WO1990015065A1 - Exonuclease-resistant oligonucleotides and methods for preparing the same - Google Patents

Exonuclease-resistant oligonucleotides and methods for preparing the same Download PDF

Info

Publication number
WO1990015065A1
WO1990015065A1 PCT/US1990/003138 US9003138W WO9015065A1 WO 1990015065 A1 WO1990015065 A1 WO 1990015065A1 US 9003138 W US9003138 W US 9003138W WO 9015065 A1 WO9015065 A1 WO 9015065A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
linkages
hydrogen
phosphoramidate
oligonucleotides
Prior art date
Application number
PCT/US1990/003138
Other languages
French (fr)
Inventor
Brian C. Froehler
Original Assignee
Gilead Sciences, 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 Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Priority to KR1019910701767A priority Critical patent/KR920701230A/en
Priority to CA002058632A priority patent/CA2058632C/en
Publication of WO1990015065A1 publication Critical patent/WO1990015065A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids

Definitions

  • the present invention is directed to oligonucleotides containing a 3'- and/or 5'-capped terminal and which are thereby rendered resistant to degradation by exonucleases.
  • the exonuclease-resistant oligonucleotides have two or more phosphoramidate
  • DNA molecules contain internucleotide phosphodiester linkages which are degraded by
  • exonucleases present in cells, culture media and human serum. For example, degradation by exonucleases in tissue culture media of DNA may be observed within about 30 minutes to about six hours.
  • Synthetic oligodeoxy- nucleotides with phosphodiester linkages are routinely used in genetic engineering, for example, to locate specific RNA or DNA fragments from a library.
  • the long- term stability of an oligonucleotide for this utility is not a major concern, since the oligonucleotide is usually not exposed to the relatively stringent environment of the culture medium, therefore exonuclease degradation is not a substantial problem.
  • oligodeoxynucleotides which are stable (i.e., for more than several hours or days) for long-term uses.
  • a oligodeoxynucleotide with phosphodiester linkages can be used to block protein synthesis by hydrogen bonding to complementary messenger RNA thereby providing a tool for use in an antisense fashion.
  • the present invention is accordingly directed to such exonuclease-stable oligonucleotides.
  • phosphodiester linkages are replaced with a specified number of phosphoramidate linkages.
  • It a further object of the invention to provide methods of making such exonuclease-resistant oligonucleotides. It is still a further object of the invention to provide a method for end-capping oligonucleotides with moieties which can perform multiple functions, such as aiding in transport, serving as chromophoric tags, or enabling cross-linking.
  • the present invention provides oligonucleotides having two or more phosphoramidate linkages at the 3' terminus and/or 5' terminus, which oligonucleotides are resistant to
  • the number of phosphoramidate linkages is at least 1 and less than a number which would interfere with hybridization to a complementary oligonucleotide strand, and/or less than a number which would interfere with RNAse activity when said oligonucleotide is hybridized to RNA.
  • phosphoramidate linkages are incorporated at either or both the 3' terminus and the 5' terminus.
  • the phosphoramidate linkages may be substituted with any one of a number of different types of moieties as will be described in detail hereinbelow.
  • exonuclease-resistant are provided which have the following formulas I, II or III, i.e., containing phosphoramidate linkages as just described as well as phosphoromonothioate and/or
  • each n, m, i, j and s is independently an integer and each s is in the range of about 2 to 10; each n and m is independently from 1 to about 50; s + n in formulas I and II is less than 100; and s + s + m in formula III is less than about 100; each i varies from 1 to n; each j varies from 1 to m; T is hydrogen or a hydroxyl- protecting group; R 1 and R 2 are moities independently selected from the group consisting of hydrogen,
  • each B is independently a protected or unprotected heterocyclic base
  • each X i and X j is independently O or S ; and each Y i and Y j is independently R, -SR or -OR, where R is as defined for R 1 and R 2 .
  • the present invention also provides methods for preparing such end-capped oligonucleotides.
  • polynucleotide and oligonucleotide shall be generic to polydeoxyribo- nucleotides (containing 2'-deoxy-D-ribose or modified forms thereof), to polyribonucleotides (containing D- ribose or modified forms thereof), and to any other type of polynucleotide which is an N-glycoside of a purine or pyrimidine bases, or modified purine or pyrimidine bases.
  • nucleoside will similarly be generic to ribonucleosides, deoxyribonucleosides, or to any other nucleoside which is an N-glycoside of a purine or
  • pyrimidine base or modified purine or pyrimidine base.
  • polynucleotide and oligonucleotide
  • nucleoside and “nucleotides” will include those moieties which contain not only the known purine and pyrimidine bases, i.e., adenine, thymine, cytosine, guanine and uracil, but also other heterocyclic bases which contain protecting groups or have been otherwise modified or derivatized.
  • modified nucleosides or “modified nucleotides” as used herein are intended to include those compounds containing one or more protecting groups such as acyl, isobutyryl, benzoyl, or the like, as well as any of the wide range of modified and derivatized bases as known in the art.
  • protecting groups such as acyl, isobutyryl, benzoyl, or the like
  • examples of such modified or deriva- tized bases include 5-fluorouracil, 5-bromouracil,
  • 5-chlorouracil 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil,
  • 5-carboxymethylaminomethyl-2-thiouridine 5-carboxy- methylaminomethyluracil, dihydroura ⁇ il, beta-D-galacto- sylqueosine, inosine, N6-isopentenyladenine, 1-methyl- adenine, 1-methylpseudouracil, 1-methylguanine,
  • Modified nucleosides or nucleotides can also include modifications on the sugar moiety, for example, wherein one or more of the hydroxyl groups are replaced with halogen or aliphatic groups, or functionalized as ethers, amines, etc.
  • the polynucleotides according to the present invention may be of any length, but lengths of about three to about fifty nucleotides are particularly useful for most genetic engineering applications. According to the present invention, the 3' end and/or the 5' end of the polynucleotide will contain at least two
  • phosphoramidate internucleotide linkages The remaining internucleotide linkages may be phosphodiester linkages, phosphorothioate linkages or phosphorodithioate linkages, or any other internucleotide linkage, other than a phosphoramidate, or combinations of these other linkages.
  • Methods for preparing such non-phosphoramidate linkages are known in the art, e.g., as taught by Froehler et al., Nuc. Acids Res. 14:5399-5467 (1986), and Froehler, B., Tet. Lett. 27:5575-5578 (1986), cited above and
  • Internucleotide phosphodiester linkages are prepared from hydrogen phosphonate linkages preferably by oxidation with, e.g., aqueous iodine.
  • Phosphoromonothioate linkages are formed from the initially present hydrogen phosphonate linkages by treatment with sulfur.
  • the reaction is carried out at approximately room temperature for on the order of 20 minutes in a solvent system which typically includes a sulfur solvent such as CS 2 along with a basic solvent such as pyridine.
  • a solvent system typically includes a sulfur solvent such as CS 2 along with a basic solvent such as pyridine.
  • CS 2 is preferred as the sulfur solvent because it acts to dissolve elemental sulfur.
  • the following scheme illustrates the postulated reaction: (See, e.g., Stein et al., cited above.)
  • the oligonucleotides of the invention are resistant to degradation under both physiological and tissue culture conditions, and in particular are resistant to degradation by exonucleases.
  • the oligonucleotide In order that the oligonucleotide be resistant to such enzymatic degradation, it is modified so that phosphodiester linkages initially present at the 3' terminus are replaced with a selected number of phosphoramidate linkages, that number being at least one and less than a number which would cause interference with hybridization to a complementary oligonucleotide strand, and/or less than a number which would interfere with RNAseH activity when said the oligonucleotide is
  • RNA hybridized to RNA.
  • Such a modification may additionally or alternatively be made at the 5' terminus.
  • the number of phosphoramidate linkages be selected such that the melting
  • any duplex formed with complement is lowered by less than about 10°C relative to that obtained with an oligonucleotide containing only the initial phosphodiester linkages.
  • the number of phosphoramidate linkages is such that the melting
  • the temperature of a duplex formed is lowered by less than about 5°C.
  • the number of phosphoramidate linkages present is typically and preferably between about 2 and 10, more preferably between about 2 and 8, and most preferably between about 2 and 6.
  • the phosphoramidate linkage has the formula
  • R 1 and R 2 moieties are substituents which must be selected so as not to interfere with
  • R 1 and R 2 are independently selected from the group consisting of hydrogen, hydrocarbyl substituents of
  • R 1 and R 2 are not both hydrogen, i.e., the phosphoramidate linkages herein are always N-substituted. In this case, it is preferred that one of the two substituents be hydrogen.
  • Suitable hydrocarbyl and oxyhydrocarbyl substituents include, for example, linear or branched alkyl of 1-20 carbon atoms, linear or branched alkenyl of 2-20 carbon atoms, cycloalkyl or cycloalkenyl of 3-20 carbon atoms, linear or branched alkoxy of 1-20 carbon atoms, or aryl of 6-18 carbon atoms.
  • the hydrocarbyl substituent may be, for example, an alkoxy substituent having the formula CH 3 O-(CH 2 ) x - or a straight chain alkyl group having the formula CH 3 (CH 2 ) y - where x is an integer in the range of 1-20, inclusive, preferably in the range of 1-10, inclusive, and y is an integer in the range of 0-15, inclusive.
  • Examples of preferred oligonucleotide linkages within the aforementioned groups are wherein one of R 1 and R 2 is H and the other is either 2-methoxyethyl, dodecyl, or n- propyl.
  • the 2-methoxyethyl and dodecyl linkages are sometimes referred to herein as "MEA” and "C12",
  • the R 1 and R 2 groups may also be, in addition to the foregoing, macromolecular species such as sugars, polypeptides, chromophoric groups, lipophilic groups, polymers, steroid hormones, or the like.
  • Lipophilic groups refer to moieties which are chemically compatible with the outer cell surface, i.e., so as to enable the oligonucleotide to attach to, merge with and cross the cell membrane. Examples of such lipophilic groups are fatty acids and fatty alcohols (in addition to the long chain hydrocarbyl groups described above).
  • R 1 and/or R 2 examples include transferrin and epidermal growth factor (EGF), while suitable non-polypeptide polymers include ionic, nonionic and zwitterionic
  • polymers examples of a particularly preferred polymer is polyethylene glycol.
  • Steroid substituents include any of the general fat ⁇ ily of lipid compounds which comprise sterols,
  • bioacids cardiac glycosides, seponans, and sex hormones, which include the following basic structure:
  • steroids examples include natural corticosteroid hormones (produced by the adrenal glands), sex hormones (progesterone, androgens, and estrogens).
  • R 1 and R 2 groups can confer any of a variety of desired properties to the oligo- nucleotide.
  • R 1 or R 2 is a polymer such as polyethylene glycol, a polypeptide or a lipophilic group such as a long-chain hydrocarbyl moiety, such a group may facilitate transport or permeation of the oligonucleotide through cell membranes, thus increasing the cellular uptake of the oligonucleotide.
  • the R 1 or R 2 group may also be a group which affects target DNA or RNA to which the oligonucleotide will bind, such as providing covalent linkages to the target strand to facilitate cleavage or intercalation of the oligonucleotide to the target strand.
  • the R 1 and R 2 groups may additionally serve a cutting function (e.g., a site for cutting the complementary strand), or a receptor function (e.g., a receptor ligand).
  • oligonucleotides of the present invention can include other phosphoramidate N-substituents not explicitly disclosed herein so long as those substituents confer exonuclease resistance and do not interfere with hybridization to a complementary oligonucleotide strand.
  • the invention also encompasses oligonucleotide compositions containing oligonucleotides of the following formula I, II or III, i.e., wherein phosphoromonothioate and/or phosphorodithioate linkages are incorporated in addition to the phosphoramidate linkages:
  • phosphoramidate linkages be in the range of 2-8, more preferably in the range of 2-6. It is also preferred that m and n be within the aforementioned ranges.
  • the 3'-capped oligonucleotides may be prepared by first preparing a polymer-bound polynucleoside with the formula IV
  • P is a solid state polymeric support, or other type of solid support
  • B the base portion of a nucleoside, i.e., a purine or pyrimidine base, or any modified purine or pyrimidine base.
  • the functional groups on the base i.e., the amine groups, will be appropriately protected during the course of the synthesis and removed after the completed polynucleotide is removed from the polymer support.
  • the linkage to the polymer support is through the 3' hydroxy group, the free hydroxy group is the 5' group of the nucleoside.
  • the group T is a
  • the polymer-bound polynucleoside hydrogen phosphonate (IV) is preferably prepared by treating the DBU
  • nucleoside hydrogen phosphonates may be added (to make the two or more internucleotide linkages at the 3' end of the
  • the oligonucleotide chain elongation will proceed in conformance with a predetermined sequence in a series of condensations, each one of which results in the addition of another nucleoside to the oligomer.
  • the condensation is typically accomplished with dehydrating agents, which are suitably phosphorylating agents or acylating agents such as isobutylchloroformate,
  • diphenylchlorophosphate organic acid anhydrides (such as acetic anhydride, isobutyric anhydride or trimethyl acetic anhydride) and organic acid halides such as pivaloyl chloride, pivaloyl bromide, 1-adamantyl- carboxylic chloride or benzoyl chloride.
  • organic acid anhydrides such as acetic anhydride, isobutyric anhydride or trimethyl acetic anhydride
  • organic acid halides such as pivaloyl chloride, pivaloyl bromide, 1-adamantyl- carboxylic chloride or benzoyl chloride.
  • the preferred condensing agent is pivaloyl chloride in pyridine
  • acetonitrile Prior to the addition of each successive nucleoside hydrogen phosphonate, the 5'-protecting group or the carrier bound nucleotide is removed. Typically, for removal of the DMT group, this is done by treatment with 2.5% volume/volume dichloroacetic acid/CH 2 Cl 2 , although 1% weight/volume trichloroacetic acid/CH 2 Cl 2 or ZnBr 2 -saturated nitromethane are also useful. Other deprotection procedures suitable for other known
  • the carrier is preferably washed with anhydrous pyridine/acetonitrile (l/l,v/v) and the condensation reaction is completed in as many cycles as are required to form the desired number of 3'-end internucleotide bonds which will be converted to phosphoramidates.
  • the carrier- bound polynucleotide hydrogen phosphonate is oxidized to convert the hydrogen phosphonate internucleotide linkages to phosphoramidate linkages, preferably by treatment with the desired amine NHR 1 R 2 with R 1 and R 2 as defined earlier and CCl 4 as described in Froehler, et al.,
  • the oligonucleotide is then completed by methods which form nonphosphoramidate linkages, such as phosphodiester linkages,
  • the preferred method for completing the oligonucleotide is to continue the sequence using 5'-protected nucleoside hydrogen- phosphonates. In the instance where the 5' end will not be capped, after the last 5'-protected nucleoside
  • hydrogen phosphonate has been added, all of the hydrogen phosphonate linkages are oxidized to produce diester linkages, preferably by aqueous iodine oxidation or oxidation using other oxidizing agents, such as
  • the oligonucleotide may be separated from the carrier, using conventional methods, which in the preferred instance is incubation with concentrated ammonium hydroxide. Any protecting groups may be removed as described above using about 2% dichloroacetic acid/CH 2 Cl 2 , or about 80% acetic acid, or by other conventional methods, depending on the nature of the protecting groups. The desired oligonucleotide is then purified by HPLC, polyacrylamide gel electro- phoresis or using other conventional techniques.
  • i varies from 1 to 5
  • B a purine or pyrimidine base
  • nucleotides (therefore having only four internucleotide linkages, two of which are phosphoramidate linkages) it is feasible to conduct the synthesis without the use of a solid state support.
  • a conventional 3'-hydroxy protecting group may be used which is
  • the 5'-protecting group may be
  • the two or more phosphoramidate linkages need not each contain the same R 1 and R 2 groups. This may be accomplished by generating the first internucleotide hydrogen phosphonate linkage, and then oxidizing it with a first amine, generating the second hydrogen phosphonate
  • a 5'-capped oligonucleotide may be made.
  • the above method may be modified by first forming a polymer-bound oligonucleotide having only hydrogen phosphonate internucleotide linkages which may then be oxidized to form phosphodiesters (or
  • the 5'-end cap is formed when the last two or more nucleosides are added, followed by reaction with the amine NHR 1 R 2 .
  • the 5' end may be added by adding a polynucleotide, such as a tri- or tetranucleotide containing the desired phosphoramidate internucleotide linkages.
  • a polynucleotide such as a tri- or tetranucleotide containing the desired phosphoramidate internucleotide linkages.
  • a combination of both of the above methods for making a 5' and a 3' end- capped oligonucleotide may be utilized.
  • the first two (or more) internucleotide linkages on the 3'-bound oligonucleotide may be oxidized to form the
  • the non-terminal portion of the oligonucleotide may be made (having
  • 5'- or 3'-phosphoramidate-capped oligonucleotides as made in accordance with the present invention may be as therapeutic agents against viral diseases (such as HIV, hepatitis B, cytomegalovirus), cancers (such as leukemias, lung cancer, breast cancer, colon cancer) or metabolic disorders, immune modulation agents, or the like, since the present end-capped oligonucleotides are stable within the environment of a cell as well as in extracellular fluids such as serum, and can be used to selectively block protein synthesis, transcription, replication of RNA and/or DNA which is uniquely associated with the disease or disorder.
  • viral diseases such as HIV, hepatitis B, cytomegalovirus
  • cancers such as leukemias, lung cancer, breast cancer, colon cancer
  • immune modulation agents or the like
  • the end-capped oligonucleotides of the invention may also be used as therapeutics in animal health care, plant gene regulation (such as plant growth promoters) or in human diagnostics, such as to stabilize DNA probes to detect microorganisms, oncogenes, genetic defects, and the like, and as research reagents to study gene functions in animal cells, plant cells, microorganisms, and viruses.
  • plant gene regulation such as plant growth promoters
  • human diagnostics such as to stabilize DNA probes to detect microorganisms, oncogenes, genetic defects, and the like
  • research reagents to study gene functions in animal cells, plant cells, microorganisms, and viruses.
  • dermatologic applications for treatment of diseases or for cosmetic purposes.
  • Polymer-bound polynucleoside H-phosphonates were prepared as described by Froehler et al., supra, on control pore glass using the DBU salt of the protected nucleoside H-phosphonate.
  • the diester linkages were generated by aqueous I 2 oxidation and the amidate
  • the oligomer was removed from the solid support, deprotected with cone. NH 4 OH (45°C/18 hr.), and purified by HPLC (PRP) using an acetonitrile (CH 3 CN) gradient in 50 mM aqueous TEAP.
  • the DMT was removed from the product fraction (80% acetic acid/R.T./2 hrs.), evaporated, desalted and evaporated. Approximately 1 ⁇ g of purified product was 5' end-labeled with T4 poly- nucleotide kmase and ⁇ - 32 P ATP for further
  • Polymer-bound polynucleoside H-phosphonates were prepared as in the preceding example on control pore glass using the DBU salt of the protected nucleoside H-phosphonate. After twelve couplings the polynucleoside H-phosphonate was oxidized with aq. I 2 (0.1 M in N-methyl morpholine/water/THF, 5:5:90) followed by two more couplings and oxidation with a solution of 2-methoxy- ethylamine in Pyr/CCl 4 (1:5:5) (20 min.) to generate a 15-mer containing twelve diester linkages at the 3' end and two phosphoramidate linkages at the 5' end.
  • aq. I 2 0.1 M in N-methyl morpholine/water/THF, 5:5:90
  • 2-methoxy- ethylamine in Pyr/CCl 4 (1:5:5) 20 min.
  • the oligomer was removed from the solid support, deprotected with cone. NH 4 OH (45°C/18 hr.) and purified by HPLC (PRP) using an acetonitrile (CH 3 CN) gradient in 50 mM aqueous TEAP. The DMT was removed from the product fraction (80% acetic acid/R.T./2 hrs.), evaporated, desalted and evaporated.
  • Polymer-bound polynucleoside H-phosphonates were prepared as described as in the preceding examples on control pore glass using the DBU salt of the protected nucleoside H-phosphonate.
  • the diester linkages were generated by aqueous I 2 oxidation and the amidate
  • polynucleoside H-phosphonate was oxidized with a solution of 2-methoxyethylamine in Pyr/CCl 4 (1:5:5) (20 min.) followed by ten more couplings and oxidation with aq. I 2 (0.1 M in N-methyl morpholine/water/THF, 5:5:90) to generate a 13-mer containing two phosphoramidate linkages at the 3' end and ten diester linkages. This was
  • Example 1 The procedure of Example 1 was repeated using dodecylamine to generate a 15-mer containing two phosphoramidate linkages at the 3' end and twelve diester linkages, wherein the phosphoramidate linkages are such that one of R 1 and R 2 as defined earlier herein is hydrogen and the other is dodecyl.
  • Example 2 The procedure of Example 2 was repeated using dodecylamine in place of 2-methoxyethylamine, so as to yield a 15-mer containing twelve diester linkages at the 3' end and two phosphoramidate linkages at the 5' end, wherein the phosphoramidate linkages are substituted as in the preceding example, i.e., one of R 1 and R 2 is hydrogen and the other is dodecyl.
  • Example 3 The procedure of Example 3 was repeated using dodecylamine in place of 2-methoxyethylamine, to give rise to a 15-mer containing two phosphoramidate linkages at the 3' end, ten diester linkages, and two phosphoramidate linkages at the 5' end, wherein the phosphoramidate is N-substituted as in the preceding two
  • Example 8 The procedure of Example 1 was repeated using propylamine to generate a 15-mer containing two phos- phoramidate linkages at the 3' end and twelve diester linkages, wherein the phosphoramidate linkages are such that one of R 1 and R 2 as defined earli.er herein is hydrogen and the other is n-propyl.
  • Example 8
  • Example 2 The procedure of Example 2 was repeated using propylamine in place of 2-methoxyethylamine, so as to yield a 15-mer containing twelve diester linkages at the 3' end and two phosphoramidate linkages at the 5' end, wherein the phosphoramidate linkages are substituted as in the preceding example, i.e., one of R 1 and R 2 is hydrogen and the other is n-propyl.
  • Example 3 The procedure of Example 3 was repeated using propylamine in place of 2-methoxyethylamine, to give rise to a 15-mer containing two phosphoramidate linkages at the 3' end, ten diester linkages, and two phosphoramidate linkages at the 5' end, wherein the phosphor- amidate is N-substituted as in the preceding two
  • Example describes hybridization stability studies performed using end-capped oligonucleotides as described and claimed herein.
  • Oligonucleotides containing end-caps were tested for their ability to form stable duplexes with complementary single-stranded DNA sequences; the various oligonucleotides tested were outlined below in Table 1. Duplex stability was measured by determining the melting temperature T m in solution over a range of temperatures. The experiment was conducted in a solution containing 150 mM NaCl, 5 mM Na 2 HPO 4 and 3 ⁇ M DNA at a pH of 7.1. The results obtained and set forth in Table 1 show that binding to complementary sequences is not materially affected by 3'-end-cap modification.
  • the acute infection assay used the MOLT-4 cell line which is susceptible to HIV infection. Measurement of HIV p24 was used to assay for inhibition of virus replication 7 days after infection with virus at a multiplicity of infection of approximately 0.1.
  • oligonucleotide Approximately 1 x 106 cells were preincubated with oligonucleotide, washed, infected with virus stock and then incubated for 7 days in oligonucleotide. HIV p24 levels in the supernatant were measured by
  • Toxieity data was obtained by incubation of 3'-end-capped oligonucleotides with uninfected cells, followed by a comparison with cell numbers with control cultures incubated in the absence of oligonucleotide. Toxieity results are expressed as the percent reduction of cell numbers obtained by incubation in oligonucleotide for 7 days compared to controls.
  • the effective inhibition of HIV replication using low levels (0.5 to 5 ⁇ M) of capped oligodeoxynucleotides supports the conclusion that significant nuclease degradation of the oligonucleotides of the invention does not occur either extracellularly or intracellulary.

Abstract

A method is provided for making 3' and/or 5' end-capped oligonucleotides so as to render the oligonucleotide resistant to degradation by exonucleases. The exonuclease degradation resistance is provided by incorporating two or more phosphoramidate and phosphoromonothioate and/or phosphorodithioate linkages at the 5' and/or 3' ends of the oligonucleotide, wherein the number of phosphoramidate linkages is less than a number which would interfere with hybridization to a complementary oligonucleotide strand and/or which would interfere with RNAseH activity when the oligonucleotide is hybridized to RNA.

Description

EXONUCLEASE-RESISTANT
OLIGONUCLEOTIDES AND METHODS FOR PREPARING THE SAME
Description
Technical Field
The present invention is directed to oligonucleotides containing a 3'- and/or 5'-capped terminal and which are thereby rendered resistant to degradation by exonucleases. The exonuclease-resistant oligonucleotides have two or more phosphoramidate
internucleotide linkages at one or both termini which render the oligonucleotides resistant to degradation. Background
DNA molecules contain internucleotide phosphodiester linkages which are degraded by
exonucleases present in cells, culture media and human serum. For example, degradation by exonucleases in tissue culture media of DNA may be observed within about 30 minutes to about six hours. Synthetic oligodeoxy- nucleotides with phosphodiester linkages are routinely used in genetic engineering, for example, to locate specific RNA or DNA fragments from a library. The long- term stability of an oligonucleotide for this utility is not a major concern, since the oligonucleotide is usually not exposed to the relatively stringent environment of the culture medium, therefore exonuclease degradation is not a substantial problem.
However, it is in fact frequently desirable to produce oligodeoxynucleotides which are stable (i.e., for more than several hours or days) for long-term uses. For example, a oligodeoxynucleotide with phosphodiester linkages can be used to block protein synthesis by hydrogen bonding to complementary messenger RNA thereby providing a tool for use in an antisense fashion.
Exonuclease-stable oligodeoxynucleotides could also be utilized to form triple-helix DNA which would interfere with the transcription process or with DNA replication, by competing with naturally occurring binding factors or by gene destruction. However, in order to utilize synthetic oligonucleotides in this manner, they must be stable to exonucleases, the major activity of which in cells and serum appears to be 3' to 5', i.e., digestion of oligonucleotides begins starting at the 3' end.
The present invention is accordingly directed to such exonuclease-stable oligonucleotides.
Related Art:
The following references relate to one or more aspects of the presently claimed invention:
Froehler, Tet. Lett. 27 (46) :5575-5578 (1986), describes polymer-bound deoxynucleoside H-phosphonate diesters as precursors to phosphoramidate, thiophosphate and phosphate triester analogs of DNA.
Froehler et al., Nuc. Acids Res. 16 (11) :4831- 4839 (1988), describe the synthesis of a 15-mer
containing 12 phosphoroamidate linkages derived from primary and secondary amines. The chemistry of the process is summarized in the figure shown on page 4833 of the reference.
Froehler et al., Nuc. Acids Res. 14 (13) :5399-
5407 (1986), describe the synthesis of deoxyoligo- nucleotides via deoxynucleoside H-phosphonate
intermediates. The chemistry of this process is
essentially shown in scheme 2 on page 5401 of the reference. Froehler, European Patent Publication No.
219342-A2, published 2 April 1987, is similar to the teachings of the latter two references in that the synthesis of DNA via deoxynucleoside H-phosphonate intermediates is shown.
Letsinger et al., Nuc. Acids Res. 14 (8) :3487- 3499 (1986), describe complexes of polyuridylic acid (poly U) and polythymidylic acid (poly dT) with
oligonucleotides possessing different pendant groups that are linked to the oligonucleotide chain at the
internucleotide phosphodiester linkages.
Stein et al., Nuc. Acids Res. 16 (8) :3209-3221, (1988) present a study of oligodeoxynucleotides modified so as to contain phosphorothioate linkages. The authors, in addition to evaluating a number of other physico- chemical properties of such oligonucleotides, study the susceptibilities of the compounds to a number of
endonucleases and exonucleases. The authors found a significant decrease in the Tm of fully substituted phosphorothioate oligodeoxynucleotides compared to diester controls (Figure 3), i.e., a 15-20°C decrease in Tm and a 30-40 Kcal/mole decrease in ΔH for fully
substituted molecules (p. 3215).
Brill et al., Tet. Lett. 29 (43) :5517-5520
(1988) describe the preparation of dinucleoside phosphorodithioates by sulfur oxidation of thiophosphate triesters.
Agrawal, Tet. Lett. 28 (311:3539-3542 (1987) describe the automated synthesis of oligodeoxynucleosides containing methylphosphonate linkages, using nucleoside methylphosphonamidites as starting materials. The authors conclude that two adjacent methylphosphonate linkages at the 3' end provides protection against degradation by snake venom phosphodiesterase and spleen phosphodiesterase (and, like Stein et al., the authors do not evaluate nuclease stability of the oligonucleotides in serum, tissue culture medium or cells).
PCT publication WO89/05358, inventors Walder et al., describe oligodeoxynucleotides modified at the 3' terminus so as to render the oligonucleotide chain resistant to degradation within cells and body fluids. Disclosed modifications at the 3'-terminal phosphodiester linkage include replacement of that linkage with an alkyl or aryl phαsphotriester, hydrogen phosphonate, an alkyl or aryl phosphonate, an alkyl or aryl phosphoramidate, a phosphorothioate, or a phosphoroselenate, although the preferred modification is stated to be the incorporation of a 3'-terminal phosphotriester linkage. Disclosure of the Invention
Accordingly, it is a primary object of the invention to address the above-mentioned need in the art and to provide exonuclease-resistant oligonucleotides.
It is another object of the invention to provide exonuclease-resistant oligonucleotides modified at the 3'-terminus so that the initially present
phosphodiester linkages are replaced with a specified number of phosphoramidate linkages.
It is still another object of the invention to provide such exonuclease-resistant oligonucleotides which additionally contain phosphoromonothioate and/or
phosphorodithioate linkages.
It is yet another object of the invention to provide exonuclease-resistant oligonucleotides which are capable of hybridizing to a complementary oligonucleotide strand.
It a further object of the invention to provide methods of making such exonuclease-resistant oligonucleotides. It is still a further object of the invention to provide a method for end-capping oligonucleotides with moieties which can perform multiple functions, such as aiding in transport, serving as chromophoric tags, or enabling cross-linking.
Additional objects, advantages and novel features of the invention will be set forth in part in the description which follows, and in part will become apparent to those skilled in the art upon examination of the following, or may be learned by practice of the invention.
In a first aspect, the present invention provides oligonucleotides having two or more phosphoramidate linkages at the 3' terminus and/or 5' terminus, which oligonucleotides are resistant to
exonuclease degradation. The number of phosphoramidate linkages is at least 1 and less than a number which would interfere with hybridization to a complementary oligonucleotide strand, and/or less than a number which would interfere with RNAse activity when said oligonucleotide is hybridized to RNA. Preferably, at least 2, and more preferably on the order of about 2 to 10, phosphoramidate linkages are incorporated at either or both the 3' terminus and the 5' terminus. The phosphoramidate linkages may be substituted with any one of a number of different types of moieties as will be described in detail hereinbelow.
In another aspect, exonuclease-resistant are provided which have the following formulas I, II or III, i.e., containing phosphoramidate linkages as just described as well as phosphoromonothioate and/or
phosphorodithioate linkages:
Figure imgf000008_0001
wherein each n, m, i, j and s is independently an integer and each s is in the range of about 2 to 10; each n and m is independently from 1 to about 50; s + n in formulas I and II is less than 100; and s + s + m in formula III is less than about 100; each i varies from 1 to n; each j varies from 1 to m; T is hydrogen or a hydroxyl- protecting group; R1 and R2 are moities independently selected from the group consisting of hydrogen,
hydrocarbyl substituents of 20 carbon atoms or less, and oxyhydrocarbyl of 20 carbon atoms or less and 1-3 oxy groups, wherein said hydrocarbyl and oxyhydrocarbyl substituents are linear or branched alkyl of 1 to 20 carbon atoms, linear or branched alkenyl of 2 to 20 carbon atoms, cycloalkyl or cycloalkenyl of 3 to 20 carbon atoms, linear or branched alkoxy of 1 to 20 carbon atoms, or aryl of 6 to 18 carbon atoms, with the proviso that R1 and R2 are not both hydrogen;
each B is independently a protected or unprotected heterocyclic base;
each Xi and X j is independently O or S ; and each Yi and Yj is independently R, -SR or -OR, where R is as defined for R1 and R2.
The present invention also provides methods for preparing such end-capped oligonucleotides.
Modes for Carrying Out the Invention
As used herein the terms "polynucleotide" and "oligonucleotide" shall be generic to polydeoxyribo- nucleotides (containing 2'-deoxy-D-ribose or modified forms thereof), to polyribonucleotides (containing D- ribose or modified forms thereof), and to any other type of polynucleotide which is an N-glycoside of a purine or pyrimidine bases, or modified purine or pyrimidine bases. The term "nucleoside" will similarly be generic to ribonucleosides, deoxyribonucleosides, or to any other nucleoside which is an N-glycoside of a purine or
pyrimidine base, or modified purine or pyrimidine base. There is no intended distinction in length between the term "polynucleotide" and "oligonucleotide" and these terms will be used interchangeably.
It will be appreciated that as used herein the terms "nucleoside" and "nucleotides" will include those moieties which contain not only the known purine and pyrimidine bases, i.e., adenine, thymine, cytosine, guanine and uracil, but also other heterocyclic bases which contain protecting groups or have been otherwise modified or derivatized.
By "modified nucleosides" or "modified nucleotides" as used herein are intended to include those compounds containing one or more protecting groups such as acyl, isobutyryl, benzoyl, or the like, as well as any of the wide range of modified and derivatized bases as known in the art. Examples of such modified or deriva- tized bases include 5-fluorouracil, 5-bromouracil,
5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil,
5-carboxymethylaminomethyl-2-thiouridine, 5-carboxy- methylaminomethyluracil, dihydrouraσil, beta-D-galacto- sylqueosine, inosine, N6-isopentenyladenine, 1-methyl- adenine, 1-methylpseudouracil, 1-methylguanine,
1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-ethylguanine, 3-methylcytosine, 5-methylcytosine,
N6-methyladenine, 7-methylguanine, 5-methylamino- methyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5'-methoxycarbonylmethyluracil,
5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, 5-methyl- 2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 3-(3-amino-3-N-2-carboxypropyl) Uracil, and 2,6-diaminopurine.
Modified nucleosides or nucleotides can also include modifications on the sugar moiety, for example, wherein one or more of the hydroxyl groups are replaced with halogen or aliphatic groups, or functionalized as ethers, amines, etc.
The polynucleotides according to the present invention may be of any length, but lengths of about three to about fifty nucleotides are particularly useful for most genetic engineering applications. According to the present invention, the 3' end and/or the 5' end of the polynucleotide will contain at least two
phosphoramidate internucleotide linkages. The remaining internucleotide linkages may be phosphodiester linkages, phosphorothioate linkages or phosphorodithioate linkages, or any other internucleotide linkage, other than a phosphoramidate, or combinations of these other linkages. Methods for preparing such non-phosphoramidate linkages are known in the art, e.g., as taught by Froehler et al., Nuc. Acids Res. 14:5399-5467 (1986), and Froehler, B., Tet. Lett. 27:5575-5578 (1986), cited above and
incorporated by reference herein.
Internucleotide phosphodiester linkages are prepared from hydrogen phosphonate linkages preferably by oxidation with, e.g., aqueous iodine. A typical
procedure involves treatment of the hydrogen phosphonate in 0.1 M iodine in Pyr/NMI/H2O/THF (5:1:5:90) for about 2-3 minutes, followed by treatment with 0.1 M iodine in Et3/H2O/THF (5:5:90) for another approximately 2-3 minutes.
Phosphoromonothioate linkages are formed from the initially present hydrogen phosphonate linkages by treatment with sulfur. The reaction is carried out at approximately room temperature for on the order of 20 minutes in a solvent system which typically includes a sulfur solvent such as CS2 along with a basic solvent such as pyridine. Other suitable solvent systems include CS2/lutidine and CS2/triethylamine; CS2 is preferred as the sulfur solvent because it acts to dissolve elemental sulfur. The following scheme illustrates the postulated reaction:
Figure imgf000012_0001
(See, e.g., Stein et al., cited above.)
To form hydrogen phosphorodithioate linkages, sulfurization of the hydrogen phosphoromonothioate linkages is effected using conditions identical to those just described for the preparation of the phosphoromono- thioate moiety. (Note: the term "phosphorothioate" as used herein is intended to encompass both "phosphoromonothioate" and "phosphorodithioate" linkages.)
Structure of the End-Capped Oligonucleotides: The oligonucleotides of the invention, as noted above, are resistant to degradation under both physiological and tissue culture conditions, and in particular are resistant to degradation by exonucleases.
In order that the oligonucleotide be resistant to such enzymatic degradation, it is modified so that phosphodiester linkages initially present at the 3' terminus are replaced with a selected number of phosphoramidate linkages, that number being at least one and less than a number which would cause interference with hybridization to a complementary oligonucleotide strand, and/or less than a number which would interfere with RNAseH activity when said the oligonucleotide is
hybridized to RNA. Such a modification may additionally or alternatively be made at the 5' terminus.
It is preferred that the number of phosphoramidate linkages be selected such that the melting
temperature of any duplex formed with complement is lowered by less than about 10°C relative to that obtained with an oligonucleotide containing only the initial phosphodiester linkages. Preferably, the number of phosphoramidate linkages is such that the melting
temperature of a duplex formed is lowered by less than about 5°C. The number of phosphoramidate linkages present is typically and preferably between about 2 and 10, more preferably between about 2 and 8, and most preferably between about 2 and 6.
The phosphoramidate linkage has the formula
Figure imgf000013_0001
wherein the R1 and R2 moieties are substituents which must be selected so as not to interfere with
hybridization with complement. In most cases, the groups
R1 and R2 are independently selected from the group consisting of hydrogen, hydrocarbyl substituents of
20 carbon atoms or less, and oxyhydrocarbyl substituents of 20 carbon atoms or less containing 1-3 oxy groups, with the proviso that R1 and R2 are not both hydrogen, i.e., the phosphoramidate linkages herein are always N-substituted. In this case, it is preferred that one of the two substituents be hydrogen. Suitable hydrocarbyl and oxyhydrocarbyl substituents include, for example, linear or branched alkyl of 1-20 carbon atoms, linear or branched alkenyl of 2-20 carbon atoms, cycloalkyl or cycloalkenyl of 3-20 carbon atoms, linear or branched alkoxy of 1-20 carbon atoms, or aryl of 6-18 carbon atoms. The hydrocarbyl substituent may be, for example, an alkoxy substituent having the formula CH3O-(CH2)x- or a straight chain alkyl group having the formula CH3(CH2)y - where x is an integer in the range of 1-20, inclusive, preferably in the range of 1-10, inclusive, and y is an integer in the range of 0-15, inclusive.
Examples of preferred oligonucleotide linkages within the aforementioned groups are wherein one of R1 and R2 is H and the other is either 2-methoxyethyl, dodecyl, or n- propyl. (The 2-methoxyethyl and dodecyl linkages are sometimes referred to herein as "MEA" and "C12",
respectively.)
The R1 and R2 groups may also be, in addition to the foregoing, macromolecular species such as sugars, polypeptides, chromophoric groups, lipophilic groups, polymers, steroid hormones, or the like.
"Lipophilic" groups refer to moieties which are chemically compatible with the outer cell surface, i.e., so as to enable the oligonucleotide to attach to, merge with and cross the cell membrane. Examples of such lipophilic groups are fatty acids and fatty alcohols (in addition to the long chain hydrocarbyl groups described above).
Examples of preferred polypeptides that can be used for R1 and/or R2 include transferrin and epidermal growth factor (EGF), while suitable non-polypeptide polymers include ionic, nonionic and zwitterionic
polymers. Examples of a particularly preferred polymer is polyethylene glycol.
Steroid substituents include any of the general fatήily of lipid compounds which comprise sterols,
bioacids, cardiac glycosides, seponans, and sex hormones, which include the following basic structure:
Figure imgf000015_0001
Examples of steroids include natural corticosteroid hormones (produced by the adrenal glands), sex hormones (progesterone, androgens, and estrogens).
These various R1 and R2 groups can confer any of a variety of desired properties to the oligo- nucleotide. For example, if R1 or R2 is a polymer such as polyethylene glycol, a polypeptide or a lipophilic group such as a long-chain hydrocarbyl moiety, such a group may facilitate transport or permeation of the oligonucleotide through cell membranes, thus increasing the cellular uptake of the oligonucleotide. The R1 or R2 group may also be a group which affects target DNA or RNA to which the oligonucleotide will bind, such as providing covalent linkages to the target strand to facilitate cleavage or intercalation of the oligonucleotide to the target strand. The R1 and R2 groups may additionally serve a cutting function (e.g., a site for cutting the complementary strand), or a receptor function (e.g., a receptor ligand).
It will be appreciated by those skilled in the art that the oligonucleotides of the present invention can include other phosphoramidate N-substituents not explicitly disclosed herein so long as those substituents confer exonuclease resistance and do not interfere with hybridization to a complementary oligonucleotide strand. The invention also encompasses oligonucleotide compositions containing oligonucleotides of the following formula I, II or III, i.e., wherein phosphoromonothioate and/or phosphorodithioate linkages are incorporated in addition to the phosphoramidate linkages:
Figure imgf000016_0001
in which B, T, R1' R2, Xj, Yi, Yj, n, m, i, j and s are as defined above. In these structures, it is
preferred that "s," which defines the number of
phosphoramidate linkages, be in the range of 2-8, more preferably in the range of 2-6. It is also preferred that m and n be within the aforementioned ranges.
Synthetic Methods:
According to one embodiment of the present invention, the 3'-capped oligonucleotides may be prepared by first preparing a polymer-bound polynucleoside with the formula IV
Figure imgf000017_0001
wherein P is a solid state polymeric support, or other type of solid support, and B the base portion of a nucleoside, i.e., a purine or pyrimidine base, or any modified purine or pyrimidine base. As is conventional in oligonucleotide syntheses, the functional groups on the base, i.e., the amine groups, will be appropriately protected during the course of the synthesis and removed after the completed polynucleotide is removed from the polymer support. As is the convention, in the formula shown above in IV, the linkage to the polymer support is through the 3' hydroxy group, the free hydroxy group is the 5' group of the nucleoside. The group T is a
conventional hydroxy-protecting group used in
oligonucleotide synthesis, preferably the DMT group
(dimethoxytrityl) or MMT group (monomethoxytrityl).
The polymer-bound polynucleoside hydrogen phosphonate (IV) is preferably prepared by treating the DBU
(1.8-diazabicyclo[5.4.0]undec-7-ene ammonium salt) of a 5'-protected (preferably, 5 DMT) nucleoside hydrogen phosphonate with a polymer-bound nucleoside, linked to support through its 3'-hydroxyl group in the presence of an activating agent, as is known in the art. Methods for preparing such polymer-bound polynucleoside hydrogen phosphonates are disclosed, for example, by Froehler, B., et al., Nuc. Acids Res. 16:4831-4839 (1988); Froehler, B., et al., Nuc. Acids Res. 14:5399-5467 (1986); and Froehler, B., et al., Nucleosides and Nucleotides 6:287- 291 (1987). Then, one or more nucleoside hydrogen phosphonates may be added (to make the two or more internucleotide linkages at the 3' end of the
polynucleotide) by sequentially deprotecting the
5'-hydroxyl group of the polymer-bound polynucleotide, and condensing with the next nucleoside hydrogen
phosphonate. The oligonucleotide chain elongation will proceed in conformance with a predetermined sequence in a series of condensations, each one of which results in the addition of another nucleoside to the oligomer. The condensation is typically accomplished with dehydrating agents, which are suitably phosphorylating agents or acylating agents such as isobutylchloroformate,
diphenylchlorophosphate, organic acid anhydrides (such as acetic anhydride, isobutyric anhydride or trimethyl acetic anhydride) and organic acid halides such as pivaloyl chloride, pivaloyl bromide, 1-adamantyl- carboxylic chloride or benzoyl chloride. The preferred condensing agent is pivaloyl chloride in pyridine
acetonitrile. Prior to the addition of each successive nucleoside hydrogen phosphonate, the 5'-protecting group or the carrier bound nucleotide is removed. Typically, for removal of the DMT group, this is done by treatment with 2.5% volume/volume dichloroacetic acid/CH2Cl2, although 1% weight/volume trichloroacetic acid/CH2Cl2 or ZnBr2-saturated nitromethane are also useful. Other deprotection procedures suitable for other known
protecting groups will be apparent to those of ordinary skill in the art.
The carrier is preferably washed with anhydrous pyridine/acetonitrile (l/l,v/v) and the condensation reaction is completed in as many cycles as are required to form the desired number of 3'-end internucleotide bonds which will be converted to phosphoramidates. After the required number of synthetic cycles, the carrier- bound polynucleotide hydrogen phosphonate is oxidized to convert the hydrogen phosphonate internucleotide linkages to phosphoramidate linkages, preferably by treatment with the desired amine NHR1R2 with R1 and R2 as defined earlier and CCl4 as described in Froehler, et al.,
Nucleic Acids Research 16:4831-4839 (1988). Although carbon tetrachloride is preferred, other mild oxidizing agents may be utilized.
After the oxidation to form the phosphoramidate internucleotide linkages, the oligonucleotide is then completed by methods which form nonphosphoramidate linkages, such as phosphodiester linkages,
phosphorothioate linkages or phosphorodithioate linkages, by methods known in the art referenced above and
incorporated by reference herein. The preferred method for completing the oligonucleotide is to continue the sequence using 5'-protected nucleoside hydrogen- phosphonates. In the instance where the 5' end will not be capped, after the last 5'-protected nucleoside
hydrogen phosphonate has been added, all of the hydrogen phosphonate linkages are oxidized to produce diester linkages, preferably by aqueous iodine oxidation or oxidation using other oxidizing agents, such as
N-chlorosuccinimide, N-bromosuccinimide or salts or periodic acid. This will result in all of the
internucleotide linkages, except for the 3'-end capped linkages which are phosphoramidate linkages, being phosphodiester linkages. Thereafter, the oligonucleotide may be separated from the carrier, using conventional methods, which in the preferred instance is incubation with concentrated ammonium hydroxide. Any protecting groups may be removed as described above using about 2% dichloroacetic acid/CH2Cl2, or about 80% acetic acid, or by other conventional methods, depending on the nature of the protecting groups. The desired oligonucleotide is then purified by HPLC, polyacrylamide gel electro- phoresis or using other conventional techniques.
The following schemes illustrate various synthetic processes within the scope of the invention:
Scheme 1
Figure imgf000021_0001
T: protecting group
Figure imgf000021_0002
protecting group or solid state carrier
i: varies from 1 to 5
Q: hydrogen or -NR1R2 (with the proviso that
at least one Qi is hydrogen) B: a purine or pyrimidine base
R 1,R2: see text Scheme 2a
1) 5'-blockednucleosideH-phosphonate and pivaloyl chloride as activator
2) remove5'-blockinggroup
3) HNR1R2/oxidizing agent
Figure imgf000022_0004
4) optional repetition of steps 1-3
Figure imgf000022_0001
Figure imgf000022_0002
T,
Figure imgf000022_0003
Xi, Yj, R1, R2 = as defined in text and in Scheme 1
Scheme 2b
Figure imgf000023_0001
T, Xi, Yi, R1 , R2 , s, n, = as defined in text and in Schemes 1 and 2a Scheme 3
Figure imgf000024_0001
Figure imgf000024_0002
1) remove T
2) condense 5' - blocked nucleoside H-phosphonate,
H-phosphorothioate or H-phosphorodithioate with pivaloyl chloride as activator
3) remove 5' - blocking group
4) repeat steps 2) & 3)
5) oxidize to form phosphodiester, phosphorothioate and/or
phosphorodithioate linkages
Figure imgf000024_0004
6) condense with additional 5' - blocked nucleoside phosphonates, optionally react with HNR1R2 in the presence of an oxidizing agent
Figure imgf000024_0003
The foregoing discussion has revolved around the consecutive addition of mononucleoside hydrogen phosphonates, but it will be understood that one or more nucleotides can be added in a given cycle by using a polynucleotide, such as a di- or trinucleotide.
It will also be understood that while the above method has been described in connection with use of a solid state carrier if the object oligonucleotide is small, i.e., containing, for example, only five
nucleotides (therefore having only four internucleotide linkages, two of which are phosphoramidate linkages) it is feasible to conduct the synthesis without the use of a solid state support. In such an instance a conventional 3'-hydroxy protecting group may be used which is
different from the 5'-protecting group used in the synthesis, so that the 5'-protecting group may be
selectively removed while the 3'-protecting group remains intact.
It will also be appreciated that the two or more phosphoramidate linkages need not each contain the same R1 and R2 groups. This may be accomplished by generating the first internucleotide hydrogen phosphonate linkage, and then oxidizing it with a first amine, generating the second hydrogen phosphonate
internucleotide linkage, and then oxidizing it in the presence of a second (different) amine. This would result in a capped oligonucleotide having mixed
phosphoramidate internucleotide linkages.
In another embodiment of the present invention, a 5'-capped oligonucleotide may be made. In such an instance, the above method may be modified by first forming a polymer-bound oligonucleotide having only hydrogen phosphonate internucleotide linkages which may then be oxidized to form phosphodiesters (or
phosphorothioate or phosphorodithioate linkages). Then for the last two (or more) cycles, the 5'-end cap is formed when the last two or more nucleosides are added, followed by reaction with the amine NHR1R2.
Alternatively, the 5' end may be added by adding a polynucleotide, such as a tri- or tetranucleotide containing the desired phosphoramidate internucleotide linkages.
In still another embodiment, a combination of both of the above methods for making a 5' and a 3' end- capped oligonucleotide may be utilized. The first two (or more) internucleotide linkages on the 3'-bound oligonucleotide may be oxidized to form the
phosphoramidate linkages, then the non-terminal portion of the oligonucleotide may be made (having
phosphodiesters, phosphorothioate or phosphorodithioate internucleotide linkages), with the final two (or more) linkages being phosphoramidates, formed as described above. Methods of Use:
The uses of 5'- or 3'-phosphoramidate-capped oligonucleotides as made in accordance with the present invention may be as therapeutic agents against viral diseases (such as HIV, hepatitis B, cytomegalovirus), cancers (such as leukemias, lung cancer, breast cancer, colon cancer) or metabolic disorders, immune modulation agents, or the like, since the present end-capped oligonucleotides are stable within the environment of a cell as well as in extracellular fluids such as serum, and can be used to selectively block protein synthesis, transcription, replication of RNA and/or DNA which is uniquely associated with the disease or disorder. The end-capped oligonucleotides of the invention may also be used as therapeutics in animal health care, plant gene regulation (such as plant growth promoters) or in human diagnostics, such as to stabilize DNA probes to detect microorganisms, oncogenes, genetic defects, and the like, and as research reagents to study gene functions in animal cells, plant cells, microorganisms, and viruses. There may also be dermatologic applications for treatment of diseases or for cosmetic purposes. There are many other potential uses which derive from the stability of the oligonucleotide to exonuclease degradation, thus prolonging oligonucleotide integrity within the
relatively stringent environment of the cells.
It is to be understood that while the invention has been described in conjunction with the preferred specific embodiments thereof, that the foregoing
description and the examples which follow are intended to illustrate and not limit the scope of the invention.
Other aspects, advantages and modifications within the scope of the invention will be apparent to those skilled to which the invention pertains.
Example 1
Polymer-bound polynucleoside H-phosphonates were prepared as described by Froehler et al., supra, on control pore glass using the DBU salt of the protected nucleoside H-phosphonate. The diester linkages were generated by aqueous I2 oxidation and the amidate
linkages by amine/CCl4 oxidation. After two couplings the polynucleoside H-phosphonate was oxidized with a solution of 2-methoxyethylamine in Pyr/CCl4 (1:5:5) (20 min.) followed by twelve more couplings and oxidation with aq. I2 (0.1 M in N-methyl morpholine/water/THF, 5:5:90) to generate a 15-mer containing two phosphoramidate linkages at the 3' end and twelve diester
linkages. The oligomer was removed from the solid support, deprotected with cone. NH4OH (45°C/18 hr.), and purified by HPLC (PRP) using an acetonitrile (CH3CN) gradient in 50 mM aqueous TEAP. The DMT was removed from the product fraction (80% acetic acid/R.T./2 hrs.), evaporated, desalted and evaporated. Approximately 1 μg of purified product was 5' end-labeled with T4 poly- nucleotide kmase and γ-32P ATP for further
characterization.
Example 2
Polymer-bound polynucleoside H-phosphonates were prepared as in the preceding example on control pore glass using the DBU salt of the protected nucleoside H-phosphonate. After twelve couplings the polynucleoside H-phosphonate was oxidized with aq. I2 (0.1 M in N-methyl morpholine/water/THF, 5:5:90) followed by two more couplings and oxidation with a solution of 2-methoxy- ethylamine in Pyr/CCl4 (1:5:5) (20 min.) to generate a 15-mer containing twelve diester linkages at the 3' end and two phosphoramidate linkages at the 5' end. The oligomer was removed from the solid support, deprotected with cone. NH4OH (45°C/18 hr.) and purified by HPLC (PRP) using an acetonitrile (CH3CN) gradient in 50 mM aqueous TEAP. The DMT was removed from the product fraction (80% acetic acid/R.T./2 hrs.), evaporated, desalted and evaporated.
Example 3
Polymer-bound polynucleoside H-phosphonates were prepared as described as in the preceding examples on control pore glass using the DBU salt of the protected nucleoside H-phosphonate. The diester linkages were generated by aqueous I2 oxidation and the amidate
linkages by amine/CCl4. After two couplings the
polynucleoside H-phosphonate was oxidized with a solution of 2-methoxyethylamine in Pyr/CCl4 (1:5:5) (20 min.) followed by ten more couplings and oxidation with aq. I2 (0.1 M in N-methyl morpholine/water/THF, 5:5:90) to generate a 13-mer containing two phosphoramidate linkages at the 3' end and ten diester linkages. This was
followed by two more couplings and oxidation with a solution of 2-methoxyethylamine in Pyr/CCl4 (1:5:5) (20 min.) to generate a 15-mer containing two phosphoramidate linkages at the 3' end, ten diester linkages, and two phosphoramidate linkages at the 5' end. The oligomer was removed from the solid support and deprotected with cone. NH4OH (45°C/18 hr.) and purified by HPLC (PRP) using an acetonitrile (CH3CN) gradient in 50 mM aqueous TEAP. The DMT was removed from the product fraction (80% acetic acid/R.T./2 hrs.), evaporated, desalted and evaporated.
Example 4
The procedure of Example 1 was repeated using dodecylamine to generate a 15-mer containing two phosphoramidate linkages at the 3' end and twelve diester linkages, wherein the phosphoramidate linkages are such that one of R1 and R2 as defined earlier herein is hydrogen and the other is dodecyl.
Example 5
The procedure of Example 2 was repeated using dodecylamine in place of 2-methoxyethylamine, so as to yield a 15-mer containing twelve diester linkages at the 3' end and two phosphoramidate linkages at the 5' end, wherein the phosphoramidate linkages are substituted as in the preceding example, i.e., one of R1 and R2 is hydrogen and the other is dodecyl.
Example 6
The procedure of Example 3 was repeated using dodecylamine in place of 2-methoxyethylamine, to give rise to a 15-mer containing two phosphoramidate linkages at the 3' end, ten diester linkages, and two phosphoramidate linkages at the 5' end, wherein the phosphoramidate is N-substituted as in the preceding two
examples.
Example 7
The procedure of Example 1 was repeated using propylamine to generate a 15-mer containing two phos- phoramidate linkages at the 3' end and twelve diester linkages, wherein the phosphoramidate linkages are such that one of R1 and R2 as defined earli.er herein is hydrogen and the other is n-propyl. Example 8
The procedure of Example 2 was repeated using propylamine in place of 2-methoxyethylamine, so as to yield a 15-mer containing twelve diester linkages at the 3' end and two phosphoramidate linkages at the 5' end, wherein the phosphoramidate linkages are substituted as in the preceding example, i.e., one of R1 and R2 is hydrogen and the other is n-propyl.
Example 9
The procedure of Example 3 was repeated using propylamine in place of 2-methoxyethylamine, to give rise to a 15-mer containing two phosphoramidate linkages at the 3' end, ten diester linkages, and two phosphoramidate linkages at the 5' end, wherein the phosphor- amidate is N-substituted as in the preceding two
examples.
Example 10
The following Example describes hybridization stability studies performed using end-capped oligonucleotides as described and claimed herein.
Oligonucleotides containing end-caps were tested for their ability to form stable duplexes with complementary single-stranded DNA sequences; the various oligonucleotides tested were outlined below in Table 1. Duplex stability was measured by determining the melting temperature Tm in solution over a range of temperatures. The experiment was conducted in a solution containing 150 mM NaCl, 5 mM Na2HPO4 and 3 μM DNA at a pH of 7.1. The results obtained and set forth in Table 1 show that binding to complementary sequences is not materially affected by 3'-end-cap modification.
5
Figure imgf000032_0001
Example 11
Several additional oligonucleotides also end-capped at the 3' terminal two internucleotide linkages were tested for their ability to form stable duplexes with complementary single stranded DNA sequences, as described in the preceding example. Results are set forth in Table 2.
Figure imgf000033_0001
Example 12
The following example was used to determine the efficacy of end-capped oligodeoxynucleotides virus inhibition and cellular toxieity using oligonucleotides capped at two terminal 3'-end internucleotide linkages with 2-methoxyethylamine and dodecylamine.
The acute infection assay used the MOLT-4 cell line which is susceptible to HIV infection. Measurement of HIV p24 was used to assay for inhibition of virus replication 7 days after infection with virus at a multiplicity of infection of approximately 0.1.
Approximately 1 x 106 cells were preincubated with oligonucleotide, washed, infected with virus stock and then incubated for 7 days in oligonucleotide. HIV p24 levels in the supernatant were measured by
radioimmunoassay and compared with control infections lacking oligonucleotide. Results are expressed as the percent of control p24 found in cultures containing oligonucleotide. Sequences of antisense oligonucleotides were complementary to HIV targets listed in Table 3.
Toxieity data was obtained by incubation of 3'-end-capped oligonucleotides with uninfected cells, followed by a comparison with cell numbers with control cultures incubated in the absence of oligonucleotide. Toxieity results are expressed as the percent reduction of cell numbers obtained by incubation in oligonucleotide for 7 days compared to controls. The effective inhibition of HIV replication using low levels (0.5 to 5 μM) of capped oligodeoxynucleotides supports the conclusion that significant nuclease degradation of the oligonucleotides of the invention does not occur either extracellularly or intracellulary.
Figure imgf000035_0001

Claims

Claims
1. An oligonucleotide resistant to degradation under physiological conditions, which oligonucleotide is modified so that the phosphodiester linkages at the 3' and/or 5' termini are replaced with phosphoramidate linkages, the number of said linkages being at least 1 and less than a number which would interfere with
hybridization to a complementary oligonucleotide strand, and/or less than a number which would interfere with
RNAseH activity when said oligonucleotide is hybridized to RNA.
2. The oligonucleotide of claim 1 wherein the phosphoramidate has the formula:
Figure imgf000036_0001
wherein R1 and R2 are substituents which do not interfere with hybridization with complement.
3. The oligonucleotide of claim 2 wherein R1 and R2 are independently selected from the group
consisting of hydrogen, hydrocarbyl substituents of 20 carbon atoms or less, and oxyhydrocarbyl substituents of 20 carbon atoms or less containing 1 to 3 oxy groups, provided that R1 and R2 are not both hydrogen.
4. The oligonucleotide of claim 3 wherein one of R1 and R2 is hydrogen and the other is an oxyhydrocarbyl substituent having the structure CH3O-(CH2)x- wherein x is an integer in the range of 1 to 20,
inclusive.
5. The oligonucleotide of claim 4 wherein x is 2 and the oxyhydrocarbyl substituent is 2-methoxyethyl.
6. The oligonucleotide of claim 3 wherein one of R1 and R2 i.s hydrogen and the other is a hydrocarbyl substituent is a straight-chain alkyl moiety having the formula CH3(CH2)y- wherein y is an integer in the range of 0 to 15, inclusive.
7. The oligonucleotide of claim 6 wherein y is 11 and said hydrocarbyl substituent is dodecyl.
8. The oligonucleotide of claim 6 wherein y is 2 and said hydrocarbyl substituent is n-propyl.
9. The oligonucleotide of claim 1 wherein the oligonucleotide is modified at both the 3' and the 5' termini.
10. The oligonucleotide of claim 2 wherein one of R1 and R2 is hydrogen and the other is a polymer.
11. The oligonucleotide of claim 2, wherein one of R1 and R2 is hydrogen and the other is a sugar moiety.
12. The oligonucleotide of claim 2, wherein one of R1 amd R2 is hydrogen and the other is a
chromophore.
13. The oligonucleotide of claim 2, wherein one of R1 and R2 is hydrogen and the other is a steroid moiety.
14. The oligonucleotide of claim 1, wherein the number of said linkages is in the range of about 2 to 10.
15. The oligonucleotide of claim 1, further including 2 to 10 phosphorothioate linkages at either the 3' terminus, the 5' terminus, or both.
16. A method for synthesizing a polynucleotide stable to degradation by 3' exonucleases comprising the steps of:
(a) treating a nucleotide of the formula
Figure imgf000038_0001
wherein T is a protecting group, P is a protecting group or a solid state carrier, and each B is independently a protected or unprotected heterocyclic base, i varies from
1 to s; each Qi I .S hydrogen or -NR1R2, provi.ded that at least one Qi is hydrogen; with an amine of the formula HNR1R2 wherein R1 and R2 are defined in Claim 1, in the presence of an oxidizing agent sufficient to form the product of the formula (b) removing the blocking group T,
(c) condensing a 5'-blocked nucleoside H-phosphonate, H-phosphorothioate or H-phosphoro- dithioate, with the 5'-terminal hydroxy group of the carrier bound nucleotide in the presence of an activating agent;
(d) removing the 5'-blocking group of the 5'-terminal nucleoside;
(e) sequentially repeating steps (c) and (d) using 5'-blocked nucleoside H-phosphonate, H-phosphorothioate and/or H-phosphorodithioate, until said
polynucleotide of desired length is obtained;
(f) forming phosphodiester, phosphorothioate and/or phosphorodithioate internucleotide linkages by oxidation;
(g) separating said polynucleotide from P.
PCT/US1990/003138 1989-06-05 1990-06-05 Exonuclease-resistant oligonucleotides and methods for preparing the same WO1990015065A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
KR1019910701767A KR920701230A (en) 1989-06-05 1990-06-05 Exonuclease-resistant oligonucleotides and methods for preparing the same
CA002058632A CA2058632C (en) 1989-06-05 1990-06-05 Exonuclease-resistant oligonucleotides and methods for preparing the same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US36104589A 1989-06-05 1989-06-05
US361,045 1989-06-05

Publications (1)

Publication Number Publication Date
WO1990015065A1 true WO1990015065A1 (en) 1990-12-13

Family

ID=23420432

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/003138 WO1990015065A1 (en) 1989-06-05 1990-06-05 Exonuclease-resistant oligonucleotides and methods for preparing the same

Country Status (5)

Country Link
EP (1) EP0476071A4 (en)
JP (1) JPH05500799A (en)
KR (1) KR920701230A (en)
CA (1) CA2058632C (en)
WO (1) WO1990015065A1 (en)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991012323A1 (en) * 1990-02-15 1991-08-22 Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US5220007A (en) * 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
WO1993012135A1 (en) 1991-12-12 1993-06-24 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5434257A (en) * 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US5576208A (en) * 1991-06-14 1996-11-19 Isis Pharmaceuticals Inc. Antisense oligonucleotide inhibition of the RAS gene
US5580972A (en) * 1993-06-14 1996-12-03 Nexstar Pharmaceuticals, Inc. Purine nucleoside modifications by palladium catalyzed methods
WO1996040159A1 (en) * 1995-06-07 1996-12-19 Merck & Co., Inc. Capped synthetic rna, analogs, and aptamers
US5719273A (en) * 1993-06-14 1998-02-17 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modifications methods using nucleophiles and carbon monoxide
US5792608A (en) * 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5929226A (en) * 1992-07-27 1999-07-27 Hybridon, Inc. Antisense oligonucleotide alkylphosphonothioates and arylphospohonothioates
US5945527A (en) * 1996-05-30 1999-08-31 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modification methods using nucleophiles and carbon monoxide
US5959100A (en) * 1996-03-27 1999-09-28 Nexstar Pharmaceuticals, Inc. Pyrimidine nucleosides as therapeutic and diagnostic agents
US5965722A (en) * 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
US6007992A (en) * 1997-11-10 1999-12-28 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US6020483A (en) * 1998-09-25 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleoside modifications by palladium catalyzed methods
US6028183A (en) * 1997-11-07 2000-02-22 Gilead Sciences, Inc. Pyrimidine derivatives and oligonucleotides containing same
US6080727A (en) * 1996-03-26 2000-06-27 Istituto Regina Elena Oligonucleotide treatments and compositions for human melanoma
US6127533A (en) * 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
WO2001068048A2 (en) * 2000-03-17 2001-09-20 Unilever Plc Non-physiologic dna fragments for tanning skin
EP0676476B1 (en) * 1994-03-16 2001-10-24 Gen-Probe Incorporated Isothermal strand displacement nucleic acid amplification
EP1159282A1 (en) * 1999-02-12 2001-12-05 Isis Pharmaceuticals, Inc. Compounds, processes and intermediates for synthesis of mixed backbone oligomeric compounds
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
US6600032B1 (en) 1998-08-07 2003-07-29 Isis Pharmaceuticals, Inc. 2′-O-aminoethyloxyethyl-modified oligonucleotides
US6670393B2 (en) 1995-06-07 2003-12-30 Promega Biosciences, Inc. Carbamate-based cationic lipids
US6673912B1 (en) 1998-08-07 2004-01-06 Isis Pharmaceuticals, Inc. 2′-O-aminoethyloxyethyl-modified oligonucleotides
US7125945B2 (en) 2003-09-19 2006-10-24 Varian, Inc. Functionalized polymer for oligonucleotide purification
WO2007038658A2 (en) 2005-09-26 2007-04-05 Medarex, Inc. Antibody-drug conjugates and methods of use
US7255874B1 (en) 2001-12-21 2007-08-14 Closure Medical Corporation Biocompatible polymers and adhesives: compositions, methods of making and uses related thereto
EP2266986A1 (en) 2001-05-31 2010-12-29 Medarex, Inc. Cytotoxins, Prodrugs, Linkers and Stabilizers useful therefor
WO2011130289A1 (en) 2010-04-12 2011-10-20 Somalogic, Inc. 5-position modified pyrimidines and their use
US8716253B2 (en) * 2000-09-26 2014-05-06 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
WO2016011174A1 (en) * 2014-07-15 2016-01-21 Life Technologies Corporation Compositions and methods for nucleic acid assembly
US9938314B2 (en) 2013-11-21 2018-04-10 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
US10316321B2 (en) 2007-01-16 2019-06-11 Somalogic Inc. Method for generating aptamers with improved off-rates
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4415732A (en) * 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4849513A (en) * 1983-12-20 1989-07-18 California Institute Of Technology Deoxyribonucleoside phosphoramidites in which an aliphatic amino group is attached to the sugar ring and their use for the preparation of oligonucleotides containing aliphatic amino groups
EP0229053A1 (en) * 1985-06-14 1987-07-22 University Patents, Inc. Method for synthesizing deoxyoligonucleotides
JP3019994B2 (en) * 1987-11-30 2000-03-15 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Novel oligodeoxynucleotide, method of blocking expression of target gene using the same, and novel oligodeoxynucleotide and method of inhibiting expression of target gene using the same

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
Nucleic Acids Research, Volume 14, No. 13, issued 1986 (Oxford, England), FROEHLER et al., "Synthesis of DNA via deoxynucleoside H-phosphonate intermediates, see pages 5399-5407. *
Nucleic Acids Research, Volume 14, No. 8, issued 1986 (Oxford, England), LETSINGER et al. "Effects of pendant groups at phosphorus on binding properties of d-ApA analogues", see pages 3487-3499, especially", pages 3493-3494. *
Nucleic Acids Research, Volume 16, No. 11, issued 1988 (Oxford, England), FROEHLER et al, "Phosphoramidate analogues of DNA; synthesis and thermal stability of heteroduplexes, see pages 4831-4839. *
Nucleic Acids Research, Volume 16, No. 8, issued 1988, (Oxford, England), STEIN et al., "Physicochemical properties of phosphorothioate oligodeoxynucleotides", see pages 3209-3221. *
Nucleosides and Nucleotides, Volume 6, No. 1 and 2, issued 1987, BRIAN C. FROEHLER et al, "The Use of Nucleoside H-phosphonates in the synthesis of Deoxyoligonucleotides", see pages 287-291. *
See also references of EP0476071A4 *
Tetrahedron Letters, Volume 27, No. 46, issued 1986, (Great Britain), BRIAN C FROEHLER, "Deoxynucleoside H-phosphonate Diester Intermediates in the Synthesis of Internucleotide Phosphate Analogues", see pages 5575-5578. *

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5635377A (en) * 1990-02-15 1997-06-03 Worcester Foundation For Experimental Biology, Inc. Method of site-specific alteration of RNA and production of encoded polypeptides
US5220007A (en) * 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
WO1991012323A1 (en) * 1990-02-15 1991-08-22 Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US5965722A (en) * 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
US5576208A (en) * 1991-06-14 1996-11-19 Isis Pharmaceuticals Inc. Antisense oligonucleotide inhibition of the RAS gene
US5792608A (en) * 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
WO1993012135A1 (en) 1991-12-12 1993-06-24 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5434257A (en) * 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US5929226A (en) * 1992-07-27 1999-07-27 Hybridon, Inc. Antisense oligonucleotide alkylphosphonothioates and arylphospohonothioates
US5783679A (en) * 1993-06-14 1998-07-21 Nexstar Pharmaceuticals, Inc. Purine nucleoside modifications by palladium catalyzed methods and oligonucleotides containing same
US5719273A (en) * 1993-06-14 1998-02-17 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modifications methods using nucleophiles and carbon monoxide
US5580972A (en) * 1993-06-14 1996-12-03 Nexstar Pharmaceuticals, Inc. Purine nucleoside modifications by palladium catalyzed methods
EP0676476B1 (en) * 1994-03-16 2001-10-24 Gen-Probe Incorporated Isothermal strand displacement nucleic acid amplification
US6359124B1 (en) 1995-04-03 2002-03-19 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
US6111095A (en) * 1995-06-07 2000-08-29 Merck & Co., Inc. Capped synthetic RNA, analogs, and aptamers
US5861501A (en) * 1995-06-07 1999-01-19 Merck & Co., Inc. Capped synthetic RNA, analogs, and aptamers
WO1996040159A1 (en) * 1995-06-07 1996-12-19 Merck & Co., Inc. Capped synthetic rna, analogs, and aptamers
US6369208B1 (en) 1995-06-07 2002-04-09 Merck & Co., Inc. Capped synthetic RNA, analogs, and aptamers
US6670393B2 (en) 1995-06-07 2003-12-30 Promega Biosciences, Inc. Carbamate-based cationic lipids
US6080727A (en) * 1996-03-26 2000-06-27 Istituto Regina Elena Oligonucleotide treatments and compositions for human melanoma
US5959100A (en) * 1996-03-27 1999-09-28 Nexstar Pharmaceuticals, Inc. Pyrimidine nucleosides as therapeutic and diagnostic agents
US6143882A (en) * 1996-03-27 2000-11-07 Nexstar Pharmaceuticals, Inc. Urea nucleosides as therapeutic and diagnostic agents
US6441161B1 (en) 1996-03-27 2002-08-27 Gilead Sciences, Inc. Urea nucleosides as therapeutic and diagnostic agents
US6914138B2 (en) 1996-03-27 2005-07-05 Gilead Sciences, Inc. Urea nucleosides as therapeutic and diagnostic agents
US5945527A (en) * 1996-05-30 1999-08-31 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modification methods using nucleophiles and carbon monoxide
US6127533A (en) * 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6194598B1 (en) 1997-02-14 2001-02-27 Isis Pharmaceuticals, Inc. Aminooxy-modified oligonucleotide synthetic intermediates
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
US6825331B2 (en) 1997-02-14 2004-11-30 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers, oligomer arrays and methods of using them
US6028183A (en) * 1997-11-07 2000-02-22 Gilead Sciences, Inc. Pyrimidine derivatives and oligonucleotides containing same
US6951931B2 (en) 1997-11-07 2005-10-04 Isis Pharmaceuticals, Inc. Pyrimidine derivatives for labeled binding partners
US6414127B1 (en) 1997-11-07 2002-07-02 Isis Pharmaceuticals, Inc. Pyrimidine derivatives for labeled binding partners
USRE39324E1 (en) * 1997-11-07 2006-10-03 Isis Pharmaceuticals, Inc. Pyrimidine derivatives and oligonucleotides containing same
US6800743B2 (en) 1997-11-07 2004-10-05 Isis Pharmaceuticals, Inc. Pyrimidine derivatives for labeled binding partners
US7511125B2 (en) 1997-11-07 2009-03-31 Carlsbad Pyrimidine derivatives for labeled binding partners
US6007992A (en) * 1997-11-10 1999-12-28 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US6673912B1 (en) 1998-08-07 2004-01-06 Isis Pharmaceuticals, Inc. 2′-O-aminoethyloxyethyl-modified oligonucleotides
US6600032B1 (en) 1998-08-07 2003-07-29 Isis Pharmaceuticals, Inc. 2′-O-aminoethyloxyethyl-modified oligonucleotides
US6512106B2 (en) 1998-09-25 2003-01-28 Gilead Sciences, Inc. Nucleoside modifications by palladium catalyzed methods
US6020483A (en) * 1998-09-25 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleoside modifications by palladium catalyzed methods
US6355787B1 (en) 1998-09-25 2002-03-12 Gilead Sciences, Inc. Purine nucleoside modifications by palladium catalyzed methods and compounds produced
US6846918B2 (en) 1998-09-25 2005-01-25 Gilead Sciences, Inc. Nucleoside modifications by palladium catalyzed methods
EP1159282A4 (en) * 1999-02-12 2002-08-28 Isis Pharmaceuticals Inc Compounds, processes and intermediates for synthesis of mixed backbone oligomeric compounds
EP1159282A1 (en) * 1999-02-12 2001-12-05 Isis Pharmaceuticals, Inc. Compounds, processes and intermediates for synthesis of mixed backbone oligomeric compounds
WO2001068048A3 (en) * 2000-03-17 2002-02-21 Unilever Plc Non-physiologic dna fragments for tanning skin
WO2001068048A2 (en) * 2000-03-17 2001-09-20 Unilever Plc Non-physiologic dna fragments for tanning skin
US8716253B2 (en) * 2000-09-26 2014-05-06 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
EP2266986A1 (en) 2001-05-31 2010-12-29 Medarex, Inc. Cytotoxins, Prodrugs, Linkers and Stabilizers useful therefor
US7255874B1 (en) 2001-12-21 2007-08-14 Closure Medical Corporation Biocompatible polymers and adhesives: compositions, methods of making and uses related thereto
US7125945B2 (en) 2003-09-19 2006-10-24 Varian, Inc. Functionalized polymer for oligonucleotide purification
WO2007038658A2 (en) 2005-09-26 2007-04-05 Medarex, Inc. Antibody-drug conjugates and methods of use
US11111495B2 (en) 2007-01-16 2021-09-07 Somalogic, Inc. Method for generating aptamers with improved off-rates
US10316321B2 (en) 2007-01-16 2019-06-11 Somalogic Inc. Method for generating aptamers with improved off-rates
EP2558478A4 (en) * 2010-04-12 2013-10-23 Somalogic Inc 5-position modified pyrimidines and their use
EP2558478A1 (en) * 2010-04-12 2013-02-20 Somalogic, Inc. 5-position modified pyrimidines and their use
EP2558586A4 (en) * 2010-04-12 2014-10-15 Somalogic Inc APTAMERS TO ß-NGF AND THEIR USE IN TREATING ß-NGF MEDIATED DISEASES AND DISORDERS
US9163056B2 (en) 2010-04-12 2015-10-20 Somalogic, Inc. 5-position modified pyrimidines and their use
WO2011130289A1 (en) 2010-04-12 2011-10-20 Somalogic, Inc. 5-position modified pyrimidines and their use
KR102185779B1 (en) 2010-04-12 2020-12-03 소마로직, 인크. 5-position modified pyrimidines and their use
EP3345915A1 (en) 2010-04-12 2018-07-11 Somalogic, Inc. 5-position modified pyramidines and their use
KR20190018750A (en) * 2010-04-12 2019-02-25 소마로직, 인크. 5-position modified pyrimidines and their use
US10221207B2 (en) 2010-04-12 2019-03-05 Somalogic, Inc. 5-position modified pyrimidines and their use
EP2558586A1 (en) * 2010-04-12 2013-02-20 Somalogic, Inc. APTAMERS TO ß-NGF AND THEIR USE IN TREATING ß-NGF MEDIATED DISEASES AND DISORDERS
US10239908B2 (en) 2013-11-21 2019-03-26 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
US9938314B2 (en) 2013-11-21 2018-04-10 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
WO2016011174A1 (en) * 2014-07-15 2016-01-21 Life Technologies Corporation Compositions and methods for nucleic acid assembly
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto

Also Published As

Publication number Publication date
EP0476071A1 (en) 1992-03-25
KR920701230A (en) 1992-08-11
CA2058632C (en) 2004-08-24
EP0476071A4 (en) 1992-11-04
CA2058632A1 (en) 1990-12-06
JPH05500799A (en) 1993-02-18

Similar Documents

Publication Publication Date Title
US5256775A (en) Exonuclease-resistant oligonucleotides
WO1990015065A1 (en) Exonuclease-resistant oligonucleotides and methods for preparing the same
US5264564A (en) Oligonucleotide analogs with novel linkages
US5264562A (en) Oligonucleotide analogs with novel linkages
AU653504B2 (en) Oligonucleotide analogs with novel linkages
JP5677716B2 (en) Kits and methods comprising oligonucleotide analogues and use of oligonucleotide analogues
US5434257A (en) Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
AU662298B2 (en) Modified internucleoside linkages
JP5342881B2 (en) 6-modified bicyclic nucleic acid analogues
EP2625186B1 (en) 5' modified nucleosides and oligomeric compounds prepared therefrom
JP2004500330A (en) Guanidinium-functionalized oligomers and their preparation
EP0431523A2 (en) Modified nucleotide compounds
KR20020013515A (en) L-Ribo-LNA analogues
MXPA96004355A (en) Oligonucleotides and used modified intermediaries in nucleic acids therapeuti
JPH05506014A (en) oligonucleotide analogs
JPH09511250A (en) Modified oligonucleotides and intermediates useful in nucleic acid therapy
EP2462153A2 (en) Bicyclic cyclohexose nucleic acid analogs
US6017895A (en) Oligonucleotides possessing zwitterionic moieties
AU2002325599B2 (en) Oligonucleotide analogues
JP3735695B2 (en) Miniribozyme, stabilized miniribozyme, target RNA cleaving method and drug using them
JPH11130793A (en) Antisense oligonucleotide
WO1993011148A1 (en) Oligonucleotides having aminohydrocarbon phosphonate moieties

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP KR US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 2058632

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1990913804

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1990913804

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1990913804

Country of ref document: EP