • a liposome or micelle may be utilized as a carrier or vehicle for the composition.
  • a phototherapeutic agent comprises a sulfenamide photosensitizer that may be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, may be on the external surface of the liposome or micelle.
  • a targeting ligand may be extemally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains the sulfenamide phototherapeutic
  • agent/photosensitizer to the desired tissue, organ, or other site in the body.
  • sulfenamide compounds may be prepared via reaction of a sulfenyl halide, such as a sulfenyl chloride, with an amine.
  • a sulfenyl halide such as a sulfenyl chloride
  • properties and methods of synthesizing and derivatizing sulfenamide compounds are disclosed in (1) "Adventures in Sulfur - Nitrogen Chemistry, F. Davis, J. Org. Chem 2006, 71 , 8993-9003; (2) Davis et. al., Org. Prep. Proc. Int., 1979, 1 1 , 33; (3), T. Greene. Protection of Amino Group. In Protective Group in Organic Synthesis.
  • Figure 2 provides a general scheme for the syntheses of a sulfenamide optical agent having an acyclic S-N bond useful in phototherapeutic methods of the invention.
  • a sulfenyl chloride is reacted with an amine in the presence of a base to form a sulfenamide compound.
  • R -R 3 and W 1 - W 3 are as described in connection with Formula (FX1).
  • At least one of R 1 -R 3 is a C5— Cso aryl or C5-C30 heteroaryl, optionally a C5— C 2 o aryl or C 5 -C 2 o heteroaryl, or C 5 - C 10 aryl or C 5 -C 0 heteroaryl.
  • Coupling a targeting ligand (Bm) to an sulfenamide compound may be accomplished by methods known in the art, for example, as disclosed in Hermanson, Bioconjugate Techniques, Academic Press, New York, 1996; Hnatowich et al., Radioactive Labeling of Antibody: A simple and efficient method, Science, 1983, 220, 613; Pelegrin et al., Photoimmunodiagnosis with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies, J. Cellular Pharm., 3 (1992) 141 ; and U.S. Patent No. 5,714,342.
  • Typical procedures for the preparation of a sulfenamide - peptide conjugate having formula are described involving reaction of an amino or carboxylated acyclic sulfenamide with a peptide targeting ligand.
  • Amino or carboxylated acyclic sulfenamides may be coupled to a targeting ligand such as a peptide by any of the standard peptide coupling methods (e.g., mixed anhydride or active ester coupling), or can be employed directly during the automated peptide synthesis procedure.
  • sulfenamides such as somatostatin, bombesin, cholecystekinin, ST, steroids, and the like
  • succinimido active esters For example, a sulfenamide compound containing a carboxyl group is activated by making a mixed anhydride in situ with isobutylchloroformate, and then reacted with any biomolecule bearing an amino group.
  • the carboxyl group can be esterified with N-hydroxysuccinimide, and reacted with the amino group to form the amide.
  • the carboxyl group containing tetrazolodisulfide can also be used directly in automated peptide synthesis procedure.
  • Figure 3A provides general schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond and peptide targeting ligand useful in phototherapeutic methods of the invention.
  • a sulfenyl chloride is reacted with an amine in the presence of a base to form a sulfenamide compound which is subsequently coupled to a peptide via formation of a peptide bond.
  • R 1 -R 3 and W 1 - W 3 are as described in connection with Formula (FX1), and wherein Peptide is a polypeptide comprising 2 to 30 amino acid units.
  • R 1 -R 3 is a C 5 - C 30 aryl or C 5 -C 30 heteroaryl, optionally a C 5 - C 20 aryl or C 5 -C 20 heteroaryl, or C 5 - C-io aryl or C 5 -C 0 heteroaryl.
  • Figure 3B provides schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond, peptide targeting ligand and phenyl group.
  • X is -CR 65 R 66 - wherein R 65 and R 66 are hydrogen or C C 2 o alkyl, C 3 -C 20 cycloalkyl, C 5 -C 20 aryl, or C 5 -C 20 heteroaryl, optionally a C1-C10 alkyl, C 3 -C 10 cycloalkyl, C 5 -C 10 aryl, or C 5 -C 0 heteroaryl.
  • each of R 1 - R 3 , R 65 and R 66 are individually an electron donating group, an electron withdrawing group or a targeting ligand (Bm).
  • Figure 4A and 4B provide schemes for the general syntheses and conjugation of photosensitizers having an acyclic S-N bond useful as phototherapeutic agents of the present invention.
  • L 1 - L 3 , W 1 - W 3 , and R 1 - R 3 are as described in connection with Formula (FX1), and wherein Peptide is a polypeptide comprising 2 to 30 amino acid units.
  • a solution of the desired amine (2.0 mmol) and triethylamine (4.0 mmol) in anhydrous methylene chloride (10 mL) is treated with 4-nitrobenzenesulfenyl chloride (2.2 mmol) in anhydrous methylene chloride (5 mL), and the mixture is stirred at ambient temperature for 2-16 hours. Thereafter, the reaction mixture is poured onto water and the organic phase was separated, washed with water, dried over sodium sulfate, and evaporated in vacuo. The crude product is purified by either crystallization or flash chromatography.
  • Figures 10 and 12 provide general schemes for the syntheses of bioconjugates of sulfenamide optical agents having formulas (FX1) and (FX49).
  • sulfenamide compounds are provided having pendant carboxyi or amine groups.
  • Subsequent reaction in the presence of a coupling agent (1) links the amino-terminus of a peptide and a carboxyi group of the sulfenamide compound in the presence of a coupling agent, or (2) links the carboxyl-terminus of a peptide and an amide group of the sulfenamide compound.
  • reaction schemes in Figures 10 and 12 illustrate conjugation of a peptide targeting ligand to the sulfenamide compound via an amide linkage.
  • coupling agents useful in the reactions of schemes of Figures 10 and 12 include hydrogen peroxide, chlorine, bromine, iodine, peracids, periodate, hypochlorite, and the like.
  • Figures 11A, 1 1 B, 13A and 13B illustrate examples of coupling reactions useful for synthesis of sulfenamide optical agents of the invention having specific targeting ligands.
  • the sulfenamide compound (1) in Figure 10 having a pendant carboxyi group is linked to octreotide, bombesin, cholecystokinin, bacterioenterotoxin and steroid targeting ligands via formation of an amide bond.
  • Figure 1 1 B the sulfenamide compound (2) in Figure 10 having a pendant amine group is linked to octreotide, bombesin, cholecystokinin,
  • the sulfenamide compound (3) in Figure 12 having a pendant carboxyl group is linked to octreotide, bombesin, cholecystokinin, bacterioenterotoxin and steroid targeting ligands via formation of an amide bond.
  • the sulfenamide compound (4) in Figure 12 having a pendant amine group is linked to octreotide, bombesin, cholecystokinin,
  • bacterioenterotoxin and steroid targeting ligands via formation of an amide bond.
  • a variety of coupling agents are useful for linking the sulfenamide backbone to the targeting ligands including dicyclohexylcarbodiimide (DCC), 1-ethyl- 3-(3-dimethylaminopropyl) carbodiimide (EDC), benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), disuccinimdyl carbonate, N-hydroxysuccinimide, methylformamide, isobutylchloroformate, etc.
  • DCC dicyclohexylcarbodiimide
  • EDC 1-ethyl- 3-(3-dimethylaminopropyl) carbodiimide
  • PyBOP benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate
  • the reaction mixture is treated with methylene chloride (e.g., 20 mL) and water (e.g., 30 mL).
  • the organic layer is separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo.
  • the crude material is purified by flash chromatography using chloroform-methanol gradien to give compound (FX51).
  • the reaction mixture is treated with methylene chloride (e.g, 20 mL) and water (e.g, 30 mL).
  • the organic layer is separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo.
  • the crude material is purified by flash chromatography using chloroform-methanol gradient to give compound (FX52).
  • the reaction mixture is treated with methylene chloride (e.g., 20 mL) and water (e.g., 30 mL).
  • the organic layer is separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo.
  • the crude material is purified by flash chromatography using chloroform-methano! gradient to give compound (FX53).
  • Peptide is a polypeptide comprising 2 to 30 amino acid units, and optionally wherein Peptide is bombesin.
  • peptide-sulfenamide conjugate (FX19) A typical procedure for the preparation of peptide-sulfenamide conjugate (FX19) using an automated peptide synthesizer is described. It should be noted that other sulfenamide compounds may be conjugated to a peptide targeting ligand, such as bombesin by the same or similar procedure.
  • the peptide-sulfenamide conjugate having formula (FX19) is prepared by
  • the first peptide cartridge contains Wang resin pre-loaded with an amide resin on 25-pmole scale.
  • the amino acid cartridges are placed on the peptide synthesizer and the product is synthesized from the C- to the N-terminal position.
  • the product is cleaved from the solid support with a cleavage mixture containing trifluoroacetic acid (85%):water (5%):phenol (5%):thioanisole (5%) for 6 hours.
  • the peptide-sulfenamide conjugate is precipitated with t-butyl methyl ether and lyophilized in watenacetonitrile (2:3) mixture.
  • the peptide-sulfenamide conjugate is purified by HPLC and analyzed with LC/MS.
  • a typical procedure for the preparation of Peptide-sulfenamide conjugate having formula (FX19) using mixed anhydride coupling procedure is described. It should be noted that other sulfenamide compounds may be conjugated to a peptide targeting ligand, such as bombesin by the same or similar procedure.
  • a solution of the sulfenamide compound having formula (FX18) (120 ⁇ ) and triethylamine (250 ⁇ ) in anhydrous dimethylformamide (DMF) (500 ⁇ _) is stirred and cooled to 0°C -10°C. Thereafter isobutylchloroformate (120 ⁇ ) is added and the mixture is stirred at 0-10 °C for about 30 minutes.
  • a polypeitde, such as Bombesin (7-14) peptide, (100 pmol) in DMF is added to the above mixture and allowed to come to ambient temperature (about 20°C to about 22°C) and stirred at ambient temperature for about one hour.
  • the product is cleaved from the solid support with a cleavage mixture containing trifluoroacetic acid (85%):water (5%):phenol (5%):thioanisole (5%) for 6 hours.
  • the peptide-sulfenamide conjugate is precipitated with t-butyl methyl ether and lyophilized in watenacetonitrile (2:3) mixture.
  • the peptide - sulfenamide conjugate is purified by reversed phase HPLC and analyzed with LC/MS. These methods are useful for preparation of an sulfenamide-bombesin (7-14) bioconjugate having the formula [FX18]-R 2 -Gln-Trp-Ala-Val-Gly-His- Leu-Met-NH 2 or [FX18]-R 3 -Gln-Trp-Ala-Val-Gly-His-Leu-Met-NH 2 .
  • a general procedure is carried out for measuring cell viability upon exposure of leukemia cells to a number of sulfenamide photosensitizers of the invention and light.
  • the sulfenamide photosensitizers evaluated include (FX18), (FX47), (FX48), (FX49) and (FX50).
  • the absorption maximum for each sulfenamide compound evaluated is provided in Table 1.
  • the cell viability analysis is carried out using a human myeloid leukemia U937 cell line by the standard WST-1 assay.
  • U397 Leukemia cells (0.5 x 10 6 ) are plated in standard T-25 cell culture flasks, and are exposed to four controls and a series of test conditions corresponding to a range of sulfenamide photosensitizer concentrations.
  • the sulfenamide photosensitizer is dissolved in DMSO at an initial concentration of about 8 mM and diluted with water to the final desired value such that amount of DMSO exposed to the cells is always kept below 0.5%.
  • the cells are incubated at 37 C with various concentrations the photosensitizer for about 30 minutes prior to the exposure of light.
  • the cells are irradiated for 5, 10 and/or 20 minute durations that radiates light with maximum output in the range of 325-425 nm with constant cooling such that the temperature at the surface of the microliter plates does not exceed 37 °C.
  • the experiments also include control conditions wherein the cells are irradiated for 5, 10 and/or 20 minute durations in the absence of photosensitizer also with constant cooling.
  • the light source is a 200-W B-100SP High Intensity Lamp from UVP. It is noted that in the present study, the light source is not optimized with respect to radiant energy, power and wavelength.
  • HBSS Hank's Balanced Salt Solution
  • Trypan blue stain Trypan blue stain
  • Viability measurements are analyzed to provide VC 50 / 2 o values which is defined as the
  • Figure 5 provides cell viability results for control conditions (Control 4, no photosensitzer, DMSO, light) wherein the cells are exposed to light and dimethyl sulfoxide.
  • Control 4 no photosensitzer, DMSO, light
  • DMSO toxicity is observed only at the highest concentrations of DMSO.
  • Cells are incubated without DMSO (0 ⁇ ) and with DMSO at concentrations of 3 mM, 6 mM, and 12 mM.
  • the cells were exposed to light from a B-100SP High Intensity Lamp for 0, 5, and 20 minutes.
  • 100% of the cells are viable with conditions of 0 mM DMSO and 3 mM DMSO for light exposure for 5 minutes and 20 minutes.
  • Figure 6 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX18) for a range of photosensitizer concentrations and exposure times.
  • the cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability.
  • the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light over the range of 5 minutes to 20 minutes.
  • cells are incubated in the absence of sulfenamide photosensitizer (0 ⁇ ) and with sulfenamide photosensitizer at concentrations of 1.5 ⁇ , 3 ⁇ , and 6 ⁇ .
  • the cells are exposed to light from a B-100SP High Intensity Lamp for 0, 5, 10, and 20 minutes.
  • the results indicate that in the absence of sulfenamide photosensitizer, the cells at all light exposure times have 100% viability.
  • cells have 100% viability with light exposure for 5 minutes, about 87% viability with light exposure for 10 minutes, and 65% viability with light exposure for 20 minutes.
  • cells For conditions of 3.0 ⁇ sulfenamide photosensitizer, cells have about 77% viability with light exposure for 5 minutes, about 46% viability with light exposure for 10 minutes, and about 15% viability with light exposure for 20 minutes. For conditions of 6.0 ⁇ sulfenamide photosensitizer, cells have about 19% viability with light exposure for 5 minutes, about 12% viability with light exposure for 10 minutes, and about 7% viability with light exposure for 20 minutes. The cell viability results yield a VC 50 /2o value of 0.2 ⁇ 0.1 ⁇ for the sulfenamide photosensitizer having Formula (FX18).
  • Figure 7 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX49) for a range of concentrations and exposure times.
  • the cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability.
  • the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes.
  • Cells are incubated in the absence of sulfenamide photosensitizer (0 ⁇ ) and with sulfenamide photosensitizer at concentrations of 0.63 ⁇ , 1.25 ⁇ , and 2.5 ⁇ .
  • the cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes.
  • the results indicate that in the absence of sulfenamide photosensitzer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes.
  • cells For conditions of 0.63 ⁇ sulfenamide photosensitizer, cells have about 80% viability for conditions of no light exposure and about 90% viability for light exposure for 20 minutes.
  • cells For conditions of 1.25 ⁇ sulfenamide photosensitizer, cells have about 78% viability for conditions of no light exposure and about 62% viability for light exposure for 20 minutes.
  • cells For conditions of 2.5 ⁇ sulfenamide photosensitizer, cells have about 78% viability for conditions of no light exposure and about 20% viability for light exposure for 20 minutes. The cell viability results yield a VC 50/2 o value of 1.8 ⁇ for the sulfenamide photosensitizer having Formula (FX49).
  • Figure 8 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX47) for a range of concentrations and exposure times.
  • the cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability.
  • the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes.
  • Cells are incubated in the absence of sulfenamide photosensitizer (0 ⁇ ) and with sulfenamide photosensitizer at concentrations of 1.25 ⁇ , 2.5 ⁇ , and 5 ⁇ .
  • the cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes.
  • the results indicate that in the absence of sulfenamide photosensitzer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes.
  • cells For conditions of 1.25 ⁇ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 85% viability for light exposure for 20 minutes.
  • cells have greater than 100% viability for conditions of no light exposure and about 55% viability for light exposure for 20 minutes.
  • cells For conditions of 5 ⁇ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 22% viability for light exposure for 20 minutes.
  • the cell viability results yield a VC 50/2 o value of 3.2 ⁇ for the sulfenamide
  • Figure 9 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX48) for a range of concentrations and exposure times.
  • the cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability.
  • the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes.
  • Cells are incubated in the absence of sulfenamide photosensitizer (0 ⁇ ) and with sulfenamide photosensitizer at concentrations of 1.25 ⁇ , 2.5 ⁇ , and 5 ⁇ .
  • the cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes.
  • the results indicate that in the absence of sulfenamide photosensitzer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes.
  • cells For conditions of 1.25 ⁇ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 75% viability for light exposure for 20 minutes.
  • cells have greater than 100% viability for conditions of no light exposure and about 42% viability for light exposure for 20 minutes.
  • cells For conditions of 5 ⁇ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 25% viability for light exposure for 20 minutes.
  • the cell viability results yield a VC 50 / 2 o value of 2.2 ⁇ for the sulfenamide
  • Figure 20 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX50) for a range of concentrations and exposure times.
  • the cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability.
  • the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes.
  • Cells are incubated in the absence of sulfenamide photosensitizer (0 ⁇ ) and with sulfenamide photosensitizer at concentrations of 0.63 ⁇ , 1.3 ⁇ , and 2.5 ⁇ .
  • the cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes.
  • the results indicate that in the absence of sulfenamide photosensitizer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes.
  • cells For conditions of 0.63 ⁇ sulfenamide photosensitizer, cells have about 90% viability for conditions of no light exposure and about 100% viability for light exposure for 20 minutes.
  • cells For conditions of 1.3 ⁇ sulfenamide photosensitizer, cells have about 94% viability for conditions of no light exposure and about 92% viability for light exposure for 20 minutes.
  • cells For conditions of 2.5 ⁇ sulfenamide photosensitizer, cells have about 90% viability for conditions of no light exposure and about 52% viability for light exposure for 20 minutes. The cell viability results yield a VC 50 /2o value of 2.5 ⁇ for the sulfenamide photosensitizer having Formula (FX50).
  • Electron Spin Resonance (ESR) spectrometry measurements are carried out to probe generation of reactive species upon optical excitation of sulfenamide compounds of the invention.
  • ESR measurements provide information relating to the mechanism of photactivation of the present compounds including the identity and abundance of radical species produced upon exposure to light. For example, strong ESR signals are observed that are indicative of stable sulfur-centered radicals and less stable nitrogen-centered radicals.
  • the ESR measurements of this example demonstrate that sulfenamide photosensitizers of the invention are effective for localized generation of reactive species, including free radicals, and generally support the applicability of the present compounds as optical agents for use in phototherapy.
  • Figure 14 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX18). Trace (a) of Figure 14 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX18) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 14 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX18) is exposed to light and indicates the production of radical species during irradiation.
  • Trace (c) of Figure 14 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX18) is exposed to light and indicates the presence of radical species during this time.
  • Trace (d) of Figure 14 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX18) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
  • Figure 15 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX47). Trace (a) of Figure 15 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 15 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of radical species during irradiation.
  • Trace (c) of Figure 15 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX47) is exposed to light and indicates the presence of radical species during this time.
  • Trace (d) of Figure 15 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX47) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
  • Figure 16 provides ESR spectra for a second set of experiments evaluating the sulfenamide compound of formula (FX47). Trace (a) of Figure 16 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 16 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of radical species during this time.
  • Trace (c) of Figure 16 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX47) is exposed to light and indicates the presence of radical species during irradiation.
  • Trace (d) of Figure 16 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX47) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
  • Figure 17 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX48). Trace (a) of Figure 17 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX48) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 17 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX48) is exposed to light and indicates the production of radical species during irradiation.
  • Trace (c) of Figure 17 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX48) is exposed to light and indicates the presence of radical species during this time.
  • Trace (d) of Figure 17 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX48) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
  • Figure 18 provides ESR spectra for a second set of experiments evaluating the sulfenamide compound of formula (FX48).
  • Trace (a) of Figure 18 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX48) is exposed to light and indicates the production of little to no radical species during this time.
  • Trace (b) of Figure 18 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula ' (FX48) is exposed to light and indicates the production of radical species during irradiation.
  • Trace (c) of Figure 18 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX48) is exposed to light and indicates the presence of radical species during this time.
  • Figure 19 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX49). Trace (a) of Figure 19 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX49) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 19 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX49) is exposed to light and indicates the production of radical species during irradiation.
  • Trace (c) of Figure 19 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX49) is exposed to light and indicates the presence of radical species during this time.
  • Trace (d) of Figure 19 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX49) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
  • Phototherapy such as photodynamic therapy (PDT) typically employs a combination of a photosensitizer (PS) and visible or near infrared light to generate reactive species that kill or otherwise degrade target cells, such as tumors or other lesions.
  • PS photosensitizer
  • the present invention provides phototherapeutic agents useful for phototherapy.
  • methods the invention are not surgical methods.
  • the invention includes phototherapy methods wherein a phototherapeutic agent comprising a compound of any one of the formulas (FX1) - (FX59) is administered to a patient, for example, wherein a therapeutically effective amount of such a component is administered to a patient in need of treatment.
  • compounds of the invention provide an optical agent capable of selective targeting and delivery to a target tissue such as a tumor, site of inflammation or other lesion.
  • the phototherapeutic agent is optionally allowed to accumulate in a target region of interest (e.g., target tissue, tumor, or organ).
  • the phototherapeutic agent is activated by exposure to electromagnetic radiation.
  • the phototherapeutic agent is activated after an effective
  • concentration of the phototherapeutic agent has accumulated in a target tissue.
  • An effective concentration of a compound of the invention depends on the nature of the formulation, method of delivery, target tissue, activation method and toxicity to the surrounding normal non-target tissue. Exposure to electromagnetic radiation and activation of the phototherapeutic agent may occur during or after administration of the phototherapeutic agent and accumulation at the target tissue.
  • the target region is illuminated with electromagnetic radiation having a wavelength in the range of about 350 nm to about 1300 nm, preferably for some applications in the range of about 400 nm to about 900 nm.
  • the wavelength of the electromagnetic radiation corresponds to a peak in the absorption spectrum of the phototherapeutic agent, for example is within 20 nanometers of a peak in the absorption spectrum of the phototherapeutic agent in the visible or NIR regions.
  • the target site is exposed to electromagnetic radiation having sufficient fluence and/or power sufficient to activate the phototherapeutic agent so as to induce cell death, for example via necrosis or apoptosis processes.
  • electromagnetic radiation having low energy, power or fluence is provided to activate the phototherapeutic agent without undesirable thermal effects.
  • the region of interest is, for example, a lesion or tumor on the skin surface, the region can be directly illuminated. Otherwise, endoscopic and/or endoluminal catheters equipped with an electromagnetic radiation source may be employed to provide a photodiagnostic and/or phototherapeutic effect.
  • Appropriate power and intensity of the electromagnetic radiation depends on the size, depth, and the pathology of the lesion, as is known to one skilled in the art.
  • the fluence of the electromagentic radiation is preferably, but not always, kept below 200 mW/cm 2 , optionally below 100 mW/cm 2 , to minimize undesirable thermal effects.
  • the intensity, power, and duration of the illumination, and the wavelength of the electromagnetic radiation may vary widely depending on the body location, the lesion site, the effect to be achieved, etc.
  • the power of the applied electromagnetic radiation is preferably is selected over the range of 1 - 500 mW/cm 2 and optionally for some applications selected over the range of 1 - 200 mW/cm 2 cm 2 and optionally for some applications selected over the range of 1 - 100 mW/cm .
  • the duration of the exposure to applied electromagentic radiation selected over the range of 1 second to 60 minutes, and optionally for some applications selected over the range of 1 second to 30 minutes, and optionally for some applications selected over the range of 1 second to 10 minutes, and optionally for some applications selected over the range of 1 second to 1 minute.
  • the invention provides a method of using a phototherapeutic agent, the method comprising: (i) administering a therapeutically effective amount of a phototherapeutic t comprising a compound being of the formula (FX1):
  • each of L 1 , L 2 , and L 3 is independently C C 10 alkylene, C 3 -C 0 cycloalkylene, C 2 -C 10 alkenylene, C 3 -C 10 cycloalkenylene, C 2 -C 10 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, -(CH 2 CH 2 0) b -, or -(CHOH) a -;
  • each of W 1 , W 2 , and W 3 is independently a single bond, -(CH 2 ) n - -(HCCH) n - -0-, -S-, -SO-, -S0 2 - -SO 3 -, -OS0 2 - -NR 11 -, -CO-, -COO-, -OCO-, -OCOO-, -CONR 12 -, - NR 13 CO- -OCONR 14 -, -NR 15 COO- -NR 16 CONR 17 -, -NR 18 CSNR 19 -, -0(CH 2 ) n - -S(CH 2 ) n - - NR" u (CH 2 ) n - -CO(CH 2 )n- -COO(CH 2 ) n -, -OCO(CH 2 ) n -, -OCOO(CH 2 ) n -, -CONR
  • each of R , R 2 , and R 3 is independently a hydrogen, -OCF 3 , C T C 20 alkyi, C 3 -C 20 cycloalkyl, C 5 -C 30 aryl, C 5 -C 30 heteroaryl, C C 20 acyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, C 5 -C 20 alkylaryl, C C 6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C0 2 R 40 , -SOR 41 , - OSR 42 , -S0 2 OR 43 , -CH 2 (CH 2 OCH 2 ) b CH 2 OH, -P0 3 R 44 R 45 , -OR 46 , -SR 47 , -NR 48 R 49 , - NR 50 COR 51 , -CN, -CONR 52 R 53 , -COR 54 , -N0 2
  • R 2 , R 3 , W 2 , W 3 , and L 2 and L 3 if present, together with the atoms to which they are attached combine to form one to five substituted or unsubstituted, carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings;
  • R 1 , R 2 , and R 3 is a C 5 -C 30 aryl or C 5 -C 30 heteroaryl, or wherein R 2 , R 3 , W 2 , W 3 , and L 2 and L 3 , if present, together with the atoms to which they are attached combine to form a C 5 -C 30 aryl or C 5 -C 30 heteroaryl;
  • each of a and b is independently an integer selected from the range of 1 to 100;
  • each n is independently an integer selected from the range of 1 to 10;
  • each of e, f, and g is independently 0 or 1 ;
  • each of R 11 - R 33 is independently hydrogen, C C 20 alkyl, C 3 -C 20 cycloalkyl, C 5 -C 20 heteroaryl, or C 5 -C 20 aryl;
  • each of R 40 -R 55 and R 58 - R 61 is independently hydrogen, C 3 -C 10 cycloalkyl or C C 10 alkyl; each of R 62 and R 63 is independently a side chain residue of a natural a-amino acid;
  • each FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
  • each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steriod, a hormone, an aptamer, a receptor, a metal chelating agent, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units; and (ii) exposing the phototherapeutic agent administered to the patient to electromagnetic radiation.
  • the phototherapeutic agent is exposed to a therapeutically effective amount of electromagnetic radiation.
  • a therapeutically effective amount of electromagentic radiation is an amount for achieving a desired therapeutic result, for example an amount for generating a therapeutically effective amount of reactive species for damaging or causing cell death of a selected target tissue.
  • the method further comprises generating one or more reactive species from said compound administered to the patient via the exposure of the phototherapeutic agent to applied electromagnetic radiation.
  • the method further comprises the step of cleaving a photolabile S-N bond so as to generate reactive species, such as free radicals and/or ions.
  • the method further comprises contacting a selected organ or selected tissue in the patient with the phototherapeutic agent.
  • a therapeutically effective dose of the phototherapeutic agent is administered to a patient in need of treatment.
  • Phototherapeutic agents useful in the present methods include sulfenamide compounds containing at least one S-N bond directly or indirectly coupled to a C 5 -C 3 o aryl or C 5 -C 30 heteroaryl comprising one or more aromatic and/or heterocyclic aromatic groups.
  • Phototherapeutic agents useful in the present methods include compounds optionally having a ligand for targeted administration.
  • Phototherapeutic agents useful in the present methods include compounds optionally having a dye component, such as a fluorophore or chromophore, for imaging and/or visualization functionality.
  • the method of the invention comprises administering to a patient a compound having any one of formula selected from (FX1) - (FX59), including any of the specific compositions classes and compounds described in connection with formula (FX1) - (FX59).
  • the present methods expressly include methods of using phototherapeutic agents wherein the phototherapeutic agent includes the compound classes, compounds, and all variations thereof, described herein, including the compound classes, compounds and variations described in connection with any one of formulas (FX1) - (FX59).
  • Embodiments of this aspect may comprise a method of carrying out an in vivo therapeutic and/or diagnostic procedure.
  • the invention comprises a method of carrying out an in vivo phototherapeutic, photoactivation, and/or photosensitizing procedure.
  • the present methods have broad clinical utility which includes, but is not limited to, phototherapy of tumors, inflammatory processes, and impaired vasculature.
  • subjects of the invention may be any mammal, such as a human, and optionally the subject of the present methods is a patient in need of treatment and/or diagnosis.
  • the present methods are also useful in ex vivo and in vitro procedures, including medical therapeutic and diagnostic procedures.
  • Methods of the invention may optionally further comprise a number of other steps.
  • the method further comprises the step of administering the phototherapeutic agent into a bodily fluid of the subject.
  • the phototherapeutic agent may be introduced into the patient by any suitable method, including intravenous, intraperitoneal or subcutaneous injection or infusion, oral administration, transdermal absorption through the skin, or by inhalation.
  • the method further comprises contacting a target tissue, such as an organ, tissue, tumor, lesion, or cell type, with a compound of any one of formulas (FX1) - (FX59) prior to or during the exposure step.
  • the method further comprises allowing the compound to accumulate in a target tissue prior to exposure of the phototherapeutic agent to electromagnetic radiation.
  • the method further comprises contacting and/or selectively targeting the diagnostic agent to a selected organ, tissue, tumor, lesion, inflammation, or cell type.
  • the phototherapeutic agent is administered to the skin, a tumor, surgical site, or a wound site.
  • the phototherapeutic agent is administered and/or delivered to a blood vessel, lung, heart, throat, ear, rectum, bladder, stomach, intestines, esophagus, liver, brain, prostrate, breast, or pancreas of the subject.
  • the optical conditions for the step of exposing the phototherapeutic agent administered to the patient to electromagnetic radiation will vary considerably with the (i) therapeutic and/or diagnostic objectives, and (ii) the condition of the subject (e.g., height, weight, state of health etc.).
  • the applied electromagnetic radiation has wavelengths, energy and/or fluence sufficient to achieve a desired therapeutic and/or diagnostic result.
  • the electromagnetic radiation has wavelengths, energy and/or fluence sufficient to activate the phototherapeutic agent, for example wavelengths, energy and/or fluence sufficient to result in generation of reactive species, including free radicals and/or singlet oxygen.
  • the electromagnetic radiation has wavelengths, energy and/or fluence sufficient to result in cleavage of at least one photolabile bond of the optical agent upon absorption.
  • the electromagnetic radiation exposed to the phototherapeutic agent has wavelengths corresponding to a maximum in the absorption spectrum of the
  • phototherapeutic agent preferably for some applications a maximum (e.g., within 20»nm of a maximum in the absorption spectrum) in the visible or NIR regions of the electromagnetic spectrum.
  • excitation is achieved using electromagnetic radiation substantially free (e.g., less than about 10% of total radiant energy), of ultraviolet radiation, for example, to minimize exposure of the subject to electromagnetic radiation capable of causing unwanted cell or tissue damage.
  • Electromagnetic radiation may be provided to the phototherapeutic agent using a range of optical sources and/or surgical instrumentation, including a laser, light emitting diodes, fiber optic device, endoscope, catheter, optical filters, or any combination of these.
  • the invention include methods for phototherapy using an optical agent providing targeted delivery to a selected target tissue.
  • Embodiments of this aspect use an optical agent, such as a photosensitizer, having a targeting ligand.
  • selection of the composition of a targeting ligand in the present methods will dependent on therapeutic and/or diagnostic objectives, the condition of the subject and the chemical composition and properties of the target tissue of interest.
  • a targeted compound can contain all or part of a steroid hormone or a steroid receptor binding compound, and therefore target steroid hormone sensitive receptors.
  • the targeted compound is administered, targets and preferably accumulates in the desired site such as breast and/or prostate lesion and is photoactivated for monitoring, imaging, or therapy remotely or at the target site.
  • Similar target binding molecules and uses will be recognized by one skilled in the art.
  • the targeted compound can be a compound that targets and binds to a somatostatin, bombesin, CCK, and/or neurotensin receptor binding molecule, or can be a carcinogenic embryonic antigen-binding compound that binds to a carcinogenic embryonic antigen.
  • lung cancer cells with CCK receptor binding molecules colorectal cancer cells with ST receptor and carcinoembryonic antigen (CEA) binding molecules, melanoma cells with dihydroxyindolecarboxylic acid, vascular sites of atherosclerotic plaque with integrin receptor binding molecules, brain lesions with amyloid plaque binding molecules, and the like.
  • CCK receptor binding molecules colorectal cancer cells with ST receptor and carcinoembryonic antigen (CEA) binding molecules
  • CEA carcinoembryonic antigen
  • the optical agents of this example can contain additional functionalities that can be used to attach various types of biomolecules, synthetic polymers, and organized aggregates for selective delivery to various organs or tissues of interest.
  • synthetic polymers include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers.
  • the invention includes, but is not limited to, phototherapeutic agents comprising an optical agent - biomolecule conjugate which provides advantages over nonspecific optical agents or the conjugation of optical agents to very large biomolecules. These conjugates provide enhanced localization in, and rapid visualization of, tumors which is beneficial for imaging, monitoring, diagnosis and therapy.
  • the agents are rapidly cleared from blood and non-target tissues so there is less concern for accumulation and for toxicity.
  • a variety of high purity compounds can be easily synthesized for combinatorial screening of new targets, e.g. , to identify receptors or targeting agents, and for the ability to affect the pharmacokinetics of the conjugates by minor structural changes.
  • a liposome or micelle can be utilized as a carrier or vehicle for the composition.
  • an optical agent comprises a compound of the invention that can be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, can be on the external surface of the liposome or micelle.
  • a targeting ligand can be externally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains a phototherapeutic agent/photosensitizer compound of the invention) to the desired tissue, organ, or other site in the body.
  • compounds of the invention are useful for both oncology and non- oncology applications.
  • Some specific targets are tumors accessible via endoscope.
  • a compound that targets a protein, polypeptide, oligonucleotide or other biomolecule associated with such a tumor is administered to the tumor via endoscope or other useful method.
  • the compounds of the invention can be used in phototherapeutic applications, monitoring applications, diagnosis applications or imaging applications.
  • Other specific target tissues include colon, lung, ovarian, cervical, esophageal, bladder, blood, stomach cancers, endometriosis, and bacterial infections.
  • the estrogen receptor is an example of a steroid receptor to which steroid receptor binding molecules would bind.
  • the following compounds are known to bind to the estrogen receptor: estratriol; 17p-aminoestrogen (AE) derivatives such as prolame and butolame; drugs such as tamoxifen, ICI-164384, raloxifene, and genistein; 17p-estradiol; glucocorticoids;
  • kits can identify compounds specific for binding to the estrogen receptor (e.g. , Estrogen Receptor-alpha Competitor Assay Kit, Red; and Estrogen Receptor-beta
  • the glucose receptor is an example of a carbohydrate receptor to which carbohydrate receptor binding molecules would bind.
  • the glucose conjugate N-palmitoyl glucosamine [NPG] is known to bind the glucose receptor (Dufes et al., Pharm. Res. 17: 1250, 2000).
  • the glycoprotein hormone receptor is another example of a carbohydrate receptor to which carbohydrate receptor binding molecules would bind.
  • Follicle stimulating hormone (FSH) is known to bind the
  • glycoprotein hormone receptor Teilly et al., Endocrinology 131 : 799, 1992.
  • Other compounds known to bind the carbohydrate receptor, and hence examples of carbohydrate receptor binding molecules, are: polysialic acid, bacterial adhesins (specialized surface proteins that mediate binding of many pathogenic bacteria, such as enterohemorrhagic E.
  • EHEC EHEC
  • Shigella dysenteriae to host cells, which allow these bacteria to colonize host cell surfaces
  • soluble carbohydrate receptor analogs artificial glycopolymers and other multivalent glycoconjugates such as an acrylamide copolymer carrying -L-fucopyranoside and 3-sulfo-D-galactopyranoside in clusters
  • isomeric carbohydrates synthetic derivatives, neoglycoproteins, neoglycolipids, glycosidases, and glycosyltransferases.
  • Carbohydrate binding proteins can be screened with phage display libraries as known to a person of ordinary skill in the art.
  • Somatostatin receptor binding molecules include somatostatin and somatostatin receptor analogs, octreotide, glycosylated somatostatin-14 (somatostatin-dextran 70 ), seglitide, and peptides P587 and P829 as described in Vallabhajosula et al., J. Nuclear Med., 37:1016, 1996.
  • Cholecystokinin receptor binding molecules include the endogenous peptides cholecystekinin (CCK)-4, CCK-8, CCK-33, and gastrin; antagonists devazepide and lorglumide; agonists BC264 [Tyr(S0 3 H)-gNle-mGly-Trp-(NMe)Nle-Asp-Phe-NH 3 ] and desulfated CCK-8; Kinevac (synthetic cholecystekinin, sincalide); and CCK analogues modified at the sulfated tyrosyl at position 27.
  • CCK cholecystekinin
  • Neurotensin receptor binding molecules include neurotensin, neuromedin N, JMV449 (H- Lysv
  • Commercially available neurotensin receptor binding kits can evaluate potential neurotensin receptor binding molecules (e.g., DELFIA Neurotensin Receptor Binding Kit, PerkinElmer (Boston MA)).
  • Bombesin receptor binding molecules include the endogenous ligands gastrin-releasing peptide (GRP), neuromedin B (NMB), and GRP-18-27, and antagonists including JMV-1458 (glycine-extended bombesin (paraphydroxy-phenyl-propionyl-Gln-Trp-Ala-Val-Gly-His-Leu-Met- Gly-OH)), JMV-641 , JMV-1799, and JMV-1802, PD165929, 1 -naphthoyl-[DAIa 24 ,DPro 26 , V 26- 27]GRP-20-27, kuwanon H, and kuwanon G.
  • Commercially available bombesin receptor binding kits can evaluate potential bombesin receptor binding molecules (e.g., DELFIA Bombesin
  • ST receptor binding molecules include native ST peptide, and SEQ ID NO:2, SEQ ID NO:3, SEQ ID NOS:5-54 and fragments and derivatives thereof from U.S. Patent No. 5,518,888.
  • Compounds of the invention can contain all or part of a targeting ligand, receptor or peptide known to bind to a specific target, such as a target tissue.
  • Targeting ligands may be linked to the backbone or other portion of the present compounds using a range of synthetic approaches known in the art, including the synthetic approaches for conjugating biomolecule targeting ligands to optical agents as disclosed in Hnatowich et al., Radiolabeling of Antibodies: A simple and efficient method, Science, 1983, 220, p. 613; Pelegrin et al., Photoimmunodiagnostics with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies, Journal of Cellular Pharmacology, 1992, 3, pp.
  • Linking of biomolecule targeting ligands having an amine group may be achieved by techniques involving succinimido active esters.
  • a carboxyl group of a compound of the invention is activated by making a mixed anhydride in situ with
  • the activated compound is subsequently reacted with any biomolecule bearing an amino group, such as a polypeptide, protein, enzyme, antibody or fragment thereof, to achieve linking of the biomolecule to the compound so as to provide a targeting ligand covalently bond to the compound.
  • a carboxyl group of the present compounds may be first esterified with N-hydroxysuccinimide, and subsequently reacted with the amino group of a biomolecule, such as a polypeptide, protein, enzyme, antibody or fragment thereof, to form an amide bond linking the biomolecule to the compound so as to provide a targeting ligand covalently bond to the compound.
  • the invention contemplates pharmaceutically active compounds either chemically synthesized or formed by in vivo biotransformation to compounds set forth herein.
  • compositions of the invention is a compound or salt or ester thereof suitable for pharmaceutical formulations.
  • Prodrugs of the compounds of the invention are useful in embodiments including compositions and methods. Any compound that will be converted in vivo to provide a biologically, pharmaceutically, diagnostically, or therapeutically active form of a compound of the invention is a prodrug.
  • Various examples and forms of prodrugs are well known in the art. Examples of prodrugs are found, inter alia, in: Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985); Methods in Enzymology, Vol. 42, at pp. 309-396, edited by K. Widder, et. al.
  • a prodrug such as a pharmaceutically acceptable prodrug
  • Prodrugs of the invention can be rapidly transformed in vivo to a parent compound of a compound described herein, for example, by hydrolysis in blood or by other cell, tissue, organ, or system processes. Further discussion is provided in: T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, V. 14 of the A.C.S. Symposium Series; and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical
  • Optical agents of the invention can be formulated with pharmaceutically-acceptable anions and/or cations.
  • Pharmaceutically-acceptable cations include among others, alkali metal cations (e.g., Li + , Na + , K + ), alkaline earth metal cations (e.g., Ca 2+ , Mg 2+ ), non-toxic heavy metal cations and ammonium (NH 4 + ) and substituted ammonium (N(R') 4 + , where R' is hydrogen, alkyl, or substituted alkyl, i.e., including, methyl, ethyl, or hydroxyethyl, specifically, trimethyl ammonium, triethyl ammonium, and triethanol ammonium cations).
  • Pharmaceutically-acceptable anions include, among others, halides (e.g., F ⁇ , CI " , Br " , At " ), sulfate, acetates (e.g., acetate,
  • Pharmaceutically acceptable salts comprise pharmaceutically-acceptable anions and/or cations.
  • pharmaceutically acceptable salt can refer to acid addition salts or base addition salts of the compounds in the present disclosure.
  • a pharmaceutically acceptable salt is any salt which retains at least a portion of the activity of the parent compound and does not impart significant deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered.
  • Pharmaceutically acceptable salts include metal complexes and salts of both inorganic and organic acids.
  • Pharmaceutically acceptable salts include metal salts such as aluminum, calcium, iron, magnesium, manganese and complex salts.
  • Pharmaceutically acceptable salts include, but are not limited to, acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, cilexetil, citric, edetic, edisylic, estolic, esyl, esylic, formic, fumaric, gluceptic, gluconic, glutamic, glycolic,
  • acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, cilexetil,
  • glycolylarsanilic hexamic, hexylresorcjnoic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, lactobionic, maleic, malic, malonic, mandelic,
  • methanesulfonic methylnitric, methylsulfuric, mucic, muconic, napsylic, nitric, oxalic, p- nitromethanesulfonic, pamoic, pantothenic, phosphoric, monohydrogen phosphoric, dihydrogen phosphoric, phthalic, polygalactouronic, propionic, salicylic, stearic, succinic, sulfamic, sulfanlic, sulfonic, sulfuric, tannic, tartaric, teoclic, toluenesulfonic, and the like.
  • Pharmaceutically acceptable salts can be derived from amino acids, including, but not limited to, cysteine. Other
  • a compound of the invention, or pharmaceutically acceptable salt thereof is administered to a subject in a diagnostically or therapeutically effective amount.
  • a diagnostically or therapeutically effective amount One skilled in the art generally can determine an appropriate dosage.
  • compositions for oral administration can be, for example, prepared in a manner such that a single dose in one or more oral preparations contains at least about 20 mg of the present compound per square meter of subject body surface area, or at least about 50, 100, 150, 200, 300, 400, or 500 mg of the present compound per square meter of subject body surface area (the average body surface area for a human is, for example, 1.8 square meters).
  • a single dose of a composition for oral administration can contain from about 20 to about 600 mg, and in certain aspects from about 20 to about 400 mg, in another aspect from about 20 to about 300 mg, and in yet another aspect from about 20 to about 200 mg of the present compound per square meter of subject body surface area.
  • compositions for parenteral administration can be prepared in a manner such that a single dose contains at least about 20 mg of the present compound per square meter of subject body surface area, or at least about 40, 50, 100, 150, 200, 300, 400, or 500 mg of the present compound per square meter of subject body surface area.
  • a single dose in one or more parenteral preparations contains from about 20 to about 500 mg, and in certain aspects from about 20 to about 400 mg, and in another aspect from about 20 to about 450 mg, and in yet another aspect from about 20 to about 350 mg of the present compound per square meter of subject body surface area.
  • these oral and parenteral dosage ranges represent generally preferred dosage ranges, and are not intended to limit the invention.
  • the dosage regimen actually employed can vary widely, and, therefore, can deviate from the generally preferred dosage regimen. It is contemplated that one skilled in the art will tailor these ranges to the individual subject.
  • Toxicity and therapeutic efficacy of such compounds and bioconjugates can be determined by standard pharmaceutical procedures in cell cultures or experimental animals for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 , (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index that can be expressed as the ratio LD 5 o/ED 50 .
  • Compounds and bioconjugates that exhibit large therapeutic indices are preferred. While compounds and bioconjugates exhibiting toxic side effects can be used, care should be taken to design a delivery system that targets such compounds and bioconjugates to the site affected by the disease or disorder in order to minimize potential damage to unaffected cells and reduce side effects.
  • the dosage can vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective amount can be estimated initially from cell culture assays.
  • a dosage can be formulated in animal models to achieve a circulating plasma
  • concentration range that includes the ED 50 (the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful dosages in humans and other mammals.
  • Compound and bioconjugate levels in plasma can be measured, for example, by high performance liquid chromatography.
  • An amount of a compound or bioconjugate that can be combined with a pharmaceutically acceptable carrier to produce a single dosage form will vary depending upon the patient treated and the particular mode of administration. It will be appreciated by those skilled in the art that the unit content of a compound/bioconjugate contained in an individual dose of each dosage form need not in itself constitute a therapeutically effective amount, as the necessary therapeutically effective amount could be reached by administration of a number of individual doses. The selection of dosage depends upon the dosage form utilized, the condition being treated, and the particular purpose to be achieved according to the determination of those skilled in the art.
  • the dosage and dosage regime for treating a disease or condition can be selected in accordance with a variety of factors, including the type, age, weight, sex, diet and/or medical condition of the patient, the route of administration, pharmacological considerations such as activity, efficacy, pharmacokinetic and/or toxicology profiles of the particular
  • the dosage regime actually employed can vary widely from subject to subject, or disease to disease and different routes of administration can be employed in different clinical settings.
  • compositions/formulations of the present invention comprise a therapeutically effective amount (which can optionally include a diagnostically effective amount) of at least one compound or bioconjugate of the present invention.
  • Subjects receiving treatment that includes a compound/bioconjugate of the invention are preferably animals (e.g., mammals, reptiles and/or avians), more preferably humans, horses, cows, dogs, cats, sheep, pigs, and/or chickens, and most preferably humans.
  • the preferred composition depends on the route of administration. Any route of administration can be used as long as the target of the compound or pharmaceutically acceptable salt is available via that route. Suitable routes of administration include, for example, oral, intravenous, parenteral, inhalation, rectal, nasal, topical (e.g., transdermal and intraocular), intravesical, intrathecal, enteral, pulmonary, intralymphatic, intracavital, vaginal, transurethral, intradermal, aural, intramammary, buccal, orthotopic, intratracheal, intralesional, percutaneous, endoscopical, transmucosal, sublingual, and intestinal administration.
  • routes of administration include, for example, oral, intravenous, parenteral, inhalation, rectal, nasal, topical (e.g., transdermal and intraocular), intravesical, intrathecal, enteral, pulmonary, intralymphatic, intracavital, vaginal, transurethral, intradermal, aural, intramamm
  • the invention provides a method for treating a medical condition comprising administering to a subject (e.g. patient) in need thereof, a therapeutically effective amount of a composition of the invention, such as a compound of any one of formulas (FX1) - (FX59).
  • a composition of the invention such as a compound of any one of formulas (FX1) - (FX59).
  • the invention provides a method for diagnosing or aiding in the diagnosis of a medical condition comprising administering to a subject in need thereof, a diagnostically effective amount of a composition of the invention.
  • the medical condition is cancer, or various other diseases, injuries, and disorders, including cardiovascular disorders such as atherosclerosis and vascular restenosis, inflammatory diseases, ophthalmic diseases and dermatological diseases.
  • the diagnostic and therapeutic formulations of this invention can be administered alone, but can be administered with a pharmaceutical carrier selected upon the basis of the chosen route of administration and standard pharmaceutical practice.
  • diagnostic and therapeutic formulations of the invention can be administered intravenously, in oral dosage forms, intraperitoneally,
  • compositions, preparations and formulations can be formulated into diagnostic or therapeutic compositions for enteral, parenteral, topical, aerosol, inhalation, or cutaneous administration.
  • Topical or cutaneous delivery of the compositions, preparations and formulations can also include aerosol formulation, creams, gels, solutions, etc.
  • the present compositions, preparations and formulations are administered in doses effective to achieve the desired diagnostic and/or therapeutic effect. Such doses can vary widely depending upon the particular compositions employed in the composition, the organs or tissues to be examined, the equipment employed in the clinical procedure, the efficacy of the treatment achieved, and the like.
  • These compositions, preparations and formulations contain an effective amount of the
  • compositions along with conventional pharmaceutical carriers and excipients appropriate for the type of administration contemplated.
  • compositions, preparations and formulations can also optionally include stabilizing agents and skin penetration enhancing agents,
  • Compounds and bioconjugates of the present invention can be formulated for parenteral administration by injection (e.g., by bolus injection or continuous infusion).
  • Formulations for injection can be presented in unit dosage form in ampoules or in multi-dose containers with an optional preservative added.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass, plastic or the like.
  • the formulation can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • a parenteral preparation can be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent (e.g., as a solution in 1 ,3-butanediol).
  • a nontoxic parenterally acceptable diluent or solvent e.g., as a solution in 1 ,3-butanediol.
  • acceptable vehicles and solvents that can be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or di-glycerides.
  • fatty acids such as oleic acid can be used in the parenteral preparation.
  • compounds and bioconjugates of the present invention can be formulated in powder form for constitution with a suitable vehicle, such as sterile pyrogen-free water, before use.
  • a compound/bioconjugate suitable for parenteral administration can include a sterile isotonic saline solution containing between 0.1 percent and 90 percent weight per volume of the compound/bioconjugate.
  • a solution can contain from about 5 percent to about 20 percent, more preferably from about 5 percent to about 17 percent, more preferably from about 8 to about 14 percent, and still more preferably about 10 percent weight per volume of the compound/bioconjugate.
  • the solution or powder preparation can also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • a solubilizing agent such as lignocaine
  • a local anesthetic such as lignocaine
  • a compound/bioconjugate of the invention can be formulated to take the form of tablets or capsules prepared by conventional means with one or more
  • pharmaceutically acceptable carriers e.g., excipients such as binding agents, fillers, lubricants and disintegrants.
  • Controlled-release (or sustained-release) preparations can be formulated to extend the activity of a compound/bioconjugate and reduce dosage frequency. Controlled-release
  • preparations can also be used to effect the time of onset of action or other characteristics, such as blood levels of the compound/bioconjugate, and consequently affect the occurrence of side effects.
  • Controlled-release preparations can be designed to initially release an amount of a compound/bioconjugate that produces the desired therapeutic effect, and gradually and continually release other amounts of the compound/bioconjugate to maintain the level of therapeutic effect over an extended period of time. In order to maintain a near-constant level of a compound/bioconjugate that produces the desired therapeutic effect, and gradually and continually release other amounts of the compound/bioconjugate to maintain the level of therapeutic effect over an extended period of time. In order to maintain a near-constant level of a
  • Controlled-release systems can include, for example, an infusion pump which can be used to administer the compound/bioconjugate in a manner similar to that used for delivering insulin or chemotherapy to the body generally, or to specific organs or tumors.
  • the compound/bioconjugate is administered in combination with a biodegradable, biocompatible polymeric implant that releases the compound/bioconjugate over a controlled period of time at a selected site.
  • a biodegradable, biocompatible polymeric implant that releases the compound/bioconjugate over a controlled period of time at a selected site.
  • polymeric materials include polyanhydrides,
  • a controlled release system can be placed in proximity of a therapeutic target (e.g., organ, tissue, or group of cells), thus requiring only a fraction of a systemic dosage.
  • a therapeutic target e.g., organ, tissue, or group of cells
  • Compounds/bioconjugates of the invention can be administered by other controlled- release means or delivery devices that are well known to those of ordinary skill in the art. These include, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or the like, or a combination of any of the above to provide the desired release profile in varying proportions. Other methods of controlled-release delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention,
  • Compounds/bioconjugates of the invention can be administered directly to the lung of a patient/subject by inhalation.
  • a compound/bioconjugate can be conveniently delivered to the lung by a number of different devices.
  • a Metered Dose Inhaler which utilizes canisters that contain a suitable low boiling point propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas can be used to deliver a compound/bioconjugate directly to the lung.
  • MDI devices are available from a number of suppliers such as 3M Corporation, Aventis, Boehringer Ingleheim, Forest Laboratories, GlaxoSmithKline, Merck & Co. and Vectura.
  • a Dry Powder Inhaler (DPI) device can be used to administer a dry Powder Inhaler (DPI) device.
  • DPI Dry Powder Inhaler
  • DPI devices typically use a mechanism such as a burst of gas to create a cloud of dry powder inside a container, which can then be inhaled by the patient.
  • DPI devices are also well known in the art and can be purchased from a number of vendors which include, for example, GlaxoSmithKline, Nektar Therapeutics, Innovata and Vectura.
  • a popular variation is the multiple dose DPI ("MDDPI") system, which allows for the delivery of more than one therapeutic dose.
  • MDDPI devices are available from companies such as AstraZeneca, GlaxoSmithKline, TEVA, Merck & Co., SkyePharma and Vectura.
  • capsules and cartridges of gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound/bioconjugate and a suitable powder base such as lactose or starch for these systems.
  • a liquid spray device supplied, for example, by Aradigm Corporation.
  • Liquid spray systems use extremely small nozzle holes to aerosolize liquid compound/bioconjugate formulations that can then be directly inhaled into the lung.
  • a nebulizer device can be used to deliver a compound/bioconjugate to the lung. Nebulizers create aerosols from liquid
  • nebulizers include devices supplied by Aventis and Battelle.
  • an electrohydrodynamic (“EHD”) aerosol device can be used to deliver a compound/bioconjugate to the lung.
  • EHD aerosol devices use electrical energy to aerosolize liquid compound/bioconjugate solutions or suspensions.
  • the electrochemical properties of the compound/bioconjugate formulation are important parameters to optimize when delivering this compound/bioconjugate to the lung with an EHD aerosol device. Such optimization is routinely performed by one of skill in the art.
  • Liquid compound/bioconjugate formulations suitable for use with nebulizers and liquid spray devices and EHD aerosol devices will typically include the compound/bioconjugate with a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier is a liquid such as alcohol, water, polyethylene glycol or a perfluorocarbon.
  • another material can be added to alter the aerosol properties of the solution or suspension of the compound/bioconjugate.
  • this material can be a liquid such as an alcohol, glycol, polyglycol or a fatty acid.
  • a compound/bioconjugate of the invention can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Accordingly, the compound/bioconjugate can be formulated with suitable polymeric or hydrophobic materials such as an emulsion in an acceptable oil or ion exchange resin, or as sparingly soluble derivatives such as a sparingly soluble salt. Other methods of depot delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
  • a compound/bioconjugate can be combined with a
  • a topical formulation of a compound/bioconjugate can be applied to the skin.
  • the pharmaceutically acceptable carrier can be in the form of, for example, and not by way of limitation, an ointment, cream, gel, paste, foam, aerosol, suppository, pad or gelled stick.
  • a topical formulation can include a therapeutically effective amount of a compound/bioconjugate in an ophthalmologically acceptable excipient such as buffered saline, mineral oil, vegetable oils such as corn or arachis oil, petroleum jelly, Miglyol 182, alcohol solutions, or liposomes or liposome-like products. Any of these formulations of such
  • compounds/bioconjugates can include preservatives, antioxidants, antibiotics,
  • immunosuppressants and other biologically or pharmaceutically effective agents that do not exert a significant detrimental effect on the compound/bioconjugate.
  • Other methods of topical delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
  • Compounds/bioconjugates of the invention can be formulated in rectal formulations such as suppositories or retention enemas that include conventional suppository bases such as cocoa butter or other glycerides and/or binders and/or carriers such as triglycerides, microcrystalline cellulose, gum tragacanth or gelatin. Rectal formulations can contain a compound/bioconjugate in the range of 0.5% to 10% by weight, for example. Other methods of rectal delivery of
  • the invention provides a medicament which comprises a
  • the invention provides a medicament which comprises a diagnostically effective amount of one or more compositions of the invention.
  • the invention provides a method for making a medicament for treatment of a condition described herein, such as the treatment of cancer, inflammation, stenosis or a vascular disease.
  • the invention provides a method for making a medicament for diagnosis or aiding in the diagnosis of a condition described herein, such as the diagnosis of cancer, inflammation, stenosis or a vascular disease.
  • the invention provides the use of one or more compositions set forth herein for the making of a medicament for the treatment of cancer, inflammation, stenosis or a vascular disease.
  • the invention provides the use of one or more compositions set forth herein for the treatment of a disease.
  • the invention provides the use of one or more compositions set forth herein for the diagnosis of a disease.
  • Compositions of the invention include formulations and preparations comprising one or more of the present optical agents provided in an aqueous solution, such as a pharmaceutically acceptable formulation or preparation.
  • compositions of the invention further comprise one or more pharmaceutically acceptable surfactants, buffers, electrolytes, salts, carriers, binders, coatings, preservatives and/or excipients.
  • the invention provides a pharmaceutical formulation having an active ingredient comprising a composition of the invention, such as a compound of any one of formulas (FX1) - (FX59).
  • the invention provides a method of synthesizing a composition of the invention or a pharmaceutical formulation thereof, such as a compound of any one of formulas (FX1) - (FX59).
  • a pharmaceutical formulation comprises one or more excipients, carriers, diluents, and/or other components as would be understood in the art.
  • the components meet the standards of the National Formulary ("NF"), United States Pharmacopoeia (“USP”; United States Pharmacopeial Convention Inc., Rockville, Maryland), or Handbook of Pharmaceutical Manufacturing Formulations (Sarfaraz K. Niazi, all volumes, ISBN: 9780849317521 , ISBN 10: 0849317525; CRC Press, 2004). See, e.g., United States
  • the formulation base of the formulations of the invention comprises
  • physiologically acceptable excipients namely, at least one binder and optionally other
  • physiologically acceptable excipients are those known to be usable in the pharmaceutical technology sectors and adjacent areas, particularly, those listed in relevant pharmacopeias (e.g. DAB, Ph. Eur., BP, NF, USP), as well as other excipients whose properties do not impair a physiological use.
  • compositions including a therapeutically effective amount of a compound or salt of this invention, as well as processes for making such compositions.
  • Such compositions generally include one or more pharmaceutically acceptable carriers (e.g., excipients, vehicles, auxiliaries, adjuvants, diluents) and can include other active ingredients.
  • pharmaceutically acceptable carriers e.g., excipients, vehicles, auxiliaries, adjuvants, diluents
  • Formulation of these compositions can be achieved by various methods known in the art. A general discussion of these methods can be found in, for example, Hoover, John E., Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA: 1975). See also, Lachman, L, eds., Pharmaceutical Dosage Forms (Marcel Decker, New York, N. Y., 1980).
  • compositions and medicaments of this invention can further comprise one or more pharmaceutically acceptable carriers, excipients, buffers, emulsifiers, surfactants, electrolytes or diluents.
  • pharmaceutically acceptable carriers such as, for example, those described in Remingtons Pharmaceutical Sciences, 17th edition, ed. Alfonoso R. Gennaro, Mack Publishing Company, Easton, Pa. (1985).
  • compositions of the invention include formulations and preparations comprising one or more of the present compounds provided in an aqueous solution, such as a pharmaceutically acceptable formulation or preparation.
  • compositions of the invention further comprise one or more pharmaceutically acceptable surfactants, buffers, electrolytes, salts, carriers, binders, coatings, preservatives and/or excipients.
  • Compounds and bioconjugates of the present invention can be formulated by known methods for administration to a subject using several routes which include, but are not limited to, parenteral, oral, topical, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and ophthalmic routes.
  • An individual compound/bioconjugate can be administered in combination with one or more additional compounds/bioconjugates of the present invention and/or together with other biologically active or biologically inert agents.
  • Such biologically active or inert agents can be in fluid or mechanical communication with the
  • administration is localized in a subject, but administration can also be systemic.
  • Compounds and bioconjugates of the present invention can be formulated by any conventional manner using one or more pharmaceutically acceptable carriers.
  • the compound(s)/bioconjugate(s) and their pharmaceutically acceptable salts and solvates can be specifically formulated for administration, e.g. , by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.
  • compounds/bioconjugates can take the form of charged, neutral and/or other pharmaceutically acceptable salt forms.
  • pharmaceutically acceptable carriers include, but are not limited to, those described in REMINGTON'S PHARMACEUTICAL SCIENCES (A.R. Gennaro, Ed.), 20th edition, Williams & Wilkins PA, USA (2000).
  • Compounds and bioconjugates of the present invention can be formulated in the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, controlled- or sustained- release formulations and the like.
  • Such formulations will contain a therapeutically effective amount of the compound/bioconjugate, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • Pharmaceutically acceptable carriers that can be used in conjunction with the compounds of the invention are well known to those of ordinary skill in the art. Carriers can be selected based on a number of factors including, for example, the particular compound(s) or pharmaceutically acceptable salt(s) used; the compound's concentration, stability, and intended bioavailability; the condition being treated; the subject's age, size, and general condition; the route of administration; etc. A general discussion related to carriers can be found in, for example, J.G. Nairn, Remington's Pharmaceutical Science, pp. 1492-1517 (A. Gennaro, ed., Mack Publishing Co., Easton, Pa. (1985)).
  • Solid dosage forms for oral administration include, for example, capsules, tablets, gelcaps, pills, dragees, troches, powders, granules, and lozenges.
  • the compounds or pharmaceutically acceptable salts thereof can be combined with one or more pharmaceutically acceptable carriers.
  • the compounds and pharmaceutically acceptable salts thereof can be mixed with carriers including, but not limited to, lactose, sucrose, starch powder, corn starch, potato starch, magnesium carbonate, microcrystalline cellulose, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, sodium carbonate, agar, mannitol, sorbitol, sodium saccharin, gelatin, acacia gum, alginic acid, sodium alginate, tragacanth, colloidal silicon dioxide, croscarmellose sodium, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration.
  • carriers including, but not limited to, lactose, sucrose, starch powder, corn starch, potato starch, magnesium carbonate, microcrystalline cellulose, cellulose esters of alkanoic acids, cellulose alkyl est
  • Such capsules or tablets can contain a controlled-release formulation, as can be provided in a dispersion of the compound or salt in hydroxypropylmethyl cellulose.
  • the dosage forms also can include buffering agents, such as sodium citrate, or magnesium or calcium carbonate or bicarbonate. Tablets and pills additionally can, for example, include a coating (e.g., an enteric coating) to delay disintegration and absorption.
  • the concentration of the present compounds in a solid oral dosage form can be from about 5 to about 50% for example, and in certain aspects from about 8 to about 40%, and in another aspect from about 10 to about 30% by weight based on the total weight of the composition.
  • Liquid dosage forms of the compounds of the invention for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also can include adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
  • the concentration of the present compounds in the liquid dosage form can be from about 0.01 to about 5 mg, and in certain aspects from about 0.01 to about 1 mg, and in another aspect from about 0.01 to about 0.5 mg per ml of the composition.
  • Low concentrations of the compounds of the invention in liquid dosage form can be prepared in the case that the compound is more soluble at low concentrations.
  • Techniques for making oral dosage forms useful in the invention are generally described in, for example, Modern Pharmaceutics, Chapters 9 and 10 (Banker & Rhodes, Editors (1979)). See also, Lieberman et al., Pharmaceutical Dosage Forms: Tablets (1981 ). See also, Ansel, Introduction to Pharmaceutical Dosage Forms (2nd Edition (1976)).
  • tablets or powders for oral administration can be prepared by dissolving the compound in a pharmaceutically acceptable solvent capable of dissolving the compound to form a solution and then evaporating when the solution is dried under vacuum.
  • a carrier can also be added to the solution before drying.
  • the resulting solution can be dried under vacuum to form a glass.
  • the glass can then be mixed with a binder to form a powder.
  • This powder can be mixed with fillers or other conventional tableting agents, and then processed to form a tablet.
  • the powder can be added to a liquid carrier to form a solution, emulsion, suspension, or the like.
  • solutions for oral administration are prepared by dissolving the compound in a pharmaceutically acceptable solvent capable of dissolving the compound to form a solution.
  • An appropriate volume of a carrier is added to the solution while stirring to form a pharmaceutically acceptable solution for oral administration.
  • a liposome or micelle can be utilized as a carrier or vehicle for the composition.
  • the compound can be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, can be on the external surface of the liposome or micelle.
  • a targeting ligand can be externally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains the optical agents) to the desired tissue, organ, or other site in the body.
  • Injectable preparations can be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
  • Acceptable vehicles for parenteral use include both aqueous and nonaqueous pharmaceutically-acceptable solvents.
  • Suitable pharmaceutically acceptable aqueous solvents include, for example, water, saline solutions, dextrose solutions (such as DW5), electrolyte solutions, etc.
  • the present compounds are formulated as nanoparticles or microparticles.
  • Use of such nanoparticle or microparticle formulations can be beneficial for some applications to enhance delivery, localization, target specificity, administration, etc. of the compound.
  • Potentially useful nanoparticles and microparticles include, but are not limited to, micelles, liposomes, microemulsions, nanoemulsions, vesicles, tubular micelles, cylindrical micelles, bilayers, folded sheets structures, globular aggregates, swollen micelles, inclusion complex, encapsulated droplets, microcapsules, nanocapsules or the like.
  • the present compounds can be located inside the nanoparticle or microparticle, within a membrane or wall of the nanoparticle or microparticle, or outside of (but bonded to or otherwise associated with) the nanoparticle or microparticle.
  • the agent formulated in nanoparticles or microparticles can be administered by any of the routes previously described. In a formulation applied topically, the compound is slowly released over time. In an injectable formulation, the liposome, micelle, capsule, etc., circulates in the bloodstream and is delivered to the desired site (e.g., target tissue).
  • liposomes can be prepared from dipalmitoyl phosphatidylcholine (DPPC) or egg phosphatidylcholine (PC) because this lipid has a low heat transition.
  • DPPC dipalmitoyl phosphatidylcholine
  • PC egg phosphatidylcholine
  • Liposomes are made using standard procedures as known to one skilled in the art (e.g., Braun-Falco et al., (Eds.), Griesbach Conference, Liposome Dermatics, Springer- Verlag, Berlin (1992), pp. 69 81 ; 91 117.
  • Polycaprolactone, poly(glycolic) acid, poly(lactic) acid, polyanhydride or lipids can be formulated as microspheres.
  • the present compounds can be mixed with polyvinyl alcohol (PVA), the mixture then dried and coated with ethylene vinyl acetate, then cooled again with PVA.
  • PVA polyvinyl alcohol
  • the present compounds can be within one or both lipid bilayers, in the aqueous between the bilayers, or within the center or core.
  • Liposomes can be modified with other molecules and lipids to form a cationic liposome. Liposomes can also be modified with lipids to render their surface more hydrophilic which increases their circulation time in the bloodstream.
  • the thus-modified liposome has been termed a "stealth" liposome, or a long-lived liposome, as described in U.S. Pat. No. 6,258,378, and in Stealth Liposomes, Lasic and Martin (Eds.) 1995 CRC Press, London.
  • Encapsulation methods include detergent dialysis, freeze drying, film forming, injection, as known to one skilled in the art and disclosed in, for example, U.S. Pat. No. 6,406,713.
  • compositions and methods include a micelle delivery system, for example, involving one or more PEG-based amphiphilic polymers developed for drug delivery including: PEG-poly(s-caprolactone), PEG-poly(amino acid), PEG-polylactide or PEG- phospholipid constructs; a cross linked poly(acrylic acid) polymer system, a phospholipid-based system and/or block copolymer systems comprising one or more of the following polymer blocks: a poly(lactic acid) polymer block; a poly(propylene glycol) polymer block; a poly(amino acid) polymer block; a poly(ester) polymer block; a poly ( ⁇ -caprolactone) polymer block; a poly(ethylene glycol) block, a poly(acrylic acid) block; a polylactide block; a polyester block; a polyamide block; a polyanhydride block; a polyurethane block; a polyimine block; a poly
  • Suitable pharmaceutically-acceptable nonaqueous solvents include, but are not limited to, the following (as well as mixtures thereof):
  • Alcohols (these include, for example, ⁇ -glycerol formal, ⁇ -glycerol formal, 1 , 3- butyleneglycol, aliphatic or aromatic alcohols having from 2 to about 30 carbons (e.g., methanol, ethanol, propanol, isopropanol, butanol, t-butanol, hexanol, octanol, amylene hydrate, benzyl alcohol, glycerin (glycerol), glycol, hexylene, glycol, tetrahydrofuranyl alcohol, cetyl alcohol, and stearyl alcohol), fatty acid esters of fatty alcohols (e.g., polyalkylene glycols, such as
  • Amides which include, for example, dimethylacetamide (DMA), benzyl benzoate DMA, dimethylformamide, N-hydroxyethyO-lactamide, N, N-dimethylacetamide-amides, 2- pyrrolidinone, 1-methyl-2-pyrrolidinone, and polyvinylpyrrolidone;
  • DMA dimethylacetamide
  • benzyl benzoate DMA dimethylformamide
  • N-hydroxyethyO-lactamide N, N-dimethylacetamide-amides
  • 2- pyrrolidinone 1-methyl-2-pyrrolidinone
  • polyvinylpyrrolidone polyvinylpyrrolidone
  • Esters which include, for example, acetate esters (e.g., monoacetin, diacetin, and triacetin), aliphatic and aromatic esters (e.g., ethyl caprylate or octanoate, alkyl oleate, benzyl benzoate, or benzyl acetate), dimethylsulfoxide (DMSO), esters of glycerin (e.g., mono, di, and tri- glyceryl citrates and tartrates), ethyl benzoate, ethyl acetate, ethyl carbonate, ethyl lactate, ethyl oleate, fatty acid esters of sorbitan, glyceryl monostearate, glyceride esters (e.g., mono, di, or triglycerides), fatty acid esters (e.g., isopropyl myristrate), aliphatic and aromatic esters
  • 20 mono ricinoleate polyoxyethylene sorbitan esters (e.g., polyoxyethylene-sorbitan monooleate, polyoxyethylene-sorbitan monopalmitate, polyoxyethylene-sorbitan monolaurate, polyoxyethylene- sorbitan monostearate, and POLYSORBATE 20, 40, 60, and 80 (from ICI Americas, Wilmington, DE)), polyvinylpyrrolidone, alkyleneoxy modified fatty acid esters (e.g., polyoxyl 40 hydrogenated castor oil and polyoxyethylated castor oils, such as CREMOPHOR EL solution or CREMOPHOR RH 40 solution), saccharide fatty acid esters (i.e., the condensation product of a monosaccharide (e.g., pentoses, such as, ribose, ribulose, arabinose, xylose, lyxose, and xylu
  • Ethers for example, alkyl, aryl, and cyclic ethers having from 2 to about 30 carbons. Examples include diethyl ether, tetrahydrofuran, dimethyl isosorbide, diethylene glycol monoethyl ether), and glycofurol (tetrahydrofurfuranyl alcohol polyethylene glycol ether);
  • Ketones which typically have from about 3 to about 30 carbons. Examples include acetone, methyl ethyl ketone, and methyl isobutyl ketone;
  • Hydrocarbons which are typically aliphatic, cycloaliphatic, or aromatic hydrocarbons having from about 4 to about 30 carbons. Examples include benzene, cyclohexane,
  • dichloromethane dioxolanes, hexane, n-decane, n-dodecane, n-hexane, sulfolane,
  • Oils which include, for example, oils of mineral, vegetable, animal, essential, or synthetic origin. These include: mineral oils, such as aliphatic and wax-based hydrocarbons, aromatic hydrocarbons, mixed aliphatic and aromatic based hydrocarbons, and refined paraffin oil; vegetable oils, such as linseed, tung, safflower, soybean, castor, cottonseed, groundnut, rapeseed, coconut, palm, olive, corn, corn germ, sesame, persic, and peanut oil; glycerides, such as mono-, di-, and triglycerides; animal oils, such as fish, marine, sperm, cod-liver, haliver, squaiene, squalane, and shark liver oil; oleic oils; and polyoxyethylated castor oil;
  • mineral oils such as aliphatic and wax-based hydrocarbons, aromatic hydrocarbons, mixed aliphatic and aromatic based hydrocarbons, and refined paraffin oil
  • vegetable oils such as l
  • Alkyl, alkenyl, or aryl halides which include, for example, alkyl or aryl halides having from 1 to about 30 carbons and one or more halogen substituents.
  • Examples include: methylene chloride; monoethanolamine; petroleum benzin; trolamine; omega-3 polyunsaturated fatty acids (e.g., alpha-linolenic acid, eicosapentaenoic acid, docosapentaenoic acid, or docosahexaenoic acid); polyglycol ester of 12-hydroxystearic acid and polyethylene glycol (SOLUTOL HS-15, from BASF, Ludwigshafen, Germany); polyoxyethylene glycerol; sodium laurate; sodium oleate; and sorbitan monooleate.
  • Solvents useful in the invention include, but are not limited to, those known to stabilize present compounds or pharmaceutically acceptable salts thereof. These can include, for example, oils rich in triglycerides, such as safflower oil, soybean oil, and mixtures thereof; and alkyleneoxy- modified fatty acid esters, such as polyoxyl 40 hydrogenated castor oil and polyoxyethylated castor oils (e.g., CREMOPHOR EL solution or CREMOPHOR RH 40 solution).
  • triglycerides include INTRALIPID emulsified soybean oil (Kabi-Pharmacia Inc.,
  • compositions of this invention for various purposes generally known in the pharmaceutical industry. These components tend to impart properties that, for example, enhance retention of the present compounds or salt thereof at the site of administration, protect the stability of the composition, control the pH, and facilitate processing of the compound or salt thereof into pharmaceutical formulations, and the like.
  • cryoprotective agents include cryoprotective agents; agents for preventing reprecipitation of the compound or salt surface; active, wetting, or emulsifying agents (e.g., lecithin, polysorbate-80, TWEEN 80, pluronic 60, and polyoxyethylene stearate); preservatives (e.g., ethyl-p- hydroxybenzoate); microbial preservatives (e.g., benzyl alcohol, phenol, m-cresol, chlorobutanol, sorbic acid, thimerosal, and paraben); agents for adjusting pH or buffering agents (e.g., acids, bases, sodium acetate, sorbitan monolaurate, etc.); agents for adjusting osmolarity (e.g., glycerin); thickeners (e.g., aluminum monostearate, stearic acid, cetyl alcohol, stearyl alcohol, guar gum, methyl cellulose, hydroxypropy
  • metabisulfite sodium bisulfite, sodium sulfite, dextrose, phenols, thiophenols, etc.
  • Formulations for parenteral administration can be prepared from one or more sterile powders and/or granules having a compound or salt of this invention and one or more of the carriers or diluents mentioned for use in the formulations for oral administration.
  • the powder or granule typically is added to an appropriate volume of a solvent (typically while agitating (e.g., stirring) the solvent) that is capable of dissolving the powder or granule.
  • Particular solvents useful in the invention include, for example, water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers.
  • Emulsions for parenteral administration can be prepared by, for example, dissolving a compound or salt of this invention in any pharmaceutically acceptable solvent capable of dissolving the compound to form a solution; and adding an appropriate volume of a carrier to the solution while stirring to form the emulsion.
  • Solutions for parenteral administration can be prepared by, for example, dissolving a compound or salt of this invention in any pharmaceutically acceptable solvent capable of dissolving the compound to form a solution; and adding an appropriate volume of a carrier to the solution while stirring to form the solution.
  • Suppositories for rectal administration can be prepared by, for example, mixing the drug with a suitable nonirritating excipient that is solid at ordinary temperatures, but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable excipients include, for example, cocoa butter; synthetic mono-, di-, or triglycerides; fatty acids; and/or polyethylene glycols.
  • Binding agents include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl
  • microcrystalline cellulose include, for example, the materials sold as AVICEL-PH-101 , AVICEL- PH-103 and AVICEL-PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pennsylvania, USA).
  • An exemplary suitable binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581 by FMC Corporation.
  • Fillers include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), lactose, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Lubricants include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, electromagnetic radiation mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laurate, agar, and mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co.
  • Disintegrants include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Tablets or capsules can optionally be coated by methods well known in the art. If binders and/or fillers are used with a compound/bioconjugate of the invention, they are typically formulated as about 50 to about 99 weight percent of the compound/bioconjugate. In one aspect, about 0.5 to about 15 weight percent of disintegrant, and particularly about 1 to about 5 weight percent of disintegrant, can be used in combination with the compound. A lubricant can optionally be added, typically in an amount of less than about 1 weight percent of the compound/bioconjugate.
  • Liquid preparations for oral administration can take the form of solutions, syrups or suspensions. Alternatively, the liquid preparations can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and/or preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid).
  • suspending agents e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats
  • emulsifying agents e.g., lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters,
  • the preparations can also contain buffer salts, flavoring, coloring, perfuming and sweetening agents as appropriate.
  • Preparations for oral administration can also be formulated to achieve controlled release of the compound/bioconjugate.
  • Oral formulations preferably contain 10% to 95% compound/bioconjugate.
  • a compound/bioconjugate of the present invention can be formulated for buccal administration in the form of tablets or lozenges formulated in a conventional manner.
  • Other methods of oral delivery of compounds/bioconjugates of the invention will be known to the skilled artisan and are within the scope of the invention.
  • Hard gelatin capsules are prepared using the following ingredients:
  • a tablet formula is prepared using the following ingredients:
  • a dry powder inhaler formulation is prepared containing the following components:
  • Lactose 95 [0316] The active ingredient is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
  • Tablets each containing 60 mg of active ingredient, are prepared as follows:
  • the active ingredient, starch and cellulose are passed through a No. 20 mesh U.S. sieve and mixed thoroughly.
  • the solution of polyvinylpyrrolidone is mixed with the resultant powders which are then passed through a 16 mesh U.S. sieve.
  • the granules as produced are dried at 50- 60 °C and passed through a 16 mesh U.S. sieve.
  • the sodium carboxymethyl starch, magnesium stearate, and talc previously passed through a No. 30 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
  • Capsules each containing 80 mg of active ingredient are made as follows:
  • Suppositories each containing 225 mg of active ingredient, are made as follows:
  • the active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed to cool.
  • Suspensions each containing 50 mg of active ingredient per 5.0 ml dose are made as follows:
  • the active ingredient, sucrose and xantham gum are blended, passed through a No. 10 mesh U.S. sieve, and mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water.
  • the sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
  • Capsules each containing 150 mg of active ingredient, are made as follows:
  • kits can include a compound/bioconjugate of the present invention, optionally one or more ingredients for preparing a pharmaceutically acceptable formulation of the compound/bioconjugate, and instructions for use (e.g., administration).
  • instructions for use e.g., administration
  • different components of a compound/bioconjugate formulation can be packaged in separate containers and admixed immediately before use.
  • Such packaging of the components separately can, if desired, be presented in a pack or dispenser device which can contain one or more unit dosage forms containing the compound/bioconjugate.
  • the pack can, for example, comprise metal or plastic foil such as a blister pack.
  • Such packaging of the components separately can also, in certain instances, permit long-term storage without losing activity of the components.
  • the different components can be packaged separately and not mixed prior to use. In various embodiments, the different components can be packaged in one combination for administration together.
  • the compounds and salts of this invention can be used in the form of a kit that is suitable for use in. performing the methods described herein, packaged in a container.
  • the kit can contain the compound or compounds and, optionally, appropriate diluents, devices or device components suitable for administration and instructions for use in accordance with the methods of the invention.
  • the devices can include parenteral injection devices, such as syringes or transdermal patch or the like.
  • Device components can include cartridges for use in injection devices and the like.
  • the kit includes a first dosage form including a compound or salt of this invention and a second dosage form including another active ingredient in quantities sufficient to carry out the methods of the invention.
  • the first dosage form and the second dosage form together can include a therapeutically effective amount of the compounds for treating the targeted condition(s).
  • kits can be supplied with instructional materials. Instructions can be printed on paper or other substrate, and/or can be supplied as an electronic-readable medium, such as a floppy disc, mini-CD-ROM, CD-ROM, DVD-ROM, Zip disc, videotape, audio tape, and the like. Detailed instructions cannot be physically associated with the kit; instead, a user can be directed to an Internet web site specified by the manufacturer or distributor of the kit, or supplied as electronic mail.
  • administration can be packaged in IV bags, vials, or other conventional containers in concentrated form, and then diluted with a pharmaceutically acceptable liquid (e.g., saline) to form an acceptable compound concentration before use.
  • a pharmaceutically acceptable liquid e.g., saline
  • Kits can include reagents in separate containers such as, for example, sterile water or saline to be added to a lyophilized active component packaged separately.
  • sealed glass ampules can contain lyophilized superoxide dismutase mimetics and in a separate ampule, sterile water, sterile saline or sterile each of which has been packaged under a neutral non- reacting gas, such as nitrogen.
  • Ampules can consist of any suitable material, such as glass, organic polymers, such as polycarbonate, polystyrene, ceramic, metal or any other material typically employed to hold reagents.
  • suitable containers include bottles that can be fabricated from similar substances as ampules, and envelopes that can consist of foil-lined interiors, such as aluminum or an alloy.
  • Other containers include test tubes, vials, flasks, bottles, syringes, and the like.
  • Containers can have a sterile access port, such as a bottle having a stopper that can be pierced by a hypodermic injection needle.
  • Other containers can have two
  • Removable membranes can be glass, plastic, rubber, and the like.
  • isotopic variants of compounds disclosed herein are intended to be encompassed by the disclosure.
  • any one or more hydrogens in a molecule disclosed can be replaced with deuterium or tritium.
  • Isotopic variants of a molecule are generally useful as standards in assays for the molecule and in chemical and biological research related to the molecule or its use. Methods for making such isotopic variants are known in the art. Specific names of compounds are intended to be exemplary, as it is known that one of ordinary skill in the art can name the same compounds differently.
  • Optical agents of the present invention may be formulated with pharmaceutically- acceptable anions and/or cations.
  • Pharmaceutically-acceptable cations include among others, alkali metal cations (e.g., Li + , Na + , K + ), alkaline earth metal cations (e.g., Ca 2+ , Mg 2+ ), non-toxic heavy metal cations and ammonium (NH 4 + ) and substituted ammonium (N(R') 4 + , where R' is hydrogen, alkyl, or substituted alkyl, i.e., including, methyl, ethyl, or hydroxyethyl, specifically, trimethyl ammonium, triethyl ammonium, and triethanol ammonium cations).
  • alkali metal cations e.g., Li + , Na + , K +
  • alkaline earth metal cations e.g., Ca 2+ , Mg 2+
  • Pharmaceutically- acceptable anions include among other halides (e.g., CI “ , Br “ ), sulfate, acetates (e.g., acetate, trifluoroacetate), ascorbates, aspartates, benzoates, citrates, and lactate.
  • a liposome or micelle may be utilized as a carrier or vehicle for the composition.
  • the sulfenamide compound may be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, may be on the external surface of the liposome or micelle.
  • a targeting ligand may be externally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains the sulfenamide optical agents) to the desired tissue, organ, or other site in the body.
  • compositions, preparations and formulations can be used both as a diagnostic agent as well as a phototherapy agent concomitantly.
  • an effective amount of the present compositions, preparations and formulations in a pharmaceutically acceptable formulation is administered to a patient.
  • Administration is followed by a procedure that combines photodiagnosis and phototherapy.
  • a composition comprising compounds for combined photodiagnosis and phototherapy is administered to a patient and its concentration, localization, or other parameters is determined at the target site of interest. More than one measurement may be taken to determine the location of the target site.
  • the time it takes for the compound to accumulate at the target site depends upon factors such as pharmcokinetics, and may range from about thirty minutes to two days.
  • the phototherapeutic part of the procedure may be done either immediately after determining the site or before the agent is cleared from the site. Clearance depends upon factors such as pharmacokinetics.
  • compositions, preparations and formulations can be formulated into diagnostic or therapeutic compositions for enteral, parenteral, topical, aerosol, inhalation, or cutaneous administration.
  • Topical or cutaneous delivery of the compositions, preparations and formulations may also include aerosol formulation, creams, gels, solutions, etc.
  • the present compositions, preparations and formulations are administered in doses effective to achieve the desired diagnostic and/or therapeutic effect. Such doses may vary widely depending upon the particular compositions employed in the composition, the organs or tissues to be examined, the equipment employed in the clinical procedure, the efficacy of the treatment achieved, and the like.
  • These compositions, preparations and formulations contain an effective amount of the
  • compositions along with conventional pharmaceutical carriers and excipients appropriate for the type of administration contemplated.
  • These compositions, preparations and formulations may also optionally include stabilizing agents and skin penetration enhancing agents.
  • Methods of this invention comprise the step of administering an "effective amount" of the present diagnostic and therapeutic compositions, formulations and preparations containing the present compounds, to diagnosis, image, monitor, evaluate, treat, reduce, alleviate, ameliorate or regulate a biological condition and/or disease state in a patient.
  • the term "effective amount,” as used herein, refers to the amount of the diagnostic and therapeutic formulation, that, when administered to the individual is effective diagnosis, image, monitor, evaluate, treat, reduce alleviate, ameliorate or regulate a biological condition and/or disease state.
  • the effective amount of a given composition or formulation will depend at least in part upon, the mode of administration (e.g.
  • any suitable form of administration can be employed in connection with the diagnostic and therapeutic formulations of the present invention.
  • the diagnostic and therapeutic formulations of this invention can be administered intravenously, in oral dosage forms, intraperitoneally, subcutaneously, or intramuscularly, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
  • the diagnostic and therapeutic formulations of this invention can be administered alone, but may be administered with a pharmaceutical carrier selected upon the basis of the chosen route of administration and standard pharmaceutical practice.
  • compositions and medicaments of this invention may further comprise one or more pharmaceutically acceptable carrier, excipient, buffer, emulsifier, surfactant, electrolyte or diluent.
  • pharmaceutically acceptable carrier such as, for example, those described in Remingtons Pharmaceutical Sciences, 17th edition, ed. Alfonoso R. Gennaro, Mack Publishing Company, Easton, Pa. (1985).
  • ranges specifically include the values provided as endpoint values of the range.
  • a range of 1 to 100 specifically includes the end point values of 1 and 100. It will be understood that any subranges or individual values in a range or subrange that are included in the description herein can be excluded from the claims herein.

Abstract

The invention relates generally to optical agents for biomedical applications, including phototherapy. Provided are sulfenamide compounds having an acyclic S-N bond with at least one aromatic and/or heterocyclic aromatic group providing phototherapeutic agents, including Type 1 phototherapeutic agents. Optical agents of the invention enable a versatile phototherapy platform for treatment of a range of pathological conditions, including the treatment of cancers, stensosis and inflammation. The invention further provides preparations and formulations comprising the sulfenamide optical agents and related methods of making and using sulfenamide optical agents in an in vivo or ex vivo biomedical procedure.

Description

SULFENA IDE COMPOUNDS FOR PHOTOTHERAPY
CROSS-REFERENCE TO RELATED APPLICATIONS
[001] This application claims the benefit of U.S. Provisional Application No. 61/260,078, entitled "Sulfenamide Compounds for Phototherapy", filed November 1 1 , 2009, which is incorporated by reference to the extent not inconsistent herewith.
BACKGROUND
[002] Optical agents currently play a central role in a large number of in vivo, in vitro and ex vivo clinical procedures including important diagnostic and therapeutic procedures.
Photodiagnostic and phototherapeutic agents, for example, include a class of molecules capable of absorbing, emitting, or scattering electromagnetic radiation applied to a biological material, particularly in the visible and near infrared regions of the electromagnetic spectrum. This property of optical agents is used in a range of biomedical applications for visualizing, imaging or otherwise characterizing biological materials and/or achieving a desired therapeutic outcome. Recent developments in targeted administration and delivery of optical agents, and advanced systems and methods for applying and detecting electromagnetic radiation in biological environments has considerably expanded the applicability and effectiveness of optical agents for clinical applications.
[003] Important applications of optical agents that absorb and/or emit in the visible and near- infrared (NIR) region of the electromagnetic spectrum include their use in biomedical imaging and visualization. For example, compounds absorbing and/or emitting light in these regions of the electromagnetic spectrum currently are useful for optical tomography, optoacoustic tomography, optical coherence tomography, confocal scanning laser tomography, optical coherence
tomography, and fluorescence endoscopy; techniques which have emerged as essential molecular imaging techniques for imaging and visualizing biological processes at the organ, cellular and subcellular (e.g., molecular) levels. Biomedical images are generated, for example, by detecting electromagnetic radiation, nuclear radiation, acoustic waves, electrical fields, and/or magnetic fields transmitted, emitted and/or scattered by components of a biological sample.
Modulation of the energy or intensity of the applied radiation yields patterns of transmitted, scattered and/or emitted radiation, acoustic waves, electrical fields or magnetic fields that contain useful anatomical, physiological, and/or biochemical information. A number of applications of biomedical imaging have matured into robust, widely used clinical techniques including planar projection and tomographic X-ray imaging, magnetic resonance imaging, ultrasound imaging, and gamma ray imaging.
[004] Established optical imaging and visualization techniques are based on monitoring spatial variations in a variety of optical parameters including the intensities, polarization states, and frequencies of transmitted, reflected, and emitted electromagnetic radiation. Given that many biological materials of interest are incompatible with ultraviolet light, research is currently directed to developing and enhancing imaging techniques using visible and near infrared (NIR) radiation (from about 400 nm to about 900 nm). In particular, NIR light (700 nm to 900 nm) is useful for visualizing and imaging deeper regions than visible light because electromagnetic radiation of this wavelength range is capable of substantial penetration (e.g. , up to four centimeters) in a range of biological media. Optical imaging and visualization using optical agents has potential to provide a less invasive and safer imaging technology, as compared to X-ray, and other widely used nuclear medicine technologies. Applications of optical imaging for diagnosis and monitoring of the onset, progression and treatment of various disease conditions, including cancer, are well established. (See, e.g., D. A. Benaron and D. K. Stevenson, Optical time-of-flight and absorbance imaging of biologic media, Science, 1993, 259, pp. 1463-1466; R. F. Potter (Series Editor), Medical optical tomography: functional imaging and monitoring, SPIE Optical Engineering Press, Bellingham, 1993; G. J. Tearney et al., In vivo endoscopic optical biopsy with optical coherence tomography, Science, 1997,276, pp. 2037-2039; B. J. Tromberg et al., Non-invasive measurements of breast tissue optical properties using frequency-domain photon migration, Phil. Trans. Royal Society London B, 1997, 352, pp. 661 -668; S. Fantini et al. , Assessment of the size, position, and optical properties of breast tumors in vivo by noninvasive optical methods, Appl. Opt, 1998, 37, pp. 1982- 1989; A. Pelegrin et al., Photoimmunodiagnosis with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies, J. Cell Pharmacol., 1992,3, pp. 141 -145).
[005] Optical agents for in vivo and in vitro biomedical imaging, anatomical visualization and monitoring organ function are described in International Patent Publication WO2008/108941 ; U.S. Patent Nos. 5,672,333; 5,698,397; 6, 167,297;6,228,344; 6,748,259; 6,838,074; 7,01 1 ,817;
7, 128,896, and 7,201 ,892. In this context, optical imaging agents are commonly used for enhancing signal-to-noise and resolution of optical images and extending these techniques to a wider range of biological settings and media. In addition, use of optical imaging agents having specific molecular recognition and/or tissue targeting functionality has also been demonstrated as effective for identifying, differentiating and characterizing discrete components of a biological sample at the organ, tissue, cellular, and molecular levels. Further, optical agents have been developed as tracers for real time monitoring of physiological function in a patient, including fluorescence-based monitoring of renal function. (See International Patent Publication
PCT/US2007/0149478). Given their recognized utility, considerable research continues to be directed toward developing improved optical agents for biomedical imaging and visualization.
[006] In addition to their important role in biomedical imaging and visualization, optical agents capable of absorption in the visible and NIR regions have also been extensively developed for clinical applications for phototherapy. The benefits of phototherapy using optical agents are widely acknowledged as this technique has the potential to provide efficacy comparable to radiotherapy, while entirely avoiding exposure of non-target organs and tissue to harmful ionizing radiation. Photodynamic therapy (PDT), in particular, has been used effectively for localized superficial or endoluminal malignant and premalignant conditions. The clinical efficacy of PDT has also been demonstrated for the treatment of various other diseases, injuries, and disorders, including cardiovascular disorders such as atherosclerosis and vascular restenosis, inflammatory diseases, ophthalmic diseases and dermatological diseases. Visudyne and Photofrin, for example, are two optical agents that have been developed for the treatment of macular degeneration of the eye and for ablation of several types of tumors, respectively. (See, e.g., Schmidt-Drfurth, U.; Bringruber, R.; Hasan, T. Phototherapy in ocular vascular disease. IEEE Journal of Selected Topics in Quantum Electronics 1996, 2, 988-996; Mlkvy, P.; Messmann, H.; Regula, J.; Conio, M.; Pauer, M.; Millson, C.E.; MacRobert, A.J.; Brown, S.G. Phototherapy for gastrointestinal tumors using three photosensitizers - ALA induced PPIX, Photofrin, and MTHPC. A pilot study. Neoplasma 1998, 45, 157-161 ; Grosjean, P.; Wagieres, G. ; Fontolliet, C.; Van Den Bergh, H.; Monnier, P. Clinical phototherapy for superficial cancer in the esophagus and the bronchi: 514 nm compared with 630 nm light irradiation after sensitization with Photofrin II. British Journal of Cancer 1998, 77, 1989-1955; Mitton, D.; Ackroyd, R. Phototherapy of Barrett's oesophagus and oesophageal carcinoma - how I do it. Photodiagnostics and Phototherapy 2006, 3, 96-98; and Li, L; Luo, R.; Liao, W. ; Zhang, M.; Luo, Y.; Miao, J. Clinical study of photofrin phototherapy for the treatment of relapse nasopharyngeal carcinoma. Photodiagnostics and Phototherapy 2006, 3, 266-271 ; See, Zheng Huang "A Review of Progress in Clinical Photodynamic Therapy", Technol Cancer Res Treat. 2005 June; 4(3): 283-293; "Photodiagnosis And Photodynamic Therapy", Brown S, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 2004;5:497-508; Triesscheijn M, Baas P, Schellens JHM. "Photodynamic Therapy in Oncology"; The Oncologist. 2006; 1 1 : 1034-1044; and Dougherty TJ, Gomer CJ, Henderson BW, Jori G, Kessel D, Korbelik M, Moan J, Peng Q. Photodynamic Therapy. J. Natl. Cancer Inst.
1998;90:899-905).
[007] Phototherapy is carried out by administration and delivery of a photosensitizer to a therapeutic target tissue (e.g., tumor, lesion, organ, etc.) followed by photoactivation of the photosensitizer by exposure to applied electromagnetic radiation. Phototherapeutic procedures require photosensitizers that are relatively chemically inert, and become activated only upon irradiation with light of an appropriate wavelength. Selective tissue injury can be induced with light when photosensitizers bind to the target tissues, either directly or through attachment to a bioactive carrier or targeting moiety. Photosensitizers essentially operate via two different pathways, classified as Types 1 and 2. A primary distinction between these classes of
photosensitizers is that the Type 1 process operates via direct energy or electron transfer from the photosensitizer to the cellular components thereby inducing cell death, whereas the Type 2 process involves first the conversion of singlet oxygen from the triplet oxygen found in the cellular environment followed by either direct reaction of singlet oxygen with the cellular components or further generating secondary reactive oxygen species (ROS) (e.g. peroxides, hydroxyl radical, etc.) which will induce cell death. Type 1 agents may also interact with oxygen, if present, to produce ROS, but this is not a necessary requirement, and does not have to be not mediated via singlet oxygen. [008] The Type 1 mechanism proceeds via a multistep process involving activation of the photosensitizer by absorption of electromagnetic radiation followed by direct interaction of the activated photosensitizer, or reactive intermediates derived from the photosensitizer, with the target tissue, for example via energy transfer, electron transfer or reaction with reactive species (e.g., radicals, ions, nitrene, carbene etc.) resulting in tissue damage. The Type 1 mechanism can be schematically represented by the following sequence of reactions:
hv PHOTOSENSITIZER* REACTIVE INTERMEDIATES
PHOTOSENSITIZER
(Excite , ions, etc.) CELL DEATH
Figure imgf000005_0001
wherein hv indicates applied electromagnetic radiation and (PHOTOSENSITIZER)* indicates excited state of the photosensitizer. The Type 2 mechanism proceeds via a multi-step process involving activation of the photosensitizer by absorption of electromagnetic radiation followed by energy transfer from the activated photosensitizer to oxygen molecules in the environment of the target tissue. This energy transfer process generates excited state oxygen ( 02) which subsequently interacts with the target tissue so as to cause tissue damage. The Type 2 mechanism can be schematically represented by the following sequence of reactions: ni lOTA r kl lTI 7rn hv PHOTOSENSITIZER* 3°2 ., _ . _ . . . PHOTOSENSITIZER (Excited State) Ό2 (Singlet Oxygen)
Figure imgf000005_0002
Collision with Cell Components REACTIVE OXYGEN SPECIES CELL DEATH (e g Hydroxy| radicals) wherein hv indicates applied electromagnetic radiation, (PHOTOSENSITIZER)* indicates photoactivated photosensitizer, 302 is ground state triplet oxygen, and 102 is excited state singlet oxygen.
[009] The biological basis of tissue injury brought about by tumor phototherapeutic agents has been the subject of intensive study. Various biochemical mechanisms for tissue damage have been postulated, which include the following: a) cancer cells up-regulate the expression of low density lipoprotein (LDL) receptors, and phototherapy (PDT) agents bind to LDL and albumin selectively; (b) porphyrin-like substances are selectively taken up by proliferative neovasculature; (c) tumors often contain increased number of lipid bodies and are thus able to bind to hydrophobic photosensitizers; (d) a combination of "leaky" tumor vasculature and reduced lymphatic drainage causes porphyrin accumulation referred to as "EPR" (enhanced permeability and retention) effect; (e) tumor cells may have increased capabilities for phagocytosis or pinocytosis of porphyrin aggregates; (f) tumor associated macrophages may be largely responsible for the concentration of photosensitizers in tumors; and (g) cancer cells may undergo apoptosis induced by
photosensitizers. Among these mechanisms, (f) and (g) are the most general and, of these two alternatives, there is a general consensus that (f) is the most likely mechanism by which the phototherapeutic effect of porphyrin-like compounds is induced.
[010] Much of the research in the past several decades has focused on developing
phototherapeutic agents based on the Type 2 (PDT) mechanism. Surprisingly, there has been considerably less attention devoted to Type 1 phototherapeutic agents despite the fact that there are numerous classes of compounds that could potentially be useful for phototherapy that function via this mechanism. Unlike Type 2, the Type 1 process does not require oxygen; and hence Type 1 photosensitizers are expected to be potentially more effective than Type 2 photosensitizers under hypoxic environments typically found in solid tumors. Second, the Type 1 mechanism involves two steps (photoexcitation and direct energy transfer), whereas the Type 2 mechanism involves three steps (photoexcitation, singlet oxygen generation, and energy transfer). Further, studies have recently shown that production of high levels of reactive oxygen species can induce an anti-inflammatory response, which may result in blood vessels to become more "leaky," thereby increasing the risk of metastasis (Chen, B.; Pogue, B.; Luna, J.M.; Hardman, R.L.; Hoopes, P.J.; Hasan, T. Tumor vascular permeabilization by vascular-targeting photosensitization: effects, mechanism, and therapeutic implications. Clinical Cancer Research 2006, 12(3, Pt.1 ), 917-923). Targeted Type 1 photosensitizers, by their very nature, are not expected to produce reactive oxygen species; rather, the reactive species produced by these photosensitizers will immediately react with the cellular component at the binding site and trigger cell death. Type 2
phototherapeutic agents, however, do have certain advantages over Type 1 agents. For example, Type 2 agents can potentially be catalytic, i.e., the Type 2 photosensitizer is regenerated once the energy transfer to the oxygen has taken place. In contrast, Type 1 process would generally be expected to require stoichiometric amounts of the photosensitizer in some clinical settings. Table B1 provides a summary of the attributes of Type 1 and Type 2 phototherapeutic agents. Given these attributes, it is clear that development of safe and effective Type 1 phototherapeutic agents would be useful to complement the existing therapeutic approaches provided by Type 2 agents, and to enhance the therapeutic portfolio available for clinicians.
Table B1 Comparison between Type 1 and Type 2 processes for phototherapy.
TYPE 1 PROCESS TYPE 2 PROCESS
Two-step process. Three-step process.
Not well explored. Very well studied.
Light of any wavelength can be used. Requires red light for optimal performance. Does not require oxygen. Requires oxygen.
Large classes of compounds. Limited classes of compounds.
Stoichiometric. Potentially catalytic.
Intramolecular energy transfer to generate Intermolecular energy transfer to generate reactive species. reactive oxygen species. No products in the market. Two products are in use.
[011] Specific optical, chemical and pharmacokinetic properties of optical agents are necessary for their effective use in Type 1 and Type 2 phototherapeutic applications. For example, optical agents for these applications preferably have strong absorption in the visible or NIR regions, and also exhibit low systemic toxicity, low mutagenicity, and rapid clearance from the blood stream. These optical agents must also be compatible with effective administration and delivery to the target tissue, for example by having reasonable solubilities and a low tendency for aggregation in solution. Upon excitation by absorption of visible and NIR electromagnetic radiation, optical agents for Type 1 and 2 phototherapy preferably provide large yields of singlet oxygen (Type 2) or other reactive species, such as free radicals or ions, capable of causing local tissue damage. Both Type 1 and Type 2 photosensitizers typically undergo photoactivation followed by intersystem crossing to their lowest triplet excited state, and therefore, a relatively long triplet lifetime is usually beneficial for providing effective tissue damage. Other useful properties of optical agents for these applications include chemical inertness and stability, insensitivity of optical properties to changes in pH, and compatibility with conjugation to ligands providing targeted delivery via molecular recognition functionality. Multifunctional optical agents have also been developed for phototherapy that are capable of providing both imaging and visual functionality upon excitation at a first range of wavelengths and phototherapeutic functionality upon excitation at a second range of wavelength. (See, US Patent No. 7,235,685 and International Patent Publication WO
2007/106436).
[012] Optical agents for some phototherapeutic applications preferably exhibit a high degree of selectivity for the target tissue. Selectivity provided by optical agents facilitates effective delivery to a target tissue of interest and provides a means of differentiating different tissue classes during therapy. Selective tissue injury can be induced with light when photosensitizers bind to the target tissues either directly, as in the case of Photofrin, or through attachment to a bioactive carrier, or through in situ biochemical synthesis of the photosensitizer in localized area, as in the case of 2- aminolevulinic acid, which is an intermediate in the biosynthesis of porphyrin. Previous studies have shown that certain dyes selectively localize in tumors and serve as a powerful probe for the detection and treatment of small cancers. (D. A. Belinier et al., Murine pharmacokinetics and antitumor efficacy of the photodynamic sensitizer 2-[l-hexyloxyethyl]-2-devinyl pyropheophorbide- a, J. Photochem. Photobiol., 1993, 20, pp. 55-61 ; G. A. Wagnieres et al., In vivo fluorescence spectroscopy and imaging for oncological applications, Photochem. Photobiol., 1998, 68, pp. 603- 632; J. S. Reynolds et al., Imaging of spontaneous canine mammary tumors using fluorescent contrast agents, Photochem. Photobiol., 1999, 70, pp. 87-94). It is recognized in some situations, however, that many dyes do not localize preferentially in malignant tissues. A number of strategies have been developed for imparting selectivity and/or targeting functionality by incorporation of a molecular recognition component in the optical agent. For example, targeting of fluorescent dyes to tumors has been demonstrated using dye conjugates with antibodies and peptides for diagnostic imaging of tumors. (See, Achilefu et al., Novel receptor-targeted fluorescent contrast agents for in vivo imaging of tumors, Investigative Radiology, 2000, 35, pp. 479-485; Ballou et al., Tumor labeling in vivo using cyanine conjugated monoclonal antibodies, Cancer Immunology and Immunotherapy, 1995, 41 , pp. 257-263; and Licha et al., New contrast agent for optical imaging: acid cleavable conjugates of cyanine dyes with biomolecules, in Biomedical Imaging: Reporters, Dyes and Instrumentation, Proceedings of SPIE, 1999, 3600, pp. 29-35). Therefore, receptor-target mediated phototherapy agents provide a promising pathway for achieving site selective activation at various target tissues.
[013] As will be generally recognized from the foregoing, a need currently exists for optical agents for biomedical applications. Specifically, optical agents for imaging, visualization and phototherapy are needed having enhanced specificity for important target tissue classes, such as tumors and other lesions. In addition, optical agents are needed having enhanced optical, physical, chemical and pharmacokinetic properties for administration, delivery and excitation with electromagnetic radiation.
SUMMARY
[014] The invention relates generally to optical agents for biomedical applications, including phototherapy. Provided are sulfenamide compounds having an acyclic S-N bond with at least one aromatic and/or heterocyclic aromatic group, including fused ring carbocyclic and/or heterocyclic aromatic groups, providing phototherapeutic agents, including Type 1 phototherapeutic agents. Optical agents of the invention enable a versatile phototherapy platform for treatment of a range of pathological conditions, including the treatment of cancers, stenosis and inflammation. The invention further provides preparations and formulations comprising the sulfenamide optical agents and related methods of making and using sulfenamide optical agents in an in vivo or ex vivo biomedical procedure.
[015] In some embodiments, for example, the invention provides sulfenamide compounds for phototherapeutic methods having one or more photolabile acyclic S-N bonds capable of undergoing photoactivated cleavage to produce reactive species, such as radicals, ions, etc., that achieve a desired therapeutic effect, such as selective and/or localized tissue damage and/or cell death. Optical agents of an aspect of the invention include compositions having one or more carbocyclic and/or heterocyclic aromatic groups, including fused ring aromatic groups, providing a chromophore directly or indirectly coupled to an acyclic S-N bond capable of undergoing photodissociation and/or photofragmentation upon exposure to electromagnetic radiation having wavelengths in the visible and/or near infrared regions of the electromagnetic spectrum. Optical agents further include conjugates, such as sulfenamide bioconjugate compositions including at least one targeting ligand such as a polypeptide, protein, oligonucleotide, carbohydrate, antibody, or other biomolecule, or fragments or fusions thereof, capable of providing molecular recognition and/or tissue specific targeting functionality. Optical agents further include multifunctional optical agents providing tandem imaging and phototherapy functionality, wherein the agent comprises a photosensitizer component having an acyclic S-N bond directly or indirectly linked to an optical dye component, such as a C5-C30 aryl or C5-C30 heteroaryl fluorophore, and optionally further comprising one or more targeting ligands.
[016] In an aspect, the invention provides sulfenamide compounds having an acyclic S-N bond useful as optical agents for phototherapeutic methods, including Type 1 phototherapy. In an embodiment, for example, the invention provides a compound for use in a phototherapy
a (FX1):
Figure imgf000009_0001
(FX1 ); or a pharmaceutically acceptable salt or ester thereof, wherein: each of L1, L2, and L3, if present, is independently CrC10 alkylene, C3-Ci0 cycloalkylene, C2-C10 alkenylene, C3-C10 cycloalkenylene, C2-Ci0 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, ~(CH2CH20)b- or -(CHOH)a-;
each of W1 , W2, and W3 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -S02- -S03- -OS02-, -NR 1-, -CO-, -COO-, -OCO-, -OCOO-, -CONR 2-, - NR 3CO- -OCONR 4-, -NR 5COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n- -S(CH2)n- - NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n- -OCO(CH2)n-, -OCOO(CH2)n-, -CONR 1(CH2)n-, - CO R22(CH2)n- -NR23CO(CH2)n-, -OCONR24(CH2)n-, -NR25COO(CH2)n-, -NR26CONR27(CH2)n- , -NR28CSNR29(CH2)n- -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-;
each of R1 , R2, and R3 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R4°, -SOR41 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R 4R45, -OR46, -SR47, -NR 8R49, - NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R6 )C02H, -CH(R63)NH2, FL or Bm;
or wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form one to five substituted or unsubstituted carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings;
wherein at least one of R1 , R2, and R3 is a C5-C30 aryl or C5-C30 heteroaryl, or wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form a C5-C30 aryl or C5-C30 heteroaryl;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of e, f, and g is independently 0 or 1 ; each of R - R33 is independently hydrogen, C C2o alkyl, C3-C2o cycloalkyl, C5-C20 heteroaryl, or C5-C2o aryl;
each of R40-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or d-C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid;
each FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a
glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steriod, a hormone, an aptamer, a receptor, a metal chelating agent, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units. In an embodiment, the composition of R , R2, R3, W1 , W2, W3, L , L2, and/or L3, is selected such that the compound undergoes cleavage of the central S-N bond upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, and optionally wavelengths selected over the range of 400 nanometers to 900 nanometers.
[017] In an embodiment, for example, the invention provides a compound having formula (FX1), wherein at least one of R1, R2 and R3 is a C5-C30 aryl or C5-C30 heteroaryl group
corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline. In an embodiment, for example, the invention provides a compound having formula (FX1), wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form one to five, and optionally one to three, carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings substituted to include one or more electron donating group, electron with drawing group, targeting group (e.g., Bm) or fluorophore group (e.g., FL).
[018] As used throughout the present description, reference to embodiments wherein e, f, and/or g is equal to 0 refers to compounds where L1, L , and/or L3, respectively, is not present; and reference to embodiments wherein e, f, and/or g is equal to 1 refers to compounds where L1, L2, and/or L3, respectively, is present. For example, W1 is directly linked to the central S-N group when e is equal to 0; and/or W2 is directly linked to the central S-N group when f is equal to 0; and/or W3 is directly linked to the central S-N group when g is equal to 0. Embodiments wherein W1 is a single bond and e is equal to 0 refer to compositions having R1 directly linked to the central S-N group via a single bond. Embodiments wherein W2 is a single bond and f is equal to 0 refer to compositions having R2 directly linked to the central S-N group via a single bond. Embodiments wherein W3 is a single bond and g is equal to 0 refer to compositions having R3 directly linked to the central S-N group via a single bond. This convention is used throughout the present description, for example, in connection with other L and W groups, such as L 20-L127 and W120- W127.
[019] As used throughout the present description, the expression "a group corresponding to" an indicated species expressly includes a radical (including a monovalent, divalent and trivalent radical), for example an aromatic radical or heterocyclic aromatic radical, of the species or group of species provided in a covalently bonded configuration, optionally with one or more substituents, including but not limited to one or more electron donating groups, electron withdrawing groups, fluorophore groups, photosensitizer groups and/or targeting ligands.
[020] In an embodiment, R1 is C5-C30 aryl or C5-C30 heteroaryl, and each of R2 and R3 is independently hydrogen, C†-C2o alkyl, C3-C2o cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, FL, or Bm. In an embodiment, R2 is C5-C30 aryl or C5-C30 heteroaryl, and each of R1 and R3 is independently hydrogen, C C2o alkyl, C3-C2o cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, FL, or Bm. In an embodiment, R3 is C5-C30 aryl or C5-C30 heteroaryl, and each of R1 and R2 is independently hydrogen, C C2o alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, FL, or Bm. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having formula (FX1 ), wherein R is C^CW alkyl, C3-C10 cycloalkyl, C5-C30 heteroaryl, or a C5-C30 aryl, wherein optionally the C5-C30 aryl or C5-C30 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R2 and R3 is independently a hydrogen, CrC2o alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, a FL or Bm, wherein optionally the C5-C30 aryl or C5-C30 heteroaryl, includes an electron donating group, electron withdrawing group, or a targeting ligand (Bm) as a substituent. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having formula (FX1), wherein R2 is C C10 alkyl, C3-Ci0 cycloalkyl, C5-C30 heteroaryl, or a C5-C30 aryl, wherein optionally the C5-C30 aryl or C5-C30 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R1 and R3 is independently a hydrogen, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 heteroaryl, C5-C30 aryl, a FL or Bm, wherein optionally the C5-C30 aryl or C5-C30 heteroaryl includes an electron donating group, electron withdrawing group, or a targeting ligand (Bm) as a substituent. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having formula (FX1 ), wherein R3 is C C 0 alkyl, C3-C10 cycloalkyl, C5-C30 heteroaryl, or a C5-C30 aryl, wherein optionally the C5-C30 aryl or C5-C30 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R1 and R2 is independently a hydrogen, Ci-C2o alkyl, C3-C20 cycloalkyl, C5-C30 heteroaryl, C5-C30 aryl, a FL or Bm, wherein optionally the C5-C30 aryl or C5-C30 heteroaryl includes an electron donating group, electron withdrawing group, or a targeting ligand (Bm) as a substituent
[021] The invention further provides compounds useful as optical agents for phototherapeutic methods having formula (FX1 ), wherein one or more of R1, R2 and R3 are independently one or more carbocylic and/or heterocyclic aromatic groups, including fused ring aromatic groups, providing a chromophore capable of excitation upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, and optionally in some embodiments wavelengths selected over the range of 400 nanometers to 900 nanometers. In an embodiment, for example, the invention provides compounds for use in a phototherapeutic methods having formula (FX2), (FX3), (FX4), (FX5), (FX6), (FX7) or (FX8):
Figure imgf000012_0001
thereof; wherein L , L2, L3, W1 , W2, W3, R1, R2, R3, e, f and g are as described in connection with formula (FX1); and wherein each of Ar1 , Ar2 and Ar3 is independently a C5-C30 aryl or C5-C30 heteroaryl group comprising one or more aromatic rings or heterocyclic aromatic rings, optionally having one or more electron donating groups and/or electron withdrawing groups provided as substituents. In an embodiment, for example, the invention provide compounds being of formula (FX2) - (FX15), wherein each of Ar1 , Ar2 and Ar3 is independently a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene,
benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline.
[022] In an embodiment, of this aspect, the invention provides compounds having formula (FX1 ) wherein one or more aromatic group (e.g., C5-C30 aryl or C5-C30 heteroaryl) is directly linked to the central S-N group. As used in the present context, the expression directly linked refers to a configuration wherein an aromatic group is covalently bonded to either the S atom or the N atom of the central S-N group, for example, via a single bond. In an embodiment, for example, the invention provides compounds for use in a phototherapeutic methods having formula (FX9),
Figure imgf000013_0001
Ar Ar3
/
\ Ar (FX15); or a pharmaceutically acceptable salt or ester thereof; wherein L1 , L2, L3, W1, W2, W3, R1 , R2, R3, e, f, g, Ar1, Ar2 and Ar3 are as described in connection with formula (FX1 ).
[023] In an embodiment, for example, the invention provides compounds for use in a phototherapeutic methods having any one of formula (FX2) - (FX15): wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar3 is independently (or optionally each of Ar1 and Ar2, Ar1 and Ar3, Ar2 and Ar3, or Ar1, Ar2 and Ar3 is):
Figure imgf000014_0001
; wherein each of R - R is independently a hydrogen, -OCF3,
Figure imgf000014_0002
alkyl, C3-C20 cycloalkyl,C5-C2o aryl, C5-C2o heteroaryl, C-i-C2o acyl, C2- C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R4°, -SOR41 , -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, - OR46, -SR47, -NR48R49, -NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, - S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; wherein a, b, FL, Bm and R40 - R55 and R58 - R63 are as described in the context of formula (FX1 ). In an embodiment, for example, the invention provides compounds for use in a phototherapeutic methods having any one of formula (FX2) - (FX1 5): wherein at least one of R1 - R3, R65 - R67 and 1 - Ar3 is independently (or optionally each of Ar1 and Ar2, Ar1 and Ar3, Ar2 and Ar3, or Ar , Ar2
Figure imgf000014_0003
- R is independently a hydrogen, -OCF3, C
C20 alkyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C20 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, - C02R40, -SOR41, -OSR42 , -SO2OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, - NR48R49, -NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; wherein a, b, FL, Bm and R40 - R55 and R58 - R63 are as described in the context of formula (FX1 ). In an embodiment, for example, the invention provides compounds for use in a phototherapeutic methods having any one of formula (FX2) - (FX1 5): wherein at least one of R - R3, R65 - R67 and Ar1 - Ar3 is independently (or optionally each of Ar1 and Ar2, Ar1 and Ar3, Ar2 and Ar3, or Ar1, Ar2 and Ar3 is):
Figure imgf000014_0004
Figure imgf000015_0001
Figure imgf000016_0001
; wherein each of R - R is independently a hydrogen, -OCF3, C C20 alkyl, C3-C2o cycloalkyl, C5-C2o aryl, C5-C20 heteroaryl, C C2o acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -
,42 ,43 44r->45 ,46
SOR41 , -OSR^ , -S02ORi,J, -CH2(CH20CH2)bCH20H, -P03R^R °, -OR -SR47, -NR 8R49, -
NR50COR51 , -CN, -CONR"Rw, -COR , -N02, -S02R , -S02NRS8Rsy, -CH2(CHOH)aR'
,62\ ,63 40
(CH2CH20)bR , -CH(RB )C02H, -CH(Re3)NH2, FL or Bm; wherein a, b, FL, Bm and R Rss and
R B - R are as described in the context of formula (FX1). In an embodiment, the invention
70
provides compounds wherein at least one of R R , or at least one of R R , or at least one of R - R is independently an electron donating group; and wherein at least one of R - R , or at least one of R75 - R77, or at least one of R78 - R85 is independently an electron withdrawing group. In an embodiment, the invention provides compounds wherein each of R70 - R85 is independently a hydrogen, -OR46, -SR47, -NR48R49, -NR50COR51 , -CN, halo, -CO2R40,-COR54, - N02, -S02R55, or -S02NR58R59. In an embodiment, the invention provides compounds wherein at
70
least one of R R , or at least one of R,b - R , or at least one of R B - R , is independently a
OR4b, -SR4 , -NR R , -NFTOOR , -CN, halo, -C02R4U,-CORS4, -N02, -S02Rt>b, or - S02NR58R59. In an embodiment, the invention provides compounds wherein at least one of R R , or at least one of R/s - R , or at least one of R'a - R8S, is independently a -OR4b, -SR , - NR48R49, or -NR50COR51; and at least one of R70 - R74, or at least one of R75 - R77, or at least one of R78 - R85, is independently -CN, halo, -CO2R40,-COR54, -N02, -S02R55, or -S02NR58R59. In an embodiment, the invention provides compounds wherein at least one of R70 - R74, or at least one of R75 - R77, or at least one of R78 - R85, is independently Bm. In an embodiment, the invention provides compounds, wherein each of R70 - R85 is hydrogen or Ci - C6 alkyl, and optionally wherein each of R70 - R85 is hydrogen or C-i - C3 alkyl, and optionally each of wherein R70 - R85 is hydrogen.
[024] The invention provides compounds for use in phototherapy having formula (FX1 ), wherein R2, W2, R3, and W2, and L2 and L3, if present, together with the atoms to which they are attached combine to form one or more alicyclic ring structures. In an embodiment, for example, the invention provides a compound for use in a phototherapy procedure, the compound being of the
Figure imgf000017_0001
(FX16); or a pharmaceutically acceptable salt or ester thereof, wherein:
X is independently -CR65R66-, -NR67-, -0-, -S-, -SO-, or -S02-;
each of Y and Y2 is independently -CR65R66-;
each of R65 , R66 and R67 is independently a hydrogen, -OCF3, CrC2o alkyl, C3-C2o cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C-|-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42, -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of i and h is independently an integer selected from the range of 0 to 6, wherein at least one of i and h is greater than or equal to 1 ; wherein R1, W , L1, e, b, FL, Bm, R40 - R55 and R58 - R63 are as described in the context of formula (FX1). In embodiments wherein i is 0, Y1 is not present and X is directly bonded to the central nitrogen of formula (FX16). In embodiments wherein h is 0, Y2 is not present and X is directly bonded to the central nitrogen of formula (FX16). In an aspect, at least one of R1, R65, R66 and R67 is independently C5-C30 aryl or C5-C30 heteroaryl. In an embodiment of this aspect, for example, the invention provides a compound for use in a phototherapy procedure, the compound being of the formula (FX17):
Figure imgf000018_0001
(FX17); or a pharmaceutically acceptable salt or ester thereof, wherein L1, W1, R1, i, h, e and X are as described in connection with formula (FX16). In an embodiment, the invention provides a compound for use in a phototherapeutic procedure having formula (FX16) or (FX17), wherein R is C5-C30 aryl or C5-C30 heteroaryl, optionally C5-C2o aryl or C5-C30 heteroaryl, or C5-C10 aryl or C5-C30 heteroaryl. In an embodiment, the invention provides a compound for use in a phototherapeutic procedure having any one of formula (FX16) or (FX17), wherein each of X, Y1 and Y2 is independently -CR65R66-, wherein optionally each of R65 and R66 is a hydrogen. In an embodiment, the invention provides a compound for use in a phototherapeutic procedure having any one of formula (FX16) and (FX17), wherein X is -0-, and wherein each of Y and Y2 is independently -CR65R66-, wherein optionally each of R65 and R66 is hydrogen. In an embodiment, the invention provides a compound for use in a phototherapeutic procedure having any one of formula (FX16) and (FX17), wherein X is -S-, and each of Y and Y2 is independently -CR65R66-, wherein optionally each of R65 and R66 is hydrogen.
[025] In an embodiment, the invention provides a compound having formula (FX16) or (FX17) wherein the sum of i and h is 2 and, therefore, a carbocyclic or heterocyclic four-membered alicyclic ring incorporating the N from the central S-N group is formed; or wherein the sum of i and h is 3 and a carbocyclic or heterocyclic five-membered alicyclic ring incorporating the N from the central S-N group is formed; or wherein the sum of i and h is 4 and a carbocyclic or heterocyclic six-membered alicyclic ring incorporating the N from the central S-N group is formed; or wherein the sum of i and h is 5 and a carbocyclic or heterocyclic seven-membered alicyclic ring
incorporating the N from the central S-N group is formed. In an embodiment, the invention provides compounds having formula (FX16) and (FX17), wherein R is a C5-C30 aryl or C5-C30 heteroaryl, and R65, R66, and R67 are independently hydrogen, C C2o alkyl, C3-C2o cycloalkyl, FL, or Bm. In an embodiment, the invention provides a compound having formula (FX16) or (FX17), wherein at least one of R65, R66, and R67 is Bm. In an embodiment, the invention provides compounds having formula (FX16) and (FX17), wherein R1 is a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene,
benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza- azulene, or anthracycline, optionally having one or more electron withdrawing groups, electron donating groups and/or targeting ligands provided as substituents.
[026] In an embodiment, the invention provides compounds for phototherapy being of the formula (FX20):
Figure imgf000019_0001
(FX20); or a pharmaceutically acceptable salt or ester thereof, wherein:
each of rings A, B, and C are independently
Figure imgf000019_0002
or ; and each of R70
- R77 is independently a hydrogen, -OCF3, C C2o aikyl, C3-C20 cycloalkyl, C5-C2o aryl, C5-C20 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, CTC6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R4°, -SOR41 , -OSR42 , -S02OR43, - CH2(CH2OCH2)BCH2OH, -P03R44R45, -OR46, -SR47, -N R48R49, -NR50COR51 , -CN , -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)AR60, -(CH2CH20)BR61 , -CH(R62)C02H, - CH(R63)NH2, FL or Bm. For example, the invention provides compounds for phototherapy being of formula (FX21 ), (FX22), (FX23), (FX24), (FX25) or (FX26):
Figure imgf000019_0003
Figure imgf000020_0001
or a pharmaceutically acceptable salt or ester thereof. In an embodiment, the invention provides a compound being of formula (FX20) - (FX26), wherein at least one of R70 - R74 is and electron donating group and at least one of R70 - R74 is and electron withdrawing group. In an embodiment, the invention provides a compound being of formula (FX20) - (FX26), wherein at least one of R75 - R77 is and electron donating group and at least one of R75 - R77 is and electron withdrawing group. In an embodiment, the invention provides a compound being of formula (FX20) - (FX26), wherein each of R70 - R77 is independently a hydrogen, -N02, -OCF3, C C2o alky!, C3-C20 cycloalkyl, C5- C2o aryl, or C5-C20 heteroaryl, and optionally wherein each of R70 - R77 is independently a hydrogen, -N02, -OCF3, or Ci-C6 alkyl, and optionally wherein each of R70 - R77 is independently a hydrogen, or C-i-C6 alkyl. In an embodiment, the invention provides a compound being of formula (FX20) - (FX26), wherein at least one of R70 - R77 is -N02.
[027] In an embodiment, the invention provides compounds being of formula (FX20) - (FX26), wherein each of R70 - R77 is independently a hydrogen,-OR46, -SR47, -NR48R49, -NR50COR51, - CN, halo, -CO2R40,-COR54, -N02, -S02R55, or -S02NR58R59. In an embodiment, the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R70 - R77 is independently a -OR46, -SR47, -NR48R49, -NR50COR51 , -CN, halo, -CO2R40,-COR54, -N02, - S02R55, or -S02NR58R59. In an embodiment, the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R70 - R77 is independently a -OR46, -SR47, -NR48R49, or -NR50COR51; and at least one of R70 - R77 is independently -CN, halo, -CO2R 0,-COR54, -N02, - S02R55, or -S02NR58R59. In an embodiment, the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R70 - R77 is independently is independently Bm. In an embodiment, the invention provides compounds being of formula (FX20) - (FX26), wherein at least one of R70 - R77 is independently hydrogen or Ci - C6 alkyl, and optionally wherein each of R70 - R77 is hydrogen or C - C3 alkyl, and optionally each of wherein R70 - R77 is hydrogen.
[028] The invention provides compounds for use in phototherapy having formula (FX1), wherein R2, W2, R3, and W2, and L2 and L3, if present, together with the atoms to which they are attached combine to form a fused ring structure comprising one to five, and optionally one to three, alicyclic and/or aromatic rings, optionally substituted with one or more electron donating groups, electron withdrawing groups, chromophore groups and/or targeting ligands. In an embodiment of this aspect, for example, the invention provides class of photosensitizers having a S-N bond with a fused ring configuration, wherein the nitrogen atom of the S-N bond is a part of the fuse ring structure. Examples of compounds of this aspect include dibenzazepine derivatives. In an embodiment, for example, the invention provides a compound for phototherapy being of the
Figure imgf000021_0001
pharmaceutically acceptable salt or ester thereof, wherein:
Z is a single bond, -CR128=CR129-, -CR130R131CR132R133-, -CR134R1350-, -CR136R137S-
_CR138R139N R140_ Q r _N = C R141_;
each of R 20 to R 4 is independently a hydrogen, -OCF3, C C2o alkyl, C3-C2o cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , -OSR42 , - S02OR43, -CH2(CH2OCH2)bCH2OH , -P03R R45, -OR46, -SR47, -NR48R49, -NR50COR51 , -CN , - CON R52R53, -COR54, -NO2, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61 , - CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
each of L120, L121 , L122, L123, L124, L 25, L126 and L127, if present, is independently C C10 alkylene, C3-C 0 cycloalkylene, C2-Ci0 alkenylene, C3-C 0 cycloalkenylene, C2-Ci0 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b- or -(CHOH)a-;
each of W120, W121 , W122, W123, W 24, W125, W 26 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -SO2-, -SO3-, -OSO2-, -NR -, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR14-, -NR15COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n-, - OCOO(CH2)n-, -CONR2 (CH2)n- -CONR22(CH2)n-, -NR23CO(CH2)n-, -OCONR24(CH2)n-, - NR25COO(CH2)n- -NR26CONR27(CH2)n-, -NR28CSNR 9(CH2)n- -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-; and wherein L1, W1, R1 , and e are as described in connection with formula (FX1).
[029] In an embodiment, for example, the invention provides a compound for phototherapy of formula (FX27) wherein at least one of R1 and R120 to R 41 is hydrogen, optionally wherein all of R and R120 to R141 are hydrogen. In another embodiment, the invention provides a compound for phototherapy of formula (FX27) wherein at least one of R1 and R120 to R141 is independently FL or Bm. In another embodiment, the invention provides a compound for phototherapy of formula
(FX27) wherein at least one of R1 and R120 to R 41 is hydrogen or C-, - C6 alkyl, and optionally hydrogen or - C3 alkyl, and optionally hydrogen.
[030] As used throughout the present description, reference to embodiments wherein o, p, q, r, s, t, u and/or v is equal to 0 refers to compounds where L120, L121 , L122, L123, L 24, L125, L 26, and/or L127, respectively, is not present; and reference to embodiments wherein o, p, q, r, s, t, u and/or v is equal to 1 refers to compounds where L120, L 21, L122, L123, L124, L125, L126, and/or L127, respectively, is present. In an embodiment, for example, the invention provides a compound for phototherapy of formula (FX27) wherein at least one of e, o, p, q, r, s, t, u, and v is independently 0, optionally wherein all of e, o, p, q, r, s, t, u, and v are 0. In a related embodiment, the invention provides a compound for phototherapy of formula (FX27) wherein at least one of e, o, p, q, r, s, t, u, and v is independently 1 , optionally wherein all of e, o, p, q, r, s, t, u, and v are 1.
[031] In an embodiment, for example, the invention provides a compound for phototherapy of
, 140 formula (FX27) wherein Z is -CR12a=CRl a- -CR134R130O-, -CR IJ0R IJ/S- -CR138R 39NR |i,u- or -N=CR141-. In an embodiment, for example, the invention provides a compound for phototherapy of formula (FX27) wherein each of R 30 to R133 is independently a hydrogen, -OCF3, C2-C20 alkyl, C3-C20 cycloalkyl, C5-C2o aryi, C5-C20 heteroaryl, C -C2o acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C-i-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R 4R45, -OR46, -SR47, -NR48R49, -
NR50COR51 , -CN, -CONR52R53, -COR54, -N02,
Figure imgf000022_0001
,62 63
(CH2CH20)bR61, -CH(RD')C02H, -CH(Roa)NH2l FL or Bm
[032] In an embodiment, for example, the invention provides a compound for phototherapy being of the formula (FX28):
Figure imgf000022_0002
pharmaceutically acceptable salt or ester thereof. [033] In an embodiment, for example, the invention provides a compound for phototherapy being of the formula : (FX29), (FX30), (FX31), (FX32), (FX33), or (FX34):
Figure imgf000023_0001
Figure imgf000024_0001
(FX34) or a pharmaceutically acceptable salt or ester thereof.
[034] In an embodiment, for example, the invention provides a compound for phototherapy being of the formula: (FX35), (FX36), (FX37), or (FX38):
Figure imgf000025_0001
thereof.
[035] In an embodiment, for example, the invention provides a compound for phototherapy being of the formula (FX39):
Figure imgf000026_0001
pharmaceutically acceptable salt or ester thereof, wherein: each of R144 to R148 is independently a hydrogen , -OCF3, CrC20 aikyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C20 heteroaryl, C C2o acyl , C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl , halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , -OSR42 , - S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R4 R45, -OR46, -SR47, -N R48R49, -NR50COR51 , -CN , - CONR52R53, -COR54, -NO2, -S02R55, -P03R56R57, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H , -CH(R63)NH2, FL or Bm.
[036] In an embodiment, for example, the invention provides a compound for phototherapy being of the formula: (FX40), (FX41 ), (FX42), (FX43), (FX44), or (FX45):
Figure imgf000027_0001
Figure imgf000028_0001
(FX45), or a pharmaceutically acceptable salt or ester thereof.
[037] In an embodiment, for example, the invention provides a compound for phototherapy being of the formula (FX46):
Figure imgf000029_0001
(FX46), or a pharmaceutically acceptable salt or ester thereof.
[038] In an embodiment, the invention provides a compound being of formula (FX55):
Figure imgf000029_0002
pharmaceutically acceptable salt or ester thereof, wherein:
, 141
Z is -CFTe=CR -, -CR ' J4R ' JO- -CR ' MR ' J'S- -CR R NR -, or -N=CR each of L120, L 21 , L 22, L123, L124, L125, L126 and L 27, if present, is independently d-C10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-C10 cycloalkenylene, C2-C 0 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b-, or -(CHOH)a-; each of W120, W121 , W122, W123, W124, W125, W126 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -S02- -S03-, -OS02- -NR11-, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR14-, -NR15COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n- -CO(CH2)n- -COO(CH2)n- -OCO(CH2)n- - OCOO(CH2)n- -CONR2 (CH2)n- -CONR2 (CH2)n- -NR23CO(CH2)n-, -OCONR24(CH2)n-, - NR25COO(CH2)n- -NR26CONR27(CH2)n- -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR3 (CH2)nNR32CO-, or -CO(CH2)nNR33CO-; each of R 20 to R129, R134 to R141 and R144 to R 48 is independently a hydrogen, -OCF3, d- C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, Ci-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, - C02R40, -SOR4 , -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, - NR48R49, -NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; each of o, p, q, r, s, t, u, and v is independently 0 or 1 ; each of a and b is independently an integer selected from the range of 1 to 100; each n is independently an integer selected from the range of 1 to 10; each of R 1 - R33 is independently hydrogen, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C20 aryl; each of R40-R55 and R58- R6 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
[039] In an embodiment, the invention provides a compound being of formula (FX56):
Figure imgf000031_0001
(FX56); or a pharmaceutically acceptable salt or ester thereof, wherein: each of L 20, L121, L122, L 23, L124, L 25, L126 and L127, if present, is independently C C10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-Ci0 cycloalkenylene, C2-Ci0 alkynylene, ethenylene, ethynyiene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b- or -(CHOH)a-; each of W 20, W 2 , W122, W123, W124, W125, W126 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -SO2-, -SO3-, -OSO2-, -NR11-, -CO-, -COO- -OCO- , -OCOO-, -CONR 2-, -NR 3CO- -OCONR14-, -NR1 COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n-, -CO(CH2)n-, -COO(CH2)n- -OCO(CH2)n-, - OCOO(CH2)n-, -CONR2 (CH2)n-, -CONR22(CH2)n- -NR23CO(CH2)n-, -OCONR24(CH2)n-, - NR25COO(CH2)n-, -NR26CONR27(CH2)n- -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-; each of R120 to R127, R 30 to R133 and R 44 to R148 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, haiomethyl, dihalomethyl, trihalomethyl, - C02R40, -SOR41 , -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, - NR 8R49, -NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61, -CH(R6 )C02H, -CH(R63)NH2, FL or Bm; wherein when R 46 is N02, at least one of R 20 to R127, R130 to R133, R144, R145, R147 and R148 is independently -0CF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, CrC20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, haiomethyl, dihalomethyl, trihalomethyl, - C02R40, -SOR41, -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R 4R45, -OR46, -SR47, - NR48R49, -NR50COR51 , -CN, -CONR5 R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm; each of o, p, q, r, s, t, u, and v is independently 0 or 1 ; each of a and b is independently an integer selected from the range of 1 to 100; each n is independently an integer selected from the range of 1 to 10; each of R1 1 - R33 is independently hydrogen, Ci-C2o alkyl, C3-C20 cycloalkyl, C5-C2o heteroaryl, or C5-C20 aryl; each of R40-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C 0 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azuiene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
[040] In an embodiment, the invention provides a compound being of formula (FX57):
Figure imgf000032_0001
pharmaceutically acceptable salt or ester thereof, wherein: each of L 20, L 2 , L122, L123, L 24, L125, L 26 and L127, if present, is independently C C10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-Ci0 cycloalkenylene, C2-C10 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b-, or -(CHOH)a-; each of W120, W 21, W 22, W123, W 24, W125, W 26 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -S02- -SO3-, -OS02-, -NR1 -, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR14-, -NR15COO- -NR 6CONR17-, -NR18CSNR19- -0(CH2)n- -S(CH2)n-, -NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n-, - OCOO(CH2)n-, -CONR21(CH2)n-, -CONR22(CH2)n-, -NR23CO(CH2)n- -OCONR24(CH2)n-, - NR25COO(CH2)n-, -NR26CONR27(CH2)n-, -NR28CSNR29(CH2)n- -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-; each of R120to R 27 and R 44 to R 48 is independently a hydrogen, -OCF3, C C20 alkyl, C3- C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, Ci-C20 acyl, C2-C20 alkenyl, C2-C2o alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm; wherein at least one of R120 to R 27 and R 44 to R148 is independently -CN, -C02R40, -COR54, -S02R55, -S02NR58R59, -OR46, -SR47, - NR48R49, or -NR50COR51; each of 0, p, q, r, s, t, u, and v is independently 0 or 1 ; each of a and b is independently an integer selected from the range of 1 to 100; each n is independently an integer selected from the range of 1 to 10; each of R1 - R33 is independently hydrogen, Ci-C20 alkyl, C3-C20 cycloalkyl, C5-C2o heteroaryl, or C5-C20 aryl; each of R 0-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units. of formula (FX58):
Figure imgf000034_0001
(FX58); or a
pharmaceutically acceptable salt or ester thereof, wherein: each of L120, L121 , L122, L 23, L124, L125, L126 and L 27, if present, is independently C C10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-C10 cycloalkenylene, C2-C10 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b-, or -(CHOH)a-; each of W120, W121, W122, W123, W 24, W 25, W 26 and W127 is independently a single bond, -(CH2)n- -(HCCH)n- -0-, -S-, -SO- -S02- -S03- -OS02- -NR 1-, -CO-, -COO- -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR14-, -NR15COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n-, -S(CH2)n- -NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n-, - OCOO(CH2)n- -CONR2 (CH2)n-, -CONR22(CH2)n-, -NR23CO(CH2)n-, -OCONR24(CH2)n- - NR25COO(CH2)n- -NR26CONR27(CH2)n-, -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n- - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-; each of R 20 to R127 is independently a hydrogen, -OCF3, C C3 alkyl, C5-C20 alkyl, C3-C20 cycloalkyl, C5 aryl, C7-C20 aryl, C5-C20 heteroaryl, C-|-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5- C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R °, - SOR41, -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR5 R53, -COR54, -N02, -S02R55 -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; each of R144, R145, R147 and R148 is independently a hydrogen, -OCF3, C1-C20 alkyl, C3-C20 cycloalkyl, C5-C2o aryl, C5-C20 heteroaryl, C-i-Cj-o acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, d-Ce alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR4 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; each of o, p, q, r, s, t, u, and v is independently 0 or 1 ; each of a and b is independently an integer selected from the range of 1 to 100; each n is independently an integer selected from the range of 1 to 10; each of R1 - R33 is independently hydrogen, C C2o alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C20 aryl; each of R40-R55 and R58- R6 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
[042] In an embodiment, the invention provides a compound being of formula (FX59):
N02 (FX59); or a pharmaceutically acceptable salt or ester thereof, wherein: each of L 20, L 2 , L 22, L123, L124, L125, L126 and L127, if present, is independently C C10 alkylene, C3-C 0 cycloalkylene, C2-Ci0 alkenylene, C3-C 0 cycloalkenylene, C2-C10 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b- or -(CHOH)a-; each of W 20, W12 , W122, W123, W124, W125, W126 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -S02- -S03- -OS02-, -NR1 -, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR 3CO- -OCONR14-, -NR15COO- -NR16CONR17-, -NR 8CSNR19-, -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n- -OCO(CH2)n-, - OCOO(CH2)n- -CONR21(CH2)n-, -CONR22(CH2)n- -NR23CO(CH2)n-, -OCONR24(CH2)n-, - NR25COO(CH2)n-, -NR26CONR 7(CH2)n- -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR3 (CH2)nNR32CO-, or -CO(CH2)nNR33CO-; each of R 20 to R 27 is independently a hydrogen, -OCF3, -CH3, -CH2CH3, C4-C20 alkyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C20 heteroaryl, Ci-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R4 R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; each of R144, R145 and R147 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C20 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C^Ce alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R4°, -SOR41 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -P03R56R57, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm; each of o, p, q, r, s, t, u, and v is independently 0 or 1 ; each of a and b is independently an integer selected from the range of 1 to 100; each n is independently an integer selected from the range of 1 to 10; each of R 1 - R33 is independently hydrogen,
Figure imgf000037_0001
alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C2o aryl; each of R40-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenyixanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
[043] In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120 to R 27 is an electron donating group and at least one of R120 to R127 is an electron withdrawing group. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120 to R127 is C C6 alkyl, -OR46, -SR47, - NR48R49, or -NR50COR51; and wherein at least one of R120to R127 is -CN, -C02R40, -S02OR43, - CONR52R53, -COR54, -N02, -SOR41 , -S02R55, -P03R44R45, halo, C C6 acyl, trihalomethyl, or - S02NR58R59. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120 to R127 is -OR46, -SR47, -NR48R49, or -NR50COR51. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120 to R127 is -NR48R49, or -NR50COR51. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120 to R127 is -CN, - CO2R40,-COR54, -N02, -S02R55, or -S02NR58R59. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59),wherein at least one of R120to R127 is -C02R4°, - COR -S02NR R or -S02R In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120to R127 is -NR48R49, and wherein at least one of R120 to R 27 is -CO2R40,-COR54, -S02NR58R59 or -S02R55. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R 20 to R127 and R144 to R148 is Bm. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of R120to R127 and R144 to R148 is FL. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein o is 0 and W 20 is a single bond; or wherein p is 0 and W 21 is a single bond; or wherein q is 0 and W122 is a single bond; or wherein r is 0 and W123 is a single bond; or wherein s is 0 and W 24 is a single bond; or wherein t is 0 and W125 is a single bond; or wherein u is 0 and W126 is a single bond; or wherein v is 0 and W 27 is a single bond. In an embodiment, the invention provides a compound being of formula (FX27) - (FX59), wherein at least one of: L 20 with W120, L121 with W12 , L122 with W122, L123 with W123, L 24 with W 24, L 25 with W 25, L 26 with W126, and L127 with W127 combine to form: -(CH2)n- -0(CH2)n- -CO(CH2)n- -OCO(CH2)n- -COO(CH2)n- -OCOO(CH2)n-, - N(R20)(CH2)n-, -CON(R22)(CH2)n-, -N(R23)CO(CH2)n-, -OCONR24(CH2)n-, -NR25COO(CH2)n-, - NR26CONR27(CH2)n-, or -NR28CSNR29(CH2)n-.
[044] In an embodiment, the invention provides a compound being of formula (FX39) - (FX59), wherein at least one of R144 to R 48 is -N02, and optionally wherein at least two of R144 to R148 are -N02. In an embodiment, the invention provides a compound being of formula (FX39) - (FX59), wherein at least one of R 44 to R148 is -CH3. In an embodiment, the invention provides a compound being of formula (FX39) - (FX59), wherein at each of R144 to R 48 is -N02 hydrogen, or C C6 alkyl, optionally hydrogen, or C-|-C3 alkyl, and optionally hydrogen. . In an embodiment, the invention provides a compound being of formula (FX39) - (FX59), wherein at least two of R144 to R148 are -N02.
[045] The present invention includes therapeutic agents for biomedical applications, including phototherapy, comprising purified stereoisomers (e.g., enantiomers and diastereomers), salts (including quarternary salts), and/or ionic forms (e.g., protonated and deprotonated forms) of the compounds of any of formula (FX1) - (FX59), and mixtures thereof. As will be understood by those having general skill in the art, acidic functional groups and basic functional groups of the compounds of any of formula (FX1) - (FX59) may be in protonated or deprotonated states depending on the molecular environment (e.g., pH, ionic strength, composition, etc.), for example during synthesis, formulation and/or administration
[046] The invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67 , R70-R85, R120-R127, and Ar - Ar3 is independently a group comprising one or more aromatic rings or heterocyclic aromatic rings having one or more electron donating groups and electron withdrawing groups provided as substituents directly or indirectly linked to the aromatic rings or heterocyclic aromatic rings. In an embodiment, for example, compounds of the present invention have any one of formula (FX1) - (FX59), wherein at least one of R - R3, R65 - R67, R70-R85, R120-R127, and Ar1 - Ar3 is a C5-C30 aryl or C5-C30 heteroaryl group, optionally a C5-C2o aryl or C5-C20 heteroaryl group, or C5-C10 aryl or C5-C10 heteroaryl group having at least one electron withdrawing group and at least one electron donating group as substituents. In an embodiment of the is aspect, the electron withdrawing group of the C5-C30 aryl or C5-C30 heteroaryl group is -CN, halo, -C02R40, -COR54, -N02, -S02R55, or -S02NR58R59. In an embodiment of this aspect, the electron donating group of the C5-C30 aryl or C5-C3o heteroaryl group is -OR46, -SR47, -NR48R49, or -NR50COR51. The invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, R120-R127, and Ar1 - Ar3 is independently a group comprising one or more aromatic rings or heterocyclic aromatic rings having one or more targeting ligands (e.g., Bm) provided as substituents directly or indirectly linked to the aromatic rings or heterocyclic aromatic rings, and optionally having one or more electron donating groups and electron withdrawing groups provided as substituents directly or indirectly linked to the aromatic rings or heterocyclic aromatic rings.
[047] In an embodiment, for example, the invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, R 20-R127, and Ar1 - Ar3 is independently a group corresponding to benzene, naphthalene, naphthoquinone,
diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene,
naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline, optionally having one or more electron donating groups, electron withdrawing groups, or targeting ligands provided as one or more substituents. In an embodiment, for example, the invention provides compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R - R3, R65 - R67, R70-R85, R120- R127, and Ar1 - Ar3 is independently a group corresponding to a cyanine, indocyanine,
phenothiazine, and phthalocyanine. In an embodiment, the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, R1 0-R127, and Ar1 - Ar3 is independently benzene or a derivative thereof. In an embodiment, the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, R120-R127, 7 and Ar1 - Ar3 is independently a polycyclic aromatic group corresponding to naphthalene, diphenylmethane, fluorene, anthracene, phenanthrene, tetracene, naphthoquinone, anthraquinone, naphthacenedione, azulene, aza-azulene or anthracycline. In an embodiment, the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one of R1 - R3, R65 - R67, R70-R85, R120-R127, and Ar1 - Ar3 is independently a heterocyclic aromatic group having a six member nitrogen-containing ring corresponding to pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, purine, acridine, acridone, or phenanthridine. In an embodiment, the invention provides a compound for use in a
phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one R1 - R3, R65 _ R67^ R7o_R85^ Ri2o_Ri27^ a nd Afi _ Af3 js jncjepencientiy a heterocyclic aromatic group having a five member nitrogen-containing ring corresponding to pyrrole, pyrazole, indole, isoindole, imidazole, oxazole, thiazole, purine, benzimidazole, or carbazole. In an embodiment, the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one of R1 - R3, R65 - R67, R70-R85, R120-R127, and Ar1 - Ar3 is
independently a heterocyclic aromatic group having a five or six member oxygen-containing ring corresponding to furan, oxazole, benzofuran, dibenzofuran, xanthene, xanthone, flavone, or coumarin. In an embodiment, the invention provides a compound for use in a phototherapy procedure having any one of formula (FX1) to (FX59); wherein at least one of R1 - R3, R65 - R67, R70-R85, R120-R127, and Ar1 - Ar3 is independently a heterocyclic aromatic group having a five member sulfur-containing ring corresponding to thiophene, thiazole, benzothiophene, or dibenzothiophene.
[048] The invention includes compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, Ar -Ar3, or R120-R127 is independently a dye (abbreviated as "FL"), such as a C5-C30 aryl or C5-C30 heteroaryl chromophore and/or C5-C30 aryl or C5-C30 heteroaryl fluorophore, that is excited upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, optionally selected over the range of 400 nanometers to 900 nanometers. Compounds of this aspect of the present invention include bifunctional optical agents capable of providing tandem functionality as a photosensitizer and an imaging agent. In an embodiment, for example, the invention provides a compound having any one of formula (FX1 ) - (FX59) that functions as a photosensitizer upon exposure to electromagnetic radiation having a first distribution of wavelengths, and wherein at least one of R1 - R3, R65 - R67, R70-R85, R1 0-R127, and Ar1 - Ar3 is independently a fluorophore that is excited upon exposure to electromagnetic radiation having a second distribution of wavelengths that is different from the first distribution of wavelengths, for example, wherein the first and second distributions of wavelengths correspond to different absorption maxima and, optionally wherein the first and second distributions of wavelengths corresponding to absorption peaks that are not overlapping, or corresponding to absorption maxima in the visible or near IR regions of the spectrum that differ by 20 nanometers or more. In an embodiment, for example, at least one of R1 - R3, R65 - R67, R70-R85, R120-R127, and Ar1 - Ar3 is independently a C5-C30 aryl or C5-C30 heteroaryl fluorophore having one or more electron donating groups as substituents, having one or more electron withdrawing groups as substituents, or having both electron donating and electron withdrawing groups as substituents. In an embodiment, at least one of R1 - R3, R65 - R67, R70-R85, R 20-R127, and Ar1 - Ar3 is independently a fluorophore group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an azaazulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a benzoindocarbocyanine, optionally having one or more electron donating groups, electron withdrawing groups, or targeting ligands provided as one or more substituents. [049] In an embodiment, the invention provides optical agents for phototherapy having a ligand component for targeting the optical agent to a selected organ, tissue, or other cell material.
Incorporation of a targeting ligand or molecular recognition component in some compounds and methods of the invention enables targeted delivery such that at least a portion of phototherapeutic agent administered to a subject preferentially accumulates at a preselected, desired site, such as the site of an organ, tissue, tumor or other lesion, prior to or during exposure to electromagnetic radiation. Targeting ligands of the present invention may be indirectly or directly linked to, or non- covalently associated with, the central S-N group of formulas (FX1) - (FX59). The invention includes, for example, compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, or R 20-R127 is independently a targeting ligand (abbreviated as "Bm" throughout this description). In an embodiment, for example, the invention includes compounds wherein R is Bm and W1 is -NR13CO- -CONR12-, -OCONR14-, -NR15COO- or -NR16CONR17- ; or R2 is Bm and W2 is -NR 3CO- -CONR12-, -OCONR14-, -NR15COO- or -NR 6CONR17-, or
R3 is Bm and W3 is -NR13CO- -, -CONR12- -, -OCONR14- -, -NR 5COO- -, or -NR16CONR17- -, or R120 is Bm and W 20 is -NR13CO- -CONR12-, -OCONR14- -NR15COO- or - NR 6CONR17-; or R121 is Bm and W12 is -NR13CO- -CONR12- -OCONR14-, -NR 5COO- or - NR16CONR17-; or R122 is Bm and W122 is -NR13CO- -CONR12- -OCONR14-, -NR15COO- or - NR16CONR17-; or R123 is Bm and W123 is -NR13CO- -CONR 2-, -OCONR14-, -NR15COO- or - NR 6CONR17-; or R124 is Bm and W 24 is -NR 3CO- -CONR12-, -OCONR14-, -NR 5COO-, or - NR16CONR17- ; or R125 is Bm and W 25 is -NR 3CO- -CONR 2- -OCONR14- -NR 5COO- or - NR16CONR17-; or R126 is Bm and W126 is -NR13CO- -CONR12- -OCONR 4-, -NR15COO-, or - NR16CONR17-; or R127 is Bm and W127 is -NR13CO- -CONR12-, -OCONR 4-, -NR15COO- or - NR 6CONR17-.
[050] In an embodiment, for example, the invention includes compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, or R 20-R127 is independently a polypeptide comprising 2 to 30 amino acid units. In an embodiment, for example, the invention includes compounds of any one of formula (FX1) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, or R120-R127 is independently an antibody or fragment thereof. In an embodiment, for example, the invention includes compounds of any one of formula (FX1 ) - (FX59), wherein at least one of R - R3, R65 - R67, R70-R85, or R120-R127 is independently a polynucleotide comprising 1 to . 50 nucleic acid units. In an embodiment, for example, the invention includes compounds of any one of formula (FX1) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, or R1 0-R127 is independently an aptamer.
[051] In an embodiment, the invention provides compounds of any one of formula (FX1) -
(FX59), wherein each of R - R33 is independently hydrogen, a C1-C10 cycloalkyl, or a C C 10 alkyl, and optionally wherein each of R11 - R33 is hydrogen, a C C10 cycloalkyl, or a C C5 alkyl, and optionally wherein each of R11 - R33 is hydrogen. In an embodiment, the invention provides compounds of any one of formulas (FX1) - (FX59), wherein each of R40 - R55 and R58 - R6 is independently hydrogen, C C 0 cycloalkyl, or C C5 alkyl, and optionally hydrogen, or C C3 alkyl, and optionally hydrogen. In an embodiment, the invention provides compounds of any one of formula (FX1) - (FX59), wherein each of R40 - R55 and R58 - R61 is hydrogen. In an embodiment, the invention provides compounds of any one of formulas (FX1 ) - (FX59), wherein at least one of R1, R2, and R3 is independently C5-C2o aryl or C5-C20 heteroaryl, optionally C5-C10 aryl or C5-C10 heteroaryl. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having any one of formula (FX1) - (FX59), wherein each of R1, R2, and R3 is independently hydrogen, C C2o alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, - CH(R62)C02H, -CH(R63)NH2, FL a dye, or a targeting Iigand (Bm); wherein at least one of R1, R2, and R3 is independently C5-C30 aryl or C5-C30 heteroaryl, and optionally C5-C20 aryl or C5-C20 heteroaryl, or C5-C10 aryl or C5-Ci0 heteroaryl. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having any one of formula
(FX1) - (FX59), wherein R1 is C C10 alkyl, or C3-Ci0 cycloalkyl, C5-C30 aryl, or C5-C30 heteroaryl wherein optionally the C5-C30 aryl or C5-C20 heteroaryl includes an electron donating group, electron withdrawing group, and/or Bm as a substituent, and each of R2, and R3 is independently a hydrogen, Ci-C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C20 heteroaryl, a FL or Bm, wherein optionally the C5-C30 aryl or C5-C20 heteroaryl includes an electron donating group, electron withdrawing group, or a targeting Iigand (Bm) as a substituent. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having any one of formulas (FX1) - (FX59), wherein at least one of R1, R2, and R3 is a FL, such as a fluorophore that is excited upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, preferably wavelengths selected over the range of 400 nanometers to 900 nanometers for some applications.
[052] L1, L2, L3 and W1, W2, W3 groups may be spacer and attaching groups, respectively, for providing an appropriate linkage between R1, R2, R3 and the central S-N bond in the compounds of (FX1) - (FX59). In some embodiments, the invention provides compounds of any one of formulas (FX1) - (FX59), wherein any one of L , L2, and L3 is independently a spacer moiety for establishing the steric environment between R , R2, and/or R3 and the central S-N group providing useful optical, pharmacokinetic, or targeting properties. In some embodiments, the invention provides compounds of any one of formulas (FX1) - (FX59), wherein any one of W , W2, and W3 is independently an attaching moiety for attaching R1 , R2, and/or R3 directly or indirectly to the central S-N group. In an embodiment, the invention provides compounds having formula (FX1) - (FX59), wherein at least one of e, f and g is 0, and optionally all of e, f and g are 0. In an embodiment, the invention provides compounds having formula (FX1) - (FX59), and related phototherapy methods, wherein e is 0, and/or f is 0, and/or g is 0. In an embodiment, the invention provides compounds useful as optical agents for phototherapeutic methods having any of formulas (FX1 ) - (FX59), wherein at least one of W1, W2, and W3 is a single bond, and optionally all of W1, W2, and W3 are single bonds. In an embodiment, the invention provides compounds having formula (FX1) - (FX59), wherein W1 is a single bond, and/or W2 is a single bond, and/or W3 is a single bond. In an embodiment, the invention provides compounds having formula (FX1) - (FX59), and related phototherapy methods, wherein e is 0 and W is a single bond, and/or f is 0 and W2 is a single bond, and/or g is 0 and W3 is a single bond.
[053] In an embodiment, at least one of L - L3 is independently -(CH2)m- -(HCCH)m-, - (CHOH)m- , or -(CH2CH20)m-, wherein each of m is independently an integer selected from the range of 1 to 10. In an embodiment, the invention provides compounds of any one of formulas (FX1) - (FX59), wherein at least one of W1 - W3 is independently a single bond, -0-, -CO-, - COO- -OCO-, -OCOO-, -NR 1-, -CONR12-, -NR13CO- -NR16CONR17-, or -NR 8CSNR19-. In an embodiment, the invention provides compounds of any one of formulas (FX1) - (FX59), wherein at least one of: L1 with W , L2 with W2, and L3 with W3, combine to form: -(CH2)n-> - 0(CH2)n- -CO(CH2)n- -OCO(CH2)n-, -COO(CH2)n-, -OCOO(CH2)n- -N(R20)(CH2)n-, - CON(R22)(CH2)n-, -N(R23)CO(CH2)n-, -OCONR24(CH2)n-, -NR25COO(CH2)n-, - NR26CONR 7(CH2)n-, or -NR28CSNR29(CH2)n-.
[054] In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1 , Ar2, and Ar3 do not include an azo group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1 , Ar2, and Ar3 do not include an diaza group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1, Ar2, and Ar3 do not include an oxaza group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1 , Ar2, and Ar3 do not include an azide group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R 20-R127, Ar1, Ar2, and Ar3 do not include a sulfenate group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R - R3, R65 - R67, R70-R85, R 20-R127, Ar1, Ar2, and Ar3 do not include a thiadiazole group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R 20- R 27, Ar1, Ar2, and Ar3 do not include a cyanate group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R - R3, R65 - R67, R70-R85, R120-R127, Ar1 , Ar2, and Ar3 do not include an isocyanide group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1 , Ar2, and Ar3 do not include an isocyanate group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1 ) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1, Ar2, and Ar3 do not include an isothiocyanate group. In an embodiment, for example, the invention provides a compound for phototherapy having any one of formula (FX1) - (FX59), wherein R1 - R3, R65 - R67, R70-R85, R120-R127, Ar1, Ar2, and Ar3 do not include a thiocyanate group. [055] In some embodiments, compounds of the invention may optionally include a
poly(ethylene glycol) (abbreviated as PEG) component. In an embodiment, for example, the invention provides a composition having any one of the formula (FX1 ) - (FX59), wherein at least one of R1 - R3, R65 - R67, R70-R85, R 20-R127, or L - L3 is a substituent comprising -(CH2OCH2) m- wherein m is an integer selected from the range of 1 to 100. Incorporation of a poly(ethylene glycol) component in some compositions of the invention provides pharmacokinetic, chemical, and/or physical properties useful for bioanalytical, diagnostic and/or phototherapeutic applications. Poly(ethylene glycol) containing compounds of some embodiments of the present invention, for example, provide enhanced biocompatibility, low toxicity and suppress immune responses upon administration. Poly(ethylene glycol) containing compounds of some embodiments of the invention facilitate formulation, administration and/or delivery of the compounds, for example, by enhancing solubility.
[056] The invention further provides a compound having any one of formula (FX1 ) - (FX59), or a pharmaceutical formulation thereof, for use in an optical imaging, diagnostic, and/or
phototherapeutic biomedical procedure such as a Type 1 or Type 2 phototherapy procedure. In an embodiment, the invention provides an optical agent comprising a pharmaceutically acceptable formulation, wherein at least one active ingredient of the formulation is a compound having any one of formula (FX1 ) - (FX59) provided in a therapeutically effective amount. The invention includes, for example, formulations comprising a compound having any one of formula (FX1 ) - (FX59) and one or more pharmaceutically acceptable carriers or excipients. In an embodiment, the invention provides a pharmaceutically acceptable formulation for combination therapy comprising a compound having any one of formula (FX1) - (FX59) and one or more additional diagnostic and/or therapeutic agents, such as anti-cancer agents, anti-inflammatory agents, and/or imaging agents (e.g., optical and/or non-optical imaging agents).
[057] In an embodiment, the invention provides methods for a medical phototherapy procedure, such as a phototherapy procedure, wherein the method comprises: (i) administering (e.g., via intravenous or intraarterial injection, oral administration, topical administration, subcutaneous administration, etc.) to a subject a therapeutically or diagnostically effective amount of a compound having any one of formula (FX1 ) - (FX59) and (ii) exposing the administered compound to electromagnetic radiation. In an embodiment, the administrating step is carried out under conditions sufficient for contacting the compound with a target tissue or cell, wherein the compound selectively binds to or otherwise preferentially associates with the target tissue or cell. In an embodiment, the administered compound is exposed to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers, optionally having wavelengths selected over the range of 400 nanometers to 900 nanometers. In an embodiment, exposing the administered compound to electromagnetic radiation cleaves a S-N bond of the compound. In an embodiment, exposing the administered compound to electromagnetic radiation generates a therapeutically effective amount of photoactivated compound. In an embodiment, exposing the administered compound to electromagnetic radiation generates a therapeutically effective amount of reactive species causing localized cell death, inactivation or injury. In an embodiment, the medical phototherapy procedure comprises administering, contacting or otherwise targeting the compound to or with a target tissue or cell of the subject, such as a tumor, lesion, site of inflammation, vasculature tissue, or organ. In an embodiment, methods of the invention further comprise exposing the administered compound at the target tissue to light having sufficient power, fluence, intensity and/or dose (net number of photons provided to the target tissue) to result in injury, inactivation and/or death to target cells or cells at the target tissue.
[058] In an embodiment, the medical phototherapy procedure comprises administering, contacting or otherwise targeting the administered compound to or with a target tissue or cell of the subject, such as a tumor, lesion, site of inflammation, vasculature tissue, or an organ. In an embodiment, for example, the target tissue is a tissue type selected from the group consisting of breast, lung, throat, cervical, colon, kidney, stomach, ovarian, testicular, prostate, gastric, esophageal, uterine, endometrial, and pancreatic tissue. In an embodiment, exposing the administered compound to electromagnetic radiation generates fluorescence, wherein the medical phototherapy procedure further comprises detecting fluorescence from the administered compound. In an embodiment, exposing the administered compound to electromagnetic radiation generates a diagnostically effective amount of fluorescence, for example an amount of fluorescence allowing for optical detection, visualizing and/or imaging of the target tissue. In an embodiment, a method of the invention further comprises exposing the administered compound at the target tissue to electromagnetic radiation having sufficient power, fluence, intensity and/or dose (net number of photons provided to the target tissue) to provide optical detection, visualization and/or imaging of the target tissue. In an embodiment, a method of the invention further comprises generating an image of the fluorescence from the compound administered to the subject. In an embodiment, a method of the invention further comprises visualizing the fluorescence from the compound.
[059] In a method, the electromagnetic radiation exposed to the compound of any one of formulas (FX1) - (FX59) does not have wavelengths in the X-ray region of the electromagnetic spectrum. In a method, the electromagnetic radiation exposed to the compound of any one of formulas (FX1) - (FX59) does not have wavelengths in the ultraviolet region of the electromagnetic spectrum, for example, not including light having wavelengths equal to or less than 380 nanometers. In an embodiment, non-ionizing electromagnetic radiation is used in the present methods. The term "non-ionizing electromagnetic radiation" as used herein refers to
electromagnetic radiation wherein a single photon does not have enough energy to completely remove at least one electron from an atom or molecule of the subject's body.
[060] Without wishing to be bound by any particular theory, there can be discussion herein of beliefs or understandings of underlying principles or mechanisms relating to the invention. It is recognized that regardless of the ultimate correctness of any explanation or hypothesis, an embodiment of the invention can nonetheless be operative and useful. BRIEF DESCRIPTION OF THE FIGURES
[061] Figures 1 A - 1 F provide schematic representations of reaction mechanisms for sulfenamide optical agents having an acyclic S-N bond, wherein photoactivation generates reactive species and cell death. Figures 1A and 1 B provide schematic representations of reaction mechanisms for phototherapeutic agents comprising a sulfenamide compound having formula (FX1). Figures 1 C and 1 D provide schematic representations of reaction mechanisms for phototherapeutic agents comprising a sulfenamide compound having formula (FX16). Figures 1 E and 1 F provide schematic representations of reaction mechanisms for phototherapeutic agents comprising a sulfenamide compound having a fused ring chromophore group.
[062] Figure 2 provides schemes for the general synthesis of sulfenamide optical agents having an acyclic S-N bond useful in phototherapeutic methods of the present invention.
[063] Figures 3A and 3B provide schemes for the preparation of sulfenamide bioconjugate optical agents. Figure 3A provides schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond and peptide targeting ligand useful in phototherapeutic methods of the invention. Figure 3B provides schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond, peptide targeting ligand and phenyl group.
[064] Figures 4A and 4B provide schemes for the preparation of sulfenamide optical agents having fused ring chromophore groups, and optionally a peptide targeting ligand.
[065] Figure 5 provides cell viability results for control conditions (no photosensitizer) wherein cells were exposed to light in the presence of dimethyl sulfoxide.
[066] Figure 6 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX18). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
[067] Figure 7 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX49). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
[068] Figure 8 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX47). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
[069] Figure 9 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX48). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
[070] Figure 10 provides general schemes for the syntheses of bioconjugates of sulfenamide optical agents having formula (FX1).
[071] Figures 11 A and 11B illustrate examples of coupling reactions useful for synthesis of sulfenamide optical agents of the invention having specific targeting ligands.
[072] Figure 12 provides general schemes for the syntheses of bioconjugates of sulfenamide optical agents having formula (FX49). [073] Figures 13A and 13B illustrate examples of coupling reactions useful for synthesis of sulfenamide optical agents of the invention having specific targeting ligands.
[074] Figure 14 provides electron spin resonance spectra for the sulfenamide of formula (FX18) for test conditions before, during, and after the exposure of the sulfenamide to light. The spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
[075] Figure 15 provides electron spin resonance spectra for the sulfenamide of formula (FX47) for test conditions before, during, and after the exposure of the sulfenamide to light. The spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
[076] Figure 16 provides electron spin resonance spectra for the sulfenamide of formula (FX47) for test conditions before, during, and after the exposure of the sulfenamide to light. The spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
[077] Figure 17 provides electron spin resonance spectra for the sulfenamide of formula (FX48) for test conditions before, during, and after the exposure of the sulfenamide to light. The spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
[078] Figure 18 provides electron spin resonance spectra for the sulfenamide of formula (FX48) for test conditions before, during, and after the exposure of the sulfenamide to light. The spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
[079] Figure 19 provides electron spin resonance spectra for the sulfenamide of formula (FX49) for test conditions before, during, and after the exposure of the sulfenamide to light. The spectra indicate that radicals are formed upon the exposure of the sulfenamide to light and are present for a period of time after the exposure to light.
[080] Figure 20 provides cell viability results for test conditions wherein cells were exposed to light in presence of a sulfenamide optical agent having formula (FX50). The results show concentration-dependent and light exposure-dependent decreases in cell viability.
STATEMENTS REGARDING CHEMICAL COMPOUNDS AND NOMENCLATURE
[081] In an embodiment, a composition or compound of the invention is isolated or purified. In an embodiment, an isolated or purified compound is at least partially isolated or purified as would be understood in the art. In an embodiment, the composition or compound of the invention has a chemical purity of 95%, optionally for some applications 99%, optionally for some applications 99.9%, optionally for some applications 99.99%, and optionally for some applications 99.999% pure. [082] Many of the molecules disclosed herein contain one or more ionizable groups, lonizable groups include groups from which a proton can be removed (e.g., -COOH) or added (e.g., amines) and groups which can be quaternized (e.g., amines). All possible ionic forms of such molecules and salts thereof are intended to be included individually in the disclosure herein. With regard to salts of the compounds herein, one of ordinary skill in the art can select from among a wide variety of available counterions that are appropriate for preparation of salts of this invention for a given application. In specific applications, the selection of a given anion or cation for preparation of a salt can result in increased or decreased solubility of that salt.
[083] The compounds of this invention can contain one or more chiral centers. Accordingly, this invention is intended to include racemic mixtures, diasteromers, enantiomers, tautomers and mixtures enriched in one or more stereoisomer. The scope of the invention as described and claimed encompasses the racemic forms of the compounds as well as the individual enantiomers and non-racemic mixtures thereof.
[084] As used herein, the term "group" may refer to a functional group of a chemical compound. Groups of the present compounds refer to an atom or a collection of atoms that are a part of the compound. Groups of the present invention may be attached to other atoms of the compound via one or more covalent bonds. Groups may also be characterized with respect to their valence state. The present invention includes groups characterized as monovalent, divalent, trivalent, etc. valence states.
[085] As used herein, the term "substituted" refers to a compound wherein a hydrogen is replaced by another functional group.
[086] As is customary and well known in the art, hydrogen atoms in formulas (FX1) - (FX59) are not always explicitly shown, for example, hydrogen atoms bonded to the carbon atoms of aromatic, heteroaromatic, and alicyclic rings are not always explicitly shown in formulas (FX1) - (FX59). The structures provided herein, for example in the context of the description of formulas (FX1 ) - (FX59), are intended to convey to one of reasonable skill in the art the chemical composition of compounds of the methods and compositions of the invention, and as will be understood by one of skill in the art, the structures provided do not indicate the specific positions of atoms and bond angles between atoms of these compounds.
[087] As used herein, the terms "alkylene" and "alkylene group" are used synonymously and refer to a divalent group derived from an alkyl group as defined herein. The invention includes compounds having one or more alkylene groups. Alkylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention may have substituted and/or unsubstituted C C20 alkylene, C-i-C-io alkylene and C C5 alkylene groups.
[088] As used herein, the terms "cycloalkylene" and "cycloalkylene group" are used
synonymously and refer to a divalent group derived from a cycloalkyi group as defined herein. The invention includes compounds having one or more cycloalkylene groups. Cycloalkyi groups in some compounds function as attaching and/or spacer groups. Compounds of the invention may have substituted and/or unsubstituted C3-C2o cycloalkylene, C3-C10 cycloalkylene and C3-C5 cycloalkylene groups.
[089] As used herein, the terms "arylene" and "arylene group" are used synonymously and refer to a divalent group derived from an aryl group as defined herein. The invention includes compounds having one or more arylene groups. In some embodiments, an arylene is a divalent group derived from an aryl group by removal of hydrogen atoms from two intra-ring carbon atoms of an aromatic ring of the aryl group. Arylene groups in some compounds function as attaching and/or spacer groups. Arylene groups in some compounds function as chromophore, fluorophore, aromatic antenna, dye and/or imaging groups. Compounds of the invention include substituted and/or unsubstituted C3-C30 arylene, C3-C20 arylene, C3-C10 arylene and Ci-C3 arylene groups.
[090] As used herein, the terms "heteroarylene" and "heteroarylene group" are used synonymously and refer to a divalent group derived from a heteroaryl group as defined herein. The invention includes compounds having one or more heteroarylene groups. In some embodiments, a heteroarylene is a divalent group derived from a heteroaryl group by removal of hydrogen atoms from two intra-ring carbon atoms or intra-ring nitrogen atoms of a heteroaromatic or aromatic ring of the heteroaryl group. Heteroarylene groups in some compounds function as attaching and/or spacer groups. Heteroarylene groups in some compounds function as chromophore, aromatic antenna, fluorophore, dye and/or imaging groups. Compounds of the invention include substituted and/or unsubstituted C3-C30 heteroarylene, C3-C2o heteroarylene, C C-i o heteroarylene and C3-C5 heteroarylene groups.
[091] As used herein, the terms "alkenylene" and "alkenylene group" are used synonymously and refer to a divalent group derived from an alkenyl group as defined herein. The invention includes compounds having one or more alkenylene groups. Alkenylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention include substituted and/or unsubstituted C2-C20 alkenylene, C2-Ci0 alkenylene and C2-C5 alkenylene groups.
[092] As used herein, the terms "cylcoalkenylene" and "cylcoalkenylene group" are used synonymously and refer to a divalent group derived from a cylcoalkenyl group as defined herein. The invention includes compounds having one or more cylcoalkenylene groups. Cycloalkenylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention include substituted and/or unsubstituted C3-C20 cylcoalkenylene, C3-C-i0 cylcoalkenylene and C3-C5 cylcoalkenylene groups.
[093] As used herein, the terms "alkynylene" and "alkynylene group" are used synonymously and refer to a divalent group derived from an alkynyl group as defined herein. . The invention includes compounds having one or more alkynylene groups. Alkynylene groups in some compounds function as attaching and/or spacer groups. Compounds of the invention include substituted and/or unsubstituted C2-C2o alkynylene, C2-C10 alkynylene and C2-C5 alkynylene groups.
[094] As used herein, the term "halo" refers to a halogen group such as a fluoro (-F), chloro (-CI), bromo (-Br), iodo (-I) or astato (-At).
[095] The term "heterocyclic" refers to ring structures containing at least one other kind of atom, in addition to carbon, in the ring. Examples of such heteroatoms include nitrogen, oxygen and sulfur. Heterocyclic rings include heterocyclic alicyclic rings and heterocyclic aromatic rings. Examples of heterocyclic rings include, but are not limited to, pyrrolidinyl, piperidyl, imidazolidinyl, tetrahydrofuryl, tetrahydrothienyl, furyl, thienyl, pyridyl, quinolyl, isoquinolyl, pyridazinyl, pyrazinyl, indolyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, pyridinyl, benzoxadiazolyl, benzothiadiazolyl, triazolyl and tetrazolyl groups. Atoms of heterocyclic rings can be bonded to a wide range of other atoms and functional groups, for example, provided as substituents.
[096] The term "carbocyclic" refers to ring structures containing only carbon atoms in the ring. Carbon atoms of carbocyclic rings can be bonded to a wide range of other atoms and functional groups, for example, provided as substituents..
[097] The term "alicyclic ring" refers to a ring, or plurality of fused rings, that is not an aromatic ring. Alicyclic rings include both carbocyclic and heterocyclic rings.
[098] The term "aromatic ring" refers to a ring, or a plurality of fused rings, that includes at least one aromatic ring. The term aromatic ring includes aromatic rings comprising carbon, hydrogen and heteroatoms. Aromatic ring includes carbocyclic and heterocyclic aromatic rings. Aromatic rings are components of aryl groups.
[099] The term "fused ring" or "fused ring structure" refers to a plurality of alicyclic and/or aromatic rings provided in a fused ring configuration, such as fused rings that share at least two intra ring carbon atoms and/or heteroatoms.
[0100] As used herein, the term "alkoxyalkyl" refers to a substituent of the formula alkyl-O-alkyl.
[0101] As used herein, the term "polyhydroxyalkyl" refers to a substituent having from 2 to 12 carbon atoms and from 2 to 5 hydroxyl groups, such as the 2,3-dihydroxypropyi, 2,3,4- trihydroxybutyl or 2,3,4,5-tetrahydroxypentyl residue.
[0102] As used herein, the term "polyalkoxyalkyl" refers to a substituent of the formula alkyl- (alkoxy)n-alkoxy wherein n is an integer from 1 to 10, preferably 1 to 4, and more preferably for some embodiments 1 to 3.
[0103] Amino acids include glycine, alanine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tryptophan, asparagine, glutamine, glycine, serine, threonine, serine, Threonine, asparagine, glutamine, tyrosine, cysteine, lysine, arginine, histidine, aspartic acid and glutamic acid. As used herein, reference to "a side chain residue of a natural a-amino acid" specifically includes the side chains of the above-referenced amino acids.
[0104] Aikyl groups include straight-chain, branched and cyclic alkyl groups. AlkyI groups include those having from 1 to 30 carbon atoms. Alkyl groups include small alkyl groups having 1 to 3 carbon atoms. Alkyl groups include medium length alkyl groups having from 4-10 carbon atoms. Alkyl groups include long alkyl groups having more than 10 carbon atoms, particularly those having 10-30 carbon atoms. The term cycloalkyi specifically refers to an alky group having a ring structure such as ring structure comprising 3-30 carbon atoms, optionally 3-20 carbon atoms and optionally 2 - 10 carbon atoms, including an alkyl group having one or more rings. Cycloalkyi groups include those having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring(s) and particularly those having a 3-, 4-, 5-, 6-, or 7-member ring(s). The carbon rings in cycloalkyi groups can also carry alkyl groups. Cycloalkyi groups can include bicyclic and tricycloalkyl groups. Alkyl groups are optionally substituted. Substituted alkyl groups include among others those which are substituted with aryl groups, which in turn can be optionally substituted. Specific alkyl groups include methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, n-butyl, s-butyl, t-butyl, cyclobutyl, n-pentyl, branched- pentyl, cyclopentyl, n-hexyl, branched hexyl, and cyclohexyl groups, all of which are optionally substituted. Substituted alkyl groups include fully halogenated or semihalogenated alkyl groups, such as alkyl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms. Substituted alkyl groups include fully fluorinated or semifluorinated alkyl groups, such as alkyl groups having one or more hydrogens replaced with one or more fluorine atoms. An alkoxy group is an alkyl group that has been modified by linkage to oxygen and can be represented by the formula R-0 and can also be referred to as an alkyl ether group. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, butoxy and heptoxy. Alkoxy groups include substituted alkoxy groups wherein the alky portion of the groups is substituted as provided herein in connection with the description of alkyl groups. As used herein MeO- refers to CH30-
[0105] Alkenyl groups include straight-chain, branched and cyclic alkenyl groups. Alkenyl groups include those having 1 , 2 or more double bonds and those in which two or more of the double bonds are conjugated double bonds. Alkenyl groups include those having from 2 to 20 carbon atoms. Alkenyl groups include small alkenyl groups having 2 to 3 carbon atoms. Alkenyl groups include medium length alkenyl groups having from 4-10 carbon atoms. Alkenyl groups include long alkenyl groups having more than 10 carbon atoms, particularly those having 10-20 carbon atoms. Cycloalkenyl groups include those in which a double bond is in the ring or in an alkenyl group attached to a ring. The term cycloalkenyl specifically refers to an alkenyl group having a ring structure, including an alkenyl group having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring(s) and particularly those having a 3-, 4-, 5-, 6- or 7-member ring(s). The carbon rings in
cycloalkenylgroups can also carry alkyl groups. Cycloalkenylgroups can include bicyclic and tricyclic alkenyl groups. Alkenyl groups are optionally substituted. Substituted alkenyl groups include among others those which are substituted with alkyl or aryl groups, which groups in turn can be optionally substituted. Specific alkenyl groups include ethenyl, prop-1 -enyl, prop-2-enyl, cycloprop-1 -enyl, but-1 -enyl, but-2-enyl, cyclobut-1 -enyl, cyclobut-2-enyl, pent-1 -enyl, pent-2-enyl, branched pentenyl, cyclopent-1 -enyl, hex-1 -enyl, branched hexenyl, cyclohexenyl, all of which are optionally substituted. Substituted alkenyl groups include fully halogenated or semihalogenated alkenyl groups, such as alkenyl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms. Substituted alkenyl groups include fully fluorinated or semifluorinated alkenyl groups, such as alkenyl groups having one or more hydrogen atoms replaced with one or more fluorine atoms.
[0106] Aryl groups include groups having one or more 5-, 6- or 7- member aromatic rings, including heterocyclic aromatic rings. The term heteroaryl specifically refers to aryl groups having at least one 5-, 6- or 7- member heterocyclic aromatic rings. Aryl groups can contain one or more fused aromatic rings, including one or more fused heteroaromatic rings, and/or a combination of one or more aromatic rings and one or more nonaromatic rings that may be fused or linked via covalent bonds. Heterocyclic aromatic rings can include one or more N, O, or S atoms in the ring. Heterocyclic aromatic rings can include those with one, two or three N atoms, those with one or two O atoms, and those with one or two S atoms, or combinations of one or two or three N, O or S atoms. Aryl groups are optionally substituted. Substituted aryl groups include among others those which are substituted with alkyl or alkenyl groups, which groups in turn can be optionally substituted. Specific aryl groups include phenyl, biphenyl groups, pyrrolidinyl, imidazolidinyl, tetrahydrofuryl, tetrahydrothienyl, furyl, thienyl, pyridyl, quinolyl, isoquinolyl, pyridazinyl, pyrazinyl, indolyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, pyridinyl, benzoxadiazolyl, benzothiadiazolyl, and naphthyl groups, all of which are optionally substituted. Substituted aryl groups include fully halogenated or semihalogenated aryl groups, such as aryl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms. Substituted aryl groups include fully fluorinated or semifluorinated aryl groups, such as aryl groups having one or more hydrogens replaced with one or more fluorine atoms. Aryl groups include, but are not limited to, aromatic group-containing or heterocylic aromatic group-containing groups corresponding to any one of the following: benzene, naphthalene, naphthoquinone,
diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, tetracenedione, pyridine, quinoline, isoquinoline, indoles, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furans, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene or anthracycline. As used herein, a group corresponding to the groups listed above expressly includes an aromatic or heterocyclic aromatic group, including monovalent, divalent and polyvalent groups, of the aromatic and heterocyclic aromatic groups listed herein are provided in a covalently bonded configuration in the compounds of the invention at any suitable point of attachment. In embodiments, aryl groups contain between 5 and 30 carbon atoms. In embodiments, aryl groups contain one aromatic or heteroaromatic six-membered ring and one or more additional five- or six-membered aromatic or heteroaromatic ring. In embodiments, aryl groups contain between five and eighteen carbon atoms in the rings. Aryl groups optionally have one or more aromatic rings or heterocyclic aromatic rings having one or more electron donating groups, electron withdrawing groups and/or targeting ligands provided as substituents.
[0107] Arylalkyl groups are alkyl groups substituted with one or more aryl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted. Specific alkylaryl groups are phenyl-substituted alkyl groups, e.g., phenylmethyl groups. Alkylaryl groups are alternatively described as aryl groups substituted with one or more alkyl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted. Specific alkylaryl groups are alkyl-substituted phenyl groups such as methylphenyl. Substituted arylalkyl groups include fully halogenated or semihalogenated arylalkyl groups, such as arylalkyl groups having one or more alkyl and/or aryl groups having one or more hydrogens replaced with one or more fluorine atoms, chlorine atoms, bromine atoms and/or iodine atoms.
[0108] As to any of the groups described herein which contain one or more substituents, it is understood that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. In addition, the compounds of this invention include all stereochemical isomers arising from the substitution of these compounds. Optional substitution of alkyl groups includes substitution with one or more alkenyl groups, aryl groups or both, wherein the alkenyl groups or aryl groups are optionally substituted. Optional substitution of alkenyl groups includes substitution with one or more alkyl groups, aryl groups, or both, wherein the alkyl groups or aryl groups are optionally substituted. Optional substitution of aryl groups includes substitution of the aryl ring with one or more alkyl groups, alkenyl groups, or both, wherein the alkyl groups or alkenyl groups are optionally substituted.
[0109] Optional substituents for any alkyl, alkenyl and aryl group includes substitution with one or more of the following substituents, among others:
halogen, including fluorine, chlorine, bromine or iodine;
pseudohalides, including -CN;
[0110] -COOR where R is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted;
[0111] -COR where R is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted;
[0112] -CON(R)2 where each R, independently of each other R, is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted; and where R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
[0113] -OCON(R)2 where each R, independently of each other R, is a hydrogen or an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group all of which groups are optionally substituted; and where R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
[0114] -N(R)2 where each R, independently of each other R, is a hydrogen, or an alkyl group, or an acyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, phenyl or acetyl group, all of which are optionally substituted; and where R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
[0115] -SR, where R is hydrogen or an alkyl group or an aryl group and more specifically where R is hydrogen, methyl, ethyl, propyl, butyl, or a phenyl group, which are optionally substituted;
-S02R, or -SOR where R is an alkyl group or an aryl group and more specifically where R is a methyl, ethyl, propyl, butyl, or phenyl group, all of which are optionally substituted;
[0116] -OCOOR where R is an alkyl group or an aryl group;
[0117] -S02N(R)2 where each R, independently of each other R, is a hydrogen, or an alkyl group, or an aryl group all of which are optionally substituted and wherein R and R can form a ring which can contain one or more double bonds and can contain one or more additional carbon atoms;
[0118] -OR where R is H, an alkyl group, an aryl group, or an acyl group all of which are optionally substituted. In a particular example R can be an acyl yielding -OCOR" where R" is a hydrogen or an alkyl group or an aryl group and more specifically where R" is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted.
[0119] Specific substituted alkyl groups include haloalkyl groups, particularly trihalomethyl groups and specifically trifluoromethyl groups. Specific substituted aryl groups include mono-, di-, tri, tetra- and pentahalo-substituted phenyl groups; mono-, di-, tri-, tetra-, penta-, hexa-, and hepta- halo-substituted naphthalene groups; 3- or 4-halo-substituted phenyl groups, 3- or 4-alkyl- substituted phenyl groups, 3- or 4-alkoxy-substituted phenyl groups, 3- or 4-RCO-substituted phenyl, 5- or 6-halo-substituted naphthalene groups. More specifically, substituted aryl groups include acetylphenyl groups, particularly 4-acetylphenyl groups; fluorophenyl groups, particularly 3-fluorophenyl and 4-fluorophenyl groups; chlorophenyl groups, particularly 3-chlorophenyl and 4- chlorophenyl groups; methylphenyl groups, particularly 4-methylphenyl groups; and
methoxyphenyl groups, particularly 4-methoxyphenyl groups. [0120] As to any of the above groups which contain one or more substituents, it is understood that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. In addition, the compounds of this invention include all stereochemical isomers arising from the substitution of these compounds.
[0121] Pharmaceutically acceptable salts comprise pharmaceutically-acceptable anions and/or cations. As used herein, the term "pharmaceutically acceptable salt" can refer to acid addition salts or base addition salts of the compounds in the present disclosure. A pharmaceutically acceptable salt is any salt which retains at least a portion of the activity of the parent compound and does not impart significant deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered. Pharmaceutically acceptable salts include metal complexes and salts of both inorganic and organic acids. Pharmaceutically acceptable salts include metal salts such as aluminum, calcium, iron, magnesium, manganese and complex salts. Pharmaceutically acceptable salts include, but are not limited to, acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, -32-cilexetil, citric, edetic, edisylic, estolic, esyl, esylic, formic, fumaric, gluceptic, gluconic, glutamic, glycolic,
glycolylarsanilic, hexamic, hexylresorcjnoic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, lactobionic, maleic, malic, malonic, mandelic,
methanesulfonic, methylnitric, methylsulfuric, mucic, muconic, napsylic, nitric, oxalic, p- nitromethanesulfonic, pamoic, pantothenic, phosphoric, monohydrogen phosphoric, dihydrogen phosphoric, phthalic, polygalactouronic, propionic, salicylic, stearic, succinic, sulfamic, sulfanlic, sulfonic, sulfuric, tannic, tartaric, teoclic, toluenesulfonic, and the like. Pharmaceutically acceptable salts may be derived from amino acids, including but not limited to cysteine. Other pharmaceutically acceptable salts may be found, for example, in Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Wiley-VCH; Verlag Helvetica Chimica Acta, Zurich, 2002. (ISBN 3-906390-26-8). Pharmaceutically-acceptable cations include among others, alkali metal cations (e.g., Li+, Na+, K+), alkaline earth metal cations (e.g., Ca2+, Mg2+), non-toxic heavy metal cations and ammonium (NH4 +) and substituted ammonium (N(R')4 +, where R' is hydrogen, alkyl, or substituted alkyl, i.e., including, methyl, ethyl, or hydroxyethyl, specifically, trimethyl ammonium, triethyl ammonium, and triethanol ammonium cations). Pharmaceutically- acceptable anions include among other halides (e.g., CI", Br"), sulfate, acetates (e.g., acetate, trifluoroacetate), ascorbates, aspartates, benzoates, citrates, and lactate.
[0122] The compounds of this invention can contain one or more chiral centers. Accordingly, this invention is intended to include racemic mixtures, diasteromers, enantiomers, tautomers and mixtures enriched in one or more stereoisomer. The scope of the invention as described and claimed encompasses the racemic forms of the compounds as well as the individual enantiomers and non-racemic mixtures thereof. DETAILED DESCRIPTION
[0123] The following definitions and methods are provided to better define the invention and to guide those of ordinary skill in the art in the practice of the invention.
[0124] Referring to the drawings, like numerals indicate like elements and the same number appearing in more than one drawing refers to the same element. Unless otherwise noted, the terms and phrases used herein have their art-recognized meaning, which can be found by reference to standard texts, journal references and contexts known to those skilled in the relevant art.
[0125] The term "inflammation" generally refers to a biological response of tissues to harmful stimuli, such as pathogens, damaged cells, irritants, etc. Inflammation can be either acute or chronic. Acute inflammation is an initial response of the body to harmful stimuli and can be achieved by the increased movement of plasma and leukocytes from the blood into injured tissues. An inflammatory response can involve the local vascular system, the immune system, and/or various cells within the injured tissue. Prolonged inflammation, referred to as chronic inflammation, can lead to a progressive shift in the type of cells which are present at the site of inflammation can be characterized by simultaneous destruction and healing of the tissue from the inflammatory process.
[0126] The term "amino acid" comprises naturally occurring amino acids as well as non-naturally occurring amino acids, including amino acid analogs and derivatives. One skilled in the art will recognize that reference herein to an amino acid includes, for example, naturally occurring proteogenic L-amino acids; D-amino acids; chemically modified amino acids such as amino acid analogs and derivatives; naturally occurring non-proteogenic amino acids, and chemically synthesized compounds having properties known in the art to be characteristic of amino acids.
[0127] The term "nucleic acid" as used herein generally refers to a molecule or strand of DNA, RNA, or derivatives or analogs thereof including one or more nucleobases. Nucleobases comprise purine or pyrimidine bases typically found in DNA or RNA (e.g., adenine, guanine, thymine, cytosine, and/or uracil). The term "nucleic acid" also comprises oligonucleotides and polynucleotides. Nucleic acids may be single-stranded molecules, or they may be double-, triple- or quadruple-stranded molecules that may comprise one or more complementary strands of a particular molecule. "Nucleic acid" includes artificial nucleic acids including peptide nucleic acids, morpholino nucleic acids, glycol nucleic acids and threose nucleic acids. Artificial nucleic acids may be capable of nucleic acid hybridization.
[0128] As used herein, "sequence" means the linear order in which monomers occur in a polymer, the order of amino acids in a polypeptide or the order of nucleotides in a polynucleotide for example. [0129] The terms "peptide" and "polypeptide" are used synonymously in the present description, and refer to a class of compounds comprising of amino acid residues chemically bonded together by amide bonds (or peptide bonds), regardless of length, functionality, environment, or associated molecule(s). Peptides and polypeptides are polymeric compounds comprising at least two amino acid residues or modified amino acid residues. Modifications can be naturally occurring or non- naturally occurring, such as modifications generated by chemical synthesis. Modifications to amino acids in peptides include, but are not limited to, phosphorylation, glycosylation, lipidation, prenylation, sulfonation, hydroxylation, acetylation, methionine oxidation, alkylation, acylation, carbamylation, iodination and the addition of cofactors. Peptides include proteins and further include compositions generated by degradation of proteins, for example by proteolyic digestion. Peptides and polypeptides can be generated by substantially complete digestion or by partial digestion of proteins. Polypeptides comprising 2 to 100 amino acid units, optionally for some embodiments 2 to 50 amino acid units and, optionally for some embodiments 2 to 20 amino acid units can be used as polypeptide targeting ligands in the invention, for example, where the polypepetide preferentially binds to proteins, peptides or other biomolecules expressed, or otherwise generated by, a target tissue, such as a tumor, precancerous tissue, site of inflammation or other lesion. Typically, the polypeptide is at least four amino acid residues in length and can range up to a full-length protein.
[0130] "Protein" refers to a class of compounds comprising one or more polypeptide chains and/or modified polypeptide chains. Proteins can be modified by naturally occurring processes such as post-translational modifications or co-translational modifications. Exemplary post- translational modifications or co-translational modifications include, but are not limited to, phosphorylation, glycosylation, lipidation, prenylation, sulfonation, hydroxylation, acetylation, methionine oxidation, the addition of cofactors, proteolysis, and assembly of proteins into macromolecular complexes. Modification of proteins can also include non-naturally occurring derivatives, analogues and functional mimetics generated by chemical synthesis. Exemplary derivatives include chemical modifications such as alkylation, acylation, carbamylation, iodination or any modification that derivatizes the protein.
[0131] As used herein, "polynucleotide" and "oligonucleotide" are used interchangeably and refer to a class of compounds composed of nucleic acid residues chemically bonded together. The invention provides optical agents having an oligonucleotide or polynucleotide targeting ligand which comprises a plurality of nucleic acid residues, such as DNA or RNA residues, and/or modified nucleic acid residues that preferentially binds to proteins, peptides or other biomolecules expressed, or otherwise generated by, a target tissue, such as a tumor, precancerous tissue, site of inflammation or other lesion. Modifications to nucleic acid residues can be naturally occurring or non-naturally occurring, such as modifications generated by chemical synthesis. Oligo- or polynucleotide targeting ligands include, for example, oligo- or poly-nucleotides comprising 2 to 100 nucleic acid units, optionally for some embodiments 2 to 50 nucleic acid units and, optionally for some embodiments 2 to 20 nucleic acid units, and optionally for some embodiments 2 to 10 nucleic acid units. Polypeptide and oligonucleotide include a polymer of at least two nucleotides joined together by phosphodiester bonds and may consist of either ribonucleotides or
deoxyribonucleotides.
[0132] The term "aptamer" refers to an oligo- or poly-nucleotide or polypeptide that binds to, or otherwise selectively or preferentially associates with, a specific target molecule. For example, the invention provides optical agents having an aptamer targeting ligand that preferentially binds to proteins, peptides or other biomolecules expressed, or otherwise generated by, a target tissue, such as a tumor, precancerous tissue, site of inflammation or other lesion.
[0133] "Peptidomimetic" refers to a molecule having activity, including biological activity, that resembles that of a polypeptide or is substantially the same as a polypeptide. Morphine, for example, is a peptidomimetic of endorphin peptide. In some embodiments, a peptidomimetic is a small protein-like polymer designed to mimic the functionality of a peptide. Peptidomimetics useful as targeting ligands for some compounds of the invention in the present invention include peptoids and β-peptides. The composition and biological activity of peptidomimetics and use of
peptidomimetics in targeted diagnostics and therapeutics are further described in the following references: (1 ) A. Giannis and T. Kolter, Peptidomimetics for Receptor Ligands - Discovery, Development, and Medical Perspectives, Angewandte Chemie International Edition In English, vol. 32, 1993, pg. 1244-1267; (3) Peptidomimetics, Accounts of Chemical Research, Vol. 41 , No. 10, October 208, 1231 -1232, by Wu and Gellman; and (3) Patch, J.A. et al., Versatile oligo(N- substitutedjglycines: The many roles of peptoids in drug discovery., Pseudo-Peptides in Drug Discovery 2004, 1 -31 P.E. Nielsen.
[0134] As used herein, "attaching moiety" refers to a component provided to attach any of R1 , R2 or R3 directly or indirectly to central sulfenamide group in compounds of the invention. In some embodiments, L , L2, L3, W1, W2 and W3 in formulas (FX1 ) - (FX59) are an attaching moieties.
[0135] As used herein, an "electron withdrawing group" (abbreviated as "EWG") refers to a chemical group that draws electrons or electron density from a center, such a C5-C30 aryl or C5-C30 heteroaryl of the sulfenamide compounds of the invention. In some embodiments, the electron withdrawing group(s) are independently selected from cyano (-CN), carbonyl (-CO), carboxylate (-C02Ra), halo (-F, -CI, -Br, -I, -At), carbamate (-CONR Rc), acyl (-CORd), nitro (-N02), sulfinyl (-SORe), sulfonyl (-S02Rf, -S02OR9, and -POaR1^, wherein in the context of this description, Ra - R' are independently selected to enhance biological and/or physiochemical properties of the optical agents of the invention. In some instances, Ra - R1 are independently selected from any one of a hydrogen atom, an anionic functional group (e.g., carboxylate, sulfonate, sulfate, phosphonate or phosphate) and a hydrophilic functional group (e.g., hydroxyl, carboxyl, sulfonyl, sulfonato or phosphonato). In other instances, Ra - R1 are independently selected from hydrogen, d.10 alkyl, aryl, heteroaryl, -(CH2)nOH, -(CH2)nC02H, -(CH2)nS03H, -(CH2)nS03 ", -(CH2)nOS03H, -(CH2)NOSCV, -(CH2)NNHS03H , -(CH2)NNHS(V, -(CH2)NP03H2, -(CH2)NP03H- -(CH2)NP03 =, - (CH2)NOP03H2, -(CH2)NOP03H" and -(CH2)NOP03, wherein n is an integer from 1 to 1 0. In one example of this embodiment, the EWG(s) are independently selected from is -CN, halo, -C02RA - CORB, -N02, -S02R°, or -S02NRDRE, wherein each of RA - RE is independently H or d - C10 alkyl. In an embodiment, an EWG is located at the terminus of a substituent arm of a C5-C30 aryl or C5-C30 heteroaryl of the sulfenamide compounds of formulas (FX1 ) - (FX59).
[0136] As used herein, an "electron donating group" (abbreviated as "EDG") refers to a chemical group that releases electrons or electron density to a center, such as a C5-C30 aryl or C5-C30 heteroaryl of the sulfenamide compounds of the invention. In some embodiments, the electron donating group(s) are independently selected from C C10 alkyl, C5-C10 aryl, -(CH2)zOH, -ORJ, - SRK, -NR'R™, -N(RN)COR°, and -P(RP), wherein in the context of this description, RJ - RP are independently selected to enhance biological and/or physiochemical properties of the optical agents of the invention and wherein z is selected from the range of 1 to 1 0. In some instances, RJ - RP are independently selected from any one of a hydrogen atom, an anionic functional group (e.g., carboxylate, sulfonate, sulfate, phosphonate or phosphate) and a hydrophilic functional group (e.g., hydroxyl, carboxyl, sulfonyl, sulfonato or phosphonato). In other instances, R1 - R are independently selected from hydrogen, C- O alkyl, aryl, heteroaryl, -(CH2)NOH, -(CH2)2C02H , - (CH2)ZS03H, -(CH2)NS03 ~, -(CH2)ZOS03H, -(CH2)ZOS03 ~ -(CH2)ZNHS03H, -(CH2)ZNHS03 ~, - (CH2)ZP03H2, -(CH2)ZP03H_, -(CH2)ZP03 =, -(CH2)ZOP03H2, -(CH2)ZOP03H~ and -(CH2)ZOP03 = where z is an integer from 1 to 1 0. In one example of this embodiment, the EDG(s) are
independently C C6 alkyl, -ORF, -SR9, -NRHR', or -NRJCORK, wherein each of RF - RK is independently H or C-i - C-|0 alkyl. In an embodiment, an EDG is located at the terminus of a substituent arm of a C5-C30 aryl or C5-C30 heteroaryl of the sulfenamide compounds of formulas (FX1) - (FX59) of the invention.
[0137] In embodiments, two substituents, such as EDG and EWG substituents, on a compound of the invention can act in what is known as a "push-pull" arrangement. In embodiments of the "push-pull" arrangement, the electron density of the compound or a portion thereof, such as an aryl or heteroaryl group, is polarized due in part to the location of an EWG and EDG on the compound. In embodiments of the "push-pull' arrangement, an EWG is positioned at a terminus of a substituent arm of the structure and an EDG is positioned at a terminus of a different substituent arm of the structure. In embodiments of the "push-pull" arrangement, an EWG is positioned at one end of a π bond and an EDG is positioned at the other end of a π bond. In an embodiment, an EWG is positioned para- to an EDG in a six-membered ring structure. In an embodiment, an EWG is positioned trans- to an EDG in an alkylene structure. In some embodiments, compounds having the "push-pull" arrangement exhibit a shift in the optical absorbance and emission spectrum as compared to compounds not having the "push-pull" arrangement.
[0138] "Optical agent" generally refers to compounds, compositions, preparations, and/or formulations that absorb, emit, or scatter electromagnetic radiation of wavelength generally in the range of 350 - 1300 nanometers, within a biologically relevant environment or condition. In some embodiments, optical agents of the invention, when excited by electromagnetic radiation, undergo emission via fluorescence or phosphorescence pathways. These pathways are useful for diagnostic imaging, visualization, or organ function monitoring. Compounds belonging to this class are commonly referred to as "optical imaging agents" or "optical contrast agents." In some other embodiments, optical agents of the invention absorb electromagnetic radiation and undergo photochemical reactions such as photofragmentation of one or more photolabile bonds to generate reactive species such as nitrenes, carbene, free radicals, ions, excited species, etc. This process is useful for a wide range of phototherapy applications, for example in the treatment of tumors or other lesions. Compounds belonging to this class are commonly referred to as "photosensitizers." The term "photosensitizer" refers to a phototherapeutic agent or a component thereof providing for photoactivation, for example, photoactivation resulting in generation of reactive species that locally kill, injure, inactivate or otherwise degrade cells (e.g., cancer cells, tumor cells, non-cancer cells, etc.). Photosensitizers of some embodiments undergo photoactivation that initiates bond cleavage reactions, such as photolysis and/or nitrogen extrusion reactions, thereby generating reactive species capable of causing localized cell death or injury. Optical agents include Type 1 and Type 2 phototherapeutic agents. Optical agents include, but are not limited to, phototherapeutic agents (Type 1 and 2), photosensitizers, imaging agents, dyes, detectable agents, photosensitizer agents, photoactivators, and photoreactive agents; and conjugates, complexes, and derivatives thereof.
[0139] As used herein, a "chromophore" is a compound or functional group of a compound that results in absorption of electromagnetic radiation, preferably for some applications electromagnetic radiation having wavelengths in the UV (e.g. 200 nm to 350 nm) or visible (e.g. 350 nm to 750 nm) of the electromagnetic spectrum.
[0140] As used herein, a "fluorophore" is a compound or functional group of a compound that results in absorption of electromagnetic radiation and subsequent fluorescence. Preferably for some applications incorporation of a fluorophore results in compounds of the invention that absorb electromagnetic radiation and generate fluorescence having wavelengths in the UV (e.g. 200 nm to 350 nm) or visible (e.g. 350 nm to 750 nm) of the electromagnetic spectrum. In some embodiment, incorporation of a fluorophore results in compounds having an appreciable quantum yield for fluorescence, such as a quantum yield over the range of 0.001 to 1 , 0.01 to 1 , optionally 0.1 to 1 . Optical agents of the present invention can contain fluorophores. Fluorophores can be functional groups in a molecule which absorb electromagnetic radiation of first specific
wavelengths and re-emit energy at second specific wavelengths. The amount and wavelengths of the emitted electromagnetic radiation depend on both the fluorophore and the chemical environment of the fluorophore. The term "fluorophore" may be abbreviated throughout the present description as "FL". In aspects of the invention, fluorophores emit energy in the visible (e.g. 350 nm to 750 nm) and NIR regions (e.g., 750 - 1300nm) of the electromagnetic spectrum. [0141] As used herein, the term "luminescence" refers to the emission of electromagnetic radiation from excited electronic states of atoms or molecules. Luminescence generally refers to electromagnetic radiation emission, such as photoluminescence, chemiluminescence, and electrochemiluminescence, among others. In photoluminescence, including fluorescence and phosphorescence, the excited electronic state is created by the absorption of electromagnetic radiation. Luminescence detection involves detection of one or more properties of the
luminescence or associated luminescence process. These properties can include intensity, excitation and/or emission spectrum, polarization, lifetime, and energy transfer, among others. These properties can also include time-independent (steady-state) and/or time-dependent (time- resolved) properties of the luminescence. Representative luminescence techniques include fluorescence intensity (FLINT), fluorescence polarization (FP), fluorescence resonance energy transfer (FRET), fluorescence lifetime (FLT), total internal reflection fluorescence (TIRF), fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), and bioluminescence resonance energy transfer (BRET), among others. By way of example, when an optical agent is used in the present invention, it is desirable that the wavelength of radiation be non-ionizing and be such that it excites the optical agent. This excitation can cause a bond of the molecule to break and can lead to creation of one or more appropriate radical(s). This excitation can also cause the molecule to emit part of the absorbed energy at a different wavelength. Such emission can be detected using fluorometric techniques as described above. One skilled in the art can readily determine the most appropriate treatment and optional detection technique based, at least in part, on the specific phototherapeutic agent(s) administered and/or the particular use (e.g., tissue to be treated).
[0142] Optical condition" refers to one or more of the following: the fluorescence quantum yield, fluorescence intensity, fluorescence excitation wavelength, wavelength distribution or spectrum, emission wavelength, wavelength distribution or spectrum, Stokes shift, color, reflectance, phosphorescence, chemiluminescence, scattering, and/or other observable and/or measurable spectral property or phenomenon.
[0143] "Phototherapy procedure" refers to a therapeutic procedure involving administration of a phototherapeutic agent to a patient followed by subsequent excitation by exposure to applied electromagnetic radiation, such as electromagnetic radiation having wavelengths in the visible and/or near IR region of the electromagnetic spectrum. Such wavelengths can be in the range of 350 - 1300 nanometers, so as to generate a therapeutically effective amount of excited phototherapeutic agent. Phototherapy includes, but is not limited to, photodynamic therapy. As used herein, "phototherapy" includes procedures involving administration of Type 1 and/or Type 2 phototherapeutic agents, optionally further including administration of one or more additional therapeutic agents. [0144] A detectable optical signal may be, for example, an observable change in absorbance, reflectance, phosphorescence, chemiluminescence, scattering, or other spectral property.
[0145] As used herein, "tumor-specific agent" refers to a compound or composition, such as an optical agent, that preferentially accumulates in a tumor at a higher level than normal tissue regardless of the particular mechanism of uptake in the tumors, for example, receptor mediated or enhanced permeability and retention (EPR). Optical agents of the invention include tumor-specific agents, including tumor specific phototherapy agents, for example having a targeting ligand providing specificity in the administration, delivery and/or binding to tumor tissue.
[0146] As used herein, "targeting ligand" (abbreviated as Bm) refers to a chemical group and/or substituent having functionality for targeting a compound of any one of formula (FX1 ) - (FX59) to an anatomical and/or physiological site of a patient, such as a selected cell, tissue or organ. For some embodiments, a targeting ligand is characterized as a ligand that selectively or preferentially binds to a specific biological site(s) (e.g., enzymes, receptors, etc.) and/or biological surface(s) (e.g., membranes, fibrous networks, etc.). In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is an amino acid, or a polypeptide comprising 2 to 30 amino acid units. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a mono- or polysaccharide comprising 1 to 50 carbohydrate units. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a mono-, oligo- or poly-nucleotide comprising 1 to 50 nucleic acid units. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a protein, an enzyme, a carbohydrate, a peptidomimetic, a glycomimetic, a glycopeptide, a glycoprotein, a lipid, an antibody (polyclonal or monoclonal), or fragment thereof. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is an aptamer. In an embodiment, the invention provides compounds having any one of formula (FX1 ) - (FX59), wherein Bm is a drug, a hormone, steroid or a receptor. In some embodiments, each occurrence of Bm in the compounds of (FX1 ) - (FX59) is
independently a monoclonal antibody, a polyclonal antibody, a metal complex, an albumin, or an inclusion compound such as a cyclodextrin. In some embodiments, each occurrence of Bm in the compounds of (FX1 ) - (FX59) is independently integrin, selectin, vascular endothelial growth factor, fibrin, tissue plasminogen, thrombin, LDL, HDL, Sialyl LewisX or a mimic thereof, or an atherosclerotic plaque binding molecule. Throughout the present description, the term
"biomolecule" can be a targeting ligand (Bm).
[0147] In the compounds of any one of formulas (FX1) - (FX59), Bm is a targeting ligand, optionally providing molecular recognition functionality. In some embodiments, the targeting ligand is a particular region of the compound that is recognized by, and binds to, a target site on an organ, tissue, tumor or cell. Targeting ligands are often, but not always, associated with biomolecules or fragments thereof which include, but are not limited to, hormones, amino acids, peptides, peptidomimetics, proteins, nucleosides, nucleotides, nucleic acids, enzymes, carbohydrates, glycomimetics, lipids, albumins, mono- and polyclonal antibodies, receptors, inclusion compounds such as cyclodextrins, and receptor binding molecules. Targeting ligands for use in the invention can also include synthetic polymers. Examples of synthetic polymers that are useful for targeting ligands include polyaminoacids, polyols, polyamines, polyacids,
oligonucleotides, aborols, dendrimers, and aptamers. Still other examples of useful targeting ligands can include integrin, selectin, vascular endothelial growth factor, fibrin, tissue plasminogen activator, thrombin, LDL, HDL, Sialyl LewisX and its mimics, and atherosclerotic plaque binding molecules.
[0148] Specific examples of targeting ligands include, but are not limited to: steroid hormones for the treatment of breast and prostate lesions; whole or fragmented somatostatin, bombesin, and neurotensin receptor binding molecules for the treatment of neuroendocrine tumors; whole or fragmented cholecystekinin receptor binding molecules for the treatment of lung cancer; whole or fragmented heat sensitive bacterioendotoxin (ST) receptor and carcinoembryonic antigen (CEA) binding molecules for the treatment of colorectal cancer; dihydroxyindolecarboxylic acid and other melanin producing biosynthetic intermediates for the treatment of melanoma; whole or fragmented integrin receptor and atherosclerotic plaque binding molecules for the treatment of vascular diseases; and whole or fragmented amyloid plaque binding molecules for the treatment of brain lesions. In some embodiments, Bm, if present, is selected from heat-sensitive bacterioendotoxin receptor binding peptide, carcinoembryonic antigen antibody (anti-CEA), bombesin receptor binding peptide, neurotensin receptor binding peptide, cholecystekinin receptor binding peptide, somastatin receptor binding peptide, ST receptor binding peptide, neurotensin receptor binding peptide, leukemia binding peptides, folate receptor binding agents, steroid receptor binding peptide, carbohydrate receptor binding peptide or estrogen. In another embodiment Bm, if present, is a ST enterotoxin or fragment thereof. In some embodiments, Bm, if present, is selected from octreotide and octreotate peptides. In another embodiment Bm, if present, is a synthetic polymer. Examples of synthetic polymers useful for some applications include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers. Examples of specific peptide targeting ligands are described in WO/2008/108941.
[0149] "Target tissue" refers to tissue of a subject to which an optical agent is administered or otherwise contacted, for example during a biomedical procedure such as an optical imaging, phototherapy, monitoring or visualization procedure. Target tissues can be contacted with an optical agent of the invention under in vivo conditions in vitro conditions or ex vivo conditions. Target tissues in some embodiments include cancerous tissue, cancer cells, precancerous tissue, a tumor, a lesion, a site of inflammation, or vasculature tissue. In some embodiments, a target tissue includes a melanoma cell, a breast lesion, a prostate lesion, a lung cancer cell, a colorectal cancer cell, an atherosclerotic plaque, a brain lesion, a blood vessel lesion, a lung lesion, a heart lesion, a throat lesion, an ear lesion, a rectal lesion, a bladder lesion, a stomach lesion, an intestinal lesion, an esophagus lesion, a liver lesion, a pancreatic lesion, and a solid tumor. Target tissue in some embodiments refers to a selected organ of the subject or component thereof, such as lung, heart, brain, stomach, liver, kidneys, gallbladder, pancreas, intestines, rectum, skin, colon, prostate, ovaries, breast, bladder, blood vessel, throat, ear, or esophagus.
[0150] Methods of this invention comprise the step of administering an "effective amount" of the present diagnostic and therapeutic compositions, formulations and preparations containing the present compounds or compositions, to diagnose, image, monitor, evaluate, treat, reduce, alleviate, ameliorate or regulate a biological condition and/or disease state in a patient. The term "effective amount," as used herein, refers to the amount of the diagnostic and therapeutic formulation, that, when administered to the individual is effective to diagnose, image, monitor, evaluate, treat, reduce alleviate, ameliorate or regulate a biological condition and/or disease state. As is understood in the art, an effective amount of a given composition or formulation will depend at least in part upon the mode of administration (e.g. intravenous, oral, topical administration), any carrier or vehicle employed, and the specific individual to whom the formulation is to be
administered (age, weight, condition, sex, etc.). The dosage requirements needed to achieve the "effective amount" vary with the particular formulations employed, the route of administration, and clinical objectives. Based on the results obtained in standard pharmacological test procedures, projected daily dosages of active compound or composition can be determined as is understood in the art.
[0151] In an embodiment, an effective amount of a compound or composition of the invention is a therapeutically effective amount. As used herein, the phrase "therapeutically effective" qualifies the amount of compound or composition administered in the therapy. This amount achieves the goal of ameliorating, suppressing, eradicating, preventing, reducing the risk of, or delaying the onset of a targeted condition. In an embodiment, an effective amount of a compound or composition of the invention is a diagnostically effective amount. As used herein, the phrase "diagnostically effective" qualifies the amount of compound or composition administered in diagnosis, for example of a disease state or other pathological condition. The amount achieves the goal of being detectable while avoiding adverse side effects found with higher doses. In an embodiment, an active ingredient or other component is included in a therapeutically acceptable amount. In an embodiment, an active ingredient or other component is included in a diagnostically acceptable amount.
[0152] It is contemplated that the compounds and pharmaceutically acceptable salts of the invention can be used as part of a combination. The term "combination" means the administration of two or more compounds directed to a target condition. The treatments of the combination generally can be co-administered in a simultaneous manner. Two compounds can be coadministered as, for example: (a) a single formulation (e.g., a single capsule) having a fixed ratio of active ingredients; or (b) multiple, separate formulations (e.g., multiple capsules) for each compound. The treatments of the combination can alternatively (or additionally) be administered at different times.
[0153] In certain embodiments, the invention encompasses administering optical agents useful in the invention to a patient or subject. A "patient" or "subject", used equivalently herein, refers to an animal. In particular, an animal refers to a mammal, preferably a human. The subject can either: (1 ) have a condition able to be monitored, diagnosed, prevented and/or treated by administration of an optical agent of the invention; or (2) is susceptible to a condition that is able to be monitored, diagnosed, prevented and/or treated by administering an optical agent of the invention.
[0154] When used herein, the terms "diagnosis", "diagnostic" and other root word derivatives are as understood in the art and are further intended to include a general monitoring, characterizing and/or identifying a state of health or disease. The term is meant to encompass the concept of prognosis. For example, the diagnosis of cancer can include an initial determination and/or one or more subsequent assessments regardless of the outcome of a previous finding. The term does not necessarily imply a defined level of certainty regarding the prediction of a particular status or outcome.
[0155] As defined herein, "administering" means that a compound or formulation thereof of the invention, such as an optical agent, is provided to a patient or subject, for example in a
therapeutically effective amount. The invention includes methods for a biomedical procedure wherein a therapeutically or diagnostically effective amount of a compound having any one of formulas (FX1) - (FX59) is administered to a patient in need of treatment, for example to a patient undergoing treatment for a diagnosed diseased state including cancer and vascular diseases. Administering can be carried out by a range of techniques known in the art including parenteral administration including intravenous, intraperitoneal or subcutaneous injection or infusion, oral administration, topical or transdermal absorption through the skin, or by inhalation, for example. The chosen route of administration may depend on such factors as solubility of the compound or composition, location of targeted condition, and other factors which are within the knowledge of one having ordinary skill in the relevant art.
[0156] "Topical administration" includes the use of transdermal administration, such as transdermal patches or iontophoresis devices.
[0157] "Parenteral administration" includes subcutaneous injections, intravenous injections, intraarterial injections, intraorbital injections, intracapsular injections, intraspinal injections, intraperitoneal injections, intramuscular injections, intrasternal injections, and infusion. Dosage forms suitable for parenteral administration include solutions, suspensions, dispersions, emulsions, and any other dosage form that can be administered parenterally.
[0158] As used herein, the term "controlled-release component" refers to an agent that facilitates the controlled-release of a compound including, but not limited to, polymers, polymer matrices, gels, permeable membranes, liposomes, microspheres, or the like, or any combination thereof. Methods for producing compounds in combination with controlled-release components are known to those of skill in the art.
[0159] As used herein, the term "pharmaceutically acceptable" means approved by a regulatory agency of an appropriate federal or state government; or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans; or does not impart significant deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered.
[0160] As will be clear to those of ordinary skill in the art, the groups and structures described herein as portions of the compounds of the invention may be defined as if they are separate valence-satisfied chemical structures. It is intended that when a group is described or shown as being a substituent of another group, that the group be viewed as having a valency to allow this binding to occur.
[0161] The invention is further detailed in the following Examples, which are offered by way of illustration and are not intended to limit the scope of the invention in any manner.
EXAMPLE 1 : SULFENAMIDE COMPOUNDS FOR PHOTOTHERAPY
1.a. Type 1 Phototherapeutic Agents
[0162] The invention provides Type 1 phototherapeutic agents, including compositions, preparations and formulations, and methods of using and making Type 1 phototherapeutic agents. Type 1 phototherapeutic agents of the invention include compounds comprising one or more photolabile, acyclic S-N bond directly or indirectly linked to a C5-C30 aryl comprising one or more aromatic and/or heterocyclic aromatic groups. Incorporation of a C5-C30 aryl comprising aromatic and/or heterocyclic aromatic groups in some compounds provides a chromophore moiety capable of absorption of electromagnetic radiation, preferably for some applications electromagnetic radiation having wavelengths in the visible (e.g. 350 nm to 750 nm) and NIR regions (e.g., 750 - 1300nm) of the electromagnetic spectrum. The C5-C30 aryl group(s) of some compositions of the invention functions as an aromatic antenna group for coupling energy from incident
electromagnetic radiation into the phototherapeutic agent. In some phototherapeutic agents of the present invention, energy coupled into the phototherapeutic agent is subsequently transferred to the surroundings to achieve a desired therapeutic outcome. Incorporation of an aromatic antenna group comprising a C5-C30 aryl having one or more aromatic and/or heterocyclic aromatic groups is useful in some embodiments for initiating cleavage of a photolabile, acyclic S-N bond upon absorption of electromagnetic radiation and subsequent internal energy transfer processes.
Cleavage of the photolabile S-N bond generates one or more reactive species capable of causing localized tissue damage, such as cell death, inactivation or injury. [0163] Some compounds of the invention operate through the Type 1 phototherapy mechanism as schematically illustrated in Figures 1 A - 1 F wherein the photosensitizer is activated upon exposure to electromagnetic radiation, thereby producing reactive species. Figures 1 A and 1 B provide schematic representations of reaction mechanisms for phototherapeutic agents having formula (FX1 ), and Figures 1 C and 1 D provide schematic representations of reaction mechanisms for phototherapeutic agents having formula (FX16). As schematically represented by the arrow and hv in Figures 1 A-1 F, compounds of the present invention are photoactivated by exposure to visible or near infrared electromagnetic radiation, for example electromagnetic radiation having wavelengths ranging from 350 nm to 1 300 nm. Absorption of at least a portion of the applied electromagnetic radiation generates a therapeutically effective amount of photoactivated phototherapeutic agent, which is schematically represented in figures 1 A-1 D by the compound provided in brackets with an asterisk symbol (*). Activation of the phototherapeutic agent may occur via a single photon absorption process, a mulitphoton absorption process or a combination of a single photon absorption process and a mulitphoton absorption process. The activated photosensitizer subsequently undergoes processes, such as internal energy transfer and/or bond cleavage processes, resulting in formation of reactive species capable of causing a desired therapeutic result. Reactive species generated by the compounds of the invention may include free radicals, intramolecular diradicals, ions, electrons, electrophiles, nitrene, vibrationally excited species, and translationally excited species. As illustrated in Figures 1 B and 1 D, excitation of the photosensitizer in these embodiments causes cleavage of the S-N bond, thereby generating two free radical species. In the mechanism shown in Figures 1 B and 1 D, excitation of a C5-C30 aryl results in rapid intramolecular energy transfer to the photolabile S-N bond resulting in bond cleavage and radical formation. The coupling between the C5-C3o aryl and the central S-N bond may be selected to enhance the intramolecular energy transfer process(es) and provide efficient photoinitated bond cleavage. In some embodiments, for example, the central S-N group is directly coupled to one or more rings of an aromatic group or heterocyclic aromatic group of a C5-C30 aryl. In some embodiments, the reactive species generated upon excitation of the photosensitizer collide, react with, or otherwise interact with cell components of a target organ or tissue class, thereby resulting in death, injury and/or damage to cells at the target tissue.
[0164] Figures 1 E and 1 F provide schematics showing mechanisms for generation of reactive species for a class of photosensitizers having a S-N bond with a fused ring configuration, wherein the nitrogen atom of the S-N bond is a part of the fuse ring structure. In the mechanism shown in Figures 1 E and 1 F, excitation results in formation of a stable free radical having a fused ring structure and a stable thiyl radical . The free radical having a fused ring structure may further undergo electron transfer to generate a cation and anion reactive species. In the scheme shown in Figure 1 E, for example, photoactivation results in formation of a moderately stable cation and a stable anion. In the scheme shown in Figure 1 F, for example, photoactivation results in formation of a very stable azatropylium cation and a stable anion. [0165] Type 1 phototherapeutic agents useful for certain phototherapy applications incorporate one or more C5-C30 aryl or C5-C30 heteroaryl groups, including aromatic groups, heterocyclic aromatic groups, polycyclic aromatic groups and polycyclic heterocyclic aromatic groups, that absorb strongly in the visible and/or NIR region of the electromagnetic spectrum. C5-C30 aryl or C5-C30 heteroaryl groups providing effective photoactivation by electromagnetic radiation having wavelengths selected over the range of 300 nm to 1300 nm include, but are not limited to, groups corresponding to azulenes, aza-azulenes, anthracenes, pyrazines, pyridazines, quinolines, quinoxalines, courmarins, phenoxazines, phenothiazines, rhodamines, and the like. The invention further includes phototherapeutic agents having one or more C5-C30 aryl or C5-C30 heteroaryl groups comprising aromatic group(s) and heterocyclic aromatic group(s) that are functionalized by incorporation of heteroatom ring members and substituents on the ring structure(s) providing excitation wavelength selection and/or tunability. In some embodiments, for example, the C5-C30 aryl or C5-C30 heteroaryl comprises one or more aromatic or heterocyclic aromatic groups independently having one or more electron donating and/or electron withdrawing groups provided as ring substituents for providing selected excitation characteristics, such as a selected absorption spectrum and/or strong absorption in the visible and/or NIR regions. Some phototherapeutic agents of the present invention operate, at least in part, via the Type 2 process involving formation of excited state oxygen (102), and optionally contain a C5-C30 aryl or C5-C30 heteroaryl that is a group corresponding to a cyanine, indocyanine, phenothiazine, or phthalocyanine.
[0166] Selection of R1 - R3, R1 - R33, R40 - R55, R58 - R63, R65 - R67, R70 - R85, R 20 - R127 and Ar1 - Ar3 in the compounds of any one of formulas (FX1) - (FX59) establishes, at least in part, the physical, chemical, optical and/or pharmacokinetic properties of optical agents for the present compositions and methods. In some embodiments, for example R1 - R3, R1 - R33, R40 - R55, R58
- R63, R65 - R67, R70 - R85, R120 - R127 and Ar1 - Ar3 are selected to provide optical properties supporting and enabling use of these compositions in phototherapeutic methods, such as providing one or more of the following: (i) large extinction coefficients; (ii) strong absorption in the visible and/or infrared regions of the electromagnetic spectrum (e.g., 350 to 1300 nanometers, preferably for some applications 350-900 nanometers); and (iii) a large quantum yield for the production of reactive species, such as free radicals or ions, capable of causing photoactivation initiated tissue damage. Selection of the composition of R1 - R3, R11 - R33, R40 - R55, R58 - R63,
R65 _
Figure imgf000068_0001
co m po u n d s Qf a ny Q ne Qf formulas (FX1) -
(FX59) may also be based, at least in part, on a number of pharmacokinetic and physical properties supporting effective delivery and clearance of the optical agents of the present methods and compositions. Such factors may include solubility, toxicity, immune response,
biocompatibility, and bioclearance considerations. In some embodiments, any one of R - R3, R11
- R33, R40 - R55, R58 - R63, R65 - R67, R70 - R85, R120 - R127 and Ar1 - Ar3 in the compounds of any one of formulas (FX1) - (FX59) comprise a hydrophilic group, a lipophilic group, hydrophobic group, or an amphiphilic group. In an embodiment, at least one of R1 - R3, R11 - R33, R40 - R55, R58 _ Re3_ R65 _ R67^ R7o _ R85> Ri2o _ R127 a nd Afi _ Af3 js g substjtuent comprjsing Poly(ethylene glycol) (PEG, -(CH2OCH2)b-), or a derivative of PEG.
[0167] In an embodiment, a phototherapeutic agent of the invention incorporates aromatic groups and/or heterocyclic aromatic groups that are derivatized by the addition of at least one electron withdrawing group and at least one electron donating group bonded directly or indirectly to a carbon atom of the ring structure. In an embodiment, for example, one or more the electron withdrawing (EWG) and electron donating (EDG) group(s) are directly attached to the ring structure of the aromatic group. In another embodiment, EWG and EDG are indirectly attached to the to the ring structure of the aromatic group through an unsaturated spacer that is in conjugation with the double bonds of a C5-C30 aryl or C5-C3o heteroaryl group. Electron donating and withdrawing groups in these dye compositions may be positioned ortho, meta or para to each other with respect to the to the ring structure of the aromatic group. In some embodiments, for example, two electron withdrawing groups are positioned para to each other on the ring structure of the aromatic group and two electron donating groups are positioned para to each other on the ring structure of the aromatic group. In some embodiments, electron withdrawing groups and electron donating groups are positioned so as to make the overall compound symmetrical.
[0168] Optical agents of the invention support a broad therapeutic platform useful for a variety of in vivo phototherapy procedures, for example for the treatment of cancer, stenosis, inflammation, infection and arthritis. Optical agents of the invention are optionally multifunctional agents capable of providing a useful combination of photodiagnostic, phototherapeutic, molecular recognition and/or targeting functionality. In an embodiment, for example, a dye component is incorporated into the phototherapeutic agent of the present compositions for imparting useful optical functionality, for example by functioning as an optical absorber, chromophore, and/or fluorophore. This functionality is useful for targeted administration and excitation of the therapeutic agent. Optionally, optical agents of the invention further comprise a targeting component, such as a targeting ligand. In an embodiment, for example, an optical agent of the invention comprises a targeting ligand integrated with a photosensitizer component to access enhanced administration, delivery and photoactivation functionality for phototherapy therapy. Optical agents and
bioconjugates thereof are provided having one or more targeting ligands covalently bonded to or noncovalently associated with the phototherapeutic agents of the present invention, thereby providing specificity for administering, targeting, delivery and/or localizing an optical agent to a specific biological environment, such as a target tissue such as a specific organ, tissue, cell type or tumor site.
[0169] In the compounds of any one of formulas (FX1) - (FX59), Bm is a targeting ligand, optionally providing molecular recognition functionality. In some embodiments, the targeting ligand is a particular region of the compound that is recognized by, and binds to, the target site on the organ, tissue, tumor or cell. Targeting ligands are often, but not always, associated with biomolecules or fragments thereof which include hormones, amino acids, peptides,
peptidomimetics, proteins, nucleosides, nucleotides, nucleic acids, enzymes, carbohydrates, glycomimetics, lipids, albumins, mono- and polyclonal antibodies, receptors, inclusion compounds such as cyclodextrins, and receptor binding molecules. Specific examples of biomolecules include steroid hormones for the treatment of breast and prostate lesions; somatostatin receptor binding molecules, bombesin receptor binding molecules, and neurotensin receptor binding molecules for the treatment of neuroendocrine tumors, cholecystekinin receptor binding molecules for the treatment of lung cancer; heat sensitive bacterioendotoxin (ST) receptor binding molecules and carcinoembryonic antigen (CEA) binding molecules for the treatment of colorectal cancer, dihydroxyindolecarboxylic acid and other melanin producing biosynthetic intermediates for melanoma, integrin receptor and atheroscleratic plaque binding molecules for the treatment of vascular diseases, amyloid plaque binding molecules for the treatment of brain lesions, cholecystokinin (CCK) receptor binding molecules, steroid receptor binding molecules,
carbohydrate receptor binding molecules, dihydroxyindole-2-carboxylic acid, and combinations thereof. Targeting ligands for use in the invention may also include synthetic polymers. Examples of synthetic polymers include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers. Still other examples of appropriate targeting ligands may include integrin, selectin, vascular endothelial growth factor, fibrin, tissue plasminogen activator, thrombin, LDL, HDL, Sialyl LewisX and its mimics, and atherosclerotic plaque binding molecules.
[0170] Successful specific targeting of fluorescent dyes to tumors using antibodies and peptides for diagnostic imaging of tumors has been demonstrated, for example, S. A. Achilefu et al., Novel receptor-targeted fluorescent contrast agents for in vivo tumor imaging, Investigative Radiology, 2000, 35(8), 479-485; B. Ballou et al., Tumor labeling in vivo using cyanine-conjugated monoclonal antibodies, Cancer Immunology and Immunotherapy, 1995,41 ,247-263; K. Licha et al., New contrast agent for optical imaging: acid-cleavable conjugates ofcyanine dyes with biomolecules, In Biomedical Imaging: Reporters, Dyes, and Instrumentation, D. J. Bomhop, C. Contag, and E. M. Sevick-Muraca (Eds.), Proceedings of SPIE, 1999, 3600, 29-35, each of which are expressly incorporated by reference herein in their entirety. Therefore, the inventive receptor-targeted phototherapeutic agents are expected to be effective in the treatment of various lesions.
[0171] In one example, a targeting ligand may contain all or part of a steroid hormone or a steroid receptor binding compound, and therefore target steroid hormone sensitive receptors. In this example, the compound is administered, targets the desired site such as breast and/or prostate lesion, is photoactivated, and forms free radicals at this site thereby effecting cell injury, damage, or death at the desired target site. Similar target binding molecules and uses will be recognized by one skilled in the art. For example, the targeting group may be a compound that targets and binds to a somatostatin, bombesin, CCK, and/or neurotensin receptor binding molecule, or may be a carcinogenic embryonic antigen-binding compound that binds to a carcinogenic embryonic antigen. These are then photoactivated for radical formation at, for example, lung cancer cells with CCK receptor binding molecules, colorectal cancer cells with ST receptor and carcinoembryonic antigen (CEA) binding molecules, melanoma cells with dihyroxyindolecarboxylic acid, vascular sites of atherosclerotic plaque with integrin receptor binding molecules, brain lesions with amyloid plaque binding molecules, and the like.
[0172] The optical agents of this example may contain additional functionalities that can be used to attach various types of biomolecules, synthetic polymers, and organized aggregates for selective delivery to various organs or tissues of interest. Examples of synthetic polymers include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers. The invention includes, but is not limited to, phototherapeutic agents comprising a photosensitizer - biomolecule conjugate which provide advantages over nonspecific
phototherapeutic agents or the conjugation of photosensitizers to very large biomolecules. These conjugates provide enhanced localization and rapid visualization of tumors which is beneficial for both diagnosis and therapy. The agents are rapidly cleared from blood and non-target tissues so there is less concern for accumulation and for toxicity. A variety of high purity compounds may be synthesized for combinatorial screening of new targets, e.g., to identify receptors or targeting agents, and for the ability to affect the pharmacokinetics of the conjugates by minor structural changes.
[0173] In some embodiments, a liposome or micelle may be utilized as a carrier or vehicle for the composition. For example, in some embodiments, a phototherapeutic agent comprises a sulfenamide photosensitizer that may be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, may be on the external surface of the liposome or micelle. As another example, a targeting ligand may be extemally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains the sulfenamide phototherapeutic
agent/photosensitizer) to the desired tissue, organ, or other site in the body.
1.b: Synthesis of Phototherapeutic Agents.
[0174] Methods for the synthesis of sulfenamide compounds are known in the art. For example, sulfenamide compounds may be prepared via reaction of a sulfenyl halide, such as a sulfenyl chloride, with an amine. Properties and methods of synthesizing and derivatizing sulfenamide compounds are disclosed in (1) "Adventures in Sulfur - Nitrogen Chemistry, F. Davis, J. Org. Chem 2006, 71 , 8993-9003; (2) Davis et. al., Org. Prep. Proc. Int., 1979, 1 1 , 33; (3), T. Greene. Protection of Amino Group. In Protective Group in Organic Synthesis. John Wiley & Sons: New York, 1981 , pp. 283-284, and (4) F. A. Davis and U.K. Nadir, Org. prep. Proc.lnt, 11 , 33 (1979). Properties and Methods of synthesizing sulfenyl halides compounds are disclosed in: (1) S. Thea ; G. Cevasco, "A Mild and Convenient Preparation of Sulfenyl Chlorides from Thiolacetates," Tetrahedron letters, vol. 29, No. 23, pp. 2865-2866, 1988, (2) S. Thea ; G. Cevasco, Tetrahedron letters, vol. 28, pp. 5193, 1987, (3) E. Kuhle, "The Chemistry of Sulfenic Acids," Georg Thieme Publishers, Stuttgart, 1973, and (4) Y.C. Aggarwal, S. Warren, Tetrahedron Lett., 27, 101 (1986), and (5) E.M. Gordon, M.A. Ondetti, J. Pluscec, CM. Cimarusti, D.P. Bonner, R.B. Sykes, J. Amer. Chem. Soc. 104, 6053, 1982. Synthetic production and physical property data for sulfenamide compounds containing a carbazole moiety are disclosed in: (1) Blanca et al., "The Chiral S-N Axis in Sulfenamides: Enantiomeric Resolution, Direction Demonstration of Optical Activity, and Kinetics of Interconversion", Angew. Chem. Int. Ed. Engl. , 1997, 36, No. 20; and (2) Miura et al., "EPR studies of nitrogen-centered free radicals. Part 50. Unusual decomposition behavior of isolable stable thioaminyl free radicals at high temperature", J. Chem. Soc. Perkin Trans 2, 1998, 1 185-1 188.
[0175] In an approach for making some of the present sulfenamide compound, for example, solution of the desired amine (e.g., 2.0 mmol) and triethylamine (e.g., 4.0 mmol) in anhydrous methylene chloride (e.g., 10 mL) is treated with 4-nitrobenzenesulfenyl chloride (e.g., 2.2 mmol) in anhydrous methylene chloride (e.g., 5 mL). The mixture is stirred at ambient temperature for 2-16 hours. Thereafter, the reaction mixture is poured onto water and the organic phase is separated, washed with water, dried over sodium sulfate, and evaporated in vacuo. The crude product is purified by either crystallization or flash chromatography to yield the desired sulfenamide compound.
[0176] Figure 2 provides a general scheme for the syntheses of a sulfenamide optical agent having an acyclic S-N bond useful in phototherapeutic methods of the invention. As shown in Figure 2 a sulfenyl chloride is reacted with an amine in the presence of a base to form a sulfenamide compound. R -R3 and W1- W3 are as described in connection with Formula (FX1). In an embodiment, at least one of R1 -R3 is a C5— Cso aryl or C5-C30 heteroaryl, optionally a C5— C2o aryl or C5-C2o heteroaryl, or C5 - C10 aryl or C5-C 0 heteroaryl.
[0177] Coupling a targeting ligand (Bm) to an sulfenamide compound may be accomplished by methods known in the art, for example, as disclosed in Hermanson, Bioconjugate Techniques, Academic Press, New York, 1996; Hnatowich et al., Radioactive Labeling of Antibody: A simple and efficient method, Science, 1983, 220, 613; Pelegrin et al., Photoimmunodiagnosis with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies, J. Cellular Pharm., 3 (1992) 141 ; and U.S. Patent No. 5,714,342. Typical procedures for the preparation of a sulfenamide - peptide conjugate having formula are described involving reaction of an amino or carboxylated acyclic sulfenamide with a peptide targeting ligand. Amino or carboxylated acyclic sulfenamides may be coupled to a targeting ligand such as a peptide by any of the standard peptide coupling methods (e.g., mixed anhydride or active ester coupling), or can be employed directly during the automated peptide synthesis procedure. The coupling of biomolecules such as somatostatin, bombesin, cholecystekinin, ST, steroids, and the like to acyclic sulfenamides can be achieved by the use of succinimido active esters. For example, a sulfenamide compound containing a carboxyl group is activated by making a mixed anhydride in situ with isobutylchloroformate, and then reacted with any biomolecule bearing an amino group. Alternatively, the carboxyl group can be esterified with N-hydroxysuccinimide, and reacted with the amino group to form the amide. The carboxyl group containing tetrazolodisulfide can also be used directly in automated peptide synthesis procedure. [0178] Figure 3A provides general schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond and peptide targeting ligand useful in phototherapeutic methods of the invention. As shown in Figure 3A, a sulfenyl chloride is reacted with an amine in the presence of a base to form a sulfenamide compound which is subsequently coupled to a peptide via formation of a peptide bond. R1 -R3 and W1- W3 are as described in connection with Formula (FX1), and wherein Peptide is a polypeptide comprising 2 to 30 amino acid units. In an embodiment, at least one of R1 -R3 is a C5 - C30 aryl or C5-C30 heteroaryl, optionally a C5 - C20 aryl or C5-C20 heteroaryl, or C5 - C-io aryl or C5-C 0 heteroaryl. Figure 3B provides schemes for the preparation of sulfenamide optical agents having an acyclic S-N bond, peptide targeting ligand and phenyl group. In some embodiments, for example, X is -CR65R66- wherein R65 and R66 are hydrogen or C C2o alkyl, C3-C20 cycloalkyl, C5-C20 aryl, or C5-C20 heteroaryl, optionally a C1-C10 alkyl, C3-C10 cycloalkyl, C5-C10 aryl, or C5-C 0 heteroaryl. In some embodiments, for example, each of R1 - R3, R65 and R66 are individually an electron donating group, an electron withdrawing group or a targeting ligand (Bm).
[0179] Figure 4A and 4B provide schemes for the general syntheses and conjugation of photosensitizers having an acyclic S-N bond useful as phototherapeutic agents of the present invention. L1 - L3, W1 - W3, and R1 - R3 are as described in connection with Formula (FX1), and wherein Peptide is a polypeptide comprising 2 to 30 amino acid units. In an embodiment, for example, a solution of the desired amine (2.0 mmol) and triethylamine (4.0 mmol) in anhydrous methylene chloride (10 mL) is treated with 4-nitrobenzenesulfenyl chloride (2.2 mmol) in anhydrous methylene chloride (5 mL), and the mixture is stirred at ambient temperature for 2-16 hours. Thereafter, the reaction mixture is poured onto water and the organic phase was separated, washed with water, dried over sodium sulfate, and evaporated in vacuo. The crude product is purified by either crystallization or flash chromatography.
[0180] Figures 10 and 12 provide general schemes for the syntheses of bioconjugates of sulfenamide optical agents having formulas (FX1) and (FX49). As shown in Figures 10 and 12, sulfenamide compounds are provided having pendant carboxyi or amine groups. Subsequent reaction in the presence of a coupling agent: (1) links the amino-terminus of a peptide and a carboxyi group of the sulfenamide compound in the presence of a coupling agent, or (2) links the carboxyl-terminus of a peptide and an amide group of the sulfenamide compound. Accordingly, the reaction schemes in Figures 10 and 12 illustrate conjugation of a peptide targeting ligand to the sulfenamide compound via an amide linkage. As will be generally understood by persons having skill in the art coupling agents useful in the reactions of schemes of Figures 10 and 12 include hydrogen peroxide, chlorine, bromine, iodine, peracids, periodate, hypochlorite, and the like.
[0181] Figures 11A, 1 1 B, 13A and 13B illustrate examples of coupling reactions useful for synthesis of sulfenamide optical agents of the invention having specific targeting ligands. As shown in Figure 11 A, the sulfenamide compound (1) in Figure 10 having a pendant carboxyi group is linked to octreotide, bombesin, cholecystokinin, bacterioenterotoxin and steroid targeting ligands via formation of an amide bond. As shown in Figure 1 1 B, the sulfenamide compound (2) in Figure 10 having a pendant amine group is linked to octreotide, bombesin, cholecystokinin,
bacterioenterotoxin and steroid targeting ligands via formation of an amide bond. As shown in Figure 13A, the sulfenamide compound (3) in Figure 12 having a pendant carboxyl group is linked to octreotide, bombesin, cholecystokinin, bacterioenterotoxin and steroid targeting ligands via formation of an amide bond. As shown in Figure 13B, the sulfenamide compound (4) in Figure 12 having a pendant amine group is linked to octreotide, bombesin, cholecystokinin,
bacterioenterotoxin and steroid targeting ligands via formation of an amide bond. As will be understood by a person having skill in the art, a variety of coupling agents are useful for linking the sulfenamide backbone to the targeting ligands including dicyclohexylcarbodiimide (DCC), 1-ethyl- 3-(3-dimethylaminopropyl) carbodiimide (EDC), benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), disuccinimdyl carbonate, N-hydroxysuccinimide, methylformamide, isobutylchloroformate, etc.
[0182] As will be understood by one of skill in the art, the synthetic approaches shown in Figures 2, 3A, 3B, 4A, 4B, 10, 1 1A, 1 1 B, 12, 13A and 13B are applicable to synthesis of other sulfenamide optical agents of the invention, including sulfenamide optical agents having formulas (FX1) - (FX59).
1 b.1 : Preparation of a Compound of Formula (FX18)
[0183] A procedure for the preparation of the sulfenamide compound having formula (FX 8) is described involving reactio -nitrobenznesulfenyl chloride with diphenylamine.
Figure imgf000074_0001
As will be understood by those of skill in the art, the same procedure, or a reasonable modification thereof, may be applied for the preparation of other sulfenamide optical agents of the present invention. A solution of diphenylamine (0.95 g, 5.0 mmol) and triethylamine (0.61 g, 5.5 mmol) in anhydrous methylene chloride (5 ml_) is treated with p-nitrobenznesulfenyl chloride (5.0 mmol) in anhydrous methylene chloride (5 ml_), and the mixture is stirred at ambient temperature for 16 hours. Thereafter, the reaction mixture is poured onto water and the organic phase is separated, washed with water, dried over sodium sulfate, and evaporated in vacuo. Flash chromatography over silica gel (gradient elution 0 to 20% ethyl acetate/hexanes) affords 1.6 grams of compound (FX18) as brown gum. LRMS, m/Z 323.4 (M+H).
1 b.2: Preparation of 9-(4-nitrophenyl)thiocarbazole (FX47).
Figure imgf000075_0001
[0184] A mixture of carbazole (250 mg) and triethylamine (0.46 ml_) in dry chloroform (20 ml_) was cooled to 0 °C and treated with 4-nitrobenzenesulfenly chloride (320 mg) in 10 mL of chloroform added dropwise. After the addition, the reaction mixture was stirred at ambient temperature for 22 hours, and treated with chloroform (20 mL) and water (30 mL). The organic layer was separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo. The crude material was purified by silica gel column chromatography to give the compound (FX47). 1H-NMR (CDCI3) δ ppm: 8.11 (d, 2H), 8.04 (d, 2H); 7.62 (d, 2H), 7.48 (m, 2H); 7.38 (m, 2H); 6.94(d, 2H); 13C-NMR (CDCI3) δ ppm: 147.7, 146.1 , 142.6, 126.9, 124.9, 124.6, 122.4, 122.1 , 120.6, 1 10.7. LRMS, m/Z 321 .4 (M+H).
1 b.3: Preparation of 9-(2,4-dinitropheyl)thiocarbazole (FX48).
Figure imgf000075_0002
[0185] A mixture of carbazole (250 mg) and triethylamine (0.46 mL) in dry chloroform (20 mL) was cooled to 0 °C and treated with 2,4-dinitrobenzenesulfenyl chloride (320 mg) in 10 mL of chloroform added dropwise. After the addition, the reaction mixture was stirred at ambient temperature for 22 hours, and treated with chloroform (20 mL) and water (30 mL). The organic layer was separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo. The crude material was purified by silica gel column chromatography to give the compound (FX48). H-NMR
(CDCI3) δ ppm: 9.19 (d, 1 H), 8.13 (d, 2H); 8.05 (dd, 1 H), 7.58 (d, 2H); 7.48 (m, 2H); 7.40 (m, 2H); 6.47 (d, 1 H); 13C-NMR (CDCI3) δ ppm: 149.2, 145.6, 142.7, 141.7, 128.2, 127.2, 125.5, 125.1 , 122.6, 121.6, 120.8, 1 10.7. LRMS, m/Z 366.4 (M+H).
1 b.4: Preparation of 1-f(4-nitropheyl)thio1-4,5-dehydrodibenzfb,flazepine (FX49).
Figure imgf000076_0001
A mixture of dibenz[b,f]azepine (480 mg) and triethyiamine (720 mg) in dry methylene chloride (10 ml_) was treated with 4-nitrobenzenesulfenly chloride (520 mg) and the entire mixture was stirred at ambient temperature for 2 hours. The reaction mixture was treated with methylene chloride (20 mL) and water (30 ml_). The organic layer was separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo to give reddish-orange solid. Recrystallization from ethyl acetate afforded pure compounds (FX49) as canary yellow needles. H-NMR (CDCI3) δ ppm: 8.15 (d, 2H), 7.58 (d, 2H); 7.56 (d, 2H); 7.48 (m, 2H); 7.27 (m, 4H); 6.92 (s, 2H); 13C-NMR (CDCI3) δ ppm: 149.7, 145.4, 132.1 , 129.9, 129.2, 126.4, 125.9, 125.3, 124.5, 124.1 , 121.8. LRMS, m/Z 347.4 (M+H).
1 b.5. Preparation of 1-r(4-nitro heyl)thioldibenzfb,f1azepine (FX50).
Figure imgf000076_0002
[0186] A mixture of 4,5-dihdrodibenz[b,f]azepine (390 mg) and triethyiamine (220 mg) in dry methylene chloride (10 mL) was treated with 4-nitrobenzenesulfenly chloride (400 mg) and the entire mixture was stirred at ambient temperature for 2 hours. The reaction mixture was treated with methylene chloride (20 mL) and water (30 mL). The organic layer was separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo. The crude material was purified by flash chromatography using chloroform-methanol gradient to give compound (FX50) as a reddish-orange solid. 1H-NMR (CDCI3) δ ppm: 8.05 (d, 2H), 7.25 (m, 2H); 7.12 (m, 2H); 7.10 (m, 2H); 6.86 (m, 2H); 6.79 (m, 2H), 3.08 (m, 4H); 13C-NMR (CDCI3) δ ppm: 148.7, 143.0, 142.3, 139.5, 136.4, 132.3, 128.7, 127.6, 127.4, 125.2, 123.6, 122.0, 1 18.7, 117.5, 116.5, 1 15.6, 33.2, 32.8. LRMS, m/Z 349.4 (M+H).
1 b.6. Preparation of H(4-nitropheyl)thioldibenzorb,f)f1 ,41oxazepine (FX51).
Figure imgf000077_0001
[0187] A mixture of dibenzo[b,f]-1 ,4-oxazepine (e.g., 2.0 mmol) and triethylamine (e.g., 4.0 mmol) in dry methylene chloride (e.g., 10 mL) is treated with 4-nitrobenzenesulfenly chloride (e.g., 2.2 mmol) and the entire mixture is stirred at ambient temperature for 2 hours. The reaction mixture is treated with methylene chloride (e.g., 20 mL) and water (e.g., 30 mL). The organic layer is separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo. The crude material is purified by flash chromatography using chloroform-methanol gradien to give compound (FX51).
1 b.7. Preparation of 1-f(4-nitropheyl)thio1dibenzorb,f1f1 ,4lthiazepine (FX52).
Figure imgf000077_0002
[0188] A mixture of dibenzo[b,f]-1 ,4-thiazepine (e.g, 2.0 mmol) and triethylamine (e.g, 4.0 mmol) in dry methylene chloride (e.g, 10 mL) is treated with 4-nitrobenzenesulfenly chloride (e.g, 2.2 mmol) and the entire mixture is stirred at ambient temperature for 2 hours. The reaction mixture is treated with methylene chloride (e.g, 20 mL) and water (e.g, 30 mL). The organic layer is separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo. The crude material is purified by flash chromatography using chloroform-methanol gradient to give compound (FX52).
1 b.8. Preparation of 1-|'(4-nitropheyl)thiol-4-methyldibenzorb,fin ,41diazepine(FX53).
Figure imgf000078_0001
[0189] A mixture of 4-methyldibenzo[b,f]-1 ,4-diazepine (e.g., 2.0 mmol) and triethylamine (e.g., 4.0 mmol) in dry methylene chloride (e.g., 10 mL) is treated with 4-nitrobenzenesulfenly chloride (e.g., 2.2 mmol) and the entire mixture is stirred at ambient temperature for 2 hours. The reaction mixture is treated with methylene chloride (e.g., 20 mL) and water (e.g., 30 mL). The organic layer is separated, washed with water, dried over sodium sulfate, filtered, and the filtrate evaporated in vacuo. The crude material is purified by flash chromatography using chloroform-methano! gradient to give compound (FX53).
1 b.9. Preparation of Sulfenamide-Peptide Conjugate (FX19) - Automated Procedure
CONH-Peptide
Figure imgf000078_0002
wherein Peptide is a polypeptide comprising 2 to 30 amino acid units, and optionally wherein Peptide is bombesin.
[0190] A typical procedure for the preparation of peptide-sulfenamide conjugate (FX19) using an automated peptide synthesizer is described. It should be noted that other sulfenamide compounds may be conjugated to a peptide targeting ligand, such as bombesin by the same or similar procedure. The peptide-sulfenamide conjugate having formula (FX19) is prepared by
fluorenylmethoxycarbonyl (Fmoc) solid phase peptide synthesis strategy with a commercial peptide synthesizer from Applied Biosystems (Model 432A SYNERGY Peptide Synthesizer). The first peptide cartridge contains Wang resin pre-loaded with an amide resin on 25-pmole scale. The amino acid cartridges are placed on the peptide synthesizer and the product is synthesized from the C- to the N-terminal position. Coupling of the Fmoc-protected amino acids (75 pmol) to the resin-bound free terminal amine (25 Mmol) is carried out with 2-(1 H-benzotriazol-1-yl)-1 ,1 ,3,3- tetramethyluronium hexafluorophosphate (HBTU, 75 Mmol)/N-hydroxybenzotriazole (HOBt, 75 μπηοΙ). Each Fmoc protecting group on solid support is removed with 20% piperidine in dimethylformamide before the subsequent amino acid is coupled to it. The last cartridge contains the sulfenamide carboxylic acid derivative having formula (FX18), which is coupled to the peptide automatically, thus avoiding the need for post-synthetic manipulations. [0191] After the synthesis is completed, the product is cleaved from the solid support with a cleavage mixture containing trifluoroacetic acid (85%):water (5%):phenol (5%):thioanisole (5%) for 6 hours. The peptide-sulfenamide conjugate is precipitated with t-butyl methyl ether and lyophilized in watenacetonitrile (2:3) mixture. The peptide-sulfenamide conjugate is purified by HPLC and analyzed with LC/MS.
[0192] A procedure similar to that described above may be used for preparation of peptide bioconjugate having formula (FX54):
Figure imgf000079_0001
(FX54); wherein Peptide is a polypeptide comprising 2 to 30 amino acid units, and optionally wherein Peptide is bombesin; and wherein X is a single bond, - CRRCRR-, -CR=CR- -CRRO-, -CRRS-, -CRRNR-, or -CR=N-i
1 b.10: Preparation of Peptide-Sulfenamide Conjugate (FX19) - Solution Phase Procedure
[0193] A typical procedure for the preparation of Peptide-sulfenamide conjugate having formula (FX19) using mixed anhydride coupling procedure is described. It should be noted that other sulfenamide compounds may be conjugated to a peptide targeting ligand, such as bombesin by the same or similar procedure. A solution of the sulfenamide compound having formula (FX18) (120 μιηοΙ) and triethylamine (250 μηηοΙ) in anhydrous dimethylformamide (DMF) (500 μΙ_) is stirred and cooled to 0°C -10°C. Thereafter isobutylchloroformate (120 μιηηΊθΙ) is added and the mixture is stirred at 0-10 °C for about 30 minutes. A polypeitde, such as Bombesin (7-14) peptide, (100 pmol) in DMF is added to the above mixture and allowed to come to ambient temperature (about 20°C to about 22°C) and stirred at ambient temperature for about one hour. After the synthesis is completed, the product is cleaved from the solid support with a cleavage mixture containing trifluoroacetic acid (85%):water (5%):phenol (5%):thioanisole (5%) for 6 hours. The peptide-sulfenamide conjugate is precipitated with t-butyl methyl ether and lyophilized in watenacetonitrile (2:3) mixture. The peptide - sulfenamide conjugate is purified by reversed phase HPLC and analyzed with LC/MS. These methods are useful for preparation of an sulfenamide-bombesin (7-14) bioconjugate having the formula [FX18]-R2-Gln-Trp-Ala-Val-Gly-His- Leu-Met-NH2 or [FX18]-R3-Gln-Trp-Ala-Val-Gly-His-Leu-Met-NH2.
A procedure similar to that described above may be used for preparation of peptide bioconjugate having formula (FX54).
1 ,b.1 1 : Phototherapeutic Methods and Cell Viability Measurements
[0194] A general procedure is carried out for measuring cell viability upon exposure of leukemia cells to a number of sulfenamide photosensitizers of the invention and light. The sulfenamide photosensitizers evaluated include (FX18), (FX47), (FX48), (FX49) and (FX50). The absorption maximum for each sulfenamide compound evaluated is provided in Table 1. The cell viability analysis is carried out using a human myeloid leukemia U937 cell line by the standard WST-1 assay. In this procedure, U397 Leukemia cells (0.5 x 106) are plated in standard T-25 cell culture flasks, and are exposed to four controls and a series of test conditions corresponding to a range of sulfenamide photosensitizer concentrations.
Table 1.b.1 : Absorption Maxima for Sulfenamide Photosensitizers
Figure imgf000080_0001
Table 1.b.2: Control and Test Conditions for Cell Viability Measurements
Figure imgf000080_0002
[0195] The sulfenamide photosensitizer is dissolved in DMSO at an initial concentration of about 8 mM and diluted with water to the final desired value such that amount of DMSO exposed to the cells is always kept below 0.5%. The cells are incubated at 37 C with various concentrations the photosensitizer for about 30 minutes prior to the exposure of light. The cells are irradiated for 5, 10 and/or 20 minute durations that radiates light with maximum output in the range of 325-425 nm with constant cooling such that the temperature at the surface of the microliter plates does not exceed 37 °C. The experiments also include control conditions wherein the cells are irradiated for 5, 10 and/or 20 minute durations in the absence of photosensitizer also with constant cooling. The light source is a 200-W B-100SP High Intensity Lamp from UVP. It is noted that in the present study, the light source is not optimized with respect to radiant energy, power and wavelength.
[0196] The viability of cells is assessed after 24 hours following light exposure. Once exposure is complete, cells are processed to determine percent viability using Hank's Balanced Salt Solution (HBSS), Trypan blue stain, and a hemacytometer to count live and dead cells. The number of viable cells is determined and percent viability is determined: Percent Viability = No. of Viable Cells Counted x 100
Total No. of Cells Counted
Viability measurements are analyzed to provide VC50/2o values which is defined as the
concentration at which 50% decrease in cell viability is observed when the cells are exposed to light and the photosensitizer for 20 minutes.
[0197] Figure 5 provides cell viability results for control conditions (Control 4, no photosensitzer, DMSO, light) wherein the cells are exposed to light and dimethyl sulfoxide. As shown in Figure 4, DMSO toxicity is observed only at the highest concentrations of DMSO. Cells are incubated without DMSO (0 μΜ) and with DMSO at concentrations of 3 mM, 6 mM, and 12 mM. For the results in Figure 4, the cells were exposed to light from a B-100SP High Intensity Lamp for 0, 5, and 20 minutes. As shown in figure 4, 100% of the cells are viable with conditions of 0 mM DMSO and 3 mM DMSO for light exposure for 5 minutes and 20 minutes. As shown in figure 4, for conditions of 6 mM DMSO and no light exposure, light exposure for 5 minutes, and light exposure for 20 minutes, at least 94% of the cells are viable. As shown in figure 4, for conditions of 12 mM DMSO and no light exposure, and for light exposure for 5 minutes, at least 95% of cells were viable; and for light exposure for 20 minutes, at least 78% of cells are viable.
[0198] Figure 6 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX18) for a range of photosensitizer concentrations and exposure times. The cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability. As shown in Figure 6, the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light over the range of 5 minutes to 20 minutes. For the results shown in Figure 6, cells are incubated in the absence of sulfenamide photosensitizer (0 μΜ) and with sulfenamide photosensitizer at concentrations of 1.5 μΜ , 3 μΜ, and 6 μΜ. The cells are exposed to light from a B-100SP High Intensity Lamp for 0, 5, 10, and 20 minutes. The results indicate that in the absence of sulfenamide photosensitizer, the cells at all light exposure times have 100% viability. For conditions of 1.5 μΜ sulfenamide photosensitizer, cells have 100% viability with light exposure for 5 minutes, about 87% viability with light exposure for 10 minutes, and 65% viability with light exposure for 20 minutes. For conditions of 3.0 μΜ sulfenamide photosensitizer, cells have about 77% viability with light exposure for 5 minutes, about 46% viability with light exposure for 10 minutes, and about 15% viability with light exposure for 20 minutes. For conditions of 6.0 μΜ sulfenamide photosensitizer, cells have about 19% viability with light exposure for 5 minutes, about 12% viability with light exposure for 10 minutes, and about 7% viability with light exposure for 20 minutes. The cell viability results yield a VC50/2o value of 0.2 ± 0.1 μΜ for the sulfenamide photosensitizer having Formula (FX18).
[0199] Figure 7 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX49) for a range of concentrations and exposure times. The cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability. As shown in Figure 7, the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes. Cells are incubated in the absence of sulfenamide photosensitizer (0 μΜ) and with sulfenamide photosensitizer at concentrations of 0.63 μΜ , 1.25 μΜ, and 2.5 μΜ. The cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes. The results indicate that in the absence of sulfenamide photosensitzer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes. For conditions of 0.63 μΜ sulfenamide photosensitizer, cells have about 80% viability for conditions of no light exposure and about 90% viability for light exposure for 20 minutes. For conditions of 1.25 μΜ sulfenamide photosensitizer, cells have about 78% viability for conditions of no light exposure and about 62% viability for light exposure for 20 minutes. For conditions of 2.5 μΜ sulfenamide photosensitizer, cells have about 78% viability for conditions of no light exposure and about 20% viability for light exposure for 20 minutes. The cell viability results yield a VC50/2o value of 1.8 μΜ for the sulfenamide photosensitizer having Formula (FX49).
[0200] Figure 8 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX47) for a range of concentrations and exposure times. The cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability. As shown in Figure 8, the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes. Cells are incubated in the absence of sulfenamide photosensitizer (0 μΜ) and with sulfenamide photosensitizer at concentrations of 1.25 μΜ , 2.5 μΜ, and 5 μΜ. The cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes. The results indicate that in the absence of sulfenamide photosensitzer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes. For conditions of 1.25 μΜ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 85% viability for light exposure for 20 minutes. For conditions of 2.5 μΜ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 55% viability for light exposure for 20 minutes. For conditions of 5 μΜ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 22% viability for light exposure for 20 minutes. The cell viability results yield a VC50/2o value of 3.2 μΜ for the sulfenamide
photosensitizer having Formula (FX47).
[0201] Figure 9 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX48) for a range of concentrations and exposure times. The cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability. As shown in Figure 9, the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes. Cells are incubated in the absence of sulfenamide photosensitizer (0 μΜ) and with sulfenamide photosensitizer at concentrations of 1.25 μΜ , 2.5 μΜ, and 5 μΜ. The cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes. The results indicate that in the absence of sulfenamide photosensitzer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes. For conditions of 1.25 μΜ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 75% viability for light exposure for 20 minutes. For conditions of 2.5 μΜ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 42% viability for light exposure for 20 minutes. For conditions of 5 μΜ sulfenamide photosensitizer, cells have greater than 100% viability for conditions of no light exposure and about 25% viability for light exposure for 20 minutes. The cell viability results yield a VC50/2o value of 2.2 μΜ for the sulfenamide
photosensitizer having Formula (FX48).
[0202] Figure 20 provides cell viability results for test conditions wherein cells are exposed to light and the sulfenamide photosensitizer having formula (FX50) for a range of concentrations and exposure times. The cell viability experiments having the sulfenamide photosensitizer present exhibit concentration-dependent and light exposure time-dependent decreases in cell viability. As shown in Figure 20, the viability of cells in contact with a sulfenamide photosensitizer decreases dramatically upon exposure to light for an exposure time of 20 minutes. Cells are incubated in the absence of sulfenamide photosensitizer (0 μΜ) and with sulfenamide photosensitizer at concentrations of 0.63 μΜ , 1.3 μΜ, and 2.5 μΜ. The cells are exposed to light from a B-100SP High Intensity Lamp for 20 minutes. The results indicate that in the absence of sulfenamide photosensitizer, the cells have 100% viability for conditions of no light exposure and an exposure time of 20 minutes. For conditions of 0.63 μΜ sulfenamide photosensitizer, cells have about 90% viability for conditions of no light exposure and about 100% viability for light exposure for 20 minutes. For conditions of 1.3 μΜ sulfenamide photosensitizer, cells have about 94% viability for conditions of no light exposure and about 92% viability for light exposure for 20 minutes. For conditions of 2.5 μΜ sulfenamide photosensitizer, cells have about 90% viability for conditions of no light exposure and about 52% viability for light exposure for 20 minutes. The cell viability results yield a VC50/2o value of 2.5 μΜ for the sulfenamide photosensitizer having Formula (FX50).
1.b.12: Phototherapeutic Methods and Electron Spin Resonance Measurements
[0203] Electron Spin Resonance (ESR) spectrometry measurements are carried out to probe generation of reactive species upon optical excitation of sulfenamide compounds of the invention. ESR measurements provide information relating to the mechanism of photactivation of the present compounds including the identity and abundance of radical species produced upon exposure to light. For example, strong ESR signals are observed that are indicative of stable sulfur-centered radicals and less stable nitrogen-centered radicals. The ESR measurements of this example demonstrate that sulfenamide photosensitizers of the invention are effective for localized generation of reactive species, including free radicals, and generally support the applicability of the present compounds as optical agents for use in phototherapy. [0204] For the present measurements, stock solutions of 20 mM to 50 mM of a sulfenamide compound in benzene and about 100 mM DMPO (5,5-dimethylpyrroline-N-oxide) in benzene are prepared. A mixture of 150 microliters of the sulfenamide compound and 50 microliters of DMPO is purged with nitrogen for 5 minutes and irradiated with light inside an ESR spectrometer for 1-3 minutes. ESR spectra are obtained as a function of magnetic field strength for the following conditions: (1) before irradiation with light (Before), (2) during irradiation with light (Light On), and (3) after irradiation with light (Light Off). The total ESR signal strength is also measured as a function of time (Decay).
[0205] Figure 14 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX18). Trace (a) of Figure 14 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX18) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 14 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX18) is exposed to light and indicates the production of radical species during irradiation. Trace (c) of Figure 14 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX18) is exposed to light and indicates the presence of radical species during this time. Trace (d) of Figure 14 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX18) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
[0206] Figure 15 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX47). Trace (a) of Figure 15 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 15 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of radical species during irradiation. Trace (c) of Figure 15 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX47) is exposed to light and indicates the presence of radical species during this time. Trace (d) of Figure 15 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX47) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
[0207] Figure 16 provides ESR spectra for a second set of experiments evaluating the sulfenamide compound of formula (FX47). Trace (a) of Figure 16 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 16 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX47) is exposed to light and indicates the production of radical species during this time. Trace (c) of Figure 16 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX47) is exposed to light and indicates the presence of radical species during irradiation. Trace (d) of Figure 16 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX47) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
[0208] Figure 17 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX48). Trace (a) of Figure 17 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX48) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 17 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX48) is exposed to light and indicates the production of radical species during irradiation. Trace (c) of Figure 17 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX48) is exposed to light and indicates the presence of radical species during this time. Trace (d) of Figure 17 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX48) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
[0209] Figure 18 provides ESR spectra for a second set of experiments evaluating the sulfenamide compound of formula (FX48). Trace (a) of Figure 18 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX48) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 18 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula' (FX48) is exposed to light and indicates the production of radical species during irradiation. Trace (c) of Figure 18 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX48) is exposed to light and indicates the presence of radical species during this time.
[0210] Figure 19 provides ESR spectra for experiments evaluating the sulfenamide compound of formula (FX49). Trace (a) of Figure 19 provides the ESR signal intensity as a function of magnetic field strength for conditions before the sulfenamide compound of formula (FX49) is exposed to light and indicates the production of little to no radical species during this time. Trace (b) of Figure 19 provides the ESR signal intensity as a function of magnetic field strength for conditions while the sulfenamide compound of formula (FX49) is exposed to light and indicates the production of radical species during irradiation. Trace (c) of Figure 19 provides the ESR signal intensity as a function of magnetic field strength for conditions after the sulfenamide compound of formula (FX49) is exposed to light and indicates the presence of radical species during this time. Trace (d) of Figure 19 provides the ESR signal intensity as a function of time before, during, and after the sulfenamide compound of formula (FX49) is exposed to light and indicates the generation of radical species during irradiation with light and the presence of radical species after irradiation with light.
[0211] As shown in the ESR spectra, compounds having formulas (FX47) and (FX48) produce strong radicals, both sulfur- and nitrogen-centered. The strong quartet observed in the ESR spectra represents sulfur-centered radicals and these appear to be more stable than the nitrogen- centered ones. Also, the radicals produced by the compound of formula (FX48) appear to be longer lived than those produced by the compound of formula (FX47).
EXAMPLE 2: PHOTOTHERAPY METHODS
[0212] Phototherapy, such as photodynamic therapy (PDT), typically employs a combination of a photosensitizer (PS) and visible or near infrared light to generate reactive species that kill or otherwise degrade target cells, such as tumors or other lesions. The present invention provides phototherapeutic agents useful for phototherapy. In an embodiment, methods the invention are not surgical methods.
[0213] The invention includes phototherapy methods wherein a phototherapeutic agent comprising a compound of any one of the formulas (FX1) - (FX59) is administered to a patient, for example, wherein a therapeutically effective amount of such a component is administered to a patient in need of treatment. In some embodiments, compounds of the invention provide an optical agent capable of selective targeting and delivery to a target tissue such as a tumor, site of inflammation or other lesion. Upon administration, the phototherapeutic agent is optionally allowed to accumulate in a target region of interest (e.g., target tissue, tumor, or organ). To induce selective tissue damage, the phototherapeutic agent is activated by exposure to electromagnetic radiation. In an embodiment, the phototherapeutic agent is activated after an effective
concentration of the phototherapeutic agent has accumulated in a target tissue. An effective concentration of a compound of the invention depends on the nature of the formulation, method of delivery, target tissue, activation method and toxicity to the surrounding normal non-target tissue. Exposure to electromagnetic radiation and activation of the phototherapeutic agent may occur during or after administration of the phototherapeutic agent and accumulation at the target tissue.
[0214] For photoactivation, the target region is illuminated with electromagnetic radiation having a wavelength in the range of about 350 nm to about 1300 nm, preferably for some applications in the range of about 400 nm to about 900 nm. In some embodiments, the wavelength of the electromagnetic radiation corresponds to a peak in the absorption spectrum of the phototherapeutic agent, for example is within 20 nanometers of a peak in the absorption spectrum of the phototherapeutic agent in the visible or NIR regions. In some phototherapeutic procedures the target site is exposed to electromagnetic radiation having sufficient fluence and/or power sufficient to activate the phototherapeutic agent so as to induce cell death, for example via necrosis or apoptosis processes. In some embodiments, electromagnetic radiation having low energy, power or fluence is provided to activate the phototherapeutic agent without undesirable thermal effects. If the region of interest is, for example, a lesion or tumor on the skin surface, the region can be directly illuminated. Otherwise, endoscopic and/or endoluminal catheters equipped with an electromagnetic radiation source may be employed to provide a photodiagnostic and/or phototherapeutic effect.
[0215] Appropriate power and intensity of the electromagnetic radiation depends on the size, depth, and the pathology of the lesion, as is known to one skilled in the art. In an embodiment, the fluence of the electromagentic radiation is preferably, but not always, kept below 200 mW/cm2 , optionally below 100 mW/cm2, to minimize undesirable thermal effects. The intensity, power, and duration of the illumination, and the wavelength of the electromagnetic radiation may vary widely depending on the body location, the lesion site, the effect to be achieved, etc. In an embodiment, the power of the applied electromagnetic radiation is preferably is selected over the range of 1 - 500 mW/cm2 and optionally for some applications selected over the range of 1 - 200 mW/cm2 cm2 and optionally for some applications selected over the range of 1 - 100 mW/cm . In an embodiment, the duration of the exposure to applied electromagentic radiation selected over the range of 1 second to 60 minutes, and optionally for some applications selected over the range of 1 second to 30 minutes, and optionally for some applications selected over the range of 1 second to 10 minutes, and optionally for some applications selected over the range of 1 second to 1 minute.
[0216] In an embodiment, the invention provides a method of using a phototherapeutic agent, the method comprising: (i) administering a therapeutically effective amount of a phototherapeutic t comprising a compound being of the formula (FX1):
Figure imgf000087_0001
(FX1); or a pharmaceutically acceptable salt or ester thereof, wherein:
each of L1, L2, and L3, if present, is independently C C10 alkylene, C3-C 0 cycloalkylene, C2-C10 alkenylene, C3-C10 cycloalkenylene, C2-C10 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, -(CH2CH20)b-, or -(CHOH)a-;
each of W1 , W2, and W3 is independently a single bond, -(CH2)n- -(HCCH)n- -0-, -S-, -SO-, -S02- -SO3-, -OS02- -NR11-, -CO-, -COO-, -OCO-, -OCOO-, -CONR12-, - NR13CO- -OCONR14-, -NR15COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n- -S(CH2)n- - NR"u(CH2)n- -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n-, -OCOO(CH2)n-, -CONR^(CH2)n- - CONR22(CH2)n- -NR23CO(CH2)n-, -OCONR24(CH2)n-, -NR25COO(CH2)n-, -NR26C0NR27(CH2)n- , -NR28CSNR29(CH2)n- -O(CH2)nNR30CO(CH2)n-, - CO(CH2yCH2OCH2)n(CH2)nNR3 CH2)nNR32CO- or -CO(CH2)nNR33CO-;
each of R , R2, and R3 is independently a hydrogen, -OCF3, CTC20 alkyi, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
or wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form one to five substituted or unsubstituted, carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings;
wherein at least one of R1, R2, and R3 is a C5-C30 aryl or C5-C30 heteroaryl, or wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form a C5-C30 aryl or C5-C30 heteroaryl;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of e, f, and g is independently 0 or 1 ;
each of R11 - R33 is independently hydrogen, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C20 aryl;
each of R40-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid;
each FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steriod, a hormone, an aptamer, a receptor, a metal chelating agent, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units; and (ii) exposing the phototherapeutic agent administered to the patient to electromagnetic radiation. In an embodiment, the phototherapeutic agent is exposed to a therapeutically effective amount of electromagnetic radiation. As used herein, a therapeutically effective amount of electromagentic radiation is an amount for achieving a desired therapeutic result, for example an amount for generating a therapeutically effective amount of reactive species for damaging or causing cell death of a selected target tissue. In an embodiment, the method further comprises generating one or more reactive species from said compound administered to the patient via the exposure of the phototherapeutic agent to applied electromagnetic radiation. In an embodiment, for example, the method further comprises the step of cleaving a photolabile S-N bond so as to generate reactive species, such as free radicals and/or ions. In an embodiment, the method further comprises contacting a selected organ or selected tissue in the patient with the phototherapeutic agent. In an embodiment, a therapeutically effective dose of the phototherapeutic agent is administered to a patient in need of treatment.
[0217] Phototherapeutic agents useful in the present methods include sulfenamide compounds containing at least one S-N bond directly or indirectly coupled to a C5-C3o aryl or C5-C30 heteroaryl comprising one or more aromatic and/or heterocyclic aromatic groups. Phototherapeutic agents useful in the present methods include compounds optionally having a ligand for targeted administration. Phototherapeutic agents useful in the present methods include compounds optionally having a dye component, such as a fluorophore or chromophore, for imaging and/or visualization functionality. In an embodiment, the method of the invention comprises administering to a patient a compound having any one of formula selected from (FX1) - (FX59), including any of the specific compositions classes and compounds described in connection with formula (FX1) - (FX59). As will be understood by one of skill in the art, the present methods expressly include methods of using phototherapeutic agents wherein the phototherapeutic agent includes the compound classes, compounds, and all variations thereof, described herein, including the compound classes, compounds and variations described in connection with any one of formulas (FX1) - (FX59).
[0218] Embodiments of this aspect may comprise a method of carrying out an in vivo therapeutic and/or diagnostic procedure. In an embodiment, the invention comprises a method of carrying out an in vivo phototherapeutic, photoactivation, and/or photosensitizing procedure. The present methods have broad clinical utility which includes, but is not limited to, phototherapy of tumors, inflammatory processes, and impaired vasculature. In embodiments, subjects of the invention may be any mammal, such as a human, and optionally the subject of the present methods is a patient in need of treatment and/or diagnosis. The present methods are also useful in ex vivo and in vitro procedures, including medical therapeutic and diagnostic procedures.
[0219] Methods of the invention may optionally further comprise a number of other steps. In an embodiment, the method further comprises the step of administering the phototherapeutic agent into a bodily fluid of the subject. The phototherapeutic agent may be introduced into the patient by any suitable method, including intravenous, intraperitoneal or subcutaneous injection or infusion, oral administration, transdermal absorption through the skin, or by inhalation. In an embodiment, the method further comprises contacting a target tissue, such as an organ, tissue, tumor, lesion, or cell type, with a compound of any one of formulas (FX1) - (FX59) prior to or during the exposure step. In an embodiment, the method further comprises allowing the compound to accumulate in a target tissue prior to exposure of the phototherapeutic agent to electromagnetic radiation. In an embodiment, the method further comprises contacting and/or selectively targeting the diagnostic agent to a selected organ, tissue, tumor, lesion, inflammation, or cell type. In an embodiment, the phototherapeutic agent is administered to the skin, a tumor, surgical site, or a wound site. In an embodiment, for example, the phototherapeutic agent is administered and/or delivered to a blood vessel, lung, heart, throat, ear, rectum, bladder, stomach, intestines, esophagus, liver, brain, prostrate, breast, or pancreas of the subject.
[0220] As will be understood by one having skill in the art, the optical conditions for the step of exposing the phototherapeutic agent administered to the patient to electromagnetic radiation will vary considerably with the (i) therapeutic and/or diagnostic objectives, and (ii) the condition of the subject (e.g., height, weight, state of health etc.). In an embodiment, the applied electromagnetic radiation has wavelengths, energy and/or fluence sufficient to achieve a desired therapeutic and/or diagnostic result. In an embodiment, the electromagnetic radiation has wavelengths, energy and/or fluence sufficient to activate the phototherapeutic agent, for example wavelengths, energy and/or fluence sufficient to result in generation of reactive species, including free radicals and/or singlet oxygen. In an embodiment, the electromagnetic radiation has wavelengths, energy and/or fluence sufficient to result in cleavage of at least one photolabile bond of the optical agent upon absorption. In an embodiment, the electromagnetic radiation exposed to the phototherapeutic agent has wavelengths corresponding to a maximum in the absorption spectrum of the
phototherapeutic agent, preferably for some applications a maximum (e.g., within 20»nm of a maximum in the absorption spectrum) in the visible or NIR regions of the electromagnetic spectrum. Optionally, excitation is achieved using electromagnetic radiation substantially free (e.g., less than about 10% of total radiant energy), of ultraviolet radiation, for example, to minimize exposure of the subject to electromagnetic radiation capable of causing unwanted cell or tissue damage. Electromagnetic radiation may be provided to the phototherapeutic agent using a range of optical sources and/or surgical instrumentation, including a laser, light emitting diodes, fiber optic device, endoscope, catheter, optical filters, or any combination of these.
EXAMPLE 3: TARGETED OPTICAL AGENTS
3.a. Targeting Methods
[0221] The invention include methods for phototherapy using an optical agent providing targeted delivery to a selected target tissue. Embodiments of this aspect use an optical agent, such as a photosensitizer, having a targeting ligand. As will be understood by one of skill in the art, selection of the composition of a targeting ligand in the present methods will dependent on therapeutic and/or diagnostic objectives, the condition of the subject and the chemical composition and properties of the target tissue of interest.
[0222] In one example, a targeted compound can contain all or part of a steroid hormone or a steroid receptor binding compound, and therefore target steroid hormone sensitive receptors. In this example, the targeted compound is administered, targets and preferably accumulates in the desired site such as breast and/or prostate lesion and is photoactivated for monitoring, imaging, or therapy remotely or at the target site. Similar target binding molecules and uses will be recognized by one skilled in the art. For example, the targeted compound can be a compound that targets and binds to a somatostatin, bombesin, CCK, and/or neurotensin receptor binding molecule, or can be a carcinogenic embryonic antigen-binding compound that binds to a carcinogenic embryonic antigen. These are then photoactivated at, for example, lung cancer cells with CCK receptor binding molecules, colorectal cancer cells with ST receptor and carcinoembryonic antigen (CEA) binding molecules, melanoma cells with dihydroxyindolecarboxylic acid, vascular sites of atherosclerotic plaque with integrin receptor binding molecules, brain lesions with amyloid plaque binding molecules, and the like.
[0223] Successful specific targeting of photoactive compounds to tumors using antibodies and peptides for diagnostic imaging of tumors has been described in Achilefu et al., Novel receptor- targeted fluorescent contrast agents for in vivo imaging of tumors, Investigative Radiology, 2000, 35, pp. 479-485; Ballou et al., Tumor labeling in vivo using cyanine conjugated monoclonal antibodies, Cancer Immunology and Immunotherapy, 1995, 41 , pp. 257-263; and Licha et al., New contrast agent for optical imaging: acid cleavable conjugates of cyanine dyes with biomolecules, in Biomedical Imaging: Reporters, Dyes and Instrumentation, Proceedings of SPIE, 1999, 3600, pp. 29-35. As such, it is widely accepted that targeted photochemicals are effective in targeting, detecting and treating a wide range of physiological and biological sites.
[0224] The optical agents of this example can contain additional functionalities that can be used to attach various types of biomolecules, synthetic polymers, and organized aggregates for selective delivery to various organs or tissues of interest. Examples of synthetic polymers include polyaminoacids, polyols, polyamines, polyacids, oligonucleotides, aborols, dendrimers, and aptamers. The invention includes, but is not limited to, phototherapeutic agents comprising an optical agent - biomolecule conjugate which provides advantages over nonspecific optical agents or the conjugation of optical agents to very large biomolecules. These conjugates provide enhanced localization in, and rapid visualization of, tumors which is beneficial for imaging, monitoring, diagnosis and therapy. The agents are rapidly cleared from blood and non-target tissues so there is less concern for accumulation and for toxicity. A variety of high purity compounds can be easily synthesized for combinatorial screening of new targets, e.g. , to identify receptors or targeting agents, and for the ability to affect the pharmacokinetics of the conjugates by minor structural changes.
[0225] In some embodiments, a liposome or micelle can be utilized as a carrier or vehicle for the composition. For example, in some embodiments, an optical agent comprises a compound of the invention that can be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, can be on the external surface of the liposome or micelle. As another example, a targeting ligand can be externally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains a phototherapeutic agent/photosensitizer compound of the invention) to the desired tissue, organ, or other site in the body.
[0226] In embodiments, compounds of the invention are useful for both oncology and non- oncology applications. Some specific targets are tumors accessible via endoscope. In an application, a compound that targets a protein, polypeptide, oligonucleotide or other biomolecule associated with such a tumor is administered to the tumor via endoscope or other useful method. Then, the compounds of the invention can be used in phototherapeutic applications, monitoring applications, diagnosis applications or imaging applications. Other specific target tissues include colon, lung, ovarian, cervical, esophageal, bladder, blood, stomach cancers, endometriosis, and bacterial infections.
3b: Targeting Ligands
[0227] The estrogen receptor is an example of a steroid receptor to which steroid receptor binding molecules would bind. The following compounds are known to bind to the estrogen receptor: estratriol; 17p-aminoestrogen (AE) derivatives such as prolame and butolame; drugs such as tamoxifen, ICI-164384, raloxifene, and genistein; 17p-estradiol; glucocorticoids;
progesterone; estrogens; retinoids; fatty acid derivatives; and phytoestrogens. In addition, commercially available kits can identify compounds specific for binding to the estrogen receptor (e.g. , Estrogen Receptor-alpha Competitor Assay Kit, Red; and Estrogen Receptor-beta
Competitor Assay Kit, Red (Invitrogen Corp., Carlsbad CA).
[0228] The glucose receptor is an example of a carbohydrate receptor to which carbohydrate receptor binding molecules would bind. The glucose conjugate N-palmitoyl glucosamine [NPG] is known to bind the glucose receptor (Dufes et al., Pharm. Res. 17: 1250, 2000). The glycoprotein hormone receptor is another example of a carbohydrate receptor to which carbohydrate receptor binding molecules would bind. Follicle stimulating hormone (FSH) is known to bind the
glycoprotein hormone receptor (Tilly et al., Endocrinology 131 : 799, 1992). Other compounds known to bind the carbohydrate receptor, and hence examples of carbohydrate receptor binding molecules, are: polysialic acid, bacterial adhesins (specialized surface proteins that mediate binding of many pathogenic bacteria, such as enterohemorrhagic E. coli (EHEC) and Shigella dysenteriae, to host cells, which allow these bacteria to colonize host cell surfaces), soluble carbohydrate receptor analogs, artificial glycopolymers and other multivalent glycoconjugates such as an acrylamide copolymer carrying -L-fucopyranoside and 3-sulfo-D-galactopyranoside in clusters, isomeric carbohydrates, synthetic derivatives, neoglycoproteins, neoglycolipids, glycosidases, and glycosyltransferases. Carbohydrate binding proteins can be screened with phage display libraries as known to a person of ordinary skill in the art.
[0229] Somatostatin receptor binding molecules include somatostatin and somatostatin receptor analogs, octreotide, glycosylated somatostatin-14 (somatostatin-dextran70), seglitide, and peptides P587 and P829 as described in Vallabhajosula et al., J. Nuclear Med., 37:1016, 1996. [0230] Cholecystokinin receptor binding molecules include the endogenous peptides cholecystekinin (CCK)-4, CCK-8, CCK-33, and gastrin; antagonists devazepide and lorglumide; agonists BC264 [Tyr(S03H)-gNle-mGly-Trp-(NMe)Nle-Asp-Phe-NH3] and desulfated CCK-8; Kinevac (synthetic cholecystekinin, sincalide); and CCK analogues modified at the sulfated tyrosyl at position 27.
[0231] Neurotensin receptor binding molecules include neurotensin, neuromedin N, JMV449 (H- Lysv|/(CH2NH)-Lys-Pro-Tyr-lle-Leu), the non-peptide antagonist SR142948A (2-([5-(2,6- dimethoxyphenyl)-1 -(4-(/V-[3-dimethylaminopropyl]-/V-methylcarbamoyl)-2-isopropylphenyl)-1 /-/- pyrazole-3-carbonyl)amino)adamantine-2-carboxylic acid hydrochloride), and levocobastine. Commercially available neurotensin receptor binding kits can evaluate potential neurotensin receptor binding molecules (e.g., DELFIA Neurotensin Receptor Binding Kit, PerkinElmer (Boston MA)).
[0232] Bombesin receptor binding molecules include the endogenous ligands gastrin-releasing peptide (GRP), neuromedin B (NMB), and GRP-18-27, and antagonists including JMV-1458 (glycine-extended bombesin (paraphydroxy-phenyl-propionyl-Gln-Trp-Ala-Val-Gly-His-Leu-Met- Gly-OH)), JMV-641 , JMV-1799, and JMV-1802, PD165929, 1 -naphthoyl-[DAIa24,DPro26,V26- 27]GRP-20-27, kuwanon H, and kuwanon G. Commercially available bombesin receptor binding kits can evaluate potential bombesin receptor binding molecules (e.g., DELFIA Bombesin
Receptor Binding Kit, PerkinElmer (Boston MA)).
[0233] ST receptor binding molecules include native ST peptide, and SEQ ID NO:2, SEQ ID NO:3, SEQ ID NOS:5-54 and fragments and derivatives thereof from U.S. Patent No. 5,518,888.
[0234] Compounds of the invention can contain all or part of a targeting ligand, receptor or peptide known to bind to a specific target, such as a target tissue.
[0235] Targeting ligands may be linked to the backbone or other portion of the present compounds using a range of synthetic approaches known in the art, including the synthetic approaches for conjugating biomolecule targeting ligands to optical agents as disclosed in Hnatowich et al., Radiolabeling of Antibodies: A simple and efficient method, Science, 1983, 220, p. 613; Pelegrin et al., Photoimmunodiagnostics with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies, Journal of Cellular Pharmacology, 1992, 3, pp. 141-145; Achilefu et al., Novel receptor-targeted fluorescent contrast agents for in vivo imaging of tumors, Investigative Radiology, 2000, 35, pp. 479-485; Ballou et al., Tumor labeling in vivo using cyanine conjugated monoclonol antibodies, Cancer Immunology and Immunotherapy, 1995, 41 , pp. 257-263; and Licha et al., New contrast agent for optical imaging: acid cleavable conjugates of cyanine dyes with biomolecules, in Biomedical Imaging: Reporters, Dyes and Instrumentation, Proceedings of SPIE, 1999, 3600, pp. 29-35; and U.S. Patent No. 5,714,342.
[0236] Linking of biomolecule targeting ligands having an amine group, for example, may be achieved by techniques involving succinimido active esters. For example, a carboxyl group of a compound of the invention is activated by making a mixed anhydride in situ with
isobutylchloroformate. The activated compound is subsequently reacted with any biomolecule bearing an amino group, such as a polypeptide, protein, enzyme, antibody or fragment thereof, to achieve linking of the biomolecule to the compound so as to provide a targeting ligand covalently bond to the compound. Alternatively, a carboxyl group of the present compounds may be first esterified with N-hydroxysuccinimide, and subsequently reacted with the amino group of a biomolecule, such as a polypeptide, protein, enzyme, antibody or fragment thereof, to form an amide bond linking the biomolecule to the compound so as to provide a targeting ligand covalently bond to the compound.
EXAMPLE 4: ADMINISTRATION AND FORMULATION
4. a: Salts and Prodrugs
[0237] The invention contemplates pharmaceutically active compounds either chemically synthesized or formed by in vivo biotransformation to compounds set forth herein.
[0238] Compounds of this invention and compounds useful in the methods of this invention include those of the compounds and formula(s) described herein and pharmaceuticaily-acceptable salts and esters of those compounds. In embodiments, salts include any salts derived from the acids and bases of the formulas herein which are acceptable for use in human or veterinary applications. In embodiments, the term ester refers to hydrolyzable esters of compounds of the names and formulas herein. In embodiments, salts and esters of the compounds of the formulas herein can include those which have the same or better therapeutic, diagnostic, or pharmaceutical (human or veterinary) general properties as the compounds of the formulas herein. In an embodiment, a composition of the invention is a compound or salt or ester thereof suitable for pharmaceutical formulations.
[0239] Compounds of the invention can have prodrug forms. Prodrugs of the compounds of the invention are useful in embodiments including compositions and methods. Any compound that will be converted in vivo to provide a biologically, pharmaceutically, diagnostically, or therapeutically active form of a compound of the invention is a prodrug. Various examples and forms of prodrugs are well known in the art. Examples of prodrugs are found, inter alia, in: Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985); Methods in Enzymology, Vol. 42, at pp. 309-396, edited by K. Widder, et. al. (Academic Press, 1985); A Textbook of Drug Design and Development, edited by Krosgaard-Larsen and H. Bundgaard, Chapter 5, "Design and Application of Prodrugs," by H. Bundgaard, at pp. 1 13-191 (1991 ); H. Bundgaard, Advanced Drug Delivery Reviews, Vol. 8, p. 1- 38 (1992); H. Bundgaard, et al., Journal of Pharmaceutical Sciences, Vol. 77, p. 285 (1988); and Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392). A prodrug, such as a pharmaceutically acceptable prodrug, can represent prodrugs of the compounds of the invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use. Prodrugs of the invention can be rapidly transformed in vivo to a parent compound of a compound described herein, for example, by hydrolysis in blood or by other cell, tissue, organ, or system processes. Further discussion is provided in: T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, V. 14 of the A.C.S. Symposium Series; and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical
Association and Pergamon Press (1987).
[0240] Optical agents of the invention can be formulated with pharmaceutically-acceptable anions and/or cations. Pharmaceutically-acceptable cations include among others, alkali metal cations (e.g., Li+, Na+, K+), alkaline earth metal cations (e.g., Ca2+, Mg2+), non-toxic heavy metal cations and ammonium (NH4 +) and substituted ammonium (N(R')4 +, where R' is hydrogen, alkyl, or substituted alkyl, i.e., including, methyl, ethyl, or hydroxyethyl, specifically, trimethyl ammonium, triethyl ammonium, and triethanol ammonium cations). Pharmaceutically-acceptable anions include, among others, halides (e.g., F~, CI", Br", At"), sulfate, acetates (e.g., acetate,
trifluoroacetate), ascorbates, aspartates, benzoates, citrates, and lactate.
[0241] Pharmaceutically acceptable salts comprise pharmaceutically-acceptable anions and/or cations. As used herein, the term "pharmaceutically acceptable salt" can refer to acid addition salts or base addition salts of the compounds in the present disclosure. A pharmaceutically acceptable salt is any salt which retains at least a portion of the activity of the parent compound and does not impart significant deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered. Pharmaceutically acceptable salts include metal complexes and salts of both inorganic and organic acids. Pharmaceutically acceptable salts include metal salts such as aluminum, calcium, iron, magnesium, manganese and complex salts.
Pharmaceutically acceptable salts include, but are not limited to, acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, cilexetil, citric, edetic, edisylic, estolic, esyl, esylic, formic, fumaric, gluceptic, gluconic, glutamic, glycolic,
glycolylarsanilic, hexamic, hexylresorcjnoic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, lactobionic, maleic, malic, malonic, mandelic,
methanesulfonic, methylnitric, methylsulfuric, mucic, muconic, napsylic, nitric, oxalic, p- nitromethanesulfonic, pamoic, pantothenic, phosphoric, monohydrogen phosphoric, dihydrogen phosphoric, phthalic, polygalactouronic, propionic, salicylic, stearic, succinic, sulfamic, sulfanlic, sulfonic, sulfuric, tannic, tartaric, teoclic, toluenesulfonic, and the like. Pharmaceutically acceptable salts can be derived from amino acids, including, but not limited to, cysteine. Other
pharmaceutically acceptable salts can be found, for example, in Stahl et al., Handbook of
Pharmaceutical Salts: Properties, Selection, and Use, Wiley-VCH, Verlag Helvetica Chimica Acta, Zurich, 2002. (ISBN 3-906390-26-8).
4.b: Efficacy [0242] Typically, a compound of the invention, or pharmaceutically acceptable salt thereof, is administered to a subject in a diagnostically or therapeutically effective amount. One skilled in the art generally can determine an appropriate dosage.
[0243] Compositions for oral administration can be, for example, prepared in a manner such that a single dose in one or more oral preparations contains at least about 20 mg of the present compound per square meter of subject body surface area, or at least about 50, 100, 150, 200, 300, 400, or 500 mg of the present compound per square meter of subject body surface area (the average body surface area for a human is, for example, 1.8 square meters). In particular, a single dose of a composition for oral administration can contain from about 20 to about 600 mg, and in certain aspects from about 20 to about 400 mg, in another aspect from about 20 to about 300 mg, and in yet another aspect from about 20 to about 200 mg of the present compound per square meter of subject body surface area. Compositions for parenteral administration can be prepared in a manner such that a single dose contains at least about 20 mg of the present compound per square meter of subject body surface area, or at least about 40, 50, 100, 150, 200, 300, 400, or 500 mg of the present compound per square meter of subject body surface area. In particular, a single dose in one or more parenteral preparations contains from about 20 to about 500 mg, and in certain aspects from about 20 to about 400 mg, and in another aspect from about 20 to about 450 mg, and in yet another aspect from about 20 to about 350 mg of the present compound per square meter of subject body surface area. It should be recognized that these oral and parenteral dosage ranges represent generally preferred dosage ranges, and are not intended to limit the invention. The dosage regimen actually employed can vary widely, and, therefore, can deviate from the generally preferred dosage regimen. It is contemplated that one skilled in the art will tailor these ranges to the individual subject.
[0244] Toxicity and therapeutic efficacy of such compounds and bioconjugates can be determined by standard pharmaceutical procedures in cell cultures or experimental animals for determining the LD50 (the dose lethal to 50% of the population) and the ED50, (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index that can be expressed as the ratio LD5o/ED50. Compounds and bioconjugates that exhibit large therapeutic indices are preferred. While compounds and bioconjugates exhibiting toxic side effects can be used, care should be taken to design a delivery system that targets such compounds and bioconjugates to the site affected by the disease or disorder in order to minimize potential damage to unaffected cells and reduce side effects.
[0245] Data obtained from the cell culture assays and animal studies can be used in formulating a range of dosages for use in humans and other mammals. The dosage of such compounds and bioconjugates lies preferably within a range of circulating plasma or other bodily fluid
concentrations that include the ED50 and provides clinically efficacious results (i.e., reduction in disease symptoms). The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound and bioconjugate of the present invention, the therapeutically effective amount can be estimated initially from cell culture assays. A dosage can be formulated in animal models to achieve a circulating plasma
concentration range that includes the ED50 (the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful dosages in humans and other mammals. Compound and bioconjugate levels in plasma can be measured, for example, by high performance liquid chromatography.
[0246] An amount of a compound or bioconjugate that can be combined with a pharmaceutically acceptable carrier to produce a single dosage form will vary depending upon the patient treated and the particular mode of administration. It will be appreciated by those skilled in the art that the unit content of a compound/bioconjugate contained in an individual dose of each dosage form need not in itself constitute a therapeutically effective amount, as the necessary therapeutically effective amount could be reached by administration of a number of individual doses. The selection of dosage depends upon the dosage form utilized, the condition being treated, and the particular purpose to be achieved according to the determination of those skilled in the art.
[0247] The dosage and dosage regime for treating a disease or condition can be selected in accordance with a variety of factors, including the type, age, weight, sex, diet and/or medical condition of the patient, the route of administration, pharmacological considerations such as activity, efficacy, pharmacokinetic and/or toxicology profiles of the particular
compound/bioconjugate employed, whether a compound/bioconjugate delivery system is utilized, and/or whether the compound/bioconjugate is administered as a pro-drug or part of a drug combination. Thus, the dosage regime actually employed can vary widely from subject to subject, or disease to disease and different routes of administration can be employed in different clinical settings.
[0248] The identified compounds/bioconjugates monitor, treat, inhibit, control and/or prevent, or at least partially arrest or partially prevent, diseases and conditions of interest and can be administered to a subject at therapeutically effective amounts and optionally diagnostically effective amounts. Compositions/formulations of the present invention comprise a therapeutically effective amount (which can optionally include a diagnostically effective amount) of at least one compound or bioconjugate of the present invention. Subjects receiving treatment that includes a compound/bioconjugate of the invention are preferably animals (e.g., mammals, reptiles and/or avians), more preferably humans, horses, cows, dogs, cats, sheep, pigs, and/or chickens, and most preferably humans.
4.c: Administration
[0249] The preferred composition depends on the route of administration. Any route of administration can be used as long as the target of the compound or pharmaceutically acceptable salt is available via that route. Suitable routes of administration include, for example, oral, intravenous, parenteral, inhalation, rectal, nasal, topical (e.g., transdermal and intraocular), intravesical, intrathecal, enteral, pulmonary, intralymphatic, intracavital, vaginal, transurethral, intradermal, aural, intramammary, buccal, orthotopic, intratracheal, intralesional, percutaneous, endoscopical, transmucosal, sublingual, and intestinal administration.
[0250] In an embodiment, the invention provides a method for treating a medical condition comprising administering to a subject (e.g. patient) in need thereof, a therapeutically effective amount of a composition of the invention, such as a compound of any one of formulas (FX1) - (FX59). In an embodiment, the invention provides a method for diagnosing or aiding in the diagnosis of a medical condition comprising administering to a subject in need thereof, a diagnostically effective amount of a composition of the invention. In an embodiment, the medical condition is cancer, or various other diseases, injuries, and disorders, including cardiovascular disorders such as atherosclerosis and vascular restenosis, inflammatory diseases, ophthalmic diseases and dermatological diseases.
[0251] The diagnostic and therapeutic formulations of this invention can be administered alone, but can be administered with a pharmaceutical carrier selected upon the basis of the chosen route of administration and standard pharmaceutical practice.
[0252] Any suitable form of administration can be employed in connection with the diagnostic and therapeutic formulations of the invention. The diagnostic and therapeutic formulations of this invention can be administered intravenously, in oral dosage forms, intraperitoneally,
subcutaneously, or intramuscularly, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
[0253] The present compositions, preparations and formulations can be formulated into diagnostic or therapeutic compositions for enteral, parenteral, topical, aerosol, inhalation, or cutaneous administration. Topical or cutaneous delivery of the compositions, preparations and formulations can also include aerosol formulation, creams, gels, solutions, etc. The present compositions, preparations and formulations are administered in doses effective to achieve the desired diagnostic and/or therapeutic effect. Such doses can vary widely depending upon the particular compositions employed in the composition, the organs or tissues to be examined, the equipment employed in the clinical procedure, the efficacy of the treatment achieved, and the like. These compositions, preparations and formulations contain an effective amount of the
composition(s), along with conventional pharmaceutical carriers and excipients appropriate for the type of administration contemplated. These compositions, preparations and formulations can also optionally include stabilizing agents and skin penetration enhancing agents,
(i) Parenteral Administration
[0254] Compounds and bioconjugates of the present invention can be formulated for parenteral administration by injection (e.g., by bolus injection or continuous infusion). Formulations for injection can be presented in unit dosage form in ampoules or in multi-dose containers with an optional preservative added. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass, plastic or the like. The formulation can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [0255] For example, a parenteral preparation can be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent (e.g., as a solution in 1 ,3-butanediol). Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid can be used in the parenteral preparation.
[0256] Alternatively, compounds and bioconjugates of the present invention can be formulated in powder form for constitution with a suitable vehicle, such as sterile pyrogen-free water, before use. For example, a compound/bioconjugate suitable for parenteral administration can include a sterile isotonic saline solution containing between 0.1 percent and 90 percent weight per volume of the compound/bioconjugate. By way of example, a solution can contain from about 5 percent to about 20 percent, more preferably from about 5 percent to about 17 percent, more preferably from about 8 to about 14 percent, and still more preferably about 10 percent weight per volume of the compound/bioconjugate. The solution or powder preparation can also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Other methods of parenteral delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
(ii) Oral Administration
[0257] For oral administration, a compound/bioconjugate of the invention can be formulated to take the form of tablets or capsules prepared by conventional means with one or more
pharmaceutically acceptable carriers (e.g., excipients such as binding agents, fillers, lubricants and disintegrants).
(iii) Controlled-Release Administration
[0258] Controlled-release (or sustained-release) preparations can be formulated to extend the activity of a compound/bioconjugate and reduce dosage frequency. Controlled-release
preparations can also be used to effect the time of onset of action or other characteristics, such as blood levels of the compound/bioconjugate, and consequently affect the occurrence of side effects.
[0259] Controlled-release preparations can be designed to initially release an amount of a compound/bioconjugate that produces the desired therapeutic effect, and gradually and continually release other amounts of the compound/bioconjugate to maintain the level of therapeutic effect over an extended period of time. In order to maintain a near-constant level of a
compound/bioconjugate in the body, the compound/bioconjugate can be released from the dosage form at a rate that will replace the amount of compound/bioconjugate being metabolized and/or excreted from the body. The controlled-release of a compound/bioconjugate can be stimulated by various inducers, e.g., change in pH, change in temperature, enzymes, water, and/or other physiological conditions or molecules. [0260] Controlled-release systems can include, for example, an infusion pump which can be used to administer the compound/bioconjugate in a manner similar to that used for delivering insulin or chemotherapy to the body generally, or to specific organs or tumors. Typically, using such a system, the compound/bioconjugate is administered in combination with a biodegradable, biocompatible polymeric implant that releases the compound/bioconjugate over a controlled period of time at a selected site. Examples of polymeric materials include polyanhydrides,
polyorthoesters, polyglycolic acid, polylactic acid, polyethylene vinyl acetate, and copolymers and combinations thereof. In addition, a controlled release system can be placed in proximity of a therapeutic target (e.g., organ, tissue, or group of cells), thus requiring only a fraction of a systemic dosage.
[0261] Compounds/bioconjugates of the invention can be administered by other controlled- release means or delivery devices that are well known to those of ordinary skill in the art. These include, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or the like, or a combination of any of the above to provide the desired release profile in varying proportions. Other methods of controlled-release delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention,
(iv) Inhalation Administration
[0262] Compounds/bioconjugates of the invention can be administered directly to the lung of a patient/subject by inhalation. For administration by inhalation, a compound/bioconjugate can be conveniently delivered to the lung by a number of different devices. For example, a Metered Dose Inhaler ("MDI") which utilizes canisters that contain a suitable low boiling point propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas can be used to deliver a compound/bioconjugate directly to the lung. MDI devices are available from a number of suppliers such as 3M Corporation, Aventis, Boehringer Ingleheim, Forest Laboratories, GlaxoSmithKline, Merck & Co. and Vectura.
[0263] Alternatively, a Dry Powder Inhaler (DPI) device can be used to administer a
compound/bioconjugate to the lung. DPI devices typically use a mechanism such as a burst of gas to create a cloud of dry powder inside a container, which can then be inhaled by the patient. DPI devices are also well known in the art and can be purchased from a number of vendors which include, for example, GlaxoSmithKline, Nektar Therapeutics, Innovata and Vectura. A popular variation is the multiple dose DPI ("MDDPI") system, which allows for the delivery of more than one therapeutic dose. MDDPI devices are available from companies such as AstraZeneca, GlaxoSmithKline, TEVA, Merck & Co., SkyePharma and Vectura. For example, capsules and cartridges of gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound/bioconjugate and a suitable powder base such as lactose or starch for these systems.
[0264] Another type of device that can be used to deliver a compound/bioconjugate to the lung is a liquid spray device supplied, for example, by Aradigm Corporation. Liquid spray systems use extremely small nozzle holes to aerosolize liquid compound/bioconjugate formulations that can then be directly inhaled into the lung. For example, a nebulizer device can be used to deliver a compound/bioconjugate to the lung. Nebulizers create aerosols from liquid
compound/bioconjugate formulations by using, for example, ultrasonic energy to form fine particles that can be readily inhaled. Examples of nebulizers include devices supplied by Aventis and Battelle.
[0265] In another example, an electrohydrodynamic ("EHD") aerosol device can be used to deliver a compound/bioconjugate to the lung. EHD aerosol devices use electrical energy to aerosolize liquid compound/bioconjugate solutions or suspensions. The electrochemical properties of the compound/bioconjugate formulation are important parameters to optimize when delivering this compound/bioconjugate to the lung with an EHD aerosol device. Such optimization is routinely performed by one of skill in the art. Other methods of intra-pulmonary delivery of
compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
[0266] Liquid compound/bioconjugate formulations suitable for use with nebulizers and liquid spray devices and EHD aerosol devices will typically include the compound/bioconjugate with a pharmaceutically acceptable carrier. In one exemplary embodiment, the pharmaceutically acceptable carrier is a liquid such as alcohol, water, polyethylene glycol or a perfluorocarbon. Optionally, another material can be added to alter the aerosol properties of the solution or suspension of the compound/bioconjugate. For example, this material can be a liquid such as an alcohol, glycol, polyglycol or a fatty acid. Other methods of formulating liquid
compound/bioconjugate solutions or suspensions suitable for use in aerosol devices are known to those of skill in the art.
(v) Depot Administration
[0267] A compound/bioconjugate of the invention can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Accordingly, the compound/bioconjugate can be formulated with suitable polymeric or hydrophobic materials such as an emulsion in an acceptable oil or ion exchange resin, or as sparingly soluble derivatives such as a sparingly soluble salt. Other methods of depot delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
(vi) Topical Administration
[0268] For topical application, a compound/bioconjugate can be combined with a
pharmaceutically acceptable carrier so that an effective dosage is delivered, based on the desired activity ranging from an effective dosage, for example, of 1 .0 μΜ to 1 .0 mM. In one aspect of the invention, a topical formulation of a compound/bioconjugate can be applied to the skin. The pharmaceutically acceptable carrier can be in the form of, for example, and not by way of limitation, an ointment, cream, gel, paste, foam, aerosol, suppository, pad or gelled stick. [0269] A topical formulation can include a therapeutically effective amount of a compound/bioconjugate in an ophthalmologically acceptable excipient such as buffered saline, mineral oil, vegetable oils such as corn or arachis oil, petroleum jelly, Miglyol 182, alcohol solutions, or liposomes or liposome-like products. Any of these formulations of such
compounds/bioconjugates can include preservatives, antioxidants, antibiotics,
immunosuppressants, and other biologically or pharmaceutically effective agents that do not exert a significant detrimental effect on the compound/bioconjugate. Other methods of topical delivery of compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
(vii) Rectal Administration
[0270] Compounds/bioconjugates of the invention can be formulated in rectal formulations such as suppositories or retention enemas that include conventional suppository bases such as cocoa butter or other glycerides and/or binders and/or carriers such as triglycerides, microcrystalline cellulose, gum tragacanth or gelatin. Rectal formulations can contain a compound/bioconjugate in the range of 0.5% to 10% by weight, for example. Other methods of rectal delivery of
compounds/bioconjugates will be known to the skilled artisan and are within the scope of the invention.
(viii) Other Systems of Administration
[0271] Various other delivery systems are known in the art and can be used to administer the compounds/bioconjugates of the invention. Moreover, these and other delivery systems can be combined and/or modified to promote optimization of the administration of
compounds/bioconjugates of the present invention. Exemplary formulations that include compounds/bioconjugates of the present invention are described elsewhere herein (the compounds/bioconjugates of the present invention are indicated as the active ingredient, but those of skill in the art will recognize that pro-drugs and compound combinations are also meant to be encompassed by this term).
4.d: Formulation
[0272] In an embodiment, the invention provides a medicament which comprises a
therapeutically effective amount of one or more compositions of the invention, such as a compound of any one of formulas (FX1) - (FX59). In an embodiment, the invention provides a medicament which comprises a diagnostically effective amount of one or more compositions of the invention. In an embodiment, the invention provides a method for making a medicament for treatment of a condition described herein, such as the treatment of cancer, inflammation, stenosis or a vascular disease. In an embodiment, the invention provides a method for making a medicament for diagnosis or aiding in the diagnosis of a condition described herein, such as the diagnosis of cancer, inflammation, stenosis or a vascular disease. In an embodiment, the invention provides the use of one or more compositions set forth herein for the making of a medicament for the treatment of cancer, inflammation, stenosis or a vascular disease. In an embodiment, the invention provides the use of one or more compositions set forth herein for the treatment of a disease. In an embodiment, the invention provides the use of one or more compositions set forth herein for the diagnosis of a disease. Compositions of the invention include formulations and preparations comprising one or more of the present optical agents provided in an aqueous solution, such as a pharmaceutically acceptable formulation or preparation. Optionally, compositions of the invention further comprise one or more pharmaceutically acceptable surfactants, buffers, electrolytes, salts, carriers, binders, coatings, preservatives and/or excipients.
[0273] In an embodiment, the invention provides a pharmaceutical formulation having an active ingredient comprising a composition of the invention, such as a compound of any one of formulas (FX1) - (FX59). In an embodiment, the invention provides a method of synthesizing a composition of the invention or a pharmaceutical formulation thereof, such as a compound of any one of formulas (FX1) - (FX59). In an embodiment, a pharmaceutical formulation comprises one or more excipients, carriers, diluents, and/or other components as would be understood in the art.
Preferably, the components meet the standards of the National Formulary ("NF"), United States Pharmacopoeia ("USP"; United States Pharmacopeial Convention Inc., Rockville, Maryland), or Handbook of Pharmaceutical Manufacturing Formulations (Sarfaraz K. Niazi, all volumes, ISBN: 9780849317521 , ISBN 10: 0849317525; CRC Press, 2004). See, e.g., United States
Pharmacopeia and National Formulary (USP 30-NF 25), Rockville, MD: United States
Pharmacopeial Convention (2007 and 2008), and each of any earlier editions; The Handbook of Pharmaceutical Excipients, published jointly by the American Pharmacists Association and the Pharmaceutical Press (Pharmaceutical Press (2005) (ISBN-10: 0853696187, ISBN-13: 978- 0853696186)); Merck Index, Merck & Co., Rahway, N.J.; and Gilman et al., (eds) (1996);
Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th Ed., Pergamon Press. In embodiments, the formulation base of the formulations of the invention comprises
physiologically acceptable excipients, namely, at least one binder and optionally other
physiologically acceptable excipients. Physiologically acceptable excipients are those known to be usable in the pharmaceutical technology sectors and adjacent areas, particularly, those listed in relevant pharmacopeias (e.g. DAB, Ph. Eur., BP, NF, USP), as well as other excipients whose properties do not impair a physiological use.
[0274] This invention also is directed, in part, to pharmaceutical compositions including a therapeutically effective amount of a compound or salt of this invention, as well as processes for making such compositions. Such compositions generally include one or more pharmaceutically acceptable carriers (e.g., excipients, vehicles, auxiliaries, adjuvants, diluents) and can include other active ingredients. Formulation of these compositions can be achieved by various methods known in the art. A general discussion of these methods can be found in, for example, Hoover, John E., Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA: 1975). See also, Lachman, L, eds., Pharmaceutical Dosage Forms (Marcel Decker, New York, N. Y., 1980).
[0275] The diagnostic and therapeutic formulations of this invention and medicaments of this invention can further comprise one or more pharmaceutically acceptable carriers, excipients, buffers, emulsifiers, surfactants, electrolytes or diluents. Such compositions and medicaments are prepared in accordance with acceptable pharmaceutical procedures, such as, for example, those described in Remingtons Pharmaceutical Sciences, 17th edition, ed. Alfonoso R. Gennaro, Mack Publishing Company, Easton, Pa. (1985).
[0276] Compositions of the invention include formulations and preparations comprising one or more of the present compounds provided in an aqueous solution, such as a pharmaceutically acceptable formulation or preparation. Optionally, compositions of the invention further comprise one or more pharmaceutically acceptable surfactants, buffers, electrolytes, salts, carriers, binders, coatings, preservatives and/or excipients.
[0277] Compounds and bioconjugates of the present invention can be formulated by known methods for administration to a subject using several routes which include, but are not limited to, parenteral, oral, topical, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and ophthalmic routes. An individual compound/bioconjugate can be administered in combination with one or more additional compounds/bioconjugates of the present invention and/or together with other biologically active or biologically inert agents. Such biologically active or inert agents can be in fluid or mechanical communication with the
compound(s)/bioconjugate(s) or attached to the compound(s)/bioconjugate(s) by ionic, covalent, Van der Waals, hydrophobic, hydrophilic or other physical forces. It is preferred that administration is localized in a subject, but administration can also be systemic.
[0278] Compounds and bioconjugates of the present invention can be formulated by any conventional manner using one or more pharmaceutically acceptable carriers. Thus, the compound(s)/bioconjugate(s) and their pharmaceutically acceptable salts and solvates can be specifically formulated for administration, e.g. , by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration. The
compounds/bioconjugates can take the form of charged, neutral and/or other pharmaceutically acceptable salt forms. Examples of pharmaceutically acceptable carriers include, but are not limited to, those described in REMINGTON'S PHARMACEUTICAL SCIENCES (A.R. Gennaro, Ed.), 20th edition, Williams & Wilkins PA, USA (2000).
[0279] Compounds and bioconjugates of the present invention can be formulated in the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, controlled- or sustained- release formulations and the like. Such formulations will contain a therapeutically effective amount of the compound/bioconjugate, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
[0280] Pharmaceutically acceptable carriers that can be used in conjunction with the compounds of the invention are well known to those of ordinary skill in the art. Carriers can be selected based on a number of factors including, for example, the particular compound(s) or pharmaceutically acceptable salt(s) used; the compound's concentration, stability, and intended bioavailability; the condition being treated; the subject's age, size, and general condition; the route of administration; etc. A general discussion related to carriers can be found in, for example, J.G. Nairn, Remington's Pharmaceutical Science, pp. 1492-1517 (A. Gennaro, ed., Mack Publishing Co., Easton, Pa. (1985)).
[0281] Solid dosage forms for oral administration include, for example, capsules, tablets, gelcaps, pills, dragees, troches, powders, granules, and lozenges. In such solid dosage forms, the compounds or pharmaceutically acceptable salts thereof can be combined with one or more pharmaceutically acceptable carriers. The compounds and pharmaceutically acceptable salts thereof can be mixed with carriers including, but not limited to, lactose, sucrose, starch powder, corn starch, potato starch, magnesium carbonate, microcrystalline cellulose, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, sodium carbonate, agar, mannitol, sorbitol, sodium saccharin, gelatin, acacia gum, alginic acid, sodium alginate, tragacanth, colloidal silicon dioxide, croscarmellose sodium, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration. Such capsules or tablets can contain a controlled-release formulation, as can be provided in a dispersion of the compound or salt in hydroxypropylmethyl cellulose. In the case of capsules, tablets, and pills, the dosage forms also can include buffering agents, such as sodium citrate, or magnesium or calcium carbonate or bicarbonate. Tablets and pills additionally can, for example, include a coating (e.g., an enteric coating) to delay disintegration and absorption. The concentration of the present compounds in a solid oral dosage form can be from about 5 to about 50% for example, and in certain aspects from about 8 to about 40%, and in another aspect from about 10 to about 30% by weight based on the total weight of the composition.
[0282] Liquid dosage forms of the compounds of the invention for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also can include adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents. The concentration of the present compounds in the liquid dosage form can be from about 0.01 to about 5 mg, and in certain aspects from about 0.01 to about 1 mg, and in another aspect from about 0.01 to about 0.5 mg per ml of the composition. Low concentrations of the compounds of the invention in liquid dosage form can be prepared in the case that the compound is more soluble at low concentrations. Techniques for making oral dosage forms useful in the invention are generally described in, for example, Modern Pharmaceutics, Chapters 9 and 10 (Banker & Rhodes, Editors (1979)). See also, Lieberman et al., Pharmaceutical Dosage Forms: Tablets (1981 ). See also, Ansel, Introduction to Pharmaceutical Dosage Forms (2nd Edition (1976)).
[0283] In some aspects of the invention, tablets or powders for oral administration can be prepared by dissolving the compound in a pharmaceutically acceptable solvent capable of dissolving the compound to form a solution and then evaporating when the solution is dried under vacuum. A carrier can also be added to the solution before drying. The resulting solution can be dried under vacuum to form a glass. The glass can then be mixed with a binder to form a powder. This powder can be mixed with fillers or other conventional tableting agents, and then processed to form a tablet. Alternatively, the powder can be added to a liquid carrier to form a solution, emulsion, suspension, or the like.
[0284] In some aspects, solutions for oral administration are prepared by dissolving the compound in a pharmaceutically acceptable solvent capable of dissolving the compound to form a solution. An appropriate volume of a carrier is added to the solution while stirring to form a pharmaceutically acceptable solution for oral administration.
[0285] In some embodiments, a liposome or micelle can be utilized as a carrier or vehicle for the composition. For example, in some embodiments, the compound can be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, can be on the external surface of the liposome or micelle. As another example, a targeting ligand can be externally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains the optical agents) to the desired tissue, organ, or other site in the body.
[0286] Injectable preparations (e.g., sterile injectable aqueous or oleaginous suspensions) can be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents. Acceptable vehicles for parenteral use include both aqueous and nonaqueous pharmaceutically-acceptable solvents. Suitable pharmaceutically acceptable aqueous solvents include, for example, water, saline solutions, dextrose solutions (such as DW5), electrolyte solutions, etc.
[0287] In one embodiment, the present compounds are formulated as nanoparticles or microparticles. Use of such nanoparticle or microparticle formulations can be beneficial for some applications to enhance delivery, localization, target specificity, administration, etc. of the compound. Potentially useful nanoparticles and microparticles include, but are not limited to, micelles, liposomes, microemulsions, nanoemulsions, vesicles, tubular micelles, cylindrical micelles, bilayers, folded sheets structures, globular aggregates, swollen micelles, inclusion complex, encapsulated droplets, microcapsules, nanocapsules or the like. As will be understood by those having skill in the art, the present compounds can be located inside the nanoparticle or microparticle, within a membrane or wall of the nanoparticle or microparticle, or outside of (but bonded to or otherwise associated with) the nanoparticle or microparticle. The agent formulated in nanoparticles or microparticles can be administered by any of the routes previously described. In a formulation applied topically, the compound is slowly released over time. In an injectable formulation, the liposome, micelle, capsule, etc., circulates in the bloodstream and is delivered to the desired site (e.g., target tissue).
[0288] Preparation and loading of nanoparticles and microparticles are well known in the art. As one example, liposomes can be prepared from dipalmitoyl phosphatidylcholine (DPPC) or egg phosphatidylcholine (PC) because this lipid has a low heat transition. Liposomes are made using standard procedures as known to one skilled in the art (e.g., Braun-Falco et al., (Eds.), Griesbach Conference, Liposome Dermatics, Springer- Verlag, Berlin (1992), pp. 69 81 ; 91 117.
Polycaprolactone, poly(glycolic) acid, poly(lactic) acid, polyanhydride or lipids can be formulated as microspheres. As an illustrative example, the present compounds can be mixed with polyvinyl alcohol (PVA), the mixture then dried and coated with ethylene vinyl acetate, then cooled again with PVA. In a liposome, the present compounds can be within one or both lipid bilayers, in the aqueous between the bilayers, or within the center or core. Liposomes can be modified with other molecules and lipids to form a cationic liposome. Liposomes can also be modified with lipids to render their surface more hydrophilic which increases their circulation time in the bloodstream. The thus-modified liposome has been termed a "stealth" liposome, or a long-lived liposome, as described in U.S. Pat. No. 6,258,378, and in Stealth Liposomes, Lasic and Martin (Eds.) 1995 CRC Press, London. Encapsulation methods include detergent dialysis, freeze drying, film forming, injection, as known to one skilled in the art and disclosed in, for example, U.S. Pat. No. 6,406,713. Optionally, the present compositions and methods include a micelle delivery system, for example, involving one or more PEG-based amphiphilic polymers developed for drug delivery including: PEG-poly(s-caprolactone), PEG-poly(amino acid), PEG-polylactide or PEG- phospholipid constructs; a cross linked poly(acrylic acid) polymer system, a phospholipid-based system and/or block copolymer systems comprising one or more of the following polymer blocks: a poly(lactic acid) polymer block; a poly(propylene glycol) polymer block; a poly(amino acid) polymer block; a poly(ester) polymer block; a poly (ε-caprolactone) polymer block; a poly(ethylene glycol) block, a poly(acrylic acid) block; a polylactide block; a polyester block; a polyamide block; a polyanhydride block; a polyurethane block; a polyimine block; a polyurea block; a polyacetal block; a polysaccharide block; and a polysiloxane block.
[0289] Suitable pharmaceutically-acceptable nonaqueous solvents include, but are not limited to, the following (as well as mixtures thereof):
[0290] (i) Alcohols (these include, for example, σ-glycerol formal, β-glycerol formal, 1 , 3- butyleneglycol, aliphatic or aromatic alcohols having from 2 to about 30 carbons (e.g., methanol, ethanol, propanol, isopropanol, butanol, t-butanol, hexanol, octanol, amylene hydrate, benzyl alcohol, glycerin (glycerol), glycol, hexylene, glycol, tetrahydrofuranyl alcohol, cetyl alcohol, and stearyl alcohol), fatty acid esters of fatty alcohols (e.g., polyalkylene glycols, such as
polypropylene glycol and polyethylene glycol), sorbitan, sucrose, and cholesterol);
[0291] (ii) Amides, which include, for example, dimethylacetamide (DMA), benzyl benzoate DMA, dimethylformamide, N-hydroxyethyO-lactamide, N, N-dimethylacetamide-amides, 2- pyrrolidinone, 1-methyl-2-pyrrolidinone, and polyvinylpyrrolidone;
[0292] (iii) Esters, which include, for example, acetate esters (e.g., monoacetin, diacetin, and triacetin), aliphatic and aromatic esters (e.g., ethyl caprylate or octanoate, alkyl oleate, benzyl benzoate, or benzyl acetate), dimethylsulfoxide (DMSO), esters of glycerin (e.g., mono, di, and tri- glyceryl citrates and tartrates), ethyl benzoate, ethyl acetate, ethyl carbonate, ethyl lactate, ethyl oleate, fatty acid esters of sorbitan, glyceryl monostearate, glyceride esters (e.g., mono, di, or triglycerides), fatty acid esters (e.g., isopropyl myristrate), fatty acid derived PEG esters (e.g., PEG- hydroxyoleate and PEG-hydroxystearate), N-methyl pyrrolidinone, pluronic 60, polyoxyethylene sorbitol oleic polyesters (e.g., poly(ethoxylated)3o-6o sorbitol poly(oleate)2-4, poly(oxyethylene) 5_2o monooleate, poly(oxyethylene)15.20 mono 12-hydroxystearate, and polyoxyethylene)! 5.20 mono ricinoleate), polyoxyethylene sorbitan esters (e.g., polyoxyethylene-sorbitan monooleate, polyoxyethylene-sorbitan monopalmitate, polyoxyethylene-sorbitan monolaurate, polyoxyethylene- sorbitan monostearate, and POLYSORBATE 20, 40, 60, and 80 (from ICI Americas, Wilmington, DE)), polyvinylpyrrolidone, alkyleneoxy modified fatty acid esters (e.g., polyoxyl 40 hydrogenated castor oil and polyoxyethylated castor oils, such as CREMOPHOR EL solution or CREMOPHOR RH 40 solution), saccharide fatty acid esters (i.e., the condensation product of a monosaccharide (e.g., pentoses, such as, ribose, ribulose, arabinose, xylose, lyxose, and xylulose; hexoses, such as glucose, fructose, galactose, mannose, and sorbose; trioses; tetroses; heptoses; and octoses), disaccharide (e.g., sucrose, maltose, lactose, and trehalose), oligosaccharide, or a mixture thereof with one or more C4-C22 fatty acids (e.g., saturated fatty acids, such as caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, and stearic acid; and unsaturated fatty acids, such as palmitoleic acid, oleic acid, elaidic acid, erucic acid, and linoleic acid), and steroidal esters;
[0293] (iv) Ethers, for example, alkyl, aryl, and cyclic ethers having from 2 to about 30 carbons. Examples include diethyl ether, tetrahydrofuran, dimethyl isosorbide, diethylene glycol monoethyl ether), and glycofurol (tetrahydrofurfuranyl alcohol polyethylene glycol ether);
[0294] (v) Ketones which typically have from about 3 to about 30 carbons. Examples include acetone, methyl ethyl ketone, and methyl isobutyl ketone;
[0295] (vi) Hydrocarbons which are typically aliphatic, cycloaliphatic, or aromatic hydrocarbons having from about 4 to about 30 carbons. Examples include benzene, cyclohexane,
dichloromethane, dioxolanes, hexane, n-decane, n-dodecane, n-hexane, sulfolane,
tetramethylenesulfone, tetramethylenesulfoxide, toluene, dimethylsulfoxide (DMSO); and tetramethylene sulfoxide;
[0296] (vii) Oils which include, for example, oils of mineral, vegetable, animal, essential, or synthetic origin. These include: mineral oils, such as aliphatic and wax-based hydrocarbons, aromatic hydrocarbons, mixed aliphatic and aromatic based hydrocarbons, and refined paraffin oil; vegetable oils, such as linseed, tung, safflower, soybean, castor, cottonseed, groundnut, rapeseed, coconut, palm, olive, corn, corn germ, sesame, persic, and peanut oil; glycerides, such as mono-, di-, and triglycerides; animal oils, such as fish, marine, sperm, cod-liver, haliver, squaiene, squalane, and shark liver oil; oleic oils; and polyoxyethylated castor oil;
[0297] (viii) Alkyl, alkenyl, or aryl halides which include, for example, alkyl or aryl halides having from 1 to about 30 carbons and one or more halogen substituents. Examples include: methylene chloride; monoethanolamine; petroleum benzin; trolamine; omega-3 polyunsaturated fatty acids (e.g., alpha-linolenic acid, eicosapentaenoic acid, docosapentaenoic acid, or docosahexaenoic acid); polyglycol ester of 12-hydroxystearic acid and polyethylene glycol (SOLUTOL HS-15, from BASF, Ludwigshafen, Germany); polyoxyethylene glycerol; sodium laurate; sodium oleate; and sorbitan monooleate.
[0298] Other pharmaceutically acceptable solvents for use in the invention are well known to those of ordinary skill in the art. General discussion relating to such solvents can be found in, for example, The Chemotherapy Source Book (Williams & Wilkens Publishing), The Handbook of Pharmaceutical Excipients, (American Pharmaceutical Association, Washington, D.C., and The Pharmaceutical Society of Great Britain, London, England, 1968), Modem Pharmaceutics 3d ed., (G. Banker et. al., eds., Marcel Dekker, Inc., New York, New York (1995)), The Pharmacological Basis of Therapeutics, (Goodman & Gilman, McGraw Hill Publishing), Pharmaceutical Dosage Forms, (H. Lieberman et. al., eds., Marcel Dekker, Inc., New York, New York (1980)), Remington's Pharmaceutical Sciences, 19th ed., (A. Gennaro, ed., Mack Publishing, Easton, PA, (1995)), The United States Pharmacopeia 24, The National Formulary 19, (National Publishing, Philadelphia, PA (2000)); Spiegel, A. J., et al., "Use of Nonaqueous Solvents in Parenteral Products," J. Pharma. Sciences, Vol. 52, No. 10, pp. 917-927 (1963).
[0299] Solvents useful in the invention include, but are not limited to, those known to stabilize present compounds or pharmaceutically acceptable salts thereof. These can include, for example, oils rich in triglycerides, such as safflower oil, soybean oil, and mixtures thereof; and alkyleneoxy- modified fatty acid esters, such as polyoxyl 40 hydrogenated castor oil and polyoxyethylated castor oils (e.g., CREMOPHOR EL solution or CREMOPHOR RH 40 solution). Commercially available triglycerides include INTRALIPID emulsified soybean oil (Kabi-Pharmacia Inc.,
Stockholm, Sweden), NUTRALIPID emulsion (McGaw, Irvine, California), LIPOSYN II 20% emulsion (a 20% fat emulsion solution containing 100 mg safflower oil, 100 mg soybean oil, 12 mg egg phosphatides, and 25 mg glycerin per ml of solution; Abbott Laboratories, Chicago, IL), LIPOSYN III 2% emulsion (a 2% fat emulsion solution containing 100 mg safflower oil, 100 mg soybean oil, 12 mg egg phosphatides, and 25 mg glycerin per ml of solution; Abbott Laboratories, Chicago, IL), natural or synthetic glycerol derivatives containing the docosahexaenoyl group at levels of from about 25 to about 100% (by weight based on the total fatty acid content) (DHASCO from Martek Biosciences Corp., Columbia, MD; DHA MAGURO from Daito Enterprises, Los Angeles, CA; SOYACAL; and TRAVEMULSION). Ethanol in particular is a useful solvent for dissolving a compound or pharmaceutically acceptable salt thereof to form solutions, emulsions, and the like.
[0300] Additional components can be included in the compositions of this invention for various purposes generally known in the pharmaceutical industry. These components tend to impart properties that, for example, enhance retention of the present compounds or salt thereof at the site of administration, protect the stability of the composition, control the pH, and facilitate processing of the compound or salt thereof into pharmaceutical formulations, and the like. Specific examples of such components include cryoprotective agents; agents for preventing reprecipitation of the compound or salt surface; active, wetting, or emulsifying agents (e.g., lecithin, polysorbate-80, TWEEN 80, pluronic 60, and polyoxyethylene stearate); preservatives (e.g., ethyl-p- hydroxybenzoate); microbial preservatives (e.g., benzyl alcohol, phenol, m-cresol, chlorobutanol, sorbic acid, thimerosal, and paraben); agents for adjusting pH or buffering agents (e.g., acids, bases, sodium acetate, sorbitan monolaurate, etc.); agents for adjusting osmolarity (e.g., glycerin); thickeners (e.g., aluminum monostearate, stearic acid, cetyl alcohol, stearyl alcohol, guar gum, methyl cellulose, hydroxypropylcellulose, tristearin, cetyl wax esters, polyethylene glycol, etc.); colorants; dyes; flow aids; non-volatile silicones (e.g., cyclomethicone); clays (e.g., bentonites); adhesives; bulking agents; flavorings; sweeteners; adsorbents; fillers (e.g., sugars such as lactose, sucrose, mannitol, sorbitol, cellulose, calcium phosphate, etc.); diluents (e.g., water, saline, electrolyte solutions, etc.); binders (e.g., gelatin; gum tragacanth; methyl cellulose;
hydroxypropyl methylcellulose; sodium carboxymethyl cellulose; polyvinylpyrrolidone; sugars; polymers; acacia; starches, such as maize starch, wheat starch, rice starch, and potato starch; etc.); disintegrating agents (e.g., starches, such as maize starch, wheat starch, rice starch, potato starch, and carboxymethyl starch; cross-linked polyvinyl pyrrolidone; agar; alginic acid or a salt thereof, such as sodium alginate; croscarmellose sodium; crospovidone; etc); lubricants (e.g., silica; talc; stearic acid and salts thereof, such as magnesium stearate; polyethylene glycol; etc.); coating agents (e.g., concentrated sugar solutions including gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, etc.); and antioxidants (e.g., sodium
metabisulfite, sodium bisulfite, sodium sulfite, dextrose, phenols, thiophenols, etc.).
[0301] Techniques and compositions for making parenteral dosage forms are generally known in the art. Formulations for parenteral administration can be prepared from one or more sterile powders and/or granules having a compound or salt of this invention and one or more of the carriers or diluents mentioned for use in the formulations for oral administration. The powder or granule typically is added to an appropriate volume of a solvent (typically while agitating (e.g., stirring) the solvent) that is capable of dissolving the powder or granule. Particular solvents useful in the invention include, for example, water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers.
[0302] Emulsions for parenteral administration can be prepared by, for example, dissolving a compound or salt of this invention in any pharmaceutically acceptable solvent capable of dissolving the compound to form a solution; and adding an appropriate volume of a carrier to the solution while stirring to form the emulsion. Solutions for parenteral administration can be prepared by, for example, dissolving a compound or salt of this invention in any pharmaceutically acceptable solvent capable of dissolving the compound to form a solution; and adding an appropriate volume of a carrier to the solution while stirring to form the solution.
[0303] Suppositories for rectal administration can be prepared by, for example, mixing the drug with a suitable nonirritating excipient that is solid at ordinary temperatures, but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Suitable excipients include, for example, cocoa butter; synthetic mono-, di-, or triglycerides; fatty acids; and/or polyethylene glycols.
[0304] Every formulation or combination of components described or exemplified herein can be used to practice the invention, unless otherwise stated,
(i) Binding Agents
[0305] Binding agents include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof. Suitable forms of
microcrystalline cellulose include, for example, the materials sold as AVICEL-PH-101 , AVICEL- PH-103 and AVICEL-PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pennsylvania, USA). An exemplary suitable binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581 by FMC Corporation.
(ii) Fillers
[0306] Fillers include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), lactose, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
(iii) Lubricants
[0307] Lubricants include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, electromagnetic radiation mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laurate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, Maryland, USA), a coagulated aerosol of synthetic silica (marketed by Deaussa Co. of Piano, Texas, USA), CAB-O- SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Massachusetts, USA), and mixtures thereof.
(iv) Disintegrants
[0308] Disintegrants include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
[0309] Tablets or capsules can optionally be coated by methods well known in the art. If binders and/or fillers are used with a compound/bioconjugate of the invention, they are typically formulated as about 50 to about 99 weight percent of the compound/bioconjugate. In one aspect, about 0.5 to about 15 weight percent of disintegrant, and particularly about 1 to about 5 weight percent of disintegrant, can be used in combination with the compound. A lubricant can optionally be added, typically in an amount of less than about 1 weight percent of the compound/bioconjugate.
Techniques and pharmaceutically acceptable additives for making solid oral dosage forms are described in Marshall, SOLID ORAL DOSAGE FORMS, Modern Pharmaceutics (Banker and Rhodes, Eds.), 7:359-427 (1979). Other formulations are known in the art.
[0310] Liquid preparations for oral administration can take the form of solutions, syrups or suspensions. Alternatively, the liquid preparations can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and/or preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations can also contain buffer salts, flavoring, coloring, perfuming and sweetening agents as appropriate. Preparations for oral administration can also be formulated to achieve controlled release of the compound/bioconjugate. Oral formulations preferably contain 10% to 95% compound/bioconjugate. In addition, a compound/bioconjugate of the present invention can be formulated for buccal administration in the form of tablets or lozenges formulated in a conventional manner. Other methods of oral delivery of compounds/bioconjugates of the invention will be known to the skilled artisan and are within the scope of the invention.
Formulation 1
[0311] Hard gelatin capsules are prepared using the following ingredients:
TABLE F1
Ingredients (mg/capsule)
Active Ingredient 250.0
Starch 305.0
Magnesium stearate 5.0
[0312] The above ingredients are mixed and filled into hard gelatin capsules in 560 mg quantities.
Formulation 2
[0313] A tablet formula is prepared using the following ingredients:
TABLE F2
Ingredients (mg/tablet)
Active Ingredient 250.0
Cellulose, microcrystalline 400.0
Colloidal silicon dioxide 10.0
Stearic acid 5.0
[0314] The components are blended and compressed to form tablets, each weighing 665 mg. Formulation 3
[0315] A dry powder inhaler formulation is prepared containing the following components:
TABLE F3
Ingredients Weight %
Active ingredient 5
Lactose 95 [0316] The active ingredient is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
Formulation 4
[0317] Tablets, each containing 60 mg of active ingredient, are prepared as follows:
TABLE F4
Ingredients Milligrams
Active ingredient 60.0
Starch 45.0
Microcrystalline cellulose 35.0
Polyvinylpyrrolidone (as 10% solution in water) 4.0
Sodium carboxymethyl starch 4.5
Magnesium stearate 0.5
Talc 1.0
Total 150.0
[0318] The active ingredient, starch and cellulose are passed through a No. 20 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with the resultant powders which are then passed through a 16 mesh U.S. sieve. The granules as produced are dried at 50- 60 °C and passed through a 16 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 30 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
Formulation 5
[0319] Capsules, each containing 80 mg of active ingredient are made as follows:
TABLE F5
Ingredients Milligrams
Active ingredient 80.0
Starch 109.0
Magnesium stearate 1.0
Total 190.0
[0320] The active ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 190 mg quantities. Formulation 6
[0321] Suppositories, each containing 225 mg of active ingredient, are made as follows:
TABLE F6
I Ingredients Milligrams I Active Ingredient 225
Saturated fatty acid glycerides to 2000
[0322] The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed to cool.
Formulation 7
[0323] Suspensions, each containing 50 mg of active ingredient per 5.0 ml dose are made as follows:
TABLE F7
Ingredients Milligrams
Active ingredient 50.0 mg
Xanthan gum 4.0 mg
Sodium carboxymethyl cellulose (11 %)
Microcrystalline cellulose (89%) 50.0 mg
Sucrose 1.75 g
Sodium benzoate 10.0 mg
Flavor q.v.
Color q.v.
Purified water to 5.0 ml
[0324] The active ingredient, sucrose and xantham gum are blended, passed through a No. 10 mesh U.S. sieve, and mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water. The sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
Formulation 8
[0325] Capsules, each containing 150 mg of active ingredient, are made as follows:
TABLE F8
Ingredients Milligrams
Active ingredient 150.0
Starch 407.0
Magnesium stearate 3.0
Total 560.0
[0326] The active ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 560 mg quantities. 4.e: Kits [0327] Various embodiments of the present invention include kits. Such kits can include a compound/bioconjugate of the present invention, optionally one or more ingredients for preparing a pharmaceutically acceptable formulation of the compound/bioconjugate, and instructions for use (e.g., administration). When supplied as a kit, different components of a compound/bioconjugate formulation can be packaged in separate containers and admixed immediately before use. Such packaging of the components separately can, if desired, be presented in a pack or dispenser device which can contain one or more unit dosage forms containing the compound/bioconjugate. The pack can, for example, comprise metal or plastic foil such as a blister pack. Such packaging of the components separately can also, in certain instances, permit long-term storage without losing activity of the components. In addition, if more than one route of administration is intended or more than one schedule for administration is intended, the different components can be packaged separately and not mixed prior to use. In various embodiments, the different components can be packaged in one combination for administration together.
[0328] It is further contemplated that the compounds and salts of this invention can be used in the form of a kit that is suitable for use in. performing the methods described herein, packaged in a container. The kit can contain the compound or compounds and, optionally, appropriate diluents, devices or device components suitable for administration and instructions for use in accordance with the methods of the invention. The devices can include parenteral injection devices, such as syringes or transdermal patch or the like. Device components can include cartridges for use in injection devices and the like. In one aspect, the kit includes a first dosage form including a compound or salt of this invention and a second dosage form including another active ingredient in quantities sufficient to carry out the methods of the invention. The first dosage form and the second dosage form together can include a therapeutically effective amount of the compounds for treating the targeted condition(s).
[0329] In certain embodiments, kits can be supplied with instructional materials. Instructions can be printed on paper or other substrate, and/or can be supplied as an electronic-readable medium, such as a floppy disc, mini-CD-ROM, CD-ROM, DVD-ROM, Zip disc, videotape, audio tape, and the like. Detailed instructions cannot be physically associated with the kit; instead, a user can be directed to an Internet web site specified by the manufacturer or distributor of the kit, or supplied as electronic mail.
[0330] If desired, the emulsions or solutions described above for oral or parenteral
administration can be packaged in IV bags, vials, or other conventional containers in concentrated form, and then diluted with a pharmaceutically acceptable liquid (e.g., saline) to form an acceptable compound concentration before use.
[0331] Kits can include reagents in separate containers such as, for example, sterile water or saline to be added to a lyophilized active component packaged separately. For example, sealed glass ampules can contain lyophilized superoxide dismutase mimetics and in a separate ampule, sterile water, sterile saline or sterile each of which has been packaged under a neutral non- reacting gas, such as nitrogen. Ampules can consist of any suitable material, such as glass, organic polymers, such as polycarbonate, polystyrene, ceramic, metal or any other material typically employed to hold reagents. Other examples of suitable containers include bottles that can be fabricated from similar substances as ampules, and envelopes that can consist of foil-lined interiors, such as aluminum or an alloy. Other containers include test tubes, vials, flasks, bottles, syringes, and the like. Containers can have a sterile access port, such as a bottle having a stopper that can be pierced by a hypodermic injection needle. Other containers can have two
compartments that are separated by a readily removable membrane that upon removal permits the components to mix. Removable membranes can be glass, plastic, rubber, and the like.
STATEMENTS REGARDING INCORPORATION BY REFERENCE AND VARIATIONS
[0332] All references cited throughout this application, for example patent documents including issued or granted patents or equivalents; patent application publications; and non-patent literature documents or other source material; are hereby incorporated by reference herein in their entireties, as though individually incorporated by reference, to the extent each reference is at least partially not inconsistent with the disclosure in this application (for example, a reference that is partially inconsistent is incorporated by reference except for the partially inconsistent portion of the reference).
[0333] The terms and expressions which have been employed herein are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments, exemplary embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims. The specific embodiments provided herein are examples of useful embodiments of the present invention and it will be apparent to one skilled in the art that the present invention may be carried out using a large number of variations of the devices, device components, methods steps set forth in the present description. As will be obvious to one of skill in the art, methods and devices useful for the present methods can include a large number of optional composition and processing elements and steps.
[0334] When a group of substituents is disclosed herein, it is understood that all individual members of that group and all subgroups, including any isomers, enantiomers, and diastereomers of the group members, are disclosed separately. When a Markush group or other grouping is used herein, all individual members of the group and all combinations and subcombinations possible of the group are intended to be individually included in the disclosure. When a compound is described herein such that a particular isomer, enantiomer or diastereomer of the compound is not specified, for example, in a formula or in a chemical name, that description is intended to include each isomers and enantiomer of the compound described individual or in any combination. Additionally, unless otherwise specified, all isotopic variants of compounds disclosed herein are intended to be encompassed by the disclosure. For example, it will be understood that any one or more hydrogens in a molecule disclosed can be replaced with deuterium or tritium. Isotopic variants of a molecule are generally useful as standards in assays for the molecule and in chemical and biological research related to the molecule or its use. Methods for making such isotopic variants are known in the art. Specific names of compounds are intended to be exemplary, as it is known that one of ordinary skill in the art can name the same compounds differently.
[0335] Optical agents of the present invention may be formulated with pharmaceutically- acceptable anions and/or cations. Pharmaceutically-acceptable cations include among others, alkali metal cations (e.g., Li+, Na+, K+), alkaline earth metal cations (e.g., Ca2+, Mg2+), non-toxic heavy metal cations and ammonium (NH4 +) and substituted ammonium (N(R')4 +, where R' is hydrogen, alkyl, or substituted alkyl, i.e., including, methyl, ethyl, or hydroxyethyl, specifically, trimethyl ammonium, triethyl ammonium, and triethanol ammonium cations). Pharmaceutically- acceptable anions include among other halides (e.g., CI", Br"), sulfate, acetates (e.g., acetate, trifluoroacetate), ascorbates, aspartates, benzoates, citrates, and lactate.
[0336] It must be noted that as used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to "a cell" includes a plurality of such cells and equivalents thereof known to those skilled in the art, and so forth. As well, the terms "a" (or "an"), "one or more" and "at least one" can be used interchangeably herein. It is also to be noted that the terms "comprising", "including", and "having" can be used interchangeably. The expression "of any of claims XX- YY" (wherein XX and YY refer to claim numbers) is intended to provide a multiple dependent claim in the alternative form, and in some embodiments is interchangeable with the expression "as in any one of claims XX- YY."
[0337] Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
[0338] In some embodiments, a liposome or micelle may be utilized as a carrier or vehicle for the composition. For example, in some embodiments, the sulfenamide compound may be a part of the lipophilic bilayers or micelle, and the targeting ligand, if present, may be on the external surface of the liposome or micelle. As another example, a targeting ligand may be externally attached to the liposome or micelle after formulation for targeting the liposome or micelle (which contains the sulfenamide optical agents) to the desired tissue, organ, or other site in the body.
[0339] Every formulation or combination of components described or exemplified herein can be used to practice the invention, unless otherwise stated. [0340] The present compositions, preparations and formulations can be used both as a diagnostic agent as well as a phototherapy agent concomitantly. For example, an effective amount of the present compositions, preparations and formulations in a pharmaceutically acceptable formulation is administered to a patient. Administration is followed by a procedure that combines photodiagnosis and phototherapy. For example, a composition comprising compounds for combined photodiagnosis and phototherapy is administered to a patient and its concentration, localization, or other parameters is determined at the target site of interest. More than one measurement may be taken to determine the location of the target site. The time it takes for the compound to accumulate at the target site depends upon factors such as pharmcokinetics, and may range from about thirty minutes to two days. Once the site is identified, the phototherapeutic part of the procedure may be done either immediately after determining the site or before the agent is cleared from the site. Clearance depends upon factors such as pharmacokinetics.
[0341] The present compositions, preparations and formulations can be formulated into diagnostic or therapeutic compositions for enteral, parenteral, topical, aerosol, inhalation, or cutaneous administration. Topical or cutaneous delivery of the compositions, preparations and formulations may also include aerosol formulation, creams, gels, solutions, etc. The present compositions, preparations and formulations are administered in doses effective to achieve the desired diagnostic and/or therapeutic effect. Such doses may vary widely depending upon the particular compositions employed in the composition, the organs or tissues to be examined, the equipment employed in the clinical procedure, the efficacy of the treatment achieved, and the like. These compositions, preparations and formulations contain an effective amount of the
composition(s), along with conventional pharmaceutical carriers and excipients appropriate for the type of administration contemplated. These compositions, preparations and formulations may also optionally include stabilizing agents and skin penetration enhancing agents.
[0342] Methods of this invention comprise the step of administering an "effective amount" of the present diagnostic and therapeutic compositions, formulations and preparations containing the present compounds, to diagnosis, image, monitor, evaluate, treat, reduce, alleviate, ameliorate or regulate a biological condition and/or disease state in a patient.. The term "effective amount," as used herein, refers to the amount of the diagnostic and therapeutic formulation, that, when administered to the individual is effective diagnosis, image, monitor, evaluate, treat, reduce alleviate, ameliorate or regulate a biological condition and/or disease state. As is understood in the art, the effective amount of a given composition or formulation will depend at least in part upon, the mode of administration (e.g. intravenous, oral, topical administration), any carrier or vehicle employed, and the specific individual to whom the formulation is to be administered (age, weight, condition, sex, etc.). The dosage requirements needed to achieve the "effective amount" vary with the particular formulations employed, the route of administration, and clinical objectives. Based on the results obtained in standard pharmacological test procedures, projected daily dosages of active compound can be determined as is understood in the art. [0343] Any suitable form of administration can be employed in connection with the diagnostic and therapeutic formulations of the present invention. The diagnostic and therapeutic formulations of this invention can be administered intravenously, in oral dosage forms, intraperitoneally, subcutaneously, or intramuscularly, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
[0344] The diagnostic and therapeutic formulations of this invention can be administered alone, but may be administered with a pharmaceutical carrier selected upon the basis of the chosen route of administration and standard pharmaceutical practice.
[0345] The diagnostic and therapeutic formulations of this invention and medicaments of this invention may further comprise one or more pharmaceutically acceptable carrier, excipient, buffer, emulsifier, surfactant, electrolyte or diluent. Such compositions and medicaments are prepared in accordance with acceptable pharmaceutical procedures, such as, for example, those described in Remingtons Pharmaceutical Sciences, 17th edition, ed. Alfonoso R. Gennaro, Mack Publishing Company, Easton, Pa. (1985).
[0346] Whenever a range is given in the specification, for example, a temperature range, a time range, or a composition or concentration range, all intermediate ranges and subranges, as well as all individual values included in the ranges given are intended to be included in the disclosure. As used herein, ranges specifically include the values provided as endpoint values of the range. For example, a range of 1 to 100 specifically includes the end point values of 1 and 100. It will be understood that any subranges or individual values in a range or subrange that are included in the description herein can be excluded from the claims herein.
[0347] As used herein, "comprising" is synonymous with "including," "containing," or
"characterized by," and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps. As used herein, "consisting of excludes any element, step, or ingredient not specified in the claim element. As used herein, "consisting essentially of does not exclude materials or steps that do not materially affect the basic and novel characteristics of the claim. In each instance herein any of the terms "comprising", "consisting essentially of and "consisting of may be replaced with either of the other two terms. The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein.
[0348] One of ordinary skill in the art will appreciate that starting materials, biological materials, reagents, synthetic methods, purification methods, analytical methods, assay methods, and biological methods other than those specifically exemplified can be employed in the practice of the invention without resort to undue experimentation. All art-known functional equivalents, of any such materials and methods are intended to be included in this invention. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various
modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred ennbodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.

Claims

What is claimed is:
cedure, the compound being of the formula (FX1 ):
Figure imgf000121_0001
FX1 ), wherein:
each of L , L2, and L3, if present, is independently C C 0 alkylene, C3-C10 cycloalkylene, C2-C-10 alkenylene, C3-C10 cycloalkenylene, C2-C 0 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, -(CH2CH20) -, or -(CHOH)a-;
each of W , W2, and W3 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -SO2-, -SO3-, -OSO2-, -NR 1-, -CO-, -COO- -OCO-, -OCOO-, -CONR12-, - NR13CO-, -OCONR14-, -NR15COO-, -NR 6CONR17-, -NR 8CSNR19-, -0(CH2)n-, -S(CH2)n- - NR20(CH2)n-, -C0(CH2)n-, -COO(CH2)n-, -OCO(CH2)n- -OCOO(CH2)n-, -CONR21(CH2)n-, - CONR2 (CH2)n-, -NR23CO(CH2)n- -OCONR24(CH2)n- -NR25COO(CH2)n-, -NR26CONR27(CH2)n- , -NR28CSNR 9(CH2)n- -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR3 (CH2)nNR32CO-, or -CO(CH2)nNR33CO-;
each of R1, R2, and R3 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C-|-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR 8R49, - NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
or wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form one to five substituted or unsubstituted, carbocyclic or heterocyclic 4, 5, 6, 7, 8 or 9 membered alicyclic or aromatic rings;
wherein at least one of R , R2, and R3 is a C5-C30 aryl or C5-C30 heteroaryl, or wherein R2, R3, W2, W3, and L2 and L3, if present, together with the atoms to which they are attached combine to form a C5-C30 aryl or C5-C30 heteroaryl;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of e, f, and g is independently 0 or 1 ;
each of R11 - R33 is independently hydrogen, C C2o alkyl, C3-C2o cycloalkyl, C5-C20 heteroaryl, or C5-C2o aryl;
each of R 0-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid;
each FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenyixanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steriod, a hormone, an aptamer, a receptor, a metal chelating agent, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
2. The compound of claim 1 , wherein the compound undergoes cleavage of a S-N bond upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers.
3. The compound of any of claims 1-2, wherein R1 is C5-C30 aryl or C5-C3o heteroaryl, and each of R2 and R3 is independently hydrogen, C C20 alkyl, C3-C2o cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, FL, or Bm; or wherein R2 is C5-C30 aryl or C5-C30 heteroaryl, and each of R and R3 is independently hydrogen, C C2o alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, FL, or Bm; or wherein R3 is C5-C30 aryl or C5-C30 heteroaryl, and each of R and R2 is independently hydrogen, C C2o alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, FL, or Bm.
4. The compound of any of claims 1-3, wherein at least one of R , R2 and R3 is independently a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline.
5. The compound of any of claims 1-4, wherein at least one of R1, R2 and R3 is a C5-C30 aryl or C5- C30 heteroaryl comprising one or more aromatic rings or heterocyclic aromatic rings having at least one electron donating group as a substituent, or having at least one electron withdrawing group as a substituent, or having at least one electron withdrawing group and at least one electron donating group as substituents.
6. The compound of any of claims 1-5, wherein at least one of R1, R2 and R3 is Bm.
7. The compound of any of claims 1-6, wherein at least one of R1 , R2 and R3 is a dye that is excited upon exposure to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers.
8. The compound of any of claims 1 -7, wherein at least one of R1 , R2 and R3 is a group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a benzoindocarbocyanine.
9. The compound of any of claims 1-8, wherein at least one of R , R2 and R3 is a group corresponding to a pyrazine, azulene or aza-azulene having at least one electron donating group provided as a substituent and at least one electron withdrawing group provided as a substituent.
10. The compound of any of claims 1-9, wherein e is 0 and W1 is a single bond; or wherein f is 0 and W2 is a single bond; or wherein g is 0 and W3 is a single bond.
1 1. The compound of any of claims 1-9, wherein at least one of: L1 with W1, L2 with W2, and L3 with W3, combine to form: -(CH2)n-, -0(CH2)n- -CO(CH2)n- -OCO(CH2)n-, -COO(CH2)n- - OCOO(CH2)n- -N(R20)(CH2)n-, -CON(R22)(CH2)n- -N(R23)CO(CH2)n- -OCONR2 (CH2)n- - NR25COO(CH2)n- -NR26CONR27(CH2)n-, or -NR28CSNR29(CH2)n-.
12. The compound of claim 1 being of the formula (FX2), (FX3), (FX4), (FX5), (FX6), (FX7) or (FX
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000124_0002
group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline.
13. The compound of claim 1 being of the formula (FX9), (FX10), (FX11 ), (FX12), (FX13), (FX14)
Figure imgf000124_0003
Ar3
\ S N /
\ Ar2 (FX15); wherein each of Ar Ar2 and Ar3 is independently a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline. of the formula (FX16):
Figure imgf000125_0001
(FX16); wherein:
X is independently -CR R -, -NR -, -0-, -S-, -SO-, or -S02-;
each of Y1 and Y2 is independently -CR65R66-;
each of R65 , R66 and R67 is independently a hydrogen, -OCF3, C^-C20 alkyi, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C^-C2o acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, Ci-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42, -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm; and
each of i and h is independently an integer selected from the range of 0 to 6, wherein at least one of i and h is greater than or equal to 1. ula (FX17):
Figure imgf000125_0002
16. The compound of any of claims 14-15, wherein R1 is C5-C30 aryl or C5-C30 heteroaryl.
17. The compound of any of claims 14-16, wherein at least one of R and R65-R67 is independently a group corresponding to benzene, naphthalene, naphthoquinone, diphenylmethane, fluorene, anthracene, anthraquinone, phenanthrene, tetracene, naphthacenedione, pyridine, quinoline, isoquinoline, indole, isoindole, pyrrole, imidazole, oxazole, thiazole, pyrazole, pyrazine, pyrimidine, purine, benzimidazole, furan, benzofuran, dibenzofuran, carbazole, acridine, acridone, phenanthridine, thiophene, benzothiophene, dibenzothiophene, xanthene, xanthone, flavone, coumarin, azulene, aza-azulene or anthracycline.
18. The compound of any of claims 14-17, wherein at least one of R1 and R65-R67 is independently Bm.
19. The compound of any of claims 14-18, wherein the sum of i and h is 2, 3 or 4.
20. The compound of claim 14, wherein X is -CR65R66-.
21. The compound of claim 14, wherein X is -0-.
22. The compound of claim 14, wherein X is -S-.
23. The compound of any of claims 14-22, wherein L with W1 independently combine to form: - (CH2)n- -0(CH2)n- -CO(CH2)n- -OCO(CH2)n- -COO(CH2)n- -OCOO(CH2)n- -N(R20)(CH2)n- -CON(R22)(CH2)n- -N(R23)CO(CH2)n- -OCONR 4(CH2)n- -NR25COO(CH2)n- - NR26CONR27(CH2)n- or -NR28CSNR29(CH2)n-.
24. The compound of any of claims 14-22, wherein e is 0 and W1 is a single bond.
25. The compound of any of claims 1-24, wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar3 is independently benzene or a derivative thereof.
26. The compound of any of claims 1 -24, wherein at least one of R - R3, R65 - R67 and Ar1 - Ar3 is independently a group corresponding to naphthalene, diphenylmethane, fluorene, anthracene, phenanthrene, tetracene, naphthoquinone, anthraquinone, naphthacenedione, azulene, aza- azulene or anthracycline.
27. The compound of any of claims 1 -24, wherein at least one of R1 - R3, R65 - RS7 and Ar1 - Ar3 is independently a group corresponding to pyridine, pyrimidine, pyrazine, quinoline, isoquinoline, purine, acridine, acridone, or phenanthridine.
28. The compound of any of claims 1-24, wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar3 is independently a group corresponding to pyrrole, pyrazole, indole, isoindole, imidazole, oxazole, thiazole, purine, benzimidazole, or carbazole.
29. The compound of any of claims 1-24, wherein at least one of R - R3, R65 - R67 and Ar - Ar is independently a group corresponding to furan, oxazole, benzofuran, dibenzofuran, xanthene, xanthone, flavone, or coumarin.
30. The compound of any of claims 1-24, wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar is independently a group corresponding to thiophene, thiazole, benzothiophene, or
dibenzothiophene.
31. The compound of any of claims 1-24, wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar
is independently:
Figure imgf000127_0001
; wherein each of R70 - R74 is independently a hydrogen, -
OCF3, C1-C20 alkyl, C3-C20 cycloalkyl,C5-C20 aryl, C5-C20 heteroaryl, Ci-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C-i-Ce alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, - OR46, -SR47, -NR48R49, -NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, - S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm.
32. The compound of any of claims 1-24, wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar
Figure imgf000127_0002
is independently: ; wherein each of R - R is independently a hydrogen, -
OCF3, C C2o alkyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C20 heteroaryl, C1-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C-|-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, - OR46, -SR47, -NR48R49, -NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, - S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm.
33. The compound of claim 1 being of the formula (FX20):
Figure imgf000127_0003
(FX20); wherein: each of rings A, B, and C are independently
Figure imgf000128_0001
each of R70 - R77 is independently a hydrogen, -OCF3, C C2o alkyl, C3-C2o cycloalkyl, C aryl, C5-C2o heteroaryl, CrC2o acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6
,42 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R , -SOR , -OSR S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, -NR50COR51, CONR52R53, -COR54, -NO2, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61
63\
CH(R6')C02H, -CH(R0J)NH2, FL or Bm
34. The compound of claim 33 being of formula (FX21 ), (FX22), (FX23), (FX24), (FX25) or
Figure imgf000128_0002
35. The compound of any of claims 33-34, wherein each of R - R is independently a hydrogen, -N02, -OCF3, C -C2o alkyl, C3-C20 cycloalkyl, C5-C2o aryl, or C5-C20 heteroaryl.
36. The compound of any of claims 1-24, wherein at least one of R1 - R3, R65 - R67 and Ar1 - Ar3 is independently:
Figure imgf000129_0001
Figure imgf000130_0001
129
Figure imgf000131_0001
; wherein each of R - R is independently a hydrogen, -OCF3, C C2o alkyl, C3-C2o cycloalkyl, C5-C2o aryl, C5-C2o heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl,
40
C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R SOR41, -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR 8R NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm.
The compound of claim 1 , being of the formula (FX27):
Figure imgf000131_0002
(FX27); wherein:
Z is a single bond, -CR128=CR129-, -CR130R131CR132R133-, -CR 3 R1350-, -CR136R137S-,
Figure imgf000131_0003
each of R120 to R141 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C2o acyl, C2-C20 alkenyl, C2-C2o alkynyl, C5-C20 alkylaryl, C^C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, -OSR42 , - S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, -NR50COR51, -CN, - CONR5 R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61, - CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
each of L120, L121 , L122, L123, L124, L125, L126 and L127, if present, is independently C C10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-C 0 cycloalkenylene, C2-C 0 alkynylene, ethenyiene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b- or -(CHOH)a-; and
each of W120, W 21 , W122, W123, W124, W 25, W126 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -S02- -S03-, -OS02- -NR1 -, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR 4-, -NR15COO- -NR16CONR17-, -NR18CSNR19- -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n- -OCO(CH2)n-, - OCOO(CH2)n- -CONR21(CH2)n-, -CONR22(CH2)n- -NR23CO(CH2)n-, -OCONR24(CH2)n-, - NR25COO(CH2)n-, -NR26CONR27(CH2)n-, -NR 8CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR3 (CH2)nNR32CO-, or -CO(CH2)nNR33CO-.
The compound of claim 37 being of the formula (FX28):
Figure imgf000132_0001
39. The compound of claim 37 being any of formula (FX29), (FX30), (FX31), (FX32), (FX33), or (FX34):
Figure imgf000132_0002
,
Figure imgf000133_0001
40. The compound of claim 37 being any of formula (FX35), (FX36), (FX37), or (FX38):
Figure imgf000134_0001
41. The compound of claim 37 being of formula (FX39):
Figure imgf000135_0001
(FX39); wherein:
each of R 44 to R 48 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C2o cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C1-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR4 , -OSR42 , - S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R 4R45, -OR46, -SR47, -NR48R49, -NR50COR51 , -CN, - CONR 2R53, -COR54, -NO2, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, -(CH2CH20)bR61 , - CH(R62)C02H,' -CH(R63)NH2, FL or Bm.
42. The compound of claim 41 being any of formula (FX40), (FX41), (FX42), (FX43), (FX44), or (FX45):
Figure imgf000136_0001

Figure imgf000137_0001
43. The compound of claim 41 being of formula (FX46):
Figure imgf000138_0001
44. A compound for use in a phototherapy procedure, the compound being of the formula (FX18), (FX19), (FX47), (FX48), (FX49), (FX50), (FX51 ), (FX52), or (FX53):
,CONH-Peptide
Figure imgf000138_0002
Figure imgf000138_0003
Figure imgf000139_0001
acid units.
45. The compound of any of claims 1-44, or a pharmaceutical formulation thereof, for use in a medical phototherapy procedure, said procedure comprising:
administering to a subject in need of treatment a therapeutically effective amount of the compound of any of claims 1-44; and
exposing the administered compound to electromagnetic radiation.
46. The compound of claim 45, wherein said procedure is a Type 1 phototherapy procedure.
47. The compound of any of claims 45-46, wherein the procedure comprises exposing the administered compound to electromagnetic radiation having wavelengths selected over the range of 350 nanometers to 1300 nanometers.
48. The compound of any of claims 45-47, wherein exposing the administered compound to electromagnetic radiation generates a therapeutically effective amount of photoactivated compound.
49. The compound of any of claims 45-48, wherein exposing the administered compound to electromagnetic radiation cleaves a S-N bond of the compound.
50. The compound of claim 49, wherein cleavage of the S-N bond of the compound generates free radicals, ions or both causing localized cell death or injury.
51. The compound of any of claims 45-50, wherein exposing the administered compound to electromagnetic radiation generates a therapeutically effective amount of reactive species causing localized cell death or injury.
52. The compound of any of claims 45-51 , wherein the procedure comprises contacting a target tissue of the subject with the administered compound.
53. The compound of claim 52, wherein the target tissue type is colon, prostate, gastric, esophageal, uterine, endometrial, pancreatic, breast, cervical, brain, skin, gallbladder, lung, throat, kidney, testicular, prostate, gastric, or ovary tissue.
54. The compound of claim 52, wherein the target tissue is cancerous tissue.
55. The compound of claim 52, wherein the target tissue is a tumor.
56. The compound of any of claims 1-55 for use in treatment of cancer or a cancer-associated disorder.
57. The compound of claim 56, wherein the cancer or cancer-associated disorder is colon cancer, prostate cancer, gastric cancer, esophageal cancer, uterine cancer, endometrial cancer, pancreatic cancer, breast cancer, cervical cancer, brain cancer, skin cancer, gallbladder cancer, lung cancer, or ovarian cancer.
58. The compound of any of claims 1-57 for use in treatment of inflammation or an inflammation - associated disorder.
59. A pharmaceutical composition comprising:
the compound of any of claims 1-58; and
one or more pharmaceutically acceptable excipients.
60. A pharmaceutical composition comprising:
the compound of any of claims 1-58; and
one or more additional therapeutic agents or diagnostic agents.
Figure imgf000141_0001
(FX55); wherein:
Z is -CR128=CR129-, -CR 34R 350- -CR136R137S- -CR138R139NR140-, or -N=CR141-; each of L120, L121 , L122, L 23, L124, L 25, L126 and L127, if present, is independently C C10 alkylene, C3-C10 cycloalkylene, C2-Ci0 alkenylene, C3-C10 cycloalkenylene, C2-C10 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b-, or -(CHOH)a-;
each of W120, W121 , W122, W123, W 24, W 25, W126 and W 27 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -SO2-, -SO3-, -OS02-, -NR 1-, -CO-, -COO-, -OCO- , -OCOO-, -CONR 2-, -NR13CO-, -OCONR 4-, -NR 5COO- -NR16CONR17-, -NR 8CSNR19-, -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n- -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n-, - OCOO(CH2)n-, -CONR21(CH2)n- -CONR22(CH2)n-, -NR23CO(CH2)n-, -OCONR24(CH2)n-, - NR25COO(CH2)n- -NR26CONR27(CH2)n-, -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR3 CO-, or -CO(CH2)nNR33CO-;
each of R120 to R129, R134 to R141 and R 44 to R148 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C-|-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, - C02R40, -SOR4 , -OSR42 , -SO2OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, - NR48R49, -NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of R11 - R33 is independently hydrogen, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C2o aryl;
each of R40-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C^C^ alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
Figure imgf000142_0001
(FX56); wherein:
each of L120, L121, L 22, L123, L124, L125, L126 and L127, if present, is independently C C10 alkylene, C3-C 0 cycloalkylene, C2-C10 alkenylene, C3-C10 cycloalkenylene, C2-C 0 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5Tdioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b- or -(CHOH)a-;
each of W120, W121 , W122, W123, W124, W125, W126 and W127 is independently a single bond, -(CH2)n-, -(HCCH)n-, -0-, -S-, -SO-, -S02- -S03- -OS02- -NR 1-, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR14-, -NR15COO- -NR 6CONR17-, -NR18CSNR19-, -0(CH2)n-, -S(CH2)n- -NR20(CH2)n- -CO(CH2)n- -COO(CH2)n- -OCO(CH2)n- - OCOO(CH2)n- -CONR (CH2)n- -CONR22(CH2)n- -NR23CO(CH2)n- -OCONR 4(CH2)n- - NR25COO(CH2)n- -NR 6CONR27(CH2)n- -NR28CSNR 9(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-;
each of R120 to R127, R130 to R133 and R144 to R148 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, - C02R , -SOFT, -OSR42 , -S02OR4J, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR4 , -SR4\ - NR48R49, -NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm; wherein when R146 is N02, at least one of R120 to R127, R130 to R 33, R144, R145, R147 and R148 is independently -OCF3, Ci-C20 aikyl, C3-C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, - C02R40, -SOR41, -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R 4R45, -OR46, -SR47, - NR48R49, -NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, - CH2(CHOH)aR60, -(CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of R - R33 is independently hydrogen, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C20 aryl;
each of R40-R55 and R58- R6 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural ot-amino acid;
each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
63. A compound being of formula (FX57):
Figure imgf000144_0001
(FX57); wherein:
each of L 20, L121 , L 122 L123] |_124] |_125_ L126 g nd |_127_ jf presenti is independently d-C 10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-C10 cycloalkenylene, C2-C 0 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b-, or -(CHOH)a-;
each of W120, W 21, W122, W 23, W 24, W125, W126 and W127 is independently a single bond,
(CH2)n- -(HCCH)n- -SO-, -S02 -S03 OS02- -NR "-, -CO-, -COO-, -OCO- OCOO-, -CONR1"-, -NR"CO- -OCONR14-, -NR 5COO- -NR16CONR17-, -NR 8CSNR19-
,20/
-0(CH2)n-, -S(CH2)n- -NR^u(CH2)n- -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n- - OCOO(CH2)n- -CONR21(CH2)n-, -CONR22(CH2)n-, -NR23CO(CH2)n-, -OCONR24(CH2)n- - NR25COO(CH2)n-, -NR26CONR27(CH2)n-, -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n- - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-;
each of R120 to R127 and R144 to R148 is independently a hydrogen, -OCF3, C C20 alkyl, C3- C20 cycloalkyl, C5-C30 aryl, C5-C30 heteroaryl, C^-C2o acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41, - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R' -OR46, -SR47, -NR48R49,
NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR' 60
-,63
(Ch CHzO^R0', -CH(R62)C02H, -CH(R0J)NH2, FL or Bm; wherein at least one of R^ 12u0 t,o R j11"27 and
,148
R144 to R1" is independently -CN, -C02R 40
-COR , -S02R -S02NR58R59, -OR46, -SR47, -
NR48R49, or -NR i 5500,COR51;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of R11 - R33 is independently hydrogen, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C20 aryl;
each of R 0-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid; each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
64. The compound of any of claims 61-63, wherein at least one of R120to R127 is an electron donating group and at least one of R 20 to R127 is an electron withdrawing group.
65. The compound of any of claims 61-64, wherein at least one of R120 to R 27 is Ci-C6 alkyl, - OR46, -SR47, -NR 8R49, or -NR50COR51; and wherein at least one of R120 to R127 is -CN, -C02R4°, -S02OR43, -CONR52R53, -COR54, -N02, -SOR41, -S02R55, -P03R 4R45, halo, d-C6 acyl, trihalomethyl, or -S02NR58R59.
66. The compound of any of claims 61-65, wherein at least one of R120 to R 27 is -OR46, -SR47, - NR 8R49, or -NR50COR51.
67. The compound of any of claims 61-65, wherein at least one of R120 to R127 is -NR48R49, or - NR50COR51.
68. The compound of any of claims 61-65, wherein at least one of R120to R127 is -CN, -C02R40,- COR54, -N02, -S02R55, or -S02NR58R59.
69. The compound of any of claims 61-65, wherein at least one of R120 to R127 is -C02R4°, - COR54, -S02NR58R59 or -S02R55.
70. The compound of any of claims 61-65, wherein at least one of R120 to R127 is -NR48R49, and wherein at least one of R120 to R127 is -CO2R 0,-COR54, -S02NR58R59 or -S02R55.
71. The compound of any of claims 61-70, wherein at least one of R120 to R127 and R144 to R148 is Bm.
72. The compound of any of claims 61-71 , wherein at least one of R120 to R 27 and R144 to R148 is FL.
73. The compound of any of claims 61-72, wherein o is 0 and W120 is a single bond; or wherein p is 0 and W 21 is a single bond; or wherein q is 0 and W 22 is a single bond; or wherein r is 0 and W123 is a single bond; or wherein s is 0 and W124 is a single bond; or wherein t is 0 and W125 is a single bond; or wherein u is 0 and W 26 is a single bond; or wherein v is 0 and W 27 is a single bond.
74. The compound of any of claims 61-72, wherein at least one of: L 20 with W120, L12 with W12 , L122 with W 22, L 23 with W123, L124 with W124, L 25 with W125, L126 with W 26, and L 27 with W127 combine to form: -(CH2)n- -0(CH2)n-, -CO(CH2)n- -OCO(CH2)n-, -COO(CH2)n-, - OCOO(CH2)n- -N(R20)(CH2)n-, -CON(R22)(CH2)n- -N(R23)CO(CH2)n- -OCONR24(CH2)n-, - NR25COO(CH2)n- -NR26CONR27(CH2)n-, or -NR28CSNR29(CH2)n-.
75. The compound of any of claims 61-63 wherein at least one of R 44 to R148 is -N02.
76. The compound of any of claims 61-63 wherein at least one of R144 to R 48 is -CH3.
Figure imgf000146_0001
(FX58); wherein:
each of L120, L121 , L122, L123, L124, L125, L126 and L127, if present, is independently C C10 alkylene, C3-C10 cycloalkylene, C2-C10 alkenylene, C3-C 0 cycloalkenylene, C2-C 0 alkynylene, ethenylene, ethynylene, phenylene, 1-aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b- or -(CHOH)a-; each of W120, W121, W 22, W123, W124, W125, W126 and W 27 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -SO2-, -SO3-, -OSO2-, -NR11-, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO-, -OCONR14-, -NR15COO- -NR16CONR17-, -NR18CSNR19-, -0(CH2)n-, -S(CH2)n-, -NR20(CH2)n- -CO(CH2)n- -COO(CH2)n- -OCO(CH2)n-, - OCOO(CH2)n- -CONR21(CH2)n-, -CONR 2(CH2)n- -NR23CO(CH2)n- -OCONR24(CH2)n- - NR25COO(CH2)n- -NR26CONR27(CH2)n-, -NR28CSNR29(CH2)n- -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-;
each of R 20 to R127 is independently a hydrogen, -OCF3, C^Cs alkyl, C5-C20 alkyl, C3-C20 cycloalkyl, C5 aryl, C7-C20 aryl, C5-C20 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5- C2o alkylaryl, C-|-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R4°, - SOR4 , -OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of R 44, R145, R147 and R148 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C2o aryl, C5-C20 heteroaryl, C-|-C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, C C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR 8R49, - NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61, -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ;
each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of R 1 - R33 is independently hydrogen, Ci-C20 alkyl, C3-C20 cycloalkyl, C5-C20 heteroaryl, or C5-C20 aryl;
each of R 0-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural ot-amino acid;
each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
Figure imgf000148_0001
(FX59); wherein:
each of L120, L 2 , L122, L 23, L124, L 25, L126 and L127, if present, is independently C C10 alkylene, C3-C 0 cycloalkylene, C2-C10 alkenylene, C3-C 0 cycloalkenylene, C2-C10 alkynylene, ethenylene, ethynylene, phenylene, 1 -aza-2,5-dioxocyclopentylene, 1 ,4-diazacyclohexylene, - (CH2CH20)b-, or -(CHOH)a-;
each of W120, W12 , W122, W123, W 24, W125, W126 and W127 is independently a single bond, -(CH2)n- -(HCCH)n-, -0-, -S-, -SO-, -S02- -S03-, -OS02-, -NR11-, -CO-, -COO-, -OCO- , -OCOO-, -CONR12-, -NR13CO- -OCONR14-, -NR15COO-, -NR16CONR17-, -NR18CSNR19-, -0(CH2)n- -S(CH2)n-, -NR20(CH2)n-, -CO(CH2)n- -COO(CH2)n-, -OCO(CH2)n-, - OCOO(CH2)n-, -CONR2 (CH2)n-, -CONR2 (CH2)n-, -NR 3CO(CH2)n- -OCONR24(CH2)n-, - NR25COO(CH2)n- -NR26CONR27(CH2)n- -NR28CSNR29(CH2)n-, -O(CH2)nNR30CO(CH2)n-, - CO(CH2)n(CH2OCH2)n(CH2)nNR31(CH2)nNR32CO-, or -CO(CH2)nNR33CO-;
each of R120 to R127 is independently a hydrogen, -OCF3, -CH3, -CH2CH3, C4-C20 alkyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C2o heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C20 alkynyl, C5-C20 alkylaryl, Ci-C6 alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51 , -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of R144, R145 and R147 is independently a hydrogen, -OCF3, C C20 alkyl, C3-C20 cycloalkyl, C5-C20 aryl, C5-C20 heteroaryl, C C20 acyl, C2-C20 alkenyl, C2-C2o alkynyl, C5-C20 alkylaryl, C^Ce alkoxycarbonyl, halo, halomethyl, dihalomethyl, trihalomethyl, -C02R40, -SOR41 , - OSR42 , -S02OR43, -CH2(CH2OCH2)bCH2OH, -P03R44R45, -OR46, -SR47, -NR48R49, - NR50COR51, -CN, -CONR52R53, -COR54, -N02, -S02R55, -S02NR58R59, -CH2(CHOH)aR60, - (CH2CH20)bR61 , -CH(R62)C02H, -CH(R63)NH2, FL or Bm;
each of o, p, q, r, s, t, u, and v is independently 0 or 1 ; each of a and b is independently an integer selected from the range of 1 to 100;
each n is independently an integer selected from the range of 1 to 10;
each of R - R33 is independently hydrogen, Ci-C2o alkyl, C3-C20 cycloalkyl, C5-C2o heteroaryl, or C5-C20 aryl;
each of R40-R55 and R58- R61 is independently hydrogen, C3-C10 cycloalkyl or C C10 alkyl; each of R62 and R63 is independently a side chain residue of a natural a-amino acid;
each of FL is independently a dye group corresponding to a pyrazine, a thiazole, a phenylxanthene, a phenothiazine, a phenoselenazine, a cyanine, an indocyanine, a squaraine, a dipyrrolo pyrimidone, an anthraquinone, a tetracene, a quinoline, an acridine, an acridone, a phenanthridine, an azo dye, a rhodamine, a phenoxazine, an azulene, an aza-azulene, a triphenyl methane dye, an indole, a benzoindole, an indocarbocyanine, a Nile Red dye, or a
benzoindocarbocyanine; and
each Bm is independently an amino acid, a peptide, a protein, a nucleoside, a nucleotide, an enzyme, a carbohydrate, a glycomimetic, an oligomer, a lipid, a polymer, an antibody, an antibody fragment, a mono- or polysaccharide comprising 1 to 50 carbohydrate units, a glycopeptide, a glycoprotein, a peptidomimetic, a drug, a steroid, a drug mimic, a hormone, a receptor, a metal chelating agent, a radioactive or nonradioactive metal complex, a mono- or polynucleotide comprising 1 to 50 nucleic acid units, or a polypeptide comprising 2 to 30 amino acid units.
PCT/US2010/056294 2009-11-11 2010-11-11 Sulfenamide compounds for phototherapy WO2011060113A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US26007809P 2009-11-11 2009-11-11
US61/260,078 2009-11-11

Publications (1)

Publication Number Publication Date
WO2011060113A1 true WO2011060113A1 (en) 2011-05-19

Family

ID=43447323

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/056294 WO2011060113A1 (en) 2009-11-11 2010-11-11 Sulfenamide compounds for phototherapy

Country Status (1)

Country Link
WO (1) WO2011060113A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9518062B2 (en) 2009-07-16 2016-12-13 Mallinckrodt Llc Compounds and compositions for use in phototherapy and in treatment of ocular neovascular disease and cancers

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5518888A (en) 1993-10-26 1996-05-21 Thomas Jefferson University ST receptor binding compounds and methods of using the same
US5672333A (en) 1996-05-13 1997-09-30 Mallinckrodt Medical, Inc. Delta1,6 bicyclo 4,4,0! functional dyes for contrast enhancement in optical imaging
US5698397A (en) 1995-06-07 1997-12-16 Sri International Up-converting reporters for biological and other assays using laser excitation techniques
US5714342A (en) 1994-10-28 1998-02-03 Oncoimmunin, Inc. Compositions for the detection of protease in biological samples and methods of use therefo
US6167297A (en) 1999-05-05 2000-12-26 Benaron; David A. Detecting, localizing, and targeting internal sites in vivo using optical contrast agents
US6228344B1 (en) 1997-03-13 2001-05-08 Mallinckrodt Inc. Method of measuring physiological function
US6258378B1 (en) 1998-02-09 2001-07-10 Bracco Research S.A. Delivery of biologically active substance to target sites in the body of patients
US6406713B1 (en) 1987-03-05 2002-06-18 The Liposome Company, Inc. Methods of preparing low-toxicity drug-lipid complexes
US20030158127A1 (en) * 2001-07-03 2003-08-21 Mallinckrodt Inc. Aromatic sulfenates for type I phototherapy
US6748259B1 (en) 2000-06-15 2004-06-08 Spectros Corporation Optical imaging of induced signals in vivo under ambient light conditions
US6838074B2 (en) 2001-08-08 2005-01-04 Bristol-Myers Squibb Company Simultaneous imaging of cardiac perfusion and a vitronectin receptor targeted imaging agent
US7011817B2 (en) 2000-01-18 2006-03-14 Mallinckrodt Inc. Hydrophilic cyanine dyes
US7128896B2 (en) 2001-10-17 2006-10-31 Mallinckrodt Inc. Pathological tissue detection and treatment employing targeted benzoindole optical agents
US20070149478A1 (en) 1996-08-09 2007-06-28 Mannatech, Inc. Compositions of plant carbohydrates as dietary supplements
WO2007106436A2 (en) 2006-03-10 2007-09-20 Mallinckrodt Inc. Photoactive compounds and compositions and uses thereof
WO2008108941A2 (en) 2007-03-01 2008-09-12 Mallinckrodt Inc. Integrated photoactive peptides and uses thereof

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6406713B1 (en) 1987-03-05 2002-06-18 The Liposome Company, Inc. Methods of preparing low-toxicity drug-lipid complexes
US5518888A (en) 1993-10-26 1996-05-21 Thomas Jefferson University ST receptor binding compounds and methods of using the same
US5714342A (en) 1994-10-28 1998-02-03 Oncoimmunin, Inc. Compositions for the detection of protease in biological samples and methods of use therefo
US5698397A (en) 1995-06-07 1997-12-16 Sri International Up-converting reporters for biological and other assays using laser excitation techniques
US5672333A (en) 1996-05-13 1997-09-30 Mallinckrodt Medical, Inc. Delta1,6 bicyclo 4,4,0! functional dyes for contrast enhancement in optical imaging
US20070149478A1 (en) 1996-08-09 2007-06-28 Mannatech, Inc. Compositions of plant carbohydrates as dietary supplements
US6228344B1 (en) 1997-03-13 2001-05-08 Mallinckrodt Inc. Method of measuring physiological function
US6258378B1 (en) 1998-02-09 2001-07-10 Bracco Research S.A. Delivery of biologically active substance to target sites in the body of patients
US6167297A (en) 1999-05-05 2000-12-26 Benaron; David A. Detecting, localizing, and targeting internal sites in vivo using optical contrast agents
US7011817B2 (en) 2000-01-18 2006-03-14 Mallinckrodt Inc. Hydrophilic cyanine dyes
US6748259B1 (en) 2000-06-15 2004-06-08 Spectros Corporation Optical imaging of induced signals in vivo under ambient light conditions
US20030158127A1 (en) * 2001-07-03 2003-08-21 Mallinckrodt Inc. Aromatic sulfenates for type I phototherapy
US7235685B2 (en) 2001-07-03 2007-06-26 Mallinckrodt, Inc. Aromatic sulfenates for type I phototherapy
US6838074B2 (en) 2001-08-08 2005-01-04 Bristol-Myers Squibb Company Simultaneous imaging of cardiac perfusion and a vitronectin receptor targeted imaging agent
US7128896B2 (en) 2001-10-17 2006-10-31 Mallinckrodt Inc. Pathological tissue detection and treatment employing targeted benzoindole optical agents
US7201892B2 (en) 2001-10-17 2007-04-10 Mallinckrodt Inc. Pathological tissue detection and treatment employing targeted optical agents
WO2007106436A2 (en) 2006-03-10 2007-09-20 Mallinckrodt Inc. Photoactive compounds and compositions and uses thereof
WO2008108941A2 (en) 2007-03-01 2008-09-12 Mallinckrodt Inc. Integrated photoactive peptides and uses thereof

Non-Patent Citations (85)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"Design of Prodrugs", 1985, ELSEVIER
"Goodman and Gilman's: The Pharmacological Bases of Therapeutics", 1996, PERGAMON PRESS
"Liposome Dermatics", 1992, SPRINGER-VERLAG, article "Griesbach Conference", pages: 69 81,91
"Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Modern Pharmaceutics", 1979
"Modern Pharmaceutics", 1995, MARCEL DEKKER, INC.
"Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
"Pharmaceutical Dosage Forms", 1980, MARCEL DEKKER, INC.
"Remingtons Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING
"REMINGTON'S PHARMACEUTICAL SCIENCES", 2000, WILLIAMS & WILKINS
"Stealth Liposomes", 1995, CRC PRESS
"The Chemotherapy Source Book", WILLIAMS & WILKENS PUBLISHING
"The Handbook of Pharmaceutical Excipients", 2005, AMERICAN PHARMACISTS ASSOCIATION AND THE PHARMACEUTICAL PRESS
"The Handbook of Pharmaceutical Excipients", AMERICAN PHARMACEUTICAL ASSOCIATION
"The National Formulary", vol. 24, 2000, NATIONAL PUBLISHING, PHILADELPHIA, article "The United States Pharmacopeia", pages: 19
"The Pharmaceutical Society of Great Britain", 1968
A. GIANNIS; T. KOLTER: "Peptidomimetics for Receptor Ligands - Discovery, Development, and Medical Perspectives", ANGEWANDTE CHEMIE INTERNATIONAL EDITION IN ENGLISH, vol. 32, 1993, pages 1244 - 1267, XP000392719, DOI: doi:10.1002/anie.199312441
A. PELEGRIN ET AL.: "Photoimmunodiagnosis with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies", J. CELL PHANNACO/., vol. 3, 1992, pages 141 - 145
ACHILEFU ET AL.: "Novel receptor-targeted fluorescent contrast agents for in vivo imaging of tumors", INVESTIGATIVE RADIOLOGY, vol. 35, 2000, pages 479 - 485
ACHILEFU ET AL.: "Novel receptor-targeted fluorescent contrast agents for in vivo imaging of tumors", INVESTIQATIVE RADIOLODV, vol. 35, 2000, pages 479 - 485
ANSEL: "Introduction to Pharmaceutical Dosage Forms", 1976
B. BALLOU ET AL.: "Tumor labeling in vivo using cyanine-conjugated monoclonal antibodies", CANCER IMMUNOLOGY AND IMMUNOTHERAPY, vol. 41, 1995, pages 247 - 263
B. J. TROMBERG ET AL.: "Non-invasive measurements of breast tissue optical properties using frequency-domain photon migration", PHIL. TRANS. ROYAL SOCIETY LONDON B, vol. 352, 1997, pages 661 - 668
BALLOU ET AL.: "Tumor labeling in vivo using cyanine conjugated monoclonal antibodies", CANCER IMMUNOLOGY AND IMMUNOTHERAPY, vol. 41, 1995, pages 257 - 263, XP002919350, DOI: doi:10.1007/BF01517001
BALLOU ET AL.: "Tumor labeling in vivo using cyanine conjugated monoclonal antibodies", CANCER IMMUNOLOQY AND IMMUNOTHERAPY, vol. 41, 1995, pages 257 - 263, XP002919350, DOI: doi:10.1007/BF01517001
BALLOU ET AL.: "Tumor labeling in vivo using cyanine conjugated monoclonol antibodies", CANCER IMMUNOLOOV AND IMMUNOTHERAPV, vol. 41, 1995, pages 257 - 263
BLANCA ET AL.: "The Chiral S-N Axis in Sulfenamides: Enantiomeric Resolution, Direction Demonstration of Optical Activity, and Kinetics of Interconversion", ANGEW. CHEM. INT. ED. ENGL., vol. 36, no. 20, 1997
BROWN S; BROWN EA; WALKER I: "Photodiagnosis And Photodynamic Therapy", THE PRESENT AND FUTURE ROLE OF PHOTODYNAMIC THERAPY IN CANCER TREATMENT. LANCET ONCOL., vol. 5, 2004, pages 497 - 508
CHEN, B.; POGUE, B.; LUNA, J.M.; HARDMAN, R.L.; HOOPES, P.J.; HASAN, T.: "Tumor vascular permeabilization by vascular-targeting photosensitization: effects, mechanism, and therapeutic implications", CLINICAL CANCER RESEARCH, vol. 12, 2006, pages 917 - 923
D. A. BELINIER ET AL.: "Murine pharmacokinetics and antitumor efficacy of the photodynamic sensitizer 2-[I-hexyloxyethyl]-2-devinyl pyropheophorbide- a", J. PHOTOCHEM. PHOTOBIOL., vol. 20, 1993, pages 55 - 61
D. A. BENARON; D. K. STEVENSON: "Optical time-of-flight and absorbance imaging of biologic media", SCIENCE, vol. 259, 1993, pages 1463 - 1466, XP000574893, DOI: doi:10.1126/science.8451643
DAVIS, ORG. PREP. PROC. INT., vol. 11, 1979, pages 33
DOUGHERTY TJ; GOMER CJ; HENDERSON BW; JORI G; KESSEL D; KORBELIK M; MOAN J; PENG Q: "Photodynamic Therapy", J. NATL.CANCER INST., vol. 90, 1998, pages 899 - 905
DUFES ET AL., PHARM. RES., vol. 17, 2000, pages 1250
E. KUHLE: "The Chemistry of Sulfenic Acids", 1973, GEORG THIEME PUBLISHERS
E.M. GORDON; M.A. ONDETTI; J. PLUSCEC; C.M. CIMARUSTI; D.P. BONNER; R.B. SYKES, J. AMER. CHEM. SOC., vol. 104, 1982, pages 6053
F. A. DAVIS; U.K. NADIR, ORG. PREP. PROC.LNT., vol. 11, 1979, pages 33
F. DAVIS: "Adventures in Sulfur - Nitrogen Chemistry", J. ORG. CHEM, vol. 71, 2006, pages 8993 - 9003
G. A. WAGNIERES ET AL.: "In vivo fluorescence spectroscopy and imaging for oncological applications", PHOTOCHEM. PHOTOBIOL., vol. 68, 1998, pages 603 - 632
G. J. TEARNEY ET AL.: "In vivo endoscopic optical biopsy with optical coherence tomography", SCIENCE, vol. 276, 1997, pages 2037 - 2039
GOODMAN; GILMAN: "The Pharmacological Basis of Therapeutics", MCGRAW HILL PUBLISHING
GROSJEAN, P.; WAGIERES, G.; FONTOLLIET, C.; VAN DEN BERGH, H.; MONNIER, P.: "Clinical phototherapy for superficial cancer in the esophagus and the bronchi: 514 nm compared with 630 nm light irradiation after sensitization with Photofrin II", BRITISH JOURNAL OF CANCER, vol. 77, 1998, pages 1989 - 1955
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38
H. BUNDGAARD: "A Textbook of Drug Design and Development", 1991, article "Design and Application of Prodrugs", pages: 113 - 191
HERMANSON: "Bioconjugate Techniques", 1996, ACADEMIC PRESS
HNATOWICH ET AL.: "Radioactive Labeling of Antibody: A simple and efficient method", SCIENCE, vol. 220, 1983, pages 613, XP002002847, DOI: doi:10.1126/science.6836304
HNATOWICH ET AL.: "Radiolabeling of Antibodies: A simple and efficient method", SCIENCE, vol. 220, 1983, pages 613
HOOVER, JOHN E.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
J. S. REYNOLDS ET AL.: "Imaging of spontaneous canine mammary tumors using fluorescent contrast agents", PHOTOCHEM. PHOTOBIOL., vol. 70, 1999, pages 87 - 94
J.G. NAIRN: "Remington's Pharmaceutical Science", 1985, MACK PUBLISHING CO., pages: 1492 - 1517
K. LICHA ET AL.: "Biomedical Imaging: Reporters, Dyes, and Instrumentation", vol. 3600, 1999, article "New contrast agent for optical imaging: acid-cleavable conjugates of cyanine dyes with biomolecules", pages: 29 - 35
LI, L.; LUO, R.; LIAO, W.; ZHANG, M.; LUO, Y.; MIAO, J.: "Clinical study of photofrin phototherapy for the treatment of relapse nasopharyngeal carcinoma", PHOTODIAGNOSTICS AND PHOTOTHERAPY, vol. 3, 2006, pages 266 - 271
LICHA ET AL.: "New contrast agent for optical imaging: acid cleavable conjugates of cyanine dyes with biomolecules", BIOMEDICAL IRNAQIR1Q: REPORTERS. DYES AND INSTRUMENTATION, vol. 3600, 1999, pages 29 - 35, XP000992961, DOI: doi:10.1117/12.351013
LICHA ET AL.: "New contrast agent for optical imaging: acid cleavable conjugates of cyanine dyes with biomolecules", BIOMEDICAL LMAQINQ: REPORTERS, DYES AND INSTRUMENTATION, PROCEEDINGS OF SPIE, vol. 3600, 1999, pages 29 - 35, XP000992961, DOI: doi:10.1117/12.351013
LICHA ET AL.: "New contrast agent for optical imaging: acid cleavable conjugates of cyanine dyes with biomolecules, in Biomedical Imaging: Reporters, Dyes and Instrumentation", PROCEEDINGS OF SPIE, vol. 3600, 1999, pages 29 - 35
LIEBERMAN ET AL., PHARMACEUTICAL DOSAGE FORMS: TABLETS, 1981
MARSHALL: "SOLID ORAL DOSAGE FORMS", vol. 7, 1979, article "Modern Pharmaceutics", pages: 359 - 427
MITTON, D.; ACKROYD, R.: "Phototherapy of Barrett's oesophagus and oesophageal carcinoma - how I do it", PHOTODIAGNOSTICS AND PHOTOTHERAPY, vol. 3, 2006, pages 96 - 98
MIURA ET AL.: "EPR studies of nitrogen-centered free radicals. Part 50. Unusual decomposition behavior of isolable stable thioaminyl free radicals at high temperature", J. CHEM. SOC. PERKIN TRANS, vol. 2, 1998, pages 1185 - 1188
MLKVY, P.; MESSMANN, H.; REGULA, J.; CONIO, M.; PAUER, M.; MILLSON, C.E.; MACROBERT, A.J.; BROWN, S.G.: "Phototherapy for gastrointestinal tumors using three photosensitizers - ALA induced PPIX, Photofrin, and MTHPC. A pilot study", NEOPLASMA, vol. 45, 1998, pages 157 - 161
NOGRADY: "Medicinal Chemistry A Biochemical Approach", 1985, OXFORD UNIVERSITY PRESS, pages: 388 - 392
PATCH, J.A. ET AL.: "Versatile oligo(N-substituted)glycines: The many roles of peptoids in drug discovery.", PSEUDO-PEPTIDES IN DRUG DISCOVERY, 2004, pages 1 - 31
PELEGRIN ET AL.: "Photoimmunodiagnosis with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies", J. CELLULAR PHARM., vol. 3, 1992, pages 141
PELEGRIN ET AL.: "Photoimmunodiagnostics with antibody-fluorescein conjugates: in vitro and in vivo preclinical studies", JOURNAL OF CELLULAR PHARMACOLOQV, vol. 3, 1992, pages 141 - 145
R. F. POTTER: "Medical optical tomography: functional imaging and monitoring", 1993, SPIE OPTICAL ENGINEERING PRESS
RAGHAVAN RAJAGOPALAN ET AL: "Novel type 1 photosensitizers: viability of leukemia cells exposed to reactive intermediates generated in situ by in vitro photofragmentation", PROCEEDINGS OF SPIE; 20090611, vol. 7380, 11 June 2009 (2009-06-11), pages 738027/1 - 8, XP009140796, ISSN: 0277-786X, [retrieved on 20090713], DOI: DOI:10.1117/12.822840 *
S. A. ACHILEFU ET AL.: "Novel receptor-targeted fluorescent contrast agents for in vivo tumor imaging", INVESTIGATIVE RADIOLOGY, vol. 35, no. 8, 2000, pages 479 - 485, XP000978923, DOI: doi:10.1097/00004424-200008000-00004
S. FANTINI ET AL.: "Assessment of the size, position, and optical properties of breast tumors in vivo by noninvasive optical methods", APPL. OPT., vol. 37, 1998, pages 1982 - 1989
S. THEA; G. CEVASCO, TETRAHEDRON LETTERS, vol. 28, 1987, pages 5193
S. THEA; G. CEVASCO: "A Mild and Convenient Preparation of Sulfenyl Chlorides from Thiolacetates", TETRAHEDRON LETTERS, vol. 29, no. 23, 1988, pages 2865 - 2866, XP022028578, DOI: doi:10.1016/0040-4039(88)85232-8
SARFARAZ K. NIAZI: "Handbook of Pharmaceutical Manufacturing Formulations", 2004, CRC PRESS
SCHMIDT-DRFURTH, U.; BRINGRUBER, R.; HASAN, T.: "Phototherapy in ocular vascular disease", IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS, vol. 2, 1996, pages 988 - 996
SPIEGEL, A.J. ET AL.: "Use of Nonaqueous Solvents in Parenteral Products", J. PHARMA. SCIENCES, vol. 52, no. 10, 1963, pages 917 - 927, XP007915361
STAHL ET AL.: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
T. GREENE: "Protective Group in Organic Synthesis", 1981, JOHN WILEY & SONS, article "Protection of Amino Group", pages: 283 - 284
T. HIGUCHI; V. STELLA: "Pro-drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES
TILLY ET AL., ENDOCRINOLOGY, vol. 131, 1992, pages 799
TRIESSCHEIJN M; BAAS P; SCHELLENS JHM: "Photodynamic Therapy in Oncology", THE ONCOLOGIST., vol. 11, 2006, pages 1034 - 1044
VALLABHAJOSULA ET AL., J. NUCLEAR MED., vol. 37, 1996, pages 1016
WU; GELLMAN, PEPTIDOMIMETICS, ACCOUNTS OF CHEMICAL RESEARCH, vol. 41, no. 10, pages 1231 - 1232
Y.C. AGGARWAL; S. WARREN, TETRAHEDRON LETT., vol. 27, 1986, pages 101
ZHENG HUANG: "A Review of Progress in Clinical Photodynamic Therapy", TECHNOL CANCER RES TREAT., vol. 4, no. 3, June 2005 (2005-06-01), pages 283 - 293

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9518062B2 (en) 2009-07-16 2016-12-13 Mallinckrodt Llc Compounds and compositions for use in phototherapy and in treatment of ocular neovascular disease and cancers
US9527858B2 (en) 2009-07-16 2016-12-27 Mallinckrodt Llc Compounds and compositions for use in phototherapy and in treatment of ocular neovascular disease and cancers

Similar Documents

Publication Publication Date Title
US20110264026A1 (en) Azo Derivatives and Uses Thereof in Phototherapy
US20110177007A1 (en) Dithienopyrrole Dyes for Imaging and Therapy
US20090035363A1 (en) Photoactive Compounds and Compositions and Uses Thereof
US8628751B2 (en) Pyrazine derivatives for optical imaging and therapy
US8664392B2 (en) Pyrazine derivatives for bioconjugation
US20110196231A1 (en) Fused Ring Thiophene Dyes for Imaging and Therapy
US20110177006A1 (en) Dithienofuran Dyes for Imaging and Therapy
US10059739B2 (en) Non-benzenoid aromatic systems for imaging, monitoring and therapy
US20110288033A1 (en) Disulfide Compounds for Phototherapy
US8731655B2 (en) Compounds containing acyclic N-N bonds for phototherapy
US9186349B2 (en) Diaza heterocyclic compounds for phototherapy
WO2013039851A1 (en) Optical agents for imaging and visualization of matrix metalloproteinase enzymes
WO2010132547A2 (en) Oxaza heterocyclic compounds for phototherapy
WO2010132525A1 (en) Compounds containing acyclic n-o bonds for phototherapy
US7888378B2 (en) Thiadiazole compounds and uses thereof
WO2011060113A1 (en) Sulfenamide compounds for phototherapy
US20120289884A1 (en) Fused Ring Diarylamino Photosensitizers for Phototherapy
WO2011084571A2 (en) Azide derivatives for phototherapy
WO2011031955A2 (en) Optical monitoring of leukemia

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10781770

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10781770

Country of ref document: EP

Kind code of ref document: A1