US20070021327A1 - Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor - Google Patents

Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor Download PDF

Info

Publication number
US20070021327A1
US20070021327A1 US11/518,321 US51832106A US2007021327A1 US 20070021327 A1 US20070021327 A1 US 20070021327A1 US 51832106 A US51832106 A US 51832106A US 2007021327 A1 US2007021327 A1 US 2007021327A1
Authority
US
United States
Prior art keywords
pdgf
nucleic acid
nucleic acids
selex
tumors
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/518,321
Inventor
Kristian Pietras
Arne Ostman
Carl-Henrik Heldin
Kristofer Rubin
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 US08/479,725 external-priority patent/US5674685A/en
Priority claimed from US08/479,783 external-priority patent/US5668264A/en
Priority claimed from US08/618,693 external-priority patent/US5723594A/en
Priority claimed from US08/991,743 external-priority patent/US6229002B1/en
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Priority to US11/518,321 priority Critical patent/US20070021327A1/en
Assigned to LUDWIG INSTITUTE FOR CANCER RESEARCH reassignment LUDWIG INSTITUTE FOR CANCER RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HELDIN, CARL-HENRIK, OSTMAN, ARNE, PIETRAS, KRISTIAN
Assigned to GILEAD SCIENCES, INC. reassignment GILEAD SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RUBIN, KRISTOFER
Assigned to GILEAD SCIENCES, INC. reassignment GILEAD SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LUDWIG INSTITUTE FOR CANCER RESEARCH
Publication of US20070021327A1 publication Critical patent/US20070021327A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B50/00Methods of creating libraries, e.g. combinatorial synthesis
    • C40B50/04Methods of creating libraries, e.g. combinatorial synthesis using dynamic combinatorial chemistry techniques
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • A61K47/544Phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/547Chelates, e.g. Gd-DOTA or Zinc-amino acid chelates; Chelate-forming compounds, e.g. DOTA or ethylenediamine being covalently linked or complexed to the pharmacologically- or therapeutically-active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/49Platelet-derived growth factor [PDGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • 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
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • 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
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • 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
    • 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
    • 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
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • 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
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/76Human chorionic gonadotropin including luteinising hormone, follicle stimulating hormone, thyroid stimulating hormone or their receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2310/13Decoys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/317Chemical structure of the backbone with an inverted bond, e.g. a cap structure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/318Chemical structure of the backbone where the PO2 is completely replaced, e.g. MMI or formacetal
    • C12N2310/3183Diol linkers, e.g. glycols or propanediols
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • 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
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F02COMBUSTION ENGINES; HOT-GAS OR COMBUSTION-PRODUCT ENGINE PLANTS
    • F02BINTERNAL-COMBUSTION PISTON ENGINES; COMBUSTION ENGINES IN GENERAL
    • F02B75/00Other engines
    • F02B75/02Engines characterised by their cycles, e.g. six-stroke
    • F02B2075/022Engines characterised by their cycles, e.g. six-stroke having less than six strokes per cycle
    • F02B2075/027Engines characterised by their cycles, e.g. six-stroke having less than six strokes per cycle four
    • 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
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • 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
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • G01N2333/163Regulatory proteins, e.g. tat, nef, rev, vif, vpu, vpr, vpt, vpx
    • 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
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/50Fibroblast growth factors [FGF]
    • G01N2333/503Fibroblast growth factors [FGF] basic FGF [bFGF]
    • 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
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/575Hormones
    • 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
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/575Hormones
    • G01N2333/62Insulins
    • 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
    • G01N2333/96433Serine endopeptidases (3.4.21)
    • G01N2333/96441Serine endopeptidases (3.4.21) with definite EC number
    • G01N2333/96455Kallikrein (3.4.21.34; 3.4.21.35)
    • 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)
    • G01N2333/972Plasminogen activators
    • G01N2333/9726Tissue plasminogen activator
    • 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
    • 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)
    • G01N2333/976Trypsin; Chymotrypsin

Definitions

  • This invention relates generally to a method for increasing the uptake of drugs into tumors by treatment with a nucleic acid ligand to PDGF in combination with a cytotoxic agent.
  • the method used for identifying nucleic acid ligands to PDGF is called SELEX, an acronym for Systematic Evolution of Ligands by Exponential enrichment.
  • the method of the present invention is useful for increasing the therapeutic effectiveness of cytotoxic agents.
  • the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process is a method for the in vitro evolution of nucleic acid molecules with highly specific binding to target molecules and is described in U.S. patent application Ser. No. 07/536,428, filed Jun. 11, 1990, entitled “Systematic Evolution of Ligands by EXponential Enrichment,” now abandoned, U.S. Pat. No. 5,475,096, entitled “Nucleic Acid Ligands,” and U.S. Pat. No. 5,270,163 (see also WO 91/19813), entitled “Methods for Identifying Nucleic Acid Ligands,” each of which is specifically incorporated herein by reference in its entirety.
  • Each of these applications, collectively referred to herein as the SELEX Patent Applications describes a fundamentally novel method for making a nucleic acid ligand to any desired target molecule.
  • the SELEX process provides a class of products which are referred to as nucleic acid ligands or aptamers, each having a unique sequence, and which has 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.
  • the SELEX process 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 or composition can serve as targets.
  • the SELEX method applied to the application of high affinity binding involves selection from a mixture of candidate oligonucleotides and step-wise iterations of binding, partitioning and amplification, using the same general selection scheme, to achieve virtually any desired criterion of binding affinity and selectivity.
  • the SELEX 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 specifically to target molecules, dissociating the nucleic acid-target complexes, amplifying the nucleic acids dissociated from the nucleic acid-target complexes 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 to yield highly specific high affinity nucleic acid ligands to the target molecule.
  • nucleic acids as chemical compounds can form a wide array of shapes, sizes and configurations, and are capable of a far broader repertoire of binding and other functions than those displayed by nucleic acids in biological systems.
  • the SELEX method encompasses the identification of high-affinity nucleic acid ligands containing modified nucleotides conferring improved characteristics on the ligand, such as improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions.
  • SELEX process-identified nucleic acid ligands containing modified nucleotides are described in U.S. Pat. No. 5,660,985, entitled “High Affinity Nucleic Acid Ligands Containing Modified Nucleotides,” that describes oligonucleotides containing nucleotide derivatives chemically modified at the 5- and 2′-positions of pyrimidines.
  • the SELEX method encompasses combining selected oligonucleotides with other selected oligonucleotides and non-oligonucleotide functional units as described in U.S. Pat. No. 5,637,459, entitled “Systematic Evolution of Ligands by EXponential Enrichment: Chimeric SELEX,” and U.S. Pat. No. 5,683,867, entitled “Systematic Evolution of Ligands by EXponential Enrichment: Blended SELEX,” respectively.
  • These applications allow the combination of the broad array of shapes and other properties, and the efficient amplification and replication properties, of oligonucleotides with the desirable properties of other molecules.
  • the SELEX method further encompasses combining selected nucleic acid ligands with lipophilic compounds or non-immunogenic, high molecular weight compounds in a diagnostic or therapeutic complex as described in U.S. Pat. No. 6,011,020, entitled “Nucleic Acid Ligand Complexes.”
  • Platelet-derived growth factor (PDGF) and the cognate tyrosine kinase receptors are potent mitogens for mesenchymal cells.
  • PDGF-BB is involved also in the regulation of IFP.
  • IFP insulin-derived growth factor
  • PDGF receptors are expressed in the tumor stroma of many common solid tumors, e.g. lung, colon and breast carcinomas. Based on these observations the effects of PDGF antagonists on tumor IFP, tumor transcapillary transport and therapeutic effects of cytotoxic drugs were investigated.
  • the present invention includes a method for treating tumors, more specifically, solid tumors comprising administering to a host a therapeutically effective dose of a composition comprising a PDGF aptamer and a cytotoxic agent.
  • the PDGF aptamer is identified using the SELEX process for the Systematic Evolution of Ligands by EXponential enrichment.
  • the present invention also includes a method for reducing the interstitial fluid pressure (IFP) of a tumor, more specifically, a solid tumor comprising administering a PDGF nucleic acid ligand.
  • IFP interstitial fluid pressure
  • the present invention includes a method for increasing the uptake of cytotoxic agents into a tumor comprising administering to a host a composition comprising a PDGF aptamer and a cytotoxic agent.
  • the present invention provides a novel method to increase drug uptake and therefore the therapeutic effectiveness of chemotherapy, by treatment with a PDGF inhibitor in conjunction with the therapeutic agent. As illustrated below, treatment with a PDGF aptamer decreases interstitial hypertension in these tumors thereby increasing the uptake of the therapeutic agent.
  • FIGS. 1A and B show the molecular description of the PDGF aptamer used in the present study ((SEQ ID NO:1), FIG. 1A ) and the aptamer that was used as the control. ((SEQ ID NO:2), FIG. 1B ).
  • FIG. 3 depicts the distribution of PDGF ⁇ -receptors and PDGF-AB/BB in PROb tumors.
  • Morphological analysis of sections from PROb tumor cells revealed three discrete zones in the investigated tumors ( FIG. 3A ). Towards the exterior a cell rich zone containing proliferative cells arranged in glandular structures was evident. A zone further towards the central part of the tumors contained apoptotic cells and cell debris, whereas the central parts were largely acellular, but contained extracellular matrix deposits. Inflammatory infiltrates were not evident in the investigated tumors. Immunohistochemical analyses showed that PDGF-AB/BB was expressed in blood vessels ( FIG.
  • FIG. 3B A weak but clearly visible staining of PDGF ⁇ -receptors was present in stromal structures in large parts of the tumors, especially around tumor glands ( FIG. 3D ). PDGF ⁇ -receptor staining was completely blocked by addition of a peptide corresponding to the amino acids 981-994 of the human/murine PDGF ⁇ -receptor at a 10-fold molar excess to the anti-PDGF ⁇ -receptor antibody ( FIG. 3E ).
  • FIG. 4 illustrates graphically the reduction of IFP upon treatment of SCID-Mice bearing subcutaneous KAT-4 tumors (human anaplastic thyroid carcinoma with PDGF aptamers.
  • FIG. 5 illustrates graphically the increased uptake of the chemotherapeutic agent Taxol upon pretreatment of KAT-4 tumors with PDGF aptamers.
  • nucleic acid ligand is a non-naturally occurring nucleic acid having a desirable action on a target.
  • Nucleic acid ligands are often referred to as “aptamers.”
  • a desirable action includes, but is not limited to, binding of the target, catalytically changing the target, reacting with the target in a way which modifies/alters the target or the functional activity of the target, covalently attaching to the target as in a suicide inhibitor, facilitating the reaction between the target and another molecule.
  • the action is specific binding affinity for a target molecule, such target molecule being a three dimensional chemical structure other than a polynucleotide that binds to the nucleic acid ligand through a mechanism which predominantly depends on Watson/Crick base pairing or triple helix binding, wherein the nucleic acid ligand does not have the known physiological function of being bound by the target molecule.
  • the target is PDGF or regions thereof.
  • Nucleic acid ligands include nucleic acids that are identified from a candidate mixture of nucleic acids, said nucleic acid ligand being a ligand of a given target, by the method comprising: a) contacting the candidate mixture with the target, 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; and c) amplifying the increased affinity nucleic acids to yield a ligand-enriched mixture of nucleic acids.
  • a “candidate mixture” is a mixture of nucleic acids of differing sequence from which to select a desired ligand.
  • the source of a candidate mixture can be from naturally-occurring nucleic acids or fragments thereof, chemically synthesized nucleic acids, enzymatically synthesized nucleic acids or nucleic acids made by a combination of the foregoing techniques.
  • each nucleic acid has fixed sequences surrounding a randomized region to facilitate the amplification process.
  • nucleic acid means either DNA, RNA, single-stranded or double-stranded, and any chemical modifications thereof. Modifications include, but are not limited to, those which provide other chemical groups that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole.
  • modifications include, but are not limited to, 2′-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil; backbone modifications, methylations, unusual base-pairing combinations such as the isobases isocytidine and isoguanidine and the like. Modifications can also include 3′ and 5′ modifications such as capping.
  • SELEX methodology involves the combination of selection of nucleic acid ligands that interact with a target in a desirable manner, for example binding to a protein, with amplification of those selected nucleic acids. Optional iterative cycling of the selection/amplification steps allows selection of one or a small number of nucleic acids which interact most strongly with the target from a pool which contains a very large number of nucleic acids. Cycling of the selection/amplification procedure is continued until a selected goal is achieved.
  • the SELEX methodology was employed to obtain nucleic acid ligands to PDGF. See U.S. Pat. No. 5,674,685, issued Oct. 7, 1997, U.S. Pat. No. 5,668,264, issued Sep. 16, 1997 and U.S.
  • SELEX target or “target” means any compound or molecule of interest for which a ligand is desired.
  • a target can be a protein, peptide, carbohydrate, polysaccharide, glycoprotein, hormone, receptor, antigen, antibody, virus, substrate, metabolite, transition state analog, cofactor, inhibitor, drug, dye, nutrient, growth factor, etc. without limitation.
  • the SELEX target was PDGF.
  • a “cytotoxic agent” is any substance used to destroy tumor cells.
  • the method of this invention can be used with any systemically administrated cytotoxic agent including, but not limited to, Bleomycin, Cisplatin, and Pt analogues; Carboplatin and Iproplatin, Cyclophosphamide, Daunorubicin, Doxofluoridine, Doxorubicin, Etoposide, Epirubicin, 5-Flurouracil, Gemzar, Ifosfamide, Melphalan, Methotrexate, Mithramycin, Mitomycin C, Mitoxanthrone, Streptozotocin, Taxol and Taxotere, Vincristine, Vinblastine, Vindesine, Vinorelbine, Topotecan and CPT-11.
  • “Therapeutic” as used herein, includes treatment and/or prophylaxis. When used, therapeutic refers to humans, as well as, other animals.
  • “Pharmaceutically or therapeutically effective dose or amount” refers to a dosage level sufficient to induce a desired biological result. That result may be the delivery of a pharmaceutical agent, alleviation of the signs, symptoms or causes of a disease or any other desirous alteration of a biological system.
  • a “host” is a living subject, human or animal, into which a drug or cytotoxic agent is administered.
  • the present invention includes a method for treating solid tumors comprising administering to a host a therapeutically effective dose of a composition comprising a PDGF aptamer and a cytotoxic agent.
  • the PDGF aptamer is identified using the SELEX methodology.
  • the SELEX process is described in U.S. patent application Ser. No. 07/536,428, entitled “Systematic Evolution of Ligands by Exponential Enrichment,” now abandoned, U.S. Pat. No. 5,475,096, entitled “Nucleic Acid Ligands,” and U.S. Pat. No. 5,270,163 (see also WO 91/19813), entitled “Methods for Identifying Nucleic Acid Ligands.”
  • the SELEX process provides a class of products that are nucleic acid molecules, each having a unique sequence, and each of which has the property of binding specifically to a desired target compound or molecule.
  • Target molecules are preferably proteins, but can also include among others carbohydrates, peptidoglycans and a variety of small molecules.
  • SELEX methodology can also be used to target biological structures, such as cell surfaces or viruses, through specific interaction with a molecule that is an integral part of that biological structure.
  • the SELEX process may be defined by the following series of steps.
  • 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 concentration 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 those nucleic acids having the strongest affinity for the target.
  • the 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 5-50%) are retained during partitioning.
  • nucleic acids selected during partitioning as having the relatively higher affinity for 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 acids 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.
  • the SELEX method encompasses the identification of high-affinity nucleic acid ligands containing modified nucleotides conferring improved characteristics on the ligand, such as improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions.
  • SELEX-identified nucleic acid ligands containing modified nucleotides are described in U.S. Pat. No. 5,660,985, entitled “High Affinity Nucleic Acid Ligands Containing Modified Nucleotides,” that describes oligonucleotides containing nucleotide derivatives chemically modified at the 5- and 2′-positions of pyrimidines.
  • the SELEX method encompasses combining selected oligonucleotides with other selected oligonucleotides and non-oligonucleotide functional units as described in U.S. Pat. No. 5,637,459, entitled “Systematic Evolution of Ligands by Exponential Enrichment: Chimeric SELEX,” and U.S. Pat. No. 5,683,867, entitled “Systematic Evolution of Ligands by Exponential Enrichment: Blended SELEX,” respectively.
  • These applications allow the combination of the broad array of shapes and other properties, and the efficient amplification and replication properties, of oligonucleotides with the desirable properties of other molecules.
  • nucleic acid ligand can be made to increase the in vivo stability of the nucleic acid ligand or to enhance or to mediate the delivery of the nucleic acid ligand. See, e.g., U.S. patent application Ser. No. 08/117,991, filed Sep. 8, 1993, now abandoned and U.S. Pat. No.
  • nucleic acid ligands contemplated in this invention include, but are not limited to, those which provide other chemical groups that incorporate additional charge, polarizability, hydrophobicity, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole.
  • Such modifications include, but are not limited to, 2′-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil; backbone modifications, phosphorothioate or alkyl phosphate modifications, methylations, unusual base-pairing combinations such as the isobases, isocytidine and isoguanidine and the like. Modifications can also include 3′ and 5′ modifications such as capping.
  • the nucleic acid ligands are RNA molecules that are 2′-fluoro (2′-F) modified on the sugar moiety of pyrimidine residues.
  • the modifications can be pre- or post-SELEX process modifications.
  • Pre-SELEX process modifications yield nucleic acid ligands with both specificity for their SELEX target and improved in vivo stability.
  • Post-SELEX process modifications made to 2′-OH nucleic acid ligands can result in improved in vivo stability without adversely affecting the binding capacity of the nucleic acid ligand.
  • nucleic acid ligands to PDGF of the invention are prepared through the SELEX methodology that is outlined above and thoroughly enabled in the SELEX applications incorporated herein by reference in their entirety.
  • the cytotoxic agent can be any substance used in the prevention, diagnosis, alleviation, treatment or cure of disease. More specifically, the cytotoxic agent can be selected from any systemically administrated agent including, but not limited to, Bleomycin, Cisplatin, and Pt analogues; Carboplatin and Iproplatin, Cyclophosphamide, Daunorubicin, Doxofluoridine, Doxorubicin, Etoposide, Epirubicin, 5-Flurouracil, Gemzar, Ifosfamide, Melphalan, Methotrexate, Mithramycin, Mitomycin C, Mitoxanthrone, Streptozotocin, Taxol and Taxotere, Vincristine, Vinblastine, Vindesine, Vinorelbine, Topotecan and CPT-11.
  • Bleomycin Cisplatin, and Pt analogues
  • Carboplatin and Iproplatin Cyclophosphamide
  • Daunorubicin Doxo
  • Various delivery systems are known in the art and can be used to administer the therapeutic composition comprising the PDGF aptamer and cytotoxic agent of the invention, e.g., aqueous solution, encapsulation in liposomes, microparticles, and microcapsules.
  • compositions of the invention may be administered parenterally by injection, although other effective administration forms, such as intraarticular injection, inhalant mists, orally active formulations, transdermal iontophoresis or suppositories are also envisioned.
  • One preferred carrier is physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers may also be used.
  • the carrier and the nucleic acid ligand constitute a physiologically-compatible, slow release formulation.
  • the primary solvent in such a carrier may be either aqueous or non-aqueous in nature.
  • the carrier may contain other pharmacologically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation.
  • the carrier may contain still other pharmacologically-acceptable excipients for modifying or maintaining the stability, rate of dissolution, release or absorption of the ligand.
  • excipients are those substances usually and customarily employed to formulate dosages for parental administration in either unit dose or multi-dose form.
  • the therapeutic composition may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such formulations may be stored either in a ready to use form or requiring reconstitution immediately prior to administration.
  • the manner of administering formulations containing the compositions for systemic delivery may be via subcutaneous, intramuscular, intravenous, intranasal or vaginal or rectal suppository.
  • the amount of the composition which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder of condition, which can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness or advancement of the disease or condition, and should be decided according to the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curved derived from in vitro or animal model test systems. For example, an effective amount of the composition of the invention is readily determined by administering graded doses of the composition of the invention and observing the desired effect.
  • the PDGF-B aptamer used in the present study ((SEQ ID NO:1), FIG. 1A ) was produced by the SELEX method. (Tuerk and Gold (1990) Science 249:505-510, which is incorporated herein by reference in its entirety).
  • the modified DNA aptamer, linked to 40 kDa polyethylene glycol, has a high affinity for PDGF-B with a Kd of ⁇ 0.1 nM. (Green et al. (1996) Biochemistry 35:14413-14424; Floege et al. (1999) Am. J. Pathol. 154:169-179; U.S. Pat. No. 6,229,002, issued May 8, 2001, ((SEQ ID NO:146), FIG.
  • Example 1 describes the method used to treat PROb tumor-bearing rats with the PDGF-B specific aptamer.
  • treatment of PROb tumor-bearing rats with the PDGF-B specific aptamer resulted in a decrease in tumor IFP when compared to rats treated with the control aptamer.
  • the mean IFP in control aptamer-treated tumors was 14.6 ⁇ 1.2 mm Hg ( ⁇ S.E.M.) and 9.7 ⁇ 1.6 mm Hg ( ⁇ S.E.M.) in tumors treated with the PDGF-B specific aptamer.
  • the method used to determine IFP is described in Example 2.
  • PROb tumors were analyzed with regard to morphology, as well as distribution of PDGF-AB/BB and PDGF ⁇ -receptors as described in Example 3.
  • the tumors displayed a heterogeneous morphology.
  • tumor cells were arranged in glandular structures, whereas more centrally, tumor cells were less abundant and less well organized ( FIG. 3A ).
  • the central part was basically acellular ( FIG. 3A ).
  • Expression of PDGF-AB/BB in PROb tumors was found in blood vessels and possibly in extravascular stromal cells surrounding tumor glands ( FIG. 3B ). In the central part of the tumors, few if any tumor cells were present, but strongly PDGF-AB/BB positive cells were seen ( FIG. 3C ).
  • PDGF-AB/BB positive tumor cells were found in vascular cells of larger vessels, and in unidentified, possibly microvascular, cells in the stroma ( FIG. 3D ).
  • Tissue culture Cells were cultured under standard conditions and all tissue culture media were supplemented with 10% fetal bovine serum (FBS) and antibiotics, unless otherwise stated.
  • FBS fetal bovine serum
  • PAE cells were maintained in F12 culture medium (Sigma).
  • PROb cells were kept in Dulbecco's Modified Eagle's Medium (Sigma).
  • the PDGF-B aptamer used in the present study was produced by the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) method. (Tuerk and Gold (1990) Science 249:505-510, which is incorporated herein by reference in its entirety).
  • the modified DNA aptamer, linked to 40 kDa polyethylene glycol, has a high affinity for PDGF-B with a Kd of ⁇ 0.1 nM. (Green et al. (1996) Biochemistry 35:14413-14424; Floege et al. (1999) Am. J. Pathol. 154:169-179, each of which is incorporated herein by reference in its entirety).
  • the aptamer shows biphasic clearance in rats following iv injection, approximately 47% is cleared with a half-life of 32 minutes, while the remainder is cleared with a half-life of 135 minutes. (Floege et al. (1999) Am. J. Pathol. 154:169-179). As a control, a sequence-scrambled analog of the PDGF-B aptamer was used. This oligonucleotide has a Kd for PDGF-BB in the micromolar range. (Floege et al. (1999) Am. J. Pathol. 154:169-179).
  • Subcutaneously growing PROb tumors were established in BDIX rats by injection of 5 ⁇ 10 6 tumor cells in 50 ⁇ L of PBS in the flank.
  • the rats were kept under pathogen-free conditions and were fed ad libitum. They were monitored regularly for tumor growth and experiments were performed 8-12 weeks after tumor cell implantation on rats bearing tumors ranging in size between 0.6 cm 3 and 7.6 cm 3 .
  • the PDGF-B specific aptamer, and a control aptamer were given as i.p. injections in 2 mL PBS twice daily for 4 consecutive days at a dose of 7 mg ⁇ kg ⁇ 1 ⁇ day ⁇ 1 . All animal experiments described in the present report were approved by the Ethical Committee for Animal Experiments in Uppsala, Sweden.
  • Tumor IFP was measured using the wick-in-needle technique (Wiig et al. (1982) Scan. J. Clin. Lab. Invest. 42:159-164). Briefly, rats were anaesthetized using isofluran in a mixture of O 2 and air. A standard 23-gauge needle filled with nylon-floss and saline, supplemented with 50 IE/mL of heparin was inserted into the center of the tumor and connected to a pressure transducer. This makeup enables continuous and stable recordings of fluid pressure. Fluid communication between the needle and the transducer was confirmed by compression and decompression of the tubing during each measurement.
  • Tumor IFP was measured once before treatment with PDGF aptamers, and again 1-2 hours after the last administration of aptamers or vehicle alone. The change in tumor IFP was calculated for each tumor. After the second IFP measurement the rats were sacrificed and the tumors were excised and snap frozen in liquid nitrogen for further analyses.
  • paraffin-embedded 4 ⁇ m sections were stained with van Gieson staining. Immunohistochemistry was performed on 6 ⁇ m cryosections from PROb tumors. Sections were fixed in acetone and blocked with 0.3% hydrogen peroxide in methanol for 15 minutes, rinsed and further incubated in a solution containing 20% human normal serum in a buffer containing 2% rat serum, 3% bovine serum albumin, 0.01% NP40 in PBS (RM buffer) for 5 hours at 4° C. Primary antibodies dissolved in RM buffer were added, either 4 ⁇ g/mL affinity-purified rabbit anti-PDGF preceptor IgG (Claesson-Welsh et al. (1988) Mol. Cell Biol.
  • SCID-mice bearing subcutaneous KAT-4 tumors (human, anaplastic thyroid carcinoma) were pre-treated for 4 consecutive days with 12 mg ⁇ kg ⁇ 1 ⁇ day ⁇ 1 SELEX aptamers (i.p. injections, three times daily). Tumor interstitial fluid pressure was measured using the wick-in-needle technique. The results are set forth in FIG. 4 . * p ⁇ 0.05, Student's t-test.

Abstract

A method is provided for treating solid tumors comprising administering a composition comprising a PDGF aptamer and a cytotoxic agent. In a preferred embodiment the PDGF aptamer is identified using the SELEX process for the Systematic Evolution of Ligands by Exponential enrichment. A method is also provided for reducing the interstitial fluid pressure (IFP) of a solid tumor comprising administering a PDGF aptamer. Finally, a method is provided for increasing the uptake of cytotoxic agents into a tumor comprising administering a composition comprising a PDGF aptamer and a cytotoxic agent.

Description

    RELATED APPLICATIONS
  • This application is a divisional of U.S. patent application Ser. No. 10/791,367, filed Mar. 2, 2004, which is a continuation of U.S. patent application Ser. No. 09/859,724, filed May 17, 2001, now U.S. Pat. No. 6,699,843, which claims the benefit of U.S. Provisional Application Ser. No. 60/205,006, filed May 17, 2000. U.S. patent application Ser. No. 09/859,724 is a continuation in part of U.S. patent application Ser. No. 08/479,725, filed Jun. 7, 1995, now U.S. Pat. No. 5,674,685, and also a continuation in part of U.S. patent application Ser. No. 08/479,783, filed Jun. 7, 1995, now U.S. Pat. No. 5,668,264, and also a continuation in part of U.S. patent application Ser. No. 08/618,693, filed Mar. 20, 1996, now U.S. Pat. No. 5,723,594, each entitled “High Affinity PDGF Nucleic Acid Ligands.” U.S. patent application Ser. No. 09/859,724 is also a continuation in part of U.S. patent application Ser. No. 08/991,743, filed Dec. 16, 1997, now U.S. Pat. No. 6,229,002, entitled “Platelet Derived Growth Factor (PDGF) Nucleic Acid Ligand Complexes.” Each of the foregoing applications is incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • This invention relates generally to a method for increasing the uptake of drugs into tumors by treatment with a nucleic acid ligand to PDGF in combination with a cytotoxic agent. The method used for identifying nucleic acid ligands to PDGF is called SELEX, an acronym for Systematic Evolution of Ligands by Exponential enrichment. The method of the present invention is useful for increasing the therapeutic effectiveness of cytotoxic agents.
  • BACKGROUND OF THE INVENTION
  • The Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process is a method for the in vitro evolution of nucleic acid molecules with highly specific binding to target molecules and is described in U.S. patent application Ser. No. 07/536,428, filed Jun. 11, 1990, entitled “Systematic Evolution of Ligands by EXponential Enrichment,” now abandoned, U.S. Pat. No. 5,475,096, entitled “Nucleic Acid Ligands,” and U.S. Pat. No. 5,270,163 (see also WO 91/19813), entitled “Methods for Identifying Nucleic Acid Ligands,” each of which is specifically incorporated herein by reference in its entirety. Each of these applications, collectively referred to herein as the SELEX Patent Applications, describes a fundamentally novel method for making a nucleic acid ligand to any desired target molecule.
  • The SELEX process provides a class of products which are referred to as nucleic acid ligands or aptamers, each having a unique sequence, and which has 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. The SELEX process 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 or composition can serve as targets. The SELEX method applied to the application of high affinity binding involves selection from a mixture of candidate oligonucleotides and step-wise iterations of binding, partitioning and amplification, using the same general selection scheme, to achieve virtually any desired criterion of binding affinity and selectivity. Starting from a mixture of nucleic acids, preferably comprising a segment of randomized sequence, the SELEX 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 specifically to target molecules, dissociating the nucleic acid-target complexes, amplifying the nucleic acids dissociated from the nucleic acid-target complexes 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 to yield highly specific high affinity nucleic acid ligands to the target molecule.
  • It has been recognized by the present inventors that the SELEX method demonstrates that nucleic acids as chemical compounds can form a wide array of shapes, sizes and configurations, and are capable of a far broader repertoire of binding and other functions than those displayed by nucleic acids in biological systems.
  • The basic SELEX method has been modified to achieve a number of specific objectives. For example, U.S. patent application Ser. No. 07/960,093, filed Oct. 14, 1992, now abandoned, and U.S. Pat. No. 5,707,796, both entitled “Method for Selecting Nucleic Acids on the Basis of Structure,” describe the use of the SELEX process in conjunction with gel electrophoresis to select nucleic acid molecules with specific structural characteristics, such as bent DNA. U.S. patent application Ser. No. 08/123,935, filed Sep. 17, 1993, entitled “Photoselection of Nucleic Acid Ligands,” now abandoned, U.S. Pat. No. 5,763,177 and U.S. Pat. No. 6,011,577, both entitled “Systematic Evolution of Ligands by Exponential Enrichment: Photoselection of Nucleic Acid Ligands and Solution SELEX,” describe a SELEX based method for selecting nucleic acid ligands containing photoreactive groups capable of binding and/or photocrosslinking to and/or photoinactivating a target molecule. U.S. Pat. No. 5,580,737, entitled “High-Affinity Nucleic Acid Ligands That Discriminate Between Theophylline and Caffeine,” describes a method for identifying highly specific nucleic acid ligands able to discriminate between closely related molecules, which can be non-peptidic, termed Counter-SELEX. U.S. Pat. No. 5,567,588, entitled “Systematic Evolution of Ligands by EXponential Enrichment: Solution SELEX,” describes a SELEX-based method which achieves highly efficient partitioning between oligonucleotides having high and low affinity for a target molecule.
  • The SELEX method encompasses the identification of high-affinity nucleic acid ligands containing modified nucleotides conferring improved characteristics on the ligand, such as improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions. SELEX process-identified nucleic acid ligands containing modified nucleotides are described in U.S. Pat. No. 5,660,985, entitled “High Affinity Nucleic Acid Ligands Containing Modified Nucleotides,” that describes oligonucleotides containing nucleotide derivatives chemically modified at the 5- and 2′-positions of pyrimidines. U.S. Pat. No. 5,580,737, supra, describes highly specific nucleic acid ligands containing one or more nucleotides modified with 2′-amino (2′-NH2), 2′-fluoro (2′-F), and/or 2′-O-methyl (2′-OMe). U.S. patent application Ser. No. 08/264,029, filed Jun. 22, 1994, entitled “Novel Method of Preparation of Known and Novel 2′ Modified Nucleosides by Intramolecular Nucleophilic Displacement,” describes oligonucleotides containing various 2′-modified pyrimidines.
  • The SELEX method encompasses combining selected oligonucleotides with other selected oligonucleotides and non-oligonucleotide functional units as described in U.S. Pat. No. 5,637,459, entitled “Systematic Evolution of Ligands by EXponential Enrichment: Chimeric SELEX,” and U.S. Pat. No. 5,683,867, entitled “Systematic Evolution of Ligands by EXponential Enrichment: Blended SELEX,” respectively. These applications allow the combination of the broad array of shapes and other properties, and the efficient amplification and replication properties, of oligonucleotides with the desirable properties of other molecules.
  • The SELEX method further encompasses combining selected nucleic acid ligands with lipophilic compounds or non-immunogenic, high molecular weight compounds in a diagnostic or therapeutic complex as described in U.S. Pat. No. 6,011,020, entitled “Nucleic Acid Ligand Complexes.” Each of the above described patent applications which describe modifications of the basic SELEX procedure are specifically incorporated by reference herein in their entirety.
  • One approach to increasing the effectiveness of existing anti-cancer drugs for the treatment of solid malignancies is to augment the uptake of the drugs into tumors, and thereby obtain increased therapeutic concentration without elevating the adverse side-effects. Most solid tumors display an increased interstitial fluid pressure (IFP). The molecular mechanisms causing increased tumor IFP are poorly understood. However, tumor stroma involvement in IFP control has been demonstrated. (Gullino et al. (1964) Cancer Res. 24:780-797; Philips et al. (1990) J. Natl Cancer Inst. 82:1457-1469; Jain (1987) Cancer Res. 47:3039-3051). It has been suggested that high tumor IFP prevents drug transport from the circulation into the tumor interstitium. The reduction of tumor IFP therefore is a target for efforts to increase tumor drug uptake. (Jain (1996) Science 271:1079-1080). Several agents which induce a lowering of IFP, and thereby increase the transcapillary transport in experimental murine tumors have been identified, including nicotinamide (Lee et al. (1992) Cancer Res. 52:3237-3240), TNF-α (Kristensen et al. (1996) Br. J. Cancer 74:533-536) and dexamethasone (Kristjansen et al. (1993) Cancer Res. 53:4764-4766).
  • Platelet-derived growth factor (PDGF) and the cognate tyrosine kinase receptors are potent mitogens for mesenchymal cells. In addition to its growth promoting effects, PDGF-BB is involved also in the regulation of IFP. After dextran-induced anaphylaxis and lowering of IFP in rat skin, local administration of PDGF-BB results in normalized IFP. PDGF receptors are expressed in the tumor stroma of many common solid tumors, e.g. lung, colon and breast carcinomas. Based on these observations the effects of PDGF antagonists on tumor IFP, tumor transcapillary transport and therapeutic effects of cytotoxic drugs were investigated.
  • SUMMARY OF THE INVENTION
  • The present invention includes a method for treating tumors, more specifically, solid tumors comprising administering to a host a therapeutically effective dose of a composition comprising a PDGF aptamer and a cytotoxic agent. In a preferred embodiment the PDGF aptamer is identified using the SELEX process for the Systematic Evolution of Ligands by EXponential enrichment. The present invention also includes a method for reducing the interstitial fluid pressure (IFP) of a tumor, more specifically, a solid tumor comprising administering a PDGF nucleic acid ligand. Finally, the present invention includes a method for increasing the uptake of cytotoxic agents into a tumor comprising administering to a host a composition comprising a PDGF aptamer and a cytotoxic agent. The present invention provides a novel method to increase drug uptake and therefore the therapeutic effectiveness of chemotherapy, by treatment with a PDGF inhibitor in conjunction with the therapeutic agent. As illustrated below, treatment with a PDGF aptamer decreases interstitial hypertension in these tumors thereby increasing the uptake of the therapeutic agent.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A and B show the molecular description of the PDGF aptamer used in the present study ((SEQ ID NO:1), FIG. 1A) and the aptamer that was used as the control. ((SEQ ID NO:2), FIG. 1B).
  • FIG. 2 illustrates graphically the reduction in tumor interstitial fluid pressure (IFP) upon treatment of PROb tumors with a PDGF specific aptamer. Average tumor IFP in the animals treated with a sequence-scrambled control oligonucleotide (n=5) and PDGF-B specific aptamer (n=7) is depicted. Bars represent S.E.M; * p<0.05
  • FIG. 3 depicts the distribution of PDGF β-receptors and PDGF-AB/BB in PROb tumors. Morphological analysis of sections from PROb tumor cells revealed three discrete zones in the investigated tumors (FIG. 3A). Towards the exterior a cell rich zone containing proliferative cells arranged in glandular structures was evident. A zone further towards the central part of the tumors contained apoptotic cells and cell debris, whereas the central parts were largely acellular, but contained extracellular matrix deposits. Inflammatory infiltrates were not evident in the investigated tumors. Immunohistochemical analyses showed that PDGF-AB/BB was expressed in blood vessels (FIG. 3B) and in the central parts of tumors by cells morphologically identified as macrophages (FIG. 3C). A weak but clearly visible staining of PDGF β-receptors was present in stromal structures in large parts of the tumors, especially around tumor glands (FIG. 3D). PDGF β-receptor staining was completely blocked by addition of a peptide corresponding to the amino acids 981-994 of the human/murine PDGF β-receptor at a 10-fold molar excess to the anti-PDGF β-receptor antibody (FIG. 3E).
  • FIG. 4 illustrates graphically the reduction of IFP upon treatment of SCID-Mice bearing subcutaneous KAT-4 tumors (human anaplastic thyroid carcinoma with PDGF aptamers.
  • FIG. 5 illustrates graphically the increased uptake of the chemotherapeutic agent Taxol upon pretreatment of KAT-4 tumors with PDGF aptamers.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Various terms are used herein to refer to aspects of the present invention. To aid in the clarification of the description of the components of this invention, the following definitions are provided.
  • As used herein a “nucleic acid ligand” is a non-naturally occurring nucleic acid having a desirable action on a target. Nucleic acid ligands are often referred to as “aptamers.” A desirable action includes, but is not limited to, binding of the target, catalytically changing the target, reacting with the target in a way which modifies/alters the target or the functional activity of the target, covalently attaching to the target as in a suicide inhibitor, facilitating the reaction between the target and another molecule. In a preferred embodiment, the action is specific binding affinity for a target molecule, such target molecule being a three dimensional chemical structure other than a polynucleotide that binds to the nucleic acid ligand through a mechanism which predominantly depends on Watson/Crick base pairing or triple helix binding, wherein the nucleic acid ligand does not have the known physiological function of being bound by the target molecule. In the present invention, the target is PDGF or regions thereof. Nucleic acid ligands include nucleic acids that are identified from a candidate mixture of nucleic acids, said nucleic acid ligand being a ligand of a given target, by the method comprising: a) contacting the candidate mixture with the target, 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; and c) amplifying the increased affinity nucleic acids to yield a ligand-enriched mixture of nucleic acids.
  • As used herein a “candidate mixture” is a mixture of nucleic acids of differing sequence from which to select a desired ligand. The source of a candidate mixture can be from naturally-occurring nucleic acids or fragments thereof, chemically synthesized nucleic acids, enzymatically synthesized nucleic acids or nucleic acids made by a combination of the foregoing techniques. In a preferred embodiment, each nucleic acid has fixed sequences surrounding a randomized region to facilitate the amplification process.
  • As used herein, “nucleic acid” means either DNA, RNA, single-stranded or double-stranded, and any chemical modifications thereof. Modifications include, but are not limited to, those which provide other chemical groups that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole. Such modifications include, but are not limited to, 2′-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil; backbone modifications, methylations, unusual base-pairing combinations such as the isobases isocytidine and isoguanidine and the like. Modifications can also include 3′ and 5′ modifications such as capping.
  • “SELEX” methodology involves the combination of selection of nucleic acid ligands that interact with a target in a desirable manner, for example binding to a protein, with amplification of those selected nucleic acids. Optional iterative cycling of the selection/amplification steps allows selection of one or a small number of nucleic acids which interact most strongly with the target from a pool which contains a very large number of nucleic acids. Cycling of the selection/amplification procedure is continued until a selected goal is achieved. In the present invention, the SELEX methodology was employed to obtain nucleic acid ligands to PDGF. See U.S. Pat. No. 5,674,685, issued Oct. 7, 1997, U.S. Pat. No. 5,668,264, issued Sep. 16, 1997 and U.S. Pat. No. 5,723,594, issued Mar. 3, 1998, each entitled “High Affinity PDGF Nucleic Acid Ligands,” and U.S. Pat. No. 6,229,002, issued, May 8, 2001, entitled “Platelet Derived Growth Factor (PDGF) Nucleic Acid Ligand Complexes.”
  • The SELEX methodology is described in the SELEX Patent Applications.
  • “SELEX target” or “target” means any compound or molecule of interest for which a ligand is desired. A target can be a protein, peptide, carbohydrate, polysaccharide, glycoprotein, hormone, receptor, antigen, antibody, virus, substrate, metabolite, transition state analog, cofactor, inhibitor, drug, dye, nutrient, growth factor, etc. without limitation. In this application, the SELEX target was PDGF.
  • A “cytotoxic agent” is any substance used to destroy tumor cells. The method of this invention can be used with any systemically administrated cytotoxic agent including, but not limited to, Bleomycin, Cisplatin, and Pt analogues; Carboplatin and Iproplatin, Cyclophosphamide, Daunorubicin, Doxofluoridine, Doxorubicin, Etoposide, Epirubicin, 5-Flurouracil, Gemzar, Ifosfamide, Melphalan, Methotrexate, Mithramycin, Mitomycin C, Mitoxanthrone, Streptozotocin, Taxol and Taxotere, Vincristine, Vinblastine, Vindesine, Vinorelbine, Topotecan and CPT-11.
  • “Therapeutic” as used herein, includes treatment and/or prophylaxis. When used, therapeutic refers to humans, as well as, other animals.
  • “Pharmaceutically or therapeutically effective dose or amount” refers to a dosage level sufficient to induce a desired biological result. That result may be the delivery of a pharmaceutical agent, alleviation of the signs, symptoms or causes of a disease or any other desirous alteration of a biological system.
  • A “host” is a living subject, human or animal, into which a drug or cytotoxic agent is administered.
  • Note, that throughout this application various citations are provided. Each citation is specifically incorporated herein in its entirety by reference.
  • The present invention includes a method for treating solid tumors comprising administering to a host a therapeutically effective dose of a composition comprising a PDGF aptamer and a cytotoxic agent. In a preferred embodiment the PDGF aptamer is identified using the SELEX methodology. The SELEX process is described in U.S. patent application Ser. No. 07/536,428, entitled “Systematic Evolution of Ligands by Exponential Enrichment,” now abandoned, U.S. Pat. No. 5,475,096, entitled “Nucleic Acid Ligands,” and U.S. Pat. No. 5,270,163 (see also WO 91/19813), entitled “Methods for Identifying Nucleic Acid Ligands.” These applications, each specifically incorporated herein by reference, are collectively called the SELEX Patent Applications.
  • The SELEX process provides a class of products that are nucleic acid molecules, each having a unique sequence, and each of which has the property of binding specifically to a desired target compound or molecule. Target molecules are preferably proteins, but can also include among others carbohydrates, peptidoglycans and a variety of small molecules. SELEX methodology can also be used to target biological structures, such as cell surfaces or viruses, through specific interaction with a molecule that is an integral part of that biological structure.
  • In its most basic form, 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 concentration 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).
  • 2. 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 those nucleic acids having the strongest affinity for the target.
  • 3. The 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 5-50%) are retained during partitioning.
  • 4. Those nucleic acids selected during partitioning as having the relatively higher affinity for 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.
  • 5. By repeating the partitioning and amplifying steps above, the newly formed candidate mixture contains fewer and fewer unique sequences, and the average degree of affinity of the nucleic acids to the target will generally increase. Taken to its extreme, 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.
  • The basic SELEX method has been modified to achieve a number of specific objectives. For example, U.S. patent application Ser. No. 07/960,093, filed Oct. 14, 1992, now abandoned, and U.S. Pat. No. 5,707,796, both entitled “Method for Selecting Nucleic Acids on the Basis of Structure,” describe the use of the SELEX process in conjunction with gel electrophoresis to select nucleic acid molecules with specific structural characteristics, such as bent DNA. U.S. patent application Ser. No. 08/123,935, filed Sep. 17, 1993, entitled “Photoselection of Nucleic Acid Ligands,” now abandoned, U.S. Pat. No. 5,763,177 and U.S. Pat. No. 6,001,577, both entitled “Systematic Evolution of Ligands by Exponential Enrichment: Photoselection of Nucleic Acid Ligands and Solution SELEX,” all describe a SELEX based method for selecting nucleic acid ligands containing photoreactive groups capable of binding and/or photocrosslinking to and/or photoinactivating a target molecule. U.S. Pat. No. 5,580,737, entitled “High-Affinity Nucleic Acid Ligands That Discriminate Between Theophylline and Caffeine,” describes a method for identifying highly specific nucleic acid ligands able to discriminate between closely related molecules, termed Counter-SELEX. U.S. Pat. No. 5,567,588, entitled “Systematic Evolution of Ligands by Exponential Enrichment: Solution SELEX,” describes a SELEX-based method which achieves highly efficient partitioning between oligonucleotides having high and low affinity for a target molecule. U.S. Pat. No. 5,496,938, entitled “Nucleic Acid Ligands to HUV-RT and HIV-1 Rev,” describes methods for obtaining improved nucleic acid ligands after SELEX has been performed. U.S. Pat. No. 5,705,337, entitled “Systematic Evolution of Ligands by Exponential Enrichment: Chemi-SELEX,” describes methods for covalently linking a ligand to its target.
  • The SELEX method encompasses the identification of high-affinity nucleic acid ligands containing modified nucleotides conferring improved characteristics on the ligand, such as improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions. SELEX-identified nucleic acid ligands containing modified nucleotides are described in U.S. Pat. No. 5,660,985, entitled “High Affinity Nucleic Acid Ligands Containing Modified Nucleotides,” that describes oligonucleotides containing nucleotide derivatives chemically modified at the 5- and 2′-positions of pyrimidines. U.S. Pat. No. 5,637,459, supra, describes highly specific nucleic acid ligands containing one or more nucleotides modified with 2′-amino (2′-NH2), 2′-fluoro (2′-F), and/or 2′-O-methyl (2′-OMe). U.S. patent application Ser. No. 08/264,029, filed Jun. 22, 1994, entitled “Novel Method of Preparation of Known and Novel 2′ Modified Nucleosides by Intramolecular Nucleophilic Displacement,” describes oligonucleotides containing various 2′-modified pyrimidines.
  • The SELEX method encompasses combining selected oligonucleotides with other selected oligonucleotides and non-oligonucleotide functional units as described in U.S. Pat. No. 5,637,459, entitled “Systematic Evolution of Ligands by Exponential Enrichment: Chimeric SELEX,” and U.S. Pat. No. 5,683,867, entitled “Systematic Evolution of Ligands by Exponential Enrichment: Blended SELEX,” respectively. These applications allow the combination of the broad array of shapes and other properties, and the efficient amplification and replication properties, of oligonucleotides with the desirable properties of other molecules.
  • In U.S. Pat. No. 5,496,938, methods are described for obtaining improved nucleic acid ligands after the SELEX process has been performed. This patent, entitled “Nucleic Acid Ligands to HIV-RT and HIV-1 Rev,” is specifically incorporated herein by reference.
  • One potential problem encountered in the diagnostic use of nucleic acids is that oligonucleotides in their phosphodiester form may be quickly degraded in body fluids by intracellular and extracellular enzymes, such as endonucleases and exonucleases, before the desired effect is manifest. Certain chemical modifications of the nucleic acid ligand can be made to increase the in vivo stability of the nucleic acid ligand or to enhance or to mediate the delivery of the nucleic acid ligand. See, e.g., U.S. patent application Ser. No. 08/117,991, filed Sep. 8, 1993, now abandoned and U.S. Pat. No. 5,660,985, both entitled “High Affinity Nucleic Acid Ligands Containing Modified Nucleotides,” and U.S. patent application Ser. No. 09/362,578, filed Jul. 28, 1999, entitled “Transcription-free SELEX,” each of which is specifically incorporated herein by reference in its entirety. Modifications of the nucleic acid ligands contemplated in this invention include, but are not limited to, those which provide other chemical groups that incorporate additional charge, polarizability, hydrophobicity, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole. Such modifications include, but are not limited to, 2′-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil; backbone modifications, phosphorothioate or alkyl phosphate modifications, methylations, unusual base-pairing combinations such as the isobases, isocytidine and isoguanidine and the like. Modifications can also include 3′ and 5′ modifications such as capping. In preferred embodiments of the instant invention, the nucleic acid ligands are RNA molecules that are 2′-fluoro (2′-F) modified on the sugar moiety of pyrimidine residues.
  • The modifications can be pre- or post-SELEX process modifications. Pre-SELEX process modifications yield nucleic acid ligands with both specificity for their SELEX target and improved in vivo stability. Post-SELEX process modifications made to 2′-OH nucleic acid ligands can result in improved in vivo stability without adversely affecting the binding capacity of the nucleic acid ligand.
  • Other modifications are known to one of ordinary skill in the art. Such modifications may be made post-SELEX process (modification of previously identified unmodified ligands) or by incorporation into the SELEX process.
  • The nucleic acid ligands to PDGF of the invention are prepared through the SELEX methodology that is outlined above and thoroughly enabled in the SELEX applications incorporated herein by reference in their entirety.
  • As noted above, the cytotoxic agent can be any substance used in the prevention, diagnosis, alleviation, treatment or cure of disease. More specifically, the cytotoxic agent can be selected from any systemically administrated agent including, but not limited to, Bleomycin, Cisplatin, and Pt analogues; Carboplatin and Iproplatin, Cyclophosphamide, Daunorubicin, Doxofluoridine, Doxorubicin, Etoposide, Epirubicin, 5-Flurouracil, Gemzar, Ifosfamide, Melphalan, Methotrexate, Mithramycin, Mitomycin C, Mitoxanthrone, Streptozotocin, Taxol and Taxotere, Vincristine, Vinblastine, Vindesine, Vinorelbine, Topotecan and CPT-11.
  • Various delivery systems are known in the art and can be used to administer the therapeutic composition comprising the PDGF aptamer and cytotoxic agent of the invention, e.g., aqueous solution, encapsulation in liposomes, microparticles, and microcapsules.
  • Therapeutic compositions of the invention may be administered parenterally by injection, although other effective administration forms, such as intraarticular injection, inhalant mists, orally active formulations, transdermal iontophoresis or suppositories are also envisioned. One preferred carrier is physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers may also be used. In one preferred embodiment, it is envisioned that the carrier and the nucleic acid ligand constitute a physiologically-compatible, slow release formulation. The primary solvent in such a carrier may be either aqueous or non-aqueous in nature. In addition, the carrier may contain other pharmacologically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation. Similarly, the carrier may contain still other pharmacologically-acceptable excipients for modifying or maintaining the stability, rate of dissolution, release or absorption of the ligand. Such excipients are those substances usually and customarily employed to formulate dosages for parental administration in either unit dose or multi-dose form.
  • Once the therapeutic composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such formulations may be stored either in a ready to use form or requiring reconstitution immediately prior to administration. The manner of administering formulations containing the compositions for systemic delivery may be via subcutaneous, intramuscular, intravenous, intranasal or vaginal or rectal suppository.
  • The amount of the composition which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder of condition, which can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness or advancement of the disease or condition, and should be decided according to the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curved derived from in vitro or animal model test systems. For example, an effective amount of the composition of the invention is readily determined by administering graded doses of the composition of the invention and observing the desired effect.
  • The following examples are provided to explain and illustrate the present invention and are not intended to be limiting of the invention.
  • The PDGF-B aptamer used in the present study ((SEQ ID NO:1), FIG. 1A) was produced by the SELEX method. (Tuerk and Gold (1990) Science 249:505-510, which is incorporated herein by reference in its entirety). The modified DNA aptamer, linked to 40 kDa polyethylene glycol, has a high affinity for PDGF-B with a Kd of ˜0.1 nM. (Green et al. (1996) Biochemistry 35:14413-14424; Floege et al. (1999) Am. J. Pathol. 154:169-179; U.S. Pat. No. 6,229,002, issued May 8, 2001, ((SEQ ID NO:146), FIG. 9A), each of which is incorporated herein by reference in its entirety). A sequence-scrambled analog of the PDGF-B aptamer was used as a control. ((SEQ ID NO:2), FIG. 1B). This oligonucleotide has a Kd for PDGF-BB in the micromolar range. (Floege et al. (1999) Am. J. Pathol. 154:169-179; U.S. Pat. No. 6,229,002, issued May 8, 2001, ((SEQ ID NO:147), FIG. 9B)).
  • Example 1 describes the method used to treat PROb tumor-bearing rats with the PDGF-B specific aptamer. As can be seen in FIG. 2 treatment of PROb tumor-bearing rats with the PDGF-B specific aptamer resulted in a decrease in tumor IFP when compared to rats treated with the control aptamer. The mean IFP in control aptamer-treated tumors was 14.6±1.2 mm Hg (±S.E.M.) and 9.7±1.6 mm Hg (±S.E.M.) in tumors treated with the PDGF-B specific aptamer. The method used to determine IFP is described in Example 2.
  • PROb tumors were analyzed with regard to morphology, as well as distribution of PDGF-AB/BB and PDGF α-receptors as described in Example 3. The tumors displayed a heterogeneous morphology. At the tumor periphery, tumor cells were arranged in glandular structures, whereas more centrally, tumor cells were less abundant and less well organized (FIG. 3A). The central part was basically acellular (FIG. 3A). Expression of PDGF-AB/BB in PROb tumors was found in blood vessels and possibly in extravascular stromal cells surrounding tumor glands (FIG. 3B). In the central part of the tumors, few if any tumor cells were present, but strongly PDGF-AB/BB positive cells were seen (FIG. 3C). In no part of the tumors could PDGF-AB/BB positive tumor cells be clearly discerned. PDGF β-receptors were found in vascular cells of larger vessels, and in unidentified, possibly microvascular, cells in the stroma (FIG. 3D). The absence of PDGF-AB/BB and P-receptor expression by carcinoma cells in PROb tumors, is in agreement with the characteristics of cultured PROb cells (data not shown).
  • Prompted by these findings, the effects of treatment with PDGF aptamer (SEQ ID NO:1) were tested on the KAT-4 tumor model (Examples 4 and 5). Both of these tumor models showed PDGF receptor expression in tumor stroma but not on tumor cells. As can be seen in FIGS. 4 and 5 treatment with PDGF aptamers lowers IFP in KAT-4 tumors and increases the uptake of Taxol.
  • EXAMPLES
  • Materials and Methods
  • Tissue culture. Cells were cultured under standard conditions and all tissue culture media were supplemented with 10% fetal bovine serum (FBS) and antibiotics, unless otherwise stated. PAE cells were maintained in F12 culture medium (Sigma). PROb cells were kept in Dulbecco's Modified Eagle's Medium (Sigma).
  • PDGF/PDGF receptor inhibitors. The PDGF-B aptamer used in the present study was produced by the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) method. (Tuerk and Gold (1990) Science 249:505-510, which is incorporated herein by reference in its entirety). The modified DNA aptamer, linked to 40 kDa polyethylene glycol, has a high affinity for PDGF-B with a Kd of ˜0.1 nM. (Green et al. (1996) Biochemistry 35:14413-14424; Floege et al. (1999) Am. J. Pathol. 154:169-179, each of which is incorporated herein by reference in its entirety). The aptamer shows biphasic clearance in rats following iv injection, approximately 47% is cleared with a half-life of 32 minutes, while the remainder is cleared with a half-life of 135 minutes. (Floege et al. (1999) Am. J. Pathol. 154:169-179). As a control, a sequence-scrambled analog of the PDGF-B aptamer was used. This oligonucleotide has a Kd for PDGF-BB in the micromolar range. (Floege et al. (1999) Am. J. Pathol. 154:169-179).
  • Example 1 Tumor Establishment and Treatment of PROb Tumors with PDGF Inhibitors
  • Subcutaneously growing PROb tumors (Martin et al. (1996) Int. J. Cancer 65:796-804) were established in BDIX rats by injection of 5×106 tumor cells in 50 μL of PBS in the flank. The rats were kept under pathogen-free conditions and were fed ad libitum. They were monitored regularly for tumor growth and experiments were performed 8-12 weeks after tumor cell implantation on rats bearing tumors ranging in size between 0.6 cm3 and 7.6 cm3. The PDGF-B specific aptamer, and a control aptamer (see above), were given as i.p. injections in 2 mL PBS twice daily for 4 consecutive days at a dose of 7 mg×kg−1×day−1. All animal experiments described in the present report were approved by the Ethical Committee for Animal Experiments in Uppsala, Sweden.
  • Example 2 Measurement of Tumor IFP
  • Tumor IFP was measured using the wick-in-needle technique (Wiig et al. (1982) Scan. J. Clin. Lab. Invest. 42:159-164). Briefly, rats were anaesthetized using isofluran in a mixture of O2 and air. A standard 23-gauge needle filled with nylon-floss and saline, supplemented with 50 IE/mL of heparin was inserted into the center of the tumor and connected to a pressure transducer. This makeup enables continuous and stable recordings of fluid pressure. Fluid communication between the needle and the transducer was confirmed by compression and decompression of the tubing during each measurement. Tumor IFP was measured once before treatment with PDGF aptamers, and again 1-2 hours after the last administration of aptamers or vehicle alone. The change in tumor IFP was calculated for each tumor. After the second IFP measurement the rats were sacrificed and the tumors were excised and snap frozen in liquid nitrogen for further analyses.
  • Example 3 Immunohistochemistry
  • For routine morphology, paraffin-embedded 4 μm sections were stained with van Gieson staining. Immunohistochemistry was performed on 6 μm cryosections from PROb tumors. Sections were fixed in acetone and blocked with 0.3% hydrogen peroxide in methanol for 15 minutes, rinsed and further incubated in a solution containing 20% human normal serum in a buffer containing 2% rat serum, 3% bovine serum albumin, 0.01% NP40 in PBS (RM buffer) for 5 hours at 4° C. Primary antibodies dissolved in RM buffer were added, either 4 μg/mL affinity-purified rabbit anti-PDGF preceptor IgG (Claesson-Welsh et al. (1988) Mol. Cell Biol. 8:3476-3486) for 5 hours at 4° C., or overnight at 4° C. with 1.3 μg/mL of the monoclonal mouse anti-PDGF-AB/BB IgG (PGF 007, Mochida Pharmaceutical Company, Tokyo, Japan). Sections were rinsed in PBS with 0.01% NP40. Bound IgG was detected with biotinylated goat anti-rabbit or biotinylated rabbit anti-mouse antibodies, respectively. Sections were developed with a Vectastain ABC elite kit (Vector, Burlingame, Calif.) using amino-ethyl-carbazole as a chromophore. Sections were counter-stained with Mayer's hematoxylin for 30 seconds.
  • Example 4 Treatment of KAT-4 Tumors with PDGF Inhibitors
  • SCID-mice bearing subcutaneous KAT-4 tumors (human, anaplastic thyroid carcinoma) were pre-treated for 4 consecutive days with 12 mg×kg−1×day−1 SELEX aptamers (i.p. injections, three times daily). Tumor interstitial fluid pressure was measured using the wick-in-needle technique. The results are set forth in FIG. 4. * p<0.05, Student's t-test.
  • Example 5 Treatment of KAT-4 Tumors with a PDGF Inhibitor in Combination with the Cytotoxic Agent Taxol
  • SCID-mice bearing subcutaneous KAT-4 tumors (human, anaplastic thyroid carcinoma) were pre-treated for 4 consecutive days with 12 mg×kg−1×day−1 SELEX aptamers (i.p. injections, three times daily). [3H]Taxol was injected s.c at a site distant from the tumor and in a mix of 5 mg×kg−1 unlabelled Taxol. Eight or 24 hours following injection of radiolabelled drug, blood was sampled, animals were sacrificed and tumors and 4 other tissues were excised. Subsequently, tissues were weighed and homogenized in a RIPA lysis buffer and the amount radioactivity in each sample was determined in a scintillation counter. Tumor uptake of Taxol was expressed as cpm/g tumor tissue divided by cpm/ml blood. The results are set forth in FIG. 5. * p<0.05, Student's t-test.
  • Statistical analysis. Statistical analysis was performed using the paired or unpaired two-sided Student's t-test. A p-value <0.05 was considered statistically significant.

Claims (4)

1. A therapeutic composition comprising a PDGF aptamer and a cytotoxic agent.
2. The therapeutic composition of claim 1 wherein said PDGF aptamer is identified according to a method comprising:
a) preparing a candidate mixture of nucleic acids;
b) contacting the candidate mixture of nucleic acids with PDGF, wherein nucleic acids having an increased affinity to PDGF relative to the candidate mixture may be partitioned from the remainder of the candidate mixture;
c) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; and
d) amplifying the increased affinity nucleic acids to yield a mixture of nucleic acids enriched for nucleic acids with relatively higher affinity and specificity for binding to PDGF, whereby a nucleic acid ligand of PDGF may be identified.
3. The therapeutic composition of claim 1 wherein said PDGF aptamer is:
Figure US20070021327A1-20070125-C00001
wherein:
U at positions 6, 20 and 30 is 2′-fluoro-2′-deoxyuridine.
C at positions 8, 21, 28, and 29 is 2′-fluoro-2′-deoxycytidine.
G at positions 9, 15, 17, and 31 is 2′-O-Methyl-2′-deoxyguanosine.
A at position 22 is 2′-O-Methyl-2′-deoxyadenosine; and
Figure US20070021327A1-20070125-C00002
4. The therapeutic composition of claim 1 wherein said cytotoxic agent is selected from the group consisting of Bleomycin, Cisplatin, and Pt analogues; Carboplatin and Iproplatin, Cyclophosphamide, Daunorubicin, Doxofluoridine, Doxorubicin, Etoposide, Epirubicin, 5-Flurouracil, Gemzar, Ifosfamide, Melphalan, Methotrexate, Mithramycin, Mitomycin C, Mitoxanthrone, Streptozotocin, Taxol and Taxotere, Vincristine, Vinblastine, Vindesine, Vinorelbine, Topotecan and CPT-11
US11/518,321 1995-06-07 2006-09-08 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor Abandoned US20070021327A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/518,321 US20070021327A1 (en) 1995-06-07 2006-09-08 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US08/479,725 US5674685A (en) 1990-06-11 1995-06-07 High affinity PDGF nucleic acid ligands
US08/479,783 US5668264A (en) 1990-06-11 1995-06-07 High affinity PDGF nucleic acid ligands
US08/618,693 US5723594A (en) 1995-06-07 1996-03-20 High affinity PDGF nucleic acid ligands
US08/991,743 US6229002B1 (en) 1995-06-07 1997-12-16 Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US20500600P 2000-05-17 2000-05-17
US09/859,724 US6699843B2 (en) 1995-06-07 2001-05-17 Method for treatment of tumors using nucleic acid ligands to PDGF
US10/791,367 US7141375B2 (en) 1995-06-07 2004-03-02 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor
US11/518,321 US20070021327A1 (en) 1995-06-07 2006-09-08 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/791,367 Division US7141375B2 (en) 1995-06-07 2004-03-02 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor

Publications (1)

Publication Number Publication Date
US20070021327A1 true US20070021327A1 (en) 2007-01-25

Family

ID=33425824

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/859,724 Expired - Lifetime US6699843B2 (en) 1995-06-07 2001-05-17 Method for treatment of tumors using nucleic acid ligands to PDGF
US10/791,367 Expired - Fee Related US7141375B2 (en) 1995-06-07 2004-03-02 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor
US11/518,321 Abandoned US20070021327A1 (en) 1995-06-07 2006-09-08 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/859,724 Expired - Lifetime US6699843B2 (en) 1995-06-07 2001-05-17 Method for treatment of tumors using nucleic acid ligands to PDGF
US10/791,367 Expired - Fee Related US7141375B2 (en) 1995-06-07 2004-03-02 Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor

Country Status (1)

Country Link
US (3) US6699843B2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050096257A1 (en) * 2003-08-27 2005-05-05 David Shima Combination therapy for the treatment of ocular neovascular disorders
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US11273171B2 (en) 2013-07-12 2022-03-15 Iveric Bio, Inc. Methods for treating or preventing ophthalmological conditions

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6699843B2 (en) * 1995-06-07 2004-03-02 Gilead Sciences, Inc. Method for treatment of tumors using nucleic acid ligands to PDGF
AU2003247576A1 (en) * 2002-06-18 2003-12-31 Archemix Corp. Aptamer-toxin molecules and methods for using same
US8039443B2 (en) 2002-11-21 2011-10-18 Archemix Corporation Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20050124565A1 (en) * 2002-11-21 2005-06-09 Diener John L. Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
WO2004047742A2 (en) * 2002-11-21 2004-06-10 Archemix Corporation Multivalent aptamer therapeutics with improved pharmacodynamic properties and methods of making and using the same
US10100316B2 (en) * 2002-11-21 2018-10-16 Archemix Llc Aptamers comprising CPG motifs
GB0305422D0 (en) * 2003-03-10 2003-04-16 Univ Open Detection, monitoring and treatment of cancer
WO2005084412A2 (en) * 2004-03-05 2005-09-15 Archemix Corp. Controlled modulation of the pharmacokinetics and biodistribution of aptamer therapeutics
WO2005087808A2 (en) * 2004-03-05 2005-09-22 Ludwig Institute For Cancer Research Growth factor binding constructs materials and methods
SI1733056T1 (en) 2004-03-31 2013-10-30 The General Hospital Corporation Method to determine responsiveness of cancer to epidermal growth factor receptor targeting treatments
EP3034089A1 (en) * 2004-11-02 2016-06-22 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
ATE474046T1 (en) * 2004-11-29 2010-07-15 Noxxon Pharma Ag VASOPRESSIN BINDING L-NUCLIC ACID
CA2628451A1 (en) 2005-11-04 2007-05-18 Biogen Idec Ma Inc. Methods for promoting neurite outgrowth and survival of dopaminergic neurons
JP5829373B2 (en) 2006-01-27 2015-12-09 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Nogo receptor antagonist
US20080026947A1 (en) 2006-02-08 2008-01-31 Gmeiner William H Cytotoxic nucleotides for targeted therapeutics
PT2005185E (en) 2006-03-22 2011-01-27 Viral Logic Systems Technology Corp Methods for identifying polypeptide targets
WO2007128109A1 (en) * 2006-05-04 2007-11-15 University Health Network Aptamers that recognize the carbohydrate n-acetylgalactosamine (galnac)
US8377448B2 (en) * 2006-05-15 2013-02-19 The Board Of Trustees Of The Leland Standford Junior University CD47 related compositions and methods for treating immunological diseases and disorders
WO2007133811A2 (en) * 2006-05-15 2007-11-22 Viral Logic Systems Technology Corp. Cd47 related compositions and methods for treating immunological diseases and disorders
EP2094268A2 (en) * 2006-05-26 2009-09-02 Bayer HealthCare, LLC Drug combinations with substituted diaryl ureas for the treatment of cancer
AU2007306139B2 (en) * 2006-10-11 2014-02-27 Fusion Antibodies Limited Combination therapy
US20100151465A1 (en) 2008-03-27 2010-06-17 Jingyue Ju Selective Capture and Release of Analytes
GB0807018D0 (en) * 2008-04-17 2008-05-21 Fusion Antibodies Ltd Antibodies and treatment
WO2009149921A2 (en) * 2008-06-11 2009-12-17 Bionucleon S.R.L. Inhibition of hrp-3 using modified oligonucleotides
US9284559B2 (en) * 2009-04-14 2016-03-15 Wake Forest University Health Sciences Multivalent aptamer complexes
US9090663B2 (en) * 2009-04-21 2015-07-28 The Trustees Of Columbia University In The City Of New York Systems and methods for the capture and separation of microparticles
FR2948663B1 (en) * 2009-07-31 2011-09-02 Centre Nat Rech Scient "APTAMERS THAT INHIBIT THE ACTIVITY OF THE HUMAN PROTEIN Rad51 AND THEIR BIOLOGICAL APPLICATIONS"
EP2510110A1 (en) 2009-11-13 2012-10-17 Pangaea Biotech S.L. Molecular biomarkers for predicting response to tyrosine kinase inhibitors in lung cancer
EP2538965B1 (en) 2010-02-25 2017-04-12 Schepens Eye Research Institute Therapeutic compositions for the treatment of dry eye disease
ES2575160T3 (en) 2010-03-15 2016-06-24 The Board Of Trustees Of The University Of Illinois Inhibitors of the interactions that bind the alpha subunit of beta integrin-protein G
US20120095085A1 (en) * 2010-10-14 2012-04-19 Layzer Juliana M Nucleic acid modulators of clec-2
US9029502B2 (en) 2010-12-20 2015-05-12 The Regents Of The University Of Michigan Inhibitors of the epidermal growth factor receptor-heat shock protein 90 binding interaction
EP2468883A1 (en) 2010-12-22 2012-06-27 Pangaea Biotech S.L. Molecular biomarkers for predicting response to tyrosine kinase inhibitors in lung cancer
EP2492688A1 (en) 2011-02-23 2012-08-29 Pangaea Biotech, S.A. Molecular biomarkers for predicting response to antitumor treatment in lung cancer
WO2013019714A1 (en) 2011-07-29 2013-02-07 The Trustees Of Columbia University In The City Of New York Mems affinity sensor for continuous monitoring of analytes
WO2013052933A2 (en) 2011-10-06 2013-04-11 The Board Of Trustees Of The University Of Illinois Myosin binding protein-c for use in methods relating to diastolic heart failure
WO2013055791A1 (en) 2011-10-10 2013-04-18 The Regents Of The University Of Michigan Polymeric nanoparticles for ultrasound imaging and therapy
WO2013190089A1 (en) 2012-06-21 2013-12-27 Pangaea Biotech, S.L. Molecular biomarkers for predicting outcome in lung cancer
CN104661664B (en) 2012-07-13 2020-07-03 波涛生命科学有限公司 Chiral control
PT2733205T (en) 2012-11-20 2018-05-09 Univ Madrid Complutense Corticospinal upper motor neurons, methods and compositions for differentiating neural stem cells by modulating cb1 cannabinoid receptor signaling and uses thereof
PT2956476T (en) 2013-02-18 2020-02-21 Vegenics Pty Ltd Ligand binding molecules and uses thereof
US9486533B2 (en) 2013-09-27 2016-11-08 Wake Forest University Health Sciences Pharmaceutical compositions for high-capacity targeted delivery
WO2015120187A1 (en) 2014-02-05 2015-08-13 The University Of Chicago Chimeric antigen receptors recognizing cancer-spevific tn glycopeptide variants
ES2845650T3 (en) 2014-04-18 2021-07-27 Acceleron Pharma Inc Procedures to increase red blood cell levels and treat sickle cell disease
TN2016000553A1 (en) 2014-06-13 2018-04-04 Acceleron Pharma Inc Methods and compositions for treating ulcers
WO2016022696A1 (en) 2014-08-05 2016-02-11 The Trustees Of Columbia University In The City Of New York Method of isolating aptamers for minimal residual disease detection
MA41119A (en) 2014-12-03 2017-10-10 Acceleron Pharma Inc METHODS OF TREATMENT OF MYELODYSPLASIC SYNDROMES AND SIDEROBLASTIC ANEMIA
US10308719B2 (en) 2015-01-26 2019-06-04 The University Of Chicago IL13Rα2 binding agents and use thereof in cancer treatment
JP6912386B2 (en) 2015-01-26 2021-08-04 ザ ユニバーシティー オブ シカゴ CAR T cells that recognize cancer-specific IL13Rα2
EP3050574B1 (en) 2015-01-28 2019-10-09 Universite De Bordeaux Use of plerixafor for treating and/or preventing acute exacerbations of chronic obstructive pulmonary disease
MA41919A (en) 2015-04-06 2018-02-13 Acceleron Pharma Inc ALK4 HETEROMULTIMERS: ACTRIIB AND THEIR USES
MA41916A (en) 2015-04-06 2021-04-14 Acceleron Pharma Inc TGF-BETA SUPERFAMILY TYPE I AND TYPE II RECEIVER HETEROMULTIMERS AND THEIR USES
CN108348578B (en) 2015-08-04 2022-08-09 阿塞勒隆制药公司 Methods for treating myeloproliferative disorders
WO2017176265A1 (en) 2016-04-06 2017-10-12 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
CN108697793B (en) 2015-11-23 2023-08-01 阿塞勒隆制药公司 Methods of treating eye diseases
BR112018073660A2 (en) 2016-05-16 2019-02-19 Baxalta Incorporated anti-factor ix padua antibodies
DK3496739T3 (en) 2016-07-15 2021-05-10 Acceleron Pharma Inc COMPOSITIONS INCLUDING ACTRIIA POLYPEPTIDES FOR USE IN THE TREATMENT OF PULMONAL HYPERTENSION
EP3490582A4 (en) 2016-07-27 2020-04-01 Acceleron Pharma Inc. Methods and compositions for treating myelofibrosis
JP2019526579A (en) 2016-09-01 2019-09-19 マヨ ファウンデーション フォー メディカル エデュケーション アンド リサーチMayo Foundation For Medical Education And Research Methods and compositions for targeting T cell cancer
KR20220151022A (en) 2016-09-01 2022-11-11 메이오 파운데이션 포 메디칼 에쥬케이션 앤드 리써치 Carrier-pd-l1 binding agent compositions for treating cancers
CN110198743B (en) 2016-10-05 2023-07-18 艾科赛扬制药股份有限公司 Compositions and methods for treating kidney disease
US11249082B2 (en) 2016-10-29 2022-02-15 University Of Miami Zika virus assay systems
CN106754935B (en) * 2016-11-30 2019-04-30 吴冬 The aptamer WYZ-5 and its screening technique of ovarian mucinous cancer cell 3AO and application
CN106754936B (en) * 2016-11-30 2019-06-07 吴冬 The aptamer WYZ-2 and its screening technique of ovarian mucinous cancer cell 3AO and application
US20200030409A1 (en) 2017-03-06 2020-01-30 Tokyo Women's Medical University Lypd1 inhibitor and method for producing biological tissue using same
US11830582B2 (en) 2018-06-14 2023-11-28 University Of Miami Methods of designing novel antibody mimetics for use in detecting antigens and as therapeutic agents
EP4180516A4 (en) 2020-07-13 2024-01-17 Univ Kyoto Skeletal muscle precursor cells and method for purifying same, composition for treating myogenic diseases, and method for producing cell group containing skeletal muscle precursor cells
TW202216778A (en) 2020-07-15 2022-05-01 美商安進公司 Tigit and cd112r blockade
WO2022051414A1 (en) 2020-09-02 2022-03-10 Mayo Foundation For Medical Education And Research Antibody-nanoparticle complexes and methods for making and using the same
WO2023137161A1 (en) 2022-01-14 2023-07-20 Amgen Inc. Triple blockade of tigit, cd112r, and pd-l1

Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4711955A (en) * 1981-04-17 1987-12-08 Yale University Modified nucleotides and methods of preparing and using same
US4828979A (en) * 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
US4863902A (en) * 1985-11-28 1989-09-05 Wakunaga Seiyaku Kabushiki Kaisha Treatment of cancer
US4904582A (en) * 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US4935363A (en) * 1987-03-30 1990-06-19 Board Of Regents, The University Of Texas System Sterol regulatory elements
US5070010A (en) * 1989-10-30 1991-12-03 Hoffman-La Roche Inc. Method for determining anti-viral transactivating activity
US5149794A (en) * 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
US5171217A (en) * 1991-02-28 1992-12-15 Indiana University Foundation Method for delivery of smooth muscle cell inhibitors
US5270163A (en) * 1990-06-11 1993-12-14 University Research Corporation Methods for identifying nucleic acid ligands
US5345022A (en) * 1992-04-17 1994-09-06 Chevron Research And Technology Company Saturate minimization in normal alpha olefins
US5425940A (en) * 1986-04-09 1995-06-20 Cetus Oncology Corporation Combination therapy using interleukin-2 and tumor necrosis factor
US5459015A (en) * 1990-06-11 1995-10-17 Nexstar Pharmaceuticals, Inc. High-affinity RNA ligands of basic fibroblast growth factor
US5475096A (en) * 1990-06-11 1995-12-12 University Research Corporation Nucleic acid ligands
US5476766A (en) * 1990-06-11 1995-12-19 Nexstar Pharmaceuticals, Inc. Ligands of thrombin
US5502037A (en) * 1993-07-09 1996-03-26 Neuromed Technologies, Inc. Pro-cytotoxic drug conjugates for anticancer therapy
US5521184A (en) * 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
US5563255A (en) * 1994-05-31 1996-10-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US5562922A (en) * 1993-03-18 1996-10-08 Cedars-Sinai Medical Center Drug incorporating and release polymeric coating for bioprosthesis
US5593974A (en) * 1991-06-28 1997-01-14 Massachusetts Institute Of Technology Localized oligonucleotide therapy
US5595877A (en) * 1990-06-11 1997-01-21 Nexstar Pharmaceuticals, Inc. Methods of producing nucleic acid ligands
US5614503A (en) * 1993-11-12 1997-03-25 Aronex Pharmaceuticals, Inc. Amphipathic nucleic acid transporter
US5631237A (en) * 1992-12-22 1997-05-20 Dzau; Victor J. Method for producing in vivo delivery of therapeutic agents via liposomes
US5668264A (en) * 1990-06-11 1997-09-16 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US5674685A (en) * 1990-06-11 1997-10-07 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US5723594A (en) * 1995-06-07 1998-03-03 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US5756291A (en) * 1992-08-21 1998-05-26 Gilead Sciences, Inc. Aptamers specific for biomolecules and methods of making
US5795910A (en) * 1994-10-28 1998-08-18 Cor Therapeutics, Inc. Method and compositions for inhibiting protein kinases
US5811533A (en) * 1990-06-11 1998-09-22 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US6011020A (en) * 1990-06-11 2000-01-04 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US6025507A (en) * 1995-02-17 2000-02-15 Schering Aktiengesellschaft Borneol derivatives, methods of manufacturing them, and their pharmaceutical use
US6054122A (en) * 1990-11-27 2000-04-25 The American National Red Cross Supplemented and unsupplemented tissue sealants, methods of their production and use
US6106866A (en) * 1995-07-31 2000-08-22 Access Pharmaceuticals, Inc. In vivo agents comprising cationic drugs, peptides and metal chelators with acidic saccharides and glycosaminoglycans, giving improved site-selective localization, uptake mechanism, sensitivity and kinetic-spatial profiles, including tumor sites
US6229002B1 (en) * 1995-06-07 2001-05-08 Nexstar Pharmaceuticlas, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US6699843B2 (en) * 1995-06-07 2004-03-02 Gilead Sciences, Inc. Method for treatment of tumors using nucleic acid ligands to PDGF

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH0229046H1 (en) 1985-03-30 1998-07-15 Stuart Alan Kauffman METHOD FOR OBTAINING DNA, RNA, PEPTIDES, POLYPEPTINIQUE. DES OR PROTEINS BY MEANS OF A DNA RECOMBINANT TECH
WO1989006694A1 (en) 1988-01-15 1989-07-27 Trustees Of The University Of Pennsylvania Process for selection of proteinaceous substances which mimic growth-inducing molecules
EP0549692A4 (en) 1990-09-21 1993-08-25 Fred Hutchinson Cancer Research Center Protein sequence-specific oligonucleotide sequences
AU1435492A (en) 1991-02-21 1992-09-15 Gilead Sciences, Inc. Aptamer specific for biomolecules and method of making

Patent Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4711955A (en) * 1981-04-17 1987-12-08 Yale University Modified nucleotides and methods of preparing and using same
US4828979A (en) * 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
US4863902A (en) * 1985-11-28 1989-09-05 Wakunaga Seiyaku Kabushiki Kaisha Treatment of cancer
US4683195B1 (en) * 1986-01-30 1990-11-27 Cetus Corp
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5425940A (en) * 1986-04-09 1995-06-20 Cetus Oncology Corporation Combination therapy using interleukin-2 and tumor necrosis factor
US4935363A (en) * 1987-03-30 1990-06-19 Board Of Regents, The University Of Texas System Sterol regulatory elements
US4904582A (en) * 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US5070010A (en) * 1989-10-30 1991-12-03 Hoffman-La Roche Inc. Method for determining anti-viral transactivating activity
US5475096A (en) * 1990-06-11 1995-12-12 University Research Corporation Nucleic acid ligands
US5811533A (en) * 1990-06-11 1998-09-22 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US5674685A (en) * 1990-06-11 1997-10-07 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US5668264A (en) * 1990-06-11 1997-09-16 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US5459015A (en) * 1990-06-11 1995-10-17 Nexstar Pharmaceuticals, Inc. High-affinity RNA ligands of basic fibroblast growth factor
US5595877A (en) * 1990-06-11 1997-01-21 Nexstar Pharmaceuticals, Inc. Methods of producing nucleic acid ligands
US5476766A (en) * 1990-06-11 1995-12-19 Nexstar Pharmaceuticals, Inc. Ligands of thrombin
US5270163A (en) * 1990-06-11 1993-12-14 University Research Corporation Methods for identifying nucleic acid ligands
US6011020A (en) * 1990-06-11 2000-01-04 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US5149794A (en) * 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
US6054122A (en) * 1990-11-27 2000-04-25 The American National Red Cross Supplemented and unsupplemented tissue sealants, methods of their production and use
US5171217A (en) * 1991-02-28 1992-12-15 Indiana University Foundation Method for delivery of smooth muscle cell inhibitors
US5593974A (en) * 1991-06-28 1997-01-14 Massachusetts Institute Of Technology Localized oligonucleotide therapy
US5521184A (en) * 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
US5345022A (en) * 1992-04-17 1994-09-06 Chevron Research And Technology Company Saturate minimization in normal alpha olefins
US5756291A (en) * 1992-08-21 1998-05-26 Gilead Sciences, Inc. Aptamers specific for biomolecules and methods of making
US5631237A (en) * 1992-12-22 1997-05-20 Dzau; Victor J. Method for producing in vivo delivery of therapeutic agents via liposomes
US5562922A (en) * 1993-03-18 1996-10-08 Cedars-Sinai Medical Center Drug incorporating and release polymeric coating for bioprosthesis
US5502037A (en) * 1993-07-09 1996-03-26 Neuromed Technologies, Inc. Pro-cytotoxic drug conjugates for anticancer therapy
US5614503A (en) * 1993-11-12 1997-03-25 Aronex Pharmaceuticals, Inc. Amphipathic nucleic acid transporter
US5563255A (en) * 1994-05-31 1996-10-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US5795910A (en) * 1994-10-28 1998-08-18 Cor Therapeutics, Inc. Method and compositions for inhibiting protein kinases
US6025507A (en) * 1995-02-17 2000-02-15 Schering Aktiengesellschaft Borneol derivatives, methods of manufacturing them, and their pharmaceutical use
US5723594A (en) * 1995-06-07 1998-03-03 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US6229002B1 (en) * 1995-06-07 2001-05-08 Nexstar Pharmaceuticlas, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US6582918B2 (en) * 1995-06-07 2003-06-24 Gilead Sciences, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US6699843B2 (en) * 1995-06-07 2004-03-02 Gilead Sciences, Inc. Method for treatment of tumors using nucleic acid ligands to PDGF
US6106866A (en) * 1995-07-31 2000-08-22 Access Pharmaceuticals, Inc. In vivo agents comprising cationic drugs, peptides and metal chelators with acidic saccharides and glycosaminoglycans, giving improved site-selective localization, uptake mechanism, sensitivity and kinetic-spatial profiles, including tumor sites

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20050096257A1 (en) * 2003-08-27 2005-05-05 David Shima Combination therapy for the treatment of ocular neovascular disorders
US20100119522A1 (en) * 2003-08-27 2010-05-13 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US20100129364A1 (en) * 2003-08-27 2010-05-27 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US7759472B2 (en) 2003-08-27 2010-07-20 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US20110200593A1 (en) * 2003-08-27 2011-08-18 Ophthotech Corporation Combination Therapy for the Treatment of Ocular Neovascular Disorders
US8187597B2 (en) 2003-08-27 2012-05-29 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US8206707B2 (en) 2003-08-27 2012-06-26 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US8685397B2 (en) 2003-08-27 2014-04-01 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US11273171B2 (en) 2013-07-12 2022-03-15 Iveric Bio, Inc. Methods for treating or preventing ophthalmological conditions

Also Published As

Publication number Publication date
US20040224335A1 (en) 2004-11-11
US20020034506A1 (en) 2002-03-21
US7141375B2 (en) 2006-11-28
US6699843B2 (en) 2004-03-02

Similar Documents

Publication Publication Date Title
US7141375B2 (en) Methods and compositions for treatment of tumors using nucleic acid ligands to platelet-derived growth factor
AU777043B2 (en) Nucleic acid ligands to CD40ligand
US6124449A (en) High affinity TGFβ nucleic acid ligands and inhibitors
ES2426160T3 (en) Platelet-derived growth factor nucleic acid ligand complexes (PDGF)
US20060105378A1 (en) Tenascin-C nucleic acid ligands
US20040258656A1 (en) High affinity TGFbeta nucleic acid ligands and inhibitors
US20040009510A1 (en) Allosteric nucleic acid sensor molecules
WO1999007724A1 (en) Systematic evolution of ligands by exponential enrichment: tissue selex
JP2004511250A (en) Nucleic acid ligand for prostate-specific membrane antigen
RU2571660C2 (en) Modifiers of glycoprotein vi representing nucleic acid
WO2001087351A1 (en) Method for treatment of tumors using nucleic acid ligands to pdgf
US20040224915A1 (en) High affinity vascular endothelial growth factor(VEGF) receptor nucleic acid ligands and inhibitors
US20160053266A1 (en) Stabilized Aptamers to Platelet Derived Growth Factor and Their Use as Oncology Therapeutics
US7005260B1 (en) Tenascin-C nucleic acid ligands
US20030065155A1 (en) Nucleic acid sensor molecules

Legal Events

Date Code Title Description
AS Assignment

Owner name: GILEAD SCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LUDWIG INSTITUTE FOR CANCER RESEARCH;REEL/FRAME:018332/0337

Effective date: 20011212

Owner name: GILEAD SCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:RUBIN, KRISTOFER;REEL/FRAME:018332/0349

Effective date: 20011211

Owner name: LUDWIG INSTITUTE FOR CANCER RESEARCH, UNITED KINGD

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PIETRAS, KRISTIAN;OSTMAN, ARNE;HELDIN, CARL-HENRIK;REEL/FRAME:018332/0174

Effective date: 20011211

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION