CA2294988C - Compositions and methods for the delivery of oligonucleotides via the alimentary canal - Google Patents

Compositions and methods for the delivery of oligonucleotides via the alimentary canal Download PDF

Info

Publication number
CA2294988C
CA2294988C CA2294988A CA2294988A CA2294988C CA 2294988 C CA2294988 C CA 2294988C CA 2294988 A CA2294988 A CA 2294988A CA 2294988 A CA2294988 A CA 2294988A CA 2294988 C CA2294988 C CA 2294988C
Authority
CA
Canada
Prior art keywords
acid
artificial sequence
pharmaceutical composition
oligonucleotide
dna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA2294988A
Other languages
French (fr)
Other versions
CA2294988A1 (en
Inventor
Ching-Leou Teng
Greg Hardee
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ionis Pharmaceuticals Inc
Original Assignee
Isis Pharmaceuticals 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 Isis Pharmaceuticals Inc filed Critical Isis Pharmaceuticals Inc
Publication of CA2294988A1 publication Critical patent/CA2294988A1/en
Application granted granted Critical
Publication of CA2294988C publication Critical patent/CA2294988C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/28Steroids, e.g. cholesterol, bile acids or glycyrrhetinic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The present invention relates to compositions and methods which enhance the transport of nucleic acids, especially oligonucleotides at various sites in the alimentary canal of an animal. The methods and compositions enhance the transport of oligonucleotides across the mucosa of the alimentary canal via the use of one or more penetration enhancers. The invention features the use of various fatty acids, bile salts, chelating agents and other penetration enhancers, as well as carrier compounds, to enhance the stability of nucleic acids and/or their transport across and/or into cells of the alimentary canal. In one preferred embodiment, the compositions and methods of the invention are utilized to effect the oral delivery of an antisense oligonucleotide to an animal in order to modulate the expression of a gene in the animal for investigative, therapeutic or prophylactic purposes.

Description

COMPOSITIONS AND METHODS FOR THE DELIVERY OF
OLIGONUCLEOTIDES VIA THE ALIMENTARY CANAL
FIELD OF THE INVENTION
The present invention relates to compositions and methods which enhance the transport of nucleic acids at various sites in the alimentary canal. More particularly, the methods and compositions enhance the transport of oligonucleotides and other nucleic acids across the mucosa of the alimentary canal through the use of one or more penetration enhancers. More specifically, the present invention is directed to the use of various fatty acids, bile salts, chelating agents and other penetration enhancers, as well as carrier compounds, to enhance the stability of oligonucleotides and other nucleic acids and/or their transport across and/or into cells of the alimentary canal. More specific objectives and advantages of the invention will hereinafter be made clear or become apparent to those skilled in the art during the course of explanation of preferred embodiments of the invention.
BACKGROUND OF THE INVENTION
Advances in the field of biotechnology have given rise to significant advances in the treatment of previously-intractable diseases such as cancer, genetic diseases, arthritis and AIDS.
Many such advances involve the administration of oligonucleotides and other nucleic acids to J
- 2 -a subject, particularly a human subject. The administration of such molecules via parenteral routes has been shown to be effective for the treatment of diseases and/or disorders. See, e.g., U.S. Patent No. 5,595,978, January 21, 1997 to Draper et a/., which discloses intravitreal injection as a means for the direct delivery of antisense oligonucleotides to the vitreous humor of the mammalian eye.
See also, Robertson, Nature Biotechnology, 1997, /5:209 and Anon., Genetic Engineering News, 1997, 15:1, each of which discuss the treatment of Crohn's disease via intravenous infusions of antisense oligonucleotides. Oligonucleotides and other nucleic acids have been administered via non-traumatic (non-parenteral) routes such as oral or rectal delivery or other mucosal routes only with difficulty. Facile non-parenteral administration of oligonucleotides and other nucleic acids offers the promise of simpler, easier and less injurious administration of such nucleic acids without the need for sterile procedures and their concomitant expenses, e.g., hospitalization and/or physician fees. There thus remains a need to provide compositions and methods to enhance the alimentary availability of novel drugs such as oligonucleotides.
It is desirable that such new compositions and methods provide for the simple, convenient, practical and optimal alimentary delivery of oligonucleotides and other nucleic acids.
OBJECTS OF THE INVENTION
To date, there are no known pharmaceutical compositions which are capable of generally enhancing the oral delivery of oligonucleotides and nucleic acids, particularly oligonucleotides having a variety of chemical modifications.
Thus, there is a long-felt need for compositions which can effectively provide for the oral delivery of nucleic acids, particularly oligonucleotides, more particularly oligonucleotides having one or more chemical modifications, together with methods for using such compositions to deliver such oligonucleotides and nucleic acids into an animal via the alimentary canal, e.g., via oral or rectal administration. It
3 is desirable that such new compositions and methods provide for the simple, convenient, practical and optimal introduction of transport and delivery of oligonucleotides and other nucleic acids across epithelial cells at various mucosal sites.
SUMMARY OF THE INVENTION
In accordance with the present disclosure, in one aspect, compositions and methods are provided for the alimentary delivery and mucosal penetration of nucleic acids in an animal. In particular, there is provided compositions and methods for modulating the production of selected proteins or other biological phenomena in an animal, which involves the administration of an oligonucleotide, especially an antisense oligonucleotide, to the alimentary canal of an animal, thereby bypassing the complications and expense which may be associated with intravenous and other modes of in vivo administration. "Administration to the alimentary canal" refers to the contacting, directly or otherwise, to all or a portion of the alimentary canal of an animal.
Because of the advantages of alimentary delivery of drugs of the antisense class, the compositions and methods described herein can be used in therapeutic methods as explained in more detail herein. However, the compositions and methods herein provided may also be used to examine the function of various proteins and genes in an animal, including those essential to animal development.
In one embodiment, there is provided a pharmaceutical composition suitable for non-parenteral administration comprising an oligonucleotide that comprises about 8 to about 30 nucleotides for use in antisense modulation of the function of DNA or messenger RNA encoding a protein, a fatty acid and a bile salt, wherein the composition modulates the in vivo expression of a gene in an animal through the alimentary canal administration of the oligonucleotide.
In another aspect, there is provided a method of modulating gene expression in cells or tissues in vitro comprising administering a composition comprising an oligonucleotide that comprises about 8 to about 30 nucleotides for use in antisense modulation of the function of DNA
or messenger RNA encoding a protein, a fatty acid and a bile salt to the cells or tissues.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The invention provides compositions and methods for the delivery of oligonucleotides and other nucleic acids to the alimentary canal of an animal.
In particular, the present invention provides compositions and methods for modulating the in vivo expression of a gene in an animal through the alimentary canal administration of an antisense oligonucleotide, thereby bypassing the complications and expense which may be associated with intravenous and other routes of administration. Enhanced bioavailability of DocsToR. 5008344\1
- 4 -oligonucleotides and other nucleic acids administered to the alimentary canal of an animal is achieved through the use of the compositions and methods of the invention.
The term "bioavailability" refers to a measurement of what portion of an administered drug reaches the circulatory system when a non-parenteral mode of administration is used to introduce the drug into an animal. The term is used for drugs whose efficacy is related to the blood concentration achieved, even if the drug's ultimate site of action is intracellular (van Berge-Henegouwen et a/., Gastroenterol., 1977, 73:300).
Traditionally, bioavailability studies determine the degree of intestinal absorption of a drug by measuring the change in peripheral blood levels of the drug after an oral dose (DiSanto, Chapter 76 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, pages 1451-1458).
The area under the curve (AUC0) is divided by the area under the curve after an intravenous (i.v.) dose (AUCiv) and the quotient is used to calculate the fraction of drug absorbed.
This approach cannot be used, however, with compounds which have a large "first pass clearance," i.e., compounds for which hepatic uptake is so rapid that only a fraction of the absorbed material enters the peripheral blood.
For such compounds, other methods must be used to determine the absolute bioavailability (van Berge-Henegouwen et a/., Gastroenterol., 1977, 73:300).
Studies suggest that oligonucleotides are rapidly eliminated from plasma and accumulate mainly in the liver and kidney after i.v. administration (Miyao et al., Antisense Res.
Dev., 1995, 5:115; Takakura et al., Antisense & Nucl. Acid Drug Dev., 1996, 6:177).
Accordingly, means for measuring and avoiding first pass clearance effects may be needed for the development of effective orally administered pharmaceutical compositions comprising these or other nucleic acids.
Another "first pass effect" that applies to orally administered drugs is degradation due to the action of gastric acid and various digestive enzymes. Furthermore, the entry of many high molecular weight active agents (such as peptides,
- 5 -proteins and oligonucleotides) and some conventional and/or low molecular weight drugs (e.g., insulin, vasopressin, leucine enkephalin, etc.) through mucosal routes (such as oral, nasal, pulmonary, buccal, rectal, transdermal, vaginal and ocular) to the bloodstream is frequently obstructed by poor transport across epithelial cells and concurrent metabolism during transport.
One means of ameliorating first pass clearance effects is to increase the dose of administered drug, thereby compensating for proportion of drug lost to first pass clearance. Although this may be readily achieved with i.v.
administration by, for example, simply providing more of the drug to an animal, other factors influence the bioavailability of drugs administered via the alimentary canal. For example, a drug may be enzymatically or chemically degraded in the alimentary canal and/or may be impermeable or semipermeable to alimentary mucosa. It has now been found that oligonucleotides can be introduced effectively into animals via the alimentary canal through coadministration of "mucosal penetration enhancers," also known as "absorption enhancers" or simply as "penetration enhancers". These are substances which facilitate the transport of a drug across the mucous membrane(s) of the alimentary canal associated with the desired mode of administration.
Many pharmaceutically acceptable penetration enhancers are known for use with certain drugs. A
"pharmaceutically acceptable" component of a formulation is one which, together with such excipients, diluents, stabilizers, preservatives and other ingredients as are appropriate to the nature, composition and mode of administration of the formulation. Accordingly it is desired to select penetration enhancers which will provide the oligonucleotides to the alimentary canal of a patient in an effective physical form, without interfering with the activity of the oligonucleotides and in an manner such that the same can be introduced into the body of an animal without unacceptable side-effects such as toxicity, irritation or allergic response. As is known in the
- 6 -medical arts, a compound that is not pharmaceutically acceptable for a given patient having a particular disease or disorder may in fact be pharmaceutically acceptable to another patient with a different set of attendant circumstances. For example, a high degree of toxicity might not be acceptable for a patient suffering from a mild, non-life-threatening disorder but be nonetheless pharmaceutically acceptable for a terminally ill patient. As another example, due to differences in human physiology during development, a composition that is pharmaceutically acceptable for most adults might be inappropriate for a child or pregnant woman.
The present invention provides compositions comprising one or more pharmaceutically acceptable alimentary penetration enhancers, and methods of using such compositions, which result in the improved bioavailability of nucleic acids administered via alimentary modes of administration.
Heretofore, certain penetration enhancers have been used to improve the bioavailability of certain drugs. See Muranishi, Crit. Rev. Ther. Drug Carrier Systems, 1990, 7:1 and Lee et al., Crit. Rev. Ther. Drug Carrier Systems, 1991, 8:91.
However, it is generally viewed to be the case that effectiveness of such penetration enhancers is unpredictable.
Accordingly, it has been surprisingly found that the administration of oligonucleotides, relatively complex molecules which are known to be difficult to administer to animals and man, can be greatly improved in the alimentary canal through the use of a number of different classes of penetration enhancers.
The following portion of this specification provides, in more detail, information concerning (1) modes of administration, (2) penetration enhancers and carriers, (3) oligonucleotides, (4) administration of pharmaceutical compositions, (5) bioequivalents and (6) exemplary utilities of the invention.
1. Modes of Administration: The present invention provides compositions and methods for alimentary delivery of one or more nucleic acids to an animal. For purposes of the
- 7 -invention, the term "animal" is meant to encompass humans as well as other mammals, as well as reptiles, fish, amphibians, and birds.
The term "alimentary delivery" refers to the administration, directly or otherwise, to a portion of the alimentary canal of an animal. The term "alimentary canal"
refers to the tubular passage in an animal that functions in the digestion and absorption of food and the elimination of food residue, which runs from the mouth to the anus, and any and all of its portions or segments, e.g., the oral cavity, the esophagus, the stomach, the small and large intestines and the colon, as well as compound portions thereof such as, e.g., the gastro-intestinal tract. Thus, the term "alimentary delivery"
encompasses several routes of administration including, but not limited to, oral, rectal, endoscopic and sublingual/buccal administration. A
common requirement for these modes of administration is absorption over some portion or all of the alimentary tract and a need for efficient mucosal penetration of the nucleic acid(s) so administered.
In addition, iontophoresis (transfer of ionic solutes through biological membranes under the influence of an electric field) (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 163), phonophoresis or sonophoresis (use of ultrasound to enhance the absorption of various therapeutic agents across biological membranes, notably the skin and the cornea) (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, at p. 166), and optimization of vehicle characteristics relative to dose deposition and retention at the site of administration (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 168) may be useful to enhance the transport of drugs across mucosal sites.
A. Buccal/Sublingual Administration: Delivery of a drug via the oral mucosa has several desirable features, including, in many instances, a more rapid rise in plasma concentration of the drug than via oral delivery (Harvey, Chapter 35 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, page 711).
- 8 -Furthermore, because venous drainage from the mouth is to the superior vena cava, this route also bypasses rapid first-pass metabolism by the liver. Both of these features contribute to the sublingual route being the mode of choice for nitroglycerin (Benet et al., Chapter 1 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al., eds., McGraw-Hill, New York, NY, 1996, page 7).
B. Endoscopic Administration: Endoscopy can be used for drug delivery directly to an interior portion of the alimentary tract. For example, endoscopic retrograde cystopancreatography (ERCP) takes advantage of extended gastroscopy and permits selective access to the biliary tract and the pancreatic duct (Hirahata et a/., Gan To Kagaku Ryoho, 1992, 19(10 Suppl.):1591). Pharmaceutical compositions, including liposomal formulations, can be delivered directly into portions of the alimentary canal, such as, e.g., the duodenum (Somogyi et al., Pharm. Res., 1995, 12, 149) or the gastric submucosa (Akamo et a/., Japanese J. Cancer Res., 1994, 85, 652) via endoscopic means. Gastric lavage devices (Inoue et a/., Artif. Organs, 1997, 21, 28) and percutaneous endoscopic feeding devices (Pennington et al., Ailment Pharmacol. Ther., 1995, 9, 471) can also be used for direct alimentary delivery of pharmaceutical compositions.
C. Rectal Administration: Drugs administered by the oral route can often be alternatively administered by the lower enteral route, i.e., through the anal portal into the rectum or lower intestine. Rectal suppositories, retention enemas or rectal catheters can be used for this purpose and may be preferred when patient compliance might otherwise be difficult to achieve (e.g., in pediatric and geriatric applications, or when the patient is vomiting or unconscious).
Rectal administration may result in more prompt and higher blood levels than the oral route, but the converse may be true as well (Harvey, Chapter 35 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, page 711). Because about 50 1 of the drug that is absorbed from the rectum will bypass the liver, administration
- 9 -by this route significantly reduces the potential for first-pass metabolism (Benet et al., Chapter 1 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et a/., eds., McGraw-Hill, New York, NY, 1996).
D. Oral Administration: The preferred method of administration is oral delivery, which is typically the most convenient route for access to the systemic circulation-:
Absorption from the alimentary canal is governed by factors that are generally applicable, e.g., surface area for absorption, blood flow to the site of absorption, the physical state of the drug and its concentration at the site of absorption (Benet et al., Chapter 1 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et a/., eds., McGraw-Hill, New York, NY, 1996, pages 5-7). A
significant factor which may limit the oral bioavailability of a drug is the degree of "first pass effects." For example, some substances have such a rapid hepatic uptake that only a fraction of the material absorbed enters the peripheral blood (Van Berge-Henegouwen et a/., Gastroenterology, 1977, 73:300).
The compositions and methods of the invention circumvent, at least partially, such first pass effects by providing improved uptake of nucleic acids and thereby, e.g., causing the hepatic uptake system to become saturated and allowing a significant portion of the nucleic acid so administered to reach the peripheral circulation. Additionally or alternatively, the hepatic uptake system is saturated with one or more inactive "carrier" nucleic acids prior to administration of the active nucleic acid.
2. Penetration Enhancers and Carriers: The present invention employs various penetration enhancers in order to effect the gastrointestinal delivery of nucleic acids, particularly oligonucleotides. Penetration enhancers may be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et a/., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of these classes is discussed in more detail in the following sections.
- 10 -Carrier substances (or simply "carriers"), which reduce first pass effects by, e.g., saturating the hepatic uptake system, are also herein described.
A. Surfactants: In connection with the present invention, surfactants (or "surface-active agents") are chemical entities which, when dissolved in an aqueous solution, reduce the surface tension of the solution or the interfacial tension between the aqueous solution and another liquid, with the result that absorption of oligonucleotides through the alimentary mucosa is enhanced. In addition to bile salts and fatty acids, these penetration enhancers include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92); and perfluorohemical emulsions, such as FC-43 (Takahashi et al., J. Pharm. Phamacol., 1988, 40:252).
B. Fatty Acids: Various fatty acids and their derivatives which act as penetration enhancers include, for example, oleic acid, lauric acid, capric acid (a.k.a. n-decanoic acid), myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein (a.k.a. 1-monooleoyl-rac-glycerol), dilaurin, caprylic acid, arachidonic acid, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, acylcarnitines, acylcholines and mono- and di-glycerides thereof and/or physiologically acceptable salts thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.) (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1; El-Hariri et al., J. Pharm.
Pharmacol., 1992, 44:651).
C. Bile Salts: The physiological roles of bile include the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 In: Goodman &
Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al., eds., McGraw-Hill, New York, NY, 1996, pages 934-935). Various natural bile salts, and their synthetic
- 11 -derivatives, act as penetration enhancers. Thus, the term "bile salt" includes any of the naturally occurring components of bile as well as any of their synthetic derivatives. The bile salts of the invention include, for example, cholic acid (or its pharmaceutically acceptable sodium salt, sodium cholate), dehydrocholic acid (sodium dehydrocholate), deoxycholic acid (sodium deoxycholate), glucholic acid (sodium glucholate), glycholic acid (sodium glycocholate), glycodeoxycholic acid (sodium glycodeoxycholate), taurocholic acid (sodium taurocholate), taurodeoxycholic acid (sodium taurodeoxycholate), chenodeoxycholic acid (sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25-dihydro-fusidate (STDHF), sodium glycodihydrofusidate and polyoxyethylene-9-lauryl ether (POE) (Lee et a/., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92;
Swinyard, Chapter 39 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, pages 782-783; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1; Yamamoto et al., J. Pharm. Exp.
Ther., 1992, 263:25; Yamashita et al., J. Pharm. Sci., 1990, 79:579).
D. Chelating Agents: Chelating agents, as used in connection with the present invention, can be defined to be compounds that remove metallic ions from solution by forming complexes therewith, with the result that absorption of oligonucelotides through the alimentary mucosa is enhanced.
With regards to their use as penetration enhancers in the present invention, chelating agents have the added advantage of also serving as DNase inhibitors, as most characterized DNA
nucleases require a divalent metal ion for catalysis and are thus inhibited by chelating agents (Jarrett, J. Chromatogr., 1993, 618, 315). Chelating agents of the invention include but are not limited to disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and homovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of beta-diketones (enamines)(Lee et al., Critical Reviews in
- 12 -Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1; Buur et al., J. Control Rel., 1990, 14:43).
E. Non-Chelating Non-Surfactants: As used herein, non-chelating non-surfactant penetration enhancing compounds can be defined as compounds that demonstrate insignificant activity as chelating agents or as surfactants but that nonetheless enhance absorption of oligonucelotides through the alimentary mucosa (Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1). This class of penetration enhancers include, for example, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92); and non-steroidal anti-inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone (Yamashita et a/., J. Pharm. Pharmacol., 1987, 39:621).
F. Carrier Compounds: As used herein, "carrier compound" refers to a nucleic acid, or analog thereof, which is inert (i.e., does not possess biological activity per se) but is recognized as a nucleic acid by in vivo processes that reduce the bioavailability of a nucleic acid having biological activity by, for example, degrading the biologically active nucleic acid or promoting its removal from circulation. The coadministration of a nucleic acid and a carrier compound, typically with an excess of the latter substance, can result in a substantial reduction of the amount of nucleic acid recovered in the liver, kidney or other extracirculatory reservoirs, presumably due to competition between the carrier compound and the nucleic acid for a common receptor. For example, the recovery of a partially phosphorothioated oligonucleotide in hepatic tissue is reduced when it is coadministered with polyinosinic acid, dextran sulfate, polycytidic acid or 4-acetamido-4'isothiocyano-stilbene-2,2'-disulfonic acid (Miyao et al., Antisense Res. Dev., 1995, 5:115; Takakura et a/., Antisense & Nucl. Acid Drug Dev., 1996, 6:177).
In contrast to a carrier compound, a "pharmaceutical
- 13 -carrier" or "excipient" is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal.
The excipient may be liquid or solid and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., when combined with a nucleic acid and the other components of a given pharmaceutical composition. Typical pharmaceutical carriers include, but are not limited to, binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc.); lubricants (e.g., magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc.); disintegrates (e.g., starch, sodium starch glycolate, etc.); or wetting agents (e.g., sodium lauryl sulphate, etc.).
The compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels. Thus, for example, the compositions may contain additional compatible pharmaceutically-active materials such as, e.g., antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the composition of present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the invention.
3. Oligonucleotides: The present invention employs oligonucleotides for use in antisense modulation of the function of DNA or messenger RNA (mRNA) encoding a protein the modulation of which is desired, and ultimately to regulate the amount of such a protein.
Hybridization of an antisense
- 14 -oligonucleotide with its mRNA target interferes with the normal role of mRNA and causes a modulation of its function in cells.
The functions of mRNA to be interfered with include all vital functions such as translocation of the RNA to the site for protein translation, actual translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, turnover or degradation of the mRNA and possibly even independent catalytic activity which may be engaged in by the RNA.
The overall effect of such interference with mRNA
function is modulation of the expression of a protein, wherein "modulation" means either an increase (stimulation) or a decrease (inhibition) in the expression of the protein. In the context of the present invention, inhibition is the preferred form of modulation of gene expression.
In the context of this invention, the term "oligonucleotide" refers to an oligomer or polymer of ribonucleic acid or deoxyribonucleic acid. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly.
Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases.
An oligonucleotide is a polymer of repeating units generically known as a nucleotides. An unmodified (naturally occurring) nucleotide has three components: (1) a nitrogenous base linked by one of its nitrogen atoms to (2) a 5-carbon cyclic sugar and (3) a phosphate, esterified to carbon 5 of the sugar. When incorporated into an oligonucleotide chain, the phosphate of a first nucleotide is also esterified to carbon 3 of the sugar of a second, adjacent nucleotide.
The "backbone" of an unmodified oligonucleotide consists of (2) and (3), that is, sugars linked together by phosphodiester linkages between the carbon 5 (5') position of the sugar of a first nucleotide and the carbon 3 (3') position of a second, adjacent
- 15 -nucleotide. A
"nucleoside" is the combination of (1) a nucleobase and (2) a sugar in the absence of (3) a phosphate moiety (Kornberg, A., DNA Replication, W.H. Freeman & Co., San Francisco, 1980, pages 4-7).
The backbone of an oligonucleotide positions a series of bases in a specific order; the written representation of this series of bases, which is conventionally written in 5' to 3' order, is known as a nucleotide sequence.
Oligonucleotides may comprise nucleotide sequences sufficient in identity and number to effect specific hybridization with a particular nucleic acid.
Such oligonucleotides which specifically hybridize to a portion of the sense strand of a gene are commonly described as "antisense." In the context of the invention, "hybridization"
means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleotides.
For example, adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds. "Complementary," as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other. Thus, "specifically hybridizable" and "complementary" are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA target. It is understood in the art that an oligonucleotide need not be 10096 complementary to its target DNA sequence to be specifically hybridizable. An oligonucleotide is specifically hybridizable when binding of the oligonucleotide to the target DNA or RNA
- 16 -molecule interferes with the normal function of the target DNA
or RNA to cause a decrease or loss of function, and there is a sufficient degree of complementarity to avoid non-specific binding of the oligonucleotide to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, or in the case of in vitro assays, under conditions in which the assays are performed.
Antisense oligonucleotides are commonly used as research reagents, diagnostic aids, and therapeutic agents.
For example, antisense oligonucleotides, which are able to inhibit gene expression with exquisite specificity, are often used by those of ordinary skill to elucidate the function of particular genes, for example to distinguish between the functions of various members of a biological pathway. This specific inhibitory effect has, therefore, been harnessed by those skilled in the art for research uses. The specificity and sensitivity of oligonucleotides is also harnessed by those of skill in the art for therapeutic uses. For example, the following U.S. patents demonstrate palliative, therapeutic and other methods utilizing antisense oligonucleotides. U.
S.
Patent No. 5,135,917 provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression. U.S. Patent No. 5,098,890 is directed to antisense oligonucleotides complementary to the c-myb oncogene and antisense oligonucleotide therapies for certain cancerous conditions.
U.S. Patent No. 5,087,617 provides methods for treating cancer patients with antisense oligonucleotides.
U.S. Patent No.
5,166,195 provides oligonucleotide inhibitors of Human Immunodeficiency Virus (HIV). U.S.
Patent No. 5,004,810 provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication. U.S. Patent No.
5,194,428 provides antisense oligonucleotides having antiviral activity against influenzavirus. U.S. Patent No. 4,806,463 provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication.
U.S. Patent No. 5,286,717 provides oligonucleotides having a complementary base sequence
- 17 -to a portion of an oncogene. U.S. Patent No. 5,276,019 and U.S. Patent No. 5,264,423 are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells. U.S. Patent No. 4,689,320 is directed to antisense oligonucleotides as antiviral agents specific to cytomegalovirus (CMV).
U.S. Patent No. 5,098,890 provides oligonucleotides complementary to at least a portion of the mRNA transcript of the human c-myb gene. U.S. Patent No.
5,242,906 provides antisense oligonucleotides useful in the treatment of latent Epstein-Barr virus (EBV) infections. Other examples of antisense oligonucleotides are provided herein.
The oligonucleotides in accordance with this invention preferably comprise from about 8 to about 30 nucleotides. It is more preferred that such oligonucleotides comprise from about 15 to 25 nucleotides. As is known in the art, a nucleotide is a base-sugar combination suitably bound to an adjacent nucleotide through a phosphodiester, phosphorothioate or other covalent linkage. In the context of this invention, the term "oligonucleotide" includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly.
Such modified or substituted oligonucleotides may be preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases.
Oligonucleotides are also useful in determining the nature, function and potential relationship to body or disease states in animals of various genetic components of the body.
Heretofore, the function of a gene has been chiefly examined by the construction of loss-of-function mutations in the gene (i.e., "knock-out" mutations) in an animal (e.g., a transgenic mouse). Such tasks are difficult, time-consuming and cannot be accomplished for genes essential to animal development since the "knock-out" mutation would produce a lethal phenotype.
Moreover, the loss-of-function phenotype cannot be transiently
- 18 -introduced during a particular part of the animal's life cycle or disease state; the "knock-out" mutation is always present.
"Antisense knockouts," that is, the selective modulation of expression of a gene by antisense oligonucleotides, rather than by direct genetic manipulation, overcomes these limitations (see, for example, Albert et a/., Trends in Pharmacological Sciences, 1994, /5:250). In addition, some genes produce a variety of mRNA transcripts as a result of processes such as alternative splicing; a "knock-out" mutation typically removes all forms of mRNA transcripts produced from such genes and thus cannot be used to examine the biological role of a particular mRNA transcript. By providing compositions and methods for the simple alimentary delivery of oligonucleotides and other nucleic acids, the present invention overcomes these and other shortcomings.
A. Modified Linkages: Specific examples of some preferred modified oligonucleotides envisioned for this invention include those containing phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages.
Most preferred are oligonucleotides with phosphorothioates and those with CH2-NH-0-042, CH2-N(CH3) -0-CH2 (known as a methylene(methylimino) or MMI
backbone), CH2-0-N (CH3) -CH.õ CH2-N (CHO -N(CH,) -CH2 and 0-N(CH3)-CH2-CH2 backbones, wherein the native phosphodiester backbone is represented as 0-P-0-CH2.
Also preferred are oligonucleotides having morpholino backbone structures (Summerton and Weller, U.S. Patent No. 5,034,506).
Further preferred are oligonucleotides with NR-C(*)-CH2-CH2, CH2-NR-C (*) -CH2, CH2-CH2-NR-C(*), C (lc) -NR-CH2-CH2 and CH2-C(*)-NR-CH2 backbones, wherein "*" represents 0 or S (known as amide backbones; DeMesmaeker et a/., WO 92/20823, published November 26, 1992). In other preferred embodiments, such as the peptide nucleic acid (PNA) backbone, the phosphodiester backbone of the oligonucleotide is replaced with a polyamide backbone, the nucleobases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone (Nielsen et al.,
- 19 -Science, 1991, 254:1497; U.S. Patent No. 5, 539, 082) .
B. Modified Nucleobases: The oligonucleotides of the invention may additionally or alternatively include nucleobase modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include adenine (A), guanine (G), thymine (T), cytosine (C) and uracil (U).
Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e.g., hypoxanthine, 6-methyladenine, 5-methylcytosine, 5-hydroxymethylcytosine (HMC), glycosyl HMC and gentiobiosyl HMC, as well synthetic nucleobases, e.g., 2-aminoadenine, 2-thiourac i 1 , 2 - thiothymine , 5 -bromouraci 1 , 5-hydroxymethyluracil, 8-azaguanine, 7-deazaguanine, 10(6-aminohexyl)adenine and 2,6-diaminopurine (Kornberg, A., DNA
Replication, W.H. Freeman & Co., San Francisco, 1980, pages 75-77; Gebeyehu, G., et al., Nucleic Acids Res., 1987, 15, 4513).
C. Sugar Modifications: The oligonucleotides of the invention may additionally or alternatively comprise substitutions of the sugar portion of the individual nucleotides. For example, oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group. Other preferred modified oligonucleotides may contain one or more substituted sugar moieties comprising one of the following at the 2' position: OH, SH, SCH,, F, OCN, OCH3OCH3, OCH30 ( CH2) 1iCH3, 0 ( CH2) nNH2 or 0 (CH2) n0-13 where n is from 1 to about 10; C, to C10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF,; OCF,; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; SOCH,; SO2CH3; ONO,; NO2; N3; NH2;
heterocycloalkyl; heterocycloalkaryl;
aminoalkylamino;
polyalkylamino; substituted silyl; an RNA cleaving group; a reporter group; an intercalator; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A preferred modification includes 2'-methoxyethoxy (2' -0-CH,CH2OCH3, also known as 2'-0-(2-methoxyethyl)) (Martin et al., Hely. Chim. Acta, 1995, 78:486).
Other preferred
- 20 -modifications include 2' -methoxy- (2' -0-CH3) , 2 ' -propoxy- (2' -OCH2CH2CH3) and 2' -fluoro- (2' -F) .
D. Other Modifications: Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide and the 5' position of the 5' terminal nucleotide.
The 5' and 3' termini of an oligonucleotide may also be modified to serve as points of chemical conjugation of, e.g., lipophilic moieties (see immediately subsequent paragraph), intercalating agents (Kuyavin et a/., WO 96/32496, published October 17, 1996; Nguyen et al., U.S. Patent No. 4,835,263, issued May 30, 1989) or hydroxyalkyl groups (Helene et a/., WO
96/34008, published October 31, 1996).
Other positions within an oligonucleotide of the invention can be used to chemically link thereto one or more effector groups to form an oligonucleotide conjugate.
An "effector group" is a chemical moiety that is capable of carrying out a particular chemical or biological function.
Examples of such effector groups include, but are not limited to, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A variety of chemical linkers may be used to conjugate an effector group to an oligonucleotide of the invention. As an example, U.S. Patent No. 5,578,718 to Cook et al. discloses methods of attaching an alkylthio linker, which may be further derivatized to include additional groups, to ribofuranosyl positions, nucleosidic base positions, or on internucleoside linkages. Additional methods of conjugating oligonucleotides to various effector groups are known in the art; see, e.g., Protocols for Oligonucleotide Conjugates (Methods in Molecular Biology, Volume 26) Agrawal, S., ed., Humana Press, Totowa, NJ, 1994.
Another preferred additional or alternative modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more
- 21 -lipophilic moieties which enhance the cellular uptake of the oligonucleotide. Such lipophilic moieties may be linked to an oligonucleotide at several different positions on the oligonucleotide.
Some preferred positions include the 3' position of the sugar of the 3' terminal nucleotide, the 5' position of the sugar of the 5' terminal nucleotide, and the 2' position of the sugar of any nucleotide. The N6 position of a purine nucleobase may also be utilized to link a lipophilic moiety to an oligonucleotide of the invention (Gebeyehu, G., et a/., Nucleic Acids Res., 1987, /5:4513). Such lipophilic moieties include but are not limited to a cholesteryl moiety (Letsinger et al., Proc. Nat/. Acad. Sci. U.S.A., 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem.
Let., 1994, 4:1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sc., 1992, 660:306;
Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3:2765), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 20:111;
Kabanov et al., FEBS Lett., 1990, 259:327; Svinarchuk et al., Biochimie, 1993, 75:49), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:3651; Shea et al., Nucl. Acids Res., 1990, /8:3777), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, /4:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923).
Oligonucleotides comprising lipophilic moieties, and methods for preparing such oligonucleotides, are disclosed in U.S. Patents Nos. 5,138,045, 5,218,105 and 5,459,255.
The present invention also includes oligonucleotides that are substantially chirally pure with regard to particular
- 22 -positions within the oligonucleotides.
Examples of substantially chirally pure oligonucleotides include, but are not limited to, those having phosphorothioate linkages that are at least 75% Sp or Rp (Cook et al., U.S. Patent No. 5,587,361) and those having substantially chirally pure (Sp or Rp) alkylphosphonate, phosphoamidate or phosphotriester linkages (Cook, U.S. Patents Nos. 5,212,295 and 5,521,302).
E. Chimeric Oligonucleotides:
The present invention also includes oligonucleotides which are chimeric.
"Chimeric" oligonucleotides or "chimeras," in the context of this invention, are oligonucleotides which contain two or more chemically distinct regions, each made up of at least one nucleotide. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA
hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of antisense inhibition of gene expression. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art. By way of example, such "chimeras" may be "gapmers,"
i.e., oligonucleotides in which a central portion (the "gap") of the oligonucleotide serves as a substrate for, e.g., RNase H, and the 5' and 3' portions (the "wings") are modified in such a fashion so as to have greater affinity for the target RNA molecule but are unable to support nuclease activity (e.g., 2'-fluoro- or 2'-methoxyethoxy- substituted). Other chimeras include "wingmers," that is, oligonucleotides in which the 5' portion of the oligonucleotide serves as a substrate for, e.g., RNase H, whereas the 3' portion is modified in such a fashion so as to have greater affinity for the target RNA molecule but
- 23 -is unable to support nuclease activity (e.g., 2'-fluoro- or 2'-methoxyethoxy- substituted), or vice-versa.
F. The oligonucleotides used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis.
Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, CA). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates and alkylated derivatives. Teachings regarding the synthesis_ of particular modified oligonucleotides can be found in _____________ ____________________________________________________________________ the following U.S. patents or pending patent applications, each of which is commonly assigned with this application: U.S. Patents Nos. 5,138,045 and 5,218,105, drawn to polyamine conjugated oligonucleotides; U.S. Patent No.
5,212,295, drawn to monomers for the preparation of oligonucleotides having chiral phosphorus linkages; U.S.
Patents Nos. 5,378,825 and 5,541,307, drawn to oligonucleotides having modified backbbnes; U.S. Patent No. 5,386,023, drawn to backbone modified oligonucleotides and the preparation thereof through reductive coupling; U.S. Patent No. 5,457,191, drawn to modified nucleobases based on the 3-deazapurine ring system and methods of synthesis thereof; U.S. Patent No. 5,459,255, drawn to modified nucleobases based on N-2 substituted purines;
U.S. Patent No. 5,521,302, drawn to processes for preparing oligonucleotides having chiral phosphorus linkages; U.S. Patent No. 5,539,082, drawn to peptide nucleic acids; U.S. Patent No.
5,554,746, drawn to oligonucleotides having P-lactam backbones;
U.S. Patent No. 5,571,902, drawn to methods and materials for the synthesis of oligonucleotides; U.S. Patent No. 5,578,718, drawn to nucleosides having alkylthio groups, wherein such groups may be used as linkers to other moieties attached at any of a variety of positions of the nucleoside; U.S. Patents Nos.
5,587,361 and 5,599,797, drawn to oligonucleotides having phosphorothioate linkages of high chiral purity; U.S. Patent . _
- 24 -No. 5,506,351, drawn to processes for the preparation of 2'-0-alkyl guanosine and related compounds, including 2,6-diaminopurine compounds; U.S. Patent No. 5,587,469, drawn to oligonucleotides having N-2 substituted purines; U.S. Patent No. 5,587,470, drawn to oligonucleotides having 3-deazapurines;
U.S. Patent No. 5,68,046, drawn to conjugated 4'-desmethyl nucleoside analogs; U.S. Patent Nos. 5,602,240, and 5,610,289, drawn to backbone modified oligonucleotide analogs; and U.S.
Patent No. 6,262,241, filed February 3, 1995, and U.S. Patent No. 5,459,255, drawn to methods of synthesizing 2'-fluorc-oligonucleotides.
4. Administration of Pharmaceutical Compositions:
The formulation of pharmaceutical compositions and their subsequent administration is believed to be within the skill of those in the art. Specific comments regarding the present invention are presented below.
A. Therapeutic Considerations: In general, for therapeutic applications, a patient (i.e., an animal, including a human, having or predisposed to a disease or disorder) is administered one or more nucleic acids, including oligonucleotides, in accordance with the invention in a pharmaceutically acceptable carrier in doses ranging from 0.01 ug to 100 g per kg of body weight depending on the age of the patient and the severity of the disorder or disease state being treated. Further, the treatment regimen may last for a period of time which will vary depending upon the nature of the particular disease or disorder, its severity and the overall condition of the patient, and may extend from once daily to once every 20 years. In the context of the invention, the term "treatment regimen" is meant to encompass therapeutic, palliative and prophylactic modalities. Following treatment, the patient is monitored for changes in his/her condition and for alleviation of the symptoms of the disorder or disease state. The dosage of the nucleic acid may either be increased in the event the patient does not respond significantly to current dosage levels, or the dose may be decreased if an alleviation of the symptoms of the disorder or disease state
- 25 -is observed, or if the disorder or disease state has been ablated.
Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient.
Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual oligonucleotides, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models.
In general, dosage is from 0.01 g to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 20 years. An optimal dosing schedule is used to deliver a therapeutically effective amount of the nucleic acid being administered via a particular mode of administration.
The term "therapeutically effective amount," for the purposes of the invention, refers to the amount of nucleic acid-containing formulation which is effective to achieve an intended purpose without undesirable side effects (such as toxicity, irritation or allergic response).
Although individual needs may vary, determination of optimal ranges for effective amounts of formulations is within the skill of the art.
Human doses can be extrapolated from animal studies (Katocs et a/., Chapter 27 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990).
Generally, the dosage required to provide an effective amount of a formulation, which can be adjusted by one skilled in the art, will vary depending on the age, health, physical condition, weight, type and extent of the disease or disorder of the recipient, frequency of treatment, the nature of concurrent therapy (if any) and the nature and scope of the desired effect(s) (Nies et al., Chapter 3 In: Goodman &
Gilman's The Pharmacological Basis of Therapeutics, 9th Ed.,
- 26 -Hardman et al., eds., McGraw-Hill, New York, NY, 1996).
As used herein, the term "high risk individual" is meant to refer to an individual for whom it has been determined, via, e.g., individual or family history or genetic testing, has a significantly higher than normal probability of being susceptible to the onset or recurrence of a disease or disorder. As art of treatment regimen for a high risk individual, the individual can be prophylactically treated to prevent the onset or recurrence of the disease or disorder.
The term "prophylactically effective amount" is meant to refer to an amount of a formulation which produces an effect observed as the prevention of the onset or recurrence of a disease or disorder. Prophylactically effective amounts of a formulation are typically determined by the effect they have compared to the effect observed when a second formulation lacking the active agent is administered to a similarly situated individual.
Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the nucleic acid is administered in maintenance doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years. For example, in the case of in individual known or suspected of being prone to an autoimmune or inflammatory condition, prophylactic effects may be achieved by administration of preventative doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years. In like fashion, an individual may be made less susceptible to an inflammatory condition that is expected to occur as a result of some medical treatment, e.g., graft versus host disease resulting from the transplantation of cells, tissue or an organ into the individual.
B. Formulation Additives: Formulations for non-parenteral administration of nucleic acids may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. Pharmaceutically acceptable organic or inorganic carrier substances suitable for non-parenteral
- 27 -administration which do not deleteriously react with nucleic acids can be used.
Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like.
The formulations can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings flavorings and/or aromatic substances and the like which do not deleteriously react with the nucleic acid(s) of the formulation. Aqueous suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. Optionally, the suspension may also contain stabilizers.
In one embodiment of the invention, a nucleic acid is administered via the rectal mode. In particular, compositions for rectal administration include foams, solutions (enemas) and suppositories. Rectal suppositories for adults are usually tapered at one or both ends and typically weigh about 2 g each, with infant rectal suppositories typically weighing about one-half as much, when the usual base, cocoa butter, is used (Block, Chapter 87 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990).
In a preferred embodiment of the invention, one or more nucleic acids are administered via oral delivery.
Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets, troches, tablets or SECs (soft elastic capsules or "caplets"). Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids, carrier substances or binders may be desirably added to such formulations. The use of such formulations has the effect of delivering the nucleic acid to the alimentary canal for exposure to the mucosa thereof. Accordingly, the formulation can consist of material
- 28 -effective in protecting the nucleic acid from pH extremes of the stomach, or in releasing the nucleic acid over time, to optimize the delivery thereof to a particular mucosal site.
Enteric coatings for acid-resistant tablets, capsules and caplets are known in the art and typically include acetate phthalate, propylene glycol and sorbitan monoleate.
Various methods for producing formulations for alimentary delivery are well known in the art. See, generally, Nairn, Chapter 83; Block, Chapter 87; Rudnic et al., Chapter 89; Porter, Chapter 90; and Longer et al., Chapter 91 In:
Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990. The formulations of the invention can be converted in a known manner into the customary formulations, such as tablets, coated tablets, pills, granules, aerosols, syrups, emulsions, suspensions and solutions, using inert, non-toxic, pharmaceutically suitable excipients or solvents. The therapeutically active compound should in each case be present here in a concentration of about 0.5% to about 95% by weight of the total mixture, that is to say in amounts which are sufficient to achieve the stated dosage range. The formulations are prepared, for example, by extending the active compounds with solvents and/or excipients, if appropriate using emulsifying agents and/or dispersing agents, and, for example, in the case where water is used as the diluent, organic solvents can be used as auxiliary solvents if appropriate.
Compositions may be formulated in a conventional manner using additional pharmaceutically acceptable carriers or excipients as appropriate.
Thus, the composition may be prepared by conventional means with additional carriers or excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrates (e.g., starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulfate).
Tablets may be coated by methods well known in the art. The preparations may also contain flavoring, coloring and/or
- 29 -sweetening agents as appropriate.
The pharmaceutical formulations, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s).
In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing predetermined amounts of the active ingredients; as powders or granules; as solutions or suspensions in an aqueous liquid or a non-aqueous liquid; or as oil-in-water emulsions or water-in-oil liquid emulsions. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredients therein.
5. Bioequivalents A. Pharmaceutically Acceptable Salts: The compounds of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite Or residue thereof.
Accordingly, for example, the disclosure is also drawn to "pharmaceutically acceptable salts" of the penetration
- 30 -enhancers and nucleic acids of the invention and prodrugs of such nucleic acids. "Pharmaceutically acceptable salts" are physiologically and pharmaceutically acceptable salts of the penetration enhancers and nucleic acids of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like.
Examples of suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine (see, for example, Berge et al., "Pharmaceutical Salts," J. of Pharma Sci., 1977, 66:1).
The base addition salts of said acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner. The free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid for purposes of the present invention.
For oligonucleotides, examples of pharmaceutically acceptable salts include but are not limited to, salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine and the like.
B. Oligonucleotide Prodrugs:The oligonucleot ides of the invention may additionally or alternatively be prepared to be delivered in a "prodrug" form.
The term "prodrug"
indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions. In particular, prodrug
- 31 -versions of the oligonucleotides of the invention are prepared as SATE ((S-acetyl-2-thioethyl) phosphate) derivatives according to the methods disclosed in WO 93/24510 to Gosselin et a/., published December 9, 1993.
C. Oligonucleotide Deletion Derivatives: During the process of oligonucleotide synthesis, nucleoside monomers are attached to the chain one at a time in a repeated series of chemical reactions such as nucleoside monomer coupling, oxidation, capping and detritylation. The stepwise yield for each nucleoside addition is above 99%. That means that less than 1% of the sequence chain failed to the nucleoside monomer addition in each step as the total results of the incomplete coupling followed by the incomplete capping, detritylation and oxidation (Smith, Anal. Chem., 1988, 60, 381A). All the shorter oligonucleotides, ranging from (n-1), (n-2), etc., to 1-mers (nucleotides), are present as impurities in the n-mer olignucleotide product. Among the impurities, (n-2)-mer and shorter oligonucleotide impurities are present in very small amounts and can be easily removed by chromatographic purification (Warren et a/., Chapter 9 In: Methods in Molecular Biology, Vol. 26: Protocols for Oligonucleotide Conjugates, Agrawal, S., Ed., 1994, Humana Press Inc., Totowa, NJ, pages 233-264). However, due to the lack of chromatographic selectivity and product yield, some (n-1)-mer impurities are still present in the full-length (i.e., n-mer) oligonucleotide product after the purification process. The (n-1) portion consists of the mixture of all possible single base deletion sequences relative to the n-mer parent oligonucleotide. Such (n-1) impurities can be classified as terminal deletion or internal deletion sequences, depending upon the position of the missing base (i.e., either at the 5' or 3' terminus or internally). When an oligonucleotide containing single base deletion sequence impurities is used as a drug (Crooke, Hematologic Pathology, 1995, 9, 59), the terminal deletion sequence impurities will bind to the same target mRNA as the full length sequence but with a slightly lower affinity. Thus, to some extent, such impurities can be considered as part of
- 32 -the active drug component, and are thus considered to be bioequivalents for purposes of the present invention.
D. Ribozymes:
Synthetic RNA molecules and derivatives thereof that catalyze highly specific endoribonuclease activities are known as ribozymes (see, generally, U.S. Patent No. 5,543,508 to Haseloff et al., issued August 6, 1996, and U.S. Patent No. 5,545,729 to Goodchild et al., issued August 13, 1996).
The cleavage reactions are catalyzed by the RNA molecules themselves. In naturally occurring RNA molecules, the sites of self-catalyzed cleavage are located within highly conserved regions of RNA secondary structure (Buzayan et al., Proc. Natl. Acad. Sci. U.S.A., 1986, 83:8859; Forster et a/., Cell, 1987, 50:9).
Naturally occurring autocatalytic RNA molecules have been modified to generate ribozymes which can be targeted to a particular cellular or pathogenic RNA molecule with a high degree of specificity. Thus, ribozymes serve the same general purpose as antisense oligonucleotides (i.e., modulation of expression of a specific gene) and, like oligonucleotides, are nucleic acids possessing significant portions of single-strandedness.
That is, ribozymes have substantial chemical and functional identity with oligonucleotides and are thus considered to be equivalents for purposes of the present invention.
E. Other Oligonucleotide Compounds: The present invention may be used to prepare pharmaceutical and other formulations of any oligonucleotide compound and is not limited to the specific oligonucleotides described herein. Moreover, the mechanism of action of an oligonucleotide formulated according to the invention does not impact the scope to which the invention may be practiced. Oligonucleotide compounds can exert their effect by a variety of means. One such means is the antisense-mediated direction of an endogenous nuclease, such as RNase H in eukaryotes or RNase P in prokaryotes, to the target nucleic acid (Chiang et al., J. Biol. Chem., 1991, 266, 18162; Forster et al., Science, 1990, 249, 783). Another means involves covalently linking a synthetic moiety having nuclease activity to an oligonucleotide having an antisense sequence,
- 33 -rather than relying upon recruitment of an endogenous nuclease.
Synthetic moieties having nuclease activity include, but are not limited to, enzymatic RNAs, lanthanide ion complexes, and the like (Haseloff et al., Nature, 1988, 334, 585; Baker et al., J. Am. Chem. Soc., 1997, 119, 8749). Regardless of their mechanism of action, such oligonucleotides are considered tq be bioequivalents for the purposes of the present invention.
6. Exemplary Utilities of the Invention:
The invention is drawn to the alimentary administration of a nucleic acid, such as an oligonucleotide, having biological activity to an animal. By "having biological activity," it is meant that the nucleic acid functions to modulate the expression of one or more genes in an animal as reflected in either absolute function of the gene (such as ribozyme activity) or by production of proteins coded by such genes.
In the context of this invention, "to modulate" means to either effect an increase (stimulate) or a decrease (inhibit) in the expression of a gene. Such modulation can be achieved by, for example, an antisense oligonucleotide by a variety of mechanisms known in the art, including but not limited to transcriptional arrest; effects on RNA processing (capping, polyadenylation and splicing) and transportation; enhancement or reduction of cellular degradation of the target nucleic acid; and translational arrest (Crooke et a/., Exp. Opin. Ther.
Patents, 1996, 6:1).
In an animal other than a human, the compositions and methods of the invention can be used to study the function of one or more genes in the animal.
For example, antisense oligonucleotides have been systemically administered to rats in order to study the role of the N-methyl-D-aspartate receptor in neuronal death, to mice in order to investigate the biological role of protein kinase C-a, and to rats in order to examine the role of the neuropeptide Y1 receptor in anxiety (Wahlestedt et al., Nature, 1993, 363:260; Dean et al., Proc.
Natl. Acad. Sci. U.S.A., 1994, 91:11762; and Wahlestedt et al., Science, 1993, 259:528, respectively).
In instances where
- 34 -complex families of related proteins are being investigated, "antisense knockouts" (i.e., inhibition of a gene by systemic administration of antisense oligonucleotides) may represent the most accurate means for examining a specific member of the family (see, generally, Albert et al., Trends Pharmacol. Sci., 1994, /5:250).
The compositions and methods of the invention are also useful therapeutically, i.e., to provide therapeutic, palliative or prophylactic relief to an animal, including a human, having or suspected of having or of being susceptible to, a disease or disorder that is treatable in whole or in part with one or more nucleic acids. The term "disease or disorder"
(1) includes any abnormal condition of an organism or part, especially as a consequence of infection, inherent weakness, environmental stress, that impairs normal physiological functioning; (2) excludes pregnancy per se but not autoimmune and other diseases associated with pregnancy; and (3) includes cancers and tumors. The term "having or suspected of having or of being susceptible to" indicates that the subject animal has been determined to be, or is suspected of being, at increased risk, relative to the general population of such animals, of developing a particular disease or disorder as herein defined. For example, a subject animal could have a personal and/or family medical history that includes frequent occurrences of a particular disease or disorder. As another example, a subject animal could have had such a susceptibility determined by genetic screening according to techniques known in the art (see, e.g., U.S. Congress, Office of Technology Assessment, Chapter 5 In: Genetic Monitoring and Screening in the Workplace, OTA-DA-455, U.S. Government Printing Office, Washington, D.C., 1990, pages 75-99). The term "a disease or disorder that is treatable in whole or in part with one or more nucleic acids" refers to a disease or disorder, as herein defined, (1) the management, modulation or treatment thereof, and/or (2) therapeutic, palliative and/or prophylactic relief therefrom, can be provided via the administration of more nucleic acids. In a preferred embodiment, such a disease or
- 35 -disorder is treatable in whole or in part with an antisense oligonucleotide.
EXAMPLES
The following examples illustrate the invention and are not intended to limit the same. Those skilled in the art will recognize, or be able to ascertain through routine experimentation, numerous equivalents to the specific substances and procedures described herein. Such equivalents are considered to be within the scope of the present invention.
Example 1: Preparation of Oligonucleotides A. General Synthetic Techniques: Oligonucleotides were synthesized on an automated DNA synthesizer using standard phosphoramidite chemistry with oxidation using iodine. Beta-cyanoethyldiisopropyl phosphoramidites were purchased from Applied Biosystems (Foster City, CA). For phosphorothioate oligonucleotides, the standard oxidation bottle was replaced by a 0.2 M solution of 3H-1,2-benzodithiole-3-one-1,1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages.
The synthesis of 2'-0-methyl- (a.k.a. 2'-methoxy-) phosphorothioate oligonucleotides is according to the procedures set forth above substituting 2'-0-methyl p-cyanoethyldiisopropyl phosphoramidites (Chemgenes, Needham, MA) for standard phosphoramidites and increasing the wait cycle after the pulse delivery of tetrazole and base to 360 seconds.
Similarly, 2'-0-propyl- (a.k.a 2'-propoxy-) phosphorothioate oligonucleotides are prepared by slight modifications of this procedure and essentially according to Procedures disclosed in U.S. patent _________________________________ Serial No.
6,262,241, filed February 3, 1995, which is assigned to the same assignee as the instant application.
The 2'-fluoro-phosphorothioate oligonucleotides of the invention are synthesized using 5'-dimethoxytrity1-3'-phosphoramidites and prepared as disclosed in U.S. patent
- 36 -No. 6,262,241, filed February 3, 1995, and U.S. Patent No.
5,459,255, which issued October 8, 1996, both of which are assigned to the same assignee as the instant application.
The 2'-fluoro-oligonucleotides are prepared using phosphoramidite chemistry and a slight modification of the standard DNA synthesis protocol (i.e., deprotection was effected using methanolic ammonia at room temperature).
PNA antisense analogs are prepared essentially as described in U.S. Patents Nos. 5,539,082 and 5,539,083, both of which (1) issued July 23, 1996, (2) are assigned to the same assignee as the instant application.
Oligonucleotides comprising 2,6-diaminopurine are prepared using compounds described in U.S. Patent No. 5,506,351 which issued April 9, 1996, and which is assigned to the same assignee as the instant application _________________________________ _____________________________________________________________________ and materials and methods described by Gaffney et al, (Tetrahedron, 1984, 40:3), Chollet et al., (Nucl. Acids Res., 1988, 26:305) and Prosnyak et al. (Genomics, 1994, 21:490). Oligonucleotides comprising 216-diaminopurine can also be prepared by enzymatic means (Bailly et al., Proc.
Natl. Acad. Sci. U.S.A., 1996, 93:13623).
The 2'-methoxyethoxy oligonucleotides of the invention were synthesized essentially according to the methods of Martin et al. (Hell/. Chim. Acta, 1995, 78, 486). For ease of synthesis, the 3' nucleotide of the 2'-methoxyethoxy oligonucleotides was a deoxynucleotide, and 2' -0-CH,CH2OCH3_ cytosines were 5-methyl cytosines, which were synthesized according to the procedures described below.
S. Synthesis of S-Methyl Cytosine Monomers:
1. 2,2'-Anhydro(1-(fl-D-arabinofuranosyl)-5-methyluridine): 5-Methyluridine (ribosylthymine, commercially available through Yamasa, Choshi, Japan) (72.0 g, 0.279 M), diphenylcarbonate (90.0 g, 0.420 M) and sodium bicarbonate (2.0 g, 0.024 M) were added to N,N-dimethylformamide (DMF, 300 mL).
The mixture was heated to reflux, with stirring, allowing the
- 37 -evolved carbon dioxide gas to be released in a controlled manner.
After 1 hour, the slightly darkened solution was concentrated under reduced pressure. The resulting syrup was poured into diethylether (2.5 L), with stirring. The product formed a gum. The ether was decanted and the residue was dissolved in a minimum amount of methanol (ca. 400 mL). The solution was poured into fresh ether (2.5 L) to yield a stiff gum. The ether was decanted and the gum was dried in a vacuum oven (60 C at 1 mm Hg for 24 h) to give a solid which was crushed to a light tan powder (57 g, 8596 crude yield). The material was used as is for further reactions.
2. 2'-0-Methoxyethy1-5-methyluridine: 2,2'-Anhydro-5-methyluridine (195 g, 0.81 M), tris(2-methoxyethyl)borate (231 g, 0.98 M) and 2-methoxyethanol (1.2 L) were added to a 2 L stainless steel pressure vessel and placed in a pre-heated oil bath at 160 C. After heating for 48 hours at 155-160 C, the vessel was opened and the solution evaporated to dryness and triturated with methanol (200 mL).
The residue was suspended in hot acetone (1 L). The insoluble salts were filtered, washed with acetone (150 mL) and the filtrate evaporated. The residue (280 g) was dissolved in CH3CN (600 mL) and evaporated. A silica gel column (3 kg) was packed in CH-,Cl /acetone/methanol (20:5:3) containing 0.59-Et3NH.
The residue was dissolved in CH qa. 2(250 mL) and adsorbed onto silica (150 g) prior to loading onto the column.
The product was eluted with the packing solvent to give 160 g (6396) of product.
3. 2'-0-Methoxyethy1-5'-0-dimethoxytrity1-5-methyluridine: 2'-0-Methoxyethy1-5-methyluridine (160 g, 0.506 M) was co-evaporated with pyridine (250 mL) and the dried residue dissolved in pyridine (1.3 L). A first aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the mixture stirred at room temperature for one hour. A second aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the reaction stirred for an additional one hour. Methanol (170 mL) was then added to stop the reaction. High pressure liquid chromatography (HPLC) showed the presence of _ _
- 38 -approximately 70% product. The solvent was evaporated and triturated with CH3CN (200 mL). The residue was dissolved in CHC13 (1.5 L) and extracted with 2x 500 mL of saturated NaHCO3 and 2x 500 mL of saturated NaCl. The organic phase was dried over Na2SO4, filtered and evaporated. 275 g of residue was obtained. The residue was purified on a 3.5 kg silica gel column, packed and eluted with Et0Ac/Hexane/Acetone (5:5:1) containing 0.5% Et3NH. The pure fractions were evaporated to give 164 g of product. Approximately 20 g additional was obtained from the impure fractions to give a total yield of 183 g (57%).
4. 3 ' -0-Acety1-2 ' -0-methoxyethy1-5 ' -0-dimethoxytrity1-5-methyluridine:
2'-0-Methoxyethy1-5'-0-dimethoxytrity1-5-methyluridine (106 g, 0.167 M), DMF/pyridine (750 mL of a 3:1 mixture prepared from 562 mL of DMF and 188 mL of pyridine) and acetic anhydride (24.38 mL, 0.258 M) were combined and stirred at room temperature for 24 hours. The reaction was monitored by thin layer chromatography (tic) by first quenching the tic sample with the addition of Me0H. Upon completion of the reaction, as judged by tic, Me0H (50 mL) was added and the mixture evaporated at 35 C. The residue was dissolved in CHC13 (800 mL) and extracted with 2x 200 mL of saturated sodium bicarbonate and 2x 200 mL of saturated NaCl.
The water layers were back extracted with 200 mL of CHC13. The combined organics were dried with sodium sulfate and evaporated to give 122 g of residue (approximately 90% product). The residue was purified on a 3.5 kg silica gel column and eluted using Et0Ac/Hexane (4:1).
Pure product fractions were evaporated to yield 96 g (84%).
5. 3 ' -0-Acety1-2 ' -0-methoxyethy1-5 ' -0-dimethoxytrity1-5-methy1-4-triazoleuridine: A first solution was prepared by dissolving 3'-0-acety1-2'-0-methoxyethy1-5'-0-dimethoxytrity1-5-methyluridine (96 g, 0.144 M) in CH3CN (700 mL) and set aside. Triethylamine (189 mL, 1.44 M) was added to a solution of triazole (90 g, 1.3 M) in CH3CN (1 L), cooled to -5 C and stirred for 0.5 h using an overhead stirrer. POC13 was added dropwise, over a 30 minute period, to the stirred
- 39 -solution maintained at 0-10 C, and the resulting mixture stirred for an additional 2 hours. The first solution was added dropwise, over a 45 minute period, to the later solution.
The resulting reaction mixture was stored overnight in a cold room. Salts were filtered from the reaction mixture and the solution was evaporated. The residue was dissolved in Et0Ac (1 L) and the insoluble solids were removed by filtration. The filtrate was washed with lx 300 mL of NaHCO3 and 2x 300 mL of saturated NaC1, dried over sodium sulfate and evaporated. The residue was triturated with Et0Ac to give the title compound.
6. 2'-0-Methoxyethy1-5'-0-dimethoxytrity1-5-methylcytidine: A solution of 3'-0-acety1-2'-0-methoxyethy1-5'-0-dimethoxytrity1-5-methy1-4-triazoleuridine (103 g, 0.141 M) in dioxane (500 mL) and NH,OH (30 mL) was stirred at room temperature for 2 hours. The dioxane solution was evaporated and the residue azeotroped with Me0H (2x 200 mL). The residue was dissolved in Me0H (300 mL) and transferred to a 2 liter stainless steel pressure vessel. Methanol (400 mL) saturated with NH3 gas was added and the vessel heated to 100 C for 2 hours (thin layer chromatography, tic, showed complete conversion). The vessel contents were evaporated to dryness and the residue was dissolved in Et0Ac (500 mL) and washed once with saturated NaC1 (200 mL). The organics were dried over sodium sulfate and the solvent was evaporated to give 85 g (95%) of the title compound.
7. N4 -Benzoy1-2 ' -0-methoxyethyl- 5 ' -0-dimethoxytrity1-5-methylcytidine:
2'-0-Methoxyethy1-5'-0-dimethoxytrity1-5-methylcytidine (85 g, 0.134 M) was dissolved in DMF (800 mL) and benzoic anhydride (37.2 g, 0.165 M) was added with stirring. After stirring for 3 hours, tic showed the reaction to be approximately 95% complete. The solvent was evaporated and the residue azeotroped with Me0H (200 mL). The residue was dissolved in CHC13 (700 mL) and extracted with saturated NaHCO3 (2x 300 mL) and saturated NaC1 (2x 300 mL), dried over MgSO4 and evaporated to give a residue (96 g). The residue was chromatographed on a 1.5 kg silica column using Et0Ac/Hexane (1:1) containing 0.5% Et3NH as the eluting
- 40 -solvent. The pure product fractions were evaporated to give 90 g (90%) of the title compound.
8. N4 -Benzoy1-2 ' -0-methoxyethy1-5 ' -0-dimethoxytrity1-5-methylcytidine-3'-amidite: N4-Benzoy1-2'-0-methoxyethy1-5'-0-dimethoxytrity1-5-methylcytidine (74 g, 0.10 M) was dissolved in CH2C12 (1 L). Tetrazole diisopropylamine (7.1 g) and 2-cyanoethoxy-tetra(isopropyl)phosphite (40.5 mL, 0.123 M) were added with stirring, under a nitrogen atmosphere.
The resulting mixture was stirred for 20 hours at room temperature (tic showed the reaction to be 95% complete). The reaction mixture was extracted with saturated NaHCO3 (1x 300 mL) and saturated NaC1 (3x 300 mL). The aqueous washes were back-extracted with CH2012 (300 mL), and the extracts were combined, dried over MgSO4 and concentrated.
The residue obtained was chromatographed on a 1.5 kg silica column using Et0Ac\Hexane (3:1) as the eluting solvent. The pure fractions were combined to give 90.6 g (87%) of the title compound.
C. Oligonucleotide Purification: After cleavage from the controlled pore glass (CPG) column (Applied Biosystems) and deblocking in concentrated ammonium hydroxide, at 55 C for 18 hours, the oligonucleotides were purified by precipitation 2x from 0.5 M NaC1 with 2.5 volumes of ethanol followed by further purification by reverse phase high liquid pressure chromatography (HPLC).
Analytical gel electrophoresis was accomplished in 20% acrylamide, 8 M urea and 45 mM Tris-borate buffer (pH 7).
D. Oligonucleotide Labeling: Antisense oligonucleotides were labeled in order to detect the presence of and/or measure the quantity thereof in samples taken during the course of the in vivo pharmacokinetic studies described herein. Although radiolabeling by tritium exchange is one preferred means of labeling antisense oligonucleotides for such in vivo studies, a variety of other means are available for incorporating a variety of radiological, chemical or enzymatic labels into oligonucleotides and other nucleic acids.
1. Tritium Exchange: Essentially, the procedure of Graham et al. (Nucleic Acids Research, 1993, 2/:3737) was
- 41 -used to label oligonucleotides by tritium exchange.
Specifically, about 24 mg of oligonucleotide was dissolved in a mixture of 200 uL of sodium phosphate buffer (pH 7.8), 400 uL of 0.1 mM EDTA (pH 8.3) and 200 uL of deionized water. The pH of the resulting mixture was measured and adjusted to pH 7.8 using 0.095 AT NaOH. The mixture was lyophilized overnight in a 1.25 mL gasketed polypropylene vial. The oligonucleotide was dissolved in 8.25 uL of p-mercaptoethanol, which acts as a free radical scavenger (Graham et al., Nucleic Acids Research, 1993, 2/:3737), and 400 uL of tritiated 1120 (5 Ci/gram). The tube was capped, placed in a 90 C oil bath for 9 hours without stirring, and then briefly centrifuged to remove any condensate from the inside lid of the tube. (As an optional analytical step, two 10 uL aliquots (one for HPLC analysis, one for PAGE
analysis) were removed from the reaction tube; each aliquot was added to a separate 1.5 mL standard microfuge tube containing 490 uL of 50 uM sodium phosphate buffer (pH 7.8).) The oligonucleotide mixture is then frozen in liquid nitrogen and transferred to a lyophilization apparatus wherein lyophilization was carried out under high vacuum, typically for 3 hours. The material was then resuspended in mL of double-distilled 1120 and allowed to exchange for 1 hour at room temperature. After incubation, the mixture was again quick frozen and lyophilized overnight. (As an optional analytical step, about 1 mg of the oligonucleotide material is removed for HPLC analysis.) Three further lyophilizations were carried out, each with approximately 1 mL of double-distilled 1120, to ensure the removal of any residual, unincorporated tritium.
The final resuspended oligonucleotide solution is transferred to a clean polypropylene vial and assayed. The tritium labeled oligonucleotide is stored at about -70 C.
2. Other Means of Labeling Nucleic Acids: As is well known in the art, a variety of means are available to label oligonucleotides and other nucleic acids and to separate unincorporated label from the labeled nucleic acid. Double-stranded nucleic acids can be radiolabeled by nick translation
- 42 -or primer extension, and a variety of nucleic acids, including oligonucleotides, are terminally radiolabeled by the use of enzymes such as terminal deoxynucleotidyl transferase or T4 polynucleotide kinase (see, generally, Chapter 3 In: Short Protocols in Molecular Biology, 2d Ed., Ausubel et al., eds., John Wiley & Sons, New York, NY, pages 3-11 to 3-38; and Chapter 10 In: Molecular Cloning: A Laboratory Manual, 2d Ed., Sambrook et al., eds., pages 10.1 to 10.70). It is also well known in the art to label oligonucleotides and other nucleic acids with nonradioactive labels such as, for example, enzymes, fluorescent moieties and the like (see, for example, Beck, Methods in Enzymology, 1992, 2/6:143; and Ruth, Chapter 6 In:
Protocols for Oligonucleotide Conjugates (Methods in Molecular Biology, Volume 26) Agrawal, S., ed., Humana Press, Totowa, NJ, 1994, pages 167-185).
Example 2: Oligonucleotide Targets and Sequences The invention is drawn to formulations comprising nucleic acids and one or more oral-gastrointestinal mucosal penetration enhancers, and methods of using such formulations.
In one embodiment, such formulations are used to study the function of one or more genes in an animal other than a human.
In a preferred embodiment, oligonucleotides are formulated into a pharmaceutical composition intended for therapeutic delivery to an animal, including a human. The following tables list, as exemplars, some preferred oligonucleotides intended for therapeutic delivery that may be administered to the oral-gastrointestinal tract according to the compositions and methods of the invention.
Such desired oligonucleotides include, but are not limited to, those which modulate the expression of cellular adhesion proteins (Table 1).
Other oligonucleotides are designed to modulate the rate of cellular proliferation (Table 2), or to have biological or therapeutic activity against miscellaneous disorders (Table 3) and diseases resulting from eukaryotic pathogens (Table 4), retroviruses including HIV (human immunodeficiency virus; Table 5) or non-retroviral viral viruses (Table 6). Further details regarding
- 43 -the sources of the following oligonucleotides are provided in the Sequence Listing.
TABLE 1: TARGET OLIGONUCLEOTIDES DESIGNED
TO MODULATE CELLULAR ADHESION PROTEINS
Cell Surface Commercial or Common Target Oligonucleotide Target Protein Name (if any) Sequence SEQ ID NO:

ICAM-1 ,GM1595 2 ELAM-1 GM1515 to GM1517 5, 6, 7 TABLE 2: OLIGONUCLEOTIDES DESIGNED TO
MODULATE THE RATE OF CELLULAR PROLIFERATION
Molecular Target Commercial or Common Target Oligonucleotide Name (if any) Sequence SEQ ID NO:
c-myb MYB-AS 8 DNA methyl transf erase 9, 10 vascular endothelial 11, 12, 13, 14, 15, 16, growth factor (VEGF) 17, 18, 19, 20 VEGF Vm 21 bc1-2 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33 bc1-2 BCL-2 34 bcl-abl 35 PKC-a,-P,-y & - oligoantinc. 36 PKc-a ISIS 3521 37 PKC-c 38 protein kinase A, 39, 40, 41 subunit RI, 3ARK1 & 3ARK2 oligoantimmu 42 Ha-ras ISIS 2503 43 MDR 44, 45, 46, 47
- 44 -A-raf kinase ISIS 9069 49 c-raf kinase ISIS 5132 50 TABLE 3: OLIGONUCLEOTIDES DESIGNED TO HAVE THERAPEUTIC ACTIVITY
AGAINST MISCELLANEOUS DISORDERS
Disorder Commercial/ Common Oligonucleotide Sequences Name (if any) SEQ ID NO:
Alzheimer's disease 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62 Beta-thalassemia 5'ss & 3'ss 63, 64 TABLE 4: OLIGONUCLEOTIDES Ig3IGNED TO HAVE THERAPEUTIC Acamrrry AGAINST EUKARYOTIC PATHOGENS
Pathogen / Disease Commercial/ Common Oligonucleotide Sequences Name (if any) SEQ ID NO:
Plasmodium / malaria PSI, PSII PSIII & RI 65, 66, 67, 68 Schistosoma / 69 bloodfluke infections TABLE 5: OLIGONUCLEOTIDES DESIGNED TO HAVE THERAPEUTIC
ACTIVITY AGAINST RETROVIRUSES, INCLUDING HIV
Virus / Molecular Commercial/ Common Oligonucleotide Sequences Target Name (if any) SEQ ID NO:
HTLV-III / primer 70, 71, 72, 73, 74, 75 binding site / gag GEM 91 76 HIV-1 / gag GEM 92, GEM 93 77, 78, 79, 80, 81, 82, 83, 84, 85 HIV / tat, vpr, 87, 88, 89 rev, env, nef
- 45 -HIV / pol, env, 90, 91, 92, 93, 94, 95, vir 96, 97 HIV-1 / tat, rev, 98, 99, 100, 101, 102, env, nef 103 HIV / gp120 ISIS 5320 104 Hepatitis C virus ISIS 6547 105 TABLE 6: OLIGONUCLEOTIDES DESIGNED TO HAVE THERAPEUTIC
ACTIVITY AGAINST NON-RETROVIRAL VIRUSES
Virus / Molecular Commercial/ Common Oligonucleotide Target Name (if any) Sequences SEQ ID NO:
influenza virus 106, 107, 108, 109, 110, 111, 112, 113, 114 Epstein-Barr Virus 115, 116, 117 Respiratory Syncytial 118, 119, 120, 121 Virus cytomegalovirus (CMV) GEM 132 122 CMV 123, 124, 125, 126, 127, 128, 129, 130 CMV _ISIS 2922 131 Example 3: Preparation of Formulations Comprising Oligonucleotides and Fatty Acids Various fatty acids and their derivatives act as penetration enhancers. These include, for example, oleic acid, a.k.a. cis-9-octadecenoic acid (or a pharmaceutically acceptable salt thereof, e.g., sodium oleate or potassium oleate); caprylic acid, a.k.a. n-octanoic acid (caprylate);
capric acid, a.k.a. n-decanoic acid (caprate); lauric acid (laurate); acylcarnitines; acylcholines; and mono- and di-glycerides (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92). In order to evaluate the ability of various fatty acids to enhance the oral delivery and/or mucosal penetration of oligonucleotides, the following formulations were prepared.
Reagents: Sources of chemical reagents were as _
- 46 -follows.
TABLE 7: Sources of Fatty Acids Compound Name Abbreviation Supplier Capric acid, Na salt caprate Sigma*
Lauric acid, Na salt laurate Sigma 'Sigma is the Sigma Chemical Company, St. Louis, MO.
Preparations: As an initial screen to evaluate the oligonucleotide penetration enhancing capacity of various fatty acids, several formulations (Table 8) of ISIS 2302 (SEQ ID
NO:1) were prepared as follows. Unless otherwise indicated, all percentages are weight per volume (w/v).
TABLE 8: Formulations 1 - 3 Formulation No. ISIS 2302 Penetration Enhancer(s) 1 1 mg/ml 1% laurate 2 1 mg/ml 1% caprate 3 1 mg/ml 0.5% laurate + 0.5% caprate Buffer: In a volumetric flask, the following were combined: 14.33 g dibasic sodium phosphate, heptahydrate (U.S.P.); 1.73 g monobasic sodium phosphate, monohydrate (U.S.P.); and 4.4 g sodium chloride (U.S.P.). The volume was brought to 1 1 with purified, deionized water. The buffer has a pH of 7.4 and an osmolality of approximately 290 mOsm/kg.
ISIS 2302 Stock Solution: In 30 ml of purified, deionized water, 10 g of pure, anhydrous ISIS 2302 (SEQ ID
NO:1) was dissolved. The solution was adjusted to pH 7.4 with 1.0 N NaOH. The volume was adjusted to 50 ml with purified water to yield a final concentration of 200 mg/ml of oligonucleotide ISIS 2302.
Formulation 1: First, 500 mg of sodium laurate was transferred to a 50 ml volumetric flask containing about 40 ml buffer. An aliquot of 250 ul of ISIS 2302 solution was then
- 47 -added to the buffer solution. The solution was titrated to pH
7.4 with 0.1 ArHC1, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 2: First, 500 mg of sodium caprate was transferred to a 50 ml volumetric flask containing about 40 ml buffer. Then, an aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was added to the buffer solution. The solution was titrated to about pH 7.7 with 0.1 Ar HC1, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 3: First, 250 mg of sodium laurate and 250 mg of sodium caprate were transferred to a 50 ml volumetric flask containing about 40 ml buffer. An aliquot of 250 ul of ISIS 2302 solution was then added to the buffer solution. The solution was titrated to pH 7.4 with 0.1 N HC1, and the volume of the solution was adjusted to 50 ml with buffer.
Example 4:
Evaluation of Formulations Comprising Fatty Acid Penetration Enhancers By In Situ Perfusion of Rat Ileum Formulations comprising fatty acid penetration enhancers were evaluated as follows.
Methods: In order to evaluate formulations comprising various fatty acids as potential penetration enhancers, in situ perfusion of rat ileum was performed essentially according to the procedure of Komiya et a/. (Int.
J. Pharmaceut., 1980, 4:249).
Specifically, male Sprague Dawley rats weighing 250-300 g were used for the study. After overnight fasting, the rats were anesthetized with 5%
pentobarbital (50 mg/kg) by intraperitoneal injection. After a midline abdominal incision was made, the small intestine was taken out and ileum section was located. An incision was made at each end of a 20 cm ileum segment. The ileum segment was laid out in a uniform multiple-S arrangement with 3 bends. The luminal contents of the section were flushed with buffer. A
10 cm piece of silicon rubber tubing was inserted into the intestinal lumen at each incision and ligated with 3-0 silk suture. The proximal end tubing was connected to a 30 mL
syringe containing oligonucleotide solution. The solution was
- 48 -perfused through the intestinal segment by using Sage model 365 syringe pump at 0.125 mL/min.
The outflow solution was collected in a 2 mL centrifuge tube over 5 min intervals for 80 mins. At the end of perfusion study, an aliquot of 0.3 mL
blood sample was collected from the portal vein.
ISIS 2302 concentration in the solution before and after passing through a 20 cm ileum segment was analyzed by' high pressure liquid chromatography (HPLC) while the plasma samples were analyzed by capillary electrophoresis (CE). In most cases, tritium labeled ISIS 2302 was used as a tracer and the radioactivity of solution was measured by liquid scintillation counter. The amount of the drug absorbed from the ileum was calculated by dividing the concentration from the average of last six outflow samples (steady state) to that of the inflow sample.
Results: No significant amount (i.e., -0%) of ISIS
2302 (SEQ ID NO:1) was absorbed at steady state when a control solution (i.e., one lacking any penetration enhancers) was used. In contrast, approximately 5% of ISIS 2302 was absorbed at steady state with a 20 cm ileum segment when Formulations 1 or 2 were perfused. The absorption increased to 15% when Formulation 3 was used. The amounts absorbed was reflected in blood samples collected from the portal veins of the rats. The plasma concentration of ISIS 2302 was 0.29 ug/ml with Formulation 1 and increased to 2.83 ug/ml with Formulation 3.
Example 5: Preparation of Formulations Comprising Oligonucleotides and Bile Salts The physiological roles of bile include the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Goodman et al., eds., McGraw-Hill, New York, NY, 1996, pages 934-935).
Various natural bile salts, and their synthetic derivatives, act as penetration enhancers. These include, for example, cholic acid, a.k.a. cholalic acid or 3a,7a,12a-trihydroxy-53-cholan-24-oic acid (or its pharmaceutically acceptable sodium
- 49 -salt); deoxycholic acid, a.k.a. desoxycholic acid, 5P-cho1an-24-oic acid-3a,12a-diol, 7-deoxycholic acid or 3a,12a-dihydroxy-5P-cholan-24-oic acid (sodium deoxycholate);
glycocholic acid, a.k.a. N-[3a,7a,12a-trihydroxy-24-oxocholan-24-yl]glycine or 3a,7a,12a-trihydroxy-5P-cho1an-24-oic acid N-(carboxymethyllamide (sodium glycocholate); glycodeoxycholic acid, a.k.a. 5P-cholan-24-oic acid N-[carboxymethyl]amide-3a,12a-diol, 3a,12a-dihydroxy-5P-cholan-24-oic acid N-[carboxymethyl]amide, N-[3a,12a-dihydroxy-24-oxocholan-24-yl)glycine or glycodesoxycholic acid (sodium glycodeoxycholate); taurocholic acid, a.k.a. 5P-cholan-24-oic acid N-[2-sulfoethyl]amide-3a,7a,12a-triol, 3a,7a,12a-trihydroxy-513-cholan-24-oic acid N-[2-sulfoethyl]amide or 2-[ ( 3a , 7a, 12a- trihydroxy- 24 - oxo- 5P- cholan- 24 -yl ) amino]
ethanesulfonic acid (sodium taurocholate); taurodeoxycholic acid, a.k.a. 3a,12a-dihydroxy-5P-cho1an-2-oic acid N[2-sulfoethyl]amide or 2-[(3a,12a-dihydroxy-24-oxo-513-cholan-24-y1)-amino]ethanesulfonic acid (sodium taurodeoxycholate, a.k.a.
sodium taurodesoxycholate); chenodeoxycholic acid, a.k.a.
chenodiol, chenodesoxycholic acid, 5P-cholanic acid-3a,7a-diol or 3a,7a-dihydroxy-5P-cho1anic acid (sodium chenodeoxycholate);
ursodeoxycholic acid, a.k.a. 5P-cho1an-24-oic acid-3a,7P-diol, 7P-hydroxylithocholic acid or 3a,7P-dihydroxy-513-cho1an-24-oic acid; sodium taurodihydro-fusidate (STDHF); and sodium glycodihydrofusidate (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Swinyard, Chapter 39 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, pages 782-783). In order to evaluate the ability of various bile salts to enhance the oral delivery and/or mucosal penetration of oligonucleotides, the following formulations (Table 10) were prepared.
- 50 -Reagents: Sources of chemical reagents were as follows.
TABLE 9: Sources of Bile Salts Compound Name Abbreviation Supplier Cholic acid, Na salt CA Sigma' Glycholic acid, Na salt GCA Sigma Glycodeoxycholic acid, Na Salt GDCA Sigma Taurocholic acid, Na salt TCA Sigma Taurodeoxycholic acid, Na salt TDCA Sigma Chenodeoxycholic acid, Na salt CDCA Sigma Ursodeoxycholic acid UDCA Aldrich' 'Sigma, Sigma Chemical Company, St. Louis, MO.
'Aldrich, Aldrich Chemical Company, Milwaukee, WI.
TABLE 10: Formulations 4 - 14 Formulation No. ISIS 2302 Penetration Enhancer(s) 4 1 mg/ml 2% GCA
5 1 mg/ml 2% GDCA
6 1 mg/ml 2% TCA
7 1 mg/ml 2% TDCA
8 1 mg/ml 2% CDCA
9 1 mg/ml 2% CA
10 1 mg/ml 1% CDCA + 1% CA
11 1 mg/ml 1% CDCA + 1% GDCA
12 1 mg/ml 1% CDCA + 1% TDCA
13 1 mg/m1 1% TDCA + 1% GDCA
14 1 mg/ml 1% CDCA + 1% UDCA
Formulation 4: First, 1.0 g of GCA was transferred to a 50 ml volumetric flask containing about 35 ml buffer. An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was then
- 51 -added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 5: First, 1.0 g of GDCA was transferred to a 50 ml volumetric flask containing about 35 ml buffer.
Then, an aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 6: First, 1.0 g of TCA was transferred to a 50 ml volumetric flask containing about 35 ml buffer. An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was then added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 7: First, 1.0 g of TDCA was transferred to a 50 ml volumetric flask containing about 35 ml buffer. An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was then added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 8: First, 1.0 g of CDCA was transferred to a 50 ml volumetric flask containing about 35 ml buffer.
Then, an aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 9: First, 1.0 g of CA was transferred to a 50 ml volumetric flask containing about 35 ml buffer. An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was then added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 10: First, 0.5 g of CDCA and 0.5 g of CA were transferred to a 50 ml volumetric flask containing about 35 ml buffer. Then, an aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was added to the buffer solution. The
- 52 -solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 11: First, 0.5 g of CDCA and 0.5 g of GDCA were transferred to a 50 ml volumetric flask containing about 35 ml buffer. An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was then added to the buffer solution.
The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 12: First, 0.5 g of CDCA and 0.5 g of TDCA were transferred to a 50 ml volumetric flask containing about 35 ml buffer. An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was then added to the buffer solution.
The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 13: First, 0.5 g of TDCA and 0.5 g of GDCA were transferred to a 50 ml volumetric flask containing about 35 ml buffer. Then an aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was added to the buffer solution. The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Formulation 14: First, 0.5 g of CDCA was transferred to a 50 ml volumetric flask containing about 35 ml buffer and dissolved. Then, 0.5 g UDCA was added to the solution (this modification to the general formulation procedure was necessary because the sodium salt of UDCA is not currently commercially available). An aliquot of 250 ul of ISIS 2302 solution (200 mg/ml) was added to the buffer solution.
The solution osmolality was adjusted to 300 mOsm/kg with of purified, deionized water, and the volume of the solution was adjusted to 50 ml with buffer.
Example 6:
Evaluation of Formulations Comprising Bile Salt Penetration Enhancers By In Situ Perfusion of Rat Ileum
- 53 -In order to evaluate formulatons comprising various bile salts as potential penentration enhancers, in situ perfusion of rat ileum was performed essentially according to the procedure of Komiya et al. (Int. J. Pharmaceut., 1980, 4:249) as in Example 4.
Results: The results of the evaluations are summarized in Table 11. No significant amount (i.e., -0%) of ISIS 2302 (SEQ ID NO:1) was absorbed at steady state when a control solution (i.e., one lacking any penetration enhancers) was used. In contrast, about 13% to 28% of ISIS 2302 (SEQ ID
NO:1) was absorbed at steady state when 2% of a single bile salt was used as a penetration enhancer (Formulations 4 through 9). The absorption generally increased when a combined bile salt solution was perfused. The blood samples collected from the portal vein at the end of perfusion were scattered.
However, the highest blood concentration of ISIS 2302 was observed when solutions of 1% CDCA and 1% UDCA (combination of bile salts) or 2% CDCA (single bile salt) were used.
Example 7: Complex Formulations Complex formulations (i.e., comprising two or more types of penetration enhancers, e.g., both bile salts and fatty acids) of ISIS 2302 were prepared as follows (see Table 12).
- 54 -TABLE 11:
Enhancement of Oligonucleotide Uptake Due to Bile Salts tormu- t--Fmsorpclon blood ConcentraLion -Enhancer(s) lation (Mean + S.D.) (Portal Vein) No. (ug/ml, Mean + S.D.) 2% GCA 4 13.3 + 1.5 3.46 + 1.98 -2% GDCA 5 20.3 + 7.4 0.70 + 0.0 , 2% TCA 6 2.0 + 1.0 0.15 + 0.21 2% TDCA 7 14.8 + 4.2 3.65 + 0.49 2% CDCA 8 28.4 + 5.0 6.67 + 2.58 -2% CA 9 13.0 + 2.8 1.65 + 0.50 1% CDCA & 1% CA , 10 31.0 + 5.7 4.90 + 1.56 _ 1% CDCA & 1% GDCA 11 26.3 + 5.7 2.00 + 0.44 _ IA CDCA & 1% TDCA 12 29.7 + 2.5 2.77 + 2.98 1% TDCA & 1% GDCA , 13 16.5 + 0.7 1.55 + 0.49 -1% CDCA & 1% UDCA _ 14 26.0 + 3.6 12.87 + 3.84 TABLE 12: Complex Formulations 15 - 17 Formulation No. ISIS 2302 Penetration Enhancers 15 1 mg/ml 2% CDCA + 0.5% Caprate + 0.5% Laurate 16 1 mg/ml 0.5% CDCA + 1% Caprate + 1% Laurate 17 1 mg/ml 1% CDCA + 1% UDCA + 0.5%
Caprate + 0.5% Laurate Formulation 15: First, 1.0 g CDCA was transferred to a 50 ml volumetric flask containing about 30 ml of buffer and mixed well. Then, 250 mg sodium caprate and 250 mg sodium laurate were added to the flask. An aliquot of 250 ul of ISIS
2302 stock solution (200 mg/ml) was added to the solution, and the osmolality of the solution was adjusted to 300 mOsm/kg with
- 55 -purified, deionized water. Finally, the volume of the solution was adjusted to 50 ml with buffer.
Formulation 16: First, 250 mg CDCA was transferred to a 50 ml volumetric flask containing about 30 ml of buffer and mixed well. Then, 500 mg sodium caprate and 500 mg sodium laurate were added to the flask. An aliquot of 250 ul of ISIS
2302 stock solution (200 mg/ml) was added to the solution, and the osmolality of the solution was adjusted to 300 mOsm/kg with purified, deionized water. Finally, the volume of the solution was adjusted to 50 ml with buffer.
Formulation 17: First, 500 mg CDCA was transferred to a 50 ml volumetric flask containing about 30 ml of buffer and mixed well. Then, 500 mg UDCA was added to the solution and dissolved by mixing. Next, 250 mg sodium caprate and 250 mg sodium laurate were added to the solution and dissolved via further mixing.
An aliquot of 250 ul of ISIS 2302 stock solution (200 mg/ml) was added to the solution, and the osmolality of the solution was adjusted to 300 mOsm/kg with purified, deionized water. Finally, the volume of the solution was adjusted to 50 ml with buffer.
Example 8:
Evaluation of Complex Formulations By In Situ Perfusion of Rat Ileum In order to evaluate formulations comprising various bile salts as potential penetration enhancers, in situ perfusion of rat ileum was performed essentially according to the procedure of Komiya et a/. (Int. J. Pharmaceut., 1980, 4:249) as in Example 4.
Results: No significant amount (i.e., -0%) of ISIS
2302 (SEQ ID NO:1) was absorbed at steady state when a control solution (i.e., one lacking any penetration enhancers) was used. In contrast, the absorption of ISIS 2302 from a 20 cm rat ileum segment ranged from about 31%, about 20% and about 23% (Formulations 15, 16 and 17, respectively) when bile salts and fatty acids were used in combination (Table 13). The blood concentration for samples collected from the portal vein at the end of the perfusion also increased significantly, with values
- 56 -ranging from about 14 ug/ml, about 36 ug/ml and about 15 ug/ml (Formulations 15, 16 and 17, respectively).
TABLE 13:
Enhancement of Oligonucleotide Uptake Due to Complex Formulations isluod Concentiaclon Penetration lation % Absorption (Portal Vein) Enhancers No. Mean + S.D. (ug/ml, Mean + S.D.) 2% CDCA
+ 0.5% Caprate 15 30.6 + 6.4 14.32 + 5.89 + 0.5% Laurate 0.5% CDCA
+ 1% Laurate 16 19.7 + 3.2 35.83 + 11.38 + 1% Caprate 1% CDCA
+ 1% UDCA 17 23.0 + 1.4 15.4 + 2.12 + 0.5% Laurate + 0.5% Caprate Example 9: Concentration Effects Methods: In order to evaluate the effect(s) of varying the concentration of either the penetration enhancer or the active agent (ISIS 2302, SEQ ID NO:1) of the formulations of the invention, the following experiments were performed. In one set of formulations, CDCA (2%) was used as the penetration enhancer for ISIS 2302, the concentration of which was, depending on the formulation, 1, 5, or 10 mg/ml.
In another set of formulations, the concentration of ISIS 2302 was held constant at 1 mg/ml and the concentration of the penetration enhancer CDCA was, depending on the formulation, 0.5, 1.0 or 2.0% (w/v). In situ perfusion of rat ileum, as described in Example 4, was then performed using the two sets of formulations.
Results: In the presence of 2% CDCA, the percentage of ISIS 2302 absorbed from a 20 cm rat ileum segment is fairly
- 57 -constant (i.e., about 25% to 28%) in the concentration range from 1 mg/ml to 10 mg/ml. The blood concentration of ISIS
2302, measured in the portal vein, increased from 6.9 ug/ml (1 mg/ml perfusion solution) to 130 mg/ml (10 mg/ml perfusion solution). The amount of ISIS 2302 absorbed from a 20 cm rat ileum segment showed no significant changes when the CDCA
concentration was increased from 0.5% to 2%.
Example 10:
Bioavailability of Formulations After In Vivo (Intrajejunum) Instillation In order to evaluate the absolute oral bioavailability of ISIS 2302 formulations containing various penetration enhancers, in vivo intrajejunum instillation was performed with the following formulations (Table 14).
Formulation 18: First, 100 mg CDCA was transferred to a 5 ml volumetric flask containing about 3 ml of buffer.
The flask was shaken until the CDCA was completely dissolved.
Next, 200 mg sodium caprate and 200 mg sodium laurate were added to the solution, and the flask was shaken until all of the solid material was completely dissolved. Then, 0.5 ml of ISIS 2302 stock solution (200 mg/ml) was added to the solution.
Finally, the volume of the solution was adjusted to 5 ml with buffer.
Formulation 19: First, 200 mg sodium caprate and 200 mg sodium laurate were transferred to a 5 ml volumetric flask containing about 3 ml of buffer. Then, 100 mg of UDCA was added and the flask was shaken until the UDCA was completely dissolved.
Then, 0.5 ml of ISIS 2302 stock solution (200 mg/ml) was added to the solution. Finally, the volume of the solution was adjusted to 5 ml with buffer.
Formulation 20: As a control, a microemulsion of ISIS 2302 was prepared essentially according to the procedures of Panayiotis (Pharm. Res., 1984, 11:1385). An aliquot of 0.6 ml of ISIS 2302 stock solution (200 mg/ml) was transferred to a 30 ml beaker containing 1.0 ml of Tween 80 (Sigma Chemical Company St. Louis, MO). Next, a mixture of 6.3 ml of Captex 355 (Abitec Corp., Janesville, WI) and 2.1 ml of Capmul MCM

M4) 99/11579
- 58 -(Abitec Corp., Janesville, WI) was added to the beaker. The resultant solution was stirred until a clear solution was formed.
TABLE 14: Intrajejunum Formulations 18 - 20 Formulation No. ISIS 2302 Penetration Enhancer(s) 18 20 mg/ml CDCA 20 mg/ml Caprate 40 mg/ml Laurate 40 mg/ml 19 20 mg/ml UDCA 20 mg/ml Caprate 40 mg/ml Laurate 40 mg/ml 12 mg/ml Microemulsion Methods: Sprague-Dawley rats weighing 250-300 g were used. After overnight fasting, the rats were anesthetized with 15 5% pentobarbital (50 mg/kg) by intraperitoneal injection.
After a midline abdominal incision was made, the small intestine was pulled out and injection site was located (2 cm after the ligament of Treitz). The intestine was put back to the body carefully. An aliquot of 1.0 mL drug solution was 20 then injected via a 27 gauge needle.
Muscle was then surgically closed and skin was clipped after injection. Three hundred uL of blood was collected from a cannula at each sampling time point. The rats were sacrificed in the CO2 chamber 24 hours after dosing. Livers and kidneys were excised and stored at -80 C until analysis. Radioactivity of plasma and tissue samples were measured. Liver and kidney were also analyzed for oligonucleotide content by CE.
Results: The results of study are summarized in Table 15. No significant amount (i.e., -0%) of ISIS 2302 (SEQ
ID NO:1) was absorbed at steady state when a control solution (i.e., one lacking any penetration enhancers) was used. In contrast, the absolute bioavailability was in the range of 8 to 29% in the examined target organs (livers and kidneys). The
- 59 -AUC(0-3h) shows 10-13% bioavailability. However, it should be noted that the AUC(0-3h) comparison tends to underestimate the bioavailability, since the blood concentration from the intestinal instillation is much higher than that from i.v.
injection at 3 hours after dosing.
TABLE 15:
Percent Absolute Bloavailability i.v.) of ISIS 2302 After Jejunum Instillation in Rats FormulationAUC(0-3h) Liver Kidney 1C No. (Dose) 2 (ug x h/mL) Formulation 18 17.4 17.8 10.7 (80 mg/kg) Formulation 19 8.8 23.0 13.5 (80 mg/kg) Formulation 20 19.8 29.1 13.6 (48 mg/kg) 2According to the CE analysis - total oligonucleotide.
According to analysis by radioactivity.
AUC(0-3h) was calculated for all in vivo instillation studies because the results from radioactivity measurements are comparable to those from HPLC analyses for the first 3 hour plasma samples.
Example 11:
Dose Proportionality After In Vivo Jejunal and Colonic Instillation of Oligonucleotides in Rats In order to evaluate the amount of ISIS 2302 absorbed as a function of dose after jejunal and colonic instillation in rats, the following studies were performed.
Methods: Sprague-Dawley rats weighing 250-300 g were used. After overnight fasting, the rats were anesthetized with 5% pentobarbital (50 mg/kg) by intraperitoneal injection.
After a midline abdominal incision was made, the small intestine was pulled out and the injection site was located (2 cm after the ligament of Treitz for jejunum and 1 cm after the ileocecal junction for colon). The intestine was put back into the body carefully. An aliquot of 1.0 mL (jejunum) or 0.5 mL
(colon) drug solution was injected via a 27 gauge needle.
Muscle was then surgically closed and skin was clipped after injection.
Three hundred uL of blood was collected from VA) 99/01579 PCT/US98/13574
- 60 -femoral vein at 0.5, 1, 2, and 3 hours after dosing. Rats were sacrificed after a period of three hours for sample collection.
Formulations:
The concentration of enhancers remained constant (2% CDCA, 4% laurate and 4% caprate) for the study. The concentration of ISIS 2302 ranged from 10 mg /mL
to 80 mg/mL for jejunal instillation and from 33.4 mg/mL to 120 mg/mL for colonic instillation. Formulations were prepared according to the procedures of the previous Examples.
Results: Results of the study are summarized in Table 16. No significant amount (i.e., -0%) of ISIS 2302 (SEQ
ID NO:1) was absorbed at steady state when a control solution (i.e., one lacking any penetration enhancers) was used. In contrast, the AUC(0-2h) of ISIS 2302 increased proportionally in the concentration range studied for the jejunal instillation. The AUC(0-2h) of ISIS 2302 increased initially (from 16.7 mg to 28.6 mg) and reached a plateau region when 30 mg and 60 mg of ISIS 2302 was given colonically.
TABLE 16:
Results of In Vivo Jejunal and Colonic Instillation of Oligonucleotides in Rats Jejunal Colonic AUC(0-Jejunal Dose AUC(0-2h) Colonic Dose 2h) (mg/rat) (Ag x h/mL) (mg/rat) (Ag x h/mL) 10 39.98 16.7 29.45 20 65.33 28.6 101.29 40 105.89 60.0 91.7 80 193.78 Example 12: Combinations of Specific Oligonucleotide Chemistries and Formulations Resulting in Enhanced Oral Bioavailability In order to evaluate the effect of oligonucleotide chemistries on bioavailability using the formulations of the invention, the following experiments were carried out. Several oligonucleotide compounds targeted to human ICAM-1 and having the same nucleobase sequence, but varying in terms of chemical modifications, were administered to rats by intrajejunal
- 61 -instillation essentially according to the procedure described in the preceding Examples. All of the following isosequence oligonucleotides have uniform phosphorothioate backbones and all have SEQ ID NO:1 (5'-GCCCAAGCTGGCATCCGTCA-3').
More specifically, the antisense compounds used in these studies are:
1. ISIS 2302: GCCCAAGCTGGCATCCGTCA (SEQ ID NO:1) ISIS 2302 is a fully 2'-deoxyoligonucleotide containing no 2'-methoxyethoxy or 5-methylcytidine residues.
2. ISIS 14725: GCCCAAGCTGGCATCCGTCA (SEQ ID NO:1) ISIS 14725 is a "hemimer"; emboldened and double-underlined residues are 2'-methoxyethoxy (2'-M0E) modified.
All 2'-MOE cytidines are 5-methylcytidine (m5c) as indicated by the double-underlined " C " characters.
3. ISIS 15839: GCCCAAGCTGGCATCCGTCA (SEQ ID NO:1) ISIS 15839 is a fully m5c "hemimer"; emboldened residues are 2'-methoxyethoxy (2'-M0E) modified. All cytidines (including 2'-deoxycytidines) are 5-methylcytidine (m5c) as indicated by the double-underlined " C " and " C " characters.
Oligonucleotides were administered to rats at 40 mg/kg in a volume of 0.5 mL, with and without penetration enhancer(s). Plasma samples were taken at 0.5, 1.0, 2.0 and 3.0 hours; tissue samples were taken 24 hours after dosing.
Oligonucleotide concentration in the tissue samples was measured, and % bioavailability was calculated, as described in the preceding Examples.
The results (Table 17) show that a formulation comprising 2% of the bile salt CDCA and the fully {C->m5c}-substituted 2'-methoxyethoxy hemimer ISIS 15839 resulted in about 18% bioavailability in plasma, compared to 11%
bioavailability in plasma when the same formulation was used
- 62 -with ISIS 2302, an isosequence 2 ' - deoxy , non-m5c oligonucleotide. Moreover, a formulation comprising bile salt (2% CDCA) and fatty acids (4% Na Caprate and 4% Na Laurate) resulted in about 32% bioavailability of ISIS 15389 in plasma, compared to about 15% bioavailability in plasma for ISIS 2302 when the same formulation is used. Compositions comprising oligonucleotides that are partially or fully {C->m5c}-substituted and, additionally or alternatively, comprise one or more 2'-methoxyethoxy modifications, are thus preferred embodiments of the invention.
- 63 -Table 17: In Vivo Bioavailability (BAV) -Plasma AIX or Tissue Compound Formulation Plasma BAV Tissue BAV
ISIS 2302 Water or saline 1-2% 1-2%
ISIS 2302 Bile salt (2% CDCA)' 11% ND3 ISIS 2302 Bile salt (2% CDCA) and 14.6% 18-30%
fatty acids (4% Na caprate + 4% Na laurate)2 ISIS 14725 rWater or saline ,5-8% 5.2%
ISIS 15839 Water or saline ND ND
ISIS 15839 Bile salt (2% CDCA)1 17.5% ND
ISIS 15839 Bile salt (2% CDCA) and 31.6% ND
fatty acids (4% Na caprate + 4% Na laurate)2 1 Corresponds to Formulation 8.
2 Corresponds to Formulation 18.
3 ND, not determined.
Example 13: In Vivo Bioavailability of Oligonucleotides in Dogs Dogs were "ported" with intestinal access catheters through which formulated drug formulations (solutions or suspensions) may be introduced into various areas of the gut.
Target areas include the proximal jejunum and distal ilium or the ileocecal junction. In addition to ported dogs, naive dogs are used for the assessment of formulations given by conventional routes, e.g., oral administration for oral dosage forms, rectal administration for enema or suppository formulations, etc.
ISIS 2302 and ISIS 15839 were administered intrajejunally to "ported" dogs at oligonucleotide doses of 10 mg/kg with or without penetration enhancers. Specifically, an aliquot of 20 mg/mL drug solution was injected into a
- 64 -subcutaneous port catheter connected to the proximal jejunum.
Bile salts (CDCA) were used alone or in combination with fatty acids (2% CDCA, 4% Na caprate, 4% Na laurate). Blood samples were collected from the femoral vein for up to 6 hours and evaluated for the presence and concentration of oligonucleotides by HPLC. Percent bioavailability (%BAV) was calculated as:
intact plasma conc. (AUC) by alimentary administration x 100%, intact plasma concentration by intravenous administration wherein "AUC" refers to the Area Under the Curve and "conc."
indicates concentration.
Table 18: Absolute Bioavailability of Oligonucleotides in Dogs After Intrajejunal Administration ,Compound n Formulation BAV
ISIS 2302 2 Water or saline 0.3 %
(no enhancer control) ISIS 2302 2 Bile salt (2% CDCA) only 1.3 %
ISIS 2302 2 Fatty acids (4% Na Caprate + 5.4 %
4% Na Laurate) only ISIS 2302 3 Bile salt (2% CDCA) and Fatty 8.4 %
acids (4% Na Caprate + 4% Na Laurate) ISIS 15839 2 Water or saline 1.5 %
(no enhancer control) ISIS 15839 3 Bile salt (2% CDCA) only 4.4 %
ISIS 15839 3 Fatty acids (4% Na Caprate + 2.5 %
4% Na Laurate) only ISIS 15839 2 Bile salt (2% CDCA) and fatty 18.0 %
acids (4% Na Caprate + 4% Na Laurate)
- 65 -The results (Table 18) confirm and extend the results from the rat experimental systems.
Specifically, for the phosphorothioate, non-{C->m50-substituted 2'-deoxyoligonucleotide ISIS 2302, the % bioavailability was maximal (8.4%) when formulated with a bile salt (2% CDCA) and fatty acids (4% sodium caprate and 4% sodium laurate). When the same formulation was prepared comprising the phosphorothioate, fully {C->m5c}-substituted, 2'-deoxy-/2'-methoxyethoxy-oligonucleotide ISIS 15839, a greater %
bioavailability (18%) resulted.
Example 14: Dose Proportionality Studies of Oligonucleotides in Dogs Dogs were "ported" with intestinal access catheters as in the previous Example and a series of varying doses of ISIS 15839 were administered. Although the concentration of oligonucleotide varied, the kinds and concentration of penetration enhancers used in these experiments were held constant (2% CDCA.Na, 4% sodium laurate and 4% sodium caprate).
Bioavailability (AUC, 0-6 h) was determined as in the preceding Examples.
The results (Table 19) show that bioavailability decreases with increasing absolute dose and drug concentration.
There is a clear trend of decreasing bioavailability as the oligonucleotide dose was increased, i.e., as the proportion of penetration enhancers was decreased. These results indicate that higher ratios of [penetration enhancer(s)] to [oligonucleotide] are preferred.
Table 19: Dose Proportionality of ISIS 15839 in Dogs Dose (mg/kg) Drug Concentration (mg/mL) BAV
10 20 -18.0 %
20 40 -7.0 %
80 -1.5%
- 66 -Example 15:
01 igonucl eot ide : Penetration Enhancer Co-Delivery Studies It is possible that oligonucleotides and penetration enhancers (PEs) are best delivered contemporaneously to one or more sites for maximal bioavailability if, for example, one or more PEs and an oligonucleotide cross a rate-limiting barrier as a complex. Alternatively, as another example, delivery of PEs to the intestinal lumen prior to the delivery of oligonucleotides might allow the PEs more time to act on the cells of the GI tract than is available when the oligonucleotide and PEs arrive at such cells at the same time;
in this case, maximal bioavailability of the oligonucleotide would occur sometime after the administration of the PEs. In order to examine some of the kinetic aspects of oligonucleotide:penetration enhancer effects, the following experiments were carried out.
In a first set of experiments, ISIS 2302 was administered intrajejunally to rats, as in Example 10, at various times after the administration of a formulation of penetration enhancers (2% CDCA.Na, 4% sodium laurate and 4%
sodium caprate), and the absolute bioavailability (AUC, 0-3 h) was determined as in the preceding Examples.
The results (Table 20) show a general trend towards decreased bioavailability as oligonucleotide is delivered at increasingly longer intervals after delivery of the penetration enhancers.
These results indicate that formulations that provide for the concomitant release of oligonucleotide and penetration enhancers at appropriate sites in vivo are preferred.
In a second set of experiments, Dogs were "ported"
with intestinal access catheters as in the preceding Examples.
ISIS 2302 was administered at various times after the administration of penetration enhancers (2% CDCA.Na, 4% sodium laurate and 4% sodium caprate), and the absolute bioavailability (AUC, 0-6 h) was determined as in the preceding Examples. The results (Table 21) demonstrate the same general trend as was seen in rats; i.e., bioavailability is maximal when the oligonucleotide and penetration enhancer are
- 67 -contemporaneously delivered and decreases when oligonucleotide is delivered after the PE delivery.
Table 20: Administration Time Studies in Rats (40 mg/kg ISIS 2302) Time After PE Administration Absolute % BAV
Oligonucleotide Deliver (n = 2 or 5) Co-Administration 14.6 (n = 5) min. 13.2, 15.2 30 min. 9.7, 9.8 10 60 min. 1.4, 8.0 Table 21: Administration Time Studies in Dogs (10 mg/kg ISIS 2302) Time After PE Administration Absolute % BAV
Oligonucleotide Deliver (n = 2) 15 15 min. 10, (*) 30 min. 10, 22 60 min. -0, 25 * Data from one animal lost due to leakage at the injection site.
Example 16:
Formulations Comprising Acid, or Acid and Salt, Forms of Penetration Enhancers In the formulations of the preceding Examples, with the exception of UDCA and other indicated exceptions, bile salts have been added to formulations as sodium (Na) salts.
As indicated in Example 5, these bile salts are also available in their acid forms.
In order to determine if effective
- 68 -oligonucleotide delivery is promoted by the acid forms of bile salts, or by combinations of the acid and salt forms of bile salts, the following studies were carried out.
In order to evaluate the ability of acid forms of bile salts to act as penetration enhancers of oligonucleotides, comparative formulations (Table 22) were prepared according to the methods described in the preceding Examples with the following modifications.
In order to minimize the proportion of water in pharmaceutical formulations, the solvents propylene glycol (PPG
400, Spectrum Quality Products, Inc., Gardena, CA) and polyethylene glycol (PEG 400, Spectrum) were tested for their ability to dissolve ISIS 2302. Although the solubility of oligonucleotide in PPG was considerable (i.e., > 160 mg/mL), oligonucleotide exhibited only limited solubility in PEG (0.08 mg/mL). Studies demonstrated that the bioavailability of oligonucleotide, alone or in combination with the penetration enhancers of the invention, was not effected by PPG. Thus, PPG
was used a solvent in the following formulations. The PPG
solutions of oligonucleotides were more viscous than water-based solutions and may provide for lower diffusion rates of oligonucleotide and penetration enhancers in vivo; if so, PPG-based solutions of oligonucleotides are expected to provide for the extended release of oligonucleotides in patients.
Inclusion of PPG in the formulations allows the water content of the formulations to be decreased to less than about 10%, preferably less than about 8% and more preferably less than about 5%. In the following PPG-based formulations, the water content 7.5% unless otherwise indicated.
- 69 -Table 22: Acid/Salt Comparative Formulations of ISIS 2302 Amount of Component in:
Component Acid Formula Salt Formula Mixed Formula Lauric Acid 200 mg 100 mg -- 5 Sodium Laurate 200 mg 100 mg Capric Acid 200 mg 100 mg Sodium Caprate 200 mg 100 mg UDCA 100 mg CDCA SO mg CDCA.Na 100 mg 50 mg ISIS 2302 100 mg 100 mg 100 mg PPG QS to 5 mL QS to 5 mL QS to 5 mL
These formulations were evaluated in rat (n = 3 or 4) by intrajejeunal instillation (0.5 mL, 40 mg/kg). Samples were taken up to 3 hours after administration and the absolute bioavailability (AUC, 3-4h) was determined.
The results (Table 23) demonstrate the unexpected result that oligonucleotide bioavailability is best enhanced by a formulation having mixtures of the sodium salts and acid forms of bile salts and fatty acids. That is, oligonucleotide bioavailability was about 17% when the Mixed Formulation was used, whereas it was only about 7% and 12% when the Acid or Salt Formulations, respectively, were used.
- 70 -Table 23: Bioavailability of Comparative Formulations Formulation Bioavailability Range ' Acid 7.3 + 0.6% 6.9% to 8.0%
Salt 12.5 + 8.1% 6.0% to 23.2%
Mixed 17.0 + 1.4% 15.9% to 18.5%
Example 17: Preparation of the Sodium Salt of UDCA
When used for gallstone dissolution, CDCA may cause diarrhea, elevated plasma transaminase activity and elevated serum cholesterol. UDCA is as effective for this use at higher doses, but causes diarrhea less frequently and does not later serum cholesterol or plasma transaminase activity (Brunton, Chapter 38 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al., eds., McGraw-Hill, New York, NY, 1996, pages 934-935). Thus, UDCA is used in place of CDCA in some preferred embodiments of the invention.
Ursodeoxycholic acid (UDCA) is commercially available in its acid form (Aldrich Chemical Company, Milwaukee, WI) but not as a sodium salt. In order to carry out experiments to evaluate the potential of the sodium salt of UDCA to act as a penetration enhancer, the following novel method of efficiently preparing the sodium salt of UDCA from its acid form was developed.
Step 1:
Dissolve 500 mg UDCA in 2.4 mL ethanol (>99%). (The volume of ethanol volume can be increased slightly with no adverse effect.) Step 2:
Dissolve 1 g of NaOH in 0.9 mL H20. (Handle reaction vessels with care, as the process generates heat.) Step 3:
Slowly transfer 46 AL NaOH solution from step 2 to
- 71 -the solution of step 1 with vigorously constant mixing (The mixing is 1:1 molar ratio).
Notes for Step 3:
(A) The volume of NaOH solution added should not be more than 50 AL; otherwise the UDCA sodium salt will be redissolved.
(B) The concentrated NaOH solution tends to settle at the bottom of the reaction vessel; as a result, constant and vigorous stirring is required during this step.
Step 4:
Filter the solution and wash the precipitate with ethanol to eliminate any remaining UDCA (acid).
The precipitate can then be air dry or dried by lyophilization.
In order to determine the ability of the sodium salt of UDCA to act as a penetration enhancer for oligonucleotides, formulations are prepared and tested as in the preceding Examples, except that UDCA.Na is used in place of CDCA.Na.
The disclosure demonstrates that a variety of formulations comprising an oligonucleotide and one or more penetration enhancers result in bioavailabilities that are typically more than about 15%, in a range from about 1.5% to about 35%, most preferably from about 17% to about 35%. Those skilled in the art will be able to prepare numerous equivalent formulations without undue experimentation upon comprehension of the present disclosure.

SEQUENCE LISTING
<110> Teng, Ching-Leou Hardee, Greg <120> Compositions And Methods For The Delivery Of Oligonucleotides Via The Alimentary Canal <130> ISIS3110 <140> PCT/US98/13574 <141> 1998-07-01 <160> 132 <170> PatentIn Ver. 2.1 <210> 1 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 1 gcccaagctg gcatccgtca 20 <210> 2 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 2 agccatagcg aggctgaggt t 21 <210> 3 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 3 aacatctccg taccatgcca 20 <210> 4 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 4 cccaggcatt ttaagttgct g 21 <210> 5 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 5 gtttaaggca gcatcctaag a 21 <210> 6 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 6 tcacccaaag gtttaggctt g 21 <210> 7 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 7 gcaatcatga cttcaagagt t 21 <210> 8 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 8 gtgccggggt cttcgggc 18 <210> 9 ____________________________ ¨

<211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 9 catctgccat tcccactcta 20 <210> 10 <211> 24 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 10 ttggcatctg ccattcccac tcta 24 <210> 11 <211> 19 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 11 catggtttcg gagggcgtc 19 <210> 12 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 12 tcgcgctccc tctctccggc 20 <210> 13 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 13 cacccaagag agcagaaagt 20 <210> 14 <211> 22 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 14 cccaagacag cagaaagttc at 22 <210> 15 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 15 tcgtgggtgc agcctgggac 20 <210> 16 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 16 ctgcccggct caccgcctcg g 21 <210> 17 <211> 19 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 17 catggtttcg gaggcccga 19 <210> 18 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 18 cacccaagac agcagaaagt 20 <210> 19 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 19 ccatgggtgc agcctgggac 20 <210> 20 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 20 ccatgggtgc agcctgggac 20 <210> 21 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 21 cagcctggct caccgccttg g 21 <210> 22 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 22 cccttcctac cgcgtgcgac 20 <210> 23 <211> 20 <212> DNA
<213> Artificial Sequence _______________________ . ¨

<220>
<223> Description of Artificial Sequence: Novel Sequence <400> 23 cctccgaccc atccacgtag 20 <210> 24 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 24 gttgacgtcc tacggaaaca 20 <210> 25 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 25 cgcgtgcgac cctcttg 17 <210> 26 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 26 tcctaccgcg tgcgacc 17 <210> 27 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 27 tcctaccgcg tgcgacc 17 <210> 28 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 28 ccttcctacc gcgtgcg 17 <210> 29 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 29 gacccttcct accgcgt 17 <210> 30 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 30 ggagaccctt cctaccg 17 <210> 31 <211> 15 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 31 gcggcggcag cgcgg 15 <210> 32 <211> 15 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 32 cggcggggcg acgga 15 <210> 33 <211> 16 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 33 cgggagcgcg gcgggc 16 <210> 34 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 34 tctcccagcg tgcgccat 18 <210> 35 <211> 19 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 35 ggcgttttga actctgctt 19 <210> 36 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 36 aaggtgggct gcttgaagaa 20 <210> 37 <211> 15 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 37 ggtcctgctg ggcat 15 <210> 38 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 38 gttctcgctg gtgagtttca 20 <210> 39 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 39 gcgtgcctcc tcactggc 18 <210> 40 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Combined DNA/RNA Molecule: U at positions 4 and 15 are RNA
<220>
<223> Description of Artificial Sequence: Novel Sequence <400> 40 gcgugcctcc tcacuggc 18 <210> 41 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Combined DNA/RNA Molecule: U at positions 8 and 11 are RNA
<220>
<223> Description of Artificial Sequence: Novel Sequence _ _____________________________________________________ <400> 41 gcgtgccucc ucactggc 18 <210> 42 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 42 accgcctcca ggtccgccat 20 <210> 43 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 43 tccgtcatcg ctcctcaggg 20 <210> 44 <211> 22 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 44 tgtgctcttc ccacagccac tg 22 <210> 45 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 45 tgtgctcttc ccacagccac 20 <210> 46 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 46 gtgctcttcc cacagccact 20 <210> 47 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 47 tgctcttccc acagccactg 20 <210> 48 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 48 tgctgttcgt gcccccgccg 20 <210> 49 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 49 ctaaggcaca aggcgggctg 20 <210> 50 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 50 tcccgcctgt gacatgcatt 20 <210> 51 <211>25 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 51 cctctctgtt taaaacttta tccat 25 <210> 52 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 52 ttcatatcct gagtcatgtc g 21 <210> 53 <211> 24 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 53 gtcccagcgc tacgacgggc caaa 24 <210> 54 <211> 13 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 54 gtcccagcgc tac 13 <210> 55 <211> 14 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 55 tacgacgggc caaa 14 <210> 56 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 56 gtcccagcgc tacgacgggc c 21 <210> 57 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 57 gtcccagcgc tacgacgg 18 <210> 58 <211> 15 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 58 gtcccagcgc tacga 15 <210> 59 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 59 ccagcgctac gacgggccaa a 21 <210> 60 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 60 gcgctacgac gggccaaa 18 <210> 61 <211> 15 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 61 ctacgacggg ccaaa 15 <210> 62 <211>24 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 62 aaaccgggca gcatcgcgac cctg 24 <210> 63 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Combined DNA/RNA Molecule: U at positions 3,5,6,10 and 11 are RNA
<220>
<223> Description of Artificial Sequence: Novel Sequence <400> 63 gcuauuaccu uaacccag 18 <210> 64 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Combined DNA/RNA Molecule: U at positions 3,4,6,7 and 12 are RNA

<220>
<223> Description of Artificial Sequence: Novel Sequence <400> 64 cauuauugcc cugaaag 17 <210> 65 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 65 taaaaagaat atgatcttca t 21 <210> 66 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 66 agcaactgag ccacctga 18 <210> 67 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 67 gtcgcagact tgttccatca t 21 <210> 68 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 68 cttggcagct gcgcgtgaca t 21 <210> 69 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 69 gccatagggg gcagggaagg c 21 <210> 70 <211> 12 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 70 ctgctagaga tt 12 <210> 71 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 71 ctgctagaga ttttccacac 20 <210> 72 <211>25 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 72 ttcaagtccc tgttcgggcg ccaaa 25 <210> 73 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 73 gcgtactcac cagtcgccgc 20 <210> 74 <211> 14 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 74 ctgctagaga ttaa 14 <210> 75 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 75 acacccaatt ctgaaaatgg 20 <210> 76 <211> 25 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 76 ctctcgcacc catctctctc cttct 25 <210> 77 <211>26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 77 ctctcgcacc catctctctc cttcta 26 <210> 78 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 78 gctctcgcac ccatctctct ccttct 26 <210> 79 <211> 27 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 79 gctctcgcac ccatctctct ccttcta 27 <210> 80 <211> 28 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 80 gctctcgcac ccatctctct ccttctag 28 <210> 81 <211>28 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 81 cgctctcgca cccatctctc tccttcta 28 <210> 82 <211>29 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 82 cgctctcgca cccatctctc tccttctag 29 ' CA 02294988 1999-12-30 <210> 83 <211> 30 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 83 cgctctcgca cccatctctc tccttctagc 30 <210> 84 <211>30 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 84 acgctctcgc acccatctct ctccttctag 30 <210> 85 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 85 ctcgcaccca tctctctcct 20 <210> 86 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 86 gtggtgggtg ggtgggt 17 <210> 87 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 87 gcctattctg ctatgtcgac acccaa 26 <210> 88 <211>26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 88 cttcgggcct gtcgggtccc ctcggg 26 <210> 89 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 89 cttcgggcct gtcgggtccc ctcggg 26 <210> 90 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 90 gctggtgatc ctttccatcc ctgtgg 26 <210> 91 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 91 ctactactcc ttgactttgg ggattg 26 <210> 92 <211> 29 <212> DNA
____________________________________ -<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 92 cctctgttag taacatatcc tgcttttcc 29 <210> 93 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 93 ggttgcttcc ttcctctctg gtaccc 26 <210> 94 <211> 41 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 94 ctagcagtgg cgcccgaaca ggttcgcctg ttcgggcgcc a 41 <210> 95 <211> 30 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 95 catcacctgc catctgtttt ccataatccc 30 <210> 96 <211> 31 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 96 cctgtctact tgccacacaa tcatcacctg c 31 <210> 97 <211> 30 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 97 actattgcta ttattattgc tactactaat 30 <210> 98 <211> 26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 98 cttcgggcct gtcgggtccc ctcggg 26 <210> 99 <211> 25 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Combined DNA/RNA Molecule: U at positions 2,3,10,12,17 and 21 are RNA
<220>
<223> Description of Artificial Sequence: Novel Sequence <400> 99 cuucgggccu gucggguccc ucggg 25 <210> 100 <211> 14 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 100 gcctgtcggg tccc 14 <210> 101 <211> 14 <212> DNA

<213> Artificial Sequence <220>
<223> Description of Combined DNA/RNA Molecule: U at positions 4,6 and 11 are RNA
<220>
<223> Description of Artificial Sequence: Novel Sequence <400> 101 gccugucggg uccc 14 <210> 102 <211>26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 102 cttcgggcct gtcgggtccc ctcggg 26 <210> 103 <211>26 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 103 gctggtgatc ctttccatcc ctgtgg 26 <210> 104 <211> 8 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 104 ttggggtt 8 <210> 105 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 105 gtgctcatgg tgcacggtct 20 <210> 106 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 106 cattcaaatg gtttgcctgc 20 <210> 107 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 107 gcaggcaaac catttgaatg 20 <210> 108 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 108 ccataatccc ctgcttctgc 20 <210> 109 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 109 gcagaagcag gggattatgg 20 <210> 110 <211> 20 <212> DNA

<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 110 gcagaagcag aggattatgg 20 <210> 111 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 111 gcataagcag aggatcatgg 20 <210> 112 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 112 ggcaagcttt attgaggctt 20 <210> 113 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 113 atcttcatca tctgagagat 20 <210> 114 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 114 cgtaagcaac agtagtccta 20 <210> 115 <211> 25 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 115 tttgggtcca tcatcttcag caaag 25 <210> 116 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 116 catcatcttc agcaaagata 20 <210> 117 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 117 tcagaagtcg agtttgggtc 20 <210> 118 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 118 acgcgaaaaa atgcgtacaa 20 <210> 119 <211>20 <212> DNA
<213> Artificial Sequence <220>

<223> Description of Artificial Sequence: Novel Sequence <400> 119 taaaccaaaa aaatggggca 20 <210> 120 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 120 aaatggggca aataagaatt 20 <210> 121 <211>20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 121 aaaaatgggg caaataaatc 20 <210> 122 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 122 tggggcttac cttgcgaaca 20 <210> 123 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 123 gacgtggggc ttaccttgcg 20 <210> 124 <211> 20 <212> DNA

<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 124 tcttcaacga cgtggggctt 20 <210> 125 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 125 gacgcgtggc atgcttggtg t 21 <210> 126 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 126 aggttggggt cgacgcgtgg c 21 <210> 127 <211>21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 127 ggctgagcgg tcatcctcgg a 21 <210> 128 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 128 cgggactcac cgtcgttctg 20 <210> 129 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 129 ggaggagagc ctacagacgg 20 <210> 130 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 130 agtaacgcac cgtcggtgcc 20 <210> 131 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 131 gcgtttgctc ttcttcttgc g 21 <210> 132 <211> 19 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence: Novel Sequence <400> 132 cacccaagac agcagaaag 19

Claims (28)

WHAT IS CLAIMED IS:
1. A pharmaceutical composition suitable for non-parenteral administration comprising an oligonucleotide that comprises about 8 to about 30 nucleotides for use in antisense modulation of the function of DNA or messenger RNA encoding a protein, a fatty acid and a bile salt, wherein the composition modulates the in vivo expression of a gene in an animal through the alimentary canal administration of the oligonucleotide.
2. The pharmaceutical composition of claim 1, wherein said oligonucleotide is an oligonucleotide in prodrug form or a bioequivalent thereof.
3. The pharmaceutical composition of claim 2 wherein said oligonucleotide has at least one chemical modification selected from the group consisting of a modified nucleobase, a modified sugar residue, and a modified backbone linkage.
4. The pharmaceutical composition of claim 2 wherein said oligonucleotide has at least one chemical modification selected from the group consisting of a cytosine to 5-methyl-cytosine substitution, a phosphorothioate linkage and a 2'-methoxyethoxy modification.
5. The pharmaceutical composition of any one of claims 1 to 4, wherein said fatty acid is arachidonic acid, oleic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a monoglyceride, a diglyceride or a phailiaceutically acceptable salt thereof.
6. The pharmaceutical composition of any one of claims 1 to 5, wherein said bile salt is present in its acid form, as a sodium salt, or in a mixture of said acid form and said sodium salt.
7. The pharmaceutical composition of any one of claims 1 to 6, wherein said bile salt is cholic acid, dehydrocholic acid, deoxycholic acid, glucholic acid, glycholic acid, glycodeoxycholic acid, taurocholic acid, taurodeoxycholic acid, chenodeoxycholic acid, ursodeoxycholic acid, sodium tauro-24,25-dihydro-fusidate, sodium glycodihydrofusidate, polyoxyethylene-9-lauryl ether or a phaimaceutically acceptable salt thereof.
8. The pharmaceutical composition of any one of claims 1 to 7, further comprising at least one carrier compound.
9. The pharmaceutical composition of claim 8, wherein said carrier compound is selected from the group consisting of polyinosinic acid, dextran sulfate, polycytidic acid and 4-acetoamido-4'isothiocyano-stilbene-2,2'-disulfonic acid.
10. The pharmaceutical composition of claim 1 wherein said pharmaceutical composition is water based.
11. The pharmaceutical composition of claim 1 wherein said pharmaceutical composition is propylene glycol based.
12. The pharmaceutical composition of claim 1 wherein said pharmaceutical composition comprises less than about 8%
water.
13. The pharmaceutical composition of claim 1 wherein said pharmaceutical composition, upon administration to a mammal, results in more than about 15% bioavailability of said oligonucleotide in said mammal.
14. The pharmaceutical composition of claim 1 wherein said pharmaceutical composition, upon administration to a mammal, results in from about 17% to about 35%
bioavailability of said oligonucleotide in said mammal.
15. The pharmaceutical composition of claim 1 further comprising an additional penetration enhancer.
16. The pharmaceutical composition of claim 15, wherein said additional penetration enhancer is a surfactant, a chelating agent or a non-chelating non-surfactant.
17. Use of a pharmaceutical composition according to any one of claims 1 to 16 in the manufacture of a medicament for modulating expression of a cellular adhesion protein in an animal to treat a disease.
18. Use of a pharmaceutical composition according to any one of claims 1 to 16 in the manufacture of a medicament for modulating the rate of cellular proliferation in an animal to treat a disease.
19. Use of a pharmaceutical composition according to any one of claims 1 to 16 in the manufacture of a medicament for administration to an animal to treat diseases resulting from eukaryotic pathogens, retroviruses or non-retroviral pathogens.
20. Use of a pharmaceutical composition according to any one of claims 1 to 16 in the manufacture of a medicament for treating a disease or disorder by transporting the oligonucleotide across the alimentary canal mucosa of an animal.
21. Use according to any one of claims 17 to 20, wherein said animal is human.
22. Use according to any one of claims 17 to 21, wherein said medicament is in a sublingual, endoscopic or rectal form.
23. Use according to any one of claims 17 to 21, wherein said medicament is in oral form.
24. A method of modulating gene expression in cells or tissues in vitro comprising administering a composition comprising an oligonucleotide that comprises about 8 to about 30 nucleotides for use in antisense modulation of the function of DNA or messenger RNA encoding a protein, a fatty acid and a bile salt to said cells or tissues.
25. Use of a pharmaceutical composition according to any one of claims 1 to 16 for modulating expression of a cellular adhesion protein in an animal to treat a disease.
26. Use of a pharmaceutical composition according to any one of claims 1 to 16 for modulating the rate of cellular proliferation in an animal to treat a disease.
27. Use of a pharmaceutical composition according to any one of claims 1 to 16 for treating a disease resulting from eukaryotic pathogens, retroviruses or non-retroviral pathogens in an animal.
28. Use according to any one of claims 25 to 27, wherein said animal is human.
CA2294988A 1997-07-01 1998-07-01 Compositions and methods for the delivery of oligonucleotides via the alimentary canal Expired - Lifetime CA2294988C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US88682997A 1997-07-01 1997-07-01
US08/886,829 1997-07-01
PCT/US1998/013574 WO1999001579A1 (en) 1997-07-01 1998-07-01 Compositions and methods for the delivery of oligonucleotides via the alimentary canal

Publications (2)

Publication Number Publication Date
CA2294988A1 CA2294988A1 (en) 1999-01-14
CA2294988C true CA2294988C (en) 2015-11-24

Family

ID=25389866

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2294988A Expired - Lifetime CA2294988C (en) 1997-07-01 1998-07-01 Compositions and methods for the delivery of oligonucleotides via the alimentary canal

Country Status (8)

Country Link
US (4) US6887906B1 (en)
EP (1) EP1012331B1 (en)
JP (1) JP2002510319A (en)
AT (1) ATE321882T1 (en)
AU (1) AU731909B2 (en)
CA (1) CA2294988C (en)
DE (1) DE69834038D1 (en)
WO (1) WO1999001579A1 (en)

Families Citing this family (334)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US20070275921A1 (en) * 1996-06-06 2007-11-29 Isis Pharmaceuticals, Inc. Oligomeric Compounds That Facilitate Risc Loading
US7235653B2 (en) 1996-12-31 2007-06-26 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
EP1012331B1 (en) * 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
JP2003524586A (en) 1998-05-21 2003-08-19 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for parenteral administration of oligonucleotides
CA2329252A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals Inc. Compositions and methods for topical delivery of oligonucleotides
US7658938B2 (en) 1999-02-22 2010-02-09 Merrion Reasearch III Limited Solid oral dosage form containing an enhancer
US8119159B2 (en) * 1999-02-22 2012-02-21 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
US20070148228A1 (en) * 1999-02-22 2007-06-28 Merrion Research I Limited Solid oral dosage form containing an enhancer
WO2000050006A2 (en) 1999-02-26 2000-08-31 Chiron Corporation Microemulsions with adsorbed macromoelecules and microparticles
JP2002542263A (en) * 1999-04-21 2002-12-10 ワイス Methods and compositions for inhibiting the function of a polynucleotide sequence
US20040138168A1 (en) * 1999-04-21 2004-07-15 Wyeth Methods and compositions for inhibiting the function of polynucleotide sequences
DE60023285D1 (en) * 1999-05-05 2006-03-02 Merrion Res I Ltd INCREASED DISTRIBUTION OF NUCLEIC ACID-BASED MEDICINAL PRODUCTS
US20080281041A1 (en) * 1999-06-07 2008-11-13 Rozema David B Reversibly Masked Polymers
US8541548B2 (en) * 1999-06-07 2013-09-24 Arrowhead Madison Inc. Compounds and methods for reversible modification of biologically active molecules
US20030036520A1 (en) * 1999-07-23 2003-02-20 Jack Gauldie Intestinal gene therapy
CN101011584A (en) * 1999-12-28 2007-08-08 诺瓦提斯公司 Method for achieving persistent transgene expression
EP1118333A1 (en) * 2000-01-18 2001-07-25 Eurand International S.P.A. Compositions with enhanced oral bioavailability
US6544555B2 (en) 2000-02-24 2003-04-08 Advancis Pharmaceutical Corp. Antibiotic product, use and formulation thereof
US20020068078A1 (en) 2000-10-13 2002-06-06 Rudnic Edward M. Antifungal product, use and formulation thereof
ATE390938T1 (en) * 2001-02-20 2008-04-15 Anges Mg Inc TOPICAL USE OF A NF-KB DECOY FOR THE TREATMENT OF ATOPIC DERMATITIS
CA2446619C (en) 2001-05-11 2011-04-26 Elan Corporation, Plc Permeation enhancers
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US20030124196A1 (en) * 2001-08-22 2003-07-03 Susan Weinbach Pulsatile release compositions and methods for enhanced intestinal drug absorption
ATE554169T1 (en) * 2001-11-02 2012-05-15 Giuliani Int Ltd SMAD7 INHIBITORS FOR THE TREATMENT OF CNS DISEASES
AU2003213635A1 (en) * 2002-02-28 2003-09-16 Mayo Foundation For Medical Education And Research Compositions for generating submucosal fluid cushions
CA2504929C (en) 2002-11-05 2014-07-22 Charles Allerson Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
DK1572615T3 (en) * 2002-12-12 2010-06-14 Novartis Ag Process for the synthesis of peptides containing a 4-hydroxyproline substructure
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
ITRM20030149A1 (en) 2003-04-02 2004-10-03 Giuliani Spa ANTISENSE OLIGONUCLEOTIDES (ODN) FOR SMAD7 AND THEIR USE IN THE MEDICAL FIELD
MXPA05011022A (en) * 2003-04-13 2006-04-27 Enzon Pharmaceuticals Inc Polymeric oligonucleotide prodrugs.
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
EP1625851A4 (en) 2003-05-16 2007-12-26 Bbk Bio Corp Preparation for preventing contact of pathogenic matter with living organism
WO2005020885A2 (en) * 2003-05-21 2005-03-10 Isis Pharmaceuticals, Inc. Compositions and methods for the treatment of severe acute respiratory syndrome (sars)
US7897582B2 (en) 2003-05-23 2011-03-01 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US7960355B2 (en) 2003-05-23 2011-06-14 Isis Pharmaceuticals, Inc. Compositions and methods for the modulation of the expression of B7 protein
WO2005000202A2 (en) * 2003-05-23 2005-01-06 Isis Pharmaceuticals, Inc. Compositions and methods for the modulation of the expression of b7 protein
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
WO2005038013A1 (en) * 2003-10-07 2005-04-28 Isis Pharmaceuticals, Inc. Artisense oligonucleotides optimized for kidney targeting
US20050191653A1 (en) * 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
FR2862235B1 (en) * 2003-11-13 2007-12-28 Rhodia Chimie Sa EMULSION FOR VEHICULATING HYDROPHOBIC ACTIVE MATERIAL TO AQUEOUS SUBSTRATE
US7468431B2 (en) * 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
US20050244869A1 (en) * 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
AU2005230684B2 (en) * 2004-04-05 2011-10-06 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
CA2464522C (en) * 2004-04-15 2011-11-15 Eng-Hong Lee Soft gel delivery system for treating poultry
JP2008501335A (en) * 2004-06-03 2008-01-24 アイシス ファーマシューティカルズ、インク. Chimeric gapped oligomer composition
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
AU2005252663B2 (en) * 2004-06-03 2011-07-07 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
EP1791567B1 (en) * 2004-08-10 2015-07-29 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
US20060046969A1 (en) * 2004-08-25 2006-03-02 Aegis Therapeutics Llc Antibacterial compositions for drug administration
US20060046962A1 (en) 2004-08-25 2006-03-02 Aegis Therapeutics Llc Absorption enhancers for drug administration
US20140162965A1 (en) 2004-08-25 2014-06-12 Aegis Therapeutics, Inc. Compositions for oral drug administration
US8268791B2 (en) * 2004-08-25 2012-09-18 Aegis Therapeutics, Llc. Alkylglycoside compositions for drug administration
US9114069B2 (en) * 2004-08-25 2015-08-25 Aegis Therapeutics, Llc Antibacterial compositions for drug administration
US9895444B2 (en) 2004-08-25 2018-02-20 Aegis Therapeutics, Llc Compositions for drug administration
US20090047347A1 (en) * 2005-07-29 2009-02-19 Aegis Therapeutics, Inc. Compositions for Drug Administration
US8642564B2 (en) * 2004-08-25 2014-02-04 Aegis Therapeutics, Llc Compositions for drug administration
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
CA2583413A1 (en) * 2004-09-21 2006-03-30 Anesiva, Inc. Delivery of polynucleotides
MX2007006591A (en) * 2004-12-02 2008-03-04 Isis Pharmaceuticals Inc Therapeutic antisense oligonucleotide composition for the treatment of inflammatory bowel disease.
US20060240108A1 (en) * 2005-04-26 2006-10-26 Bernard Bobby L Cellulosic films incorporating a pharmaceutically acceptable plasticizer with enhanced wettability
GT200600405A (en) * 2005-09-07 2007-04-16 MICROEMULSION FORMULA
EP1764107A1 (en) * 2005-09-14 2007-03-21 Gunther Hartmann Compositions comprising immunostimulatory RNA oligonucleotides and methods for producing said RNA oligonucleotides
EP2202239A1 (en) * 2005-11-01 2010-06-30 Alnylam Pharmaceuticals Inc. RNAI inhibition of influenza virus replication
WO2007051303A1 (en) 2005-11-02 2007-05-10 Protiva Biotherapeutics, Inc. MODIFIED siRNA MOLECULES AND USES THEREOF
CA2630602A1 (en) 2005-11-21 2007-05-31 Isis Pharmaceuticals, Inc. Modulation of eif4e-bp2 expression
US8229398B2 (en) * 2006-01-30 2012-07-24 Qualcomm Incorporated GSM authentication in a CDMA network
BRPI0710503A2 (en) 2006-04-07 2011-08-16 Merrion Res Iii Ltd use of a pharmaceutical composition, pharmaceutical composition, and oral dosage form
CN101437943A (en) * 2006-05-03 2009-05-20 波罗的科技发展有限公司 Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease
BRPI0712034A2 (en) * 2006-05-19 2012-01-10 Alnylam Pharmaceuticals Inc aha rnai modulation and therapeutic uses thereof
WO2007137301A2 (en) * 2006-05-23 2007-11-29 Isis Pharmaceuticals, Inc. Modulation of chrebp expression
JP2009539862A (en) * 2006-06-09 2009-11-19 メリオン リサーチ Iii リミテッド Solid oral dosage form with toughener
US8226949B2 (en) 2006-06-23 2012-07-24 Aegis Therapeutics Llc Stabilizing alkylglycoside compositions and methods thereof
WO2008011473A2 (en) 2006-07-19 2008-01-24 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hbxip
AU2007285782B2 (en) * 2006-08-18 2010-06-24 Arrowhead Research Corporation Polyconjugates for in vivo delivery of polynucleotides
US8017109B2 (en) * 2006-08-18 2011-09-13 Roche Madison Inc. Endosomolytic poly(acrylate) polymers
EP2063709A2 (en) * 2006-09-15 2009-06-03 Enzon Pharmaceuticals, Inc. Hindered ester-based biodegradable linkers for oligonucleotide delivery
MX2009002854A (en) * 2006-09-15 2009-03-27 Enzon Pharmaceuticals Inc Polyalkylene oxides having hindered ester-based biodegradable linkers.
US8110559B2 (en) * 2006-09-15 2012-02-07 Enzon Pharmaceuticals, Inc. Hindered ester-based biodegradable linkers for oligonucleotide delivery
US20100279408A1 (en) * 2006-11-27 2010-11-04 Enzon Pharmaceuticals, Inc. Polymeric short interfering rna conjugates
EP2118118B1 (en) * 2007-01-19 2017-09-27 Exiqon A/S Mediated cellular delivery of lna oligonucleotides
AR066984A1 (en) 2007-06-15 2009-09-23 Novartis Ag INHIBITION OF THE EXPRESSION OF THE ALFA SUBUNITY OF THE SODIUM EPITELIAL CHANNEL (ENAC) THROUGH ARNI (INTERFERENCE RNA)
WO2009045536A2 (en) * 2007-10-05 2009-04-09 The University Of North Carolina At Chapel Hill Receptor targeted oligonucleotides
JP2011502502A (en) * 2007-11-05 2011-01-27 バルティック テクロノジー デヴェロプメント,リミテッド Use of oligonucleotides containing modified bases in nucleic acid hybridization
US7834172B2 (en) * 2007-11-14 2010-11-16 Novagali Pharma Sa Composition comprising at least one nucleosidic moiety as a therapeutic agent, and CKC
EP2238251B1 (en) 2007-12-27 2015-02-11 Protiva Biotherapeutics Inc. Silencing of polo-like kinase expression using interfering rna
KR101397407B1 (en) 2008-03-05 2014-06-19 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibiting expression of Eg5 and VEGF genes
EP2105145A1 (en) 2008-03-27 2009-09-30 ETH Zürich Method for muscle-specific delivery lipid-conjugated oligonucleotides
CA2756690C (en) * 2008-03-28 2016-08-16 Hale Biopharma Ventures, Llc Administration of benzodiazepine compositions
CA2721380A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Silencing of csn5 gene expression using interfering rna
US8324366B2 (en) 2008-04-29 2012-12-04 Alnylam Pharmaceuticals, Inc. Compositions and methods for delivering RNAI using lipoproteins
JP5671451B2 (en) 2008-05-07 2015-02-18 メリオン・リサーチ・Iii・リミテッド Compositions and preparation processes for GnRH related compounds
WO2010008582A2 (en) 2008-07-18 2010-01-21 Rxi Pharmaceuticals Corporation Phagocytic cell drug delivery system
EP2317847B1 (en) 2008-07-29 2019-04-17 The Board of Regents of The University of Texas System Selective inhibition of polyglutamine protein expression
EP2323667A4 (en) * 2008-08-07 2012-07-25 Isis Pharmaceuticals Inc Modulation of transthyretin expression for the treatment of cns related disorders
EP2331690B1 (en) 2008-09-02 2016-01-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of mutant egfr gene
CA2753338A1 (en) 2008-09-22 2010-03-25 Rxi Pharmaceuticals Corporation Neutral nanotransporters
EP3109321B1 (en) 2008-09-25 2019-05-01 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
EP3848461A1 (en) 2008-10-20 2021-07-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
US8715715B2 (en) 2008-11-03 2014-05-06 Nal Pharmaceuticals Ltd. Dosage form for insertion into the mouth
US20100222417A1 (en) * 2008-11-26 2010-09-02 Alnylam Pharmaceuticals Compostions and methods for enhancing oligonucleotide delivery across and into epithelial tissues
JP5855462B2 (en) 2008-12-10 2016-02-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. DsRNA compositions targeting GNAQ and methods for inhibiting expression
US8440631B2 (en) 2008-12-22 2013-05-14 Aegis Therapeutics, Llc Compositions for drug administration
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
AR075613A1 (en) * 2009-02-25 2011-04-20 Merrion Res Iii Ltd COMPOSITION AND SUPPLY OF BIFOSPHONATE DRUGS. TREATMENT METHOD
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
EP2406376A1 (en) 2009-03-12 2012-01-18 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
CA2761106C (en) 2009-05-05 2019-01-15 Miragen Therapeutics Lipophilic polynucleotide conjugates
JP2012526840A (en) * 2009-05-13 2012-11-01 プロテイン デリヴァリー ソリューションズ エルエルシー Formulation system for transmembrane delivery
US9029338B2 (en) 2009-08-14 2015-05-12 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
CA2780741C (en) 2009-10-12 2023-04-04 Smith Holdings, Llc Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
US8841429B2 (en) 2009-11-03 2014-09-23 Vivonics, Inc. Nucleic acid ligands against infectious prions
US8236570B2 (en) 2009-11-03 2012-08-07 Infoscitex Methods for identifying nucleic acid ligands
AU2010329847A1 (en) 2009-12-11 2012-07-26 Genecode As Methods of facilitating neural cell survival using GDNF family ligand (GFL) mimetics or RET signaling pathway activators
AU2010339907A1 (en) 2009-12-16 2012-07-05 Nod Pharmaceuticals, Inc. Compositions and methods for oral drug delivery
US20110142889A1 (en) * 2009-12-16 2011-06-16 Nod Pharmaceuticals, Inc. Compositions and methods for oral drug delivery
US20110182985A1 (en) * 2010-01-28 2011-07-28 Coughlan David C Solid Pharmaceutical Composition with Enhancers and Methods of Preparing thereof
WO2011097388A1 (en) 2010-02-03 2011-08-11 Alnylam Pharmaceuticals, Inc. Selective inhibition of polyglutamine protein expression
PL2539451T3 (en) * 2010-02-24 2016-08-31 Arrowhead Res Corporation Compositions for targeted delivery of sirna
EP2550000A4 (en) 2010-03-24 2014-03-26 Advirna Inc Reduced size self-delivering rnai compounds
WO2011120033A1 (en) 2010-03-26 2011-09-29 Merrion Research Iii Limited Pharmaceutical compositions of selective factor xa inhibitors for oral administration
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
JP5058351B2 (en) * 2010-04-13 2012-10-24 株式会社 資生堂 Oil-in-water emulsion composition
SI2563920T1 (en) 2010-04-29 2017-05-31 Ionis Pharmaceuticals, Inc. Modulation of transthyretin expression
CA3102008A1 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
EP2582387A2 (en) 2010-06-17 2013-04-24 The United States Of America As Represented By The Secretary, National Institutes Of Health Compositions and methods for treating inflammatory conditions
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2012023291A1 (en) * 2010-08-20 2012-02-23 国立大学法人東京医科歯科大学 Pharmaceutical composition for transcolonic absorption
NZ608972A (en) 2010-10-01 2015-09-25 Moderna Therapeutics Inc Engineered nucleic acids and methods of use thereof
JP2012111933A (en) * 2010-11-02 2012-06-14 Nitto Denko Corp Thermoplastic silicone resin
CA2817002C (en) 2010-11-05 2019-01-15 Miragen Therapeutics Base modified oligonucleotides
CN110123830A (en) 2010-11-09 2019-08-16 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting the lipid of the expression of Eg5 and VEGF gene to prepare
EP2649182A4 (en) 2010-12-10 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
BR112013012319A2 (en) 2010-12-15 2019-09-24 Miragen Therapeutics micro rna inhibitors comprising blocked nucleotides
JP2014501784A (en) 2011-01-07 2014-01-23 メリオン・リサーチ・Iii・リミテッド Pharmaceutical composition of iron for oral administration
ES2528472T3 (en) 2011-02-03 2015-02-10 The Government Of The U.S.A, As Represented By The Secretary, Dept. Of Health And Human Services Multivalent vaccines for rabies virus and filovirus
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
EP2697374A2 (en) 2011-04-12 2014-02-19 Beth Israel Deaconess Medical Center, Inc. Micro-rna inhibitors and their uses in disease
KR101970634B1 (en) 2011-06-02 2019-04-19 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Methods and uses for ex vivo tissue culture systems
EP2720699B1 (en) 2011-06-14 2018-05-16 Hale Biopharma Ventures, Llc Administration of benzodiazepine
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
EP3564393A1 (en) 2011-06-21 2019-11-06 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
CN103890000B (en) 2011-06-21 2017-09-01 阿尔尼拉姆医药品有限公司 (ANGPTL3) the iRNA compositions of angiopoietin-like 3 and its application method
CA3191066A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP4134433A1 (en) 2011-06-23 2023-02-15 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
EP2724156B1 (en) 2011-06-27 2017-08-16 The Jackson Laboratory Methods and compositions for treatment of cancer and autoimmune disease
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
CA2844577A1 (en) 2011-09-13 2013-03-21 Ottawa Hospital Research Institute Microrna inhibitors
DE19216461T1 (en) 2011-10-03 2021-10-07 Modernatx, Inc. MODIFIED NUCLEOSIDES, NUCLEOTIDES AND NUCLEIC ACIDS AND USES THEREOF
JP2014533953A (en) 2011-11-17 2014-12-18 ザ ユナイテッド ステイツ オブ アメリカ, アズ リプレゼンテッド バイ ザ セクレタリー, デパートメント オブ ヘルス アンド ヒューマン サービシーズ Therapeutic RNA switch compositions and methods of use
WO2013090648A1 (en) 2011-12-16 2013-06-20 modeRNA Therapeutics Modified nucleoside, nucleotide, and nucleic acid compositions
US9035039B2 (en) 2011-12-22 2015-05-19 Protiva Biotherapeutics, Inc. Compositions and methods for silencing SMAD4
JP2015509943A (en) 2012-02-21 2015-04-02 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル TAM receptor as a viral entry cofactor
WO2013124327A1 (en) 2012-02-21 2013-08-29 Institut National De La Sante Et De La Recherche Medicale (Inserm) Tim receptors as virus entry cofactors
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
AU2013243948A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
GB201209895D0 (en) 2012-06-01 2012-07-18 Atlantic Pharmaceuticals Holdings Ltd Use of a composition
MX355408B (en) 2012-06-21 2018-04-18 Miragen Therapeutics Inc Oligonucleotide-based inhibitors comprising locked nucleic acid motif.
EP2877492A1 (en) 2012-07-27 2015-06-03 Institut National de la Santé et de la Recherche Médicale (INSERM) Cd147 as receptor for pilus-mediated adhesion of meningococci to vascular endothelia
RU2657749C2 (en) 2012-09-21 2018-06-15 Интенсити Терапьютикс, Инк Methods of treating cancer
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
WO2014089313A1 (en) 2012-12-05 2014-06-12 Alnylam Pharmaceuticals PCSK9 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
FI3732994T3 (en) 2013-02-08 2023-01-13 Reduced sodium food products
PE20160046A1 (en) 2013-03-14 2016-02-14 Alnylam Pharmaceuticals Inc COMPOSITION OF RNAi AGAINST THE C5 COMPONENT OF THE COMPLEMENT AND METHODS FOR ITS USE
DK2970968T3 (en) 2013-03-15 2018-03-05 Miragen Therapeutics Inc CONNECTED BICYCLIC Nucleosides
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP2994167B1 (en) 2013-05-06 2020-05-06 Alnylam Pharmaceuticals, Inc. Dosages and methods for delivering lipid formulated nucleic acid molecules
SG10201913872XA (en) 2013-05-22 2020-03-30 Alnylam Pharmaceuticals Inc SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
EP3047023B1 (en) 2013-09-19 2019-09-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for inhibiting jc virus (jcv)
EP3052626A1 (en) 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
TW202310853A (en) 2013-10-04 2023-03-16 美國西奈山伊坎醫學院 Compositions and methods for inhibiting expression of the alas1 gene
CA2930693A1 (en) 2013-11-15 2015-05-21 The Board Of Trustees Of The Leland Stanford Junior Unversity Methods of treating heart failure with agonists of hypocretin receptor 2
US10772974B2 (en) 2013-11-18 2020-09-15 Beth Israel Deaconess Medical Center, Inc. Compositions and methods for cardiac regeneration
CA2931090A1 (en) 2013-12-12 2015-06-18 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
GB201407822D0 (en) 2014-05-02 2014-06-18 Atlantic Pharmaceuticals Holdings Ltd Use of a composition
TW201607559A (en) 2014-05-12 2016-03-01 阿尼拉製藥公司 Methods and compositions for treating a SERPINC1-associated disorder
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
US9752144B2 (en) 2014-08-04 2017-09-05 MiRagen Therapeutics, Inc. Inhibitors of MYH7B and uses thereof
MX2017003018A (en) 2014-09-08 2018-01-24 Miragen Therapeutics Inc Mir-29 mimics and uses thereof.
WO2016040589A1 (en) 2014-09-12 2016-03-17 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
AU2015328012A1 (en) 2014-10-02 2017-05-11 Arbutus Biopharma Corporation Compositions and methods for silencing Hepatitis B virus gene expression
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CN113846101A (en) 2014-11-17 2021-12-28 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
TW201702218A (en) 2014-12-12 2017-01-16 美國杰克森實驗室 Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
US9885042B2 (en) 2015-01-20 2018-02-06 MiRagen Therapeutics, Inc. miR-92 inhibitors and uses thereof
CA2974369A1 (en) 2015-01-20 2016-07-28 The Children's Medical Center Corporation Anti-net compounds for treating and preventing fibrosis and for facilitating wound healing
WO2016120378A1 (en) 2015-01-29 2016-08-04 Novo Nordisk A/S Tablets comprising glp-1 agonist and enteric coating
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
US10627340B2 (en) 2015-04-09 2020-04-21 Bikanta Corporation Imaging systems and methods using fluorescent nanodiamonds
WO2016179634A1 (en) 2015-05-11 2016-11-17 Murdoch University Multiple sclerosis treatment
WO2016197132A1 (en) 2015-06-04 2016-12-08 Protiva Biotherapeutics Inc. Treating hepatitis b virus infection using crispr
KR102034619B1 (en) 2015-06-05 2019-11-11 미라젠 세러퓨틱스 인코포레이티드 MIR-155 inhibitor for the treatment of cutaneous T cell lymphoma (CTCL)
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
CA2989970A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
US20180208932A1 (en) 2015-07-29 2018-07-26 Arbutus Biopharma Corporation Compositions and methods for silencing hepatitis b virus gene expression
US10889813B2 (en) 2015-09-02 2021-01-12 Alnylam Pharmaceuticals, Inc. Programmed cell death 1 ligand 1 (PD-L1) iRNA compositions and methods of use thereof
JP6875685B2 (en) * 2015-10-08 2021-05-26 日油株式会社 O / W type emulsion
CA3002744A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
EP3377073A4 (en) 2015-11-16 2019-06-26 Ohio State Innovation Foundation Methods and compositions for treating disorders and diseases using survival motor neuron (smn) protein
US20170360815A1 (en) 2016-02-25 2017-12-21 Applied Biological Laboratories, Inc. Compositions and methods for protecting against airborne pathogens and irritants
CN109069520A (en) 2016-02-25 2018-12-21 应用生物实验室公司 Protect the composition and method of airborne pathogen and stimulant
WO2017176596A1 (en) 2016-04-04 2017-10-12 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Multivalent vaccines for rabies virus and coronaviruses
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
MA45290A (en) * 2016-05-04 2019-03-13 Wave Life Sciences Ltd PROCESSES AND COMPOSITIONS OF BIOLOGICALLY ACTIVE AGENTS
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
UY37376A (en) 2016-08-26 2018-03-23 Amgen Inc ARNI CONSTRUCTIONS TO INHIBIT EXPRESSION OF ASGR1 AND METHODS FOR USE
ES2659845B1 (en) 2016-09-19 2019-01-04 Univ Valencia Modulation of microRNAs against myotonic dystrophy type 1 and antagonists of microRNAs for this
EP3538654A1 (en) 2016-11-11 2019-09-18 Janssen BioPharma, Inc. Oligonucleotide targeting strategy for hbv cccdna
US20190070109A1 (en) * 2016-11-18 2019-03-07 National Tsing Hua University Pharmaceutical composition for oral delivery
TW201818926A (en) * 2016-11-18 2018-06-01 國立清華大學 Pharmaceutical composition
TW202313978A (en) 2016-11-23 2023-04-01 美商阿尼拉製藥公司 Serpina1 irna compositions and methods of use thereof
TWI790217B (en) 2016-12-16 2023-01-21 美商阿尼拉製藥公司 METHODS FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES USING TRANSTHYRETIN (TTR) iRNA COMPOSITIONS
GB201700257D0 (en) * 2017-01-06 2017-02-22 Atlantic Pharmaceuticals (Holdings) Ltd New formulation
US11459563B2 (en) 2017-02-03 2022-10-04 The University Of Western Australia Treatment for NEAT1 associated disease
TWI801377B (en) 2017-04-18 2023-05-11 美商阿尼拉製藥公司 Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
CN110832077A (en) 2017-07-06 2020-02-21 箭头药业股份有限公司 RNAi agents for inhibiting α -ENaC expression and methods of use
EP3652317A1 (en) 2017-07-13 2020-05-20 Alnylam Pharmaceuticals, Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
WO2019051173A1 (en) 2017-09-08 2019-03-14 Ionis Pharmaceuticals, Inc. Modulators of smad7 expression
US11555189B2 (en) 2017-10-18 2023-01-17 Sarepta Therapeutics, Inc. Antisense oligomer compounds
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
WO2019099610A1 (en) 2017-11-16 2019-05-23 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
WO2019101882A1 (en) 2017-11-23 2019-05-31 INSERM (Institut National de la Santé et de la Recherche Médicale) New method for treating dengue virus infection
AR113490A1 (en) 2017-12-12 2020-05-06 Amgen Inc RNAi CONSTRUCTIONS TO INHIBIT THE EXPRESSION OF PNPLA3 AND METHODS OF USE OF THE SAME
JP7359396B2 (en) 2017-12-15 2023-10-11 ジ・オーストラリアン・ナショナル・ユニヴァーシティ Compounds for treating and preventing pathologies mediated by extracellular histones
MX2020006012A (en) 2017-12-18 2020-09-14 Alnylam Pharmaceuticals Inc High mobility group box-1 (hmgb1) irna compositions and methods of use thereof.
WO2019126542A1 (en) 2017-12-20 2019-06-27 Allergan, Inc. Botulinum toxin cell binding domain polypeptides and methods of use for treatments of fibrosis associated disorders
CN111801317A (en) 2017-12-21 2020-10-20 卡耐基梅隆大学 Template-directed nucleic acid-targeting compounds
PT3514235T (en) * 2018-01-18 2024-02-12 Sterna Biologicals Gmbh Composition for the treatment of a patient suffering from ulcerative colitis and utilisation of said composition as medicament
CA3093572A1 (en) 2018-03-14 2019-09-19 Beth Israel Deaconess Medical Center Inhibitors of micro-rna 22
EP3837367A1 (en) 2018-08-16 2021-06-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
JP2022500003A (en) 2018-09-18 2022-01-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA composition and its usage
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
US20220049252A1 (en) 2018-12-10 2022-02-17 Amgen Inc. CHEMICALLY-MODIFIED RNAi CONSTRUCTS AND USES THEREOF
AR117297A1 (en) 2018-12-10 2021-07-28 Amgen Inc ARNI CONSTRUCTS TO INHIBIT THE EXPRESSION OF PNPLA3 AND METHODS OF USE OF THEM
HUE064532T2 (en) 2018-12-20 2024-03-28 Vir Biotechnology Inc Combination hbv therapy
SG11202112144RA (en) 2019-05-14 2021-11-29 Univ Monash Modulators and modulation of the receptor for advanced glycation end-products rna
AU2020284254A1 (en) 2019-05-30 2021-12-23 Amgen Inc. RNAi constructs for inhibiting SCAP expression and methods of use thereof
JP2022544238A (en) 2019-08-13 2022-10-17 アムジエン・インコーポレーテツド RNAi constructs for inhibiting SLC30A8 expression and methods of use thereof
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
JP2022546570A (en) 2019-09-03 2022-11-04 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting expression of the LECT2 gene
EP4038189A1 (en) 2019-10-04 2022-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
US20210106525A1 (en) 2019-10-11 2021-04-15 Massachusetts Institute Of Technology Formulations for gastrointestinal delivery of oligonucleotides
US20230040920A1 (en) 2019-11-01 2023-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
MX2022004388A (en) 2019-11-01 2022-05-06 Alnylam Pharmaceuticals Inc HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF.
US20230056569A1 (en) 2019-11-22 2023-02-23 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
JP2023504744A (en) 2019-12-09 2023-02-06 アムジエン・インコーポレーテツド RNAi constructs and methods for inhibiting LPA expression
EP4073251A1 (en) 2019-12-13 2022-10-19 Alnylam Pharmaceuticals, Inc. Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
TW202204615A (en) 2020-03-26 2022-02-01 美商阿尼拉製藥公司 Coronavirus irna compositions and methods of use thereof
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
BR112022020145A2 (en) 2020-04-06 2023-01-03 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR SILENCING THE MYOC EXPRESSION
TW202204617A (en) 2020-04-07 2022-02-01 美商艾爾妮蘭製藥公司 Compositions and methods for silencing scn9a expression
EP4133076A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Angiotensin-converting enzyme 2 (ace2) irna compositions and methods of use thereof
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
EP4143319A1 (en) 2020-04-27 2023-03-08 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
EP4150086A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
WO2021231675A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
WO2021234607A1 (en) 2020-05-20 2021-11-25 St. Jude Children's Research Hospital Detection of alzheimer's disease using specific biomarkers
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
US20230279399A1 (en) 2020-06-01 2023-09-07 Amgen Inc. Rnai constructs for inhibiting hsd17b13 expression and methods of use thereof
WO2021252557A1 (en) 2020-06-09 2021-12-16 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
BR112022026316A2 (en) 2020-06-24 2023-03-07 Vir Biotechnology Inc ENGINEERED HEPATITIS B VIRUS NEUTRALIZING ANTIBODIES AND THEIR USES
EP3929295A1 (en) 2020-06-26 2021-12-29 Universitat Pompeu Fabra Artificial rnas for modulating rna fragments
MX2023001786A (en) 2020-08-13 2023-03-10 Amgen Inc RNAi CONSTRUCTS AND METHODS FOR INHIBITING MARC1 EXPRESSION.
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
AU2021365822A1 (en) 2020-10-21 2023-06-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
IL302330A (en) 2020-11-05 2023-06-01 Amgen Inc METHODS FOR TREATING ATHEROSCLEROTIC CARDIOVASCULAR DISEASE WITH LPA-TARGETED RNAi CONSTRUCTS
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
BR112023015761A2 (en) 2021-02-12 2023-11-07 Alnylam Pharmaceuticals Inc SUPEROXIDE DISMUTASE 1 (SOD1) IRNA COMPOSITIONS AND METHODS OF USE THEREOF TO TREAT OR PREVENT NEURODEGENERATIVE DISEASES ASSOCIATED WITH SUPEROXIDE DISMUTASE 1 (SOD1)
WO2022182864A1 (en) 2021-02-25 2022-09-01 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods and methods of use thereof
EP4305169A1 (en) 2021-03-12 2024-01-17 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
KR20230162024A (en) 2021-03-29 2023-11-28 알닐람 파마슈티칼스 인코포레이티드 Huntingtin (HTT) iRNA preparation composition and method of use thereof
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
EP4341401A1 (en) 2021-05-18 2024-03-27 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
IL308743A (en) 2021-06-04 2024-01-01 Alnylam Pharmaceuticals Inc HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
AR126070A1 (en) 2021-06-08 2023-09-06 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR TREATING OR PREVENTING STARGARDT DISEASE AND/OR DISORDERS ASSOCIATED WITH RETINOL BORDER PROTEIN 4 (RBP4)
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
TW202333748A (en) 2021-07-19 2023-09-01 美商艾拉倫製藥股份有限公司 Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
KR20240037293A (en) 2021-07-23 2024-03-21 알닐람 파마슈티칼스 인코포레이티드 Beta-catenin (CTNNB1) iRNA composition and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
IL310244A (en) 2021-08-03 2024-03-01 Alnylam Pharmaceuticals Inc Transthyretin (ttr) irna compositions and methods of use thereof
CA3228255A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing angiotensinogen (agt)
AR126771A1 (en) 2021-08-13 2023-11-15 Alnylam Pharmaceuticals Inc RNAi COMPOSITIONS AGAINST FACTOR XII (F12) AND THEIR METHODS OF USE
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
AU2022345881A1 (en) 2021-09-20 2024-03-21 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
AU2022359289A1 (en) 2021-10-05 2024-04-04 Amgen Inc. Compositions and methods for enhancing gene silencing activity of oligonucleotide compounds
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023069754A2 (en) 2021-10-22 2023-04-27 Amgen Inc. Rnai constructs for inhibiting gpam expression and methods of use thereof
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023144163A1 (en) 2022-01-25 2023-08-03 Albert-Ludwigs-Universität Freiburg A glun2d inhibitor for use in the treatment or relapse prevention of a depressive episode
WO2023177870A1 (en) 2022-03-18 2023-09-21 Baylor College Of Medicine Potentiation of durable antitumor immunity by multifactorial immune modulation
WO2023183556A2 (en) * 2022-03-24 2023-09-28 Amcyte Pharma, Inc. Treatment for retinal disorders
US20240084301A1 (en) 2022-07-25 2024-03-14 Amgen Inc. Rnai constructs and methods for inhibiting fam13a expression
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof

Family Cites Families (244)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA218654A (en) 1922-05-16 The Canadian Westinghouse Company Heat regulating attachment for tube machines
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
AR205589A1 (en) 1974-10-09 1976-05-14 Reckitt & Colmann Prod Ltd INTRODUCING DEVICE OF AN AQUEOUS COMPOSITION INTO A BODY CAVITY
JPS5780314U (en) 1980-11-04 1982-05-18
JPS5780314A (en) * 1980-11-09 1982-05-19 Kyoto Yakuhin Kogyo Kk Pharmaceutical preparation to be medicated to rectum
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
JPS5927900A (en) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
FR2540122B1 (en) 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4948882A (en) 1983-02-22 1990-08-14 Syngene, Inc. Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
NZ209840A (en) 1983-10-17 1988-11-29 Kaji Akira A method of inhibiting viral propagation by hybridising dna with the viral rna thus blocking its action
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
US5298614A (en) 1986-01-06 1994-03-29 Nippon Shinyaku Co. Ltd. Size limited double stranded poly I poly(cytidylate/4-thiouridylate)
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
US4878979A (en) 1986-04-25 1989-11-07 United Technologies Corporation Method of reusably sealing a silicone rubber vacuum bag to a mold for composite manufacture
US5194428A (en) 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US4806463A (en) 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
JPS638396A (en) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd Poly-labeled oligonucleotide derivative
DE3788914T2 (en) 1986-09-08 1994-08-25 Ajinomoto Kk Compounds for cleaving RNA at a specific position, oligomers used in the preparation of these compounds and starting materials for the synthesis of these oligomers.
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
EP0366685B1 (en) 1987-06-24 1994-10-19 Howard Florey Institute Of Experimental Physiology And Medicine Nucleoside derivatives
US5585481A (en) 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5525465A (en) 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
EP0348458B1 (en) 1987-11-30 1997-04-09 University Of Iowa Research Foundation Dna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
US5403711A (en) 1987-11-30 1995-04-04 University Of Iowa Research Foundation Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved
US5543508A (en) 1987-12-15 1996-08-06 Gene Shears Pty. Limited Ribozymes
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
JPH03503894A (en) 1988-03-25 1991-08-29 ユニバーシィティ オブ バージニア アランミ パテンツ ファウンデイション Oligonucleotide N-alkylphosphoramidate
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5004810A (en) 1988-09-30 1991-04-02 Schering Corporation Antiviral oligomers
US5098890A (en) 1988-11-07 1992-03-24 Temple University-Of The Commonwealth System Of Higher Education Antisence oligonucleotides to c-myb proto-oncogene and uses thereof
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5087617A (en) 1989-02-15 1992-02-11 Board Of Regents, The University Of Texas System Methods and compositions for treatment of cancer using oligonucleotides
US5599923A (en) 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US5108921A (en) 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
ATE190981T1 (en) 1989-10-24 2000-04-15 Isis Pharmaceuticals Inc 2'-MODIFIED NUCLEOTIDES
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5223168A (en) 1989-12-12 1993-06-29 Gary Holt Surface cleaner and treatment
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5591623A (en) 1990-08-14 1997-01-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5859221A (en) 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5506351A (en) 1992-07-23 1996-04-09 Isis Pharmaceuticals Process for the preparation of 2'-O-alkyl guanosine and related compounds
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5514788A (en) 1993-05-17 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5212295A (en) 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
US5955589A (en) 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5149797A (en) 1990-02-15 1992-09-22 The 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
AU7579991A (en) 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
ATE167523T1 (en) 1990-05-11 1998-07-15 Microprobe Corp IMMERSIBLE TEST STRIPS FOR NUCLEIC ACID HYBRIDIZATION ASSAY AND METHOD FOR COVALENT IMMOBILIZATION OF OLIGONUCLEOTIDES
US5166195A (en) 1990-05-11 1992-11-24 Isis Pharmaceuticals, Inc. Antisense inhibitors of the human immunodeficiency virus phosphorothioate oligonucleotides
US5135917A (en) 1990-07-12 1992-08-04 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
ATE154246T1 (en) 1990-07-27 1997-06-15 Isis Pharmaceuticals Inc NUCLEASE RESISTANT PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECTE AND MODULATE GENE EXPRESSION
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
WO1992002534A2 (en) 1990-08-03 1992-02-20 Sterling Drug, Inc. Compounds and methods for inhibiting gene expression
US6262241B1 (en) 1990-08-13 2001-07-17 Isis Pharmaceuticals, Inc. Compound for detecting and modulating RNA activity and gene expression
US6096722A (en) * 1990-08-14 2000-08-01 Isis Pharmaceuticals Inc. Antisense modulation of cell adhesion molecule expression and treatment of cell adhesion molecule-associated diseases
US5843738A (en) * 1990-08-14 1998-12-01 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US6111094A (en) 1990-08-14 2000-08-29 Isis Pharmaceuticals Inc. Enhanced antisense modulation of ICAM-1
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5595978A (en) 1990-08-16 1997-01-21 Isis Pharmaceuticals, Inc. Composition and method for treatment of CMV retinites
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
JPH06505704A (en) 1990-09-20 1994-06-30 ギリアド サイエンシズ,インコーポレイテッド Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
KR930702373A (en) 1990-11-08 1993-09-08 안토니 제이. 페이네 Addition of Multiple Reporter Groups to Synthetic Oligonucleotides
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5674530A (en) 1991-01-31 1997-10-07 Port Systems, L.L.C. Method for making a multi-stage drug delivery system
US5656272A (en) 1991-03-18 1997-08-12 New York University Medical Center Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies
US5242906A (en) 1991-04-22 1993-09-07 University Of North Carolina At Chapel Hill Antisense oligonucleotides against Epstein-Barr virus
WO1992020813A1 (en) 1991-05-17 1992-11-26 Dana Farber Cancer Institute Assays for factors affecting circularization of dna, assays for factors affecting dna integration, factors, and uses thereof
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
EP0587749A4 (en) 1991-05-31 1995-08-30 Genta Inc Compositions and delivery systems for transdermal administration of neutral oligomers
WO1993000446A1 (en) 1991-06-27 1993-01-07 Genelabs Technologies, Inc. Screening assay for the detection of dna-binding molecules
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
US5697248A (en) 1991-07-25 1997-12-16 The Whitaker Corporation Liquid level sensor
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
US5599797A (en) 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5338837A (en) 1991-12-13 1994-08-16 The Trustees Of Princeton University Glycosylated steroid derivatives for transport across biological membranes and process for making same
US5780444A (en) 1991-12-13 1998-07-14 Trustees Of Princeton University Compositions and methods for cell transformation
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5595726A (en) 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
US5565552A (en) 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US5948898A (en) 1992-03-16 1999-09-07 Isis Pharmaceuticals, Inc. Methoxyethoxy oligonucleotides for modulation of protein kinase C expression
WO1993019660A1 (en) 1992-04-03 1993-10-14 Baylor College Of Medicine Gene therapy using the intestine
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
FR2692265B1 (en) 1992-05-25 1996-11-08 Centre Nat Rech Scient BIOLOGICALLY ACTIVE COMPOUNDS OF THE PHOSPHOTRIESTER TYPE.
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
IT1255449B (en) * 1992-06-30 1995-10-31 Fabio Berlati USE OF NOR- AND HOMO-DERIVATIVES OF BILE ACIDS AS DRUGS ABSORPTION PROMOTERS.
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
RU95104940A (en) 1992-07-27 1997-01-10 Хайбрайдон Method of incorporation of alkylphosphonothioate or arylphosphonothioate internucleotide linkage in oligonucleotide, method of oligonucleotide synthesis, method of gene expression inhibition, treatment method
WO1994005333A1 (en) 1992-09-02 1994-03-17 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
CA2105595A1 (en) 1992-09-23 1994-03-24 Ramaswamy Narayanan Antisense polynucleotides
IL107150A0 (en) 1992-09-29 1993-12-28 Isis Pharmaceuticals Inc Oligonucleotides having a conserved g4 core sequence
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
US5631237A (en) 1992-12-22 1997-05-20 Dzau; Victor J. Method for producing in vivo delivery of therapeutic agents via liposomes
DE69400208T2 (en) 1993-01-25 1996-11-28 Hybridon Inc OLIONUCLEOTIDALKYLPHOSPHONATES AND PHOSPHONOTHIOATES
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
CA2156512C (en) 1993-02-19 2002-11-19 Mary Ellen Harper Treatment of androgen-associated baldness using antisense oligomers
JP3189279B2 (en) 1993-02-19 2001-07-16 日本新薬株式会社 Pharmaceutical composition containing nucleic acid copolymer
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
CA2159631A1 (en) 1993-03-30 1994-10-13 Sanofi Acyclic nucleoside analogs and oligonucleotide sequences containing them
CA2159629A1 (en) 1993-03-31 1994-10-13 Sanofi Oligonucleotides with amide linkages replacing phosphodiester linkages
DE4311944A1 (en) 1993-04-10 1994-10-13 Degussa Coated sodium percarbonate particles, process for their preparation and detergent, cleaning and bleaching compositions containing them
AU7966694A (en) 1993-07-21 1996-05-02 University Of Kentucky Research Foundation, The A multicompartment hard capsule with control release properties
US5571902A (en) 1993-07-29 1996-11-05 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
CA2170869C (en) 1993-09-03 1999-09-14 Phillip Dan Cook Amine-derivatized nucleosides and oligonucleosides
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5801154A (en) 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US5429807A (en) 1993-10-28 1995-07-04 Beckman Instruments, Inc. Method and apparatus for creating biopolymer arrays on a solid support surface
EP0736041B1 (en) * 1993-11-17 2006-02-08 Athena Neurosciences, Inc. Transparent liquid for encapsulated drug delivery
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
NZ278490A (en) 1993-12-09 1998-03-25 Univ Jefferson Chimeric polynucleotide with both ribo- and deoxyribonucleotides in one strand and deoxyribonucleotides in a second strand
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5641754A (en) * 1994-01-10 1997-06-24 The Board Of Regents Of The University Of Nebraska Antisense oligonucleotide compositions for selectively killing cancer cells
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5554746A (en) 1994-05-16 1996-09-10 Isis Pharmaceuticals, Inc. Lactam nucleic acids
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
JPH07330614A (en) 1994-06-02 1995-12-19 L T T Kenkyusho:Kk Fat emulsion for transduction of gene into cell
US5597696A (en) 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
GB9417524D0 (en) 1994-08-31 1994-10-19 Cortecs Ltd Pharmaceutical compositions
US5591721A (en) * 1994-10-25 1997-01-07 Hybridon, Inc. Method of down-regulating gene expression
US5545729A (en) 1994-12-22 1996-08-13 Hybridon, Inc. Stabilized ribozyme analogs
AU5527696A (en) 1995-03-31 1996-10-16 Children's Hospital Medical Center Use of surfactants for introducing genetic material into lun g cells
US5801155A (en) 1995-04-03 1998-09-01 Epoch Pharmaceuticals, Inc. Covalently linked oligonucleotide minor grove binder conjugates
FR2733500B1 (en) 1995-04-28 1997-07-25 Centre Nat Rech Scient NOVEL ANTISENS DIRECTED AGAINST RAS, PREPARATION AND USES
US5955059A (en) 1995-06-06 1999-09-21 Trustees Of Boston University Use of locally applied DNA fragments
HUP9900910A2 (en) 1995-06-07 1999-07-28 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
KR19990022184A (en) 1995-06-07 1999-03-25 스톤 스티븐 에프. Oral administration of gene constructs
GB9516268D0 (en) 1995-08-08 1995-10-11 Danbiosyst Uk Compositiion for enhanced uptake of polar drugs from the colon
US5652356A (en) 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
US5855911A (en) * 1995-08-29 1999-01-05 Board Of Regents, The University Of Texas System Liposomal phosphodiester, phosphorothioate, and P-ethoxy oligonucleotides
US5994062A (en) * 1995-10-02 1999-11-30 The United States Of America As Represented By The Department Of Health And Human Services Epithelial protein and DNA thereof for use in early cancer detection
DE69634696T2 (en) 1995-10-06 2006-01-19 PerSeptive Biosystems, Inc., Framingham METHOD AND TEST SYSTEM FOR HYBRIDIZATION ANALYSIS USING PEPTIDE NUCLEIC ACID PROBLEMS
EP0871489A1 (en) 1995-10-12 1998-10-21 Gs Development Ab A pharmaceutical composition for administration of an active substance to or through a skin or mucosal surface
US5846525A (en) * 1995-12-19 1998-12-08 Trega Biosciences, Inc. Protected biopolymers for oral administration and methods of using same
US5994316A (en) 1996-02-21 1999-11-30 The Immune Response Corporation Method of preparing polynucleotide-carrier complexes for delivery to cells
US5980861A (en) * 1996-03-12 1999-11-09 University Of Massachusetts Chelator compositions and methods of synthesis thereof
US5877162A (en) * 1996-03-14 1999-03-02 Innovir Laboratories, Inc. Short external guide sequences
WO1997038728A1 (en) 1996-04-15 1997-10-23 Dyad Pharmaceutical Corporation Combination therapy for androgenic alopecia with antisense oligonucleotides and minoxidil
GB9613858D0 (en) 1996-07-02 1996-09-04 Cortecs Ltd Hydrophobic preparations
AU727447B2 (en) 1996-07-03 2000-12-14 University Of Pittsburgh Emulsion formulations for hydrophilic active agents
GB9619894D0 (en) 1996-09-24 1996-11-06 Celltech Therapeutics Ltd Pharmaceutical products
KR19980067138A (en) 1997-01-31 1998-10-15 박원훈 Fat emulsions for efficiently delivering genes or biologically active drugs into cells and methods for preparing the same
US6127533A (en) 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
EP0979309A4 (en) 1997-04-30 2000-10-18 Isis Pharmaceuticals Inc Oligonucleotides for enhanced bioavailability
EP1012331B1 (en) * 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
WO1999007342A1 (en) 1997-08-11 1999-02-18 Alza Corporation Prolonged release active agent dosage form adapted for gastric retention
US5877309A (en) 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
ATE269710T1 (en) 1997-09-26 2004-07-15 Medeva Europ Ltd PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF INFLAMMATORY BOWEL DISEASE
US20030228597A1 (en) 1998-04-13 2003-12-11 Cowsert Lex M. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
JP2003524586A (en) 1998-05-21 2003-08-19 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for parenteral administration of oligonucleotides
CA2329252A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals Inc. Compositions and methods for topical delivery of oligonucleotides
US6228642B1 (en) * 1998-10-05 2001-05-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of tumor necrosis factor-(α) (TNF-α) expression
US6080580A (en) * 1998-10-05 2000-06-27 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of tumor necrosis factor-α (TNF-α) expression
US6133031A (en) * 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
US6008048A (en) * 1998-12-04 1999-12-28 Isis Pharmaceuticals Inc. Antisense inhibition of EGR-1 expression
US6159694A (en) * 1999-04-08 2000-12-12 Isis Pharmaceuticals Inc. Antisense modulation of stat3 expression
US6037176A (en) * 1999-06-25 2000-03-14 Isis Pharmaceuticals Inc. Antisense inhibition of integrin beta 3 expression
US6610674B1 (en) * 1999-09-28 2003-08-26 University Of Pennsylvania Method of treating inflammatory conditions with progesterone analogs

Also Published As

Publication number Publication date
AU8378698A (en) 1999-01-25
US6887906B1 (en) 2005-05-03
EP1012331A4 (en) 2002-06-26
ATE321882T1 (en) 2006-04-15
JP2002510319A (en) 2002-04-02
US20040229831A1 (en) 2004-11-18
WO1999001579A1 (en) 1999-01-14
US6747014B2 (en) 2004-06-08
EP1012331B1 (en) 2006-03-29
US20130274309A1 (en) 2013-10-17
US8691785B2 (en) 2014-04-08
EP1012331A1 (en) 2000-06-28
CA2294988A1 (en) 1999-01-14
US20030040497A1 (en) 2003-02-27
DE69834038D1 (en) 2006-05-18
AU731909B2 (en) 2001-04-05

Similar Documents

Publication Publication Date Title
CA2294988C (en) Compositions and methods for the delivery of oligonucleotides via the alimentary canal
AU745880B2 (en) Compositions and methods for non-parenteral delivery of oligonucleotides
AU2002329825B2 (en) Bioadhesive compositions and methods for enhanced mucosal drug absorption
IL160117A (en) Delayed release oral formulation for enhanced intestinal oligonucleotide absorption
WO2005020885A2 (en) Compositions and methods for the treatment of severe acute respiratory syndrome (sars)
US8946178B2 (en) Compositions and methods for treatment of pouchitis
US20030096770A1 (en) Enhancement of the stability of oligonucleotides comprising phosphorothioate linkages by addition of water-soluble antioxidants
CA2286396C (en) Oligonucleotides for enhanced bioavailability
AU755786B2 (en) Compositions and methods for the delivery of oligonucleotides via the alimentary canal
AU2002316662A1 (en) Enhancement of the stability of oligonucleotides comprising phosphorothioate linkages by addition of water-soluble antioxidants

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20180703