CA2329130A1 - Compositions and methods for non-parenteral delivery of oligonucleotides - Google Patents

Compositions and methods for non-parenteral delivery of oligonucleotides Download PDF

Info

Publication number
CA2329130A1
CA2329130A1 CA002329130A CA2329130A CA2329130A1 CA 2329130 A1 CA2329130 A1 CA 2329130A1 CA 002329130 A CA002329130 A CA 002329130A CA 2329130 A CA2329130 A CA 2329130A CA 2329130 A1 CA2329130 A1 CA 2329130A1
Authority
CA
Canada
Prior art keywords
isis
pharmaceutical composition
acid
oligonucleotide
animal
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002329130A
Other languages
French (fr)
Inventor
Ching-Leou Teng
Phillip D. Cook
Lloyd Tillman
Gregory E. Hardee
David J. Ecker
Muthiah Manoharan
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
Individual
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 Individual filed Critical Individual
Publication of CA2329130A1 publication Critical patent/CA2329130A1/en
Abandoned 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
    • A61K48/0008Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • 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
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic 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
    • A61K48/0008Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0041Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being polymeric
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0031Rectum, anus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification

Abstract

The present invention relates to compositions and methods which enhance the local and systemic uptake and delivery of oligonucleotides and nucleic acids via non-parenteral routes of administration. Pharmaceutical compositions comprising oligonucleotides disclosed herein include, for systemic delivery, emulsion and microemulsion formulations for a variety of applications and oral dosage formulations. It has also suprisingly been discovered that oligonucleotides may be locally delivered to colonic sites be rectal enemas and suppositories in simple solutions, e.g., neat or in saline. Such pharmaceutical compositions of olignonucleotides may further include one or more penetration enhancers for the transport of oligonucleotides and other nucleic acids across mucosal membranes. The compositions and methods of the invention are utilized to effect the oral, buccal, rectal or vaginal administration of an antisense oligonucleotide to an animal in order to modulate the expression of a gene in the animal for investigative, therapeutic, palliative or prophylactic purposes.

Description

WO 99160012 PC'f/US99111394 COMPOSITIONS AND METHODS FOR
NON-PARENTERAL DELIVERY OF OLIGONUCLEOTIDES
CROSS REFERENCE TO RELATED APPLICATIONS
This application is a cont:inuation-in-part of U.S.
patent application serial no. 09/082,624 filed on May 21, 1998, the disclosure of which is incorporated by reference herein in its entirety.
FIELD OF THE INVENTION
The present invention relates to compositions and methods which enhance the local and systemic uptake and delivery of nucleic acids via non-parenteral routes of administration. More particularly, the methods and compositions enhance the transport of oligonucleotides and other nucleic acids across mucosal membranes through the use of one or more penetration enhancer:a. The compositions of the present invention are solutions, emulsions, and related mixtures that facilitate the uptake and delivery of oligonucleotides and other nucleic acids. 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 cell walls and/or into cells. 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.

WO 99!60012 PCT/US99/11394 BACKGROUND OF THE INVENTION
Advances in the field of biotechnology have Ied to significant advances in the treatment of diseases such as cancer, genetic diseases, arthritis and AIDS that were previously difficult to treat. Many such advances involve the administration of oligonucleotic~es and other nucleic acids to 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., Draper et al., U.S. Patent No.
5,595,978, January 21, 1997, 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, I5, 209, and Genetic Engineering News, 1997, 15, 1, each of which discuss the treatment of Crohn~s disease via intravenous infusions of antisense oligonucleoti.des. Non-parenteral routes for administration of oligonucleotides and other nucleic acids (such as oral or rectal delivery or other mucosal routes) 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 is a need to provide compositions and methods to enhance the availability of novel drugs such as oligonucleotides when administered via non-parenteral routes. It is desirable that such new compositions and methods provide for the simple, convenient, practical and optimal non-parenteral delivery of oligonucleotides and other nucleic acids.
SUI~lARY OF THE INVENTION
In accordance with the present invention, compositions and methods are provided for the non-parenteral delivery and mucosal penetration of nucleic acids in an animal. In particular, the present invention provides 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, via non-parenteral means to an animal, thereby circumventing the complications and expense which may be associated with intravenous and other parenteral modes of in vivo administration. "Non-parenteral administration" refers to the contacting, directly or otherwise, to all or a portion of the alimentary canal, skin, eyes, pulmonary tract, urethra or vagina of an animal. Compositions of the present invention may be a mixture of components or phases as are present in emulsions (including microemulsions and creams), and related formulations comprising two or more phases.
In one aspect, the present invention provides pharmaceutical compositions comprising at least one nucleosidic moiety such as a nucleoside, nucleotide, or nucleic acid in a solution or emulsion. The nucleic acid can be a ribozyme, a PNA, or an aptamer, but preferably is an oligonucleotide such as, for example, an oligonucleotide that modulates expression of a cellular adhesion protein, modulates a rate of cellular proliferation, or has biological activity against eukaryotic pathogens or retroviruses.
In certain embodiments, solutions according to the invention consist essentially of the nucleosidic moiety and a solvent comprising, for example, saline solution or cocoa butter. Emulsions according to the invention include oil-in-water emulsions, water-in-oil emulsions, oil-in-water-in-oil emulsions, and water-in-oil-in-water emulsions.
In certain embodiments, th.e pharmaceutical compositions of the invention further comprise a penetration enhancer such as a fatty acid, a bile salt, a chelating agent, a surfactant, and a non-surfactant such as an unsaturated cyclic urea, a 1-alkyl-alkanone, a 1-WO 99/b0012 PCTIUS99/11394 alkenylazacyclo-alakanone, or a stei-oidal anti-inflammatory agent.
Also provided are methods for treating an animal comprising administering to t:he animal a therapeutically effective amount of a pharmaceutical composition according to the invention. The composition can be administered by, for example, buccal, sublingual, endoscopic, rectal, oral, vaginal, topical, pulmonary, or urethral routes. In preferred embodiments, the compositions of the invention are administered re:ctally means of an enema or a suppository.
Because of the advantages of non-parenteral delivery of drugs. of the antisense class, the compositions and methods of the invention can be used in therapeutic methods as explained in more detail herein. 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.
The methods of the invention can be used, for example, for the treatments of animals that are known or suspected to suffer from diseases such as ulcerative colitis, Chrohn's disease, inflammatory bowel disease, or undue cellular proliferation.
DETAILED DESCRIPTION OF THE INVENTION
The invention provides compositions and methods for the local as well as systemic delivery of oligonucleotides and other nucleic acids to an animal via non-parenteral means. In particular, the present invention provides compositions and methods for modulating the in vivo expression of a gene in an animal through the non-parenteral administration of an antisense oligonucleotide, thereby circumventing the complications and expense which may be associated with intravenous and other parenteral routes of administration.
Enhanced bioavailability of oligonucleotides and other nucleic acids is achieved via the non-parenteral administration of the compositions and methods of the present 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 al., Gastroenterol., 1977, 73, 300). Traditionally, l0 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 C;o., Easton, PA, 1990, pages 1451-1458). The area under the curve (AUCo) is divided by the area under the curve after ara intravenous (i.v.) dose (AUCi~) 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 i.s 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 al., Gastroenterol., 1977, 73, 300). With regards to oligonucleotides, studies suggest that they 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; T'akakura et al., Antisense & Nucl. Acid Drug Dev., 1996, 6, 177).
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, 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, pulmonary, buccal, rectal, transdermal, vaginal and ocular) to the bloodstream is frequently obstructed by poor transport across epithelial cells and concurrent metabolism during transport. This type of degradative metabolism is known for oligonucleotides and nucleic acids. For example, phosphodiesterases are known to cleave the phosphodiester linkages of oligonucleotides and many other modified linkages present in synthetic oligonucleotides and nucleic acids.
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 non-parenteral means. For example, a drug may be enzymatically or chemically degraded in the alimentary canal or blood stream and/or may be impermeable or semipermeable to various mucosal membranes.
It has now been found that oligonucleotides can be introduced effectively into animals via non-parenteral means 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 mucous membranes) associated with the desired mode of administration.
A "pharmaceutically acceptable" component of a formulation of the invention is one which, when used together with excipients, diluents, stabilizers, preservatives and other ingredients are appropriate to the nature, composition and mode of administration of a formulation. Accordingly it is desired to select penetration enhancers which facilitate the uptake of oligonucleotides, without interfering with the activity of the oligonucleotides and in a 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.
The present invention provides compositions comprising one or more pharmaceutically acceptable penetration enhancers, and methods of using such compositions, which result in the improved bioavailability of nucleic acids administered via non-parenteral 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 a~., 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. Therefore, it has been surprisingly found that the uptake and delivery of oligonucleotides, relatively complex molecules which are known to be difficult to administer to animals and man, can be greatly improved even when administered by non-parenteral means through the use of a number of different classes of penetration enhancers.
The effective non-parenteral use and administration of compositions of the present invention involves consideration of a number of different aspects about drug therapy. One important consideration when using the compositions and methods of the present invention is the mode of administration of the pharmaceutical composition containing the therapeutic oligonucl.eotide or other nucleic acid. Administration typically is either parenteral or non-parenteral. Non-parenteral modes of administration include, but are not limited to, buccal, sublingual, endoscopic, oral, rectal, transdermal, topical, nasal, intratracheal, pulmonary, urethral, vaginal, and ocular. When administered by such non-parenteral modes the methods and composition of the present invention can deliver drug both locally and systemically as desired.

_ g _ A second consideration of importance when using the compositions and methods of the present invention is the use and nature of penetration enhan.cers and carriers.
Penetration enhancers facilitate th.e transport of drug molecues, for example, oligonucleotides and other nucleic acids, across mucosal and other epithelial cell membranes.
Penetration enhancers include, but are not limited to, members of molecular classes such as surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactant molecules. Carriers are inert molecules that may be included in the compositions of the present invention to interfere with processes that lead 'to reduction in the levels of bioavailable nucleic acid or oligonucleotide drug.
A third consideration of :importance to the compositions and methods of the present invention is the nature of oligonucleotide or other nucleic acid used.
Oligonucleotides of the present invE.ntion may be, but are not limited to, those nucleic acids bearing modified linkages, modified nucleobases, or modified sugars, and chimeric nucleic acids.
A fourth consideration of importance in the present invention is the nature of t:he composition.
Pharmaceutical compositions of the present invention include, but are not limited to, so7.utions, emulsions (including microemulsions and creama), and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids. Tree compositions of the present invention may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, liquid syrups, soft gels, suppositories, and enemas.
A fifth consideration of importance to the compositions and methods of the present invention is the means by which such compositions may be administered. Thus the dose, method of administration or application, and the _ g _ use of additives are all worthy of consideration in this regard. Further, the methods and compositions of the present invention may be used to ameliorate a variety of diseases via local or systemic treatment. Such local or systemic treatment may be accomplished using the methods and compositions of the present invention via modes of administration that include, but are not limited to, buccal, sublingual, endoscopic, oral, rectal, transdermal, topical, nasal, pulmonary, urethral, vaginal, and ocular modes.
A sixth consideration of importance to the compositions and methods of the present invention is their applicability to bioequivalents of oligonucleotides and other nucleic acids such as, but not limited to, oligonucleotide prodrugs, deletion .derivatives, conjugates, aptamers, and ribozymes.
The present invention provides compositions and methods for local and systemic delivery of one or more nucleic acids to an animal via non-parenteral administration. For purposes of the invention, the term "animal" is meant to encompass humans as well as other mammals, as well as reptiles, fish, amphibians, and birds.
The term "non-parenteral delivery" refers to the administration, directly or otherwise, of the drug via a non-invasive procedure which typica7_ly does not entail the use of a syringe and needle. Non-parenteral administration may be, but is not limited to, delii~ery of the drug via the alimentary canal or via transdermal, topical, nasal, pulmonary, urethral, vaginal or ocular routes. 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 a.ll 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 WO 99/b0012 PCT/1JS99/11394 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 acids) 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 a1. , Cri ti cal Revi ews in Therapeu ti c Drug Carri er Systems, 1991, p. 166), and optimizai~ion of vehicle characteristics relative to dose deposition and retention at the site of administration (Lee et a1. , Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 168) may be useful methods for enhancing the transport of drugs across mucosal sites in accordance with the' present invention.
Delivery of a drug via the oral mucosa, as in the case of buccal and sublingual administration, 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). 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 drugs like 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).

Endoscopy may be used for drug delivery directly to an interior portion of the alimentary tract. For example, endoscopic retrograde cystopancreatography (FRCP) takes advantage of extended gastroscopy and permits selective access to the biliary tract and the pancreatic duct (Hirahata et al . , Gan To Kagaku Ryoho, 1992 , 19 (10 Suppl.), 1591). Pharmaceutical compositions, including liposomal formulations, can be delivered directly into portions of the alimentary canal, SLlch as, e.g., the duodenum (Somogyi et al., Pharm. Re:~., 1995, 12, 149) or the gastric submucosa (Akamo et al., Japanese J. Cancer Res., 1994, 85, 652) via endoscopic means. Gastric lavage devices ( moue et al . , Artif. Organs, 1997, 21, 28 ) and percutaneous endoscopic feeding devices (Penningt.on et al., Ailment Pharmacol. Ther. , 1995, 9, 471) can also be used for direct alimentary delivery of pharmaceutical compositions.
Drugs administered by the oral route can often be alternatively administered by the lower enteral route, i.e., through the anus 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 can result in more prompt and higher blood levels than the oral route.
(Harvey, Chapter 35 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, page 711). Because about 50% of the drug that is absorbed from the rectum will bypass the liver, administration 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 al., eds., McGraw-Hill, New York, NY, 1996).

The preferred method of non-parenteral administration for most drugs is oral delivery. This 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 e~t al., 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 al., 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 by, 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.
Topical administration is often chosen when local delivery of a drug is desired at, or immediately adjacent to the point of application of the drug composition or formulation. Although occasionally enough drug is absorbed into the systemic circulation to cause systemic effects, topical routes generally are not effective for systemic therapy. Three general types of topical routes of administration are recognized, topical administration of a drug composition to mucous membranes, skin or eyes.
Drugs that are applied to the mucous membranes produce primarily local effects. This route of administration includes application of drug compositions to mucous membranes of the conjunctiva, nasopharynx, oropharynx, vagina, colon, urethra, and urinary bladder.

Absorption of drugs occurs rapidly through mucous membranes and is an effective route for localized therapy and, on occasion, for systemic therapy.
Transdermal drug delivery is a valuable route for the administration of lipid soluble therapeutics. It has been recognized that the dermis is more permeable than the epidermis and therfore absorption of drugs is much more rapid through abraded, burned or denuded skin. Inflammation and other physiologic conditions that increase blood flow to the skin also enhance absorption via the transdermal route.
Absorption by this route may be enhanced via the use of an oily vehicle (inunction) or through the use of penetration enhancers. Hydration of the skin and the use of controlled release topical patches are also effective ways to administer drugs via the transdermal route. This route provides a means to deliver the drug for both systemic and local therapy.
Ocular delivery of drugs is especially useful for the local treatment of eye infections or abnormalities. The drug is typically administered via instillation and absorption of the drug occurs through the cornea. Corneal infection or trauma may thus result in more rapid absorption. Opthalmic delivery systems that provide prolonged duration of action (e.g., suspensions and ointments) and ocular inserts that provide continuous delivery of low amounts of drugs are useful additions to ophthalmic therapy. The ocular delivery of drugs results in predominantly local effects. Systemic absorption that reults from drainage via the nasolachrimal canal is limited and few systemic side effects are typically observed.
The present invention employs various penetration enhancers in order to effect transport of oligonucleotides and other nucleic acids across mucosal and epithelial membranes. Penetration enhancers may be classified as belonging to one of five broad categories - surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et al., Critica.I Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of these classes is discussed in more detail in the following paragraphs.
Carrier substances (or simply "carriers"), which reduce first pass effects by, e.g., saturating the hepatic uptake system, are also herein described.
In connection with the present invention, surfactants (or "surface-active agents") are chemical entities which, when dissolved in a.n 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 and other epithelial membranes is enhanced. In addition to bile salts and fatty acids, surfactants 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) .
Fatty acids and their derivatives which act as penetration enhancers and may be used in compositions of the present invention include, for example, oleic acid, lauric acid, capric acid (n-decanoic acid),, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein (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, myx-istate, 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; E1-Hariri et a1 . , J. Pharm. Praarmacol . , 1992, 44, 3S 651) .

A variety of bile salts also function as penetration enhancers to facilitate the uptake and bioavailability of drugs. 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 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 (CDCA, sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25-dihydro-fusidate (STDHF), sodium glycodihydrofusidate and polyoxyeth:ylene-9-lauryl ether (POE) (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;
Murani shi , Cri ti caI Revi ews in Therapeu ti c Drug Carri er Systems, 1990, 7, 1; Yamamoto et al., J. Pharm. Exp. Ther., 1992, 263, 25; Yamashita et al., J. Pharm. Sci., 1990, 79, 579) .
In a particular embodiment., penetration enhancers useful in the present invention are mixtures of penetration enhancing compounds. For example, a particularly preferred penetration enhancer is a mixture of. UDCA (and/or CDCA) with capric and/or lauric acids or salts thereof e.g. sodium.
Such mixtures are useful for enhancing the delivery of biologically active substances across mucosal membranes, in particular intestinal mucosa. Preferred penetration enhancer mixtures comprise about 5-95°s of bile acid or salts) UDCA and/or CDCA with 5-95% capric and/or lauric acid. Particularly preferred are mixtures of the sodium salts of UDCA, capric acid and laur:ic acid in a .ratio of about 1:2:2 respectively. In another particularly preferred embodiment 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 and other 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 recc~uuire a divalent metal ion for catalysis and are thus inhibited by chelating agents (Jarrett, J. Chromatogr., 1993, 6I8, 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 a1. , Cri ti cal Revi ews in Therapeu ti c Drug Carri er Sys terns , 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, l; Buur et a1. , J. Control Rel. , 1990, I4, 43) .
As used herein, non-chelating non-surfactant penetration enhancers may be defined as compounds that demonstrate insignificant activity a,s chelating agents or as surfactants but that nonetheless enhance absorption of oligonucelotides through the alimentary and other mucosal membranes (Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1). This class of penetration enhancers includes, but is not limited to, 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 al., ~T. Pharm.
Pharmacol., 1987, 39, 621).
Agents that enhance uptake of oligonucleotides at the cellular level may also be added to the pharmaceutical and other compositions of the present invention. For example, cationic lipids, such as l:ipofectin (Junichi et al, U.S. Patent No. 5,705,188), cationic glycerol derivatives, and polycationic molecules, such as polylysine (Lollo et al., PCT Application WO 97/30731), can be used.
Certain compositions of the present invention also incorporate carrier compounds in the' formulation. As used herein, "carrier compound" or "carrier" can refer 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 phosphorothioate oligonucleotide in hepatic tissue can be 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 al., Ant:isense & Nucl. Acid Drug Dev. , 1996, 6, 177) .
In contrast to a carrier compound, a "pharmaceutical 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 vec3etable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc.); disintegrants (e. g., starch, sodium starch glycolate, EXPLOTAB); and 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, for example, 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 present invention.
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 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 modified 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 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 ~>' 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 100°s complementary to its target DNA sequence to be specifically hyb:ridizable. An oligonucleotide is specifically hyb:ridizable when binding of the oligonucleotide to the target DNA or RNA 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 complementaritw 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 oligonucleotide~s are commonly used as research reagents, diagnostic aids, and therapeutic agents.
For example, antisense oligonucleoti.des, 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. Antisense oligonucleotides have also been used as diagnostic aids based on their specific binding or hybridization to DNA or mRNA that are present in certain disease states and due to the high degree of sensitivity that hybridization based assays and amplified assays that utilize some of polymerase chain reaction afford. 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 a.ntisense 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 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 mR.NA 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 Esxamples of antisense oligonucleotides are provided herein.
Further, oligonucleotides used in the compositions of the present invention may be directed to modify the effects of mRNAs or DNAs involved in the synthesis of proteins that regulate adhesion of white blood cells and to other cell types. The adherence of white blood cells to vascular endothelium appears to be mediated in part if not in toto by five cell adhesion molecules ICAM-1, ICAM-2, ELAM-1, VCAM-1 and GMP-140. Dustin and Springer, J. Cell.
Biol. 1987, 107, 321. Such antisense oligonucleotides are designed to hybridize either directly to the mRNA or to a selected DNA portion encoding intercellular adhesion molecule-1 (ICAM-1), endothelial leukocyte adhesion molecule-1 (ELAM-1, or E-selectin), and vascular cell adhesion molecule-1 (VCAM-1) as disclosed in U.S. Patents 5,514,788 (Bennett et al., May 7, 1996) and 5,591,623 (Bennett et al., January 7, 1997), and pending U.S. patent applications Serial Nos. 08/440,740 (filed May 12, 1995) and 09/062,416 (filed April 17, 1998). These oligonucleotides have been found to modulate the activity of the targeted mRNA or DNA, leading to the modulation of the synthesis and metabolism of specific cell adhesion molecules, and thereby result in palliative and therapeutic effects. Inhibition of ICAM-1, VCAM-1 and SLAM-1 expression is expected to be useful for the treatment of inflammatory diseases, diseases with an inflammatory component, allograft rejection, psoriasis and other skin diseases, inflammatory bowel disease, cancers and their metastases, and viral infection.
Methods of modulating cell adhesion comprising contacting the animal with an oligonucleotide composition of the present invention are provided.
The antisense compounds used as exemplary biologically active nucleic acids in the studies detailed herein are as follows:
ISIS 2302 is a 2'-deoxyoligonucleotide having a phosphorothioate backbone and the sequence 5'-GCC-CAA-GCT-GGC-ATC-CGT-CA-3' (SEQ ID N0:1). ISIS 2302 is targeted to the 3'-untranslated region (3'-UTR) of the human ICAM-1 gene. ISIS 2302 is described in U.S. Patents 5,514,788 and 5,591,623, hereby incorporated by reference.

- a4 -ISIS 15839 is a phosphorothioate isosequence "hemimer" derivative of ISIS 2302 h;wing the structure 5'-GCC-CAA-GCT-GGC-ATC-CGT-CA-3' (SEQ :ID NO:1), wherein emboldened "C" residues have 5-methylcytosine (m5c) bases and wherein the emboldened, double-underlined residues further comprise a 2'-methoxyethoxy modification (other residues are 2'-deoxy). ISIS 15839 is described in co-pending U.S. Patent application Serial No. 09/062,416, filed April 17, 1998, hereby incorporated by reference.
ISIS 1939 is a 2'-deoxyoligonucleotide having a phosphorothioate backbone and the sequence 5'-CCC-CCA-CCA-CTT-CCC-CTC-TC-3' (SEQ ID N0:2). ISIS 1939 is targeted to the 3'-untranslated region (3'-UTR) of the human ICAM-1 gene. ISIS 1939 is described in U.~~. Patents 5,514,788 and 5,591,623, hereby incorporated by reference.
Examination of the predicted RNA secondary structure of the human ICAM-1 mRNA 3'-untranslated region (M. Zuker, Science 1989, 244, 48) surprisingly suggested that both ISIS 1939 and ISIS 2302 hybridize to sequences predicted to be in a stable stem-loop structure of the mRNA.
Current dogma suggests that when designing antisense oligonucleotides regions of RNA secondary structure should be avoided. Thus, ISIS 1939 and ISIS 2302 would not have been predicted to inhibit ICAM-1 expression.
ISIS 2302 (SEQ ID N0: 1) has been found to inhibit ICAM-1 expression in human umbilical vein cells, human lung carcinoma cells (A549), human epidermal carcinoma cells (A431), and human keratinocytes. ISIS 2302 has also demonstrated specificity for its target ICAM-1 over other potential nucleic acid targets such as HLA-A and HLA-B.
ISIS 1939 (SEQ ID N0:2) and ISIS 2302 markedly reduced ICAM-1 expression, as detected by northern blot analysis to determine mRNA levels, in C8161 human melanoma cells. In an experimental metastasis assay, ISIS 2302 decreased the metastatic potential of C8161 cells, and eliminated the enhanced metastatic ability of C8161 cells resulting from TNF-a treatment. ISIS 2302 has also shown significant biological activity in animal models of inflammatory disease. The data from animal testing has revealed strong anti-inflammatory effects of ISIS 2302 in a number of inflammatory diseases including Crohn's disease, rheumatoid arthritis, psoriasis, ulcerative colitis, and kidney transplant rejection. When tested on humans, ISIS 2302 has shown good safety and activity against Crohn~s disease.
Further ISIS 2302 has demonstrated a statistically significant steroid-sparing effect on treated subjects such that even after five months post-treatment subjects have remained weaned from steroids and i.n disease remission.
This is a surprising and significant finding of TSIS 2302's effects.
The oligonucleotides used in the compositions of the present invention preferably comprise from about 8 to about 30 nucleotides. It is more preferred that such oligonucleotides comprise from about 10 to about 25 nucleotides.
Antisense oligonucleotides employed in the compositions of the present invention may also be used to determine the nature, function and potential relationship of various genetic components of the body to normal or abnormal body states of animals. 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 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, Albeit et al., Trends in Pharmacological Sciences, 1994, I5, 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 non-parenteral delivery of oligonucleot:ides and other nucleic acids, the present invention overcomes these and other shortcomings.
Specific examples of some preferred modified oligonucleotides envisioned for use in the compositions of the present invention include oligonucleotides containing modified backbones or non-natural intersugar linkages. As defined in this specification, oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that have an atom (or group of atoms) other than a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their intersugar backbone, including peptide nucleic acids (PNAs) are also be considered to be oligonucleotides.
Specific oligonucleotide chemical modifications are described in the following subsections. It is not necessary for all positions in a given compound to be uniformly modified, and in fact more than one of the following modifications may be incorporated in a single antisense compound or even in a single residue thereof, for example, at a single nucleoside within an oligonucleotide.
A. Modified Liakagess Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphoro-dithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thiono-alkylphosphonates, thionoalklyphosphotriesters, and borano-phosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'. Various salts, mixed salts and free acid forms are also included.
Representative United States Patents that teach the preparation of the above phospharus atom containing linkages include, but are not limited to, U.S. Patents Nos.
3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196;
5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717;
5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233;
5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306;
5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,625,050; and 5,697,248, certain of which are commonly owned with this application, and each of which is herein incorporated by reference.
Preferred modified oligonucleotide backbones that do not include a phosphorus atom therein (i.e., oligonucleosides) have backbones that are formed by short chain alkyl or cycloalkyl intersugar linkages, mixed heteroatom and alkyl or cycloalkyl intersugar linkages, or one or more short chain heteroatomic or heterocyclic intersugar linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones;
methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, 0, S
and CHZ component parts.
Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Patents Nos. 5,034,506; 5,166,315;
5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562;

WO 991b0012 PCTIUS99/11394 5,264,564; 5,405,938; 5,434,257; 5,466,677 ; 5,470,967;
5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240;
5,610,289; 5,602,240; 5,608,046; 5,610,289 ;5,618,704;
5,623,070; 5,663,312; 5,633,360; 5,E~77,437; and 5,677,439, certain of which are commonly owned with this application, and each of which is herein incorporated by reference.
In other preferred oligonucleotide mimetics, both the sugar and the intersugar linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA
compounds, the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Patents Nos.
5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA
compounds can be found in Nielsen et al., Scierzce, 1991, 254, 1497.
Some preferred embodiments of the present invention may employ oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CHZ-NH-0-CHZ-, -CHZ-N(CH3) -0-CHZ- [known as a methylene (methylimino) or MMI backbone], -CHZ-O-N(CH3)-CH2-, - CHz-N(CH3) -N(CH3) -CHZ- and -0-N(CH3) -CHZ-CHZ- [wherein the native phosphodiester backbone is represented as -O-P-O-CHz-] of the above referenced U.S. Pa.tent 5,489,677, and the amide backbones of the above referenced U.S. Patent No.
5,602,240. Also preferred are oligonucleotides having morpholino backbone structures of then above-referenced U.S.
Patent No. 5,034,506.

H. Modified Nucleobasess The oligonucleotides employed in the compositions of the present invention may additionally or alternatively comprise nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uraci:L (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-met.hyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in United States Patent No.
3,687,808, those disclosed in the Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J.I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y.S., Chapter 15, Antisense Research and Applications, pages 289-302, Crooke, S.T. and Lebleu, B., ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C
(Id., pages 276-278) and are presently preferred base substitutions, even more particularly when combined with 2~-methoxyethyl sugar modifications.
Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Patent 3,687,808, as well as U.S. Patents 4,845,205; 5,130,302;
ZO 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187;
5,459,255; 5,484,908; 5,502,177; 5,~i2S,711; 5,552,540;
5,587,469; 5,594,121, 5,596,091; 5,E>14,617; and 5,681,941, certain of which are commonly owned, and each of which is herein incorporated by reference, and commonly owned United States patent application 08/762,488, filed on December 10, 1996, also herein incorporated by reference.
C. Sugar Modificationss The oligonucleotides employed in the compositions of the present invention may additionally or alternatively comprise one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following at the 2' position: OH; F; 0-, S-, or N-alkyl, 0-, S-, or N-alkenyl, or 0, S- or N-alkynyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to Clo alkyl or CZ to Clo alkenyl and alkynyl.
Particularly preferred are 0 [ (CHZ) n0] mCH3, 0 (CHZ) nOCH3, O ( CHZ ) nNH2, O ( CHZ ) nCH3, O ( CH2 ) nONH2, and 0 ( CHZ ) ~ON [ ( CHZ ) ,;CHj ) ] .,, where n and m are from 1 to about 10. Other preferred' oligonucleotides comprise one of the following at the 2~
position: C, to Clo lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or 0-ara:lkyl, SH, SCH,, OCN, C1, Br, CN, CF3, OCF3, SOCH3, SOZCH3, ONOz, 1V02, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, poly-alkylamino, 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' -O-CHZCHZOCH3, also known as 2' -O- (2-methoxyethyl) or 2'-MOE] (Martin et al., Helv. Chim. Acta, 1995, 78, 486), i.e., an alkoxyalkoxy group. A further preferred modification includes 2'-dimethylaminooxyethoxy, i . a . , a O (CHZ) 20N (CH3) 2 group, also known as 2' -DMAOE, as described in co-owned United States patent application Serial Number 09/016,520, filed on January 30, 1998, the contents of which are herein incorporated by reference.
Other preferred modifications include 2'-methoxy ( 2 ' -0-CH3 ) , 2 ' -aminopropoxy ( 2 ' -OCHzCH2CH2NH2 ) and 2 ' - f luoro (2'-F). Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugars structures include, but are not limited to, U.S. Patents Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878;
5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811;
5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,0531 5,639,873; 5,646,265; 5,658,873; 5,6'70,633; and 5,700,920, certain of which are commonly owned, and each of which is herein incorporated by reference, and commonly owned United States patent application 08/468,037, filed on June 5, 1995, also herein incorporated by reference.
D. Other Modifications: Additional 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 5' terminal nucleotide. For example, one additional modification of the oligonucleotides employed in the compositions of the present invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci.
USA, 1989, 86, 6553), cholic acid (Manoharan et al., Bioorg.
Med. Chem. Lett., 1994, 4, 1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 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, 10, 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 t:riethylammonium 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, I8, 3777), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, I4, 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 ) .
Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patents Nos. 4,828,979;
4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730;
5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584;
5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603;
5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735;
4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263;
4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963;
5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022;

5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873;
5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463;
5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810;
5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696;
5,599,923; 5,599,928 and 5,688,941, certain of which are commonly owned, and each of which is herein incorporated by reference.
A preferred conjugate imparting improved absorption of oligonucleotides in the gut is folic acid.
Accordingly, there is provided a composition for oral administration comprising an oligonucleotide and a carrier wherein said oligonucleotide is conjugated to folic acid.
Folic acid (folate) may be conjugated to the 3' or 5' termini of oligonucleotides, to a nucleobase or to a 2' position of any of the sugar residues in the chain.
Conjugation may be via any suitable chemical linker utilitizing functional groups on the oligonucleotide and folate. Oligonucleotide-folate conjugates and methods in preparing are described in copending United States patent applications 09/098,166 (filed June 16, 1998) and 09/275,505 (filed March 24, 1999) both incorporated herein by reference.
E. Chimeric Oligoaucleotidess The present invention also includes compositions employing antisense compounds which are chimeric compounds. ~~Chimeric'~
antisense compounds or ~~chimeras,~~ i.n the context of this invention, are antisense compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound. 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 oligonucleotide inhibition of gene expression.
Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate oligodeoxynucleotides hybridizing to the same target region. Cleavage of the RNA
target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art. RNase H-mediated target cleavage is distinct from the use of ribozymes to cleave nucleic acids.
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, or stability when duplexed with, the target RNA molecule but are unable to support nuclease activity (e. g., 2'-fluoro- or 2'-methoxyethoxy- substituted). Other chimeras include "hemimers," 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, or stability when duplexed with, the target RNA
molecule but is unable to support nuclease activity (e. g., 2'-fluoro- or 2'-methoxyethoxy- substituted), or vice-versa.
A number of chemical modi:Eications to oligonucleotides that confer greater oligonucleotide:RNA
duplex stability have been described by Freier et a1. (Nucl.
Acids Res., 1997, 25, 4429). Such modifications are preferred for the RNase H-refractory portions of chimeric oligonucleotides and may generally be used to enhance the affinity of an antisense compound for a target RNA.

WO 99/60012 PCTlUS99/11394 Chimeric antisense compounds of the invention may be formed as composite structures of t:wo or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleatide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Patents Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878;
5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355;
5,652,356; and 5,700,922, certain of which are commonly owned, and each of which is herein incorporated by reference, and commonly owned and allowed United States patent application serial number 08/465,880, filed on June 6, 1995, also herein incorporated by reference.
The present invention also includes compositions employing oligonucleotides that are substantially chirally pure with regard to particular 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).
The present invention further encompasses compositions employing 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 Auqust 13, 1996.) The cleavaaP
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 a1 . , CeI3, 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 a.ntisense 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.
Other biologically active oligonucleotides may be formulated in the compositions of the invention and used for therapeutic, palliative or prophylactic purposes according to the methods of the invention. Such other biologically active oligonucleotides include, but are not limited to, antisense compounds including, inter alia, antisense oligonucleotides, antisense PNAs and ribozymes (described supra) and EGSs, as well as aptamers and molecular decoys (described infra).
Sequences that recruit RNase P are known as External Guide Sequences, hence the abbreviai~ion ~~EGS. ~~ EGSs are antisense compounds that direct of <~n endogenous nuclease (RNase P) to a targeted nucleic acid (Forster et al., Science, 1990, 249, ?83; Guerrier-Takada et al., Proc. Natl.
Acad. Sci . USA, 1997, 94, 8468 ) .
Antisense compounds may alternatively or additionally comprise a synthetic moiety having nuclease activity covalently linked to an oligonucleot:ide having an antisense sequence instead of relying upon recruitment of an endogenous nuclease. Synthetic moieties having nuclease activity include, but are not limited to, enzymatic RNAs (as in ribozymes), lanthanide ion comlexes, and the like (Haseloff et al., Nature, 1988, 334, 585; Baker et al., J.
Am. Chem. Soc. , 1997, 119, 8749) .
Aptamers are single-stranded oligonucleotides that bind specific ligands via a mechanism other than Watson-Crick base pairing. Aptamers are typically targeted to, e.g., a protein and are not designed to bind to a nucleic acid (Ellington et a1. , Nature, 1990, 346, 818 ) .
Molecular decoys are short double-stranded nucleic acids (including single-stranded nucleic acids designed to ~~fold back~~ on themselves) that mimic a site on a nucleic acid to which a factor, such as a protein, binds. Such decoys are expected to competitively inhibit the factor;
that is, because the factor molecules are bound to an excess of the decoy, the concentration of factor bound to the cellular site corresponding to the decoy decreases, with resulting therapeutic, palliative or prophylactic effects.
Methods of identifying and constructing nucleic acid decoy molecules are described in, e.g., U.S. Patent 5,716,780 to Edwards et al.
Another type of bioactive oligonucleotide is an RNA-DNA hybrid molecule that can direct gene conversion of an endogenous nucleic acid (Cole-Strauss et al., Science, 1996, 273, 1386) .
Examples of specific oligonucleotides and the target genes to which they inhibit, which may be employed in formulations of the present invention include:
ISIS-2302 GCCCA AGCTG GCATC CGTCA (SEQ ID N0:1) ICAM-1 ISIS-15839 GCCCA AGCTG GCATC CGTCA (SEQ ID N0:1) ICAM-1 ISIS-1939 CCCCC ACCAC TTCCC CTCTC (SEQ ID N0:2) ICAM-1 ISIS-2922 GCGTT TGCTC TTCTT CTTGC (SEQ ID N0:48) HCMV
G

ISIS-13312 GCGTT TGCTC TTCTT CTTGG SEQ ID 0:48) CMV
( G H
N

ISIS-3521 GTTCT CGCTG GTGAG TTTCA (SEQ ID N0:49) PKCa ISIS-9605 GTTCT CGCTG GTGAG TTTCA SEQ ID N0:49) KCa ( P

ISIS-9606 GTTCT CGCTG GTGAG TTTCA SEQ ID N0:49) KCa ( P

ISIS-14859 AACTT GTGCT TGCTC N (SEQ ID 0:50) PKCa WO 99/b0012 PCT/US99111394 ISIS-2503 TCCGT CATCG CTCCT CAGGG (SEQ ID N0:16) Ha-ras ISIS-5132 TCCCG CCTGT GACAT GCATT (SEQ ID N0:19) c-raf ISIS-14803 GTGCT _CATGGTGCAC GGTCT (SEQ ID 0:51) CV
N H

ISIS-28089 GTGTG CCAGA CACCC TATCT (SEQ ID 0:52) T'NFa _ N

ISIS-104838 GCTGA TTAGA GAGAG GTCCC (SEQ ID N0:53) TNFa ISIS-2105 TTGCT TCCAT CTTCC TCGTC (SEQ ID N0:54) HPV
wherein (i) each oligo backbone linkage is a phosphorothioate linkage (except ISIS-9605) and (ii) each sugar is 2~-deoxy unless represented in bold font in which case it incorporates a 2~-0-methoxyethyl group and iii) underlined cytosine nucleosides incorporate a 5-methyl substituent on their nucleobase. ISIS-9605 incorporates natural phosphodiester bonds at the' first five and last five linkages with the remainder being phosphorothioate linkages.
F. Synthesise The oligonucleotides used in the compositions of the present 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.
1. Teachings regarding the synthesis of particular modified oligonucleotide:s may 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. F~atent 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 o~ligonucleotides having modified backbones; 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 ~i-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 No. 5,506,351,. drawn to processes for the preparation of 2'-O-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. Patents Nos.
5,223,168, issued June 29, 1993, and 5,608,046, both 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 application Serial No. 08/383,666, filed February 3, 1995, and U.S. Patent No.
5,459,255, drawn to, inter alia, methods of synthesizing 2'-fluoro-oligonucleotides.
2. Bioequivalentss The compositions of the present invention encompass any pharmaceutically acceptable 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 "prodrugs" and 'pharmaceutically acceptable salts" of the antisense compounds of the invention and other bioequivalents.

A. Oligoaucleotide Prodrugs: The oligonucleotide and nucleic acid compounds employed in the compositions of the present 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 versions of the antisense compounds may be prepared as SATE [(S-acetyl-2-thioethyl) phosphate] derivatives according to the methods disclosed in WO 93/24510 (Gosselin et al., published December 9, 1993).
B. Pharmaceutically Acceptable Salts: The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the oligonucleotide and nucleic acid compounds employed in the compositions of the present 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, ammonium, polyamines such as spermine and spermidine, and the like. Examples of suitable amines are chloroprocaine, choline, N,N'-dibenzylethylenediamine, 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 con~Tentional 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.
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 be generated from 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 (nucleot;ides), are present as impurities in the n-mer oligonucleatide 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 al., 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 the active drug component, and are thus considered to be bioequivalents for purposes of the present invention.
Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids.
The compositions of the present invention may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, liquid syrups, soft gels, suppositories, and enemas.
Pharmaceutically acceptable organic or inorganic carrier substances suitable for non-parenteral administration which do not deleteriously react with nucleic acids can also be used to formulate the compositions of the present invention. Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohols, polyethylene g:Lycols, gelatin, lactose, amylose, magnesium stearate, talc, ailicic 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 interact with the nucleic acids) of the formulation.
The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. Aqueous suspensions may further contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.
In one embodiment of the present invention the pharmaceutical compositions may be formulated and used as foams. Pharmaceutical foams include formulations such as, but not limited to, emulsions, microemulsions, creams, jellies and formulations containing liposomes. While basically similar in nature these formulations vary in the components and the consistency of the final product. The know-how on the preparation of such compositions and formulations is generally known to those skilled in the pharmaceutical and formulation arts and may be applied to the formulation of the compositions of the present invention.
The compositions of the present invention may be prepared and formulated as emulsion;. Emulsions are typically heterogenous systems of one liquid dispersed in another in the form of droplets usually exceeding O.l um in diameter.
(Idson, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marvel Dekker, Inc., New York, NY, 1988, p. 199; Rosoff, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marvel Dekker, Inc., New York, NY, 1988, p. 245; Block, in Pharmaceutica3 Dosage Forms: Disperse Systems, Vol. 2, Lieberman, Rieger and Banker, Eds., Marvel Dekker, Inc., New York, NY, 1988, p. 335; Higuchi et al., in "Remington~s Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 1985, p. 301). Emulsions are often biphasic systems comprising of two immiscible: liquid phases intimately mixed and dispersed with each other. In general, emulsions may be either water in oil (w/o) or of the oil in water (o/w) variety. When an aqueous phase is finely divided into and dispersed as minute droplets into a bulk oily phase the resulting composition. is called a water in oil (w/o) emulsion. Alternatively, when an oily phase is finely divided into and dispersed as minute droplets into a bulk aqueous phase the resulting composition is called an oil in water (o/w) emulsion.
Emulsions may contain additional components in addition to the dispersed phases arid the active drug which may be present as a solution in either the aqueous phase, oily phase or itself as a separate phase. Pharmaceutical excipients such as emulsifiers, stabilizers, dyes, and anti-oxidants may also be present in emulsions as needed.
Pharmaceutical emulsions may also be multiple emulsions that are comprised of more than two phases such as, for example, in the case of oil in water in oil (o/w/o) and water in oil in water (w/o/w) emulsions. Such complex formulations often provide certain advantages that simple binary emulsions do not. Multiple emulsions in which individual oil droplets of an o/w emulsion enclose small water droplets constitute a w/o/w emulsion. Likewise a system of oil droplets enclosed in globules of water stabilized in an oily continuous provides an o/w/o emulsion.
Emulsions are characterized by little or no thermodynamic stability. Often, the dispersed or discontinuous phase of the emulsion is well dispersed into the external or continuous phase and maintained in this form through the means. of emulsifiers or the viscosity of the formulation. Either of the phases of the emulsion may be a semisolid or a solid, as is the case of emulsion-style ointment bases and creams. Other means of stabilizing emulsions entail the use of emulsifiers that may be incorporated into either phase of the emulsion. Emulsifiers may broadly be classified into four categories: synthetic surfactants, naturally occurring emulsifiers, absorption bases, and finely dispersed solids (Idson, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 199) .
Synthetic surfactants, also known as surface active agents, have found wide applicability in the formulation of emulsions arid have been reviewed in the literature (Rieger, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p., 285; Idson,in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 199) . Surfactant:s are typically amphiphilic and comprise a hydrophi7.ic and a hydrophobic portion. The ratio of the hydrophilic to the hydrophobic nature of the surfactant has been teamed the hydrophile/lipophile balance (HLB) and is a valuable tool in categorizing and selecting surfactants in the preparation of formulations. Surfactants may be classified into different classes based on the nature of the hydrophilic group into:
nonionic, anionic, cationic and amphoteric (Rieger, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 285).
Naturally occuring emulsifiers used in emulsion formulations include lanolin, beeswax, phosphatides, lecithin and acacia. Absorption bases possess hydrophilic properties such that they can soak up water to form w/o emulsions yet retain their semisolid consistencies, such as anhydrous lanolin and hydrophilic petrolatum. Finely divided solids have also been used as good emulsifiers especially in combination with surfactants and in viscous preparations. These include polar inorganic solids, such as heavy metal hydroxides, nonswelling clays such as bentonite, attapulgite, hectorite, kaolin, montmorillonite, colloidal aluminum silicate and colloidal magnesium aluminum silicate, pigments and nonpolar solids such as carbon or glyceryl tristearate.
A large variety of non-emulsifying materials are also included in emulsion formulations and contribute to the properties of emulsions. These include fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hydrophilic colloids, preservatives and antioxidants (Block, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marvel Dekker, Inc., New York, NY, 1988, p. 335; Idson, Id., p. 199).
Hydrophilic colloids or hydrocolloids include naturally occuring gums and synthetic polymers such as polysaccharides (for example, acacia, agar, alginic acid, carrageenan, guar gum, karaya gum, and tragacanth), cellulose derivatives (for example, carboxymethylc cellulose and carboxypropyl cellulose), and synthetic polymers (for example, carbomers, cellulose ethers, and carboxyvinyl polymers). These disperse or swell in water to form colloidal solutions that stabilize emulsions by forming strong interfacial films around the dispersed-phase droplets and by increasing the viscocity of the external phase.
Since emulsions often contain a number of ingredients such as carbohydrates, proteins, sterols and phosphatides that may readily support the growth of microbes, these formulations often incorporate preservatives. Commonly used preservatives included in emulsion formulations include methyl paraben, propyl paraben, quaternary ammonium salts, benzalkonium chloride, esters of p-hydroxybenzoic acid, and boric acid.
Antioxidants are also commonly added to emulsion formulations to prevent deterioration of the formulation.
Antioxidants used may be free radical scavengers such as tocopherols, alkyl gallates, butylated hydroxyanisole, butylated hydroxytoluene, or reducing agents such as ascorbic acid and sodium metabisulfite, and antioxidant synergists such as citric acid, tartaric acid, and lecithin.
The application of emulsion formulations via dermatological, oral and parenteral :routes and methods for their manufacture have been reviewed in the literature (Idson, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marvel Dekker, Inc., New York, NY, 1988, p. 199). Emulsion formulations for oral delivery have been very widely used because of reasons of ease of formulation, efficacy from an absorption and bioavailability standpoint. (Rosoff, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York,. NY, 1988, p. 245; Idson, Id., p. 199). Mineral-oil base laxatives, oil-soluble vitamins and high fat nutritive preparations are among the materials that have commonly been administered orally as o/w emulsions.
In one embodiment of the present invention, the compositions of oligonucleotides and nucleic acids are formulated as microemulsions. A microemulsion may be defined as a system of water, oil and amphiphile which is a single optically isotropic and thermodynamically stable liquid solution (Rosoff, in Pharmaceutical Dosage Forms:
Disperse Systems, Vol. l, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 245).
Typically microemulsions are systems that are prepared by first dispersing an oil in an aqueous surfactant solution and then adding a sufficient amount of a fourth component, generally an intermediate chain-length alcohol to form a transparent system. Therefore, microemulsions have also been described as thermodynamically stable, isotropically clear dispersions of two immiscible liquids that are stabilized by interfacial films of surface-active molecules (Leung and Shah, in: Controlled Release of Drugs: Polymers and Aggregate Systems, Rosoff, M., Ed., 1989, VCH
Publishers, New York, pages 185-215). Microemulsions commonly are prepared via a combination of three to five components that include oil, water, surfactant, cosurfactant and electrolyte. Whether the microemulsion is of the water-in-oil (w/o) or an oil-in-water (o/w) type is dependent on the properties of the oil and surfactant used and on the structure and geometric packing of the polar heads and hydrocarbon tails of the surfactant molecules (Schott, in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 1985, p. 271).

The phenomenological approach utilizing phase diagrams has been extensively studied and has yielded a comprehensive knowledge, to one skilled in the art, of how to formulate microemulsions (Rosoff, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 245; Block, Id., p. 335). Compared to conventional emulsions, microemulsions offer the advantage of solubilizing water-insoluble drugs in a formulation of thermodynamically stable droplets that are formed spontaneously.
Surfactants used in the preparation of microemulsions include, but are not limited to, ionic surfactants, non-ionic surfactants, Brij 96, polyoxyethylene oleyl ethers, polyglycerol fatty acid esters, tetraglycerol monolaurate (ML310), tetraglycerol monooleate (M0310), hexaglycerol monooleate (P0310), hexaglycerol pentaoleate (P0500), decaglycerol monocaprate (MCA750), decaglycerol monooleate (M0750), decaglycerol sequioleate (50750), decaglycerol decaoleate (DA0750), alone or in combination with cosurfactants. The cosurfactant, usually a short-chain alcohol such as ethanol, 1-propanol, and 1-butanol, serves to increase the interfacial fluidity by penetrating into the surfactant film and consequently creating a disordered film because of the void space generated among surfactant molecules. Microemulsions may, however, be prepared without the use of cosurfactants and alcohol-free self-emulsifying microemulsion systems are known in t:he art. The aqueous phase may typically be, but is not limited to, water, an aqueous solution of the drug, glycerol, PEG300, PEG400, polyglycerols, propylene glycols, and derivatives of ethylene glycol. The oil phase may include, but is not limited to, materials such as Captex 300, Captex 355, Capmul MCM, fatty acid esters, medium chain (C8-C12) mono, di, and tri-glycerides, polyoxyethylated glyceryl fatty acid esters, fatty alcohols, polyglycolized glyce;rides, saturated polyglycolized C8-C10 glycerides, vegetable oils and silicone oil.
Microemulsions are particularly of interest from the standpoint of drug solubilization and the enhanced absorption of drugs. Lipid based microemulsions (both o/w and w/o) have been proposed to enhance the oral bioavailability of drugs, including peptides (Constantinides et al., Pharmaceutical Research, 1994, lI, 1385; Ritschel, Meth. Find. Exp. Clin. Pharmacol., 1993, 13, 205).
Microemulsions afford advantages of improved drug solubilization, protection of drug :from enzymatic hydrolysis, possible enhancement of drug absorption due to surfactant-induced alterations in membrane fluidity and permeability, ease of preparation, ease of oral administration over solid dosage forms, improved clinical potency, and decreased toxicity (Constantinides et al., Pharmaceutical Research, 1994, 11, :1385; Ho et al., J.
Pharm. Sci., 1996, 85, 138). Often microemulsions may form spontaneously when their components are brought together at ambient temperature. This may be particularly advantageous when formulating thermolabile drugs,, peptides or oligonucleotides. Microemulsions have also been effective in the transdermal delivery of active components in both cosmetic and pharmaceutical applications. It is expected that the microemulsion compositions and formulations of the present invention will facilitate the increased systemic absorption of oligonucleotides and nucleic acids from the gastrointestinal tract, as well as _Lmprove the local cellular uptake of oligonucleotides and nucleic acids within the-gastrointestinal tract, vagina, buccal cavity and other areas of administration.
Microemulsions of the present invention may also contain additional components and additives such as sorbitan manostearate (Grill 3), Labrasol, and penetration enhancers to improve the properties of the fo~.-mulation and to enhance the absorption of the oligonucleotides and nucleic acids of the present invention. Penetration enhancers used in the microemulsions of the present invention may be classified as belonging to one of five broad categories - surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of these classes has been discussed above.
There are many organized surfactant structures besides microemulsions that have been studied and used for the formulation of drugs. These include monolayers, micelles, bilayers and vesicles. Vesicles, such as liposomes, have attracted great interest because of their specificity and the duration of action they offer from the standpoint of drug delivery. Further advantages are that liposomes obtained from natural phospholipids are biocompatible and biodegradable, liposomes can incorporate a wide range of water and lipid soluble drugs, liposomes can protect encapsulated drugs in their internal compartments from metabolism and degradation (Rosoff, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 245). Important considerations i:n the preparation of liposome formulations are the lipid surface charge, vesicle size and the aqueous volume of the liposomes. Liposomes can be administered orally and in aerosols and topical applications.
Surfactants find wide application in formulations such as emulsions (including microemulsions) and liposomes.
The most common way of classifying and ranking the properties of the many different types of surfactants, both natural and synthetic, is by the use of the hydrophile/lipophile balance (HLB). The nature of the hydrophilic group (also known as the "head") provides the most useful means for categorizing the different surfactants used in formulations (Rieger, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. l, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p.
285) .
If the surfactant molecule is not ionized, it is classified as a nonionic surfactant. Nonionic surfactants find wide application in pharmaceutical and cosmetic products and are usable over a wide range of pH values. In general their HLB values range from 2 to about 18 depending on their structure. Nonionic surfactants include nonionic esters such as ethylene glycol esters, propylene glycol esters, glyceryl esters, polyglyceryl esters, sorbitan esters, sucrose esters, and ethoxylated esters. Nonionic alkanolamides and ethers such as fatty alcohol ethoxylates, propoxylated alcohols, and ethoxylated/ propoxylated block polymers are also included in this class. The polyoxyethylene surfactants are the most popular members of the nonionic surfactant class.
If the surfactant molecules carries a negative charge when it is dissolved or dispersed in water, the surfactant is classified as anionic. Anionic surfactants include carboxylates such as soaps, acyl lactylates, acyl amides of amino acids, esters of sulfuric acid such as alkyl sulfates and ethoxylated alkyl sulfates, sulfonates such as alkyl benzene sulfonates, acyl iseth:ionates, acyl taurates and sulfosuccinates, and phosphates. The most important members of the anionic surfactant class are the alkyl sulfates and the soaps.
If the surfactant molecule carries a positive charge when it is dissolved or dispersed in water, the surfactant is classified as cationic. Cationic surfactants include quaternary ammonium salts and ethoxylated amines.
The quaternary ammonium salts are the most used members of this class.
If the surfactant molecule has the ability to carry either a positive or negative r_harge, the surfactant is classified as amphoteric. Amphoteric surfactants include acrylic acid derivatives, substituted alkylamides, N-alkylbetaines and phosphatides.

The use of surfactants in drug products, formulations and in emulsions has been reviewed (Rieger, in Pharmaceutical Dosage Forms: Disperse Systems, Vol. 1, Lieberman, Rieger and Banker, Eds., Marcel Dekker, Inc., New York, NY, 1988, p. 285).
In one embodiment of the invention, a nucleic acid is administered via the rectal mode. In particular, compositions for rectal administration include solutions (enemas and suppositories) and emulsions. 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).
The use of absorption-promoting adjuvants is known in the art for the modification of t:he barrier function of the rectal membrane and has been reviewed (Nishihata and Rytting, Advanced Drug Delivery Rev.~ews, 1997, 28, 205).
Absorption-promoting adjuvants have shown promising effects on the performance of formulations of poorly absorbed drugs such as moderately large water-soluble drugs and peptides.
Enamine derivatives of amino acids have exhibited absorption promoting properties but the mechanism by which they increase rectal absorption is unclear. Compounds such as chelating agents, and sulfhydryl depleters have been shown to increase the rectal absorption of drugs through the paracellular route as well as the transcellular route.
Salicylate and its derivatives also increase absorption of drugs administered via the rectal route via both paracellular and transcellular paths. Fatty acids show properties similar to salicylates when enhancing rectal absorption of drugs. Lectin is alsc> known to increase rectal absorption of drugs via induction of microvillus infusion.
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. 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 (PVP or gums such as tragecanth, acacia, carrageenan), lubricant (e. g. stearates such as magnesium stearate), glidant (talc, colloidal silica dioxide), inert diluent, preservative, surface active or dispersing agent.
Preferred binders/disintegrants include EMDEX (dextrate), PRECIROL (triglyceride), PEG, and AVICEL (cellulose).
Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound rnoistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as t:o provide slow or controlled release of the active ingredients therein.
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 contain an enteric material 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 muc;osal site. Enteric materials for acid-resistant tablets, capsules and caplets are known in the art and typically include acetate phthalate, propylene glycol, sorbitan monoleate, cellulose acetate phthalate (CAP), cellulose acetate trimellitate and hydroxy propyl methyl cellulose phthalate (HPMCP). Enteric materials may be incorporated within the dosage form or may be a coating substantially covering the entire surface of tablets, capsules or caplets. Enteric materials may also be accompanied by plasticizers which impart flexible resiliency to the material for resisting fracturing, for example during tablet curing or aging. Plasticizers are known in the art and typically include diethyl phthalate (DEP), triacetin, dibutyl sebacate (DBS), dibutyl phthalate (DBP) and triethyl citrate (TEC) .
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 compositions of this invention .can be converted in a known manner into the customary foronulations, such as tablets, coated tablets, pills, granules, capsules, 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. Compositions may be formulated in a conventional manner using additiona_L pharmaceutically acceptable carriers or excipients as appropriate. Thus, the composition may be prepared by conventional means with 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);
disintegrants (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 sweetening agents as appropriate.

Capsules used for oral delivery may include formulations that are well known in the art. Further, multicompartment hard capsules with control release properties as described by Digenis et al., U.S. Patent No.
5,672,359, and water permeable capsules with a multi-stage drug delivery system as described by Amidon et al., U.S.
Patent No. 5,674,530 may also be used to formulate the compositions of the present invention.
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.
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, arid may extend from once daily to once every 20 years. In th.e context of the invention, the term "treatment regimen" is meant to encompass therapeutic, palliative anal 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 if 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 is observed, or if the disorder or disease state has been abated.
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 rnay vary depending on the relative potency of individual oligonucleotides, and can generally be estimated based on ECsovalues found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 ug to 7.00 g per kg of body weight, and may be given once or mane 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 al., Chapter 27 In: Remington's Pharmaceutical Sciences, IBth 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 effects) (Vies et al., Chapter 3 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al., eds., McGraw-Hill, New York, NY, 1996).

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.
Formulations for non-parenteral administration of nucleic acids may include sterile and non-sterile aqueous solutions, non-aqueous solutions in common solvents such as alcohols, or solutions of the nucleic acids in liquid or solid oil bases. The solutions may also contain buffers, diluents and other suitable additives. Pharmaceutically acceptable organic or inorganic carrier substances suitable for non-parenteral 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 interact with the nucleic acids) of the formulation. Aqueous suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.
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 t:echnigues include the step of bringing into association the active ingredients with the pharmaceutical carriers) 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.
A number of bioequivalents of oligonucleotides and other nucleic acids may also be employed in accordance with the present invention. The invention therefore, also encompasses oligonucleotide and nucleic acid equivalents such as, but not limited to, prodrugs of oligonucleotides and nucleic acids, deletion derivatives, conjugates of oligonucleotides, aptamers, and ribozymes.
The methods and compositions of the present invention also encompass the myriad deletion oligonucleotides, both internal and terminal deletion oligonucleotides, that are synthesized during the process of solid-phase manufacture of oligonucleotides for such deletion sequences are for all practical purposes bioequivalents. Synthetic RNA molecules and their derivatives that possess specific catalytic activities are known as ribozymes and are also considered bioequivalents of oligonucleotides for the purposes of the methods and compositions of the present invention. Also considered bioequivalents of oligonucleotides, for the purposes of the methods and compositions of the pres<~nt invention, are peptide nucleic acids (PNAs) and apt<~mers (see, generally, Ellington et al., Nature, 1990, 346, 818; U.S..Patent 5, 523, 389 (Esker et aI. , June 4, 199E~) ) .

The name aptamer has been coined by Ellington and Szostak (Nature, 1990, 346, 818) fox- nucleic acid molecules that fit and therefore bind with significant specificity to non-nucleic acid ligands such as peptides, proteins and small molecules such as drugs and dyes. Because of these specific ligand binding properties, nucleic acids and oligonucleotides that may be classified as aptamers may be readily purified or isolated via affinity chromatography using columns that bear immobilized ligand. Aptamers may be nucleic acids that are relatively short to those that are as large as a few hundred nucleotides. For example, Ellington and Szostak have reported the discovery of RNA aptamers that are 155 nucleotides long and that bind dyes such as Cibacron Blue and Reactive Blue 4 (Ellington and Szostak, Nature, 1990, 346, 818) with very good selectivity. While RNA
molecules were first referred to as aptamers, the term as used in the present invention refers to any nucleic acid or oligonucleotide that exhibits specific binding to small molecule ligands including, but not limited to, DNA, RNA, DNA derivatives and conjugates, RNA derivatives and conjugates, modified oligonucleotides, chimeric oligonucleotides, and gapmers.
The invention is drawn to the non-parenteral 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 translatio:nal arrest (Crooke et al. , 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 Yl receptor in anxiety (Wahlestedt et al., Nature, 1993, 363, 260; Dean et al., Proc. Natl. Acad. Sci.
U. S.A. , 1994, 91, 11762; and Wahlest:edt et al . , Science, 1993, 259, 528, respectively). In instances where complex families of related proteins are being investigated, "antisense knockouts" (.i . e. , inhibit:ion 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, 15, 250) .
As stated, the compositions and methods of the invention are 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-BA-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 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 t:o 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 phosphate linkages.
The synthesis of 2'-0-methyl- (2'-methoxy-) phosphorothioate oligonucleotides is according to the procedures set forth above substituting 2"-O-methyl b-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'-O-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 application Serial No.
08/383,666, filed February 3, 1995, which is assigned to the same assignee as the instant application and which is incorporated by reference herein.
The 2'-fluoro-phosphoroth:ioate oligonucleotides of the invention are synthesized using 5'-dimethoxytrityl-3'-phosphoramidites and prepared as disclosed in U.S. patent application Serial No. 08/383,666, filed February 3, 1995, and U.S. Patent 5,459,255, which issued October 8, 1996, both of which are assigned to the same assignee as the instant application and which are incorporated by reference herein. 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 and (3) are incorporated by reference herein.
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 incorporated by reference herein, and materials and methods described by Gaffney et al. (Tetrahedron, 1984, 40, 3), Chollet et al., (Nucl. Acids Res., 1988, 16, 305} and Prosnyak et a1. (Genomics, 1994, 21, 490). Oligonucleotides comprising 2,6-diaminopurine can also be prepared by enzymatic means (Bailly et al., Proc. Natl. Acad. Sci.
U.S.A. , 1996, 93, 13623) .
2~-Methoxyethoxy oligonucleotides of the invention are synthesized essentially according to the methods of Martin et al. (Helv. Chim. Acta, 1995, 78, 486) .
B. Oligonucleotide Purification: After cleavage from the controlled pore glass (CPG;1 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 NaCl 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).
C. 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 pharmacokinet.ic studies described herein. Although radiolabeling by tritium exchange is one preferred means of 7_abeling 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, 21, 3737) was used to label oligonuc:leotides by tritium exchange. Specifically, about 24 mc~ of oligonucleotide was dissolved in a mixture of 200,uL of sodium phosphate buffer (pH 7 . 8 ) , 400 ~eL of 0 .1 mM EDTA (pH 8 .3 ) and 200 ~.L of deionized water. The pH of the resulting mixture was measured and adjusted to pH 7.8 using 0.095 N NaOH. The mixture was lyophilized overnight in a 1.25 mL gasketed polypropylene vial. The oligonucleotide was dissolved in 8.25 JCL of b-mercaptoethanol, which acts as a free radical scavenger (Graham et al., Nucleic Acids Research, 1993, 2I, 3737), and 400 ~.L of tritiated H20 (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 ~.L 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 ~eL 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 H20 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 H20, to ensure the removal of any residual, unincorporated tritium. The final resuspended olic~onucleotide solution is transferred to a clean polypropylene vial and assayed. The tritium labeled oligonucleotide is stored at about -70°C.
2. Other Meaas of Lalbeling Nucleic Acidss 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.
For example, double-stranded nucleic acids can be radiolabeled by nick translation and primer extension, and a variety of nucleic acids, including oligonucleotides, can be terminally radiolabeled by the use of enzymes such as T4 polynucleotide kinase or terminal d.eoxynucleotidyl transferase (see, generally, Chapter 3 In: Short Protoco.Is in Molecular Biology, 2d Ed., Ausubel et al., eds., John Wiley & Sons, New York, NY, pages 3-11 to 3-38; and Chapter 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 10 nucleic acids with nonradioactive labels such as, for example, enzymes,, fluorescent moieties and the like (see, for example, Beck, Methods in Enzymology, 1992, 216, 143;
and Ruth, Chapter 6 In: Protocols for 0ligonuc3eotide Conjugates (Methods in Molecular Biology, Volume 26t Agrawal, S., ed., Humana Press, Totowa, NJ, 1994, pages 167-185) .
Example 2: Oligonucleotide Targets and Sequences The present invention is drawn to compositions and formulations comprising oligonucleot~ides or nucleic acids and one or more 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 local or systemic therapeutic delivery, as desired, that may be administered via non-parenteral means 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) and non-retroviral viruses (Table 6). Further details regarding these oligonucleotides are provided in the Sequence Listing.
TABLE 1: TARGET OLIGONUCLEOTIDES DESIGNED
TO MODULATE CELLULAR ADHESION
Cell Surface Commercial or Common Oligonucleotide Sequence Target Proteia Name (if any) of SEQ ID NO(S):
Oligonucleotide ELAM-1 GM1515, GM1516, GM15176, 7, 8 ~

TABLE 2s OLIGONUCLEOTIDES DESIGNED TO
MODULATE THE RATE OF CELLULAR PROLIFERATION
Molecular Target Commercial or CommoaOligonucleotide Sequence Name (if any) of SEQ ID NO(S):
Oligonucleotide c-myb MYB-AS !3 2 vascular endothelial :LO, 11, 12 0 growth factor (VEGF) bc1-2 :L3, 14, 15 Ha-ras ISIS 2503 16 2 A-raf kinase ISIS 9069 18 c-raf kinase ISIS 5132 1~9 WO 99/60012 PCTlUS99l11394 TABLE 3s OLIGONUCLEOTIDES DESIGNED TO HAVE TH$RApEtITIC
ACTIVITY AGAINST MISCELLANEOUS DISORDERS
Disorder Commercial or CommonOligonucleotide Sequence Name (if any) of SEQ ID NO(S)s Oligoaucleotide Alzheimer's disease 20, 21 Beta-thalassemia 5~ss & 3'as 22, 23 TABLE 4s OLIGONLTCLEOTIDES DESIGNED TO HAVE THERAPEUTIC
ACTIVITY AGAINST EUKARYOTIC PATHOGENS
Pathogen / Disease Commercial or CommonOligonucleotide Sequence Name (if any) of SEQ ID NO (S) s Oligonucleotide Plasmodium / malariaPSI, PSII 24, 25 Schistosoma / 26 bloodfluke infections TABLE 5s OLIGONUCLEOTIDES DESIGNED TO HAVE THERAPEUTIC
ACTIVITY AGAINST RETROVIRUSES. INCLUDING HIV
Virus / Molecular Commercial or Common Oligonucleotide Sequence Target Name (if nay) of SEQ ID NO(S)s Oligoaucleotide HIV-1 / gag GEM 91 27 HIV-1 / gag 28, 29 HIV / tat, vpr, rev, 31, 32 2 env, nef HIV / pol, env, vir 33, 34 HIV-1 / tat, rev, 35, 36 env, nef HIV / gp120 ISIS 5320 37 2 Hepatitis C virus ISIS 6547 ~ 38 TABLE 6: OLIGONUCLEOTIDES DESIGNED TO HAVE THERAPEUTIC
ACTIVITY AGAINST NON-RETROVIRAL VIRUSES
Virus / Molecular Commercial or CommonOligoaucleotide Sequeace Target Name (if any) of SEQ ID NO(S):
Oligoaucleotide influenza virus 39, 40 Epstein-Barr Virus 41, 42 Respiratory Syncytial 43, 44 Virus cytomegalovirus (CMV)GEM 132 45 CMV 46, 47 Additional oligonucleotides that may be formulated in the compositions of the invention include, for example, ribozymes, aptamers, molecular decoys, External Guide Sequences (EGSs) and peptide nucleic acids (PNAs).
Example 3: Evaluation of Formulations By In Situ Perfusion of Rat Ileum The formulations of the invention may be evaluated as follows.
Methods: In order to evaluate the formulations, in situ perfusion of rat ileum is performed essentially according to the procedure of Komiya et: al. (Int. J.
Pharmaceut., 1980, 4:249). Specifically, male Sprague Dawley rats weighing 250-300 g are used. After overnight fasting, the rats are anesthetized with. 5% pentobarbital (50 mg/kg) by intraperitoneal injection. After a midline abdominal incision is made, the small intestine is taken out and ileum section located. An incision is made at each end of a 20 cm ileum segment. The ileum segment is laid out in a uniform multiple-S arrangement with 3 bends. The luminal contents of the section are flushed with buffer. A 10 cm piece of silicon rubber tubing is inserted into the intestinal lumen at each incision and Iigated with 3-0 silk suture. The proximal end tubing is connected to a 30 mL
syringe containing oligonucleotide solution. The formulation is perfused through the intestinal segment by using Sage model 365 syringe pump at: 0.125 mL/min. The outflow solution is collected in a 2 mL centrifuge tube over 5 min intervals for 80 rains. At the end of perfusion study, an aliquot of 0.3 mL blood sample is collected from the portal vein.
The oligonucleotide concentration in the solution before and after passing through a 20 cm ileum segment is analyzed by high pressure liquid chromatography (HPLC) while the plasma samples are analyzed by capillary electrophoresis (CE). In most cases, tritium labeled ISIS 2302 is used as a tracer and the radioactivity of solution measured by a liquid scintillation counter. The amount of the drug absorbed from the ileum is calculated by dividing the concentration from the average of last six outflow samples (steady state) to that of the inflow sample.
Example 4: Evaluation of the Bioavailability of Oligonucleotide from Formulations Folloaring Ia 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 7).

TABLE 7: Intrajejunum Formulations 4a - 4c Formulation No. ISIS 2302 Penetration Enhancer(s) Concentration 4a 20 mg/ml CDCA 20 mg/ml Solution Caprate 40 mg/ml Laurate 40 mg/ml 4b 20 mg/ml UDCA 20 mg/ml Solution Caprate 40 mg/ml Laurate 40 mg/ml 4c - 12 mg/ml Microemulsion Formulation 4a: First, 100 mg CDCA was transferred to a 5 rnl 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 4b: 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 4cs 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 (Abitec Corp.) was added to the beaker. The resultant solution was stirred until. a clear solution was formed.
Methodss Precannulated 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 injection site was located (2 cm after the ligament of Treitz). An aliquot of 1.0 mL drug solution was then injected via a 27 gauge needle. The intestine was put back to the body carefully. Muscle was then surgically closed and skin was clipped after injection.. Three hundred ~.L of blood was collected from a cannula at each sampling time point. The rats were sacrificed in the C02 chamber 24 hours after dosing. Livers and kidneys were excised and stored at -80EC until analysis. Radioactivity of plasma and tissue samples were measured. Liver and kidney were also analyzed for oligonucleotide content by capillary gel electrophoresis (CGE) .
Results: The results of study are summarized in Table 8. No significant amount (i.e~., ~0%) of ISIS 2302 (SEQ ID NO:1) was absorbed when a control solution (i.e., one lacking any penetration enhancers) was used. In contrast, when formulated as a solution containing a mixture of penetration enhancers (Formulations 4a and 4b) the absolute bioavailability of ISIS 2302 was in the range of 8 to 23% in the examined target organs (liver and kidneys).
The AUC(0-3h) shows 10-13% bioavailability. When formulated as a microemulsion (Formulation 4c), in the absence of any penetration enhancers, the absolute bioavailability was surprisingly found to be 19-29% in the target organs (liver and kidneys). Formulation 4c provided an AUC(0-3h) showing about 13% bioavailability that is comparable to the bioavailability seen with Formulations 4a and 4b that used - ~a -penetration enhancers in solution. 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 8:
Percent Absolute Bioavailability (~ i.v.) of ISIS 2302 Following Intrajejunum Instillation in Rats Formulation AUC(0-3h) 1 No. (Dose) Liverl Kidrxeyl (ug x h/mL) Formulation 4a 17.4 17.8 10.7 (80 m /k ) Formulation 4b 8.8 23.0 13.5 (80 m /k }

1 Formulation 4c 19.8 29.1 13.6 (48 m k ) 1 According to the CGE analysis - total oligonucleotide.
2 According to analysis by radioactivity. AUC(0-3h) was 20 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 5s Preparation of Microemulsion Formulations 25 In order to evaluate the bioavailability of oligonucleotide microemulsions the following microemulsion formulations of ISIS 2302 were prepared (Table 9).
TABLE 9s Microemulsion Formulations 5a and 5b Formulation ISIS 2302 30 No. Concentration Components 5a 12 mg/ml Captex 355 3 parts Capmul MCM 1 part Tween 80 0.5 part 5b 4 mg/ml Captex 355 3 parts 35 Capmul MCM 1 part Tween 80 0.04 art WO 99160012 PCT/U~99/11394 _ 73 Formulation 5as 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 (Abitec Corp., Janesville, WI) was added to the beaker. The resultant solution was stirred until a clear solution was formed.
Formulation 5bs A water-in-oil microemulsion of ISIS 2302 was prepared essentially :by adding the oil phase to the aqueous phase with adequate 'mixing. The aqueous phase was prepared by mixing 1 ml of a 100 mg/ml solution of ISIS 2302 and 1 ml of Tween 80 (Sigma Chemical Company St.
Louis, MO). The oil phase was prepared by mixing 3 parts of Captex 355 (Abitec Corp., Janesville, WI) and 1 part of Capmul MCM (Abitec Corp., Janesville, WI). The oil phase was added to the aqueous phase with adequate stirring until the resultant mixture was a clear solution.
8xample 6s Preparation of Water-in-Gil (rn/o) Cream Formulations In order to evaluate the bioavailability of oligonucleotide emulsions the water-in-oil cream formulations of ISIS 2302 described in Table 10 were prepared as follows.
Formulation Gals A water-in-oil cream formulation of ISIS 2302 was prepared by first preparing the two phases.
A 2 ml aliquot of the ISIS 2302 stock solution (100 mg/ml) was mixed with 2 ml water in a 10 m:1 beaker and warmed to 70°C. A mixture of 1 gram of Grill 3 (Croda, U.S.), 3 ml Captex 355 (Abitec Corp., Janesville, WI) and 3 ml of Labrasol (Gattefosse, France) was prepared in a 30 ml beaker and this mixture was also warmed to 70°C. The aqueous solution of oligonucleotide was then poured slowly into the oil phase with vigorous mixing. Upon cooling to ambient temperature the desired water-in-oi:l cream formulation of oligonucleotide (~20 mg/mL) was obtained.
Formulation 6a2: A water-in-oil cream formulation of ISIS 2302 was prepared by first preparing the two phases.
A 1.5 ml aliquot of the ISIS 2302 stock solution (200 mg/ml) was transferred to a 10 ml beaker and warmed to 70°C. In a 30 ml beaker were placed 0.5 gram of Grill 3 (Croda, U.S.), 1.5 ml Captex 355 (Abitec Corp., Janesville, WI), and 1.5 ml of Labrasol (Gattefosse, France) and this mixture also warmed to 70°C. The aqueous solution of oligonucleotide was then poured slowly into the oil phase with vigorous mixing.
Upon cooling to ambient temperature the desired water-in-oil cream formulation oligonucleotide ('r60 mg/mL) was obtained.
TABLE lOs Water-in-Oil Cream Formulations 6a1 (~20 mg/mL):
Aqueous Phase 2 ml ISIS 2302 solution (100 mg/ml) 2 mL water Oil Phase 1 g. Grill 3 (Sorbitan Monostearate) 3 ml Captex 355 3 ml Labrasol ~6a2 (~60 mg/mL) Aqueous Phase 1.5 ml ISIS 2302 solution (200 mg/ml) Oil Phase 0.5 g. Grill 3 (Sorbitan Monostearate) 1.5 ml Captex 355 1.5 ml Labrasol Example 7: Preparation of Oil-in-Water (o/w) Cream Formulations In order to evaluate the bioavailability of oligonucleotide emulsions the following oil-in-water cream formulations of ISIS 2302 were prepared (Table 1Z).
TABLE 11: Oil-ia-water Cream Formulation 7a Aqueous Phase 0.5 ml ISIS 2302 :solution (200 mg/ml) 0.5 ml Tween 80 1.8 ml water Oil Phase 100 mg. Grill 3 (Sorbitan Monostearate) 1 ml Captex 355 1 ml Labrasol Formulation 7as An oil-ira-water cream formulation of ISIS 2302 was prepared by first preparing the two phases.
A 0.5 ml aliquot of the ISIS 2302 stock solution (200 mg/ml) was mixed with 0.5 ml of Tween 80 (Sigma Chemical Company St. Louis, MO) and 1.8 ml water in a 30 ml beaker and warmed to about 70°C. In a 10 ml beaker were placed 100 mg. of Grill 3 (Croda, U.S.), 1 ml Captex :355 (Abitec Corp., Janesville, WI), and 1 ml of Labrasol (Gattefosse, France) and this mixture also warmed to about 70°C. The oil phase was then poured into the aqueous solution of oligonucleotide with vigorous mixing. Upon cooling to ambient temperature the desired oil-in-water cream formulation was obtained.
Example 8: In Vivo Evaluation of Emulsion Formulations of (A) Following In Vivo Intrajejunum Instillation In order to determine the ability of emulsion formulations to effectively deliver oligonucleotide drugs with adequate bioavailability the emulsion formulations of the invention were administered via intrajejunum instillation and the plasma concentrations of the oligonucleotide and AUC(0-3h) were measured.
Methods: Sprague-Dawley z:ats 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 injection site was located (2 cm after the ligament of Treitz). An aliquot of 0.5 mL drug formulation was then injected via a 27 gauge needle. The intestine was put back into the body carefully. The incision portion was then covered with a wet gauze. 300 ~L of blood was collected by a 27 gauge needle from the femoral vein at each sampling time point. The rats were sacrificed in a carbon dioxide chamber 3 hours after dosing. Plasma samples were analyzed by HPLC.
Formulations: A 20 mg/ml :solution of ISIS 2302 was used as a control formulation. Water-in-oil cream formulations (Formulation Gal and 6a2) were used as the test formulation at two dosage levels - 10 mg/rat (6a1) and 30 mg/rat (6a2).
Results: The results of ~~tudy are summarized in Table 12. No significant amount (i.e., ~0%) of ISIS 2302 (SEQ ID N0:1) was absorbed when a control solution of the oligonucleotide was used. In contrast, when formulated as a water-in-oil cream (Formulation 6al), the blood concentration of ISIS 2302 was found to reach a high of about 34 ~,g/ml (as determined by HPLC) within 0.5 h of dosing at 10 mg/rat. The concentration of total oligonucleotides, which includes n-1 oligo and related ISIS
2302 metabolites, was observed to be as high as about 36 ~.g/ml within 0.5 h of dosing. When the rats were dosed at 30 mg/animal (formulation 6a2) the blood concentration of _ 77 -ISIS 2302 was found to reach a high of about 64 ~g/ml (and total oligonucleotides reached 81 ~.g/ml) within 0.5 h of dosing. The AUC(0-3h) of ISIS 2302 was determined to be 55 ug x h/mL from the 10 mg/rat dose and 81 ug x h/mL from the 30 mg/rat dosing.
TABLE 12s Blood Concentrations) aad AUC(0-3h) of ISIS 2302 After Intrajejuaum Instillation in Rats Formula- ISIS 2302 Total Oligo ISIS 2302 Total tion No. Time Blood Conc. Blood Conc. AUC(0-3h) AUC(0-3h) 10/ Dose (h.) (ug/ml) (ug/ml) (ug.h/ml) (ug.h/ml) 6a1 0.5 33.73 35.98 55.24 50.64 1 29.53 29.11 mg/rat 2 16.45 15.15 3 0.33 ~~ .

6a2 0.5 64. 80.63 81.52 101.

1 48.49 61.77 30 mQ/rat 2 17.67 21.28 3 6.89 6.70 lAs determined by HPLC analysis.
(B) Follovviag Rectal Administration In order to determine the ability of emulsion formulations to effectively deliver oligonucleotide drugs with adequate bioavailability the emulsion formulations of the invention were administered via the rectum and the blood concentrations of the oligonucleotide and AUC(0-3h) were measured.
Methodsz 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. Test rats were first administered a cleansing enema and then dosed with a sample of the test _ 78 _ formulation. A 0.5 ml formulation was applied via a 2 cm length catheter. The rat was lifted up by the bottom in a 15 degree angle to prevent the sample leakage during the experimental period. 300 ~L of blood was collected by a 27 guage needle from the femoral vein at each sampling time point. The rats were sacrificed in a carbon dioxide chamber 3 hours after dosing. Plasma samples were analyzed by HPLC.
Formulations: A 20 mg/ml solution of ISIS 2302 was used as a control formulation. Three different emulsions of 2302 were evauated as test formulations: a water-in-oil microemulsion (Formulation 5b), a water-in-oil cream formulation (Formulations 6a1 and 6a2) and an oil-in-water cream formulation (Formulation 7a).
Results: The results of study are summarized in Table 13. No significant amount (i.e., ~0%) of ISIS 2302 (SEQ ID N0:1) was absorbed when a control solution of the oligonucleotide was used. In contrast, when the oligonucleotide was administered rectally as a water-in-oil microemulsion (Formulation 5b) at 10 mg/rat significant absorption of the oligonucleotide occured as observed from blood levels of about 21 ~,g/mL within 0.5 h and an AUC(0-2h) of about 28 ug x h/mL.
When formulated as a water-in-oil cream (Formulation 6a1) the plasma concentration of ISIS 2302 was found to reach a high of about 34 ~.c~/ml (as determined by HPLC) within 0.5-1.0 h of dosing at 10 mg/rat. The concentration of total oligonucleotides, which includes N-1 oligo and related ISIS 2302 metabolites was observed to be as high as about 45 ~.g/ml within 0.5-1.0 h of dosing. When the rats were dosed at 30 mg/animal (formulation 6a2) the plasma concentration of ISIS 2302 was found to reach a high of about 105 ~Cg/ml (and total oligonucleotides reached 136 ~.g/ml) within 0.5 h of dosing. The AUC(0-3h) was determined to be 64 ug x h/mL from the 10 mg/rat dose and 143 ug x h/mL
from the 30 mg/rat dosing. A similar increase in delivery _ 79 _ of the oligonucleotide into the blood circulation was observed with the oil-in-water cream (Formulation 7a) when administered rectally at a dose of 10 mg/rat, but the AUC(0-3h) was lower than that observed wit:h the water-in-oil cream.
Hxample 9s Determination of Hioavailability of Oligonucleotides folloyving Intrajejunal and Rectal Administration of Formulations In order to determine the bioavailability of formulations of oligonucleotide drugs, two different modes of administration of oligonucleotide formulated in Formulation 6a1 were studied.
Methodss Intrajejunal Instillation: Sprague-Dawley rats weighing 250-300 g were used. After. overnight fasting, the rats were anesthetized with 5~ pentabarbital (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). An aliquot of 0.5 mL drug formulation was then injected via a 27 gauge needle. The intestine was put back into the body carefully.

WO 99/60012 PCT/US99/I i 394 TABLE 13s Plasma Conceatrationsl aad AUC of ISIS 2302 After Rectal Administration of Formulations to Rats ISIS 2302 Total Oligo Formula- Time plasma Plasma ISIS 2302 Total tion No. Conc. Coac. AUC AUC
& Dose (h. ) (~,g/ml) (~.g/ml) (~ug.h/ml) (~cg.h/ml) 5b 0.5 21.31 29.00 27.952 40.852 1 19.00 25.05 ma/rat 2 5.71 11.19 6a1 0.5 34.37 45.07 64.313 84.52' 1 33.93 44.69 10 mQ/rat 2 20.84 27.42 3 10.26 13.37 10 .."6a2 0.5 105.84 136.49 115.85' 143.29' ~

1 71.64 98.49 30 ma/rat 2 20.29 15.47 3 4.28 5.54 7a 0.5 33.24 43.42 34.96' 46.033 1 21.09 27.42 10 ma/rat 2 4.87 6.74 3 3.61 4.89 1 As determined by FiPLC analysis .
2AUC(0-2h).
3 AUC ( 0-3h) .
Rectal Administrations Following a period of overnight fasting, the rats were anesthetized with 5%
pentobarbital (50 mg/kg). Test rats were first administered a cleansing enema and then dosed with a sample of the test formulation. The enema formulation was applied via a 2 cm length catheter. The bottom part of rat was lifted up in a 15 degree angle to hold the solution.
In order to assess bioavailability of oligonucleotide, samples were processed and the amount of oligonucleotide present was assessed by HPLC analyses.
Resultss The absolute bioavailability of ISIS 2302 (SEQ ID N0: 1) was determined following intrajejunal instillation in five Sprague-Daley rats and following rectal administration in seven rats. The results are shown in Table 14.
TABLE 14: Bioavailability of ISIS 2302 Following Intrajejunal and Rectal Administration in Rats Route of Absolute Administration Bioavailability Intrajejunal 20.3% (n=5) Rectal 24.5% (n=7) 8xample 10: Preparation of Emulsion Formulations Containing Penetration Enhancers Various fatty acids, their salts 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); capryli.c 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 ) .
Various natural bile salts, and their synthetic derivatives act as penetration enhancers. 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). Bile salt derived penetration enhancers include, for example, cholic acid, cholalic acid or 3a,7a,12a-trihydroxy-5b-cholan-24-oic acid (or its pharmaceutically acceptable sodium salt);
deoxycholic acid, desoxycholic acid, 5b-cholan-24-oic acid-3a,12a-diol, 7-deoxycholic acid or 3a,12a-dihydroxy-5b-cholan-24-oic acid (sodium deoxycholate); glycocholic acid, (N-[3a,7a,12a-trihydroxy-24-oxocholan-24-yl]glycine or 3a,7a,12a-trihydroxy-5b-cholan-24-oic acid N-[carboxymethyl]amide or sodium glycocholate);
glycodeoxycholic acid, (5b-cholan-24-oic acid N-[carboxymethyl]amide-3a,12a-diol), 3a,12a-dihydroxy-5b-cholan-24-oic acid N-[carboxymethyl]amide, N-[3a,12a-dihydroxy-24-oxocholan-24-yl]glycine or glycodesoxycholic acid (sodium glycodeoxycholate); taurocholic acid, (5b-cholan-24-oic acid N-[2-sulfoethyl]amide-3a,7a,12a-triol), 3a,7a,12a-trihydroxy-5b-cholan-24-oi.c acid N-[2-sulfoethyl]amide or 2-[(3a,7a,12a-trihydroxy-24-oxo-5b-cholan-24-yl)amino] ethanesulfonic acid (sodium taurocholate); taurodeoxycholic acid, (3a,12a-dihydroxy-5b-cholan-2-oic acid N[2-sulfoethyl]amide or 2-[(3a,12a-dihydroxy-24-oxo-5b-cholan-24-yl)-amino]ethanesulfonic acid, or sodium taurodeoxycholate, or sodium taurodesoxycholate);
chenodeoxycholic acid (chenodiol, chenodesoxycholic acid, 5b-cholanic acid-3a,7a-diol, 3a,7a-dihydroxy-5b-cholanic acid, or sodium chenodeoxycholate, or CDCA); ursodeoxycholic acid, (5b-cholan-24-oic acid-3a,7b-c~iol, 7b-hydroxylithocholic acid or 3a,7b-dihydroxy-5b-cholan-24-oic acid, or UDCA); 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 C:o., Easton, PA, 1990, WO 99160012 PCT/lJS99/11394 pages 782-783). Commercial sources of the penetration enhancers are listed in Table 15.
TABLE 15s Sources of Penetration Enhancers A. FATTY ACIDS AND DERIVATIVES
Compound Name Abbreviation Supplier Capric acid, Na salt caprate S igmat Lauric acid, Na salt laurate Sigma B. BILE SALTS AND DERIVATIVES
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*
tSigma, Sigma Chemical Company, St. Louis, M0.
*Aldrich, Aldrich Chemical Company, Milwaukee, WI.
In order to evaluate the ability of various penetration enhancers to enhance the oral delivery and/or mucosal penetration of oligonucleotides, and the bioavailability of oligonucleotides the following formulations were prepared (Table 16). Formulations 9a and 9c were prepared as emulsions of ISIS 2302 containing a combination of fatty acid and bile salt penetration enhancers. Formulations 9b and 9d were prepared such that the concentration of ISIS 2302 and the penetration enhancers remained the same as in Formulations 9a and 9c, respectively. However, formulations 9b and 9d are merely solution preparations and serve as comparison points for the emulsion formulations.
TABLE 16s ISIS 2302 Formulations Containing Peaetration Enhancers (PE's) 9a-9d Formulation Oil Phase Aqueous Phase 9a Captex 355 1.25 ml ISIS 2302 0.5 ml Labrasol 1.25 ml Mix. Of PE~s 1.0 ml Grill 3 0.5 g. Water 0.5 ml The aqueous phase contains 2% CDCA, 4% Sodium Laurate, and 4%~ Sodium Caprate 9b Solution of ISIS 2302 (20 mg/ml) containing 2% CDCA, 4% Sodium Laurate, and 4% Sodium Caprate 9c Captex 355 1.25 ml ISIS 2302 0.5 ml Labrasol 1.25 ml Mix. Of PE~s 1.0 ml Grill 3 0.5 g. Water 0.5 ml The aqueous phase contains 2% UDCA, 4% Sodium Laurate, and 4%~ Sodium Caprate 9d Solution of ISIS 2302 (20 mg/ml) containing 2% UDCA, 4% Sodium Laurate, and 4% Sodium Caprate Note: ISIS 2302 Stock Solution used was 200 mg/ml.

Formulation gas An emulsion formulation of ISIS
2302 was prepared by first preparing the two phases. A 0.5 ml aliquot of the ISIS 2302 stock solution (200 mg/ml) was mixed with 1.0 ml of the mixture of penetration enhancers (chenodeoxycholic acid sodium salt, sodium laurate and sodium caprate) and 0.5 ml water, and warmed to about 70°C
(aqueous phase). A separate mixture of 500 mg. of Grill 3 (Croda International Plc., East Yorkshire, U.K.), 1.25 ml Captex 355 (Abitec Corp., Janesville, WI), and 1.25 ml of Labrasol (Gattefosse Corp., Westwood, N.J.) was also prepared and warmed to about 70°C (ail phase). The aqueous phase was then transferred to the o:il phase with vigorous mixing to afford the desired emulsion concentration of 20 mg/ml ISIS 2302. The aqueous phase of emulsion contained 2%
CDCA, 4% Sodium laurate, and 4% Sodium caprate.
Formulation 9bs Aliquots of the stock solution of ISIS 2302 (200 mg/ml) and mixture of penetration enhancers were mixed to afford a solution formulation comprising ISIS
2302 at a concentration of 20 mg/ml and a final concentration of 2% CDCA, 4% Sodium laurate and 4% Sodium caprate.
Formulation 9cs An emulsion formulation of ISIS
2302 was prepared by first preparing the two phases. A 0.5 ml aliquot of the ISIS 2302 stock solution (200 mg/ml) was mixed with 1.0 ml of the mixture of penetration enhancers (ursodeoxycholic acid sodium salt, aodium laurate and sodium caprate) and 0.5 ml water, and warmed to about 70°C. A
separate mixture of 500 mg. of Gril:L 3 (Croda, U.S.), 1.25 ml Captex 355 (Abitec Corp., Janesv:ille, WI), and 1.25 ml of Labrasol (Gattefosse, France) was a:Lso prepared and warmed to about 70°C. The aqueous phase was then transferred to the oil phase with vigorous mixing to a:Eford the desired emulsion concentration of 20 mg/ml ISIS 2302. The aqueous phase of emulsion contained 2% UDCA, 4% Sodium laurate, and 4% Sodium caprate.

Formulation 9da Aliquots of the stock solution of ISIS 2302 (200 mg/ml) and mixture of penetration enhancers were mixed to afford a solution formulation comprising ISIS
2302 at a concentration of 20 mg/ml and a final concentration of 2% UDCA, 4% Sodium laurate, and 4% Sodium caprate.
Example lls Evaluation of Emulsion Formulations Containing Penetration Enhancers In order to determine the ability of penetration enhancers to improve the absorption and delivery of oligonucleotide drugs the emulsion formulations of Example 9 were administered via intrajejunum instillation and the blood concentrations of the oligonucleotide and AUC(0-3h) were measured.
Methodss 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 injection site was located (2 cm after the ligament of Treitz). An aliquot of 0.5 mL drug formulation was then injected via a 27 gauge needle. ThES intestine was put back into the body carefully. The incision portion was covered with a wet gauze. 300 ~.L of blood was collected by a 27 gauge needle from the femoral vein <~t each sampling time point.
Formulations: Two emulsions (Formulations 9a and 9c) were evaluated at a dosage leve:L of 10 mg/rat. The delivery of ISIS 2302 using these emulsions (Formulations 9a and 9c) formulated with a combination of penetration enhancers was compared to the performance of solutions (Formulations 9b and 9d, respectively) of ISIS 2302 that were formulated with the same combination and concentration of penetration enhancers.

_ 87 _ Results: The results of study are summarized in Table 17. When a control solution of ISIS 2302 (SEQ ID
N0:1) was administered no significant amount of oligonucleotides was found to be absorbed. In contrast, when ISIS 2302 was formulated as a solution that contained a mixture of fatty acid and bile salts (Formulations 9b and 9d) a significant amount of oligonucleotide was found to be absorbed and present in the systemic circulation. The concentrations of oligonucleotide in plasma were found to be in the 42-73 ~g/ml concentration range 30 mins after administration of Formulation 9b (at a dose of 10 mg/rat) which contains 2% CDCA. At the same time point 33-87 ~,g/ml of oligonucleotide was delivered into the blood circulation when Formulation 9d containing 2% UDCA was administered (at a dose of 10 mg/rat) . The AUC(0-3h) was observed to be in the 48-58 ug x h/mL range from Formulation 9b and the 30-101 ug x h/mL range from administration of Formulation 9d.
Further increases in the amount of oligonucleotides delivered into systemic circulation were observed when the emulsions containing 2% bile salt together with 4% each of sodium laurate and sodium caprate were evaluated. Thus, Formulation 9a which incorporates the bile salt CDCA afforded 34-52 ~,g/ml concentrations of total oligonucleotide in the plasma within 30 minutes of administration and an AUC(0-3h) in the 63-151 ug x h/mL
range. Likewise, Formulation 9c which incorporates the bile salt UDCA afforded 54-79 ~Cg/ml concentrations of total oligonucleotide in the plasma within 30 minutes of administration and an AUC(0-3h) in the 84-127 ~.g.h/ml range.

_ 88 TABLE 17: Plasma Coaceatrationsl and AUC(0-3h) of ISIS
2302 After Iatrajejuaum Instillation is Rats Formul-Total Time Hormul- Total atioa Coac. (h~) ation Coac.
Img/ml) (mg/ml) Animal Animal #1 #2 #3 #1 #2 #3 9a 52.2 34.0 96.04 0.5 9b 42.8 72.9 45.7 50.1 33.2 89.62 1 26.1 15.0 29.9 19.7 17.6 41.67 2 10.4 15.4 7.9 7.7 14.6 12.89 3 4.3 7.4 4.7 . ....... . _:. ....:....::::. .. ... . . ,...>......
. ...... , .: : : . -'.: k ...
.... : .~..~.~... .. .. . ....: :. > .
: . f .'x...Ø....3::: x ..
............... . ~........: ~,. >. 4 $ 1 .. . ......
. .. .>. ~3 .1.:..2.............. ::::.. 7 :'7 ..
;. .... ... .....:_.. ,.:...
. .. .... .............. ....
.. 3~ ,... :::: . :>.
....~ . ........:... ....::..... ....:: ..
~ ..~~~. . ~ ..:... ..
::: .. . .
..... .:::.
.

54-8 5 78, a.5 9x 3 8 3.7 ~ 3.3 7.3 53.4 56.6 84.6 1 17.1 63.0 18.0 17.4 32.0 2 4.2 18.6 10.6 5.1 5.0 3 0.6 5.1 k f:: .:5 .. ..k .tf : >3 <::y r::: ..r .:: 2:.
.. .. .J,.....:>
..k... 3 .>:._.
k:' , C. .
~.>.,...~........:::::
,. .
,-. ..T-:. .5 .
r '33' ..
.r. : . s;., .. : ~.
' ,,. ...:,. .::.;.>.:
. y>k,. ,:~' !,,,~ ..
. .. .> > . ,~
5:3; ...>~ .:~~ k. .5,. " -s.3;..
>..
. . '.
. x. :: ni>; . . f.3:.5.2..
:5< ~.. . ' .>
.f ... S>. k~~~.
i:: '. :.. 3: . .3.
.'.i.. . . -,. ...~. ...:>.
....:~>... ,:. ,....3. .~ .r.:
.... ~ ....y. x. ~.. i:
~,W~ . ..
.... .:.,. .. :i ,... .. . . ::. ,:......
, .. - a .. ..
;_.. .,.>...~.>. ,,:::>: r::>,..':::.....:...-.... ...
;. :.....
t : ..... .. x: .~ ..... .. .....>
.....> .. . . - ....
.............. . -.. > .....:. ... ........~~
...>,.........
~2f' .... .. . ..m>. . .........,~~...............,....>.,.....
~k.....: ..... .3,.............. .>...:
.... : :.. ...,....,.. ........
... . ... .._.......
. . ..v.. ......
.. ....
."....... ........... ...s:..,..
., .....,-..

lAs determined by HPLC analysis.
Example 11: Comparison of ISIS 2302 Bioavailability From Formulations Administered by Intrejenuaal Instillation In order to determine the effectiveness of formulations the absolute bioavailability of oligonucleotide was assessed following intrajejunal instillation.
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 injection site was located (2 cm after the ligament of Treitz). An aliquot of 0.5 mL drug formulation was then injected via a 27 gauge needle. The intestine was put back into the body carefully. The incision portion was covered with a wet gauze. 300 JCL of blood was collected by a 27 gauge needle from the femoral vein at each sampling time point. The rats were sacrificed in a carbon dioxide chamber 3 hours after dosing. Plasma samples were analyzed by HPLC.
Formulationss Five formulations were evaluated.
Two solution formulations were prepared. Formulation 9b was prepared by dissolving ISIS 2302 and a combination of CDCA
and fatty acid penetration enhancers to the desired concentrations. Formulation 9d was prepared by dissolving ISIS 2302 and a combination of UDCA and fatty acid penetration enhancers to the desired concentrations.
Formulation 6a1 was prepared as an emulsion of ISIS 2302 in a mixture of labrasol, captex and Grill 3.
Formulation 9a was also prepared as an emulsion that is similar to Formulation 6a1 with one difference in that CDCA
and fatty acid penetration enhancers were incorporated into aqueous phase of Formulation 9a at the same concentrations as were present in Formulation 9b. Formulation 9c was likewise prepared as an emulsion that is similar to Formulation 6a1 with one difference in that UDCA and fatty acid penetration enhancers were incorporated into the aqueous phase of Formulation 9c at t:he same concentrations as were present in Formulation 9d.
All five formulations contained ISIS 2302 at a final concentration of 20 mg/ml.
Results: The results of absolute bioavailability of ISIS 2302 as determined in this study are summarized in Table 18.

TABLE 18= Absolute Bioavailability of ISIS 2302 Following Intrajejunal Instillation in Rats Formulation Ca~mposition Absolute Bioavailability 9b ISIS 2302 CDCA + Fatty acids 14.6% (n 5}
+ =

solution 9a ISIS 2302 CDCA + Fatty acids 27.7% (n 3) + =

emuZs3on + Labrasol + Captex +
Grill 3 9d ISIS 2302 UDCA + Fatty acids 12.4% (n 2) + =

solut.ioa 9c ISIS 2302 UDCA + Fatty acids 27.7% (n 3) + =

emulsion + Labrasol + Captex +
Grill 3 6a1 ISIS 2302 20.3% (n 5}
+ =

emulsion Labrasol Captex + Grill3 +

When a control solution of ISIS 2302 (SEQ ID N0:1) was administered, no significant amount of oligonucleotides was found to be absorbed. In contrast, when ISIS 2302 was formulated as a solution that contained a mixture of fatty acids and a bile salt (Formulations 9b and 9d), a significant amount of oligonucleotide was found to be absorbed and bioavailable in the systemic circulation. The absolute bioavailability of ISIS 2302 was found to be 14.6%
from Formulation 9b (containing a mixture of CDCA and fatty acid penetration enhancers) and 12.4% from Formulation 9d (containing a mixture of UDCA and fatty acid penetration enhancers}. The simple emulsion, Formulation 6a1, that is devoid of any penetration enhancers was also effective in making a significant portion of the ISIS 2302 oligonucleotide bioavailable (absolute bioavailability of 20.4%} .
When formulated as emulsions that contained a combination of penetration enhancers, the bioavailability of ISIS 2302 was found to increase even further. Formulation 9a, which is an emulsion containing a combination of CDCA

and fatty acid penetration enhancers, afforded an absolute bioavailability of ISIS 2302 to the tune of 27.7%. A
similar bioavailability of the oligonucleotide was also found when emulsion Formulation 9c containing UDCA instead of CDCA, as in Formulation 9a, was Evaluated.
Example 13: Preparation of Enema Formulations To evaluate the delivery and mucosal penetration of oligonucleotides into the colon following rectal delivery, the following formulations were prepared (Table 19). Solution and emulsion formulations of ISIS 2302 (SEQ
ID NO: 1) were prepared. Additives used in the formulations included saline, hydroxypropyl methyl cellulose (HPMC), carrageenan, Vitamin E a-tocopheryl polyethyelene glycol 1000 succinate (TPGS), Tween 80 and sorbitol.
TABLE 19: ISIS 2302 Formulations 12a-12f Formulation Comyositioa 12a ISIS 2302 in Saline 12b ISIS 2302 + 1.5% Hydroxypropyl Methyl Ce llulose (HPMC) 12c ISIS 2302 + 1.0% Carrageenan + 2.5%

Vitamin E a-Tocopheryl Polyethylene Glycol 10 00 Succinate (TPGS) (Source: Eastman Chemical Company, NY ) 12d ISIS 2302 in a w/o emulsion 12e ISIS 2302 + 0.5% Tween 80 + 0.75%

HPMC

12f ISIS 2302 + 5% Sorbitol + 0.63%

HPMC

12g ISIS 2302 in water Formulation 12a: A solution of ISIS 2302 was prepared by mixing 5 ml ISIS 2302 stock solution(100 mg/mL) with 95 ml sterile saline to have a final cincentration of 5 mg/ml.
Formulation l2bs The solution was prepared by mixing 7.5 ml ISIS 2302 stock solution (100 mg/mL) with 142.5 ml hydroxypropyl methyl cellulose solution (HPMC) to have final concentrations of ISIS 2302 5 mg/ml and HPMC 15 mg/ml (1.5%). The HPMC solution was prepared by dissolving 2.25 g HPMC in 30 ml of 80°C water and Q.S. to 142.5 mL with cold water.
Formulation l2cs The solution was prepared by mixing 7.5 ml ISIS 2302 stock solution (100 mg/ml) with 142.5 ml carrageenan/Vitamine E TPG~i solution to have a final concentration of ISIS 2302 5 mg/ml, carrageenan 10 mg/ml (1%), and Vitamine E TPGS 25 mg/ml (2.5%). The carrageenan/Vitamine E TPGS solution was prepared by dissolving 1.5g carrageenan and 3.75 g Vitamine E TPGS in 142.5 ml water. The solution was then heated to 60°C to form a gel and cooled down to room temperature before the addition of ISIS 2302 stock solution.
Formulation 12d: A water-in-oil emulsion (w/o) of ISIS 2302 was prepared following the general methods in Examples 5 and 6. The emulsion containing 5 mg/ml of ISIS
2302.
Formulation 12e: The solution was prepared by mixing 6.0 ml ISIS 2302 stock solution (100 mg/ml) with 0.6 ml tween 80, and 113.4 ml HPMC solution to have final concentrations of ISIS 2302 5 mg/ml, tween 80 50 ~1/ml (0.5%), and HPMC 7.5 mg/ml (0.75%). The HPMC solution was prepared by dissolving 0.9 g HPMC in 60 ml of 80°C water and Q.S. to 113.4 ml with cool water.
Formulation 12f: The solution was prepared by mixing 6.0 ml ISIS 2302 stock solution (100 mg/ml) with 6 g sorbitol, and 108 ml HPMC solution to have final concentrations of ISIS 2302 5 mg/ml, sorbitol 50 mg/ml (5%), and HPMC 6.3 mg/ml (0.63%). The HPMC aolution was prepared WO 99!60012 PCT/US99/11394 by dissolving 0.75 g HPMC in 50 ml of 80°C water and Q.S. to 108 ml with cool water.
Formulation 12g: A solution of ISIS 2302 was prepared by mixing 5 ml ISIS 2302 stock solution(100 mg/ml) with 95 ml water to have a final concentration of 5 mg/ml Example 14: Evaluation of Enema Formulations for Local Delivery of Oligonucleotide Formulations of oligonucleotide were evaluated via rectal administration as enemas to laboratory beagle dogs.
Methods: Following a period of overnight fasting, test dogs were first administered a cleansing enema and then dosed with a sample of the test formulation. The enema formulation was applied via a Foley catheter and hold for a period of 1 h. In order to assess colonic tissue delivery and uptake of oligonucleotide, colon tissue biopsies were performed on the test animal, 3h and 24 h after dosing.
Tissue samples were processed and the amount of oligonucleotide present in the tissue assessed by capillary gel electrophoresis (CGE) and immunohistochemical (IHC) analyses.
Results: Seven formulations of ISIS 2302 (SEQ ID
N0: 1) as prepared in Example 12 (Formulations 12a-12g) were administered to dogs via rectal enemas and the local distribution of ISIS 2302 in colonic tissue was determined by CGE and IHC at 3h and 24h following dosing. The results are shown in Table 20.

TABLE 20: Local Colonic Tissue Distribution of ISIS 2302 Follov~ing Rectal Enema in Dog Formu- IIiC CGE (lug/g) lation 3 h 24 h 3 h 24 h 12a ++++ - 782 + 664 NA

12b ++++ - 660 440 6.8 5.0 12c ++++ - 558 212 2.5 + 1.4 12d ++++ - 224 78 1.2 + 0.7 12e ++++ - 62:L 368 6.0 5.9 12f ++++ - 417 + 127 1.3 + 0.5 12g ++++ - 143 NA

Note: "++++" indicates strong staining in IHC using a primary antibody to ISIS 2302; "-" indicates no significant staining compared to background levels.
Surprisingly, effective local colonic delivery of oligonucleotide was seen for a variety of formulations.
Even oligonucleotide that was merely suspended in sterile saline (Formulation 12a) or water (Formulation 12g). The presence of HPMC, a protective colloid that is useful as a general dispersing and thickening excipient, did not hinder the localized delivery of oligonucleotide (Formulation 12b), nor did other additives, such as Carrageenan and TPGS (12c) Tween 80 (12e) or Sorbitol (12f).
The present disclosure thus provides for localized delivery of oligonucleotides and other small nucleic acids to the lower portion of the G.I. tract. Such delivery can be via means of an enema using a solution comprising an effective concentration of oligonucl.eotide. Alternatively, suppositories comprising oligonucleotides suspended in an agent that disperses its contents when exposed to the physical and/or chemical conditions of the colon. In addition to HMPC, a preferred dispersing agent for localized colconic delivery of oligonucleotides is cocoa butter.
Immunohistochemical (IHC) analyses were used to confirm and extend these results by determining the histopathology and cellular localization of rectally administered oligonucleotides in does. Biopsy samples were taken 10 to 20 centimeters from (i.e~., approximately 18, 20, 21 and 22 cm from) the dorsal side of the colon and evaluated for the distribution of priosphorothioate oligodeoxynucleotide (P=S ODN) ISIS-2302 three hours after rectal administration. The biopsy samples were fixed in 10%
neutral buffered formalin for 24 hours and transferred to 70°s for storage. The tissues were embedded in paraffin and sectioned at 5 um for immunohistochemical detection of P=S
ODN. The affinity purified antibody used for this work, 2E1-B5, is a mouse IgGl (Berkeley Antibody Company, Richmond, CA) which specifically recognizes P=S ODN.
Tissues were deparaffinized and pre-treated with proteinase K (Dako Corp., Carpenteri.a, CA) for 10 minutes at room temperature prior to incubation in the primary antibody. The antibody was detected with donkey anti mouse f(ab')2 IgG conjugated to horseradish peroxidase (Jackson Laboratories, West Grove, PA) and di.aminobenzidene (DAB, Dako Corp.) was used as a substrate. All slides were stained on the Dako automated immuno-stainer.
Staining of the P=S ODN ins seen in the nucleus of the surface epithelial layer at the tips of the colonic villi in alI of the biopsies. The staining is strongest in the 20-22 cm samples and some staining is seen at the luminal surface of the epithelium, which is most likely associated with mucinous material in the colon.
Example 15: Bioavailability of Oligonucleotide Tablet Formulations: ISIS 2302 and ISIS 15839 In order to evaluate the potential for delivering oligonucleotides via various oral dosage forms, the following experiments were carried out.

A. Composition of and Preparation of Oral Dosage Formulations The following oral dosage formulations of oligonucleotides were prepared as follows.
Oral Dosage Formulation a: ISIS 2302 with penetration enhancers (CDCA, SC, SL) and excipient precirol.
Oligonucleotide (ISIS 2302) was passed through a 60 mesh screen, 12.4 g of which was then mixed with 10 g sodium chenodeoxycholate (CDCA), 20 g of sodium caprate (SC), 20 g sodium laurate (SL) and 47.5 g precirol (WL 2155 ATO, prescreened on 60 mesh). The powder was then placed in a plastic bag, mixed thoroughly and then sifted through a 20 mesh screen. Powder blend was then compressed into tablets at slight weight overage using round flat-faced tooling.
The resulting 1100 ~ 50 mg tablets contained 124 mg oligonucleotide (as is by weight), :L00 mg CDCA, 200 mg SL, 200 mg SC, and 476 mg precirol. Resultant tablets may be used as is (core tablets) or may be enteric film coated as described below under "Enteric Coating".
Oral Dosage Formulation b: ISIS 2302 with penetration enhancers (CDCA, SC, SL) and excipient (PEG).
Oligonucleotide (ISIS 2302) was passed through a 60 mesh screen and 9.3 g of which was mixed with 7.5 g sodium chenodeoxycholate (CDCA), 15 g of sodium caprate (SC), 15 g sodium laurate (SL) and :35.7 g polyethyleneglycol (20,000 mw PEG, prescreened 20 mesh:?. The powder was then placed in a plastic bag, mixed thoroughly and sifted through 20 mesh screen. Powder blend was then compressed into tablets at slight weight overage using round flat-faced tooling. The resulting 1100 ~ 50 mg tablets contained 124 mg oligonucleotide (as is by weight), 100 mg CDCA, 200 mg SL, 200 mg SC, and 476 mg PEG. Resultant tablets may be used as is (core tablets) or may be enteric film coated as described below under "Enteric Coating".
Oral Dosage Formulation c: ISIS 15839 with penetration enhancers (CDCA, SC, SL) and excipient (PEG).

_ 97 ISIS 15839 is a phosphorothioate isosequence "hemimer" derivative of ISIS 2302 having the structure 5'-GCC-CAA-GCT-GGC-ATC-CGT-CA-3' (SEQ I:D NO:1), wherein emboldened "C" residues have 5-methylcytosine (m5c) bases and wherein the emboldened, double-underlined residues further comprise a 2'-methoxyethoxy modification (other residues are 2'-deoxy). ISIS 15839 is described in co-pending U.S. Patent application Serial No. 09/062,416, filed April 17, 1998, hereby incorporated by reference.
ISIS 15839 was passed through a 60 mesh screen and 2.323 g of which was mixed with 2.0 g sodium chenodeoxycholate (CDCA), 4.0 g of sodium caprate (SC), 4.0 g sodium laurate (SL) and 9.523 g polyethyleneglycol (20,000 mw PEG, prescreened 20 mesh). The powder was then sifted through a 20 mesh screen and placed in a plastic bag and mixed thoroughly. Powder blend was then compressed into tablets at slight weight overage using 12 mm round tooling.
The resulting 728.4 ~ 10 mg tablets contained 77.44 mg oligonucleotide, 66.67 mg CDCA, 133.4 mg SL, 133.4 mg SC, and 317.5 mg PEG. Resultant tablets may be used as is (core tablets) may be enteric film coated as described below under "Enteric Coating".
Oral Dosage Formulation d: ISIS 2302 with penetration enhancers (CDCA,SC, SL) without excipient.
Oligonucleotide ISIS 2302 was passed through a 60 mesh screen and 3.72 g of which was mixed with 3 g sodium chenodeoxycholate (CDCA), 6 g of sodium caprate (SC) and 6 g sodium laurate (SL). The powder was sifted through a 20 mesh screen and placed in plastic bag and mixed thoroughly.
This powder blend was then compressed into tablets at slight weight overage using 12 mm diameter tooling. The resulting 624 ~ 10 mg tablets contained 124 m<3 oligonucleotide (as is by weight), 100 mg CDCA, 200 mg SL <~nd 200 mg SC. Resultant tablets may be used as is (core tablets) or may be enteric film coated as described below under "Enteric Coating".
Enteric Coating (EC) _ 98 _ A cellulose acetate phthalate (CAP) enteric coating solution was prepared by slowly adding 7.0 g of CAP
powder to 90.0 g stirred acetone. Before dissolving, 3.0 g diethyl phthalate was added and the solution covered with aluminum foil and continued to stir until dissolution was complete after approximately 30 minutes.
Core tablets were coated by hand by dipping into a stirred CAP solution using vacuum tubing to hold the tablet.
As the coating dried the tablets were inverted and redipped to effect completion of a single coat over entire surface.
This method may be repeated to impart an adequate enteric film coverage of 2 to 5°s weight gain and, depending on tablet size and configuration, to allow for a uniform coat thickness and performance quality.
B. Evaluation of Oral Dosage Formulations The tablet formulations were evaluated according to the following methods.
In Vitro: In order to evaluate the integrity of the enteric film coat, tablets were placed in 500 mL aqueous 0.1 N HCL acid solution (pH 1.5) using USP method II
(paddles) at 150 rpm and 37°C for up to 1 hour. Filtered samples were periodically taken and analyzed for presence of oligonucleotide as described below. Absence of oligonucleotide and visual inspection verified enteric coat integrity. Tablets were then placed into 500 mL of 0.2 M
phosphate buffer solution, pH 6.5, to evaluate the rate at which oligonucleotides were released out of the tablets.
Dissolution was monitored at regular time intervals by analyzing sample filtrate using UV light at 260 nm. This analysis was suitable for formulations devoid of interfering components (i.e., dissolved excipien.ts capable of absorbing at 260 nm). Alternatively, samples may be analyzed by any of various separation methods (e. g., HPLC).
Ia Vivo: In order to measure the bioavailability of oligonucleotides from tablet formulations, tablets were administered orally (p.o.) to healthy beagle dogs of ~12 kg average weight at an approximate dose of 15 mg/kg. Blood samples were taken at regular time intervals and plasma harvested then subsequently analyzed for presence of oligonucleotide by either high pressure liquid chromatography (HPLC)~for screening purposes or capillary gel electrophoresis (CGE) for purposes of confirmation and/or quantitation. Baseline pharmacokinetic intravenous (i.v.) data were obtained by administration of sterile drug solution (2 mg/kg) by slow i.v. push via antecubital vein followed by phlebotomies and analysis as described above.
Percent bioavailability was calculated from the resulting data according to the following formula Bioavailability = (AUCP',/Do) / (AUCi~/Do) x 100 0, wherein AUCpo is area under the plasma concentration curve for formulated oligonucleotide tableas administered orally, AUCi" is area under the plasma concentration curve for oligonucleotide administered as an i..v. solution (control), and Do is the respective dosages for these two regimens.
Results:
The results of the study are summarized in Table 21. As expected, enteric coated (EC') tablets had longer times of dissolution in acidic conditions (i.e., 4 minutes for "'50% dissolution for tablets lacking an enteric coating, from 7 to 14 minutes for EC tablets).
As indicated by the CmaX values, all of the oral dosage formulations tested can result in plasma concentrations of at least 1 ug/mL. Oral dosage formulation a, with or without an enteric coating, had CmaX values of 2.4 ug/mL and 2.0 mg/mL, respectively. A similar Cma~ value was observed with enteric coated tablet:> of oral dosage formulation c. Oral dosage formulation b had the lowest C~"al_ value (1.2 ug/mL) when no enteric coating was provided but gave the highest CmaX value (3.3 mg/mL) when an enteric coating was used.
The °sBioavailability values generally followed the trend established by the Cma,; values.. For example, Oral dosage formulation b had the lowest %Bi.oavailability value when no enteric coating was provided but gave the highest %Bioavailability value when an enteric coating was used. As with CmaX~ the remaining oral dosage formulations had %Bioavailability values that are comparable to each other.
Table als Ia Vitro Dissolution of, aad Ia V3voo Plasma Levels in Dogs Resultiag from, Oral Dosage Formulations of ISIS 2302 aad 15839 Oral Dosage Dsol ~2 ~ Bioavail-Formulatioa (min) (~Cg/mL) ability 3 a core tablets 4 2.4 2.0 b core tablets 4 1.2 0.9 a with CAP EC4 12 2.0 2.3 b with CAP EC 14 3.3 2.8 c with CAP EC 7 2.2 2.2 1 Dso - approximate time for 50% tablet: dissolution.
2 C",~,~ = maximum oligonucleotide concentration in plasma.
3 Calculated as dose normalized AUC relative to i.v. AUC.
4 CAP EC - cellulose acetate phthalates (CAP) enteric coating.
8xample 16s Use of Other Aaimal Models to Evaluate Formulations In order to further evaluate the bioavailability of the formulations of the invention, various animal models are used. For example, rectal formulations are tested in rats essentially according to the method of Aungst et aI. (Pharm.
Res., 1988, 5:305) or in rabbits essentially according to the methods of Buur et a1. (J. Control Rel., 1990, 14:43) and Yamamoto et al. (J. Pharmacol. Exper. Therapeutics, 1992, 263:25).

- iai -The formulations of the invention are further evaluated in larger animals for optimization of the penetration enhancer (PE) systems in terms of, E~.g., concentration and temporal effects on the absorption of oligonucleotides (ODN). Dogs will be "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 iliocecal junction. These respective areas provide for ideal assessment of the systemic oligonucleotide bioavailability and for local tissue (e.g., colonic) absorption. This latter objective is assessed on the basis of both tissue biopsy drug levels and/or inferred by the presence of drug in the plasma. In addition to ported dogs, naive dogs will be 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.
Dogs are dosed at 10 mg/kg of oligonucleotides, which are appropriately labeled as necessary, and blood samples are collected and evaluated for the presence and concentration of oligonucleotides. The absolute bioavailability is calculated and, if necessary, anima:Ls are sacrificed and tissue samples are collected and analyzed.
Those skilled in the art will <~ppreciate that numerous changes and modifications may be made to the preferred embodiments of the invention and that such changes and modifications may be made without departing from the spirit of the invention. It is therefore :intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention. Also, it is intended that each of the patents and patent applications referenced above be incorporated by reference herein.
A further preferred oligonucleotide modification includes 2'-dimethylamino oxyethoxy, i.e., a 0(CH~)~ON(CH~):
group, also known as 2'-DMAOE, as described in co-owned United States patent application Serial Number 09/016,520, filed on January 30, 1998, the contents of which are herein incorporated by reference. Other preferred modifications include 2'-methoxy (2'-0-CH3), 2'-aminopropoxy (2'-OCHZCHZCHZNHZ) and 2' -fluoro (2' -F) . Similar modifications may also be made at other positions on the sugar group, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide. The nucleosides of the oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
Unsubstituted and substituted phosphodiester oligonucleotides are alternately synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphorarnidite chemistry with oxidation by iodine.
Phosphorothioates are synthesized as per the phosphodiester oligonucleotides except the standard oxidation bottle was replaced by 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 thiation wait step was increased to 68 sec and was followed by the capping step. After cleavage from the CPG column and deblocking in concentrated ammonium hydroxide at 55°C (18 hr), the oligonucleotides were purified by precipitating twice with 2.5 volumes of ethanol from a 0.5 M NaCl solution.
Phosphinate oligonucleotides are prepared as described in U.S. Patent 5,508,270, herein incorporated by reference.
Alkyl phosphonate oligonucleotides are prepared as described in U.S. Patent 4,469,863, herein incorporated by reference.
3'-Deoxy-3'-methylene phosphonate oligonucleotides are prepared as described in U.S. Patents 5,610,289 or 5,625,050, herein incorporated by reference.

Phosphoramidite oligonucleotides are prepared as described in U.S. Patent, 5,256,775 or U.S. Patent 5,366,878, hereby incorporated by reference.
Alkylphosphonothioate oligonucleotides are prepared as described in published PCT applications PCT/US94/00902 and PCT/US93/06976 (published as WO 94/17093 and WO 94/02499, respectively).
3'-Deoxy-3'-amino phosphoramidate oligonucleotides are prepared as described in U.S. Patent 5,476,925, herein incorporated by reference.
Phosphotriester oligonucleotides are prepared as described in U.S. Patent 5,023,243, herein incorporated by reference.
Boranophosphate oligonucleotides are prepared as described in U.S. Patents 5,130,302 and 5,177,198, both herein incorporated by reference.
Methylenemethylimino linked oligonucleosides, also identified as MMI linked oligonucleosides, methylenedi-methylhydrazo linked oligonucleoside;s, also identified as MDH linked oligonucleosides, and methylenecarbonylamino linked oligonucleosides, also identified as amide-3 linked oligonucleosides, and methyleneaminocarbonyl linked oligo-nucleosides, also identified as amide-4 linked oligonucleo-sides, as well as mixed backbone compounds having, for instance, alternating MMI and PO or PS linkages are prepared as described in U.S. Patents 5,378,825; 5,386,023;
5,489,677; 5,602,240 and 5,610,289, all of which are herein incorporated by reference.
Formacetal and thioformacetal linked oligonucleo-sides are prepared as described in U.S. Patents 5,264,562 and 5,264,564, herein incorporated x>y reference.
Ethylene oxide linked oligonucleosides are prepared as described in U.S. Patent: 5,223,618, herein incorporated by reference.
Peptide nucleic acids (PNAs) are prepared in accordance with any of the various procedures referred to in Peptide Nucleic Acids (PNA): Synthesis, Properties and Potential Applications, Bioorganic & Medicinal Chemistry, 1996, 4, 5. They may also be prepared in accordance with U.S. Patents 5,539,082; 5,700,922, and 5,719,262, herein incorporated by reference.
A further preferred oligonucleotide modification includes 2'-dimethylamino oxyethoxy, i.e., a O(CHZ)~ON(CH3)~
group, also known as 2'-DMAOE, as described in co-owned United States patent application Serial Number 09/016,520, filed on January 30, 1998, the contents of which are herein incorporated by reference. Other preferred modifications include 2'-methoxy (2'-O-CH3), 2'-aminopropoxy (2'-OCHZCHZCHZNH~) and 2' -fluoro (2' -F) . Similar modifications may also be made at other positions on the sugar group, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligon.ucleotides and the 5' position of 5' terminal nucleotide. The nucleosides of the oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
Unsubstituted and substituted phosphodiester oligonucleotides are alternately synthesized on an automated DNA synthesizer (Applied Biosystems model 3808) using standard phosphoramidite chemistry with oxidation by iodine.
Phosphorothioates are synthesized as per the phosphodiester oligonucleotides except the standard oxidation bottle was replaced by 0.2 M solution of 3H-1,2-benzodithiole-3-one 1,1-dioxide in a.cetonitrile for the stepwise thiation of the phosphite linkages. The thiation wait step was increased to 68 sec anal was followed by the capping step. After cleavage from the CPG column and deblocking in concentrated ammonium hydroxide at 55°C (18 hr), the oligonucleotides were purified by precipitating twice with 2.5 volumes of ethanol from a 0.5 M NaCl solution.
Phosphinate oligonucleotides are prepared as described in U.S. Patent 5,508,270, herein incorporated by reference.

Alkyl phosphonate oligonuc~leotides are prepared as described in U.S. Patent 4,469,863, herein incorporated by reference.
3'-Deoxy-3'-methylene phosphonate oligonucleotides are prepared as described in U.S. Pa.tents 5,610,289 or 5,625,050, herein incorporated by reference.
Phosphoramidite oligonucleotides are prepared as described in U.S. Patent, 5,256,775 or U.S. Patent 5,366,878, hereby incorporated by reference.
Alkylphosphonothioate oligonucleotides are prepared as described in published F'CT applications PCT/US94/00902 and PCT/US93/06976 (published as WO 94/17093 and WO 94/02499, respectively).
3'-Deoxy-3'-amino phosphoramidate oligonucleotides are prepared as described in U.S. Patent 5,476,925, herein incorporated by reference.
Phosphotriester oligonucleotides are prepared as described in U.S. Patent 5,023,243, herein incorporated by reference.
Boranophosphate oligonucleotides are prepared as described in U.S. Patents 5,130,302 and 5,177,198, both herein incorporated by reference.
Methylenemethylimino linked oligonucleosides, also identified as MMI linked oligonucleosides, methylenedi-methylhydrazo linked oligonucleosides, also identified as MDH linked oligonucleosides, and methylenecarbonylamino linked oligonucleosides, also identified as amide-3 linked oligonucleosides, and methyleneaminocarbonyl linked oligo-nucleosides, also identified as amide-4 linked oligonucleo-sides, as well as mixed backbone compounds having, for instance, alternating MMI and PO or PS linkages are prepared as described in U.S. Patents 5,378,825; 5,386,023;
5,489,677; 5,602,240 and 5,610,289, all of which are herein incorporated by reference.
Formacetal and thioformacetal linked oligonucleo-sides are prepared as described in tT.S. Patents 5,264,562 and 5,264,564, herein incorporated x>y reference.

Ethylene oxide linked oligonucleosides are prepared as described in U.S. Patent: 5,223,618, herein incorporated by reference.
Peptide nucleic acids (PNAs) are prepared in accordance with any of the various procedures referred to in Peptide Nucleic Acids (PNA): Synthesis, Properties and Potential Applications, B.ioorganic 2K Medicinal Chemistry, 1996, 4, 5. They may also be prepared in accordance with U.S. Patents 5,539,082; 5,700,922, and 5,719,262, herein incorporated by reference.
Those skilled in the art will appreciate that numerous changes and modifications may be made to the preferred embodiments of the invention and that such changes and modifications may be made without departing from the spirit of the invention. It is therefore intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention.
It is intended that each of the patents, applications, printed publications, and other published documents mentioned or referred to in this specification be herein incorporated by reference in their entirety.

WO 99/60012 PCT/US99l11394 SEQUENCE FISTING
<110> ISIS Pharmaceuticals, Inc, et al.
<120> Compositions and Methods for Non-parenteral Delivery of Oligonucleotides <130>
<140>
<141>
<150> U.S. serial no. 09/082,624 <151> 1998-05-21 <160> 54 <210> 1 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 1 gcccaagctg gcatccgtca 20 <210> 2 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 2 cccccaccac ttcccctctc 20 <210> 3 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 3 agccatagcg aggctgaggt t 21 <210> 4 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 4 aacatctccg taccatgcca 20 <210> 5 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 5 cccaggcatt ttaagttgct g 21 <210> 6 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 6 gtttaaggca gcatcctaag a 21 <210> 7 <211> 21 <212> DNA
<213> Artificial Sequence <220>

<223> antisense sequence <400> 7 tcacccaaag gtttaggctt g 21 <210> 8 <211> 21 <212> DNA

<213> Artificial Sequence <220>
<223> antisense sequence <400> 8 gcaatcatga cttcaagagt t 21 <210> 9 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 9 gtgccggggt 18 cttcgggc <210> 10 <211> 19 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 10 catggt ttcg gagggcgtc 19 <210> 11 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 11 tcgcgc tccc tctctccggc 20 <210> 12 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 12 cacccaagag 20 agcagaaagt <210> 13 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 13 cccttcctac 2'0 cgcgtgcgac <210> 14 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 14 cctccgaccc 20 atccacgtag <210> 15 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 15 gttgacgtcc 20 tacggaaaca <210> 16 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 16 tccgtcatcg ctcctcaggg 20 <210> 17 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 17 tgctgttcgt 20 gcccccgccg <210> 18 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 18 ctaaggcaca 20 aggcgggctg <210> 19 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 19 tcccgcctgt gacatgcatt 20 <210> 20 <211> 25 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 20 cctctctgtt taaaacttta tccat 25 <210> 21 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 21 ttcatatcct gagtcatgtc g 21 <210> 22 <211> 18 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 22 gcuauuaccu uaacccag 18 <210> 23 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 23 cauuauugcc cugaaag 17 <210> 24 <211> 21 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 24 taaaaagaat 21 atgatcttca t <210> 25 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 25 agcaactgag ccacctga 18 <210> 26 <211> 21 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 26 gccatagggg gcagggaagg c 21 <210> 27 <211> 25 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 27 ctctcgcacc 25 catctctctc cttct <210> 28 <211> 26 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 28 ctctcgcacc 26 catctctctc cttcta <210> 29 <211> 26 _ 7 _ <212> DNA
<213> Artificial Sequence <220>

<223> antisense sequence <400> 29 gctctcgcac 26 ccatctctct ccttct <210> 30 <211> 17 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 30 gtggtgggtg 17 ggtgggt <210> 31 <211> 26 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 31 gcctattctg ctatgtcgac acccaa 26 <210> 32 <211> 26 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 32 cttcgggcct 26 gtcgggtccc ctcggg <210> 33 <211> 26 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 33 gctggtgatc ctttccatcc ctgtgg 26 <210> 34 <211> 26 _ g _ <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 34 ctactactcc ttgactttgg ggattg 26 <210> 35 <211> 26 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 35 cttcgggcct 26 gtcgggtccc ctcggg <210> 36 <211> 26 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 36 cuucgggccu 26 gucggguccc cucggg <210> 37 <211> 8 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 37 ttggggtt 8 <210> 38 <211> 20 <212> DNA

<213> Artificial Sequence <220>
<223> antisense sequence <400> 38 gtgctcatgg tgcacggtct 20 <210> 39 <211> 20 _ g _ <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 39 cattcaaatg gtttgcctgc 20 <210> 40 <211> 20 <212> DNA
<213> Artificial Sequence <220>

<223> antisense sequence <400> 40 gcaggcaaac 20 catttgaatg <210> 41 <211> 25 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 41 tttgggtcca 25 tcatcttcag caaag <210> 42 <211> 20 , <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 42 catcatcttc agcaaagata 20 <210> 43 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 43 acgcgaaaaa 20 atgcgtacaa <210> 44 <211> 20 WO 99!60012 PCT/US99I11394 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 44 taaaccaaaa aaatggggca 20 <210> 45 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 45 tggggcttac 20 cttgcgaaca <210> 46 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 46 gacgtggggc 20 ttaccttgcg <210> 47 <211> 20 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 47 tcttcaacga cgtggggctt 20 <210> 48 <211> 21 <212> DNA

<213> Artificial Sequence <220>

<223> antisense sequence <400> 48 gcgtttgctc ttcttcttgc g 21 <210> 49 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 49 gttctcgctg gtgagtttca 20 <210> 50 <211> 15 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 50 aacttgtgct tgctc 15 <210> 51 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 51 gtgctcatgg tgcacggtct 20 <210> 52 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 52 gtgtgccaga caccctatct 20 <210> 53 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> antisense sequence <400> 53 gctgattaga gagaggtccc 20 <210> 54 <211> 20 <212> DNA
<2I3> Artificial Sequence <220>
<223> antisense sequence <400> 54 ttgcttccat cttcctcgtc 20

Claims

What is claimed is:
1. A pharmaceutical composition comprising at least one oligonucleotide in an emulsion.
2. The pharmaceutical composition of claim 1 wherein said oligonucleotide is an antisense oligonucleotide.
3. The pharmaceutical composition of claim 1 wherein said oligonucleotide modulates expression of a cellular adhesion protein, modulates a rate of cellular proliferation, or has biological activity against eukaryotic pathogens or retroviruses.
4. The pharmaceutical composition of claim 1 wherein said oligonucleotide is a ribozyme, a peptide nucleic acid, a molecular decoy, an external guide sequence or an aptamer.
5. The pharmaceutical composition of claim 1 wherein said emulsion is an oil-in-water emulsion, a water-in-oil emulsion, an oil-in-water-in-oil emulsion or a water-in-oil-in-water emulsion.
6. The pharmaceutical composition of claim 1 wherein said emulsion is a microemulsion.
7. The pharmaceutical composition of claim 6 wherein said microemulsion is an oil-in-water microemulsion, a water-in-oil microemulsion, an oil-in-water-in-oil microemulsion or a water-in-oil-in-water microemulsion.
8. The pharmaceutical composition of claim 1 further comprising at least one penetration enhancer.

9. The pharmaceutical composition of claim 8 wherein said penetration enhancer is a fatty acid.
10. The pharmaceutical composition of claim 9 wherein said fatty acid is selected from a group consisting of 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 pharmaceutically acceptable salt thereof.
11. The pharmaceutical composition of claim 8 wherein said penetration enhancer is a bile salt.
12. The pharmaceutical composition of claim 11 wherein said bile salt is selected from a group consisting of 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 pharmaceutically acceptable salt thereof.
13. The pharmaceutical composition of claim 8 wherein said penetration enhancer is a combination of at least one fatty acid and at least one bile salt.
14. The pharmaceutical composition of claim 8 wherein said penetration enhancer is a chelating agent.
15. The pharmaceutical composition of claim 14 wherein said chelating agent is selected from a group consisting of EDTA, citric acid, a salicyclate, a N-acyl derivative of collagen, laureth-9, an N-amino aryl derivative of a beta-diketone or a mixture thereof.

15. The pharmaceutical composition of claim 8 wherein said penetration enhancer is a surfactant.
17. The pharmaceutical composition of claim 16 wherein said surfactant is selected from a group consisting of sodium lauryl sulfate, polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether, a perfluorchemical emulsion or a mixture thereof.
18. The pharmaceutical composition of claim 8 wherein said penetration enhancer is selected from a group consisting of unsaturated cyclic ureas, 1-alkyl-alkanones, 1-alkenylazacyclo-alakanones, steroidal anti-inflammatory agents and mixture thereof.
19. The pharmaceutical composition of claim 1 further comprising at least one carrier compound.
20. The pharmaceutical composition of claim 19 wherein said carrier compound is selected from a group consisting of polyinosinic acid, dextran sulfate, polycytidic acid, lipofectin, cationic glycerol derivatives, polylysine and 4-acetamido-4'isothiocyano-stilbene-2,2'-disulfonic acid.
21. A method of treating an animal comprising administering to said animal a therapeutically effective amount of a pharmaceutical composition according to claim 1.
22. The method of claim 21 wherein said administration is buccal, sublingual, endoscopic, rectal, oral, vaginal, topical, pulmonary ox urethral.
23. The method of claim 22 wherein said administration is rectal.

24. The method of claim 23 wherein said rectal administration is by means of an enema.
25. The method of claim 23 wherein said rectal administration is by means of a suppository.
26. The method of claim 25 wherein suppository includes cocoa butter or hydroxpropylmethylcellulose.
27. The method of claim 21 wherein said animal is known or suspected to suffer from ulcerative colitis.
28. The method of claim 21 wherein said animal is known or suspected to suffer from Chrohn's disease.
29. The method of claim 21 wherein said animal is known or suspected to suffer from inflammatory bowel disease.
30. The method of claim 21 wherein said animal is known or suspected to suffer from undue cellular proliferation.
31. A rectal enema comprising an oligonucleotide in a solution.
32. The rectal enema of claim 31 wherein said solution is a saline solution.
33. The rectal enema of claim 31 wherein said solution is a buffered solution.
34. A method of treating an animal comprising administering to said animal a therapeutically effective amount of the rectal enema of claim 31.

35. The method of claim 34 wherein said animal is known or suspected to suffer from ulcerative colitis.
36. The method of claim 34 wherein said animal is known or suspected to suffer from Chrohn's disease.
37. The method of claim 34 wherein said animal is known or suspected to suffer from inflammatory bowel disease.
38. The method of claim 34 wherein said animal is known or suspected to suffer from undue cellular proliferation.
39. A rectal suppository comprising an oligonucleotide and an excipient.
40. The rectal suppository of claim 31 wherein said excipient comprises cocoa butter or hydroxpropylmethylcellulose.
41. A method of treating an animal comprising administering to said animal a therapeutically effective amount of the rectal suppository of claim 39.
42. The method of claim 41 wherein said animal is known or suspected to suffer from ulcerative colitis.
43. The method of claim 41 wherein said animal is known or suspected to suffer from Chrohn's disease.
44. The method of claim 41 wherein said animal is known or suspected to suffer from inflammatory bowel disease.

45. The method of claim 41 wherein said animal is known or suspected to suffer from undue cellular proliferation.
46. A pharmaceutical composition comprising an oligonucleotide in oral dosage form.
47. The pharmaceutical composition of claim 46 wherein at least one of said covalent linkages of said oligonucleotide is a modified covalent linkage.
48. The pharmaceutical composition of claim 47 wherein said modified covalent linkage is selected from the group consisting of a phosphorothioate linkage, a phosphotriester linkage, a methyl phosphonate linkage, a methylene(methylimino) linkage, a morpholino linkage, an amide linkage, a polyamide linkage, a short chain alkyl intersugar linkage, a cycloalkyl intersugar linkage, a short chain heteroatomic intersugar linkage and a heterocyclic intersugar linkage.
49. The pharmaceutical composition of claim 46 wherein at least one of the nucleotides of said oligonucleotide has a modified sugar moiety.
50. The pharmaceutical composition of claim 49 wherein said modified sugar moiety has a substitution or addition at the 2' position of a moiety selected from the group consisting of -OH, -SH, -SCH3, -F, -OCN, -OCH3OCH3, -OCH3O(CH2)n CH3, -O(CH2)n NH2 or -O(CH2)n CH3 where n is from 1 to about 10, a C1 to C10 lower alkyl group, an alkoxyalkoxy group, a substituted lower alkyl group, a substituted alkaryl group, a substituted aralkyl group, -Cl, -Br, -CN, -CF3, -OCF3, an -O-alkyl group, an -S-alkyl group, an N-alkyl group, an O-alkenyl group, an S-alkenyl group, an N-alkenyl group, -SOCH3, -SO2CH3, -ONO2, -NO2, -N3, -NH2, a heterocycloalkyl group, a heterocycloalkaryl group, an aminoalkylamino group, a polyalkylamino group, a substituted silyl group, an RNA cleaving group, a reporter group, a DNA
intercalating group, a group for improving the pharmacokinetic properties of an oligonucleotide, a group for improving the pharmacodynamic properties of an oligonucleotide, a methoxyethoxy group and a methoxy group.
51. The pharmaceutical composition of claim 46 wherein at least one of the nucleotides of said oligonucleotide has a modified nucleobase.
52. The pharmaceutical composition of claim 46 wherein said oral dosage form is selected from the group consisting of tablets, capsules and gel capsules.
53. The pharmaceutical composition of claim 46 wherein said oligonucleotide is an antisense oligonucleotide.
54. The pharmaceutical composition of claim 46 wherein said oligonucleotide modulates expression of a cellular adhesion protein, modulates a rate of cellular proliferation, or has biological activity against eukaryotic pathogens or retroviruses.
55. The pharmaceutical composition of claim 46 wherein said nucleic acid is a ribozyme, a peptide nucleic acid, an external guide sequence, a molecular decoy or an aptamer.
56. The pharmaceutical composition of claim 46 further comprising an enteric material that substantially prevents dissolution of said tablets, capsules or gel capsules in a mammalian stomach.
57. The pharmaceutical composition of claim 56 wherein said enteric material is a coating.

58. The pharmaceutical composition of claim 57 wherein said enteric coating is acetate phthalate, propylene glycol, sorbitan monoleate, cellulose acetate trimellitate, hydroxy propyl methyl cellulose phthalate or cellulose acetate phthalate.
59. The pharmaceutical composition of claim 46 further comprising a penetration enhancer.
60. The pharmaceutical composition of claim 59 wherein said penetration enhancer is selected from the group consisting of bile salts and fatty acids.
61. The pharmaceutical composition of claim 60 wherein said bile salt is selected from ursodeoxycholic acid, chenodeoxycholic acid, and salts thereof.
62. The pharmaceutical composition of claim 60 wherein said fatty acids are selected from capric acid, lauric acid, and salts thereof.
63. The pharmaceutical composition of claim 46 further comprising an excipient.
64. The pharmaceutical composition of claim 63 wherein said excipient is selected from the group consisting of polyethyleneglycol and precirol.
65. The pharmaceutical composition of claim 46 further comprising a plasticizer.
66. The pharmaceutical composition of claim 65 wherein said plasticizer is diethyl phthalate, triacetin, dibutyl sebacate, dibutyl phthalate or triethyl citrate.

67. A method of treating an animal comprising administering to said animal a therapeutically effective amount of the pharmaceutical composition of claim 46.
68. The method of claim 67 wherein said animal is known or suspected to suffer from ulcerative colitis.
69. The method of claim 67 wherein said animal is known or suspected to suffer from Chrohn's disease.
70. The method of claim 67 wherein said animal is known or suspected to suffer from inflammatory bowel disease.
71. The method of claim 67 wherein said animal is known or suspected to suffer from undue cellular proliferation.
72. A method of modulating the expression of a gene in an animal comprising administering to said animal the pharmaceutical composition of claim 1.
73. A method of modulating the expression of a gene in an animal comprising administering to said animal the rectal enema of claim 31.
74. A method of modulating the expression of a gene in an animal comprising administering to said animal the rectal suppository of claim 39.
75. A method of modulating the expression of a gene in an animal comprising administering to said animal the pharmaceutical composition of claim 46.
76. A composition for oral administration comprising an oligonucleotide and a carrier wherein said oligonucleotide is conjugated to folic acid.

77. The composition according to claim 76 wherein the folic acid is conjugated to the 5' terminus of said oligonucleotide.
78. The composition according to claim 77 further comprising an enteric substance resistant to degradation by gastric acids.
79. A method for introducing an intact oligonucleotide into a mammal, the method comprising the step of orally administering an oligonucleotide, wherein said oligonucleotide comprises a folic acid conjugated thereto.
80. The pharmaceutical composition according to claim 1, wherein said oligonucleotide is selected from the group consisting of ISIS-2302, ISIS-15839, ISIS-1939, ISIS-2922, ISIS-13312, ISIS-3521, ISIS-9605, ISIS-9606, ISIS-14859, ISIS-2503, ISIS-5132, ISIS-14803, ISIS-28089, ISIS-104838 and ISIS-2105.
81. The rectal enema according to claim 31, wherein said oligonucleotide is selected from the group consisting of ISIS-2302, ISIS-15839, ISIS-1939, ISIS-2922, ISIS-13312, ISIS-3521, ISIS-9605, ISIS-9606, ISIS-14859, ISIS-2503, ISIS-5132, ISIS-14803, ISIS-28089, ISIS-104838 and ISIS-2105.
82. The rectal suppository according to claim 39, wherein said oligonucleotide is selected from the group consisting of ISIS-2302, ISIS-15839, ISIS-1939, ISIS-2922, ISIS-13312, ISIS-3521, ISIS-9605, ISIS-9606, ISIS-14859, ISIS-2503, ISIS-5132, ISIS-14803, ISIS-28089, ISIS-104838 and ISIS-2105.
83. The pharmaceutical composition according to claim 46, wherein said oligonucleotide is selected from the group consisting of ISIS-2302, ISIS-15839, ISIS-1939, ISIS-2922, ISIS-13312, ISIS-3521, ISIS-9605, ISIS-9606, ISIS-14859, ISIS-2503, ISIS-5132, ISIS-14803, ISIS-28089, ISIS-104838 and ISIS-2105.
CA002329130A 1998-05-21 1999-05-20 Compositions and methods for non-parenteral delivery of oligonucleotides Abandoned CA2329130A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US8262498A 1998-05-21 1998-05-21
US09/082,624 1998-05-21
PCT/US1999/011394 WO1999060012A1 (en) 1998-05-21 1999-05-20 Compositions and methods for non-parenteral delivery of oligonucleotides

Publications (1)

Publication Number Publication Date
CA2329130A1 true CA2329130A1 (en) 1999-11-25

Family

ID=22172335

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002329130A Abandoned CA2329130A1 (en) 1998-05-21 1999-05-20 Compositions and methods for non-parenteral delivery of oligonucleotides

Country Status (6)

Country Link
US (1) US8377897B2 (en)
EP (2) EP1469009A2 (en)
JP (1) JP2003524586A (en)
AU (1) AU745880B2 (en)
CA (1) CA2329130A1 (en)
WO (1) WO1999060012A1 (en)

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU731909B2 (en) 1997-07-01 2001-04-05 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
AU745880B2 (en) 1998-05-21 2002-04-11 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US20030036520A1 (en) * 1999-07-23 2003-02-20 Jack Gauldie Intestinal gene therapy
US20020155084A1 (en) * 2000-06-02 2002-10-24 The Regents Of The University Of The Michigan Nanoemulsion formulations
JP2004537516A (en) * 2001-05-11 2004-12-16 オラセンス リミテッド Antisense penetration enhancer
US20030124196A1 (en) * 2001-08-22 2003-07-03 Susan Weinbach Pulsatile release compositions and methods for enhanced intestinal drug absorption
US20030083286A1 (en) * 2001-08-22 2003-05-01 Ching-Leou Teng Bioadhesive compositions and methods for enhanced intestinal drug absorption
CN100438908C (en) * 2001-10-06 2008-12-03 梅瑞尔有限公司 CpG formulations and related methods
ITRM20030149A1 (en) 2003-04-02 2004-10-03 Giuliani Spa ANTISENSE OLIGONUCLEOTIDES (ODN) FOR SMAD7 AND THEIR USE IN THE MEDICAL FIELD
EP1687410A4 (en) * 2003-10-07 2008-04-09 Isis Pharmaceuticals Inc Antisense oligonucleotides optimized for kidney targeting
JP2005119986A (en) * 2003-10-15 2005-05-12 Japan Science & Technology Agency Method for administering physiologically active substance, physiologically active substance-containing emulsion, and method for producing physiologically active substance-containing emulsion
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
AU2005209242A1 (en) * 2004-01-26 2005-08-11 Psivida Inc. Controlled and sustained delivery of nucleic acid-based therapeutic agents
DE102004038535B4 (en) * 2004-08-06 2012-10-04 Universitätsklinikum Schleswig-Holstein Cellular introduction of nucleic acid agents
US20060069055A1 (en) * 2004-09-21 2006-03-30 Maya Dajee Delivery of polynucleotides
FR2878746B1 (en) * 2004-12-02 2007-02-23 Vetoquinol Sa Sa NOVEL PHARMACEUTICAL COMPOSITION FOR USE IN THE FIELD OF VACCINES
GB0505081D0 (en) * 2005-03-14 2005-04-20 Genomica Sau Downregulation of interleukin-12 expression by means of rnai technology
US8017159B2 (en) * 2005-11-16 2011-09-13 Idexx Laboratories, Inc. Phospholipid gel compositions for delivery of aptamers and methods of treating conditions using same
US8530436B2 (en) * 2007-01-29 2013-09-10 Transderm, Inc. Methods and compositions for transdermal delivery of nucleotides
US8637478B2 (en) 2007-11-13 2014-01-28 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
ATE503485T1 (en) 2007-11-14 2011-04-15 Novagali Pharma Sa COMPOSITION WITH AT LEAST ONE NUCLEOSIDE PORTION AS A CURE AND CKC
EP2105145A1 (en) * 2008-03-27 2009-09-30 ETH Zürich Method for muscle-specific delivery lipid-conjugated oligonucleotides
US10138485B2 (en) 2008-09-22 2018-11-27 Rxi Pharmaceuticals Corporation Neutral nanotransporters
LT2364360T (en) 2008-11-13 2017-08-10 Nogra Pharma Limited Antisense compositions and methods of making and using same
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
KR102175230B1 (en) * 2009-09-11 2020-11-06 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of huntingtin expression
EP2488210A4 (en) 2009-10-12 2014-04-30 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
EP2550000A4 (en) 2010-03-24 2014-03-26 Advirna Inc Reduced size self-delivering rnai compounds
EP2606910B1 (en) * 2010-08-20 2018-10-17 National University Corporation Tokyo Medical and Dental University Pharmaceutical composition for transcolonic absorption
WO2014110225A1 (en) * 2013-01-09 2014-07-17 The Feinstein Institute For Medical Research Hmgb1-binding beads and uses thereof
ES2675215T3 (en) 2013-02-08 2018-07-09 General Mills, Inc. Sodium Reduced Food Products
CN105492011A (en) 2013-04-08 2016-04-13 丹尼斯·M·布朗 Therapeutic benefit of suboptimally administered chemical compounds
US10934550B2 (en) 2013-12-02 2021-03-02 Phio Pharmaceuticals Corp. Immunotherapy of cancer
US9587268B2 (en) * 2014-01-29 2017-03-07 Agilent Technologies Inc. Fast hybridization for next generation sequencing target enrichment
CA2982175A1 (en) 2015-04-08 2016-10-13 Carl Magnus SCHOELLHAMMER Systems, apparatus, and assembling method thereof for administering a substance
GB201700257D0 (en) * 2017-01-06 2017-02-22 Atlantic Pharmaceuticals (Holdings) Ltd New formulation
EP3612152A4 (en) * 2017-04-19 2021-02-17 Phio Pharmaceuticals Corp. Topical delivery of nucleic acid compounds
GB201807312D0 (en) * 2018-05-03 2018-06-20 Index Pharmaceuticals Ab Formulation
US20210106525A1 (en) 2019-10-11 2021-04-15 Massachusetts Institute Of Technology Formulations for gastrointestinal delivery of oligonucleotides

Family Cites Families (226)

* 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
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
US5258506A (en) * 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
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
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5185444A (en) * 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5166315A (en) * 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
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
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
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
DE3851889T2 (en) 1987-06-24 1995-04-13 Florey Howard Inst NUCLEOSIDE DERIVATIVES.
US5585481A (en) * 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
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
US5188897A (en) * 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
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
DE3855864T2 (en) * 1987-11-30 1997-09-25 Univ Iowa Res Found DNA MOLECULES STABILIZED BY MODIFICATIONS ON THE 3'-TERMINAL PHOSPHODIESTERBINDING, THEIR USE AS NUCLEIC ACID PROBE AND AS A THERAPEUTIC AGENT FOR INHIBITING THE EXPRESSION OF SPECIFIC TARGET 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
US5264564A (en) * 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
EP0497875B1 (en) * 1989-10-24 2000-03-22 Isis Pharmaceuticals, Inc. 2' modified oligonucleotides
US5264562A (en) * 1989-10-24 1993-11-23 Gilead Sciences, Inc. 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
US5514788A (en) * 1993-05-17 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5623065A (en) * 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5591623A (en) * 1990-08-14 1997-01-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5457191A (en) * 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5587361A (en) * 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5212295A (en) * 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
US5587470A (en) * 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5859221A (en) * 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5459255A (en) * 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5506351A (en) * 1992-07-23 1996-04-09 Isis Pharmaceuticals Process for the preparation of 2'-O-alkyl guanosine and related compounds
US5681941A (en) * 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5578718A (en) * 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5955589A (en) 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
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
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
US5214136A (en) * 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
AU7579991A (en) * 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
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
US5166195A (en) * 1990-05-11 1992-11-24 Isis Pharmaceuticals, Inc. Antisense inhibitors of the human immunodeficiency virus phosphorothioate oligonucleotides
ES2116977T3 (en) * 1990-05-11 1998-08-01 Microprobe Corp SOLID SUPPORTS FOR NUCLEIC ACID HYBRIDIZATION TESTS AND METHODS TO IMMOBILIZE OLIGONUCLEOTIDES IN A COVALENT WAY.
US5135917A (en) 1990-07-12 1992-08-04 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
DE69126530T2 (en) * 1990-07-27 1998-02-05 Isis Pharmaceuticals Inc NUCLEASE RESISTANT, PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECT AND MODULE GENE EXPRESSION
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
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
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
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
US5677437A (en) * 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5610289A (en) * 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
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
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5623070A (en) * 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5245022A (en) * 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
EP0541722B1 (en) * 1990-08-03 1995-12-20 Sterling Winthrop 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
US6111094A (en) 1990-08-14 2000-08-29 Isis Pharmaceuticals Inc. Enhanced antisense modulation of ICAM-1
US5843738A (en) * 1990-08-14 1998-12-01 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5595978A (en) * 1990-08-16 1997-01-21 Isis Pharmaceuticals, Inc. Composition and method for treatment of CMV retinites
US5177196A (en) * 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
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
US5596086A (en) * 1990-09-20 1997-01-21 Gilead Sciences, Inc. Modified internucleoside linkages having one nitrogen and two carbon atoms
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
WO1992008728A1 (en) * 1990-11-08 1992-05-29 Hybridon, Inc. Incorporation of multiple reporter groups on 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
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
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
AU2169992A (en) * 1991-05-31 1993-01-08 Genta Incorporated Compositions and delivery systems for transdermal administration of neutral oligomers
AU655839B2 (en) * 1991-06-27 1995-01-12 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
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
US5359044A (en) * 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
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
US5565552A (en) * 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
US5595726A (en) * 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
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
US5633360A (en) * 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
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
AU673193B2 (en) * 1992-09-02 1996-10-31 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
CA2105595A1 (en) * 1992-09-23 1994-03-24 Ramaswamy Narayanan Antisense polynucleotides
US5523389A (en) 1992-09-29 1996-06-04 Isis Pharmaceuticals, Inc. Inhibitors of human immunodeficiency virus
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
US5476925A (en) * 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
WO1994018835A1 (en) * 1993-02-19 1994-09-01 Genta Incorporated Treatment of androgen-associated baldness using antisense oligomers
CA2156288C (en) * 1993-02-19 2005-10-18 Junichi Yano Glycerol derivative, device and pharmaceutical composition
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
ATE155467T1 (en) * 1993-03-30 1997-08-15 Sanofi Sa ACYCLIC NUCLEOSIDE ANALOGUES AND OLIGONUCLEOTIDE SEQUENCES CONTAINING THEM
JPH08508491A (en) 1993-03-31 1996-09-10 スターリング ウインスロップ インコーポレイティド Oligonucleotides with phosphodiester bonds replaced by amide bonds
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
ATE193892T1 (en) * 1993-06-29 2000-06-15 Ferring Bv SYNTHESIS OF CYCLIC PEPTIDES
WO1996010996A1 (en) * 1993-07-21 1996-04-18 The University Of Kentucky Research Foundation A multicompartment hard capsule with control release properties
US5571902A (en) * 1993-07-29 1996-11-05 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
DE69433036T2 (en) * 1993-09-03 2004-05-27 Isis Pharmaceuticals, Inc., Carlsbad AMINODERIVATIZED 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
JPH09510182A (en) 1993-11-17 1997-10-14 エルディーエス・テクノロジーズ・インコーポレーテッド Encapsulated transparent liquid for drug delivery
US5457187A (en) * 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
JP3585238B2 (en) * 1993-12-09 2004-11-04 トーマス ジェファーソン ユニバーシティー Compounds and methods for site-directed mutagenesis in eukaryotic cells
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
US5597696A (en) * 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5580731A (en) * 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) * 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
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
US5955059A (en) 1995-06-06 1999-09-21 Trustees Of Boston University Use of locally applied DNA fragments
US5591091A (en) * 1995-08-03 1997-01-07 Hackman; Lloyd E. Method of matching a golfer to a golf club
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
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
AU702030B2 (en) * 1995-10-12 1999-02-11 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
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
GB9613858D0 (en) 1996-07-02 1996-09-04 Cortecs Ltd Hydrophobic preparations
CA2259179C (en) * 1996-07-03 2008-09-23 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
AU731909B2 (en) 1997-07-01 2001-04-05 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
PT1003476E (en) 1997-08-11 2005-05-31 Alza Corp ACTIVE AGGREGATE AGGREGATE DOSAGE FORM ADAPTED FOR GASTRIC RETENTION
US5877309A (en) * 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
WO1999016454A1 (en) * 1997-09-26 1999-04-08 Medeva Europe Limited 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
AU745880B2 (en) 1998-05-21 2002-04-11 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
JP2002515514A (en) * 1998-05-21 2002-05-28 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 local delivery of oligonucleotides
US6080580A (en) 1998-10-05 2000-06-27 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of tumor necrosis factor-α (TNF-α) expression
US6228642B1 (en) 1998-10-05 2001-05-08 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
EP1080103A4 (en) 2003-07-02
US20070249551A1 (en) 2007-10-25
AU4095599A (en) 1999-12-06
EP1469009A2 (en) 2004-10-20
WO1999060012A1 (en) 1999-11-25
JP2003524586A (en) 2003-08-19
AU745880B2 (en) 2002-04-11
US8377897B2 (en) 2013-02-19
EP1080103A1 (en) 2001-03-07

Similar Documents

Publication Publication Date Title
AU745880B2 (en) Compositions and methods for non-parenteral delivery of oligonucleotides
US8691785B2 (en) Compositions and methods for non-parenteral delivery of oligonucleotides
AU2002329825B2 (en) Bioadhesive compositions and methods for enhanced mucosal drug absorption
US7576067B2 (en) Pulsatile release compositions and methods for enhanced intestinal oligonucleotide drug absorption
AU2002329825A1 (en) Bioadhesive compositions and methods for enhanced mucosal drug absorption
CA2329252A1 (en) Compositions and methods for topical delivery of oligonucleotides
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
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
FZDE Discontinued