EP0724647A4 - Acides nucleiques servant de ligands et procedes de production ameliores - Google Patents

Acides nucleiques servant de ligands et procedes de production ameliores

Info

Publication number
EP0724647A4
EP0724647A4 EP94927409A EP94927409A EP0724647A4 EP 0724647 A4 EP0724647 A4 EP 0724647A4 EP 94927409 A EP94927409 A EP 94927409A EP 94927409 A EP94927409 A EP 94927409A EP 0724647 A4 EP0724647 A4 EP 0724647A4
Authority
EP
European Patent Office
Prior art keywords
ligand
nucleic acid
seq
rna
ligands
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.)
Ceased
Application number
EP94927409A
Other languages
German (de)
English (en)
Other versions
EP0724647A1 (fr
Inventor
Larry Gold
Wolfgang Pieken
Diane Tasset
Nebojsa Janjic
Gary P Kirschenheuter
Barry Polisky
Sumedha Jayasena
Greg Biesecker
Drew Smith
Robert D Jenison
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.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/199,507 external-priority patent/US5472841A/en
Priority claimed from US08/233,012 external-priority patent/US5849479A/en
Priority claimed from US08/234,997 external-priority patent/US5683867A/en
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Priority to EP06006842A priority Critical patent/EP1793006A3/fr
Publication of EP0724647A1 publication Critical patent/EP0724647A1/fr
Publication of EP0724647A4 publication Critical patent/EP0724647A4/fr
Ceased legal-status Critical Current

Links

Classifications

    • 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
    • C12N15/115Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/001Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1048SELEX
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6811Selection methods for production or design of target specific oligonucleotides or binding molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/535Production of labelled immunochemicals with enzyme label or co-enzymes, co-factors, enzyme inhibitors or enzyme substrates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/16Aptamers
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/81Protease inhibitors
    • G01N2333/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • G01N2333/811Serine protease (E.C. 3.4.21) inhibitors
    • G01N2333/8121Serpins
    • G01N2333/8125Alpha-1-antitrypsin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96433Serine endopeptidases (3.4.21)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96433Serine endopeptidases (3.4.21)
    • G01N2333/96436Granzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96486Metalloendopeptidases (3.4.24)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/966Elastase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/974Thrombin

Definitions

  • VEGF vascular endothelial growth factor
  • SELEX an acronym for Systematic Evolution of Ligands by Exponential enrichment .
  • counter-SELEX methods for identifying highly specific nucleic acid ligands able to discriminate between closely related molecules
  • methods for combining nucleic acids with other functional units for generation of high affinity ligands termed “blended SELEX” and methods for preparing modified oligonucleotides capable of binding target molecules with high affinity.
  • modified oligonucleotides of the present invention contain one or more modified nucleotide bases, which include 5-X and/or 2' -Y substitutions in pyrimidine bases and 8-X and/or 2'Y substitutions in purine bases.
  • the invention includes nuclease-resistant oligonucleotide ligands containing the modified nucleotides of the present invention.
  • the invention further includes methods for synthesizing the substituted nucleotides, bases and intermediates described herein.
  • the oligonucleotides of the present invention are modified by incorporation of chemically-modified nucleotide derivatives.
  • the nucleotide derivatives incorporated into the oligonucleotides of the present invention also introduce means for incorporating additional functional groups into the nucleic acid ligands.
  • This invention includes modified high affinity nucleic acid ligands which are single-stranded DNA and RNA ligands.
  • oligonucleotides containing nucleotide derivatives chemically modified at the 5- and 2'- positions of pyrimidine. Further disclosed are specific RNA ligands to thrombin containing 2' -NH 2 -modifications.
  • the modified oligonucleotides of the present invention increase the chemical diversity of the candidate mixture for the SELEX process, producing improved nucleic acid ligands to specific target molecules. In many cases, the modifications also provide the oligonucleotide with increased relative resistance to endonucleases in serum.
  • the modified oligonucleotides of the present invention are useful as pharmaceuticals, diagnostic agents, and as part of gene therapy treatments.
  • the blended nucleic acid ligands of the present invention consist of at least one nucleic acid ligand unit and at least one functional unit.
  • functional units that may be coupled to nucleic acids include proteins, peptides, photoreactive groups, chemically-reactive groups, active site directed compounds, lipids, biotin and fluorescent compounds.
  • the nucleic acid ligand unit (s) of the blended nucleic acid ligand serve in whole or in part as ligands to a given target.
  • the functional unit(s) can be designed to serve in a large variety of functions.
  • the functional unit may independently or in combination with the nucleic acid ligand have specific affinity for the target, and in some cases may be a ligand to a different site of interaction with the target than the nucleic acid ligand.
  • the functional unit (s) may be added which covalently react and couple the ligand to the target molecule, catalytic groups may be added to aid in the selection of protease or nuclease activity, and reporter molecules such as biotin- or fluorescence-tagged oligonucleotides may be added for use as diagnostic reagents.
  • SELEX SELEX
  • the SELEX process provides a class of products which are referred to as nucleic acid ligands, such ligands having a unique sequence, and which have the property of binding specifically to a desired target compound or molecule.
  • Each SELEX-identified nucleic acid ligand is a specific ligand of a given target compound or molecule.
  • SELEX is based on the unique insight that nucleic acids have sufficient capacity for forming a variety of two- and three-dimensional structures and sufficient chemical versatility available within their monomers to act as ligands (form specific binding pairs) with virtually any chemical compound, whether monomeric or polymeric. Molecules of any size can serve as targets.
  • the SELEX method involves selection from a mixture of candidates and step-wise iterations of binding, partitioning, and amplification, using the same general selection theme, to achieve virtually any desired criterion of binding affinity and selectivity.
  • the method includes steps of contacting the mixture with the target under conditions favorable for binding, partitioning unbound nucleic acids from those nucleic acids which have bound to target molecules, dissociating the nucleic acid- arget pairs, amplifying the nucleic acids dissociated from the nucleic acid-target pairs to yield a ligand-enriched mixture of nucleic acids, then reiterating the steps of binding, partitioning, dissociating and amplifying through as many cycles as desired.
  • a variety of techniques can be used to partition members in the pool of nucleic acids that have a higher affinity to the target than the bulk of the nucleic acids in the mixture.
  • SELEX is based on the inventors' insight that within a nucleic acid mixture containing a large number of possible sequences and structures there is a wide range of binding affinities for a given target.
  • a nucleic acid mixture comprising, for example, a 20 nucleotide randomized segment, can have 4 20 candidate possibilities. Those which have the higher affinity constants for the target are most likely to bind to the target.
  • a second nucleic acid mixture is generated, enriched for the higher binding affinity candidates. Additional rounds of selection progressively favor the best ligands until the resulting nucleic acid mixture is predominantly composed of only one or a few sequences.
  • nucleic acids of the test mixture preferably include a randomized sequence portion as well as conserved sequences necessary for efficient amplification. Nucleic acid sequence variants can be produced in a number of ways including synthesis of randomized nucleic acid sequences and size selection from randomly cleaved cellular nucleic acids.
  • variable sequence portion may contain a fully or partially random sequence; it may also contain subportions of conserved sequence incorporated with randomized sequence. Sequence variation in test nucleic acids can be introduced or increased by mutagenesis before or during the selection/partition/amplification iterations.
  • the selection process is so efficient at isolating those nucleic acid ligands that bind most strongly to the selected target, that only one cycle of selection and amplification is required.
  • Such an efficient selection may occur, for example, in a chromatographic-type process wherein the ability of nucleic acids to associate with targets bound on a column operates in such a manner that the column is sufficiently able to allow separation and isolation of the highest affinity nucleic acid ligands.
  • the target-specific nucleic acid ligand solution may include a family of nucleic acid structures or motifs that have a number of conserved sequences and a number of sequences which can be substituted or added without significantly affecting the affinity of the nucleic acid ligands to the target.
  • nucleic acid primary, secondary and tertiary structures are known to exist .
  • the structures or motifs that have been shown most commonly to be involved in non-Watson-Crick type interactions are referred to as hairpin loops, symmetric and asymmetric bulges, pseudoknots and myriad combinations of the same.
  • Almost all known cases of such motifs suggest that they can be formed in a nucleic acid sequence of no more than 30 nucleotides. For this reason, it is often preferred that SELEX procedures with contiguous randomized segments be initiated with nucleic acid sequences containing a randomized segment of between about 20-50 nucleotides.
  • the basic SELEX method may be modified to achieve specific objectives. For example, U.S. Patent Application Serial No. 07/960,093, filed October 14,
  • the SELEX Patent Applications describe methods for obtaining nucleic acid ligands that bind to more than one site on the target molecule, and to nucleic acid ligands that include non-nucleic acid species that bind to specific sites on the target.
  • the SELEX method provides means for isolating and identifying nucleic acid ligands which bind to any envisionable target.
  • the SELEX method is applied to situations where the target is a protein, including both nucleic acid-binding proteins and proteins not known to bind nucleic acids as part of their biological function.
  • RNA ligand inhibitors of bFGF discloses RNA ligand inhibitors of bFGF and U.S. Patent Application Serial No. 07/973,333, filed November 6, 1992, entitled Ligands of Thrombin, herein specifically incorporated by reference, describes nucleic acid ligands to thrombin.
  • Basic fibroblast growth factor is a multifunctional effector for many cells of mesenchymal and neuroectodermal origin (Rifkin & Moscatelli (1989) J. Cell Biol. 109:1; Baird & Bohlen (1991) in Peptide Growth Factors and Their Receptors (Sporn, M. B. & Roberts, A. B., eds . ) ; pp. 369-418, Springer, N.Y. ; Basilico & Moscatelli (1992) Adv. Cancer Res. 5_9:115) . It is one of the most studied and best characterized members of a family of related proteins that also includes acidic FGF (Jaye et al .
  • it is one of the most potent inducers of neovascularization.
  • angiogenic activity in vivo suggests a role in tissue remodeling and wound healing, as well as, in some disease states that are characterized by pathological neovascularization such as tumor proliferation, tumor metastasis, diabetic retinopathy and rheumatoid arthritis (Folkman & Klagsbrun (1987) Science 115:442; Gospodarowitz (1991) Cell Biology Reviews 15:307) .
  • bFGF is also known to play a key role in the development of smooth-muscle cell lesions following vascular injury (Reidy et al . Circulation, Suppl. Ill
  • heparin preparation is heterogeneous with respect to size, degree of sulfonation and iduronic acid content. Additionally, heparin also affects many enzymes and growth factors. Thus, apart from monoclonal antibodies, specific antagonists of bFGF are not known.
  • Thrombin is a multifunctional serine protease that has important procoagulant and anticoagulant activities.
  • thrombin cleaves fibrinogen, activates clotting factors V, VIII, and XIII, and activates platelets.
  • the specific cleavage of fibrinogen by thrombin initiates the polymerization of fibrin monomers, a primary event in blood clot formation.
  • the central event in the formation of platelet thrombi is the activation of platelets from the "nonbinding" to the "binding" mode and thrombin is the most potent physiologic activator of platelet aggregation (Berndt and Phillips (1981) in Platelets in Biology and Pathology, J.L. Gordon, ed.
  • thrombin plays a key role in the arrest of bleeding (physiologic hemostasis) and formation of vasoocclusive thrombi (pathologic thrombosis) .
  • thrombomodulin a glycoprotein expressed on the surface of vascular endothelial cells.
  • TM alters substrate specificity from fibrinogen and platelets to protein C through a combination of an allosteric change in the active site conformation and an overlap of the TM and fibrinogen binding sites on thrombin.
  • Activated protein C in the presence of a phospholipid surface, Ca 2+ , and a second vitamin K-dependent protein cofactor, protein S, inhibits coagulation by proteolytically degrading factors Va and Villa.
  • the formation of the thrombin-TM complex converts thrombin from a procoagulant to an anticoagulant enzyme, and the normal balance between these opposing activities is critical to the regulation of hemostasis. It is therefore of interest to produce a high affinity nucleic acid ligand of thrombin capable of inhibiting its anticoagulant activity.
  • Integrin gpllbllla is a protein expressed on activated platelets which mediate platelet adhesion to fibrinogen and fibrin clots (Phillips et al . (1988) Blood 11:831; Frojmovic et al . (1991) Blood 21:369) .
  • Members of the integrin superfamily of cell adhesion receptors, including gpllbllla, are known to recognize the peptide arginine-glycine-aspartic acid sequence (RGD) .
  • RGD arginine-glycine-aspartic acid sequence
  • Inhibitors of gpllbllla-mediated platelet clot formation may have therapeutic potential in a variety of vascular diseases including reducing the occurrence of heart attacks following angioplasty.
  • elastase Human neutrophil elastase
  • elastase Human neutrophil elastase
  • Elastase is a serine protease with broad substrate specificity able to digest many macromolecules found in connective tissues.
  • elastase can hydrolyze macromolecules such as elastin, type III and type IV collagen, and fibronectin.
  • Elastase is a single-chain glycoprotein 218 amino acids in length (Sinha et al . (1987) Proc. Natl. Acad. Sci. USA 14:2228) . Elastase has two N-glycosylation sites at positions Asn-95 and Asn-144. Its molecular weight is about 29,500 daltons, and its isoelectric point (pi) lies between 8-9.
  • the crystal structure of elastase complexed with an inhibitor has been determined by Navia et al . (1989) Proc. Natl. Acad. Sci.
  • SLPI Secretory leucocyte proteinase inhibitor
  • elafin another natural elastase inhibitor, was characterized from human skin (Wiedow et al . (1990) J. Biol. Chem. 26_5:14791) .
  • An excess of elastase activity has been implicated in several diseases, including pulmonary emphysema
  • elastase-specific inhibitors have been a major goal in the pharmaceutical industry for some time. As a result, different types of inhibitors have been developed. These include irreversible inhibitors such as peptide chloromethyl ketones (Powers et al . (1977) Biochim. Biophys. Acta 484 :156) , reversible inhibitors such as peptide boronic acids (Stone et al . (1990) Am. Rev. Respir. Dis. 141 :47) , cephalosporins (Doherty et al . (1986) Nature 322 :192) , and peptide aldehydes (Kennedy et al . (1987) Eur. J. Respir.
  • irreversible inhibitors such as peptide chloromethyl ketones (Powers et al . (1977) Biochim. Biophys. Acta 484 :156)
  • reversible inhibitors such as peptide boronic acids (Stone e
  • biosynthetically derived naturally occurring inhibitors such as ⁇ ;-l-proteinase (Gadek et al . (1981) J. Clin. Invest. £1:1158) , eglin C (Snider et al . (1985) Am. Rev. Respir. Dis. 131:1155) , and SLPI
  • RNA homopolymers Synthetic RNA homopolymers (Simon et al . (1988) Exp. Lung Res. 14 . :85) , tRNAs and
  • DNAs (Lestienne & Bieth (1983) Biochimie £5:49) have been shown to inhibit elastase to some extent .
  • the enzyme inhibition caused by these polyanions is not likely due to simple electrostatic interactions, because other polyanions lacking hydrophobic constituents such as heparin and polyanionic polysaccharides have been shown to be ineffective inhibitors.
  • Cell extracts of certain pneumococcal species (Pneumococci type I, type II smooth, and type II rough) yield high molecular weight RNAs upon autolysis that act as elastase inhibitors (Vered et al . (1988) Exp. Lung Res. 14:67) .
  • VEGF Vascular endothelial growth factor
  • bFGF vascular endothelial growth factor
  • Neovascularization or angiogenesis is the process in which sprouting new blood vessels are formed from the existing endothelium in response to external stimuli that signal inadequate blood supply.
  • Angiogenesis is generally rare under normal physiological conditions, but frequently accompanies certain pathological conditions such as psoriasis, rheumatoid arthritis, hemangioma, solid tumor growth and metastasis (Folkman S_ Klagsbrun, 1987) Science 235, 442- 447; Kim et al . , (1993) Nature 362, 841-844) .
  • fibroblast growth factors aFGF, bFGF
  • TGF ⁇ , TGF ⁇ transforming growth factors c_ and ⁇
  • PDGF platelet derived growth factor
  • angiogenin platelet-derived endothelial cell growth factor
  • IL-8 interleukin-8
  • VEGF vascular endothelial growth factor
  • VEGF was originally purified from guinea pig ascites and tumor cell cultures as a factor that increases vascular permeability (Senger, D.R. et al . (1983) , Science 219 :983-985) and it has therefore also been referred to as vascular permeability factor (VPF) .
  • VEGF is a heat and acid-stable, disulfide-linked homodimer. Four isoforms have been described (121, 165, 189 and 206 amino acids, respectively) and are believed to be the result of alternative splicing of mRNA.
  • VEGF vascular endothelial growth factor
  • the predominant VEGF isoform in most cells and tissues is the 165 amino acid species.
  • VEGF is typically glycosylated, glycosylation is required only for efficient secretion, not for activity (Yeo, T-.K. et al . (1991) Biochem. Biophys. Res. Commun. 179:1568-1575; Peretz, D. et al . (1992) Biochem. Biophys. Res. Commun.
  • VEGF vascular endothelial growth factor
  • fltl Three high-affinity receptors for VEGF have been cloned to date: fltl, kdr/flk-1 and flt4 (Vaisman, N. et al . (1990) J. Biol. Chem. 265:19461-19466; de Vries, C. et al . (1992) Science 255 :989-991; Galland, F. et al . (1993) Oncogene 1:1233-1240) . These receptors belong to a family of transmembrane tyrosine kinases and bind VEGF with dissociation constants between 10 "11 M to 10 "12 M.
  • VEGF is also known to induce the proteolytic enzymes interstitial collagenase, urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) (Unemori E. et al . (1993) J. Biol. Chem. in press; Pepper, M.S. et al . (1992) Biochem. Biophys. Res. Commun. 181:902) . These enzymes are known to play a prominent role in angiogenesis-related extracellular matrix degradation.
  • uPA urokinase-type plasminogen activator
  • tPA tissue-type plasminogen activator
  • VEGF may be one of the major angiogenesis inducers in vivo.
  • angiogenesis associated with (i) embryonic development (Breier, G. et al . (1992) Development 114 :521-532) ; (ii) hormonally-regulated reproductive cycle; and (iii) tumor growth (Dvorak, H.F. (1991) J. Exp. Med. 174:1275-1278; Shweiki, D. et al .
  • VEGF plays a key role in hypoxia-induced angiogenesis.
  • Theophylline (1, 3-dimethylxanthine) ( Figure 47) is a naturally occurring alkaloid that is widely used as an effective bronchodilator in the treatment of asthma, bronchitis, and emphysema (Hendeles and Weinberger (1983) Pharmacotherapy 1:2) . Because of its narrow therapeutic index, serum levels must be monitored carefully to avoid serious toxicity.
  • Theophylline is closely related structurally to caffeine (1, 3 , 7-trimethylxanthine) ( Figure 47) and theobromine (3, 7-dimethylxanthine) , both of which are often present in serum samples. Analytical diagnostic techniques utilizing spectroscopic characteristics or immunological reagents must therefore discriminate efficiently among these and other alkaloids (Jolley et al . (1981) Clin. Chem. 17:1575; Broussard
  • Native oligonucleotides are sensitive to degradation by nucleases .
  • Two kinds of ribonucleases are known. The first, termed exonucleases, degrade an oligoribonucleotide sequentially from either the 3'- or the 5' -end. Exonucleases cleave the phosphodiester chain through catalyzing direct hydrolysis, with water as the attacking agent . The more prevailing mode of degradation of oligoribonucleotides proceeds through catalysis by endonucleases . Endonucleases cleave RNA within the chain, 3' to the specific base they recognize.
  • the mechanism of cleavage involves activation of the 2'- hydroxyl (2' -OH) to attack the phosphorous of the internucleotidic linkage (Saenger (1984) in: Principles of Nucleic Acid Structure, Springer Verlag, New York, p. 174) .
  • This initial step leads to chain cleavage with formation of the 2',3'-cyclic phosphate end on the 5'- product and a free 5' -OH end on the 3' -product .
  • the major degradation of oligoribonucleotides in serum proceeds through pyrimidine-specific endonuclease (Pieken et al . (1990) Science 251:314) .
  • RNAse A pancreatic ribonuclease
  • Acids Res. H:239 also described the synthesis of mixed oligonucleotide sequences containing 2'-OCH 3 at every nucleotide unit.
  • the stability of oligoribonucleotides against endonuclease degradation may be achieved by replacement of the 2' -OH group of the ribose moiety with an alternate substituent such as an amino group or a fluoro group (Pieken et al . (1990) Science 251:314) .
  • Both 2'-amino and 2'-fluoro nucleoside 5-triphosphates are substrates for T7 RNA polymerase, albeit with somewhat decreased incorporation efficiency (Aurup et al . (1992) Biochemistry 11:9636) .
  • Other 2' -substituted nucleotides such as 2'-0-methyl, 2'-0-alkyl, or 2'-deoxy nucleoside 5-triphosphates are not recognized as substrates by T7 RNA polymerase.
  • the present invention includes methods for identifying and producing nucleic acid ligands using the
  • the invention also includes the nucleic acid ligands so identified and produced.
  • the SELEX method described above allows for identification of a single nucleic acid ligand or a family of nucleic acid ligands to a given target.
  • the methods of the present invention allow for the analysis of the nucleic acid ligand or family of nucleic acid ligands obtained by SELEX in order to identify and produce improved nucleic acid ligands.
  • nucleic acid ligands are desired for their ability to inhibit one or more of the biological activities of the target . In such cases, methods are provided for determining whether the nucleic acid ligand effectively inhibits the desired biological activity.
  • the present invention further includes methods for producing high affinity nucleic acid ligands which incorporate chemically-modified nucleotides.
  • SELEX is performed with a candidate mixture of oligonucleotides containing modified nucleotides.
  • SELEX is performed with a candidate mixture of oligonucleotides not containing modified nucleotides and the selected high affinity ligands are subsequently modified by incorporation of modified nucleotides. Incorporation of modified nucleotides into oligonucleotides provides means for introducing additional functional groups into the nucleic acid ligands via the modified nucleotides.
  • the method of the present invention provides increased enrichment of the chemical diversity of a nucleic acid candidate mixture from which ligands to specific targets are identified through the SELEX process. Further, the method of the present invention provides nucleic acid ligands with increased in vivo stability relative to the non-modified ligand.
  • the present invention includes oligonucleotides containing one or more modified bases.
  • the modified pyrimidine bases of the present invention have substitutions of the general formula 5-X and/or 2'-Y, and the modified purine bases have modifications of the general formula 8-X and/or 2'-Y.
  • the group X includes the halogens I, Br, Cl, or an azide or amino group.
  • the group Y includes an amino group, fluoro, or a methoxy group as shown in Figure 1. Other functional substitutions that would serve the same function are also included.
  • the oligonucleotide ligands of the present invention may have one or more X-modified bases, or one or more Y- modified bases, or a combination of X- and Y-modified bases.
  • the present invention encompasses derivatives of these substituted pyrimidines and purines such as 5'- triphosphates, and 5' -dimethoxytrityl, 3' -b-cyanoethyl, N, N-diisopropyl phosphoramidites with isobutyryl protected bases in the case of adenosine and guanosine, or acyl protection in the case of cytosine.
  • oligonucleotides bearing any of the nucleotide analogs herein disclosed.
  • the present invention encompasses specific nucleotide analogs modified at the 5 and 2' positions, including 5- (3-aminoallyl)uridine triphosphate (5-AA-UTP) , 5- (3- aminoallyl) deoxyuridine triphosphate (5-AA-dUTP) , 5- fluorescein-12-uridine triphosphate (5-F-12-UTP) , 5- digoxygenin-11-uridine triphosphate (5-Dig-ll-UTP) , 5- bromouridine triphosphate (5-Br-UTP) , 2' -amino-uridine triphosphate (2' -NH 2 -UTP) , 2' -amino-cytidine triphosphate (2'-NH 2 -CTP) , 2' -fluoro-cytidine triphosphate (2'-F-CTP) and 2' -fluro-uridine triphosphat
  • nuclease-resistant oligonucleotide ligands containing the modified nucleotides of the present invention.
  • the present invention further includes a method for generating blended nucleic acid ligands comprised of functional unit(s) added to provide a nucleic acid ligand with additional functions.
  • the functional unit provides additional affinity or a desired effect such as inhibition or induction between the blended nucleic acid ligand and the target molecule.
  • This method for combining nucleic acids with other functional groups to use in molecular evolution is herein referred to as blended SELEX.
  • the method of this invention provides novel means for generating nucleic acid ligands with specifically selected functionalities.
  • high affinity ligands are generated by the method of this invention which are highly specific inhibitors of a target enzyme.
  • the present invention encompasses nucleic acid ligands coupled to a non-nucleic acid functional unit.
  • a peptide- conjugated nucleotide was produced by coupling the peptide Gly-Arg-Gly-Asp-Thr-Pro (SEQ ID NO:40) to the derivatized base 5- (3-aminoallyl) -uridine triphosphate (RGD-UTP) .
  • RGD-UTP containing oligonucleotides (RGD-RNA) were then generated by the method of this invention and shown to bind the RGD-binding protein integrin gpllbllla.
  • RGD-RNA is a highly specific inhibitor of gpllbllla.
  • blended nucleic acid ligands produced by the method of this invention a blended nucleic acid ligand to elastase with the ability to specifically inhibit elastase activity was generated.
  • An inhibitory peptide was coupled to a single-stranded DNA ligand to elastase and the blended nucleic acid ligand shown to specifically inhibit elastase activity.
  • RNA ligands to VEGF, identified through the SELEX method. These ligands were selected from an initial pool of about 10 14 RNA molecules randomized at thirty contiguous positions. The evolved RNA ligands bind VEGF with affinities in the low nanomolar range. Also included herein are modified RNA ligands to
  • modified RNA ligands may be prepared after the identification of 2' -OH RNA ligands or by performing SELEX using a candidate mixture of modified RNAs.
  • 2'-NH 2 pyrimidine RNA ligands to VEGF are described herein.
  • the present invention includes the method of identifying nucleic acid ligands and ligand sequences to VEGF comprising the steps of a) contacting a candidate mixture of nucleic acids with VEGF; b) partitioning between members of said candidate mixture on the basis of affinity to VEGF; and c) amplifying the increased affinity nucleic acids to yield a mixture of nucleic acids enriched for nucleic acid sequences with relatively higher affinity for binding to VEGF. Also included herein are nucleic acid ligands to human neutrophil elastase identified through the SELEX procedure.
  • This invention includes 2'-NH 2 RNA and single-stranded DNA ligands to elastase, specifically the 2' -NH 2 RNA sequences shown in Table 4 and the DNA sequences shown in Table 9. Also included are RNA and DNA ligands to elastase identified through the SELEX method that are substantially homologous to those shown in Tables 4 and 7 and that have substantially the same ability to bind elastase. This invention also includes nucleic acid ligands which inhibit the biological activity of elastase.
  • nucleic acid ligands and ligand sequences to elastase comprising the steps of a) contacting a candidate mixture of single-stranded nucleic acids with elastase; b) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; and c) amplifying the increased affinity nucleic acids to yield a ligand-enriched mixture of nucleic acids, whereby nucleic acid ligands to elastase may be identified.
  • the present invention further includes methods for identifying and producing nucleic acid ligands of theophylline and caffeine, and the nucleic acid ligands so identified and produced.
  • Nucleic acid sequences identified through the SELEX process are provided that are ligands of theophylline.
  • RNA sequences are provided that are capable of binding with high affinity to theophylline. Included within the invention are the nucleic acid ligand sequences shown in Figure 48 (TR8 and TCT8) (SEQ ID
  • RNA ligands to theophylline identified through the SELEX method that are substantially homologous to those shown in Figure 48 and that have substantially the same ability to bind theophylline.
  • Nucleic acid sequences identified through the SELEX process are provided that are ligands of caffeine. Specifically, RNA sequences are provided that are capable of binding with high affinity to caffeine. Included within the invention are the nucleic acid ligand sequences shown in Figure 54 (CR8) (SEQ ID NOS:16-27) . Also included are RNA ligands to caffeine identified through the SELEX method that are substantially homologous to those shown in Figure 54 and that have substantially the same ability to bind caffeine.
  • Counter-SELEX is a method for improving the specificity of nucleic acid ligands to a target molecule by eliminating nucleic acid ligand sequences with cross-reactivity to one or more non-target molecules.
  • Counter-SELEX is comprised of the steps of (a) contacting a candidate mixture with a target molecule, wherein nucleic acids having an increased affinity to the target relative to the candidate mixture may be partitioned from the remainder of the candidate mixture; b) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; c) contacting the increased affinity nucleic acids with one or more non-target molecules such that nucleic acid ligands with specific affinity for the non-target molecule (s) are removed; and d) amplifying the nucleic acids with specific affinity to the target molecule to yield a mixture of nucleic acids enriched for nucleic acid sequences with a relatively higher affinity and specificity for binding to the target molecule.
  • the present invention includes the nucleic acid ligands to theophylline identified according to the above-described counter-SELEX method, including those ligands listed in Figure 48 (TCT8) . Also included are nucleic acid ligands to theophylline that are substantially homologous to any of the given ligands and that have substantially the same ability to bind theophylline.
  • FIGURE 1 illustrates some of the 2'- and 5- substituted nucleosides which may be utilized by the method of the present invention.
  • FIGURE 2 illustrates the structures of 5-allylamino- UTP (5-AA-UTP) , 5-bromo-UTP (5-Br-UTP) , 5-fluorescein-12- UTP (5-F-12-UTP) , 5-digoxygenin-ll-UTP (5-Dig-ll-UTP) .
  • FIGURE 3 shows the autoradiogram of a 15% denaturing polyacrylamide gel for transcription of an 87-mer of defined sequence. Four time points are 0.5, 1.0, 2.0, and 3.0 hours .
  • FIGURE 4 shows examples of side chains on 5-AA-UTP
  • RNA SELEX for RNA SELEX
  • 5-AA-dUTP for DNA SELEX
  • FIGURE 5 shows how a variety of functional groups can be introduced into the primary amine group of 5-AA- UTP and 5-AA-dUTP by the carbodiimide/N-hydroxy succinimide coupling.
  • FIGURE 6 depicts nucleotide sequences of RNA ligands isolated by SELEX for human thrombin. Each sequence is divided into 3 blocks from left to right: 1) the 5' fixed region, 2) the 30 base pair (30N) variable region, and 3) the 3' fixed region.
  • Figure 6A depicts 2' -OH RNA ligands separated into class I and II.
  • Figure 6B depicts 2 ' -NH 2 RNA ligands separated into groups I, II, and III.
  • FIGURE 7 shows proposed secondary structures of RNA ligands.
  • Figure 7A shows the sequence of the class I 2'- OH RNA clone 16 and of the class II 2' -OH RNA clone 27.
  • Figure 7B shows the sequence of the group I 2 ' -NH 2 RNA clone 32, the group II sub A 2'-NH 2 RNA clone 37 and sub B clone 17, and the group III 2' -NH 2 RNA clone 29.
  • FIGURE 8 depicts binding curves for thrombin ligands.
  • Figure 8A shows thrombin binding curves for nonselected 30N3 RNA (•) , and for the 2' -OH RNA ligand class I clone 16 (0) and class II clone 27 (x) .
  • Figure 8B shows the thrombin binding curves for 2'-NH 2 -30N3 (nonselected) (•) and for 2 ' -NH 2 RNA ligand clones 32, 37 , and 17 ( 0) .
  • FIGURE 9 illustrates the four step synthesis of 5- iodo-2' -amino,2' -deoxyuridine.
  • FIGURE 10 shows the synthesis of 2' -amino, 2'- deoxyuridine 5' -triphosphate.
  • FIGURE 11 shows the plasma kinetic curves obtained for each of 5 experimental rats injected with 32 P labelled RNA ligand for thrombin. Blood samples were collected at 1, 2, 4, 7, and 24 hours.
  • FIGURE 12 illustrates the structure of 5- (3- aminoallyl) -uridine triphosphate (AA-UTP) and the synthetic scheme for production of Arg-Gly-Asp-UTP (RGD- UTP) . Details of the reaction conditions are as described in Example 8.
  • FIGURE 13A shows the isolation of succinyl-UTP by
  • FIGURE 13B shows the fractions versus the optical density at 290 nm. As described in Example 8, succinyl-UTP eluted in fractions 43-49.
  • FIGURE 14A shows the isolation of RGD-UTP by Mono Q anion exchange column chromatography.
  • FIGURE 14B shows the fractions versus the optical density at 290 nm. As described in Example 8, RGD-UTP eluted in fractions 35- 38.
  • FIGURE 15 shows the results of thin layer chromatography of aminoallyl-UTP, succinyl-UTP, and RGD- UTP.
  • FIGURE 16 shows the results of the RGD-UTP RNA transcript purification by anion exchange chromatography. The elution of RGD-UTP RNA (D) and RNA ( ⁇ ) from a Mono Q anion exchange column is shown.
  • FIGURE 17 shows the separation of RGD-30n7 RNA and gpllbllla by size exclusion chromatography on Superdex 200 (Pharmacia) .
  • the elution profiles of RGD-30n7 bound to gpllbllla (D) and RGD-30n7 ( ⁇ ) are shown.
  • FIGURE 18 shows the chemistry of the attachment of the inhibitory substrate peptide N-methoxysuccinyl Ala- Ala-Pro-Val-chloromethyl (SEQ ID NO:41) ketone to a high affinity single-stranded DNA (DNA-17) .
  • FIGURE 19 shows the inactivation of 1.95 mM N- methoxysuccinyl Ala-Ala-Pro-Val-chloromethyl (SEQ ID NO:41) ketone by 50 mM DTT.
  • FIGURE 20 shows the inhibition of the blended nucleic acid ligand in the presence of 20 mM DTT.
  • FIGURE 21 shows the inhibition of elastase by DNA-17 conjugated at the 5' end with chloromethyl ketone (O) , and DNA-17 conjugated at the 3' end with chloromethyl ketone (#) .
  • FIGURE 22 is a Lineweaver-Burk plot of the inhibition of elastase by N-methoxysuccinyl Ala-Ala-Pro- Val (SEQ ID NO:44) (t) , peptide-conjugated oligonucleotide ( ⁇ ) and substrate alone (O) .
  • FIGURE 23 shows the inhibition of elastase by DNA ligands with and without the inhibitors conjugated at the 3' end compared with the effect of conjugated and non- conjugated ligands on urokinase and thrombin.
  • FIGURE 24 depicts the valyl phosphonate moiety attached to a nucleic acid segment as used in Example 11 below, and the reaction of the species with human neutrophil elastase.
  • FIGURE 25 depicts the first order rate constant of a pool of splint blended SELEX nucleic acid ligands after five rounds of SELEX measured by gel electrophoresis; TBE pH 8 (I) and MAE pH 6 (•) .
  • FIGURE 26 depicts the starting RNA and PCR primers used in the VEGF SELEX experiment described in Examples 12 and 13.
  • FIGURE 27 depicts the aligned sequences and predicted secondary structures for the six families (grouped by primary sequence homology) of RNA ligands to VEGF. Arrows underline the inverted repeats of the double stranded (stem) regions. Lowercase and uppercase letters are used to distinguish nucleotides in the constant and the evolved sequence regions, respectively. Positions are numbered consecutively starting (arbitrarily) with the evolved nucleotide closest to the 5' end of the shown window.
  • FIGURE 28 shows the consensus sequences and predicted secondary structures for certain of the VEGF ligand families. Plain text is used to designate positions that occur at >60% but ⁇ 80% frequencies. Positions where individual nucleotides are strongly conserved (frequencies >80%) are outlined. Residues in parenthesis occur at that position with equal frequencies to gaps. The numbering system described in the legend to Figure 27 is used.
  • FIGURE 29 depicts the binding curves for a representative set of high-affinity ligands to VEGF.
  • Full-length (o) and truncated ( ⁇ ) ligands tested were 100 (SEQ ID NO:55) and lOOt (SEQ ID NO:95) (family 1, Figure 29A) , 44 (SEQ ID NO:64) and 44t (SEQ ID NO:96) (family 2, Figure 29B) , 12 (SEQ ID NO:66) (SEQ ID NO: ) and 12t (SEQ ID NO:97) (family 3, Figure 29), 40 (SEQ ID NO:28) and 40t (SEQ ID NO:98) (family 4, Figure 29D) , 84 (SEQ ID NO:80) and 84t (SEQ ID NO:99) (family 5, Figure 29E) , and 126 (SEQ ID NO:82) and 126t (SEQ ID NO:100) (family 6, Figure 29F) .
  • RNA concentrations were determined from their absorbance reading at 260 nm (and were typically ⁇ 50 pM) . Binding reactions were done at 37°C in phosphate buffered saline containing 0.01% human serum albumin.
  • FIGURE 30 depicts the results of the determination of the 3'- and 5' -boundaries for a representative high- affinity VEGF ligand (ligand 12 (SEQ ID NO:66)) .
  • the 3'- boundary determination ( Figure 30A) showing partially hydrolyzed 5' -end labeled RNA (lane 4), hydrolytic fragments retained on nitrocellulose filters following incubation of the partially hydrolyzed RNA with VEGF at 5 nM (lane 1) , 0.5 nM (lane 2) , or 0.125 nM (lane 3) and partial digest of the 5' -end labeled RNA with RNAse T (lane 5) resolved on an 8% denaturing polyacrylamide gel.
  • the 5' -boundary (Figure 30B) was determined in an identical manner except that RNA radiolabeled at the 3 ' - end was used. Shown are RNase T x digest (lane 1) , partial alkaline hydrolysis (lane 2) , and hydrolytic fragments retained on nitrocellulose filters following incubation with VEGF at 5 nM (lane 3) , 0.5 nM (lane 4) , or 0.125 nM (lane 5) . Arrows indicate the 3'- and the 5' -boundaries of the minimal ligand 12 (italized) .
  • FIGURE 31 depicts the Scotchard analysis of 125 I-VEGF binding to HUVECS. Data points are averages of two determinations. Increasing concentrations of 125 I-VEGF were incubated with 2X10 5 cells in the presence or absence of 50-fold excess of unlabeled VEGF to determine the amount of total (o) , specific (D) and non-specific ( ⁇ ) binding of 125 I-VEGF as a function of free 125 I-VEGF concentration (insert) .
  • FIGURE 32 depicts the effect of random RNA (o) and representative high affinity RNA ligands lOOt (SEQ ID NO: 95) (family 1) ( ⁇ ) and 44t (SEQ ID NO: 96) (family 2)
  • FIGURE 33 depicts the starting random RNAs for experiments A and B, and PCR primers used in identifying 2'-NH 2 -RNA ligands to VEGF (Example 15) .
  • FIGURE 34 depicts 2'-NH 2 -RNA ligands to VEGF identified via the SELEX technology as described in Example 15.
  • FIGURE 36 shows the elastase inhibitory activity of representative ligand 34 (SEQ ID NO:197) .
  • Generation of p-nitroanilide was measured in the absence of elastase (o) , in the presence of the irreversible inhibitor N- methoxysuccinyl-Ala-Ala-Pro-Val-chloromethyl ketone (f) , in the absence of any inhibitor (D) , and in the presence of 180 nM ligand 34 (0) .
  • FIGURE 37A shows the results of competition experiments carried out between ligands 24 (SEQ ID NO:198) and 30 (SEQ ID NO:191) (Classes I and II) .
  • Figure 37B shows the results of competition experiments carried out between ligands 56 (SEQ ID NO:195) and 19 (SEQ ID NO:205) (Classes II and IV) .
  • FIGURE 38 shows the binding of representative ligand
  • FIGURE 39 shows the binding of representative ligand
  • FIGURE 40 shows the additive effect of representative ligand 19 (SEQ ID NO:205) on a-IPI-induced inhibition of elastase.
  • Generation of p-nitroanilide was measured in the presence of elastase alone ( ⁇ ) , 5 nM ⁇ __- IPI (o and •) , 5 nM ⁇ -lPI and 325 nM ligand 19 (D and ⁇ ) .
  • FIGURE 41 shows the results of a cell detachment assay conducted with elastase only, or elastase in the presence of an inhibitory ligand 24 (SEQ ID NO:198) , 30 (SEQ ID NO:191) or 19 (SEQ ID NO:205) .
  • FIGURE 42 shows the predicted secondary structure of the G-quartet sequence (SEQ ID NO:197) .
  • FIGURE 43 shows the binding affinities of ligands 21 (SEQ ID NO:195) , 24 (SEQ ID NO:198), and 34 (SEQ ID NO:197) to elastase in the presence and absence of 6 mM KC1.
  • FIGURE 44 shows the effect of pH on binding of 2'- NH 2 RNA ligand 14 (SEQ ID NO:200) (3'-fixed sequence truncate) and non-modified single-stranded DNA ligand 17 (SEQ ID NO:191) to elastase.
  • FIGURE 45 shows a predicted G-quartet structure and the mimetic molecule wherein the connecting loop nucleotides are replaced by a synthetic ethylene glycol linker tether.
  • FIGURE 46 shows the 2'-NH 2 -RNA SELEX primer- template constructs (Primer-Template-Mixture I) and ssDNA SELEX primer template constructs (Primer-Template-Mixture II) •
  • FIGURE 47 shows the structures of theophylline and caffeine.
  • FIGURE 48 shows the aligned sequences for two classes of RNA molecules with affinity for theophylline selected through the SELEX method (designated TR8 (SEQ ID NOS:251-253, 256-258)) and counter-SELEX method (designated TCT8 (SEQ ID NOS:247-250, 254-255)) .
  • Figure 2A shows the Class I sequences
  • Figure 2B shows the Class II sequences. The clone number from which the sequence was derived is shown at the left of the sequence. In some cases, multiple isolates were obtained. Sequences shown comprised the 40 nucleotide sequence that was initially random at the start of the SELEX process. The bold uppercase sequence is conserved and provides the basis for the alignment.
  • the arrows overlay regions of potential intramolecular complementary bases. The asterisk marks the single position in motif 1 that shows variability. Dashes represent absence of a nucleotide. Class I and II are related by circular permutation.
  • FIGURE 49 illustrates the predicted secondary structures for theophylline binding RNA species obtained from the TR8 SELEX and TCT8 SELEX experiments. Bases in shadow text were initially present in either the fixed 5' or 3' regions which flanked the random region. Note that either fixed region can contribute to the proposed structure.
  • the arrows in the TCT8-4 ligand represent the termini of the mini-derivative mTCT8-4 with the exception that the position 1-38 AU pair was changed to a GC pair in mTCT8-4.
  • FIGURE 50 illustrates the predicted secondary structure of theophylline binding RNA species.
  • N and N' are any complementary base pair. Numbers represent the size range of the various domains observed in the ligands obtained.
  • FIGURE 51 shows the binding properties of TCT8-4 (SEQ ID NO:249) and mTCT8-4 RNAs.
  • FIGURE 52 shows the predicted secondary structure for mTCT8-4.
  • FIGURE 53 shows the competition binding analysis of xanthine derivatives with TCT8-4 RNA (SEQ ID NO:249) .
  • FIGURE 54 shows the sequences of RNA ligands for caffeine (CR8 (SEQ ID NOS :259-270) ) . These ligands were identified after 8 SELEX selection rounds.
  • FIGURE 55 illustrates the chemical structure of theophylline with a series of derivatives that were used in competitive binding experiments with TCT8-4 RNA as described in Example 27.
  • the number in parenthesis represents the effectiveness of the competitor relative to theophylline, Kc(c)/Kc(t) .
  • Kc(c) is the individual competitor dissociation constant and Kc(t) is the competitive dissociation constant of theophylline.
  • 1-cp theophylline is 1-carboxypropyl theophylline. Certain data, denoted by ">" are minimum values that were limited by solubility of the competitor. Each experiment was carried out in duplicate.
  • SELEX This application is an extension and an improvement of the method for identifying nucleic acid ligands referred to as the SELEX method.
  • the SELEX method (hereinafter termed SELEX) was first described in U.S. Application Serial No. 07/536,428 filed June 11, 1990, entitled Systematic Evolution of Ligands by Exponential Enrichment, now abandoned, U.S. Patent Application Serial No. 07/714,131, filed June 10, 1991 and U.S. Patent Application Serial No. 07/931,473, filed August 17, 1992, now U.S. Patent No. 5, 270,163, both entitled Nucleic Acid Ligands. (See also PCT/US91/04078 and PCT/US93/09296) .
  • the full text of these applications, collectively referred to as the SELEX applications, including but not limited to, all definitions and descriptions of the SELEX process, are specifically incorporated herein by reference.
  • the SELEX process may be defined by the following series of steps: 1) A candidate mixture of nucleic acids of differing sequence is prepared.
  • the candidate mixture generally includes regions of fixed sequences (i.e., each of the members of the candidate mixture contains the same sequences in the same location) and regions of randomized sequences.
  • the fixed sequence regions are selected either: a) to assist in the amplification steps described below; b) to mimic a sequence known to bind to the target; or c) to enhance the potential of a given structural arrangement of the nucleic acids in the candidate mixture.
  • the randomized sequences can be totally randomized (i.e., the probability of finding a base at any position being one in four) or only partially randomized (e.g., the probability of finding a base at any location can be selected at any level between 0 and 100 percent) .
  • the candidate mixture is contacted with the selected target under conditions favorable for binding between the target and members of the candidate mixture. Under these circumstances, the interaction between the target and the nucleic acids of the candidate mixture can be considered as forming nucleic acid-target pairs between the target and the nucleic acids having the strongest affinity for the target.
  • nucleic acids with the highest affinity for the target are partitioned from those nucleic acids with lesser affinity to the target. Because only an extremely small number of sequences (and possibly only one molecule of nucleic acid) corresponding to the highest affinity nucleic acids exist in the candidate mixture, it is generally desirable to set the partitioning criteria so that a significant amount of the nucleic acids in the candidate mixture (approximately 5-10%) is retained during partitioning.
  • nucleic acids selected during partitioning as having the relatively higher affinity to the target are then amplified to create a new candidate mixture that is enriched in nucleic acids having a relatively higher affinity for the target.
  • the newly formed candidate mixture contains fewer and fewer unique sequences, and the average degree of affinity of the nucleic acid mixture to the target will generally increase.
  • the SELEX process will yield a candidate mixture containing one or a small number of unique nucleic acids representing those nucleic acids from the original candidate mixture having the highest affinity to the target molecule.
  • Partitioning means any process whereby ligands bound to target molecules can be separated from nucleic acids not bound to target molecules. More broadly stated, partitioning allows for the separation of all the nucleic acids in a candidate mixture into at least two pools based on their relative affinity to the target molecule. Partitioning can be accomplished by various methods known in the art. Nucleic acid-protein pairs can be bound to nitrocellulose filters while unbound nucleic acids are not . Columns which specifically retain nucleic acid- target complexes can be used for partitioning. For example, oligonucleotides able to associate with a target molecule bound on a column allow use of column chromatography for separating and isolating the highest affinity nucleic acid ligands.
  • Liquid-liquid partitioning can be used as well as filtration gel retardation, and density gradient centrifugation.
  • the SELEX Patent Applications describe and elaborate on this process in great detail. Included are targets that can be used in the process; methods for the preparation of the initial candidate mixture; methods for partitioning nucleic acids within a candidate mixture; and methods for amplifying partitioned nucleic acids to generate enriched candidate mixtures.
  • the SELEX Patent Applications also describe ligand solutions obtained to a number of target species, including both protein targets wherein the protein is and is not a nucleic acid binding protein.
  • SELEX provides high affinity ligands of a target molecule. This represents a singular achievement that is unprecedented in the field of nucleic acids research.
  • the present invention includes the application of the SELEX procedure to the specific targets of VEGF, elastase, thrombin, theophylline and caffeine.
  • the nucleic acid ligand 1) binds to the target in a manner capable of achieving the desired effect on the target; 2) be as small as possible to obtain the desired effect; 3) be as stable as possible; and 4) be a specific ligand to the chosen target. In most, if not all, situations it is preferred that the nucleic acid ligand have the highest possible affinity to the target.
  • the present invention includes methods for producing improved nucleic acid ligands based on modifications of the basic SELEX process.
  • the application includes separate sections covering the following embodiments of the invention: I. High Affinity Nucleic Acid Ligands Containing Modified Nucleotides; II. High Affinity
  • Nucleic Acid Ligands Containing Blended Nucleotides - Blended SELEX III. High-Affinity Oligonucleotide Ligands to Vascular Endothelial Growth Factor (VEGF) ; IV. Nucleic Acid Ligand Inhibitors of Human Neutrophil Elastase; and V. High-Affinity Nucleic Acid Ligands that Discriminate Between Theophylline and Caffeine - Counter-SELEX.
  • VEGF Vascular Endothelial Growth Factor
  • Modified and non-modified nucleic acid ligands to VEGF, elastase, thrombin, theophylline and caffeine are disclosed and claimed herein.
  • the scope of the ligands covered by the invention extends to all ligands of VEGF, elastase, thrombin, theophylline and caffeine identified herein.
  • This invention also includes modified and non- modified nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind thrombin as the specific nucleic acid ligands disclosed herein.
  • substantially homologous it is meant, a degree of primary sequence homology in excess of 70%, most preferably in excess of 80%.
  • Substantially the same ability to bind VEGF, elastase, thrombin, theophylline and caffeine means that the affinity is within two orders of magnitude of the affinity of the substantially homologous sequences described herein. It is well within the skill of those of ordinary skill in the art to determine whether a given sequence -- substantially homologous to those specifically described herein -- has substantially the same ability to bind VEGF, elastase, thrombin, theophylline and caffeine.
  • sequences that have little or no primary sequence homology may still have substantially the same ability to bind the target molecule. It is clear that binding is controlled by the secondary or tertiary structure of the nucleic acid ligand.
  • the present invention includes nucleic acid ligands that have substantially the same structural form as the ligands presented herein and that have substantially the same ability to bind VEGF, elastase, thrombin, theophylline and caffeine as the modified and unmodified nucleic acid ligands disclosed herein.
  • substantially the same structure includes all nucleic acid ligands having the common structural elements as the ligands disclosed herein that lead to the affinity to VEGF, elastase, thrombin, theophyline and caffeine.
  • This invention further includes ligands containing a variety of modified nucleotides, such as 2'- fluoro modifications.
  • the present invention is directed at methods for producing improved nucleic acid ligands.
  • SELEX is performed with a candidate mixture of oligonucleotides containing one or more modified nucleotides. The presence of the modified nucleotides increases the chemical diversity of the candidate mixture, allowing improved ligands to a particular target molecule to be identified.
  • nucleic acid ligands selected through the SELEX process are modified by incorporation of modified nucleotides.
  • Important modifications or derivatizations of the ligand to which the method of the present invention are directed are those that confer resistance to degradation and clearance in vivo during therapy. Further improvements conferred by the method of the present invention may include enhanced capacity to cross various tissue or cell membrane barriers, or any other accessory properties that do not significantly interfere with affinity for the target molecule.
  • the increased chemical diversity achieved by carrying out SELEX with nucleotide analogs can introduce ligand properties such as enhanced stability against nucleases, incorporation of reporter groups, introduction of moieties capable of covalent crosslinking to a target, or introduction of intra-ligand crosslinks for the generation of stable conformers and novel oligonucleotide shapes.
  • oligonucleotides containing 2' -amino, 2' -deoxy pyrimidines by T7 RNA polymerase transcription of DNA templates has also been previously reported (Id. ; Pieken et al . (1990) Science 253 :314) .
  • Homopolymers of the 2' -amino, 2' -deoxy pyrimidine nucleotides have also been prepared by polymerization of their 5' -diphosphate derivatives (Hobbs et al . (1973) Biochem 2:5138) .
  • Oligoribonucleotides containing 2' -amino, 2' -deoxy pyrimidines have also been prepared by automated solid phase synthesis. The trifluoroacetyl group has been used for protection of the 2'-amino group in preparation of phosphoramidite monomers (Pieken et al . (1990) supra) .
  • the SELEX technology identifies specific high affinity oligonucleotide ligands to a given molecular target by iterative enrichment from a vast pool of species.
  • the amplified oligodeoxyribonucleotide sequences are transcribed to its oligoribonucleotide homolog with T7 RNA polymerase.
  • the library is reassembled from its nucleoside triphosphate building blocks. This feature allows the introduction of chemically modified nucleoside triphosphates, and thus the enrichment of ligands bearing chemical functionalities not found in native RNA.
  • the increased chemical diversity achieved by the method of the present invention requires the chemical synthesis of a number of compounds. It is desirable to have a highly divergent synthetic methodology at hand that allows preparation of a variety of desired nucleotide analogs from a common intermediate. These analogs in all cases need to infer nuclease resistance to the oligonucleotide.
  • the 5-iodo, 2 ' -deoxyuridine is introduced into the SELEX library of candidate oligonucleotides as the 5' -triphosphate derivative.
  • the 5-iodo substituent is not compatible with the reaction conditions used in standard phosphorylation of 5-iodo, 2' amino, 2' -deoxy pyrimidines.
  • the 5' -triphosphate derivative which has not been previously described, is prepared from the 2' -amino, 2' -deoxy pyrimidine 5' -triphosphate by mercuration of the 5-position (Dale et al . (1975)
  • modified nucleic acid ligands of the present invention may include one or more modified nucleotides.
  • the proportion of modified to non-modified nucleotides contained in the nucleic acid ligands of the present invention may range from 1-100%.
  • the desired amount of modified nucleotide incorporation in the oligonucleotides is achieved by including one or more modified nucleotides in the oligonucleotide synthesis mixture.
  • SELEX is performed to completion with an initial candidate mixture of 2' -OH oligonucleotides, such that high-affinity nucleic acid ligands or family of ligands to a target molecule are identified. These ligands are then transcribed in the presence of modified nucleotides such that high-affinity ligands to the target molecule containing modified nucleotides are produced.
  • the present invention includes oligonucleotide ligands modified at specific positions. Nucleic acids ligands containing modified nucleotides at specific positions may be produced by synthesis.
  • SELEX is performed for a few selection rounds but not to completion, such that a candidate mixture of 2' -OH oligonucleotides is selected for partially enhanced affinity to the target molecule.
  • Modified nucleotides are then incorporated into the partially-selected oligonucleotides by transcription in the presence of modified nucleotides.
  • Example 1 describes the general experimental procedures for performing SELEX. In order to be useful in the existing SELEX protocol, the modified nucleotides must be substrates for polymerase (s) in their triphosphate forms and the resulting modified oligonucleotides must be templates for amplification.
  • Example 2 demonstrates that four prototypic 5-modified uridines, 5-AA-UTP, 5-F-12-UTP, 5-Dig-ll-UTP, and 5-Br- UTP, shown in Figure 2, meet the above requirements and can be used in SELEX.
  • modified nucleotides are incorporated into RNA by the T7 RNA polymerase under standard transcription conditions. Modified transcripts were reversed transcribed into cDNA by AMV reverse transcriptase and amplified by PCR. These results show that modified nucleotides can be directly incorporated into the SELEX procedure.
  • Example 3 describes SELEX selection of 2'-NH 2 ligands to the human thrombin. The affinities of SELEX identified 2' -OH and 2'-NH 2 ligands to thrombin are compared. While Example 3 describes SELEX identification of RNA ligands, the same procedure may be performed for the SELEX identification of DNA ligands to a specific target molecule.
  • the only enzymatic requirement for DNA SELEX is that the modified deoxynucleoside triphosphates serve as substrates for Taq DNA polymerase or another suitable polymerase. It is known that digoxygenin-11- deoxyuridine triphosphate can be used as a replacement substrate for TPP in PCR (Lanzillo (1990) BioTechniques 1:621) .
  • This invention includes the specific 2' -OH and 2'- NH 2 nucleic acid ligands of thrombin shown in Figure 6.
  • This invention also includes modified and non-modified nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind thrombin as the specific nucleic acid ligands shown in Figure 6.
  • nucleic acid ligands that have substantially the same structural form as the ligands presented herein and that have substantially the same ability to bind thrombin as the modified and unmodified nucleic acid ligands shown in Figure 6.
  • substantially the same structure includes all nucleic acid ligands having the common structural elements shown in Figure 7 that lead to the affinity to thrombin.
  • This invention further includes ligands containing a variety of modified nucleotides, such as 2'-fluoro modifications.
  • Example 4 describes a novel method for the synthesis of 5-iodo, 2' -amino deoxyuridine.
  • Example 5 describes the synthesis of 5-iodo, 2'- amino, 2' -deoxy pyrimidine 5' -triphosphate.
  • 5-iodo, 2'- amino, 2' -deoxy pyrimidine 5' -triphosphates cannot be prepared from their 5-iodo, 2' -amino, 2' -deoxy pyrimidine nucleoside precursors because the conditions employed in the phosphorylation reaction are not compatible with the 5-iodo group. However, they may be prepared from the 2'- amino, 2' -deoxy pyrimidine 5' -triphosphates by mercuration and subsequent iodination of the C-5 position.
  • Example 6 describes the in vivo stability of 2' -NH 2 modified ligands of thrombin.
  • the method of the present invention further includes incorporation of functional groups into oligonucleotides via the modified nucleotides.
  • One of the products of the SELEX procedure is a consensus of primary and secondary structures that enables the chemical or enzymatic synthesis of oligonucleotide ligands whose design is based on that consensus. Because the replication machinery of SELEX requires that rather limited variation at the subunit level (ribonucleotides, for example) , such ligands imperfectly fill the available atomic space of a target molecule's binding surface.
  • the present invention further includes nucleic acid ligands containing additional functional groups introduced via the modified nucleotides. A strategy for introduction of functional groups is described in Example 7.
  • the present invention further includes a method for generating high affinity blended nucleic acid ligands to specific target molecules.
  • the method generates blended nucleic acid molecules comprised of at least one functional unit.
  • Functional units that can be coupled to nucleotides or oligonucleotides include peptides, amino acids, aliphatic groups and lipid chains, or larger compounds such as peptide motifs, recognizable by the target molecule. These non-nucleic acid components of oligonucleotides may fit into specific binding pockets to form a tight binding via appropriate hydrogen bonds, salt bridges, or van der Walls interactions.
  • the blended nucleic acid ligands generated may guide SELEX- generated ligands to specific sites of the target molecule.
  • Further blended nucleic acid ligands may be prepared after the SELEX process for post-SELEX modification to add functionality to the ligand, for example, to increase RNA hydrophobicity and enhance binding, membrane partitioning and/or permeability, or to add reporter molecules, such as biotin- or fluorescence- tagged reporter oligonucleotides, for use as diagnostics.
  • Blended nucleic acid ligands may be generated by the addition of chemical groups which covalently react and couple the SELEX ligand to the target molecule.
  • catalytic groups can be added to nucleic acids to aid in the selection of SELEX ligands with protease or nuclease activity.
  • the functional units may also serve as toxins or radiochemicals that are delivered to specific locations in the body determined by the specificity of the SELEX devised nucleic acid ligand.
  • Blended nucleic acid ligands are defined herein as comprising at least one nucleic acid ligand and at least one functional unit.
  • a nucleic acid ligand is defined as any nucleic acid identified generally according to the SELEX process as described in the SELEX Patent
  • the functional unit may be any chemical species not naturally associated with nucleic acids, and may have any number of functions as enumerated herein.
  • the blended nucleic acid ligand is prepared by performing the SELEX method utilizing an initial candidate mixture wherein each nucleic acid sequence of the candidate mixture contains at least one functional unit, or a "blended candidate mixture".
  • each nucleic acid sequence of the candidate mixture has 1) a single functional unit attached at either the 5' or 3' end of nucleic acid sequence; 2) functional units at both the 5' and 3' ends of the nucleic acid sequence; 3) functional units added to individual nucleic acid residues; 4) functional units attached to all or a portion of all pyrimidine or purine residues; or 5) functional units attached to all or a portion of all nucleotides of a given type.
  • the functional units may also be attached only to the fixed or to the randomized regions of each nucleic acid sequence of the candidate mixture.
  • one or more functional units may be attached to a nucleic acid ligand identified according to the SELEX method wherein a blended candidate mixture is not used.
  • the points of attachment of the functional unit(s) to the nucleic acid ligand may vary depending on the ultimate requirements for the blended nucleic acid ligands.
  • Examples 8-11 below describe methods for generating the blended nucleic acid ligands of the present invention. As these examples establish, nucleotides and oligonucleotides containing a new functional unit are useful in generating blended nucleic acid ligands to specific sites of a target molecule.
  • peptide molecules Two examples are described for coupling peptide molecules to SELEX nucleic acid ligands in order to target specific peptide binding pockets.
  • a peptide containing the sequence arginine- glycine-aspartic acid (RGD) is coupled to a nucleotide and enzymatically incorporated into unselected polyribonucleotide.
  • the RGD-containing peptide is recognized and bound by the gpllbllla integrin protein, causing gpllbllla-mediated platelet aggregation.
  • a SELEX RGD-nucleic acid ligand may be generated with high specificity for gpllbllla that would not cross react with other integrins such as receptors for fibronectin, vetronectin, collagen, or laminin.
  • SELEX blended nucleic acid ligands containing the RGD peptide could bind at or near the gpllbllla ligand site and specifically inhibit gpllbllla activity.
  • a SELEX- identified single stranded DNA ligand to elastase was produced and coupled to inhibitory substrate peptide chloromethyl ketone. The resulting blended nucleic acid ligand was shown to specifically inhibit elastase.
  • Example 8 describes the generation of a peptide- conjugated RNA.
  • the peptide Gly-Arg-Gly-Asp-Thr-Pro (SEQ ID NO:40) was coupled to the derivatized base, 5- (3- aminoallyl) -uridine triphosphate to produce the peptide- conjugated UTP (RGD-UTP) , as shown in Figure 12.
  • RGD-UTP may be used in SELEX T7-catalyzed template-dependent transcription to produce an RNA oligonucleotide containing the RGD peptide at every uridine position.
  • the Gly-Arg-Gly-Asp-Thr-Pro (SEQ ID NO:40) peptide was chosen because the Arg-Gly-Asp (RGD) motif in matrix proteins is recognized and bound specifically by proteins of the integrin superfamily of cell adhesion receptors. Integrins bind to such proteins as fibronectin, laminin and vitronectin, at sites containing the RGD sequence. This binding is inhibited by short RGD-containing peptides which bind integrin proteins with a Kd of approximately 10 "5 M.
  • an RGD-containing peptide conjugated to an oligonucleotide may facilitate selection of high-affinity ligands to a RGD-binding target molecule.
  • peptide-conjugated oligonucleotides must also be compatible with the SELEX method, that is, the peptide-conjugated nucleotide must be incorporated with reasonable efficiency into transcribed RNA and partitioned RNA in turn must be reasonably efficiently transcribed into complementary DNA for amplification and additional rounds of SELEX.
  • Examples 8 and 9 below demonstrate that all of the conditions required for successful SELEX identification of nucleic acid ligands containing new functional groups are met by the method of this invention.
  • the peptide-UTP derivative was enzymatically incorporated into a blended RNA oligonucleotide as shown by increased size and altered charge compared with the native or unmodified oligonucleotide and by the UV shoulder at 310 nm.
  • Example 9 describes the binding of RGD-RNA to RGD- binding integrin gpllbllla. After separation, the bound RGD-RNA was reversed transcribed into DNA using normal SELEX protocols. The efficient transcription, partitioning, and reverse transcription shows that the site-directed blended nucleic acid ligands of this invention are compatible with the basic SELEX procedure.
  • the methods described herein do not include all of the schemes for introducing non-nucleic acid functional units, such as peptides, into an oligonucleotide. However, such methods would be well within the skill of those ordinarily practicing in the art. Putting a peptide on every uridine, as done in Example 8, has several advantages as compared with other labelling methods for use in the SELEX procedure.
  • the peptide is introduced throughout both the random and fixed regions, so that evolved RNA ligands could bind close to the peptide binding site.
  • Post-transcription modification may require additional time and expertise and introduces the additional variable of coupling efficiency.
  • Non-nucleic acid functional units may be used to yield evolved ligands with a non-overlapping spectrum of binding sites. For instance, a peptide could be placed at the 5' or 3' end of SELEX identified RNA ligands.
  • Another embodiment of this invention for introducing a non-nucleic acid functional unit at random positions and amounts is by use of a template-directed reaction with non-traditional base pairs. This method uses molecular evolution to select the best placement of the non-nucleic acid group on the SELEX identified ligand.
  • a X-dY base pair could be used, where X is a derivatizable ribonucleotide and the deoxynucleotide dY would pair only with X.
  • the X-RNA would contain the non- nucleic acid functional unit only at positions opposite dY in the dY-DNA template; the derivatized X base could be positioned in either the fixed or random regions or both; and the amount of X at each position could vary between 0-100%.
  • the sequence space of non-evolved SELEX ligands would be increased from N 4 to N 5 by substituting this fifth base without requiring changes in the SELEX protocol.
  • This embodiment may be used with photo-SELEX (U.S. Patent Application Serial No. 08/123,935, filed September 17, 1993, specifically incorporated herein by reference), where photo-active bases are placed at random sites, and these blended ligands partitioned after photolysis, then high affinity ligands are selected with the minimum number of substituted bases needed to crosslink with the target.
  • functional units that react covalently at enzyme active sites such as chloromethylketones, may be incorporated to produce irreversible enzyme inhibitors.
  • directed incorporation could be used also for post-SELEX incorporation of fluorescence tags, biotin, radiolabel, lipid groups, or to cap the oligonucleotide with a uniquely modified base for protection against nuclease digestion .
  • incorporación of non-nucleic acid functional units to produce blended SELEX ligands increases the repertoire of structures and interactions available to produce high affinity binding ligands.
  • Various types of functional units can be incorporated to produce a spectrum of molecular structures. At one end of this structural spectrum are normal polynucleic acids where the ligand interactions involve only nucleic acid functional units. At the other, are fully substituted nucleic acid ligands where ligand interactions involve only non-nucleic acid functional units. Since the nucleic acid topology is determined by the sequence, and sequence partitioning and amplification are the basic SELEX steps, the best ligand topology is selected by nucleic acid evolution.
  • Example 10 describes the preparation of a blended nucleic acid ligand to elastase to act as an elastase inhibitor.
  • N-methoxysuccinyl-Ala-Ala-Pro-Val- chloromethyl (SEQ ID NO:41) ketone is an effective irreversible inhibitor for elastase.
  • the nonspecific highly reactive nature of the chloromethyl ketone functionality in conjunction with mM range Kd of the tetrapeptide makes the inhibitor molecule unsuitable as a therapeutic agent.
  • the enzyme inhibition of nucleic acid ligands to elastase may be improved by coupling such ligands to the substrate tetrapeptide in a nonhydrolyzable manner such that the blended nucleic acid will inhibit the enzyme by occupying the substrate binding pocket.
  • a blended nucleic acid affinity and specificity is provided by the SELEX-derived nucleic acid ligand, whereas inhibition is provided by the peptide.
  • the functional unit of the blended nucleic acid ligand is attached to the SELEX derived nucleic acid ligand via the attachment of the functional unit to a nucleic acid that hybridizes to a region of the nucleic acid sequence of the ligand.
  • the functional unit oligonucleotide is DNA, and hybridizes to the fixed region of the nucleic acid ligand or at least a region of the nucleic acid ligand that is not involved in the binding reaction to the target.
  • the SELEX process is accomplished by the preparation of a candidate mixture of nucleic acid sequences comprised of fixed and randomized regions.
  • the candidate mixture also contains an oligonucleotide attached to a selected functional group.
  • the oligonucleotide is complementary to the fixed region of the nucleic acid candidate mixture, and is able to hybridize under the conditions employed in SELEX for the partitioning of high affinity ligands from the bulk of the candidate mixture. Following partitioning, the conditions can be adjusted so that the oligo-functional unit dissociates from the nucleic acid sequences.
  • Advantages to this embodiment include the following: 1) it places a single functional unit, such as a peptide analog, at a site where it is available for interaction with the random region of nucleic acid sequences of the candidate mixture; 2) because the functional unit is coupled to a separate molecule, the coupling reaction need only be performed once, whereas when the functional unit is coupled directly to the SELEX ligand, the coupling reaction must be performed at every SELEX cycle (aliquots from this reaction can be withdrawn for use at every cycle of SELEX) ; 3) the coupling chemistry between the functional unit and the oligonucleotide need not be compatible with RNA integrity or solubility -- thus simplifying the task of coupling; 4) in cases where the functional unit forms a covalent complex with the target, the SELEX derived nucleic acid ligand portion of the selected members of the candidate mixture can be released from the target for amplification or identification; and 5) following the successful identification of a blended nucleic ligand, the tethered portion of nu
  • VASCULAR ENDOTHELIAL GROWTH FACTOR VEGF
  • the present invention applies the SELEX and modified SELEX procedures to the specific target of vascular endothelial growth factor (VEGF) .
  • VEGF vascular endothelial growth factor
  • Examples 12-15 below the experimental parameters used to isolate and identify the nucleic acid ligand solutions to VEGF are described.
  • This invention includes the specific RNA ligands to VEGF shown in Figure 27 (SEQ ID NOS:48-82) .
  • the scope of the ligands covered by this invention extends to all RNA ligands of VEGF identified according to the SELEX procedure. More specifically, this invention includes nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind
  • VEGF as the specific nucleic acid ligands shown in Figure 27.
  • This invention also includes the 2' -NH 2 modified RNA ligands to VEGF as shown in Figure 34.
  • the scope of the present invention extends, therefore, to all modified nucleic acid ligands identified according to the SELEX method as well as to all sequences that are substantially homologous to and that have substantially the same ability to bind VEGF as the specific nucleic acid ligands shown in Figure 34.
  • This invention encompasses the use of the disclosed ligands to identify a second ligand.
  • a first SELEX identified ligand which binds to a specific site of the target molecule is used to elute secondary ligands binding to the same site.
  • a first SELEX identified ligand binding to a specific site of the target molecule is used to select secondary ligands which do not bind to the same site.
  • SELEX is conducted in the presence of the first ligand such that the binding site is saturated with the first ligand and selection occurs for ligands binding elsewhere on the target molecule.
  • a SELEX identified ligand to VEGF may itself be used as a target molecule to identify secondary ligands resembling the VEGF binding site. Such secondary ligands may compete with VEGF-substrate binding and inhibit the biological activity of VEGF.
  • this invention also includes nucleic acid ligands that have substantially the same structure as the ligands presented herein and that have substantially the same ability to bind VEGF as the nucleic acid ligands shown in Figures 27 and 34.
  • Example 12 describes the experimental procedures used to generate high-affinity nucleic acid ligands to VEGF.
  • Example 13 describes the high-affinity RNA ligands to VEGF shown in Figure 27.
  • Example 14 describes the specificity of truncated RNA ligands to VEGF.
  • Example 15 describes the experimental procedures used to generate 2'-NH 2 pyrimidine modified RNA ligands to VEGF.
  • the present invention also applies the SELEX and modified SELEX procedures to the specific target of human neutrophil elastase (elastase) .
  • elastase human neutrophil elastase
  • Examples 16-22 below the experimental parameters used to isolate and identify the nucleic acid ligands to elastase are described.
  • the invention includes the 2'-NH 2 RNA ligands of elastase shown in Table 4.
  • the scope of the ligands covered extends to all RNA ligands of elastase, modified and unmodified, identified according to the SELEX procedure, and further, ligands obtained by use of the ligands of the present invention.
  • this invention includes nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind elastase as the specific nucleic acid ligands shown in Table 4.
  • This invention also includes the specific DNA ligands of elastase shown in Table 9, identified by the method described in Example 16.
  • the scope of the ligands covered extends to all DNA ligands of elastase, modified and unmodified, identified according to the SELEX procedure, and further, ligands obtained by use of the ligands of the present invention. More specifically, this invention includes nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind elastase as the specific nucleic acid ligands shown in Table 9.
  • This invention encompasses the use of the disclosed ligands to identify a second ligand.
  • a first SELEX identified ligand which binds to a specific site of the target molecule is used to elute secondary ligands binding to the same site.
  • a first SELEX identified ligand binding to a specific site of the target molecule is used to select secondary ligands which do not bind to the same site.
  • SELEX is conducted in the presence of the first ligand such that the binding site is saturated with the first ligand and selection occurs for ligands binding elsewhere on the target molecule.
  • a SELEX identified ligand to elastase may itself be used as a target molecule to identify secondary ligands resembling the elastase binding site.
  • Such secondary ligands may compete with elastase-substrate binding and inhibit the biological activity of elastase.
  • this invention also includes nucleic acid ligands that have substantially the same three dimensional structure as the ligands presented herein and substantially the same ability to bind elastase as the nucleic acid ligands shown in Tables 4 and 9.
  • the present invention identifies and describes 2'- NH 2 -modified RNA ligands which inhibit elastase activity (Example 18) .
  • Several of these inhibitory ligands were truncated by removal of the 3'-fixed sequence and the ability of the truncated ligands to inhibit elastase examined (Example 19) .
  • the ligands of elastase have been grouped into a number of classes. The results show that for class I and II ligands, 3 '-truncated ligands had similar abilities to inhibit elastase as the full-length ligands.
  • RNA ligands is pH dependent effects how the ligands may best be utilized. For example, by using two buffers with different pH, it may be possible to develop two ligands for a given target, each of which binds at a different pH. Likewise, one may tailor ligands with high affinity binding for a given target molecule in one cell compartment with specific pH, but not to the same protein in another cell compartment with a different pH. Similarly, ligands that are active specifically in certain cell types, for example tumor cells which are known to have lower internal pH than normal cells, may be discovered with 2'-NH 2 modified RNAs.
  • Nucleic Acid Mimetics Nucleic Acid Mimetics.
  • Several classes of elastase ligands identified herein appear to assume a "G-quartet" type structure. Sequences rich in repeated runs of contiguous guanines undergo structural rearrangement to form planar quartets joined by Hoogstein hydrogen bonds of four guanine residues. Based on X-ray fiber diffraction data, a G-quartet was described for poly rG (Zimmerman et al . (1975) J. Mol. Biol. 12:181-192) . In the late 1980s, this structure was identified in DNA sequences containing short runs of guanines interrupted by several bases, and such DNA sequences are found in immunoglobulin switch regions (Sen, D. and Gilbert, W.
  • the G-quartet structure can occur within a single DNA strand (intramolecular G- quartet) or between two (Sundquist et al . (1989) Nature 342:825-829: Kang et al . (1992) Nature 356:126-131) or four strands (intermolecular G-quartets) (Sen, (1988) supra) .
  • the present invention applies the SELEX and counter- SELEX procedures to the specific targets of caffeine and theophylline.
  • Examples 23-29 describe the experimental parameters used to isolate and identify the nucleic acid ligands to theophylline and caffeine.
  • this invention includes the nucleic acid ligands of theophylline shown in Figures 48A and 48B and the specific nucleic acid ligands of caffeine shown in Figure 54.
  • the scope of the ligands covered extends to all ligands of theophylline and caffeine modified and unmodified, identified according to the SELEX and counter-SELEX procedures.
  • this invention includes nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind theophylline and caffeine as the specific nucleic acid ligands shown in Figures 48 and 54.
  • this invention also includes nucleic acid ligands that have substantially the same three dimensional structure as the ligands presented herein and substantially the same ability to bind theophylline and caffeine as the nucleic acid ligands shown in Figures 48 and 54.
  • nucleic acid ligands that have substantially the same three dimensional structure as the ligands presented herein and substantially the same ability to bind theophylline and caffeine as the nucleic acid ligands shown in Figures 48 and 54.
  • RNA ligands of theophylline and caffeine that contain 2'-NH 2 and/or 2'-F modifications of certain riboses of the RNA ligand.
  • nucleic acid ligands and nucleic acid ligand solutions to theophylline and caffeine described herein are useful as diagnostic reagents. Further, the nucleic acid ligands to theophylline and caffeine described herein may be used beneficially for therapeutic purposes.
  • the present invention shows how SELEX can be applied to generate oligonucleotides with binding and discriminatory properties for theophylline and caffeine superior in certain respects to those of monoclonal antibodies. As is shown below, SELEX selected nucleic acid ligands to theophylline have an affinity for theophylline comparable to that of monoclonal antibodies but with a superior selectivity.
  • the present invention includes a novel modification of the SELEX method, herein termed "counter-SELEX".
  • the counter-SELEX method is a powerful means of eliminating undesired sequences from a SELEX experiment .
  • the significance of this technique is that it provides a means for increased specificity of SELEX pools. Also it provides a methodology for decreasing the number of SELEX selection rounds required to identify a high affinity and high specificity nucleic acid ligand. Further, it provides a methodology for identifying a nucleic acid ligand of a target molecule that does not cross-react with other molecules, including closely related molecules.
  • the method of counter-SELEX is comprised of the steps of: a) preparing a candidate mixture of nucleic acids (The candidate mixture may contain nucleic acids with one or more types of modified nucleotides . ) ; b) contacting the candidate mixture with the target molecule, wherein nucleic acids having an increased affinity to the target relative to the candidate mixture will bind the target molecule and may be partitioned from the remainder of the candidate mixture (The target molecule may be, but is not necessarily attached to a column) ; c) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; d) contacting the increased affinity nucleic acids with one or more non-target molecules such that nucleic acids bound to the target molecule with affinity to a non- target molecule are removed; and e) amplifying the nucleic acids with specific affinity to the target molecule to yield a mixture of nucleic acids enriched for sequences with a relatively higher affinity and specificity to the target molecule.
  • counter-SELEX is conducted with oligonucleotides bound to a column.
  • This embodiment is used to eliminate oligonucleotides with undesired cross-reactivities which bind the target molecule.
  • the target molecule is bound to a column, and a candidate mixture of oligonucleotides applied to the column such that oligonucleotides binding the target molecule are retained in the column.
  • the column is then washed with the binding buffer to remove non-binding oligonucleotides.
  • one or more non-target molecules, or counter- ligands, to which cross-reactivity is undesirable are applied in solution to the column in vast molar excess.
  • Molar excess may be defined as 10 3 -10 4 times the amount bound to the column. This results in the removal of bound oligonucleotides from the column which also bind the counter-ligand(s) .
  • the target molecule is then applied to the column in solution, in vast molar excess, to remove oligonucleotides with the desired ligand binding specificity.
  • Counter-SELEX may be conducted with a single counter-ligand molecule, with sequential exposure to more than one counter-ligand, or with a cocktail of several related counter-ligands, to eliminate oligonucleotides with cross-reactivity to several compounds. Further, counter-SELEX may be conducted after pre-selection of oligonucleotides for affinity for a target molecule by one or more selection rounds of SELEX.
  • counter-SELEX is conducted in solution. This approach is designed to eliminate oligonucleotides with undesired cross-reactivity in solution.
  • the binding of oligonucleotides to a target on a column is performed with the counter-ligand present in the binding buffer. Once the oligonucleotides are bound to the target, the column is vigorously washed. This procedure prevents oligonucleotides with cross- reactivity to the counter-ligand from binding to the column.
  • This embodiment of the counter-SELEX method may be used in SELEX experiments for binding to peptides with sequences related to other peptides .
  • This embodiment is intended to eliminate possible side effects due to cross- reactivity of SELEX selected oligonucleotides with other target molecules in vivo .
  • SELEX may be performed to identify nucleic acid ligands to a peptide hormone such as vasointestinal peptide (VIP) .
  • VIP vasointestinal peptide
  • the amino acid sequence of VIP is similar to that of the peptide hormone glucagon, which has several essential roles in vivo .
  • nucleic acid ligands to VIP are identified which do not cross-react with glucagon in solution, and therefore, would not cross-react with glucagon in vivo .
  • This embodiment of the counter-SELEX procedure may be performed with one or more counter-ligand molecules in solution to eliminate nucleic acid ligands with cross- reactivity to more than one target molecule in vivo . Further, counter-SELEX may be conducted after one or more selection rounds of SELEX.
  • the utility of the counter-SELEX procedure for accelerating the enrichment of the target molecule binding nucleic acid species is illustrated by comparing the binding properties of pooled RNA after an identical number of selection rounds of SELEX and counter-SELEX. After three SELEX selection rounds, 12% of RNA initially bound to immobilized theophylline was resistant to subsequent elution with caffeine (TR8) . After three counter-SELEX selection rounds with caffeine, 45% of initially bound RNA was resistant to caffeine elution (TCT8) .
  • the ligands selected to bind to theophylline show a remarkable 15 base sequence which is completely conserved at 14 positions and present in each TCT8 and TR8 clone.
  • This sequence is comprised of two separate motifs: motif 1 is 5'-AUACCA-3' (SEQ ID NO:244) and motif 2 is 5' -CCUUGG(C/A)AG-3' (SEQ ID N0:245) .
  • the sequences and spacing between motifs 1 and 2 are variable among the ligands, ranging from 8 to 20 residues.
  • each ligand contains two stems of variable length and sequence loop, termed "vsteml” and “vstem2”, and a variable length and sequence loop, termed "vloop” . These structural features flank the conserved motifs in the model.
  • Motif 1 and 2 are based-paired through interaction of the CCA triplet of motif 2 with the UGG triplet of motif 1.
  • the CCU triplet of motif 1 is depicted as an asymmetric bulge.
  • the model accommodates the remaining six nucleotides of the conserved region as a symmetric bulge consisting of the AUA of motif 2 opposed to (C/A)AG of motif 1. This bulge is closed by a stem of variable length and sequence.
  • the relative spatial orientation of the CCU and (C/A)AG components is clearly specific in this family of ligands since their orientations are preserved whether the CCU component is located 3' (e.g., TCT8-5, TCT8-7, TCT8-8, TCT8-1, TCT8-6 (SEQ ID NOS:247, 250, 252, 248)) or 5' (e.g., TCT8-11, TCT8-3 (SEQ ID NOS:254-255) ) . It is notable that closure of the symmetric bulge often involves participation of the fixed sequences, either from the 5' (e.g., TCT8-5 (SEQ ID NO:247)) or 3' regions (e.g., TCT8-7 (SEQ ID NO:250)) .
  • the present invention shows that nucleic acids can interact specifically with a therapeutically important small molecule, further confirming the possible use of these oligonucleotides as diagnostic reagents.
  • monoclonal antibodies are commonly used to determine serum theophylline levels (Poncelet et al . (1990) J. Immunoassay ⁇ :77) .
  • the affinity of the RNA ligands for theophylline is comparable to that of the antibodies used in clinical assays (Poncelet et al . (1990) supra) .
  • these antibodies vary in their ability to discriminate amongst related compounds, typically they show 0.1-1% cross-reactivity with compounds such as caffeine and theobromine.
  • the discrimination of the RNA ligands described here is at least an order of magnitude greater than such antibodies.
  • RNA In addition to the work described here, the structural versatility of RNA is attested to by its ability to interact with other small molecules. These include the interaction of type I introns with guanosine (Bass and Cech (1984) Nature 308 : 820) and arginine (Yarus (1988) Science 240:1751) , the binding of arginine by HIV TAR RNA (Tao and Frankel (1992) Proc. Natl. Acad. Sci.
  • the counter-SELEX procedure described herein may be used to accelerate the rate at which specific ligands are obtained, and more importantly, to selectively remove from the oligonucleotide population those species with affinity for targets that are structurally closely related to the target of interest.
  • the present invention verifies the potential application of SELEX to include virtually any molecule of biomedical interest, irrespective of size and chemical class.
  • Example 23 describes the general experimental procedures used in the present work, including the equilibrium filtration analysis and the selection of the theophylline family of ligands TR8 after 8 rounds of selection of SELEX.
  • Example 24 describes the selection of the theophylline ligand family TCT8, selected after 5 selection rounds of SELEX and 3 selection rounds of counter-SELEX with caffeine.
  • Example 25 describes the identification of the caffeine ligand family CR8 after eight rounds of selection of SELEX.
  • Example 26 describes the binding characteristics of a representative theophylline RNA ligand (TCT8-4 (SEQ ID N0:249)) . The results show that TCT8-4 bound theophylline with a Kd of 0.58 ⁇ M, similar to that observed for some monoclonal antibodies.
  • a truncated ligand was constructed containing the conserved motif of TCT8-4 (mTCT8-4) , and found to bind theophylline with a Kd of 0.11 ⁇ M, approximately 5.8-fold greater than the full-length ligand.
  • Example 27 describes competition binding studies conducted with xanthine derivatives for RNA ligands for theophylline. These studies allow investigation of the relative contributions of various constituents to the overall binding.
  • Example 28 describes aspects of molecular modeling of high affinity ligands for theophylline.
  • DNA templates for in vi tro transcription that contain a region of thirty random positions flanked by constant sequence regions
  • the corresponding PCR primers were synthesized chemically (Operon) .
  • the random region was generated by utilizing an equimolar mixture of the four nucleotides during oligonucleotide synthesis.
  • the two constant regions were designed to contain PCR primer annealing sites, a primer annealing site for cDNA synthesis, T7 RNA polymerase promoter region, and restriction enzyme sites that allow cloning into vectors.
  • Nitrocellulose Filter Binding Assay Oligonucleotides bound to proteins can be effectively separated from the unbound species by filtration through nitrocellulose membrane filters (Yarus & Berg (1970) Anal. Biochem. 15:450; Lowary & Uhlenbeck (1987) Nucleic Acids Res. 11:10483; Tuerk & Gold (1990) supra) . Nitrocellulose filters (Millipore, 0.45 urn pore size, type HA) were secured on a filter manifold and washed with 4-10 ml of buffer.
  • RNA polymerase under standard transcription conditions (40 mM Tris-Cl, pH 8.0, 12 mM MgCl 2 , 5 mM DTT, 1 mM spermidine, 0.002% Triton X- 100, 4% PEG, 37°C) .
  • Autoradiogram of a 15% denaturing polyacrylamide gel showing products of transcription done with UTP, 5-AA-UTP, and 5-Br-UTP is shown in Figure 3.
  • the efficiency of transcription was estimated from the total amounts of 32 P-radiolabelled transcript produced after three hours of incubation with 2 mM ribonucleotide triphosphates, 100 nM double-stranded DNA template, and 5 units/ul of T7 RNA polymerase (NEB) .
  • 5-AA-UTP, 5-F-12- UTP, 5-Dig-11-UTP, and 5-Br-UTP were incorporated with 28%, 51%, 23%, and 50% efficiency, respectively, as compared to UTP. No transcription was detectable when UTP was omitted from the transcription mixture.
  • UTP, 5-Dig-ll-UTP, or 5-Br-UTP were reverse transcribed into cDNA by AMV reverse transcriptase with efficiencies comparable to that of the unmodified RNA (done in 50 mM Tris, pH 8.3, 60 mM NaCl, 6 mM Mg(OAc) 2 , 10 mM DTT, 37°C) (data not shown) .
  • These cDNAs were then amplified by
  • RNA was transcribed with T' RNA polymerase from double-stranded DNA in a reaction containing ATP, GTP, 2'-NH 2 -UTP, and 2'-NH 2 -CTP.
  • Clone 17 (SEQ ID NO:36) is representative of the A subgroup of the group II 2'-NH 2 ligands.
  • Clone 16 (SEQ ID NO:7) is representative of the class I ligands of the 2' -OH ligands and clone 27 (SEQ ID NO:18) is representative of the class II ligands of the 2' -OH ligands.
  • Binding Analysis All binding analyses were done by nitrocellulose filter binding. Binding curves as determined by increasing protein concentration in the presence of a fixed RNA concentration lower than the protein concentration are shown for the 2' -OH ligands in Figure 8A and for the bulk 2'-NH 2 ligands in Figure 8B. Under these conditions, the RNAs 16 and 27 have kDs of 30 nM and 60 nM, respectively. The kD for the bulk 2' -NH 2 round 15 ligands which had been sequenced is approximately 200 nM.
  • EXAMPLE 4 SYNTHESIS OF 5-IODO, 2' -AMINO DEOXYURIDINE.
  • the reported synthesis of 5-iodo, 2' -amino,2' - deoxyuridine involved five steps and gave the desired compound in an overall yield of 3%.
  • Presented herein is a method of preparing 5-iodo, 2' -amino, 2' -deoxyuridine in four steps from uridine with an overall yield of 46%.
  • the synthesis of 5-iodo, 2' -amino, 2' -deoxyuridine is shown in Figure 9.
  • uridine is initially dehydrated to the 2,2' -anhydrouridine by the standard method in 92% yield. Id. This intermediate is then converted to the 2' -azido, 2' -deoxyuridine in 84% yield according to Example 7 below (Synthesis of 2' - Azido-2' -Deoxyuridine (ADU)) .
  • ADU is iodinated at the C- 5 position without protection of the 5'- and 3' -OH groups in 76% yield in adaptation of the method of Robins et al . (1982) Can. J. Chem. £0:554. Reduction of the 5-iodo,2'- amino, 2' -deoxyuridine with triphenylphosphine gave 78% yield of the desired compound.
  • Lithium fluoride (0.93 g, 36 mmol) was suspended in 20 ml of DMF heated to 105 °C. To the stirred suspension was added 20 ml of TMEDA followed by azidotrimethylsilane (4.15 g, 36 mmol) . After stirring for 30 min, anhydrouridine (4.52 g, 20 mmol) was added and the reaction allowed to proceed for 48 h. The solvents were removed under vacuum and the residue co-evaporated from methanol three times. The residue was dissolved in 10 ml of methanol and 40 ml of ethyl acetate was added to precipitate most of the salt and residual starting material.
  • PYRIMIDINE 5' -TRIPHOSPHATES The synthesis of 2' -amino,2' -deoxy pyrimidine 5'- triphosphates has not previously been reported in full detail.
  • the 2' -amino group of 2' -amino, 2' -deoxyuridine is trifluoroacetylated (Imazawa et al . (1979) J. Org. Chem. 44 . :2039) in 82% yield. This intermediate is phosphorylated at the 5' -position (Ludwig and Eckstein (1989) J. Org. Chem. 54.:631) in 61% yield.
  • the 5-iodo derivative of the 2' -amino, 2' -deoxyuridine 5'- triphosphate was prepared by mercuration (Dale et al . (1975) Biochemistry 14:2447) in a single step.
  • FIG. 11 shows the plasma kinetic curves obtained for each experimental animal. The shape of the plasma kinetic curve was similar to that observed with a 2'-OCH 3 stabilized oligonucleotide of similar length which distributes rapidly into total body water and remains stable in vivo for at least 6 hours.
  • EXAMPLE 7 INTRODUCTION OF FUNCTIONAL GROUPS.
  • 5-AA-UTP and 5-AA-dUTP contain a primary amine functionality which is useful since a wide variety of other functional groups can be readily introduced by the carbodiimide/N-hydroxysuccinimide coupling, as shown by Muhlegger et al . (1989) Nucleosides & Nucleotides 8.: 1161 ( Figure 5) .
  • Global physico-chemical properties of the modified oligonucleotides such as hydrophobicity or the overall charge can be modified with the addition of appropriate "side chains".
  • molecules that are known to bind at well-defined sites on given target molecules may be used in this context to anchor the binding of nucleic acid ligands to specific regions on the target molecule.
  • Such moieties themselves need only modest affinities for their target molecules since the oligonucleotide framework would be evolved to provide additional binding energy.
  • An example of this strategy is given in Figure 5.
  • Gly-Arg-Gly-Asp-Thr-Pro was obtained from GIBCO-BRL (Gaithersburg, MD) .
  • l-ethyl-3- (dimethylaminopropyl) carbodiimide hydrochloride (EDC) was obtained from Pierce Chemical Co. (Rockford, IL) .
  • Other chemicals were of highest quality available from Aldrich Chemical Company (Milwaukee, WI) .
  • the peptide Gly-Arg-Gly- Asp-Thr-Pro (SEQ ID NO:40) was coupled to UTP via a two- step reaction through the N-terminal ⁇ -amino group of the peptide.
  • This ⁇ -amino group is the only reactive amine in the peptide and is not required for integrin binding. Condensation with peptide carboxyl groups could function to block the aspartic acid residue, thus preventing integrin binding.
  • 5- (3-aminoallyl)UTP (5 mg) was dissolved in 250 ⁇ l of 0.2 M sodium borate, pH 8.5.
  • the solution was mixed with 750 ⁇ l succinic anhydride (97 mg/1.14 ml DMF) and reacted for 2 h at 4°C.
  • the sample was applied in 0.1 M triethylammonium bicarbonate, and eluted with a linear gradient to 1.0 M triethylammonium bicarbonate.
  • the column was eluted at 1.0 ml/min over 20 min.
  • the major peak was pooled, lyophilized, and taken up in dry DMF for the next reaction.
  • the sample was analyzed by TLC by spotting on PEI-F chromatography plates (J.T. Baker) and developing with 0.2 M sodium phosphate, pH 3.5.
  • succinyl-UTP Chromatographic isolation of succinyl-UTP.
  • the succinylation reaction mixture was diluted 1:1 with 0.1 M triethylammonium-bicarbonate, pH 7.6 (Buffer A) , filtered and applied to a 0.5 cm x 5 cm Mono Q anion exchange column which was equilibrated with the same buffer.
  • the column was eluted with a gradient to 1.0 M triethylammonium-bicarbonate (Buffer B) at a flow rate of 0.5 ml/ml (1 min/fraction) ( Figure 13) .
  • Succinyl-UTP was easily separated by ion exchange from the starting material.
  • the activated nucleotide was added to 14 mg of lyophilized GRGDTP and the mixture reacted for 2 h at 4°C.
  • the reaction mixture was diluted 1:1 with 0.1 M triethylammonium bicarbonate, purified on a Mono Q column as above, and analyzed by TLC and Fast Atom Bombardment (FAB) Mass Spectrometry (University of Colorado Structural Division) .
  • FAB Fast Atom Bombardment
  • RGD-UTP Chromatographic isolation of RGD-UTP.
  • RGD-UTP was purified by ion exchange chromatography. The peptide coupling reaction mixture was diluted with Buffer A, filtered and purified on the Mono Q column described above. RGD-UTP eluted from the Mono Q column similar to AA-UTP, but ahead of succinyl-UTP, suggesting the arginine side chain also interacts with the phosphate charge. The major peak eluted at approximately 70% of Buffer B, ahead of the peak of residual succinyl-UTP (which eluted at approximately 90% of Buffer B) ( Figure 14) . Only trace amounts of other peaks were detected, except for the EDC which eluted earlier. Fractions 35-38 were pooled and lyophilized. The sample was dissolved in dry DMF for thin layer chromatography (TLC) and FAB mass spectroscopy. The sample had a maximum adsorption at 290 nm.
  • TLC thin layer chromat
  • EXAMPLE 9 BINDING OF RGD-RNA TO gpllbllla.
  • RNA transcription reaction mixture was phenol:chloroform extracted, ethanol precipitated and washed, and dissolved in 50 mM sodium phosphate, 0.2 M NaCl, 6 M urea, 50% formamide (Fisher, Ultrapore, low-UV absorbance) , pH 6.0. Following sample application, the column was developed with a linear gradient to 1 M NaCl.
  • RNA peak was precipitated by addition of 2 :1 v/v ethanol, the pellet was washed with 80% ethanol and dried in a speed-vac, then dissolved in SELEX binding buffer (150 mM NaCl, 10 nM HEPES, 2 mM CaCl 2 , 1 mM MgCl 2 , 0.1% Tween, pH 7.4) .
  • SELEX binding buffer 150 mM NaCl, 10 nM HEPES, 2 mM CaCl 2 , 1 mM MgCl 2 , 0.1% Tween, pH 7.4
  • RNA and RNA-gpIIbllla (Enzyme Research) complexes were prepared by incubation at 37°C for 10 min, and separated by size exclusion chromatography on a 16 x 100 Superdex 200 column (Pharmacia) which was equilibrated and run in the binding buffer.
  • RNA transcript was purified by adsorption onto an anion exchange column (Mono Q) in a phosphate buffer containing 6 M urea and 50% formamide, and eluted with a gradient to 1 M NaCl ( Figure 16) .
  • RGD-RNA eluted as one major peak from the column at a position in the gradient slightly ahead of the elution position of an equivalent RNA transcript made with normal UTP, consistent with the altered charge and perhaps altered conformation of the derivatized RNA.
  • the amount of truncated RNA material was only slightly increased compared to normal RNA transcription.
  • Binding of RGD-30n7 RNA to gpllbllla is a method for identifying high affinity RNA ligands to gpllbllla (Kd ⁇ 10 "8 ) .
  • RGD-RNA was incubated with gpllbllla (2 mg/ml) in 0.15 M NaCl, 2 mM CaCl 2 , 1 mM MgCl 2 , 20 mM HEPES, 0.1% Tween 20, pH 7.4.
  • ssDNA ligand 17 (DNA-17) , having the sequence TAGCGAT ACTGCGTGGGTTGGGGCGGGTAGGGCCAGCAGTCTCGTGCGGTACTTGAGCA (SEQ ID NO:42) has a Kd for elastase of 15 nM.
  • blended nucleic acid molecules were prepared in which the substrate peptide was attached to both the 3' and 5' ends of the DNA. Peptide conjugation. The chemistry of the attachment is shown in Figure 18.
  • An oligonucleotide with four 18-atom ethylene glycol moieties (synthesized by using spacer phosphoramidite, Clonetech) and a thiol group at the 3' end was synthesized by automated DNA synthesis, deprotected by standard methods, and gel purified.
  • the oligonucleotide was passed through a Sephadex-G50 spin column equilibrated in 0.5 M triethylammonium acetate buffer (pH 7.5) to remove excess DTT, and then mixed with N-methoxysuccinyl Ala-Ala-Pro- Val-chloromethyl (SEQ ID NO:41) ketone (25 mg/200 ul of DMF) . The mixture was incubated at 37°C overnight. 20 ⁇ l of 1 M DTT was added to inactivate the unreacted chloromethyl ketone inhibitor. The peptide conjugated DNA was finally purified from the unconjugated peptide either by three successive Sephadex-G50 spin columns or by reverse phase HPLC.
  • Elastase inhibition assay The assay for elastase inhibition is based on the use of a chromogenic tetrapeptide substrate.
  • the assay was conducted in a buffer containing 10 nM elastase, 0.5 mM N- methoxysuccinyl-Ala-Ala-Pro-Val-p-nitroanilide (SEQ ID NO:43) , 150 mM NaCl, 26 mM KCl, 2 mM MgCl 2 , 0.02% HSA, 0.05% DMSO, 0.01% Triton X-100, and 100 mM Tris-HCl, at 25°C.
  • the assay measured the generation of elastase- induced release of p-nitroanilide as a function of time by spectroscopy (OD 405 nm) .
  • the rate of p-nitroanilide generation in the absence of inhibitor was used as the control.
  • Positive control was established in the presence of the irreversible inhibitor N-methoxysuccinyl- Ala-Ala-Pro-Val-chloromethyl (SEQ ID NO:41) ketone.
  • the partial inhibition seen with the inactivated inhibitor may be due to competitive inhibition due to the high peptide concentration.
  • Figure 20 shows the inhibition of the blended nucleic acid ligand in the presence of 20 mM DTT.
  • the inhibition of elastase by the 5' end blended nucleic acid ligand and the 3' end blended nucleic acid ligand is shown in Figure 21.
  • a Lineweaver-Burk plot of elastase inhibition by N-methoxysuccinyl Ala-Ala-Pro-Val (SEQ ID NO:44) or the blended nucleic acid ligand is shown in Figure 22.
  • the resulting Km of 0.16 mM for the substrate is in good agreement with published values.
  • the Ki of the blended nucleic acid ligands was 30 nM versus 920 uM for the inhibitor peptide alone, or a 30,000 fold improvement.
  • the splint blended SELEX process was performed by preparing a standard SELEX candidate mixture and a single compound containing a valyl phosphonate attached to a nucleic acid sequence that hybridizes to a portion of the fixed region of the candidate mixture of nucleic acid sequences as shown in Figure 2 .
  • Figure 24 also shows the chemical reaction that occurs between the valyl phosphonate and human neutrophil elastase (HNE) .
  • HNE human neutrophil elastase
  • the valyl phosphonate was activated via an NHS ester. This compound was coupled to the 5' hexyl amine linker of a 19-mer DNA oligo complementary to the 5'- fixed region of 40N7.1.
  • a candidate mixture composed of pyrimidine 2'NH 2 -substituted RNA was hybridized to the splint, and reacted with HNE at subsaturating protein concentrations. Covalent complexes were enriched by diluting the reaction 100-fold, then filtering through nitrocellulose. After five rounds of selection for species that reacted with the HNE, the reactivity of the pool was assayed by gel electrophoresis (5% polyacrylamide/TBE pH 8 or MAE pH 6/0.25% SDS) . The results are shown in Figure 25. The analysis of reaction rates indicates biphasic kinetics, with k- j ⁇ obs ⁇ 0.1 min -1 , and k 2 obs ⁇ 0.01 min -1 .
  • the second-order rate constant for the thrombin reaction is estimated to be ⁇ 200 M _1 min -1 , 10 4 fold lower than the reaction rate with elastase.
  • RNA ligands with an affinity for VEGF an initial pool of random RNAs were generated (each containing a 30 nucleotide random region flanked by fixed sequences for transcription and PCR amplification) by in vi tro transcription of approximately 200 picomoles (10 14 molecules) of the double stranded DNA template using T7 RNA polymerase.
  • the DNA templates and the corresponding PCR primers were prepared chemically using established solid phase oligonucleotide synthesis protocols.
  • the random region was generated by utilizing an equimolar mixture of the four unmodified nucleotides during oligonucleotide synthesis.
  • the two constant regions were designed to contain PCR primer annealing sites, primer annealing site for cDNA synthesis, T7 RNA polymerase promoter region and restriction enzyme sites that allow cloning into vectors (Figure 26) (SEQ ID NOS:45-47) .
  • Transcription mixtures consisting of 100-300 nM template, 5 units/ ⁇ l T7 RNA polymerase, 40 mM Tris-Cl buffer (pH 8.0) containing 12 mM MgCl 2 , 5 mM DTT, 1 mM spermidine, 0.002% Triton X-100, 4% PEG were incubated at 37°C for 2-3 hours. These conditions typically resulted in transcriptional amplification of 10 to 100-fold.
  • PBS phosphate buffered saline
  • RNA (which typically amounted to 5-10% of the total input RNA) was then extracted from the filters and reverse transcribed into cDNA by avian myeloblastoma virus reverse transcriptase (AMV RT) . Reverse transcriptions were done at 48°C (60 min) in 50 mM Tris buffer (pH 8.3) , 60 mM NaCl, 6 mM Mg(0Ac) 2 , 10 mM DTT and 1 unit/ ⁇ l AMV RT. Amplification of the cDNA by PCR under standard conditions yielded a sufficient amount of double-stranded DNA for the next round of in vi tro transcription. Nitrocellulose Filter Binding Assays.
  • Nitrocellulose filters (0.2 ⁇ m pore size, Schleicher and Schuell, Keene, NH) were secured on a filter manifold and washed with 4-10 ml of buffer. Following incubations of 32 P labeled RNA with serial dilutions of the protein for 10 min at 37°C in buffer (PBS) containing 0.01% human serum albumin (HSA) , the solutions were applied to the filters under gentle vacuum in 45 ml aliquots and washed with 5 ml of PBS. The filters were than dried under an infrared lamp and counted in a scintillation counter.
  • PBS buffer
  • HSA human serum albumin
  • RNA ligands exhibited biphasic binding to VEGF. For those ligands, binding of RNA to VEGF is described by a model where total RNA is assumed to be partitioned between two non-interconverting components (Rl and R2) that bind to VEGF with different affinities (eqs 3 and 4) .
  • ⁇ l and ⁇ 2 are the mole fractions of Rl and R2 and Kdl and Kd2 are the corresponding dissociation constants .
  • RNA ligands used for binding studies were prepared by in vi tro transcription using T7 RNA polymerase (Milligan et al . (1987) Nucl . Acids Res. 11:8783) and were purified on denaturing polyacrylamide gels to ensure size homogeneity. All RNA ligands were diluted to about 1 nM in PBS, denatured at 90°C for 2 minutes, and then cooled on ice prior to incubation with the protein. This denaturation/renaturation cycle performed at high dilution is necessary to ensure that the RNA is essentially free from dimers and other higher order aggregates.
  • VEGF ligands were radiolabeled at the 5' -end with ⁇ - 3 P- ATP (New England Biolabs, Beverly, MA) and T4 polynucleotide kinase (New England Biolabs, Beverly, MA) for the 3 ' -boundary determinations, or at the 3'-end with c_- 32 PCp and T4 RNA ligase (New England Biolabs) for the 5'-boundary determination.
  • Radiolabeled RNA ligands were subjected to partial alkaline hydrolysis and then selectively bound in solution to VEGF at 5, 0.5, or 0.125 nM before being passed through nitrocellulose filters.
  • oligonucleotides were resolved on 8% denaturing polyacrylamide gels.
  • the smallest radiolabeled oligonucleotide bound by VEGF at the lowest protein concentration defines the information boundary.
  • Partial digests of the 5'- or the 3' -labelled RNA ligands with RNAse T-L were used to mark the positions of labeled oligonucleotides ending with a guanosine.
  • VEGF Receptor Binding.
  • VEGF was radioiodinated by the Iodegen method (Jakeman et al . (1992) J. Clin. Invest. 8_9:244) to a specific activity of 2.4 x 10 4 cpm/ng.
  • Human umbilical vein endothelial cells (HUVECs) were plated in 24-well plates at a density of 1-2 x 10 5 cells/well and grown to confluence in EGM (Clonetics, San Diego, CA) media (24-48 hrs) .
  • VEGF vascular endothelial growth factor
  • EGF EGF
  • RNA unlabeled competitor
  • placental RNase inhibitor Promega, Madison, Wl
  • HUVECs were then lysed with 1% triton X-100/1 M NaOH and the amount of cell-associated 125 I-VEGF determined by gamma counting.
  • RNA molecules randomized at thirty contiguous positions were used in the initial selection targeting VEGF. Random RNA bound to VEGF with an affinity of approximately 0.2 uM. After 13 rounds of SELEX, the observed improvement in affinity of the evolved RNA pool was about two orders of magnitude (data not shown) . 64 isolates were cloned and sequenced from this evolved pool, and 37 unique sequences found (sequences differing at only one or two positions were not considered unique) . 34 of the 37 unique sequences could be classified into six families based on sequence similarity in the evolved region (Figure 27) .
  • Consensus Structures In addition to allowing determination of consensus primary structures, groups of similar sequences consisting of members that share a defined functional property often contain useful clues for secondary structure prediction (James et al . (1988) Meth. Enzymol. 180 :227) .
  • the underlying assumption is that ligands with similar primary structures are capable of adopting similar secondary structures in which the conserved residues are organized in unique, well-defined motifs.
  • ligands which have strong, unambiguous secondary structures can provide good structural leads for other sequences within a similar set where consensus folding may be less obvious.
  • conserved elements of secondary structure such as base-pairing, may also be detected through covariation analysis of aligned sequence sets (James et al .
  • Base-pairing covariation is noted between positions 5 and 36 (C-G, 2/7; G-C, 2/7; U-A, 1/7; G-U, 1/7) , 6 and 35 (A-U, 4/7; C-G, 1/7; G-C, 1/7) , 7 and 34 (A-U, 4/7; G-C, 1/7) , 11 and 28 (C-G, 6/7; G-C, 1/7) , 12 and 27 (G-U, 6/7; C-G, l/7) , 13 and 26 (A-U, 5/7; G-C, 1/7; G-U, 1/7) , 14 and 25 (G-C, 4/7; C-G, 2/7) and 15 and 24 (C-G, 4/7; G-C, 2/7) .
  • Family 3 and family 4 sequence sets are characterized by highly conserved contiguous stretches of 21 (GGGAACCUGCGU(C/U)UCGGCACC (SEQ ID NO:92) , positions 11-31) and 15 (GGUUGAGUCUGUCCC (SEQ ID NO: 93) , positions 15-29) arranged in bulged hairpin motifs ( Figure 28) (SEQ ID NOS:88-89) .
  • Base-pairing covariation is detected in family 3 between positions 8 and 33 (A-U, 2/4; G-C, 2/4) , 9 and 32 (A-U, 2/4; U-A, 1/4; G-C, 1/4) , and 10 and 31 (A-U, 1/4; G-C, 3/4) and in family 4 between positions 13 and 31 (A-U, 4/7; C-G, 2/7; U-A, 1/7) and 14 and 30 (C-G, 3/7; U-A, 3/7; A-U, 1/7) .
  • EXAMPLE 14 SPECIFICITY OF TRUNCATED RNA LIGANDS TO VEGF. Binding of two truncated high-affinity ligands, lOOt (SEQ ID NO:95) and 44t (SEQ ID NO:96), to four other heparin binding proteins (bFGF, PDGF, antithrombin III and thrombin) was tested in order to address the question of specificity. Dissociation constants were determined using the nitrocellulose filter partitioning technique. Results are shown in Table 2. Binding of these ligands to VEGF in a buffer containing 10 mM dithiothreitol is at least 1000-fold weaker.
  • RNA ligands representing each of the sequence families (lOOt (SEQ ID NO:95) , family 1; 44t (SEQ ID NO:96) , family 2; 12t (SEQ ID NO:97), family 3; 40t (SEQ ID NO:98), family 4; 84t (SEQ ID NO:99) , family 5; and 126t (SEQ ID NO:100), family 6) , as well as random RNA were tested for their ability to inhibit binding of VEGF to its cell-surface receptors. All high- affinity ligands, but not random RNA, inhibited VEGF-VEGF receptor interaction in a concentration-dependent manner with half-inhibition occurring in the 20-40 nM range ( Figure 32) .
  • VEGF vascular endothelial growth factor
  • a and B SELEX experiments targeting VEGF were initiated with separate pools of randomized RNA containing amino (NH 2 ) functionalities at the 2' -position of each pyrimidine.
  • Starting ligand pools for the two experiments contained approximately 10 14 molecules (500 pmols) of modified RNA randomized at 30 (SELEX experiment A) and 50 (SELEX experiment B) contiguous positions.
  • the starting RNAs and the corresponding PCR primers are defined in Figure 33 (SEQ ID NOS :101-106) . Sequences corresponding to the evolved regions of modified RNA are shown in Figure 34.
  • Ligands with similar primary structures were grouped into 5 families and their consensus sequences are shown below each sequence set Figure 34 (SEQ ID NOS:107-190) . Groups of sequences with similar primary structure (families) have been aligned in Figure 34 and their consensus sequences are shown below each set. Pairs of similar/related sequences, sequences that could not be included in any of the families ("other sequences") and sequences that correspond to ligands that bind additionally to nitrocellulose filters with high affinity have been shown in separate groups. Letter N in a sequence indicates an ambiguous position on a sequencing gel. Italicized letter N in a consensus sequence indicates a position that is not conserved (i.e., any nucleotide may be found at that position) . Dissociation constants for Random RNA A (30N8), Random RNA B (50N7) and a set of modified 2' -amino pyrimidine high-affinity RNA ligands for VEGF are shown in Table 3.
  • the first round random pool of RNA was derived from the transcription of a DNA template containing a forty-nucleotide random region flanked by defined regions; the complexity of the pool was in the order of 10 14 molecules.
  • the DNA template and the corresponding PCR primers were synthesized chemically by standard techniques using cyanoethyl phosphoramidites (Operon) .
  • the random region was generated by reacting with all four phosphoramidites at optimized concentrations of each to obtain equimolar coupling during oligonucleotide synthesis.
  • nucleotide sequences in the flanking regions of the template served as primer annealing sites for PCR, for reverse transcriptase and as a promoter for T7 RNA polymerase, and restriction enzyme sites that allow cloning into vectors.
  • the SELEX was done with the primer-template mixture I, shown in Figure 46.
  • 2' -NH 2 RNA transcriptions were carried out in 100 ⁇ l transcription reactions containing 2 mM each ATP, GTP, 2'-NH 2 CTP and 2'-NH 2 UTP, 40 mM Tris- HC1 (pH 8.0) , 12 mM MgCl 2 , 1 mM Spermidine, 5 mM DTT, 0.002% Triton X-100 and 4% polyethylene glycol (w/v) at room temperature for 2 hr.
  • the full length transcripts were gel purified, resuspended in binding buffer (150 mM NaCl, 100 mM Tris-HCl (pH 7.0) , 2 mM MgCl 2 and 6 mM KC1) , heated to 70°C for 3 min., chilled on ice, and incubated with HNE at 37°C for 10 min.
  • binding buffer 150 mM NaCl, 100 mM Tris-HCl (pH 7.0) , 2 mM MgCl 2 and 6 mM KC1
  • RNA pool was incubated with elastase (1.7-0.003 ⁇ M) in binding buffer containing 150 mM NaCl, 100 mM Tris-HCl (pH 7.0) , 2 mM MgCl 2 , and 6 mM KCl for 10 minutes at 37°C.
  • the RNA-protein mixture was passed through a nitrocellulose filter (0.45 ⁇ , Millipore) and the RNA from RNA-protein complexes retained on the filter was eluted (Tuerk & Gold (1990) Science 249 :505) and recovered.
  • the recovered RNA was reverse transcribed into cDNA by AMV reverse transcriptase at 48°C.
  • the resulting cDNA was amplified by PCR and the amplified DNA used as the template for T7 RNA polymerase to obtain a pool of RNA for the subsequent round of SELEX.
  • the newly transcribed RNA pool was passed through several layers of nitrocellulose filters (negative selection) before the next round of selection with HNE. The procedure was reiterated until the enriched pool of RNA showed significantly improved affinity to elastase over the initial random pool.
  • PCR-amplified DNA was digested with HindiII and BamHI, and cloned into pUC18 previously digested with the same enzymes by standard techniques (Tuerk & Gold (1990) Science 249 :505) . Recombinant plasmids were isolated and sequenced.
  • Single-Stranded DNA SELEX Single-Stranded DNA SELEX.
  • SELEX was conducted with a candidate library of single-stranded DNA (ssDNA) molecules.
  • the SELEX experiment was carried out by using the primer template-mixture II, shown in Figure 46. Briefly, after nitrocellulose filter partitioning, filter bound ssDNA was recovered by incubating the filter in a mixture of neutralized phenol and 7 M urea at 37°C followed by extraction of the aqueous phase. Recovered ssDNA was precipitated with ethanol, and an aliquot used as a template in a PCR reaction to identify the optimum number of cycles needed to produce double-stranded DNA (dsDNA) molecules of the correct size without creating high molecular weight DNAs.
  • dsDNA double-stranded DNA
  • the optimum number of cycles was used for a bulk PCR reaction (800 ul) carried out with the remaining recovered ssDNA as the template and a biotinylated primer (BIO-3G2) and a non-biotinylated primer (5G2) .
  • the PCR products are gel purified to remove unincorporated biotinylated primer.
  • the non- biotinylated strand of the double-stranded DNA was isolated from the complementary strand by incubation of the PCR product with 200 ul of streptavidin beads (Pierce) followed by alkali denaturation (0.15 N NaOH, 30 min at room temperature) .
  • the recovered single-stranded DNA was neutralized, precipitated with ethanol, and used for the next round of SELEX.
  • ssDNA was passed through a layer of 4-5 nitrocellulose filters after every SELEX round to remove those DNA molecules that bound to the filter, not elastase (filter binders) .
  • filter binders elastase
  • the percentage of filter binders increased from 0.2% to as high as 12-15%.
  • the filters were washed with 0.5 M urea solution immediately after the filtration of the protein-DNA mixture.
  • the urea treatment brought background binding levels to 0.1-0.2%. Therefore, during each SELEX round as well as during determination of binding curves, the filters were washed with 10 ml of 0.5 M urea and 10 ml of binding buffer.
  • the assay was conducted in a solution containing 10 nM elastase, 0.5 mM N- methoxysuccinyl-Ala-Ala-Pro-Val-p-nitroanilide, 150 mM NaCl, 26 mM KCl, 2 mM MgCl 2 , 0.02% HSA, 0.05% DMSO, 0.01% Triton X-100, and 100 mM Tris-HCl, at 25°C.
  • the assay measured by spectroscopy (OD 405 nm) the generation of elastase-induced release of p-nitroanilide as a function of time.
  • the rate of p-nitroanilide generation in the absence of inhibitor was used as the negative control; the positive control contained the irreversible inhibitor N-methoxysuccinyl-Ala-Ala-Pro-Val-chloromethyl ketone. OD values were plotted against time, and the slope used to determine the rate of enzyme activity.
  • Cell detachment assay An assay was developed in which elastase-induced endothelial cell detachment is measured in cultured endothelial cells. Endothelial cells were grown under standard culture conditions in 24- well plates to obtain a confluent monolayer.
  • the cells were washed with solution A (PBS, 20 mM HEPES, 10 mM glucose) prior to addition of elastase with and without the test ligand. After incubation at 37°C for 45 minutes, the supernatant was removed and counted for detached cells.
  • solution A PBS, 20 mM HEPES, 10 mM glucose
  • EXAMPLE 18 IDENTIFICATION OF INHIBITORY LIGANDS TO ELASTASE.
  • RNA ligands with inhibitory activity toward elastase were identified. Molecules belonging to class I and II were found to inhibit elastase. Clone 9 (SEQ ID NO:202) of class III and clone 19 (SEQ ID NO:205) of class IV also exhibited inhibitory activity.
  • Figure 36 shows the elastase inhibitory activity of representative ligand 34 (SEQ ID NO:197) . The elastase inhibitory activity of all RNA ligands tested was dependent on RNA concentration and had a maximum inhibition of elastase of approximately 30%.
  • the chloromethyl ketone portion of the inhibitor irreversibly binds at the active site of elastase, with the tetrapeptide portion occupying the substrate binding pocket (Navia et al . (1989) Proc. Natl. Acad. Sci. USA 8£:7) .
  • the results, shown in Figure 38, show that ligand 34 (SEQ ID NO:197) bound to complexed elastase to the same extent as to the native enzyme. These results support the conclusion that the ligands do not bind at the active site or the substrate binding pocket of elastase.
  • Ligand-mediated elastase inhibition may come from either ligand binding near the active site or to a remote site of the enzyme surface, inducing an allosteric change in the enzyme which impairs catalytic activity to some extent.
  • a natural inhibitor for elastase, Qf-1-antitrypsin (0.-1PI) forms a tight irreversible enzyme-inhibitor complex with elastase.
  • the binding of ligand 34 SEQ ID NO: 197) to o;-IPI-complexed elastase was examined. The results show ( Figure 39) that ligand 34 did not bind the complexed enzyme.
  • ligand binds to a site on the face of elastase that interacts with ⁇ -lPI.
  • c_- 1PI binding may induce a conformation change in elastase that abolishes ligand binding.
  • Representative ligand 19 (SEQ ID NO:205) was found to act additively with 0.-1PI inhibition, enhancing the inhibitory activity of the natural inhibitor ( Figure 40) .
  • a cell detachment assay was conducted with inhibitory ligands 24, 30, and 19 (SEQ ID NOS:198, 191 &. 205) . As shown in Figure 41, all of the ligands substantially inhibited elastase activity.
  • the binding buffer employed with 2' -NH 2 SELEX contained 6 mM KCl, a monovalent ion known to stabilize the G-quartet structure.
  • the absence of KCl in the binding buffer affects the binding affinity of three clones (21, 24 and 34) (SEQ ID NOS:195, 198 & 197) .
  • SEQ ID NOS:195, 198 & 197 As shown in Figure 43, there was a significant change in Kd values in the presence and absence of KCl.
  • clone 21 (SEQ ID NO:195) had a Kd for elastase of 39 nM, in the presence of KCl, which dropped to 192 nM in the absence of KCl. This suggests that KCl has an important role in ligand binding, perhaps by favoring the formation of secondary structures with high binding affinity to elastase.
  • Ligand binding to elastase was then investigated with ligands 30 (SEQ ID N0:191) (class I) , 34 (SEQ ID NO:
  • ligand 9 SEQ ID NO:202 (class III) had lower elastase affinity in the presence of high KCl, suggesting that the secondary structure of ligand 9 may be different from that assumed by class I and II ligands.
  • the lowest affinity of class I and II ligands to elastase was seen in the presence of Li, the monovalent cation that does not favor formation of G-quartet structures.
  • Tris buffer all three ligands exhibited a moderately high affinity for elastase (however, lower than that of class I and II ligands in KCl buffer) .
  • Mg ++ ions may be involved in establishment of secondary structures in the absence of a high concentration of a monovalent ion.
  • EXAMPLE 21 EFFECT OF pH ON 2' -NH RNA AND NON- MODIFIED SINGLE-STRANDED DNA LIGAND BINDING TO ELASTASE.
  • the pKa of 2'-NH 2 -NTPs is approximately 6.5, not distant from the physiological pH range of 7.35-7.45. Therefore, at a physiological pH, a given RNA molecule containing 2' -NH 2 -modified NTPs should have a distribution of protonated and unprotonated 2'-NH 2 groups. Consequently, it is likely that a small change in pH can affect the degree of protonation of a ligand, which may affect its binding to a target molecule.
  • the binding to elastase of representative 2'-NH 2 RNA ligand 14 and non-modified single-stranded DNA ligand 17 was examined over a pH range of 5.0-8.0. The results are shown in Figure 44.
  • the binding of the 2' -NH 2 RNA ligand was profoundly affected by pH. For example, the Kd increased more than 10 fold from pH 6.5 to pH 6.25.
  • the optimum binding for the 2'-NH 2 ligand is seen at pH 7.0, the pH at which SELEX was conducted.
  • the connecting loop nucleotides of the G-quartet structure was replaced with a synthetic linker tether group ( Figure 45) .
  • DNA-17 (TAGCGATACTGCGTGGGTTGGGGCGGGT AGGGCCAGCAGTCTCGTGCGGTACTTGAGCA) (SEQ ID NO:52) was synthesized with two loop nucleotides replaced by a 18- carbon ethylene glycol linker (Spacer phosphoramidite; Clonetech) to obtain a mimetic sequence, 17-LQT(TAGCGAT
  • L stands for the ethylene glycol linker.
  • the length of the internucleotide phosphate is about a 3- carbon distance.
  • the 18-carbon linker was expected to have enough flexibility to loop back into a G-quartet.
  • 17-LQT was found not to bind elastase with significant affinity, however.
  • the absence of high affinity binding could be due to lack of loop nucleotides that are crucial for protein recognition or due to the synthetic tether restricting the oligonucleotide to fold into a tetraplex.
  • EXAMPLE 23 METHODS AND MATERIALS -NUCLEIC ACID LIGANDS TO THEOPHYLLINE AND CAFFEINE. Materials . 1-carboxypropyl theophylline was provided by Abbott Laboratories, Abbott Park, IL.
  • SELEX The essential features of the SELEX protocol are described in Example 1. Briefly, DNA templates for in vi tro transcription (that contain a region of forty random positions flanked by constant sequence regions) and the corresponding PCR primers were synthesized chemically (Operon) . The random region was generated by utilizing an equimolar mixture of the four nucleotides during oligonucleotide synthesis. The two constant regions were designed to contain PCR primer annealing sites, a primer annealing site for cDNA synthesis, T7 RNA polymerase promoter region, and restriction enzyme sites that allow cloning into vectors. Theophylline SELEX experiments utilized 100 mM HEPES buffer (100 mM HEPES, pH 7.3, 0.5 M NaCl and 5 mM MgCl 2 ) .
  • Eguilibrium filtration analysis A rapid procedure was developed to assess theophylline binding by oligonucleotides, called “equilibrium filtration” . These assays were performed by the addition of [ 14 C] - theophylline and RNA at the indicated concentrations (see Figure 51) in a 150 ⁇ l reaction mixture containing 100 mM HEPES (pH 7.3), 5 mM MgCl 2 , and 50 mM NaCl. Each binding mixture was incubated 5 min at 25 °C. The mixture was then placed on a Microcon 10 (Amicon) filtration device and centrifuged 4 min at 14,000 x g.
  • RNA is retained by the filter while theophylline passes freely through the filter.
  • a 25 ⁇ l sample was removed from each side of the filter and the radioactivity determined by scintillation counting. Each reaction was performed in triplicate. Other experiments established that this procedure provided results similar to that obtained by equilibrium dialysis. Equilibrium dialysis analysis of binding was carried out under identical reaction conditions to those described above. Each binding mixture was placed in a Spectra dialyzer (Spectrum) for 2 hr at 25 °C. Radioactivity retained inside the chamber was determined. Data were fit by least squares to a quadratic binding equation assuming a 1:1 stoichiometry (Gill et al . (1991) J. Mol. Biol. 220:307) ( Figure 51) .
  • RNA ligands to Theophylline To screen for RNA molecules with affinity for theophylline, a pool of 10 14 random RNAs were generated, each containing a 40 nucleotide random region flanked by fixed sequences for transcription and PCR amplification. These RNAs were generated by transcription of double-stranded DNA by T7 RNA polymerase as described above, and were radiolabelled with [ex] 32 P-ATP.
  • RNA was added to an EAH Sepharose column to which 1- carboxypropyl theophylline (1-cp theophylline) was covalently bound via an amide linkage using the coupling reagents l-ethyl-3 (3-dimethylaminopropyl carbodimide (EDC) and 60 N-hydroxysuccinimide (NHS) .
  • EDC electrophilicity-sensitive polymer
  • NHS N-hydroxysuccinimide
  • Bound RNA was partitioned by eluting with 0.1 M theophylline.
  • the eluted RNA was precipitated, reverse transcribed with AMV reverse transcriptase, and the resulting DNA amplified by PCR as described above.
  • This set of procedures constituted one SELEX selection round.
  • the general progress of the experiment over eight selection rounds was monitored by determining the percent of the labelled RNA input to each selection round that was specifically eluted from the theophylline column. About 0.05% of the random RNA in the first selection round was eluted from the column. After eight rounds of selection, approximately 62% of the input RNA was eluted by theophylline, representing an affinity enrichment of about 1200-fold, relative to the starting population.
  • This SELEX experiment was designated TR8.
  • LIGANDS TCT8.
  • a variation on the initial SELEX partition protocol was carried out after the fifth SELEX selection round.
  • a different elution protocol was used to increase the stringency of the SELEX process. Rather than eluting with theophylline directly after washing, bound RNAs were first subjected to challenge with 0.1 M caffeine after the fifth SELEX selection round. Remaining RNAs were eluted with 0.1 M theophylline.
  • TCT8 RNAs Approximately 80% of the TCT8 RNAs bound directly to theophylline columns, representing an affinity enrichment of approximately 4000-fold relative to that of the starting population. Of the bound RNAs, 54% were resistant to caffeine challenge and were eluted subsequently by theophylline.
  • RNA ligands with high affinity to caffeine were identified in a similar fashion to the experiments described in Example 23. Eight selection rounds of SELEX were performed using Sepharose-bound caffeine as the molecular target . Bound RNAs were eluted with excess free caffeine. Enrichment of the populations of RNAs for those with affinity for caffeine proceeded similarly to the theophylline SELEX experiment. In the first selection round, about 0.01% of the random RNA was eluted with caffeine. After eight selection rounds, 52.5% of the input RNA was eluted with caffeine. Twenty-one bacterial clones were generated from this population, designated CR8 and sequenced as described in Example 23. CR8 sequences are shown in Figure 54.
  • RNAs selected for high affinity to caffeine are distinct from those selected to bind to theophylline.
  • the binding properties of the CR8 RNA pool with 14 C-caffeine were investigated by equilibrium dialysis. The CR8 RNA pool bound caffeine with a Kd of 15 ⁇ M, an affinity not dissimilar from that of the theophylline binding RNA population for theophylline after eight selection rounds.
  • Truncated RNA ligand to Theophylline (mTCT8-4 (SEQ ID NO:286)) .
  • the mini- TCT8-4 RNA construct (mTCT8-4 (SEQ ID NO:286)) ( Figure 52) is a 39 nucleotide T7 RNA polymerase transcript produced from a DNA template designed from the sequence of TCT8-4 RNA.
  • TCT8-4 present in mTCT8- 4 are shown in Figure 49.
  • mTCT8-4 contains motifs 1 and 2, and the 6 bp vsteml and 4 bp vstem2 regions of TCT8-4. It lacks the 24 nucleotide 3' fixed region of the TCT8-4 RNA.
  • mTCT8-4 RNA was produced by T7 RNA polymerase in vi tro, gel purified, and tested for its ability to bind to 14 C-theophylline by equilibrium filtration analysis. A Kd of 0.11 ⁇ M was determined for mTCT8-4 binding to theophylline ( Figure 51) .
  • EXAMPLE 27 COMPETITION BINDING STUDIES WITH XANTHINE DERIVATIVES.
  • RNA binding surface Several features of the RNA binding surface are revealed by these studies. First, these results emphasize the major role played by the 7-position of xanthine derivative in recognition by the SELEX RNA. 7- methylxanthine competes at least 1500-fold less efficiently than xanthine, which itself is about 23-fold less effective than theophylline. The relatively good competition by xanthine indicates that the unadorned planar ring system is tightly accommodated by the RNA binding surface. As expected from the behavior of 7- methylxanthine, theobromine (3, 7-dimethylxanthine) is also recognized poorly by TCT8-4 RNA.
  • the 3-position provides positive binding energy as evidenced by the approximately 5-fold better competition of 3- methylxanthine relative to xanthine.
  • TCT8-4 RNA is relatively blind to the 1-position methyl, relative to xanthine.
  • Hypoxanthine is bound about 5-fold more poorly by the RNA relative to xanthine, indicating that the 2-position keto oxygen is also important in the recognition.
  • 1,3-dimethyl uric acid is very poorly recognized compared to theophylline, indicating that the binding site is highly sensitive to the 8- position.
  • TCT8-4 (SEQ ID NO:249) can discriminate between theophylline and caffeine at least 9000-fold, corresponding to a ⁇ G of about 5.6 kcal/mol. Relative to the truncated version of TCT8-4, mTCT8-4 (SEQ ID NO:286) , the discrimination is approximately 60, 000-fold.
  • TCT8-4 RNA contains a binding surface or pocket that closely inspects the 7- and 8-positions of these xanthine derivatives, and discriminates between theophylline and caffeine largely on the basis of its inability to accommodate the 7-position methyl group of caffeine.
  • the conserved sequences present in each of the class I and II molecules may be arranged in space in a way that ultimately rationalizes their high discrimination in recognizing theophylline.
  • molecular modeling, energy minimization and molecular dynamics of the conserved region and flanking sequences were used to model a small oligonucleotide composed of the conserved sequences and limited flanking regions.
  • This model incorporates A-form helical parameters derived from tRNA crystal data to model those portions predicted to be helical. Bulged nucleotides are believed to introduce local kinking into the helical axis (Riordan et al .
  • the three base pyrimidine bulge was modeled as stacked within the flanking helical regions based on similar tracts that have been analyzed in the HIV TAR RNA by NMR analysis (Puglisi et al . (1992) Science 257 :76) . In addition, preliminary nuclease mapping experiments indicate that the two C residues in this bulge are not accessible to nuclease CL3 (data not shown) .
  • the three base symmetric bulge was modeled using a central UA Watson-Crick base pair, flanked by two non- Watson-Crick CA base pairs, AC and AG.
  • Target Molecule lOOt Kd 44t (Kd) (SEQ ID NO: 5) (SEQ ID N0:96) bFGF 1 uM 0.6 uM

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Analytical Chemistry (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Virology (AREA)

Abstract

L'invention concerne des procédés d'identification et de production d'acides nucléiques servant de ligands avec une molécule cible par le procédé SELEX (évolution systématique de ligands par enrichissement exponentiel). On peut utiliser de tels procédés avec des acides nucléiques modifiés ou mélangés, selon des phases séparées destinées à mettre le mélange d'acides nucléiques condidat en contact avec des molécules cibles et non cibles. L'invention englobe des procédés d'identification et de préparation d'acides nucléiques servant de ligands avec la thrombine, le facteur de croissance endothéliale vasculaire, l'élastase de granulocytes neutrophiles humains, la caféine et la théophylline, ainsi qu'avec la thrombine quand ils sont identifiés par le procédé SELEX.
EP94927409A 1993-09-08 1994-09-08 Acides nucleiques servant de ligands et procedes de production ameliores Ceased EP0724647A4 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP06006842A EP1793006A3 (fr) 1993-09-08 1994-09-08 Ligands d'acide nucléique et méthode pour les produire

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US117991 1987-11-09
US11799193A 1993-09-08 1993-09-08
US13402893A 1993-10-07 1993-10-07
US134028 1993-10-07
US08/199,507 US5472841A (en) 1990-06-11 1994-02-22 Methods for identifying nucleic acid ligands of human neutrophil elastase
US199507 1994-02-22
US08/233,012 US5849479A (en) 1990-06-11 1994-04-25 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US08/234,997 US5683867A (en) 1990-06-11 1994-04-28 Systematic evolution of ligands by exponential enrichment: blended SELEX
US234997 1994-04-28
PCT/US1994/010306 WO1995007364A1 (fr) 1993-09-08 1994-09-08 Acides nucleiques servant de ligands et procedes de production ameliores
US233012 1999-01-19

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP06006842A Division EP1793006A3 (fr) 1993-09-08 1994-09-08 Ligands d'acide nucléique et méthode pour les produire

Publications (2)

Publication Number Publication Date
EP0724647A1 EP0724647A1 (fr) 1996-08-07
EP0724647A4 true EP0724647A4 (fr) 2003-09-17

Family

ID=27537502

Family Applications (2)

Application Number Title Priority Date Filing Date
EP94927409A Ceased EP0724647A4 (fr) 1993-09-08 1994-09-08 Acides nucleiques servant de ligands et procedes de production ameliores
EP06006842A Withdrawn EP1793006A3 (fr) 1993-09-08 1994-09-08 Ligands d'acide nucléique et méthode pour les produire

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP06006842A Withdrawn EP1793006A3 (fr) 1993-09-08 1994-09-08 Ligands d'acide nucléique et méthode pour les produire

Country Status (5)

Country Link
EP (2) EP0724647A4 (fr)
JP (2) JPH09502354A (fr)
AU (1) AU692469B2 (fr)
CA (1) CA2169536A1 (fr)
WO (1) WO1995007364A1 (fr)

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6001577A (en) 1998-06-08 1999-12-14 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: photoselection of nucleic acid ligands and solution selex
US6395888B1 (en) * 1996-02-01 2002-05-28 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US5789163A (en) * 1990-06-11 1998-08-04 Nexstar Pharmaceuticals, Inc. Enzyme linked oligonucleotide assays (ELONAS)
US5705337A (en) * 1990-06-11 1998-01-06 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chemi-SELEX
US6344321B1 (en) * 1990-06-11 2002-02-05 Gilead Sciences, Inc. Nucleic acid ligands which bind to hepatocyte growth factor/scatter factor (HGF/SF) or its receptor c-met
US5972599A (en) * 1990-06-11 1999-10-26 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of cytokines
US6762290B1 (en) * 1999-07-29 2004-07-13 Gilead Sciences, Inc. High affinity vascular endothelial growth factor (VEGF) receptor nucleic acid ligands and inhibitors
US6458539B1 (en) 1993-09-17 2002-10-01 Somalogic, Inc. Photoselection of nucleic acid ligands
EP0736105B1 (fr) * 1993-09-17 2007-11-21 SomaLogic, Inc. Evolution systematique de ligands par enrichissement exponentiel : photoselection de ligands d'acide nucleique
ATE274520T1 (de) * 1995-05-03 2004-09-15 Gilead Sciences Inc Systematische evoultion von liganden durch exponentielle anreicherung: gewebe-selex
US6013443A (en) 1995-05-03 2000-01-11 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue SELEX
CA2221318C (fr) * 1995-06-02 2012-01-24 Nexstar Pharmaceuticals, Inc. Ligands oligonucleotidiques ayant une affinite elevee pour les facteurs de croissance
CA2223003A1 (fr) * 1995-06-07 1996-12-19 Nexstar Pharmaceuticals, Inc. Ligands de cytokines a acide nucleique et d'une affinite elevee
AU773815B2 (en) * 1995-06-07 2004-06-10 Gilead Sciences, Inc. High-affinity nucleic acid ligands of cytokines
US6229002B1 (en) 1995-06-07 2001-05-08 Nexstar Pharmaceuticlas, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
DE69739880D1 (de) * 1996-02-01 2010-07-01 Gilead Sciences Inc NUKLEINSÄURELIGANDEN MIT HOHER AFFINITÄT FÜR DAS C1q PROTEIN DES KOMPLEMENTSYSTEMS
US6838238B1 (en) 1996-10-17 2005-01-04 Invitrogen Corporation Morphatides: novel shape and structure libraries
US6426335B1 (en) 1997-10-17 2002-07-30 Gilead Sciences, Inc. Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
US6287765B1 (en) * 1998-05-20 2001-09-11 Molecular Machines, Inc. Methods for detecting and identifying single molecules
DE19909156A1 (de) * 1999-03-02 2000-09-07 Aventis Res & Tech Gmbh & Co Testsystem zum Nachweis einer Spleißreaktion, sowie dessen Verwendung
US7074586B1 (en) 1999-06-17 2006-07-11 Source Precision Medicine, Inc. Quantitative assay for low abundance molecules
EP1111069A1 (fr) * 1999-12-22 2001-06-27 BioChip Technologies GmbH Des acides nucléiques modifiés et leur utilisation
US8030465B2 (en) 2001-06-29 2011-10-04 Medimolecular Pty Ltd Nucleic acid ligands to complex targets
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
WO2004050899A2 (fr) 2002-12-03 2004-06-17 Archemix Corporation Methode de selection in vitro d'acides nucleiques 2'-substitues
US7803931B2 (en) 2004-02-12 2010-09-28 Archemix Corp. Aptamer therapeutics useful in the treatment of complement-related disorders
SG157417A1 (en) * 2004-11-29 2009-12-29 Noxxon Pharma Ag Vasopressin-binding l-nucleic acids
CA2607185A1 (fr) * 2005-05-04 2006-11-09 Noxxon Pharma Ag Nouvelle utilisation de spiegelmers
WO2007004748A1 (fr) 2005-07-05 2007-01-11 Ribomic Inc. Acide nucléique capable de se lier à une immunoglobuline g et son utilisation
US7947447B2 (en) 2007-01-16 2011-05-24 Somalogic, Inc. Method for generating aptamers with improved off-rates
EP2172566B2 (fr) * 2007-07-17 2022-05-18 Somalogic, Inc. Procédé de génération d'aptamères avec des vitesses d'arrêt améliorées
EP2316935B1 (fr) * 2008-07-14 2016-08-31 The University of Tokyo Aptamère à l'encontre d'il-17 et son utilisation
RU2011116175A (ru) 2008-09-24 2012-10-27 Рибомик Инк. (Jp) Аптамер к ngf и его применение
AR077049A1 (es) 2009-06-11 2011-07-27 Otsuka Pharma Co Ltd Aptamero contra quimasa y su uso
US8772259B2 (en) 2010-02-12 2014-07-08 Ribomic Inc. Aptamer to FGF2 and use thereof
US9175292B2 (en) 2010-03-24 2015-11-03 Fujimoto Pharmaceutical Corporation Aptamer for NGF and use thereof
DK2762569T3 (en) 2011-09-28 2018-08-27 Ribomic Inc NGF aptamer and its use.
SG11201402402SA (en) 2011-11-18 2014-09-26 Tagcyx Biotechnologies Nucleic acid fragment binding to target protein
EP2977454B1 (fr) 2013-03-22 2018-05-02 The University of Tokyo Aptamère pour il-17 et utilisation de celui-ci
CN106103719B (zh) 2014-03-24 2019-12-13 力博美科股份有限公司 针对fgf2的适体及其应用
CN106232817A (zh) 2014-04-24 2016-12-14 力博美科股份有限公司 结合自分泌运动因子并且抑制自分泌运动因子的生物学活性的适体及其应用
GB201615989D0 (en) * 2016-09-20 2016-11-02 Imp Innovations Ltd Drug Delivery
EP3708666A4 (fr) 2017-11-09 2021-07-21 Ribomic Inc Aptamer pour adamts5 et utilisation pour aptamer pour adamts5
US11639503B2 (en) 2017-11-30 2023-05-02 Ribomic Inc. Anti-chymase aptamer and use for same
IL299193A (en) 2020-06-17 2023-02-01 Ribomic Inc Aptamer for IL-21 and its use

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991019813A1 (fr) * 1990-06-11 1991-12-26 The University Of Colorado Foundation, Inc. Ligands d'acide nucleique
WO1994008050A1 (fr) * 1992-09-29 1994-04-14 Nexagen, Inc. Ligands d'acide nucleique et procedes de production

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5476766A (en) * 1990-06-11 1995-12-19 Nexstar Pharmaceuticals, Inc. Ligands of thrombin

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991019813A1 (fr) * 1990-06-11 1991-12-26 The University Of Colorado Foundation, Inc. Ligands d'acide nucleique
WO1994008050A1 (fr) * 1992-09-29 1994-04-14 Nexagen, Inc. Ligands d'acide nucleique et procedes de production

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
See also references of WO9507364A1 *
TUERK C ET AL: "RNA PSEUDOKNOTS THAT INHIBIT HUMAN IMMUNODEFICIENCY VIRUS TYPE 1 REVERSE TRANSCRIPTASE", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 89, no. 15, 10 August 1992 (1992-08-10), pages 6988 - 6992, XP002911411, ISSN: 0027-8424 *

Also Published As

Publication number Publication date
AU7686594A (en) 1995-03-27
AU692469B2 (en) 1998-06-11
WO1995007364A1 (fr) 1995-03-16
EP0724647A1 (fr) 1996-08-07
JP2005065701A (ja) 2005-03-17
EP1793006A2 (fr) 2007-06-06
JPH09502354A (ja) 1997-03-11
EP1793006A3 (fr) 2007-08-22
CA2169536A1 (fr) 1995-03-16

Similar Documents

Publication Publication Date Title
AU692469B2 (en) Nucleic acid ligands and improved methods for producing the same
US5958691A (en) High affinity nucleic acid ligands containing modified nucleotides
US9873879B2 (en) Nucleic acid fragment binding to target protein
AU781234B2 (en) High affinity TGF beta nucleic acid ligands and inhibitors
US5811533A (en) High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US6083696A (en) Systematic evolution of ligands exponential enrichment: blended selex
US5580737A (en) High-affinity nucleic acid ligands that discriminate between theophylline and caffeine
AU714469B2 (en) Parallel selex
KR970002255B1 (ko) 핵산 리간드
US5840867A (en) Aptamer analogs specific for biomolecules
EP1683871B1 (fr) Ligands d'acides nucléiques et procédé de leur production
JPH11502105A (ja) 指数濃縮によるリガンドの体系的進化:ケミセレックス
IE920561A1 (en) Aptamer specific for thrombin and methods of use
US5849479A (en) High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US7153948B2 (en) High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US20030032785A1 (en) High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US20060084797A1 (en) High affinity TGFbeta nucleic acid ligands and inhibitors
AU2004242532A1 (en) High affinity TGF beta nucleic acid ligands and inhibitors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19960403

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: GILEAD SCIENCES, INC.

RIC1 Information provided on ipc code assigned before grant

Ipc: 7C 12N 15/10 B

Ipc: 7C 07H 21/04 B

Ipc: 7C 07H 21/02 B

Ipc: 7C 12Q 1/68 A

A4 Supplementary search report drawn up and despatched

Effective date: 20030804

17Q First examination report despatched

Effective date: 20031211

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20080307