US20040161816A1 - Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique - Google Patents

Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique Download PDF

Info

Publication number
US20040161816A1
US20040161816A1 US10/138,213 US13821302A US2004161816A1 US 20040161816 A1 US20040161816 A1 US 20040161816A1 US 13821302 A US13821302 A US 13821302A US 2004161816 A1 US2004161816 A1 US 2004161816A1
Authority
US
United States
Prior art keywords
stochastic
process according
protein
peptide
polypeptide
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
US10/138,213
Inventor
Stuart Kauffman
Marc Ballivet
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=4209046&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040161816(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US08/464,569 external-priority patent/US6492107B1/en
Application filed by Individual filed Critical Individual
Priority to US10/138,213 priority Critical patent/US20040161816A1/en
Publication of US20040161816A1 publication Critical patent/US20040161816A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1093General methods of preparing gene libraries, not provided for in other subgroups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • C07K14/485Epidermal growth factor [EGF], i.e. urogastrone
    • 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
    • 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
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/66General methods for inserting a gene into a vector to form a recombinant vector using cleavage and ligation; Use of non-functional linkers or adaptors, e.g. linkers containing the sequence for a restriction endonuclease
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • C12N15/68Stabilisation of the vector
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • 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
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • C12N9/2468Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1) acting on beta-galactose-glycoside bonds, e.g. carrageenases (3.2.1.83; 3.2.1.157); beta-agarase (3.2.1.81)
    • C12N9/2471Beta-galactosidase (3.2.1.23), i.e. exo-(1-->4)-beta-D-galactanase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01023Beta-galactosidase (3.2.1.23), i.e. exo-(1-->4)-beta-D-galactanase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention has as its object a process for obtaining DNA, RNA, peptides, polypeptides, or proteins, through use of transformed host cells containing genes capable of expressing these RNAs, peptides, polypeptides, or proteins; that is to say, by utilization of recombinant DNA technique.
  • the invention aims in particular at the production of stochastic genes or fragments of stochastic genes in a fashion to permit obtaining simultaneously, after transcription and translation of these genes, a very large number (on the order of at least 10,000) of completely new proteins, in the presence of host cells (bacterial or eukaryotic) containing these genes respectively capable of expressing these proteins, and to carry out thereafter a selection or screen among the said clones, in order to determine which of them produce proteins with desired properties, for example, structural, enzymatic, catalytic, antigenic, pharmacologic, or properties of liganding, and more generally, chemical, biochemical, biological, etc. properties.
  • desired properties for example, structural, enzymatic, catalytic, antigenic, pharmacologic, or properties of liganding, and more generally, chemical, biochemical, biological, etc. properties.
  • the invention also has as its aim procedures to obtain, sequences of DNA or RNA with utilizable properties notably chemical, biochemical, or biological properties.
  • the process for production of peptides or polypeptides according to the invention is characterized in that one produces simultaneously, in the same medium, genes which are at least partially composed of synthetic stochastic polynucleotides, that one introduces the genes thus obtained into host cells, that one cultivates simultaneously the independent clones of the transformed host cells containing these genes in such a manner so as to clone the stochastic genes and to obtain the production of the proteins expressed by each of these stochastic genes, that one carries out selection and/or screening of the clones of transformed host cells in a manner to identify those clones producing peptides or polypeptides having at least one desired activity, that one thereafter isolates the clones thus identified and that one cultivates them to produce at least one peptide or polypeptide having the said property.
  • stochastic genes are produced by stochastic copolymerization of the four kinds of deoxyphosphonucleotides, A, C, G and T from the two ends of an initially linearized expression vector, followed by formation of cohesive ends in such a fashion as to form a stochastic first strand of DNA constituted by a molecule of expression vector possessing two stochastic sequences whose 3′ ends are complementary, followed by the synthesis of the second strand of the stochastic DNA.
  • stochastic genes are produced by copolymerization of oligonucleotides without cohesive ends, in a manner to form fragments of stochastic DNA, followed by ligation of these fragments to a previously linearized expression vector.
  • the expression vector can be a plasmid, notably a bacterial plasmid. Excellent results have been obtained using the plasmid pUC8 as the expression vector.
  • the expression vector can also be viral DNA or a hybrid of plasmid and viral DNA.
  • the host cells can be prokaryotic cells such as HB 101 and C 600, or eukaryotic cells.
  • the property serving as the criterion for selection of the clones of host cells can be the capacity of the peptides or polypeptides, produced by a given clone, to catalyze a given chemical reaction.
  • the said property can be the capacity to catalyze a sequence of reactions leading from an initial group of chemical compounds to at least one target compound.
  • the said property can be the capacity to catalyze the synthesis of the same ensemble from amino acids and/or oligopeptides in an appropriate milieu.
  • the said property can also be the capacity to modify selectively the biological or chemical properties of a given compound, for example, the capacity to selectively modify the catalytic activity of a polypeptide.
  • the said property can also be the capacity to stimulate, inhibit, or modify at least one biological function of at least one biologically active compound, chosen, for example, among the hormones, neurotransmitters, adhesion factors, growth factors, and specific regulators of DNA replication and/or transcription and/or translation of RNA.
  • the said property can equally be the capacity of the peptide or polypeptide to bind to a given ligand.
  • the invention also has as its object the use of the peptide or polypeptide obtained by the process specified above, for the detection and/or the titration of a liquid.
  • the criterion for selection of the clones of transformed host cells is the capacity of these peptides or polypeptides to simulate or modify the effects of a biologically active molecule, for example, a protein, and screening and/or selection for clones of transformed host cells producing at least one peptide or polypeptide having this property, is carried out by preparing antibodies against the active molecule, then utilizing these antibodies after their purification, to identify the clones containing this peptide or polypeptide, then by cultivating the clones thus identified, separating and purifying the peptide or polypeptide produced by these clones, and finally by submitting the peptide or polypeptide to an in vitro assay to verify that it has the capacity to simulate or modify the effects of the said molecule.
  • a biologically active molecule for example, a protein
  • the property serving as the criterion of selection is that of having at least one epitope similar to one of the epitopes of a given antigen.
  • the invention carries over to obtaining polypeptides by the process specified above and utilizable as chemotherapeutically active substances.
  • the invention permits obtaining polypeptides usable for chemotherapeutic treatment of epitheliomas.
  • one identifies and isolates the clones of transformed host cells producing peptides or polypeptides having the property desired, by affinity chromatography against antibodies corresponding to a protein expressed by the natural pan of the DNA hybrid.
  • the natural part of the hybrid DNA contains a gene expressing B-galactosidase
  • the invention also applies to a use of the process specified above for the preparation of a vaccine; the application is characterized by the fact that antibodies against the pathogenic agent are isolated, for example, antibodies formed after injection of the pathogenic agent in the body of an animal capable of forming antibodies against this agent, and these antibodies are used to identify the clones producing at least one protein having at least one epitope similar to one of the epitopes of the pathogenic agent, the transformed host cell corresponding to these clones are cultured to produce these proteins, this protein is isolated and purified from the clones of cells, then this protein is used for the production of a vaccine against the pathogenic agent.
  • antibodies against the pathogenic agent are isolated, for example, antibodies formed after injection of the pathogenic agent in the body of an animal capable of forming antibodies against this agent, and these antibodies are used to identify the clones producing at least one protein having at least one epitope similar to one of the epitopes of the pathogenic agent, the transformed host cell corresponding to these clones are cultured to produce these proteins, this protein is
  • an anti-HVB vaccine For example in order to prepare an anti-HVB vaccine, one can extract and purify at least one capsid protein of the HVB virus, inject this protein into an animal capable of forming antibodies against this protein having at least one epitope similar to one of the epitopes of the HVB virus, then cultivate the clones of transformed host cells corresponding to these clones in a manner to produce this protein, isolate and purify the protein from culture of these clones of cells and utilize the protein for the production of an anti-HVB vaccine.
  • the host cells consist in bacteria such as Escherichia coli whose genome contains neither the natural gene expressing ⁇ -galactosidase, nor the EBG gene, that is to say, Z ⁇ , EBG ⁇ E. coli .
  • the transformed cells are cultured in the presence of X gal and the indicator IPTG in the medium, and cells positive for ⁇ -galactosidase functions are detected; thereafter, the transforming DNA is transplanted into an appropriate clone of host cells for large scale culture to produce at least one peptide or polypeptide.
  • the property serving as the criterion for selection of the transformed host cells can also be the capacity of the polypeptides or proteins produced by the culture of these clones to bind to a given compound.
  • This compound can be in particular chosen advantageously among peptides, polypeptides, and proteins, notably among proteins regulating the transcription activity of DNA.
  • the said compound can also be chosen among DNA and RNA sequences.
  • the invention has also as its object those proteins which are obtained in the case where the property serving as criterion of selection of the clones of transformed host cells consist in the capacity of these proteins to bind to regulatory proteins controlling transcription activity of the DNA, or else to DNA and RNA sequences.
  • the invention has, in addition, as an object, the use of a protein which is obtained in the first particular case above mentioned, as a cis-regulatory sequence controlling replication or transcription of a neighboring DNA sequence.
  • the aim of the invention also includes utilization of proteins obtained in the second case mentioned to modify the properties of transcription or replication of a sequence of DNA, in a cell containing the sequence of DNA, and expressing this protein.
  • the invention has as its object as well as a process of production of DNA, characterized by simultaneous production in the same medium, of genes at least partially composed of stochastic synthetic polynucleotides, in that the genes thus obtained are introduced into host cells to produce an ensemble of transformed host cells, in that screening and/or selection on this ensemble is carried out to identify those host cells containing in their genome stochastic sequences of DNA having at least one desired property, and finally, in that the DNA from the clones of host cells thus identified is isolated.
  • the invention further has as its object a procedure to produce RNA, characterized by simultaneous production in the same medium, of genes at least partially composed of stochastic synthetic polynucleotides, in that the genes thus obtained are introduced into host cells to produce an ensemble of transformed host cells, in that the host cells so produced are cultivated simultaneously, and screening and/or selection of this ensemble is carried out in a manner to identify those host cells containing stochastic sequences of RNA having at least one desired property, and in that the RNA is isolated from the host cells thus identified.
  • the said property can be the capacity to bind a given compound, which might be, for example, a peptide or polypeptide or protein, or also the capacity to catalyze a given chemical reaction, or the capacity to be a transfer RNA.
  • 30 micrograms that is, approximately 10 13 molecules of the pUC8 expression vector are linearized by incubation for 2 hours at 37° C. with 100 units of the Pst1 restriction enzyme in a volume of 300 ⁇ l of the appropriate standard buffer.
  • the linearized vector is treated with phenol-chloroform then precipitated in ethanol, taken up in volume of 30 ⁇ l and loaded onto a 0.8% agarose gel in standard TEB buffer. After migration in a field of 3V/cm for three hours, the linearized vector is electro-eluted, precipitated in ethanol, and taken up in 30 ⁇ l of water.
  • the reaction attains or passes a mean value of 300 nucleotides added per 3′ extremity, it is stopped and the free nucleotides are separated from the polymer by differential precipitation or by passage over a column containing a molecular sieve such as Biogel P60. After concentration by precipitation in ethanol, the polymers are subjected to a further polymerization with TdT, first in the presence of dATP, then in the presence of dTTP. These last two reactions are separated by a filtration on a gel and are carried out for short intervals (30 seconds to 3 minutes) in order to add sequentially 10-30 A followed by 10-30 T to the 3′ ends of the polymers.
  • Each molecule of vector possess, at the end of the preceding operation, two stochastic sequences whose 3′ ends are complementary.
  • the mixture of polymers is therefore incubated in conditions favoring hybridization of the complementary extremities (150 mM NaCl, 10 mM Tris-HCl, pH 7.6, 1 mM EDTA at 650 for 10 minutes, followed by lowering the temperature to 22° C. at a rate of 3 to 4° C. per hour).
  • the hybridized polymers are then reacted with 60 units of the large fragment (Klenow) of polymerase 1, in the presence of the four nucleotide triphosphates (200 mM) at 4° C. for two hours. This step accomplishes the synthesis of the second strand from the 3′ ends of the hybrid polymers.
  • the molecules which result from this direct synthesis starting from linearized vector are thereafter utilized transform competent cells.
  • the amplified culture is centrifuged, and the pellet of transformed cells is lyophilysed and stored at ⁇ 70° C.
  • Such a culture contains 3 ⁇ 10 7 to 10 8 independent transformants, each containing a unique stochastic gene inserted into the expression vector.
  • [0058] is composed of 5 palindromes (thus self-complementary sequences) where it is easy to verify that their stochastic polymerization does not generate any “stop” codons, and specifies all the amino acids.
  • a mixture containing 5 ⁇ g each of the oligonucleotides indicated above is reacted in a 100 ul volume containing 1 mM ATP, 10% polyethyleneglycol, and 100 units of T4 DNA ligase in the appropriate buffer at 13° C. for six hours.
  • This step carries out the stochastic polymerization of the oligomers in the double stranded state and without cohesive ends.
  • the resulting polymers are isolated by passage over a molecular sieve (Biogel P60) recovering those with 20 to 100 oligomers. After concentration, this fraction is again submitted to catalysis or polymerization by T4 DNA ligase under the conditions described above. Thereafter, as described above, those polymers which have assembled at least 100 oligomers are isolated.
  • the pUC8 expression vector is linearized by Sma1 enzyme in the appropriate buffer, as described above.
  • the vector linearized by Sma1 does not have cohesive ends.
  • the linearized vector is treated by calf intestine alkaline phosphatase (CIP) at a level of one unit per microgram of vector in the appropriate buffer, at 37° C. for 30 minutes.
  • CIP calf intestine alkaline phosphatase
  • the CIP enzyme is thereafter inactivated by two successive extractions with phenol-chloroform.
  • the linearized and dephosphorylated vector is precipitated in ethanol, then redissolved in water at 1 mg/ml.
  • This procedure differs from that just discussed in that it utilizes palindromic heptamers which have variable cohesive ends, in place of stochastic sequences containing a smaller number of identical motifs.
  • the polymers thus obtained have one unpaired base on their two 5′ extremities. Thus, it is necessary to add the complementary base to the corresponding 3′ extremities.
  • This is carried out as follows: 10 micrograms of the double stranded polymers are reacted with 10 units of the Klenow enzyme, in the presence of the four deoxynucleotidephosphates (200 mM) in a volume of 100 ⁇ l, at 4° C., for 60 minutes.
  • the enzyme is inactivated by phenol chloroform extraction, and the polymers are cleansed of the residual free nucleotides by differential precipitation.
  • the polymers are then ligated to the host plasmid (previously linearized and dephosphorylated) by following the procedures described above.
  • the two last procedures which were described utilize palindromic octamers or heptamers which constitute specific sites of certain restriction enzymes. These sites are absent, for the most part, from the pUC8 expression vector.
  • the plasmid DNA derived from the culture of 10 7 independent transformants obtained by one of the two last procedures described above is isolated. After this DNA is purified, it is partially digested by Cla1 restriction enzyme (procedure II) or by the Pst1 restriction enzyme (procedure III).
  • the polymerization can be carried out directly on a cloning expression vector, which was previously linearized; or it is possible to proceed sequentially to the polymerization then the ligation of the polymers to the expression vector.
  • the next step is transformation or transfection of competent bacterial cells (or cells in culture). This step constitutes cloning the stochastic genes in living cells where they are indefinitely propagated and expressed.
  • the further step of the procedure according to the invention consists in examining the transformed or transfected cells by selection or screening, in order to isolate one or several cells whose transforming or transfecting DNA leads to the synthesis of a transcription product (RNA) or translation product (protein) having desired property.
  • RNA transcription product
  • protein translation product
  • properties can be, for example, enzymatic, functional, or structural.
  • RNA or protein an exploitable product
  • DNA synthesized and cloned as described can be selected or screened in order to isolate sequences of DNA constituting products in themselves, having exploitable biochemical properties.
  • EGF epithelial growth factor
  • KLH keyhole limpet hemocyanin
  • the purified anti-EGF antibodies are then used as probes to screen a large number of bacterial clones lysed by chloroform, and on a solid support.
  • the anti-EGF antibodies bind those stochastic peptides or proteins whose epitopes resemble those of the initial antigen.
  • the clones containing such peptides or proteins are shown by autoradiography after incubation of the solid support with radioactive protein A, or after incubation with a radioactive antibody.
  • a variant of this procedure consists in purifying these stochastic peptides, polypeptides, or proteins, which can be used as vaccines or more generally, to confer an immunity against a pathogenic agent or to exercise other effects on the immunological system, for example, to create a tolerance or diminish hypersensitivity with respect to a given antigen, in particular, due to binding of these peptides, polypeptides, or proteins with the antibodies directed against this antigen. It is clear that it is possible to use such peptides, polypeptides, or proteins in vitro as well as in vivo.
  • each has at least one epitope in common with X, thus the ensemble has an ensemble of epitopes in common with X.
  • This permits utilization of the ensemble or sub-ensemble as a vaccine to confer immunity against X. It is, for example, easy to purify one or several of the capsid proteins of the hepatitis B virus. These proteins can then be injected into an animal, for example, a rabbit, and the antibodies corresponding to the initial antigen can be recovered by affinity column purification. These antibodies may be used, as described above, to identify clones producing at least one protein having an epitope resembling at least one of the epitopes of the initial antigen. After purification, these proteins are used as antigens (either alone or in combination) with the aim of conferring protection against hepatitis B. The final production of the vaccine does not require further access to the initial pathogenic agent.
  • the first step of the process consists in generating a very large ensemble of expression vectors, each expressing a distinct novel protein.
  • the pUC8 expression vector with cloning of stochastic sequences of DNA in the Pst1 restriction site.
  • the plasmids thus obtained are then introduced into a clone of E. coli from whose genome the natural gene for ⁇ -galactosidase, Z, and a second gene EBG, unrelated to the first but able to mutate towards ⁇ -gal function, have both been eliminated by known genetic methods.
  • Such host cells (Z ⁇ , EBG ⁇ ) are not able by themselves to catalyse lactose hydrolysis, and as a consequence, to use lactose as a carbon source for growth. This permits utilization of such host clones for screening or selection for ⁇ -gal function.
  • a convenient biological assay to analyze transformed E. coli clones for those which have novel genes expressing a ⁇ -gal function consists in the culture of bacteria transformed as described in petri dishes containing X-gal in the medium. In this case, all bacterial colonies expressing a ⁇ -gal function are visualized as blue colonies. By using such a biological assay, it is possible to detect even weak catalytic activity.
  • the specific activity of characteristic enzymes ranges from 10 to 10,000 product molecules per second.
  • each cell would need to catalyse cleavage of between 10,000 and 100 of X-gal to be detected, which would require between 2.7 and 270 hours. Since under selective conditions it is possible to amplify the number of copies of each plasmid per cell from 5 to 20 copies per cell, or even to 100 or 1000, and because up to 10% of the protein of the cell can be specified by the new gene, the duration needed to detect a blue colony in the case of 100 enzyme molecules of weak activity per cell is on the order of 0.27 to 2.7 hours.
  • bacterial colonies which appear blue on X-gal Petri dishes might be false positives due to a mutation in the bacterial genome which confers upon it the capacity to metabolize lactose, or for other reasons than those which result from a catalytic activity of the novel protein expressed by the cells of the colony.
  • Such false positives can be directly eliminated by purifying the DNA of the expression vector from the positive colony, and retransforming Z ⁇ , EBG ⁇ E. coli host cells. If the ⁇ -gal activity is due to the novel protein coded by the new gene in the expression vector, all those cells transformed by that vector will exhibit ⁇ -gal function.
  • the initial blue colony is due to a mutation in the genome of the host cell, it is a rare event and independent of the transformation, thus the number of cells of the new clone of the transformed E. coli capable of expressing gal function will be small or zero.
  • Mass purification of the expression vectors from the total of 220 positive bacterial clones followed by retransformation of the naive bacteria with the mixture of these expression vectors will produce a large number of positive clones consisting of all those bacteria transformed with the 20 expression vectors which code for the novel proteins having the desired function, and a very small number of bacterial clones resulting from genomic mutations and containing the 200 expression vectors which are not of interest.
  • a small number of cycles of purification of expression vectors from positive bacterial colonies, followed by such retransformation allows the detection of very rare expression vectors truly positive for a desired catalytic activity, despite a high background rate of mutations in the host cells for the same function.
  • selection for ⁇ -galactosidase function shall be described.
  • An appropriate clone of Z ⁇ EBG ⁇ E. coli is not able to grow on lactose as the sole carbon source.
  • selection techniques When both selection techniques and convenient bioassay techniques exist at the same time, as in the present case, it is possible to use selection techniques initially to enrich the representation of host bacteria expressing the ⁇ -gal function, then carry out a screening on a Petri plate on X-gal medium to establish efficiently which are the positive cells. In the absence of convenient bioassays, application of progressively stricter selection is the easiest route to purify one or a small number of distinct host cells whose expression vectors code for the proteins catalyzing the desired reaction.
  • a clone mutant in the repressor of the lactose operon expresses ⁇ -gal function constitutively due to the fact the lactose operator is not repressed. All cells of this type produce blue clones on Petri plates containing X-gal medium. It is possible to transform such host cells with expression vectors synthesizing novel proteins and carry out a screen on X-gal Petri plates in order to detect those clones which are not blue. Among those, some represent the case where the new protein binds to the lactose operator and represses the synthesis of ⁇ -gal. It is then feasible to mass isolate such plasmids, retransform, isolate those clones which do not produce ⁇ -gal, and thereafter carry out a detailed verification.
  • the process can be utilized in order to create, then isolate, not only exploitable proteins, but also RNA and DNA as products in themselves, having exploitable properties.
  • the procedure consists in creating stochastic sequences of DNA which may interact directly with other cellular or biochemical constituents, and on the other hand, these sequences cloned in expression vectors are transcribed into RNA which are themselves capable of multiple biochemical interactions.
  • This example illustrates selection for a useful DNA, and the purification and study of the mechanism of action of regulatory proteins which bind to the DNA.
  • oestradiol receptor a protein obtained by standard techniques. In the presence of oestradiol, a steroid sexual hormone, the receptor changes conformation and binds tightly to certain specific sequences in the genomic DNA, thus affecting the transcription of genes implicated in sexual differentiation and the control of fertility.
  • RNA transcribed from these sequences in the transformed host cells can be useful products themselves.
  • tRNA suppressor transfer RNA
  • Each of these novel proteins is a candidate to catalyse one or another of the possible reactions, in the set of all the possible reactions leading from the ensemble of building blocks to the target compound. If a sufficiently large number of stochastic proteins is present in a reaction mixture containing the building block compounds, such that a sufficiently large number of the possible reactions are catalyzed, there is a high probability that one connected sequence of reactions leading from the set of building block compounds to the target compound will be catalyzed by a subset of the novel proteins. It is clear that this procedure can be extended to the catalysis not only of one, but of several target compounds simultaneously.
  • Formation of a peptide bond requires energy in a dilute aqueous medium, but if the peptides participating in the condensation reactions are adequately concentrated, formation of peptide bonds is thermodynamically favored over hydrolysis and occurs efficiently in the presence of an appropriate enzymatic catalyst, for example pepsin or trypsin, without requiring the presence of ATP or other high energy compounds.
  • an appropriate enzymatic catalyst for example pepsin or trypsin
  • the pentapeptide is formed in the conditions described above, but not when an extract is utilized which is derived from host cells transformed by an expression vector containing no stochastic genes, the formation of the pentapeptide is not the result of bacterial contaminants and thus requires the presence of a subset of the novel proteins in the reaction mixture.
  • the following step consists in the separation of the particular subset of cells which contain expression vectors with the novel proteins catalyzing the sequence of reactions leading to the target pentapeptide. As an example, if the number of reactions forming this sequence is 5, there are about 5 novel proteins which catalyse the necessary reactions. If the clone bank of bacteria containing the expression vectors which code for the novel genes has a number of distinct novel genes which is on the order of 1,000,000 all these expression vectors are isolated en masse and retransformed into 100 distinct sets of 10 8 bacteria at a ratio of vectors to bacteria which is sufficiently low that, on average, the number of bacteria in each set which are transformed is about half the number of initial genes, i.e. about 500,000.
  • the total number of new genes is only 500,000 rather than 1,000,000.
  • this procedure isolates the 5 critical novel genes.
  • mutagenesis and selection on this set of 5 stochastic genes allows improvement of the necessary catalytic functions.
  • the hydrolysis of a given stochastic peptide but at least one member of the set of stochastic peptides and proteins can be followed and measured by radioactive marking of the given protein followed by an incubation with a mixture of the stochastic proteins in the presence of ions such as Mg, Ca, Zn, Fe and ATP or GTP. The appearance of radioactive fragments of the marked protein is then measured as described.
  • the stochastic protein(s) which catalyse this reaction can again be isolated, along with the gene(s) producing them, by sequential diminution of the library of transformed clones, as described above.
  • An extension of the procedure consists in the selection of an ensemble of stochastic peptides and polypeptides capable of catalyzing a set of reactions leading from the initial building blocks (amino acids and small peptides) to some of the peptides or polypeptides of the set. It is therefore also possible to select an ensemble capable of catalyzing its own synthesis; such a reflexively autocatalytic set can be established in a chemostat where the products of the reactions are constantly diluted, but where the concentration of the building blocks is maintained constant. Alternatively, synthesis of such a set is aided by enclosing the complex set of peptides in liposomes by standard techniques.
  • the peptides and polypeptides which constitute an autocatalytic set may have certain elements in common with the large initial ensemble (constituted of coded peptides and polypeptides as given by our procedure) but can also contain peptides and polypeptides which are not coded by the ensemble of stochastic genes coding for the initial ensemble.
  • an autocatalytic set can contain coded peptides initially coded by the stochastic genes and synthesized continuously in the autocatalytic set. To isolate this coded subset of peptides and proteins, the autocatalytic set can be used to obtain, through immunization in an animal, polyclonal sera recognizing a very large number of the constituents of the autocatalytic set.
  • This set of stochastic genes expresses a large number of coded stochastic proteins which persist in the autocatalytic set.
  • the remainder of the coded constituents of such an autocatalytic set can be isolated by serial diminution of the library of stochastic genes, from which the subset detected by immunological methods has first been subtracted.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Virology (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Saccharide Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention is directed to a process for the production of a peptide, polypeptide, or protein having a predetermined property. In accordance with one embodiment, the process begins by producing by way of synthetic polynucleotide coupling, stochastically generated polynucleotide sequences. A library of expression vectors containing such stochastically generated polynucleotide sequences is formed. Next, host cells containing the vectors are cultured so as to produce peptides, polypeptides, or proteins encoded by the stochastically generated polynucleotide sequences. Screening or selection is carried out on such host cells to identify a peptide, polypeptide, or protein produced by the host cells which has the predetermined property. The stochastically generated polynucleotide sequence which encodes the identified peptide, polypeptide, or protein is then isolated and used to produce the peptide, polypeptide, or protein having the predetermined property.

Description

    CROSS REFERENCE TO RELATED APPLICATION
  • The present application is a continuation of U.S. patent application Ser. No. 942,630 filed Nov. 20, 1986, the entire disclosure of which is incorporated herein by reference.[0001]
  • The present invention has as its object a process for obtaining DNA, RNA, peptides, polypeptides, or proteins, through use of transformed host cells containing genes capable of expressing these RNAs, peptides, polypeptides, or proteins; that is to say, by utilization of recombinant DNA technique. [0002]
  • The invention aims in particular at the production of stochastic genes or fragments of stochastic genes in a fashion to permit obtaining simultaneously, after transcription and translation of these genes, a very large number (on the order of at least 10,000) of completely new proteins, in the presence of host cells (bacterial or eukaryotic) containing these genes respectively capable of expressing these proteins, and to carry out thereafter a selection or screen among the said clones, in order to determine which of them produce proteins with desired properties, for example, structural, enzymatic, catalytic, antigenic, pharmacologic, or properties of liganding, and more generally, chemical, biochemical, biological, etc. properties. [0003]
  • The invention also has as its aim procedures to obtain, sequences of DNA or RNA with utilizable properties notably chemical, biochemical, or biological properties. [0004]
  • It is clear, therefore, that the invention is open to a very large number of applications in very many areas of science, industry, and medicine. [0005]
  • The process for production of peptides or polypeptides according to the invention is characterized in that one produces simultaneously, in the same medium, genes which are at least partially composed of synthetic stochastic polynucleotides, that one introduces the genes thus obtained into host cells, that one cultivates simultaneously the independent clones of the transformed host cells containing these genes in such a manner so as to clone the stochastic genes and to obtain the production of the proteins expressed by each of these stochastic genes, that one carries out selection and/or screening of the clones of transformed host cells in a manner to identify those clones producing peptides or polypeptides having at least one desired activity, that one thereafter isolates the clones thus identified and that one cultivates them to produce at least one peptide or polypeptide having the said property. [0006]
  • In a first mode of carrying out this process, stochastic genes are produced by stochastic copolymerization of the four kinds of deoxyphosphonucleotides, A, C, G and T from the two ends of an initially linearized expression vector, followed by formation of cohesive ends in such a fashion as to form a stochastic first strand of DNA constituted by a molecule of expression vector possessing two stochastic sequences whose 3′ ends are complementary, followed by the synthesis of the second strand of the stochastic DNA. [0007]
  • In a second mode of carrying out this process, stochastic genes are produced by copolymerization of oligonucleotides without cohesive ends, in a manner to form fragments of stochastic DNA, followed by ligation of these fragments to a previously linearized expression vector. [0008]
  • The expression vector can be a plasmid, notably a bacterial plasmid. Excellent results have been obtained using the plasmid pUC8 as the expression vector. [0009]
  • The expression vector can also be viral DNA or a hybrid of plasmid and viral DNA. [0010]
  • The host cells can be prokaryotic cells such as HB 101 and C 600, or eukaryotic cells. [0011]
  • When utilizing the procedure according to the second mode mentioned above, it is possible to utilize oligonucleotides which for a group of palindromic octamers. [0012]
  • Particularly good results are obtained by utilizing the following group of palindromic octamers: [0013]
    5′ GGAATTCC 3′
    5′ GGTCGACC 3′
    5′ CAAGCTTG 3′
    5′ CCATATGG 3′
    5′ CATCGATG 3′
  • It is also possible to use oligonucleotides which form a group of palindromic heptamers. [0014]
  • Very good results are obtained utilizing the following group of palindromic heptamers: [0015]
    5′ XTCGCGA 3′
    5′ XCTGCAG 3′
    5′ RGGTACC 3′
  • where X=A, G, C, or T, and R=A or T [0016]
  • According to a method to utilize these procedures which is particularly advantageous, one isolates and purifies the transforming DNA of the plasmids from a culture of independent clones of the transformed host cells obtained by following the procedures above, then the purified DNA is cut by at least one restriction enzyme corresponding to specific enzymatic cutting site present in the palindromic octamers or heptamers but absent from the expression vector which was utilized; this cutting is followed by inactivation of the restriction enzyme, then one simultaneously treats the ensemble of linearized stochastic DNA fragments thus obtained with T4 DNA ligase, in such a manner to create a new ensemble of DNA containing new stochastic sequences, this new ensemble can therefore contain a number of stochastic genes larger than the number of genes in the initial ensemble. One then utilizes this new ensemble of transforming DNA to transform the host cells and clone these genes, and finally utilizes screening and/or selection and isolates the new clones of transformed host cells and finally these are cultivated to produce at least one peptide or polypeptide, for example, a new protein. [0017]
  • The property serving as the criterion for selection of the clones of host cells can be the capacity of the peptides or polypeptides, produced by a given clone, to catalyze a given chemical reaction. [0018]
  • For instance, for the production of several peptides and/or polypeptides, the said property can be the capacity to catalyze a sequence of reactions leading from an initial group of chemical compounds to at least one target compound. [0019]
  • With the aim of producing an ensemble constituted by a plurality of peptides and polypeptides which are reflexively autocatalytic, the said property can be the capacity to catalyze the synthesis of the same ensemble from amino acids and/or oligopeptides in an appropriate milieu. [0020]
  • The said property can also be the capacity to modify selectively the biological or chemical properties of a given compound, for example, the capacity to selectively modify the catalytic activity of a polypeptide. [0021]
  • The said property can also be the capacity to stimulate, inhibit, or modify at least one biological function of at least one biologically active compound, chosen, for example, among the hormones, neurotransmitters, adhesion factors, growth factors, and specific regulators of DNA replication and/or transcription and/or translation of RNA. [0022]
  • The said property can equally be the capacity of the peptide or polypeptide to bind to a given ligand. [0023]
  • The invention also has as its object the use of the peptide or polypeptide obtained by the process specified above, for the detection and/or the titration of a liquid. [0024]
  • According to a particularly advantageous mode of carrying out the invention, the criterion for selection of the clones of transformed host cells is the capacity of these peptides or polypeptides to simulate or modify the effects of a biologically active molecule, for example, a protein, and screening and/or selection for clones of transformed host cells producing at least one peptide or polypeptide having this property, is carried out by preparing antibodies against the active molecule, then utilizing these antibodies after their purification, to identify the clones containing this peptide or polypeptide, then by cultivating the clones thus identified, separating and purifying the peptide or polypeptide produced by these clones, and finally by submitting the peptide or polypeptide to an in vitro assay to verify that it has the capacity to simulate or modify the effects of the said molecule. [0025]
  • According to another mode of carrying out the process according to the invention, the property serving as the criterion of selection is that of having at least one epitope similar to one of the epitopes of a given antigen. [0026]
  • The invention carries over to obtaining polypeptides by the process specified above and utilizable as chemotherapeutically active substances. [0027]
  • In particular, in the case where the said antigen is EGF, the invention permits obtaining polypeptides usable for chemotherapeutic treatment of epitheliomas. [0028]
  • According to a variant of the procedure, one identifies and isolates the clones of transformed host cells producing peptides or polypeptides having the property desired, by affinity chromatography against antibodies corresponding to a protein expressed by the natural pan of the DNA hybrid. [0029]
  • For example, in the case where the natural part of the hybrid DNA contains a gene expressing B-galactosidase, one can advantageously identify and isolates the said clones of transformed host cells by affinity chromatography against anti-B-galactosidase antibodies. After expression and purification of hybrid peptides or polypeptides, one can separate and isolate their novel parts. [0030]
  • The invention also applies to a use of the process specified above for the preparation of a vaccine; the application is characterized by the fact that antibodies against the pathogenic agent are isolated, for example, antibodies formed after injection of the pathogenic agent in the body of an animal capable of forming antibodies against this agent, and these antibodies are used to identify the clones producing at least one protein having at least one epitope similar to one of the epitopes of the pathogenic agent, the transformed host cell corresponding to these clones are cultured to produce these proteins, this protein is isolated and purified from the clones of cells, then this protein is used for the production of a vaccine against the pathogenic agent. [0031]
  • For example in order to prepare an anti-HVB vaccine, one can extract and purify at least one capsid protein of the HVB virus, inject this protein into an animal capable of forming antibodies against this protein having at least one epitope similar to one of the epitopes of the HVB virus, then cultivate the clones of transformed host cells corresponding to these clones in a manner to produce this protein, isolate and purify the protein from culture of these clones of cells and utilize the protein for the production of an anti-HVB vaccine. [0032]
  • According to an advantageous mode of carrying out the process according to the invention, the host cells consist in bacteria such as [0033] Escherichia coli whose genome contains neither the natural gene expressing β-galactosidase, nor the EBG gene, that is to say, Z, EBG− E. coli. The transformed cells are cultured in the presence of X gal and the indicator IPTG in the medium, and cells positive for β-galactosidase functions are detected; thereafter, the transforming DNA is transplanted into an appropriate clone of host cells for large scale culture to produce at least one peptide or polypeptide.
  • The property serving as the criterion for selection of the transformed host cells can also be the capacity of the polypeptides or proteins produced by the culture of these clones to bind to a given compound. [0034]
  • This compound can be in particular chosen advantageously among peptides, polypeptides, and proteins, notably among proteins regulating the transcription activity of DNA. [0035]
  • On the other hand, the said compound can also be chosen among DNA and RNA sequences. [0036]
  • The invention has also as its object those proteins which are obtained in the case where the property serving as criterion of selection of the clones of transformed host cells consist in the capacity of these proteins to bind to regulatory proteins controlling transcription activity of the DNA, or else to DNA and RNA sequences. [0037]
  • The invention has, in addition, as an object, the use of a protein which is obtained in the first particular case above mentioned, as a cis-regulatory sequence controlling replication or transcription of a neighboring DNA sequence. [0038]
  • On the other hand, the aim of the invention also includes utilization of proteins obtained in the second case mentioned to modify the properties of transcription or replication of a sequence of DNA, in a cell containing the sequence of DNA, and expressing this protein. [0039]
  • The invention has as its object as well as a process of production of DNA, characterized by simultaneous production in the same medium, of genes at least partially composed of stochastic synthetic polynucleotides, in that the genes thus obtained are introduced into host cells to produce an ensemble of transformed host cells, in that screening and/or selection on this ensemble is carried out to identify those host cells containing in their genome stochastic sequences of DNA having at least one desired property, and finally, in that the DNA from the clones of host cells thus identified is isolated. [0040]
  • The invention further has as its object a procedure to produce RNA, characterized by simultaneous production in the same medium, of genes at least partially composed of stochastic synthetic polynucleotides, in that the genes thus obtained are introduced into host cells to produce an ensemble of transformed host cells, in that the host cells so produced are cultivated simultaneously, and screening and/or selection of this ensemble is carried out in a manner to identify those host cells containing stochastic sequences of RNA having at least one desired property, and in that the RNA is isolated from the host cells thus identified. The said property can be the capacity to bind a given compound, which might be, for example, a peptide or polypeptide or protein, or also the capacity to catalyze a given chemical reaction, or the capacity to be a transfer RNA. [0041]
  • Now the process according to the invention will be described in more details, as well as some of its applications, with reference to non-limitative embodiments. [0042]
  • First, we shall describe particularly useful procedures to carry out the synthesis of stochastic genes, and the introduction of those genes in bacteria to produce clones of transformed bacteria. [0043]
  • I) Direct Synthesis on an Expression Vector. [0044]
  • a) Linearization of the Vector [0045]
  • 30 micrograms, that is, approximately 10[0046] 13 molecules of the pUC8 expression vector are linearized by incubation for 2 hours at 37° C. with 100 units of the Pst1 restriction enzyme in a volume of 300 μl of the appropriate standard buffer. The linearized vector is treated with phenol-chloroform then precipitated in ethanol, taken up in volume of 30 μl and loaded onto a 0.8% agarose gel in standard TEB buffer. After migration in a field of 3V/cm for three hours, the linearized vector is electro-eluted, precipitated in ethanol, and taken up in 30 μl of water.
  • b) Stochastic Synthesis Using the Enzyme Terminal Transferase (TdT) [0047]
  • 30 ug of the linearized vector are reacted for 2 hours at 37° C. with 30 units of TdT in 300 μl of the appropriate buffer, in the presence of 1 mM dGTP, 1 mM dCTP, 0.3 mM dTTP and 1 mM dATP. The lower concentration of dTTP is chosen in order to reduce the frequence of “stop” codons in the corresponding messenger RNA. A similar result, although somewhat less favorable, can be obtained by utilizing a lower concentration for dATP than for the other deoxynucleotide triphosphates. The progress of the polymerization on the 3′ extremity of the Pst1 sites is followed by analysis on a gel of aliquots taken during the course of the reaction. [0048]
  • When the reaction attains or passes a mean value of 300 nucleotides added per 3′ extremity, it is stopped and the free nucleotides are separated from the polymer by differential precipitation or by passage over a column containing a molecular sieve such as Biogel P60. After concentration by precipitation in ethanol, the polymers are subjected to a further polymerization with TdT, first in the presence of dATP, then in the presence of dTTP. These last two reactions are separated by a filtration on a gel and are carried out for short intervals (30 seconds to 3 minutes) in order to add sequentially 10-30 A followed by 10-30 T to the 3′ ends of the polymers. [0049]
  • c) Synthesis of the Second Strand of Stochastic DNA [0050]
  • Each molecule of vector possess, at the end of the preceding operation, two stochastic sequences whose 3′ ends are complementary. The mixture of polymers is therefore incubated in conditions favoring hybridization of the complementary extremities (150 mM NaCl, 10 mM Tris-HCl, pH 7.6, 1 mM EDTA at 650 for 10 minutes, followed by lowering the temperature to 22° C. at a rate of 3 to 4° C. per hour). The hybridized polymers are then reacted with 60 units of the large fragment (Klenow) of polymerase 1, in the presence of the four nucleotide triphosphates (200 mM) at 4° C. for two hours. This step accomplishes the synthesis of the second strand from the 3′ ends of the hybrid polymers. The molecules which result from this direct synthesis starting from linearized vector are thereafter utilized transform competent cells. [0051]
  • d) Transformation of Competent Clones [0052]
  • 100 to 200 ml of competent HB 101 of C 600 at a concentration of 10[0053] 10 cells/ml, are incubated with the stochastic DNA preparation (from above) in the presence of 6 mM CaCl2, 6 mM Tris-HCl pHG, 6 mM MgCl2 for 30 minutes at 0° C. A temperature shock of 3 minutes at 37° C. is imposed on the mixture, followed by the addition of 400 to 800 ml of NZY culture medium, without antibiotics. The transformed culture is incubated at 37° C. for 60 minutes, then diluted to 10 litres by addition of NZY medium containing 40 μg/ml of ampicillin. After 3-5 hours of incubation at 37° C., the amplified culture is centrifuged, and the pellet of transformed cells is lyophilysed and stored at −70° C. Such a culture contains 3×107 to 108 independent transformants, each containing a unique stochastic gene inserted into the expression vector.
  • II) Synthetic of Stochastic Genes Starting from Oligonucleotides Without Cohesive Ends. [0054]
  • This procedure is based on the fact that polymerization of judiciously chosen palindromic oligonucleotides permits construction of stochastic genes which have no “stop” codon in any of the six possible reading frames, while at the same time assuring a balanced representation of triplets specifying all amino acids. Further, and to avoid a repetition of sequence motifs in the proteins which result, the oligonucleotides can contain a number of bases which is not a multiple of three. The example which follows describes the use of one of the possible combinations which fulfil these criteria: [0055]
  • a) Choice of a Group of Octamers [0056]
  • The group of oligonucleotides following: [0057]
    5′ GGAATTCC 3′
    5′ GGTCGACC 3′
    5′ CAAGCTTG 3′
    5′ CCATATGG 3′
    5′ CATCGATG 3′
  • is composed of 5 palindromes (thus self-complementary sequences) where it is easy to verify that their stochastic polymerization does not generate any “stop” codons, and specifies all the amino acids. [0058]
  • Obviously, one can utilize other groups of palindromic octamers which do not generate any “stop” codons and specify all the amino acids found in polypeptides. Clearly, it is also possible to utilize non-palindromic groups of octamers, or other oligomers, under the condition that their complements forming double stranded DNA are also used. [0059]
  • b) Assembly of a Stochastic Gene from a Group of Octamers. [0060]
  • A mixture containing 5 μg each of the oligonucleotides indicated above (previously phosphorylated at the 5′ position by a standard procedure) is reacted in a 100 ul volume containing 1 mM ATP, 10% polyethyleneglycol, and 100 units of T4 DNA ligase in the appropriate buffer at 13° C. for six hours. This step carries out the stochastic polymerization of the oligomers in the double stranded state and without cohesive ends. The resulting polymers are isolated by passage over a molecular sieve (Biogel P60) recovering those with 20 to 100 oligomers. After concentration, this fraction is again submitted to catalysis or polymerization by T4 DNA ligase under the conditions described above. Thereafter, as described above, those polymers which have assembled at least 100 oligomers are isolated. [0061]
  • c) Preparation of the Host Plasmid [0062]
  • The pUC8 expression vector is linearized by Sma1 enzyme in the appropriate buffer, as described above. The vector linearized by Sma1 does not have cohesive ends. Thus the linearized vector is treated by calf intestine alkaline phosphatase (CIP) at a level of one unit per microgram of vector in the appropriate buffer, at 37° C. for 30 minutes. The CIP enzyme is thereafter inactivated by two successive extractions with phenol-chloroform. The linearized and dephosphorylated vector is precipitated in ethanol, then redissolved in water at 1 mg/ml. [0063]
  • d) Ligation of Stochastic Genes to the Vector [0064]
  • Equimolar quantities of vector and polymers are mixed and incubated in the presence of 1000 units of T4 DNA ligase, 1 mM ATP, 10% polyethylene glycol, in the appropriate buffer, for 12 hours at 13° C. This step ligates the stochastic polymers in the expression vector and forms double stranded circular molecules which are, therefore, capable of transforming. [0065]
  • Transformation of Competent Clones. [0066]
  • Transformation of competent clones in carried out in the manner previously described. [0067]
  • III) Assembly of Stochastic Genes Starting from a Group of Heptamers. [0068]
  • This procedure differs from that just discussed in that it utilizes palindromic heptamers which have variable cohesive ends, in place of stochastic sequences containing a smaller number of identical motifs. [0069]
  • a) Choice of a Group of Heptamers [0070]
  • It is possible, as an example, to use the following three palindromic heptamers: [0071]
    5′ XTCGCGA 3′
    5′ XCTGCAG 3′
    5″ RGGTACC 3′
  • where X=A, G, C, or T and R=A or T, and where polymerization cannot generate any “stop” codons and forms triplets specifying all the amino acids. Clearly, it is possible to use other groups of heptamers fulfilling these same conditions. [0072]
  • b) Polymerization of a Group of Heptamers [0073]
  • This polymerization is carried out exactly in the fashion described above for octamers. [0074]
  • c) Elimination of Cohesive Extremities [0075]
  • The polymers thus obtained have one unpaired base on their two 5′ extremities. Thus, it is necessary to add the complementary base to the corresponding 3′ extremities. This is carried out as follows: 10 micrograms of the double stranded polymers are reacted with 10 units of the Klenow enzyme, in the presence of the four deoxynucleotidephosphates (200 mM) in a volume of 100 μl, at 4° C., for 60 minutes. The enzyme is inactivated by phenol chloroform extraction, and the polymers are cleansed of the residual free nucleotides by differential precipitation. The polymers are then ligated to the host plasmid (previously linearized and dephosphorylated) by following the procedures described above. [0076]
  • It is to be noted that the two last procedures which were described utilize palindromic octamers or heptamers which constitute specific sites of certain restriction enzymes. These sites are absent, for the most part, from the pUC8 expression vector. Thus, it is possible to augment considerably the complexity of an initial preparation of stochastic genes by proceeding in the following way: the plasmid DNA derived from the culture of 10[0077] 7 independent transformants obtained by one of the two last procedures described above, is isolated. After this DNA is purified, it is partially digested by Cla1 restriction enzyme (procedure II) or by the Pst1 restriction enzyme (procedure III). After inactivation of the enzyme, partially digested DNA is treated with T4 DNA ligase, which has the effect of creating a very large number of new sequences, while conserving the fundamental properties of the initial sequences. This new ensemble of stochastic sequences can then be used to transform competent cells. In addition, the stochastic genes cloned by procedure II and III can be excised intact from the pUC8 expression vector by utilizing restriction sites belonging to the cloning vector and not represented in the stochastic DNA sequences.
  • Recombination within the stochastic genes generated by the two procedures just described, which result from the internal homology due to the recurrent molecular motifs, is an important additional method to achieve in vivo mutagenesis of the coding sequences. This results in an augmentation of the number of the new genes which can be examined. [0078]
  • Finally, for all the procedures to generate novel synthetic genes, it is possible to use a number of common techniques to modify genes in vivo or in vitro, such as a change of reading frame, inversion of sequences with respect to their promotor, point mutations, or utilization of host cells expressing one or several suppressor tRNAs. [0079]
  • In considering the above description, it is clear that it is possible to construct, in vitro, an extremely large number (for example, greater than a billion) different genes, by enzymatic polymerization of nucleotides or of oligonucleotides. This polymerization is carried out in a stochastic manner, as determined by the respective concentrations of the nucleotides or oligonucleotides present in the reaction mixture. [0080]
  • As indicated above, two methods can be utilized to clone such genes (or coding sequences): the polymerization can be carried out directly on a cloning expression vector, which was previously linearized; or it is possible to proceed sequentially to the polymerization then the ligation of the polymers to the expression vector. [0081]
  • In the two cases, the next step is transformation or transfection of competent bacterial cells (or cells in culture). This step constitutes cloning the stochastic genes in living cells where they are indefinitely propagated and expressed. [0082]
  • Clearly, in addition to the procedures which were described above, it is feasible to use all other methods which are appropriate for the synthesis of stochastic sequences. In particular, it is possible to carry out polymerization, by biochemical means, of single stranded oligomers of DNA or RNA obtained by chemical synthesis, then treat these segments of DNA or RNA by established procedures to generate double stranded DNA (cDNA) in order to clone such genes. [0083]
  • Screening or Selection of Clones of Transformed Host Cells [0084]
  • The further step of the procedure according to the invention consists in examining the transformed or transfected cells by selection or screening, in order to isolate one or several cells whose transforming or transfecting DNA leads to the synthesis of a transcription product (RNA) or translation product (protein) having desired property. These properties can be, for example, enzymatic, functional, or structural. [0085]
  • One of the most important aspects of the process, according to the invention, is that it permits the simultaneous screening or selection of an exploitable product (RNA or protein) and the gene which produces that product. In addition, the DNA synthesized and cloned as described, can be selected or screened in order to isolate sequences of DNA constituting products in themselves, having exploitable biochemical properties. [0086]
  • We shall now describe, as non-limiting examples, preferred procedures for screening and/or selection of clones of transformed cells such that the novel proteins are of interest from the point of view of industrial or medical applications. [0087]
  • One of these procedures rests in the idea of producing, or obtaining, polyclonal or monoclonal antibodies, by established techniques, directed against a protein or another type of molecule of biochemical or medical interest, where that molecule is, or has been rendered, immunogenic, and thereafter using these antibodies as probes to identify among the very large number of clones transformed by stochastic genes, those whose protein react with these antibodies. This reaction is a result of a structural homology which exists between the polypeptide synthesized by the stochastic gene and the initial molecule. It is possible in this way to isolate numbers of novel proteins which behave as epitopes or antigenic determinants on the initial molecule. Such novel proteins are liable to simulate, stimulate, modulate, or block the effect of the initial molecule. It will be clear that this means of selection or screening may itself have very many pharmacologic and biochemical applications. Below we describe, as a non-limiting example, this first mode of operation in a concrete case: [0088]
  • EGF (epidermal growth factor) is a small protein present in the blood, whose role is to stimulate the growth of epithelial cells. This effect is obtained by the interaction of EGF with a specific receptor situated in the membrane of epithelial cells. Antibodies directed against EGF are prepared by injecting animals with EGF coupled to KLH (keyhole limpet hemocyanin) to augment the immunogenicity of the EGF. The anti-EGF antibodies of the immunized animals are purified, for example, by passage over an affinity column, where the ligand is EGF or a synthetic peptide corresponding to a fragment of EGF. The purified anti-EGF antibodies are then used as probes to screen a large number of bacterial clones lysed by chloroform, and on a solid support. The anti-EGF antibodies bind those stochastic peptides or proteins whose epitopes resemble those of the initial antigen. The clones containing such peptides or proteins are shown by autoradiography after incubation of the solid support with radioactive protein A, or after incubation with a radioactive antibody. [0089]
  • These steps identify those clones, each of which contains one protein (and its gene) reacting with the screening antibody. It is feasible to screen among a very large number of colonies of bacterial cells or viral plaques (for example, on the order of 1,000,000) and it is feasible to detect extremely small quantities, on the order of 1 nanogram, of protein product. Thereafter, the identified clones are cultured and the proteins so detected are purified in conventional ways. These proteins are tested in vitro in cultures of epithelial cells to determine if they inhibit, simulate, or modulate the effects of EGF on these cultures. Among the proteins so obtained, some may be utilized for the chemotherapeutic treatment of epitheliomas. The activities of the proteins thus obtained can be improved by mutation of the DNA coding for the proteins, in ways analogous to those described above. A variant of this procedure consists in purifying these stochastic peptides, polypeptides, or proteins, which can be used as vaccines or more generally, to confer an immunity against a pathogenic agent or to exercise other effects on the immunological system, for example, to create a tolerance or diminish hypersensitivity with respect to a given antigen, in particular, due to binding of these peptides, polypeptides, or proteins with the antibodies directed against this antigen. It is clear that it is possible to use such peptides, polypeptides, or proteins in vitro as well as in vivo. [0090]
  • More precisely, in the ensemble of novel proteins which react with the antibodies against a given antigen X, each has at least one epitope in common with X, thus the ensemble has an ensemble of epitopes in common with X. This permits utilization of the ensemble or sub-ensemble as a vaccine to confer immunity against X. It is, for example, easy to purify one or several of the capsid proteins of the hepatitis B virus. These proteins can then be injected into an animal, for example, a rabbit, and the antibodies corresponding to the initial antigen can be recovered by affinity column purification. These antibodies may be used, as described above, to identify clones producing at least one protein having an epitope resembling at least one of the epitopes of the initial antigen. After purification, these proteins are used as antigens (either alone or in combination) with the aim of conferring protection against hepatitis B. The final production of the vaccine does not require further access to the initial pathogenic agent. [0091]
  • Note that, during the description of the procedures above, a number of means to achieve selection or screening have been described. All these procedures may require the purification of a particular protein from a transformed clone. These protein purifications can be carried out by established procedures and utilize, in particular, the techniques of gel chromatography, by ion exchange, and by affinity chromatography. In addition, the proteins generated by the stochastic genes can be cloned in the form of hybrid proteins having, for example, a sequence of the β-galactosidase enzyme which permits affinity chromatography against anti-β-galactosidase antibodies, and allows the subsequent cleavage of the hybrid part; that is to say, allowing separation of the novel part and the bacterial part of the hybrid protein. Below we describe the principles and procedures for selection of peptides or polypeptides and the corresponding genes, according to a second method of screening or selection based on the detection of the capacity of these peptides or polypeptides to catalyse a specific reaction. [0092]
  • As a concrete and non-limiting example, screening or selection in the particular case of proteins capable of catalyzing the cleavage of lactose, normally a function fulfilled by enzyme β-galactosidase (β-gal) will be described. [0093]
  • As above described, the first step of the process consists in generating a very large ensemble of expression vectors, each expressing a distinct novel protein. To be concrete, for example, one may choose the pUC8 expression vector with cloning of stochastic sequences of DNA in the Pst1 restriction site. The plasmids thus obtained are then introduced into a clone of [0094] E. coli from whose genome the natural gene for β-galactosidase, Z, and a second gene EBG, unrelated to the first but able to mutate towards β-gal function, have both been eliminated by known genetic methods. Such host cells (Z, EBG) are not able by themselves to catalyse lactose hydrolysis, and as a consequence, to use lactose as a carbon source for growth. This permits utilization of such host clones for screening or selection for β-gal function.
  • A convenient biological assay to analyze transformed [0095] E. coli clones for those which have novel genes expressing a β-gal function consists in the culture of bacteria transformed as described in petri dishes containing X-gal in the medium. In this case, all bacterial colonies expressing a β-gal function are visualized as blue colonies. By using such a biological assay, it is possible to detect even weak catalytic activity. The specific activity of characteristic enzymes ranges from 10 to 10,000 product molecules per second.
  • Supposing that a protein synthesized by a stochastic gene has a weak specific activity, on the order of one molecule per 100 seconds, it remains possible to detect such catalytic activity. In a petri dish containing X-gal in the medium, and the presence of the non-metabolizable inducer IPTG (isopropyl-D-thiogalactoside) visualization of a blue region requires cleavage of about 10[0096] 10 to 1011 molecules of X-gal per square millimeter. A bacterial colony expressing a weak enzyme and occupying a surface area of 1 mm2 has about 107 to 108 cells. If each cell has only one copy of the weak enzyme, each cell would need to catalyse cleavage of between 10,000 and 100 of X-gal to be detected, which would require between 2.7 and 270 hours. Since under selective conditions it is possible to amplify the number of copies of each plasmid per cell from 5 to 20 copies per cell, or even to 100 or 1000, and because up to 10% of the protein of the cell can be specified by the new gene, the duration needed to detect a blue colony in the case of 100 enzyme molecules of weak activity per cell is on the order of 0.27 to 2.7 hours.
  • As a consequence of these facts, screening a very large number of independent bacterial colonies, each expressing a different novel gene, and using the capacity to express a β-gal function as the selection criterion, is fully feasible. It is possible to carry out screening of about 2000 colonies in one Petri dish of 10 cm diameter. Thus, about 20 million colonies can be screened on a sheet of X-gal agar of 1 square meter. [0097]
  • It is to be noted that bacterial colonies which appear blue on X-gal Petri dishes might be false positives due to a mutation in the bacterial genome which confers upon it the capacity to metabolize lactose, or for other reasons than those which result from a catalytic activity of the novel protein expressed by the cells of the colony. Such false positives can be directly eliminated by purifying the DNA of the expression vector from the positive colony, and retransforming Z[0098] , EBG E. coli host cells. If the β-gal activity is due to the novel protein coded by the new gene in the expression vector, all those cells transformed by that vector will exhibit β-gal function. In contrast, if the initial blue colony is due to a mutation in the genome of the host cell, it is a rare event and independent of the transformation, thus the number of cells of the new clone of the transformed E. coli capable of expressing gal function will be small or zero.
  • The power of mass simultaneous purification of all the expression vectors from all the positive clones (blue) followed by retransformation of naive bacteria should be stressed. Suppose that the aim is to carry out a screening to select proteins having a catalytic function, and that the probability that a new peptide or polypeptide carries out this function at least weakly is 10[0099] −6, while the probability that a clone of the E. coli bacterial host undergoes a mutation rendering it capable of carrying out the same function is 10−5, then it can be calculated that among 20 million transformed bacteria which are screened, 20 positive clones will be attributable to the novel genes in expression vectors which each carries, while 200 positive clones will be the result of genomic mutation. Mass purification of the expression vectors from the total of 220 positive bacterial clones followed by retransformation of the naive bacteria with the mixture of these expression vectors will produce a large number of positive clones consisting of all those bacteria transformed with the 20 expression vectors which code for the novel proteins having the desired function, and a very small number of bacterial clones resulting from genomic mutations and containing the 200 expression vectors which are not of interest. A small number of cycles of purification of expression vectors from positive bacterial colonies, followed by such retransformation, allows the detection of very rare expression vectors truly positive for a desired catalytic activity, despite a high background rate of mutations in the host cells for the same function.
  • Following screening operations of this type, it is possible to purify the new protein by established techniques. The production of that protein in large quantity is made possible by the fact that identification of the useful protein occurs together with simultaneous identification of the gene coding for the same protein. Consequently, either the same expression vector can be used, or the novel gene can be transplanted into a more appropriated expression vector for its synthesis and isolation in large quantity. [0100]
  • It is feasible to apply this method of screening for any enzymatic function for which an appropriate biological assay exists. For such screenings, it is not necessary that the enzymatic function which is sought be useful to the host cell. It is possible to carry out screenings not only for an enzymatic function but for any other desired property for which it is possible to establish an appropriate biological assay. It is thus feasible to carry out, even in the simple case of β-gal function visualized on an X-gal Petri plate, a screening of on the order of 100 million, or even a billion novel genes for a catalytic activity or any other desired property. [0101]
  • Selection of Transformed Host Cells. [0102]
  • On the other hand, it is possible to use selection techniques for any property, catalytic or otherwise, where the presence or absence of the property can be rendered essential for the survival of the host cells containing the expression vectors which code for the novel genes, or also can be used to select for those viruses coding and expressing the desired novel gene. As a non-limiting, but concrete example, selection for β-galactosidase function shall be described. An appropriate clone of Z[0103] EBG E. coli is not able to grow on lactose as the sole carbon source. Thus, after carrying out the first step described above, it is possible to culture a very large number of host cells transformed by the expression vectors coding for the novel genes, under selective conditions, either by progressive diminution of other sources of carbon, or utilization of lactose alone from the start. During the course of such selection, in vivo mutagenesis by recombination, or by explicitly recovering the expression vectors and mutagenizing their novel genes in vitro by various mutagens, or by any other common technique, permits adaptive improvements in the capacity to fulfill the desired catalytic function. When both selection techniques and convenient bioassay techniques exist at the same time, as in the present case, it is possible to use selection techniques initially to enrich the representation of host bacteria expressing the β-gal function, then carry out a screening on a Petri plate on X-gal medium to establish efficiently which are the positive cells. In the absence of convenient bioassays, application of progressively stricter selection is the easiest route to purify one or a small number of distinct host cells whose expression vectors code for the proteins catalyzing the desired reaction.
  • It is possible to utilize these techniques to find novel proteins having a large variety of structural and functional characteristics beyond the capacity to catalyse a specific reaction. For example, it is possible to carry out a screen or select for novel proteins which bind to cis-regulatory sites on the DNA and thereby block the expression of one of the host cell's functions, or block transcription of the DNA, stimulate transcription, etc. [0104]
  • For example, in the case of [0105] E. coli, a clone mutant in the repressor of the lactose operon (i−) expresses β-gal function constitutively due to the fact the lactose operator is not repressed. All cells of this type produce blue clones on Petri plates containing X-gal medium. It is possible to transform such host cells with expression vectors synthesizing novel proteins and carry out a screen on X-gal Petri plates in order to detect those clones which are not blue. Among those, some represent the case where the new protein binds to the lactose operator and represses the synthesis of β-gal. It is then feasible to mass isolate such plasmids, retransform, isolate those clones which do not produce β-gal, and thereafter carry out a detailed verification.
  • As mentioned above, the process can be utilized in order to create, then isolate, not only exploitable proteins, but also RNA and DNA as products in themselves, having exploitable properties. This results from the fact that, on one hand, the procedure consists in creating stochastic sequences of DNA which may interact directly with other cellular or biochemical constituents, and on the other hand, these sequences cloned in expression vectors are transcribed into RNA which are themselves capable of multiple biochemical interactions. [0106]
  • An Example of the Use of the Procedure to Create and Select for a DNA Which is Useful in Itself. [0107]
  • This example illustrates selection for a useful DNA, and the purification and study of the mechanism of action of regulatory proteins which bind to the DNA. Consider a preparation of the oestradiol receptor, a protein obtained by standard techniques. In the presence of oestradiol, a steroid sexual hormone, the receptor changes conformation and binds tightly to certain specific sequences in the genomic DNA, thus affecting the transcription of genes implicated in sexual differentiation and the control of fertility. By incubating a mixture containing oestradiol, its receptor, and a large number of different stochastic DNA sequences inserted in their vectors, followed by filtration of the mixture across a nitrocellulose membrane, one has a direct selection for those stochastic DNA sequences binding to the oestrodiol-receptor complex, where only those DNAs bound to a protein are retained by the membrane. After washing and elution, the DNA liberated from the membrane is utilized as such to transform bacteria. After culture of the transformed bacteria, the vectors which they contain are again purified and one or several cycles of incubation, filtration, and transformation are carried out as described above. These procedures allow the isolation of stochastic sequences of DNA having an elevated affinity for the oestradiol-receptor complex. Such sequences are open to numerous diagnostic and pharmacologic applications, in particular, for developing synthetic oestrogens for the control of fertility and treatment of sterility. [0108]
  • Creation and Selection of an RNA Useful in Itself [0109]
  • Let there be a large number of stochastic DNA sequences, produced as has been described and cloned in an expression vector. It follows that the RNA transcribed from these sequences in the transformed host cells can be useful products themselves. As a non-limiting example, it is possible to select a stochastic gene coding for a suppressor transfer RNA (tRNA) by the following procedure: a large number (≧10[0110] 8) of stochastic sequences are transformed into competent bacterial hosts carrying a “nonsense” mutation in the arg E. gene. These transformed bacteria are plated on minimal medium without arginine and with the selective antibiotic for that plasmid (ampicillin if the vector is pUC8). Only those transformed bacteria which have become capable of synthesizing arginine will be able to grow. This phenotype can result either from a back mutation of the host genome, or the presence in the cell of a suppressor. It is easy to test each transformed colony to determine if the arg+ phenotype is or is not due to the presence of the stochastic gene in its vector; it suffices to purify the plasmid from this colony and verify that it confers an arg+ phenotype on all arg E. cells transformed by it.
  • Selection of Proteins Capable of Catalyzing a Sequence of Reactions [0111]
  • Below we describe another means of selection, open to independent applications, based on the principle of simultaneous and parallel selection of a certain number of novel proteins capable of catalyzing a connected sequence of reactions. [0112]
  • The basic idea of this method is the following: given an initial ensemble of chemical compounds considered as building blocks or elements of construction from which it is hoped to synthesize one or several desired chemical compounds by means of a catalyzed sequence of chemical reactions, there exists a very large number of reaction routes which can be partially or completely substituted for one another, which are all thermodynamically possible, and which lead from the set of building blocks to the desired target compound(s). Efficient synthesis of a target compound is favored if each step of at least one reaction pathway leading from the building block compounds to the target compound is comprised of reactions each of which is catalyzed. On the other hand, it is relatively less important to determine which among the many independent or partially independent reaction pathways is catalyzed. In the previous description, we have shown how it is possible to obtain a very large number of host cells each of which expresses a distinct novel protein. [0113]
  • Each of these novel proteins is a candidate to catalyse one or another of the possible reactions, in the set of all the possible reactions leading from the ensemble of building blocks to the target compound. If a sufficiently large number of stochastic proteins is present in a reaction mixture containing the building block compounds, such that a sufficiently large number of the possible reactions are catalyzed, there is a high probability that one connected sequence of reactions leading from the set of building block compounds to the target compound will be catalyzed by a subset of the novel proteins. It is clear that this procedure can be extended to the catalysis not only of one, but of several target compounds simultaneously. [0114]
  • Based on this principle it is possible to proceed as follows in order to select in parallel a set of novel proteins catalyzing a desired sequence of chemical reactions: [0115]
  • 1. Specify the desired set of compounds constituting the building blocks, utilizing preferentially a reasonably large number of distinct chemical species in order to increase the number of potential concurrent reactions leading to the desired target compound. [0116]
  • 2. Using an appropriate volume of reaction medium, add a very large number of novel stochastic proteins isolated from transformed or transfected cells synthesizing these proteins. Carry out an assay to determine if the target compound is formed. If it is, confirm that this formation requires the presence of the mixture of novel proteins. If so, then the mixture should contain a subset of proteins catalyzing one or several reaction pathways leading from the building block set to the target compound. Purify and divide the initial ensemble of clones which synthesize the set of novel stochastic proteins, the subset which is required to catalyse the sequence of reactions leading to the target compound. [0117]
  • More precisely, as a non-limiting example, below we describe selection of novel proteins capable of catalyzing the synthesis of a specific small peptide, in particular, a pentapeptide, starting from a building block set constituted of smaller peptides and amino acids. All peptides are constituted by a linear sequence of 20 different types of amino acids, oriented from the amino to the carboxy terminus. Any peptide can be formed in a single step by the terminal condensation of two smaller peptides (or of two amino acids), or by hydrolysis of a larger peptide. A peptide with M residues can thus be formed by M−1 condensation reactions. The number of reactions, R, by which a set of peptides having length 1, 2, 3, . . . M residues can be interconverted is larger than the number of possible molecular species, T. This can be expressed as R/T=M−2. Thus, starting from a given ensemble of peptides, a very large number of independent or partially independent reaction pathways leads to the synthesis of a specific target peptide. Choose a pentapeptide whose presence can be determined conveniently by some common assay technique for example HPLC (liquid phase high pressure chromatography), paper chromatography, etc. Formation of a peptide bond requires energy in a dilute aqueous medium, but if the peptides participating in the condensation reactions are adequately concentrated, formation of peptide bonds is thermodynamically favored over hydrolysis and occurs efficiently in the presence of an appropriate enzymatic catalyst, for example pepsin or trypsin, without requiring the presence of ATP or other high energy compounds. Such a reaction mixture of small peptides whose amino acids are marked radioactively to act as tracers with [0118] 3H, 14C, 35S, constituting the building block set can be used at sufficiently high concentrations to lead to condensation reactions.
  • For example, it is feasible to proceed as follows: 15 mg of each amino acid and small peptides having 2 to 4 amino acids, chosen to constitute the building block set, are dissolved in a volume of 0.25 ml to 11.0 ml of a 0.1M pH 7.6 phosphate buffer. A large number of novel proteins, generated and isolated as described above are purified from their bacterial or other host cells. The mixture of these novel proteins is dissolved to a final concentration on the order of 0.8 to 1.0 mg/ml in the same buffer. 0.25 ml to 0.5 ml of the protein mixture is added to the mixture of building blocks. This is incubated at 25° C. to 40° C. for 1 to 40 hours. Aliquots of 8 μl are removed at regular intervals, the first is used as a “blank” and taken before addition of the mixture of novel proteins. These aliquots are analyzed by chromatography using n-butanol-acetic acid-pyridine-water (930:6:20:24 by volume) as the solvent. The chromatogram is dried and analyzed by ninhydrin or autoradiography (with or without intensifying screens). Because the compound constituting the building block set are radioactively marked, the target compound will be radioactive and it will have specific activity high enough to permit detection at the level of 1-10 ng. In place of standard chromatographic analysis, it is possible to use HPLC (high pressure liquid chromatography) which is faster and simpler to carry out. More generally, all the usual analytic procedures can be employed. Consequently it is possible to detect a yield of the target compound of less than one part per million by weight compared to the compounds used as initial building blocks. [0119]
  • If the pentapeptide is formed in the conditions described above, but not when an extract is utilized which is derived from host cells transformed by an expression vector containing no stochastic genes, the formation of the pentapeptide is not the result of bacterial contaminants and thus requires the presence of a subset of the novel proteins in the reaction mixture. [0120]
  • The following step consists in the separation of the particular subset of cells which contain expression vectors with the novel proteins catalyzing the sequence of reactions leading to the target pentapeptide. As an example, if the number of reactions forming this sequence is 5, there are about 5 novel proteins which catalyse the necessary reactions. If the clone bank of bacteria containing the expression vectors which code for the novel genes has a number of distinct novel genes which is on the order of 1,000,000 all these expression vectors are isolated en masse and retransformed into 100 distinct sets of 10[0121] 8 bacteria at a ratio of vectors to bacteria which is sufficiently low that, on average, the number of bacteria in each set which are transformed is about half the number of initial genes, i.e. about 500,000. Thus, the probability that any given one of the 100 sets of bacteria contains the entire set of 5 critical novel proteins is (½)5={fraction (1/32)}. Among the 100 initial sets of bacteria, about 3 will contain the 5 critical transformants. In each of these sets, the total number of new genes is only 500,000 rather than 1,000,000. By successive repetitions, the total number of which is about 20 in the present case, this procedure isolates the 5 critical novel genes. Following this, mutagenesis and selection on this set of 5 stochastic genes allows improvement of the necessary catalytic functions. In a case where it is necessary to catalyse a sequence of 20 reactions and 20 genes coding novel proteins need to be isolated in parallel, it suffices to adjust the multiplicity of transformation such that each set of 108 bacteria receives 80% of the 106 stochastic genes, and to use 200 such sets of bacteria. The probability that all 20 novel proteins are found in one set is 0.820≅0.015. Thus, about 2 among the 200 sets will have the 20 novel genes which are needed to catalyse the formation of the target compound. The number of cycles required for isolation of the 20 novel genes is on the order of 30.
  • The principles and procedures described above generalize from the case of peptides to numerous areas of chemistry in which chemical reactions take place in aqueous medium, in temperature, pH, and concentration conditions which permit general enzymatic function. In each case it is necessary to make use of an assay method to detect the formation of the desired target compound(s). It is also necessary to choose a sufficiently large number of building block compounds to augment the number of reaction sequences which lead to the target compound. [0122]
  • The concrete example which was given for the synthesis of a target pentapeptide can also be generalized as follows: [0123]
  • The procedure as described, generates among other products, stochastic peptides and proteins. These peptides or proteins can act, catalytically or in other ways, on other compounds. They can equally constitute the substrates on which they act. Thus, it is possible to select (or screen) for the capacity of such stochastic peptides or proteins to interact among themselves and thereby modify the conformation, the structure or the function of some among them. Similarly, it is possible to select (or screen) for the capacity of these peptides and proteins to catalyse among themselves, hydrolysis, condensation, transpeptidation or other reactions modifying the peptides. For example, the hydrolysis of a given stochastic peptide but at least one member of the set of stochastic peptides and proteins can be followed and measured by radioactive marking of the given protein followed by an incubation with a mixture of the stochastic proteins in the presence of ions such as Mg, Ca, Zn, Fe and ATP or GTP. The appearance of radioactive fragments of the marked protein is then measured as described. The stochastic protein(s) which catalyse this reaction can again be isolated, along with the gene(s) producing them, by sequential diminution of the library of transformed clones, as described above. [0124]
  • An extension of the procedure consists in the selection of an ensemble of stochastic peptides and polypeptides capable of catalyzing a set of reactions leading from the initial building blocks (amino acids and small peptides) to some of the peptides or polypeptides of the set. It is therefore also possible to select an ensemble capable of catalyzing its own synthesis; such a reflexively autocatalytic set can be established in a chemostat where the products of the reactions are constantly diluted, but where the concentration of the building blocks is maintained constant. Alternatively, synthesis of such a set is aided by enclosing the complex set of peptides in liposomes by standard techniques. In a hypertonic aqueous environment surrounding such lipsomes, condensation reactions forming larger peptides lowers the osmotic pressure inside the lipsomes, drives water molecules produced by the condensation reactions out of the lipsomes, hence favoring synthesis of larger polymers. Existence of such an autocatalytic ensemble can be verified by two dimensional gel electrophoresis and by HPLC, showing the synthesis of a stable distribution of peptides and polypeptides. The appropriate reaction volume depends on the number of molecular species used, and the concentrations necessary to favor the formation of peptide bonds over their hydrolysis. The distribution of molecular species of an autocatalytic ensemble is free to vary or change due to the emergence of variant autocatalytic ensembles. The peptides and polypeptides which constitute an autocatalytic set may have certain elements in common with the large initial ensemble (constituted of coded peptides and polypeptides as given by our procedure) but can also contain peptides and polypeptides which are not coded by the ensemble of stochastic genes coding for the initial ensemble. [0125]
  • The set of stochastic genes whose products are necessary to establish such an autocatalytic set can be isolated as has been described, by sequential diminution of the library of transformed clones. In addition, an autocatalytic set can contain coded peptides initially coded by the stochastic genes and synthesized continuously in the autocatalytic set. To isolate this coded subset of peptides and proteins, the autocatalytic set can be used to obtain, through immunization in an animal, polyclonal sera recognizing a very large number of the constituents of the autocatalytic set. [0126]
  • These sera can be utilized to screen the library of stochastic genes to find those genes expressing proteins able to combine with the antibodies present in the sera. [0127]
  • This set of stochastic genes expresses a large number of coded stochastic proteins which persist in the autocatalytic set. The remainder of the coded constituents of such an autocatalytic set can be isolated by serial diminution of the library of stochastic genes, from which the subset detected by immunological methods has first been subtracted. [0128]
  • Such autocatalytic sets of peptides and proteins, obtained as noted, may find a number of practical applications. [0129]

Claims (66)

We claim:
1. A process for the production of a peptide, polypeptide, or protein having a predetermined property, comprising the steps of:
producing by synthetic polynucleotide coupling, stochastically generated polynucleotide sequences;
forming a library of expression vectors containing such stochastically generated polynucleotide sequences;
culturing host cells containing the vectors to produce peptides, polypeptides, or proteins encoded by the stochastically generated polynucleotide sequences;
carrying out screening or selection on such host cells, to identify a peptide, polypeptide, or protein produced by the host cells having the predetermined property;
isolating a stochastically generated polynucleotide sequence which encodes the identified peptide, polypeptide, or protein;
using the isolated sequence to produce the peptide, polypeptide, or protein having the predetermined property.
2. A process for the production of a peptide, polypeptide, or protein having a predetermined property, comprising the steps of:
producing at least partially stochastic synthetic polynucleotide sequences,
introducing the at least partially stochastic polynucleotide sequences thus obtained into host cells,
cultivating the transformed host cells containing these at least partially stochastic polynucleotide sequences so as to clone the stochastic polynucleotide sequences and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these stochastic polynucleotide sequences,
carrying out screening and/or selection methods on such clones of transformed host cells to identify those clones producing the peptide, polypeptide, or protein having the predetermined property,
isolating the clones so identified, and
growing the isolated clones in a manner so as to produce the peptide, polypeptide, or protein having the predetermined property.
3. The process according to claim 2 wherein the at least partially stochastic polynucleotide sequences are introduced into host cells by an expression vector.
4. A process for the production of a peptide, polypeptide, or protein having a predetermined property by microbiological means, comprising the steps of:
simultaneously producing in a common milieu expression vectors which include at least one stochastic sequence of polynucleotides,
introducing the expression vectors thus obtained into host cells,
cultivating the host cells containing these expression vectors so as to clone the stochastic sequences and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these stochastic sequences,
carrying out screening and/or selection methods on such clones to identify those clones producing the peptide, polypeptide, or protein having the predetermined property,
isolating the clones so identified, and
growing the isolated clones in a manner so as to produce the peptide, polypeptide, or protein having the predetermined property.
5. A process for the production of a peptide, polypeptide, or protein having a predetermined property by microbiological means, comprising the steps of:
simultaneously producing in a common milieu expression vectors which include at least one stochastic sequence of polynucleotides,
introducing the expression vectors thus obtained into host cells,
cultivating the host cells containing these expression vectors so as to clone the expression vector and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these expression vectors,
carrying out screening and/or selection methods on such clones producing the peptide, polypeptide, or protein having the predetermined property,
isolating the clones so identified, and
growing the isolated clones in a manner so as to produce the peptide, polypeptide, or protein having the predetermined property.
6. A process for the production of a peptide, polypeptide, or protein having a predetermined property, comprising the steps of:
producing a library of at least partially stochastic synthetic polynucleotide sequences,
amplifying and translating these at least partially stochastic polynucleotide sequences to produce peptides, polypeptides, or proteins expressed by at least some of these stochastic polynucleotide sequences, and
carrying out screening and/or selection methods to obtain the peptide, polypeptide, or protein having at least the predetermined property.
7. The process according to claim 6 wherein the at least partially stochastic polynucleotide sequences undergo extracellular amplification.
8. The process according to claim 1 wherein polynucleotide sequences are produced by stochastic copolymerization of desired ratios of the deoxyphosphonucleotides guanine, cytosine, thymidine, and adenine, starting from the two extremities of an expression vector which was previously linearized, then by formation of cohesive extremities to create a first strand of stochastic DNA constituting a molecule of expression vector possessing two stochastic sequences whose 3′ extremities are complementary, followed by synthesis of the second strand of the stochastic DNA.
9. The process according to claim 1 wherein the polynucleotide sequences are produced by stochastic copolymerization of double stranded oligonucleotides which do not have cohesive ends, in a manner so as to form fragments of stochastic DNA, followed by ligation of these fragments in an expression vector which was previously linearized.
10. The process according to claim 1 wherein the host cells are prokaryotic cells.
11. The process according to claim 10 wherein the prokaryotic cells are HB 101 or C 600.
12. The process according to claim 1 wherein the host cells are eukaryotic cells.
13. The process according to claim 1 wherein the expression vector is a plasmid.
14. The process according to claim 13 wherein the plasmid is pUC8.
15. The process according to claim 1 wherein the expression vector is viral DNA.
16. The process according to claim 1 wherein the expression vector is a hybrid of plasmid and viral DNA.
17. The process according to claim 1 wherein the expression vector is a phage.
18. The process according to claim 1 wherein during selection, the DNA which encodes for the peptide, polypeptide, or protein is mutagenized.
19. The process according to claim 18 wherein the DNA undergoes in vivo mutagenesis by recombination.
20. The process according to claim 18 wherein the DNA undergoes in vitro mutagenesis by mutagens.
21. The process according to claim 18 wherein the predetermined property is an adaptive improvement in the capacity to fulfill a desired function.
22. The process according to claim 1 wherein the predetermined property is the capacity to catalyse a given chemical reaction.
23. The process according to claim 1 wherein the predetermined property is the capacity to simulate at least one biologically active compound.
24. The process according to claim 23 wherein the biologically active compound is a member of the group consisting of hormones, neurotransmitters, growth factors, and specific regulators of replication, transcription, or translation of nucleic acids.
25. The process according to claim 1 wherein the predetermined property is the capacity to modify at least one biological function of at least one biologically active compound.
26. The process according to claim 25 wherein the biologically active compound is a member of the group consisting of hormones, neurotransmitters, growth factors, and specific regulators of replication, transcription, or translation of nucleic acids.
27. The process according to claim 1 wherein the predetermined property is that of having at least one epitope similar to one of the epitopes of a given antigen.
28. The process according to claim 27 wherein the antigen is epidermal growth factor.
29. The process according to claim 27 wherein the said property is the capacity to simulate an epitope of a given antigen, and that screening and/or selection of clones producing a peptide, polypeptide, or protein having this property is carried out by obtaining molecules which bind to that epitope, utilizing these molecules so obtained to identify those clones containing the peptide, polypeptide, or protein, growing the clones thus identified, separating and purifying the peptide, polypeptide, or protein produced by the clones, and then submitting these peptide(s), polypeptide(s), or protein(s) to an in vitro assay to verify that it has the said property.
30. The process according to claim 27 wherein the peptide, polypeptide, or protein having said property is used to make a vaccine.
31. The process according to claim 27 wherein an ensemble of peptides, polypeptides, or proteins having said property is used to make a vaccine.
32. The process according to claim 1 wherein the clones producing a peptide, polypeptide, or protein having the predetermined property are identified and isolated by affinity chromatography on antibodies corresponding to a protein expressed by a natural fragment of DNA.
33. The process according to claim 32 wherein after expression and purification of the identified peptide, polypeptide, or protein, novel fragments of the peptide, polypeptide, or protein having the predetermined property are separated and isolated.
34. The process according to claim 32 wherein the natural fragment of DNA contains a gene expressing B-galactosidase, and that the peptide, polypeptide, or protein is identified and isolated by affinity chromatography with anti-β-galactosidase antibodies.
35. The process according to claim 34 wherein after expression and purification of the identified peptide, polypeptide, or protein, novel fragments of the peptide, polypeptide, or protein having the predetermined property are separated and isolated.
36. The process according to claim 1 wherein the predetermined property is the capacity to bind to a given compound.
37. The process according to claim 36 wherein the said given compound is a member of the group consisting of peptides, polypeptides, and proteins.
38. The process according to claim 36 wherein the said given compound is a member of the group consisting of sequences of DNA and RNA.
39. The process according to claim 38 wherein the sequence of DNA acts as a cis-regulatory sequence of replication or transcription of a neighboring sequence of DNA.
40. The process according to claim 1 wherein the number of different peptides, polypeptides, or proteins expressed by all of the at least partially stochastic polynucleotide sequences is greater than 10,000.
41. The process according to claim 2 wherein the number of different peptides, polypeptides, or proteins expressed by all of the at least partially stochastic polynucleotide sequences is greater than 10,000.
42. The process according to claim 6 wherein the number of different peptides, polypeptides, or proteins expressed by all of the at least partially stochastic polynucleotide sequences is greater than 10,000.
43. The process according to claim 1 wherein DNA having the predetermined property is isolated from the clones.
44. The process according to claim 1 wherein RNA having the predetermined property is isolated from the clones.
45. A process for the production of an expression vector which includes at least one stochastic sequence of polynucleotides, comprising the steps of:
providing, in a common milieu, at least three oligonucleotides, said oligonucleotides comprising at least 7 nucleotides,
polymerizing said nucleotides to form stochastic double stranded DNA fragments, and
ligating said stochastic double stranded DNA fragments into an expression vector.
46. The process according to claim 45 wherein the oligonucleotides are heptamers.
47. The process according to claim 46 wherein the transforming DNA derived from a culture of clones is isolated and purified, the purified DNA is cut by means of at least one restriction enzyme which corresponds to a specific restriction site present in the palindromic heptamers or octamers but absent from the expression vector, then the ensemble of linearized stochastic DNA fragments obtained are simultaneously treated with T4 ligase in a manner to create a new ensemble of DNA containing new stochastic sequences, and this new ensemble of DNA is used to transform host cells.
48. The process according to claim 46 wherein the heptamers are palindromic.
49. The process according to claim 48 wherein the palindromic heptamers are selected from the group consisting of:
5′ XTCGCGA 3′ 5′ XCTGCAG 3′ 5′ RGGTACC 3′
where X=A, G, C, or T, and R=A or T.
50. The process according to claim 46 wherein the oligonucleotides are octamers.
51. The process according to claim 50 wherein the transforming DNA derived from a culture of clones is isolated and purified, the purified DNA is cut by means of at least one restriction enzyme which corresponds to a specific restriction site present in the palindromic heptamers or octamers but absent from the expression vector, then the ensemble of linearized stochastic DNA fragments obtained are simultaneously treated with T4 ligase in a manner to create a new ensemble of DNA containing new stochastic sequences, and this new ensemble of DNA is used to transform host cells.
52. The process according to claim 50 wherein the octamers are palindromic.
53. The process according to claim 52 wherein the palindromic octamers are selected from the group consisting of:
5′ GGAATTCC 3′ 5′ GGTCGACC 3′ 5′ CAAGCTTG 3′ 5′ CCATATGG 3′ 5′ CATCGATG 3′
54. The process according to claim 45 wherein the stochastic double stranded DNA fragments are about 160 to 800 base pairs in length.
55. A process for the production of an expression vector which includes at least one stochastic sequence of polynucleotides, comprising the steps of:
linearizing an expression vector,
reacting said linearized vector with terminal transferase in the presence of desired ratios of deoxynucleotide-triphosphates of guanine, cytosine, thymidine, and adenine to form polymers,
hybridizing said polymers, and
treating said polymers to form expression vectors consisting of stochastic double stranded DNA.
56. A process for the production of an expression vector comprising the steps of:
producing a library expression vectors which include at least one stochastic synthetic sequence of polynucleotides,
introducing the expression vectors thus obtained into host cells,
cultivating the host cells containing these expression vectors so as to clone the expression vector and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these expression vectors,
carrying out screening and/or selection methods on such clones producing the peptide, polypeptide, or protein having the predetermined property, and
isolating the cloned expression vectors so identified.
57. A process for the production of a peptide, polypeptide, or protein having a predetermined property, comprising the steps of:
producing by synthetic polynucleotide coupling, stochastically generated polynucleotide sequences;
forming a library of expression vectors containing such stochastically generated polynucleotide sequences;
culturing host cells containing the vectors to produce peptides, polypeptides, or proteins encoded by the stochastically generated polynucleotide sequences;
carrying out screening or selection on such host cells, to identify a peptide, polypeptide, or protein produced by the host cells having the predetermined property.
58. A process for the production of a peptide, polypeptide, or protein having a predetermined property, comprising the steps of:
producing at least partially stochastic polynucleotide sequences,
introducing the at least partially stochastic polynucleotide sequences thus obtained into host cells,
cultivating the transformed host cells containing these at least partially stochastic polynucleotide sequences so as to clone the stochastic polynucleotide sequences and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these stochastic polynucleotide sequences,
carrying out screening and/or selection methods on such clones of transformed host cells to identify the peptide, polypeptide, or protein having the predetermined property.
59. A process for the production of a host cell containing an expression vector which includes at least one stochastic sequence of polynucleotides, comprising the steps of:
producing a library of expression vectors which include at least one stochastic sequence of polynucleotides, said expression vectors being produced by the following steps:
providing, in a common milieu, at least three oligonucleotides, said oligonucleotides comprising at least 7 nucleotides;
polymerizing said nucleotides to form stochastic double stranded DNA fragments; and
ligating said stochastic double stranded DNA fragments into an expression vector;
introducing the expression vectors thus obtained into host cells; and
cultivating the host cells containing these expression vectors so as to clone the stochastic sequences and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these stochastic sequences.
60. A process for the production of a host cell containing an expression vector which includes at least one stochastic sequence of polynucleotides, comprising the steps of:
producing a library of expression vectors which include at least one stochastic sequence of polynucleotides, said expression vectors being produced by the following steps:
linearizing an expression vector,
reacting said linearized vector with terminal transferase in the presence of desired ratios of deoxynucleotide-triphosphates of guanine, cytosine, thymidine, and adenine to form polymers,
hybridizing said polymers, and
treating said polymers to form expression vectors consisting of stochastic double stranded DNA;
introducing the expression vectors thus obtained into host cells; and
cultivating the host cells containing these expression vectors so as to clone the stochastic sequences and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these stochastic sequences.
61. A process for the production of a host cell containing an expression vector which includes at least one stochastic sequence of polynucleotides, comprising the steps of:
producing a library of expression vectors which include at least one stochastic synthetic sequence of polynucleotides,
introducing the expression vectors thus obtained into host cells,
cultivating the host cells containing these expression vectors so as to clone the stochastic sequences and lead to the production of peptides, polypeptides, or proteins expressed by at least some of these stochastic sequences,
carrying out screening and/or selection methods on such clones to identify those clones producing the peptide, polypeptide, or protein having the predetermined property, and
isolating the cloned host cells so identified.
62. A library of expression vectors comprising stochastic polynucleotide sequences which encode at least 10,000 peptides, polypeptides, or proteins.
63. An expression vector produced in accordance with the process of claim 45, 55, or 56.
64. A host cell produced in accordance with the process of claim 59, 60, or 61.
65. The peptide, polypeptide, or protein produced in accordance with claim 1, 2, 4, 5, 6, 57, or 58.
66. A process for the production of an ensemble of peptides, polypeptides, or proteins, which ensemble has the capacity to catalyze a sequence of chemical reactions to thereby produce a desired compound, comprising the steps of:
a. producing a library of at least partially stochastic polynucleotide sequences,
b. introducing the at least partially stochastic polynucleotide sequences thus obtained into host cells,
c. simultaneously cultivating the independent transformed host cells containing these at least partially stochastic polynucleotide sequences so as to clone the stochastic polynucleotide sequences and lead to the production of a first set of peptides, polypeptides, or proteins expressed by at least some of these stochastic polynucleotide sequences,
d. combining the first set of peptides, polypeptides, or proteins with a collection of precursors to said desired compound under conditions favorable for said sequence of chemical reactions;
e. determining if said desired compound is produced, and if so, dividing the first set of peptides, polypeptides, or proteins into a plurality of subsets;
f. combining each of the subsets with the collection of precursors under conditions favorable for said sequence of chemical reactions;
g. determining, subset by subset, if said desired compound is produced with each of said subsets, and if so, dividing that subset into further subsets; and
h. repeating steps f. and g. until the ensemble of peptides, polypeptides, or proteins which catalyze the sequence of chemical reactions to produce the desired compound is identified.
US10/138,213 1985-03-30 2002-05-01 Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique Abandoned US20040161816A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/138,213 US20040161816A1 (en) 1985-03-30 2002-05-01 Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CH1379/85-8 1985-03-30
CH137985 1985-03-30
PCT/CH1985/000099 WO1986005803A1 (en) 1985-03-30 1985-06-17 Method for obtaining dna, rna, peptides, polypeptides or proteins by means of a dna recombinant technique
US94263086A 1986-11-20 1986-11-20
US61631990A 1990-11-21 1990-11-21
US97730792A 1992-11-16 1992-11-16
US13395293A 1993-10-08 1993-10-08
US08/349,510 US5723323A (en) 1985-03-30 1994-12-02 Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US08/464,569 US6492107B1 (en) 1986-11-20 1995-06-05 Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US10/138,213 US20040161816A1 (en) 1985-03-30 2002-05-01 Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/464,569 Continuation US6492107B1 (en) 1985-03-30 1995-06-05 Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique

Publications (1)

Publication Number Publication Date
US20040161816A1 true US20040161816A1 (en) 2004-08-19

Family

ID=4209046

Family Applications (7)

Application Number Title Priority Date Filing Date
US08/349,510 Expired - Lifetime US5723323A (en) 1985-03-30 1994-12-02 Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US08/468,477 Expired - Lifetime US5814476A (en) 1985-03-30 1995-06-05 Process for the production of stochastically-generated transcription or translation products
US08/464,142 Expired - Lifetime US5824514A (en) 1985-03-30 1995-06-05 Process for the production of expression vectors comprising at least one stochastic sequence of polynucleotides
US08/464,141 Expired - Lifetime US5817483A (en) 1985-03-30 1995-06-05 Process for the production of stochastically-generated peptides,polypeptides or proteins having a predetermined property
US08/464,327 Expired - Lifetime US5976862A (en) 1985-03-30 1995-06-05 Process for obtaining DNA, RNA, peptides, polypeptides, or proteins, by recombinant DNA technique
US08/468,468 Expired - Lifetime US6569641B1 (en) 1985-03-30 1995-06-05 Process for obtaining DNA, RNA, peptides, polypeptides, or protein by recombinant DNA technique
US10/138,213 Abandoned US20040161816A1 (en) 1985-03-30 2002-05-01 Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US08/349,510 Expired - Lifetime US5723323A (en) 1985-03-30 1994-12-02 Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US08/468,477 Expired - Lifetime US5814476A (en) 1985-03-30 1995-06-05 Process for the production of stochastically-generated transcription or translation products
US08/464,142 Expired - Lifetime US5824514A (en) 1985-03-30 1995-06-05 Process for the production of expression vectors comprising at least one stochastic sequence of polynucleotides
US08/464,141 Expired - Lifetime US5817483A (en) 1985-03-30 1995-06-05 Process for the production of stochastically-generated peptides,polypeptides or proteins having a predetermined property
US08/464,327 Expired - Lifetime US5976862A (en) 1985-03-30 1995-06-05 Process for obtaining DNA, RNA, peptides, polypeptides, or proteins, by recombinant DNA technique
US08/468,468 Expired - Lifetime US6569641B1 (en) 1985-03-30 1995-06-05 Process for obtaining DNA, RNA, peptides, polypeptides, or protein by recombinant DNA technique

Country Status (14)

Country Link
US (7) US5723323A (en)
EP (3) EP0229046B1 (en)
JP (4) JP2584613B2 (en)
CN (1) CN86102090A (en)
AU (1) AU4434585A (en)
CA (2) CA1341595C (en)
CH (1) CH0229046H1 (en)
DE (7) DE3546806C2 (en)
FR (1) FR2579618B1 (en)
GB (1) GB2183661B (en)
HK (1) HK20292A (en)
IN (2) IN165561B (en)
SG (1) SG7992G (en)
WO (1) WO1986005803A1 (en)

Families Citing this family (747)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5866363A (en) 1985-08-28 1999-02-02 Pieczenik; George Method and means for sorting and identifying biological information
US20060008806A1 (en) * 1986-07-17 2006-01-12 University Of Washington Method for producing novel DNA sequence with biological activity
US5824469A (en) * 1986-07-17 1998-10-20 University Of Washington Method for producing novel DNA sequences with biological activity
US5266684A (en) * 1988-05-02 1993-11-30 The Reagents Of The University Of California Peptide mixtures
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
CA2009996A1 (en) * 1989-02-17 1990-08-17 Kathleen S. Cook Process for making genes encoding random polymers of amino acids
US7413537B2 (en) 1989-09-01 2008-08-19 Dyax Corp. Directed evolution of disulfide-bonded micro-proteins
ATE168416T1 (en) * 1989-10-05 1998-08-15 Optein Inc CELL-FREE SYNTHESIS AND ISOLATION OF GENES AND POLYPEPTIDES
US5747334A (en) * 1990-02-15 1998-05-05 The University Of North Carolina At Chapel Hill Random peptide library
US5498538A (en) * 1990-02-15 1996-03-12 The University Of North Carolina At Chapel Hill Totally synthetic affinity reagents
JP3043407B2 (en) * 1990-02-15 2000-05-22 ザ ユニバーシティー オブ ノースカロライナ アット チャペル ヒル Complete synthetic affinity reagent
US5639595A (en) * 1990-05-01 1997-06-17 Isis Phamaceuticals, Inc. Identification of novel drugs and reagents
US5427908A (en) * 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US6465188B1 (en) 1990-06-11 2002-10-15 Gilead Sciences, Inc. Nucleic acid ligand complexes
US5567588A (en) * 1990-06-11 1996-10-22 University Research Corporation Systematic evolution of ligands by exponential enrichment: Solution SELEX
US5849890A (en) * 1990-06-11 1998-12-15 Nexstar Pharmaceuticals, Inc. High affinity oligonucleotide ligands to chorionic gonadotropin hormone and related glycoprotein hormones
US5972599A (en) * 1990-06-11 1999-10-26 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of cytokines
US5869641A (en) * 1990-06-11 1999-02-09 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of CD4
US6569620B1 (en) 1990-06-11 2003-05-27 Somalogic, Inc. Method for the automated generation of nucleic acid ligands
US5683867A (en) * 1990-06-11 1997-11-04 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: blended SELEX
US5543293A (en) * 1990-06-11 1996-08-06 Nexstar Pharmaceuticals, Inc. DNA ligands of thrombin
US6127119A (en) * 1990-06-11 2000-10-03 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands of tissue target
US20040132067A1 (en) * 1990-06-11 2004-07-08 Somalogic, Inc. Systematic evolution of ligands by exponential enrichment: photoselection of nucleic acid ligands and solution selex
US5853984A (en) * 1990-06-11 1998-12-29 Nexstar Pharmaceuticals, Inc. Use of nucleic acid ligands in flow cytometry
US5731144A (en) * 1990-06-11 1998-03-24 Nexstar Pharmaceuticals, Inc. High affinity TGFβ nucleic acid ligands
US5837456A (en) * 1990-06-11 1998-11-17 Nexstar Pharmaceuticals, Inc. High affinity oligonucleotide ligands to chorionic gonadotropin hormone and related glycoprotein hormones
US5527894A (en) * 1990-06-11 1996-06-18 Nexstar Pharmacueticals, Inc. Ligands of HIV-1 tat protein
US5789163A (en) * 1990-06-11 1998-08-04 Nexstar Pharmaceuticals, Inc. Enzyme linked oligonucleotide assays (ELONAS)
US6696252B2 (en) 1990-06-11 2004-02-24 Gilead Sciences, Inc. High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US5864026A (en) * 1990-06-11 1999-01-26 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US5780228A (en) * 1990-06-11 1998-07-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
US5763173A (en) * 1990-06-11 1998-06-09 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US5270163A (en) * 1990-06-11 1993-12-14 University Research Corporation Methods for identifying nucleic acid ligands
US5693502A (en) * 1990-06-11 1997-12-02 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US6716580B2 (en) 1990-06-11 2004-04-06 Somalogic, Inc. Method for the automated generation of nucleic acid ligands
US5459015A (en) * 1990-06-11 1995-10-17 Nexstar Pharmaceuticals, Inc. High-affinity RNA ligands of basic fibroblast growth factor
US6261774B1 (en) * 1990-06-11 2001-07-17 Gilead Sciences, Inc. Truncation selex method
US5874557A (en) * 1990-06-11 1999-02-23 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US5472841A (en) * 1990-06-11 1995-12-05 Nexstar Pharmaceuticals, Inc. Methods for identifying nucleic acid ligands of human neutrophil elastase
US6001988A (en) * 1990-06-11 1999-12-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
DK0786469T3 (en) * 1990-06-11 2006-07-10 Gilead Sciences Inc Nucleic acid
US5660985A (en) * 1990-06-11 1997-08-26 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands containing modified nucleotides
US5637682A (en) * 1990-06-11 1997-06-10 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to the tachykinin substance P
US6344321B1 (en) * 1990-06-11 2002-02-05 Gilead Sciences, Inc. Nucleic acid ligands which bind to hepatocyte growth factor/scatter factor (HGF/SF) or its receptor c-met
US5648214A (en) * 1990-06-11 1997-07-15 University Research Corporation High-affinity oligonucleotide ligands to the tachykinin substance P
US5654151A (en) * 1990-06-11 1997-08-05 Nexstar Pharmaceuticals, Inc. High affinity HIV Nucleocapsid nucleic acid ligands
US5496938A (en) * 1990-06-11 1996-03-05 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands to HIV-RT and HIV-1 rev
US6395888B1 (en) 1996-02-01 2002-05-28 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US5962219A (en) * 1990-06-11 1999-10-05 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chemi-selex
US5726017A (en) * 1990-06-11 1998-03-10 Nexstar Pharmaceuticals, Inc. High affinity HIV-1 gag nucleic acid ligands
US6011020A (en) * 1990-06-11 2000-01-04 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US5503978A (en) * 1990-06-11 1996-04-02 University Research Corporation Method for identification of high affinity DNA ligands of HIV-1 reverse transcriptase
US5795721A (en) * 1990-06-11 1998-08-18 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of ICP4
US5861254A (en) * 1997-01-31 1999-01-19 Nexstar Pharmaceuticals, Inc. Flow cell SELEX
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
US5705337A (en) * 1990-06-11 1998-01-06 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chemi-SELEX
US5811533A (en) * 1990-06-11 1998-09-22 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US5837834A (en) * 1990-06-11 1998-11-17 Nexstar Pharmaceuticals, Inc. High affinity HKGF nucleic acid ligands and inhibitors
US5587468A (en) * 1990-06-11 1996-12-24 University Research Corporation High affinity nucleic acid ligands to HIV integrase
US6280932B1 (en) 1990-06-11 2001-08-28 Gilead Sciences, Inc. High affinity nucleic acid ligands to lectins
US5723289A (en) * 1990-06-11 1998-03-03 Nexstar Pharmaceuticals, Inc. Parallel selex
US5731424A (en) * 1990-06-11 1998-03-24 Nexstar Pharmaceuticals, Inc. High affinity TGFβ nucleic acid ligands and inhibitors
US5707796A (en) * 1990-06-11 1998-01-13 Nexstar Pharmaceuticals, Inc. Method for selecting nucleic acids on the basis of structure
US5641629A (en) * 1990-06-11 1997-06-24 Nexstar Pharmacueticals Inc Spectroscopically detectable nucleic acid ligands
US6030776A (en) * 1990-06-11 2000-02-29 Nexstar Pharmaceuticals, Inc. Parallel SELEX
US6083696A (en) * 1990-06-11 2000-07-04 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands exponential enrichment: blended selex
US5688935A (en) * 1990-06-11 1997-11-18 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands of tissue target
US6232071B1 (en) * 1990-06-11 2001-05-15 Gilead Sciences, Inc. Tenascin-C nucleic acid ligands
US6331394B1 (en) 1991-06-10 2001-12-18 Gilead Sciences, Inc. Nucleic acid ligands to integrins
US6001577A (en) 1998-06-08 1999-12-14 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: photoselection of nucleic acid ligands and solution selex
US5766853A (en) * 1990-06-11 1998-06-16 Nexstar Pharmaceuticals, Inc. Method for identification of high affinity nucleic acid ligands to selectins
US5635615A (en) * 1990-06-11 1997-06-03 Nexstar Pharmaceuticals, Inc. High affinity HIV nucleocapsid nucleic acid ligands
US5763177A (en) * 1990-06-11 1998-06-09 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: photoselection of nucleic acid ligands and solution selex
US5763566A (en) * 1990-06-11 1998-06-09 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue SELEX
US5686592A (en) * 1990-06-11 1997-11-11 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to immunoglobulin E (IgE)
US5637459A (en) * 1990-06-11 1997-06-10 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chimeric selex
US6759392B1 (en) 1990-06-11 2004-07-06 Gilead Sciences, Inc. High affinity RNA ligands of basic fibroblast growth factor
US6124449A (en) * 1990-06-11 2000-09-26 Nexstar Pharmaceuticals, Inc. High affinity TGFβ nucleic acid ligands and inhibitors
US5874218A (en) * 1990-06-11 1999-02-23 Nexstar Pharmaceuticals, Inc. Method for detecting a target compound in a substance using a nucleic acid ligand
US6610841B1 (en) 1997-12-18 2003-08-26 Gilead Sciences, Inc. Nucleotide-based prodrugs
US6140490A (en) * 1996-02-01 2000-10-31 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of complement system proteins
US5846713A (en) * 1990-06-11 1998-12-08 Nexstar Pharmaceuticals, Inc. High affinity HKGF nucleic acid ligands and inhibitors
US5750342A (en) * 1990-06-11 1998-05-12 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands of tissue target
US20060084797A1 (en) * 1990-06-11 2006-04-20 Gilead Sciences, Inc. High affinity TGFbeta nucleic acid ligands and inhibitors
US5580737A (en) * 1990-06-11 1996-12-03 Nexstar Pharmaceuticals, Inc. High-affinity nucleic acid ligands that discriminate between theophylline and caffeine
US6177557B1 (en) 1990-06-11 2001-01-23 Nexstar Pharmaceuticals, Inc. High affinity ligands of basic fibroblast growth factor and thrombin
US5756287A (en) * 1990-06-11 1998-05-26 Nexstar Pharmaceuticals, Inc. High affinity HIV integrase inhibitors
US5712375A (en) * 1990-06-11 1998-01-27 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US6346611B1 (en) 1990-06-11 2002-02-12 Gilead Sciences, Inc. High affinity TGfβ nucleic acid ligands and inhibitors
US5629155A (en) * 1990-06-11 1997-05-13 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to immunoglobulin E (IgE)
US5789157A (en) * 1990-06-11 1998-08-04 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US6465189B1 (en) 1990-06-11 2002-10-15 Gilead Sciences, Inc. Systematic evolution of ligands by exponential enrichment: blended selex
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
US6916605B1 (en) * 1990-07-10 2005-07-12 Medical Research Council Methods for producing members of specific binding pairs
US7063943B1 (en) 1990-07-10 2006-06-20 Cambridge Antibody Technology Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5843701A (en) * 1990-08-02 1998-12-01 Nexstar Pharmaceticals, Inc. Systematic polypeptide evolution by reverse translation
CA2105300C (en) 1991-03-01 2008-12-23 Robert C. Ladner Process for the development of binding mini-proteins
US6762290B1 (en) 1999-07-29 2004-07-13 Gilead Sciences, Inc. High affinity vascular endothelial growth factor (VEGF) receptor nucleic acid ligands and inhibitors
US6028186A (en) * 1991-06-10 2000-02-22 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of cytokines
AU2313392A (en) * 1991-08-01 1993-03-02 University Research Corporation Systematic polypeptide evolution by reverse translation
US5270170A (en) * 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
US5733731A (en) * 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
PT1024191E (en) 1991-12-02 2008-12-22 Medical Res Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5656467A (en) * 1992-01-09 1997-08-12 The Trustees Of The University Of Pennsylvania Methods and materials for producing gene libraries
CA2132306C (en) * 1992-03-16 2012-07-10 Jacob N. Wohlstadter Selection methods
US5869644A (en) * 1992-04-15 1999-02-09 The Johns Hopkins University Synthesis of diverse and useful collections of oligonucleotidies
US5719273A (en) * 1993-06-14 1998-02-17 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modifications methods using nucleophiles and carbon monoxide
US5998142A (en) * 1993-09-08 1999-12-07 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chemi-SELEX
US6458539B1 (en) 1993-09-17 2002-10-01 Somalogic, Inc. Photoselection of nucleic acid ligands
JPH09504522A (en) * 1993-10-12 1997-05-06 グリコメド・インコーポレイテッド Library of glyco-peptides useful for identifying cell adhesion inhibitors
US20060078561A1 (en) * 1994-01-31 2006-04-13 The Trustees Of Boston University Polyclonal antibody libraries
AU1736495A (en) 1994-01-31 1995-08-15 Trustees Of Boston University Polyclonal antibody libraries
US6995017B1 (en) 1994-02-17 2006-02-07 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6395547B1 (en) 1994-02-17 2002-05-28 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6309883B1 (en) * 1994-02-17 2001-10-30 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US6406855B1 (en) 1994-02-17 2002-06-18 Maxygen, Inc. Methods and compositions for polypeptide engineering
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US6117679A (en) 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US20060257890A1 (en) 1996-05-20 2006-11-16 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US6335160B1 (en) * 1995-02-17 2002-01-01 Maxygen, Inc. Methods and compositions for polypeptide engineering
US6165793A (en) * 1996-03-25 2000-12-26 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US7153948B2 (en) 1994-04-25 2006-12-26 Gilead Sciences, Inc. High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US6682886B1 (en) 1994-04-28 2004-01-27 Gilead Sciences, Inc. Bivalent binding molecules of 7 transmembrane G protein-coupled receptors
US6010861A (en) * 1994-08-03 2000-01-04 Dgi Biotechnologies, Llc Target specific screens and their use for discovering small organic molecular pharmacophores
US6048698A (en) * 1994-09-20 2000-04-11 Nexstar Pharmaceuticals, Inc. Parallel SELEX™
AU3719395A (en) * 1994-09-21 1996-04-09 Cytogen Corporation Antigen binding peptides (abtides) from peptide libraries
US5885577A (en) * 1994-09-21 1999-03-23 Cytogen Corporation Antigen binding peptides (abtides) from peptide libraries
US6013443A (en) * 1995-05-03 2000-01-11 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue SELEX
US6114120A (en) * 1995-05-03 2000-09-05 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US8071737B2 (en) 1995-05-04 2011-12-06 Glead Sciences, Inc. Nucleic acid ligand complexes
US5859228A (en) * 1995-05-04 1999-01-12 Nexstar Pharmaceuticals, Inc. Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
US6207816B1 (en) 1995-06-02 2001-03-27 Nexstar Pharmaceuticals, Inc. High affinity oligonucleotide ligands to growth factors
US5723594A (en) * 1995-06-07 1998-03-03 Nexstar Pharmaceuticals, Inc. High affinity PDGF nucleic acid ligands
US20010053523A1 (en) * 1995-06-02 2001-12-20 M&E Biotech A/S. Method for identification of biologically active peptides and nucleic acids
CA2223078C (en) * 1995-06-07 2008-11-25 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands that bind to and inhibit dna polymerases
US6475806B1 (en) 1995-06-07 2002-11-05 Praecis Pharmaceuticals, Inc. Anchor libraries and identification of peptide binding sequences
US6699843B2 (en) 1995-06-07 2004-03-02 Gilead Sciences, Inc. Method for treatment of tumors using nucleic acid ligands to PDGF
US6183967B1 (en) 1995-06-07 2001-02-06 Nexstar Pharmaceuticals Nucleic acid ligand inhibitors to DNA polymerases
US6229002B1 (en) 1995-06-07 2001-05-08 Nexstar Pharmaceuticlas, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US5797870A (en) * 1995-06-07 1998-08-25 Indiana University Foundation Pericardial delivery of therapeutic and diagnostic agents
US20090118481A1 (en) * 1995-06-07 2009-05-07 Gilead Sciences, Inc. High Affinity Nucleic Acid Ligands To Lectins
US6740506B2 (en) 1995-12-07 2004-05-25 Diversa Corporation End selection in directed evolution
US6713279B1 (en) 1995-12-07 2004-03-30 Diversa Corporation Non-stochastic generation of genetic vaccines and enzymes
US6361974B1 (en) 1995-12-07 2002-03-26 Diversa Corporation Exonuclease-mediated nucleic acid reassembly in directed evolution
US5830696A (en) 1996-12-05 1998-11-03 Diversa Corporation Directed evolution of thermophilic enzymes
US6537776B1 (en) * 1999-06-14 2003-03-25 Diversa Corporation Synthetic ligation reassembly in directed evolution
US6939689B2 (en) 1995-12-07 2005-09-06 Diversa Corporation Exonuclease-mediated nucleic acid reassembly in directed evolution
US5965408A (en) 1996-07-09 1999-10-12 Diversa Corporation Method of DNA reassembly by interrupting synthesis
US20030219752A1 (en) * 1995-12-07 2003-11-27 Diversa Corporation Novel antigen binding molecules for therapeutic, diagnostic, prophylactic, enzymatic, industrial, and agricultural applications, and methods for generating and screening thereof
US20020164580A1 (en) * 1995-12-07 2002-11-07 Diversa Corporation Combinatorial screening of mixed populations of organisms
US6358709B1 (en) 1995-12-07 2002-03-19 Diversa Corporation End selection in directed evolution
US6352842B1 (en) 1995-12-07 2002-03-05 Diversa Corporation Exonucease-mediated gene assembly in directed evolution
US7018793B1 (en) * 1995-12-07 2006-03-28 Diversa Corporation Combinatorial screening of mixed populations of organisms
US20020028443A1 (en) * 1999-09-27 2002-03-07 Jay M. Short Method of dna shuffling with polynucleotides produced by blocking or interrupting a synthesis or amplification process
US6455247B1 (en) 1996-01-23 2002-09-24 Board Of Trustees Of The Leland Stanford Junior University Methods for screening for transdominant effector peptides and RNA molecules
US6365344B1 (en) 1996-01-23 2002-04-02 The Board Of Trustees Of The Leland Stanford Junior University Methods for screening for transdominant effector peptides and RNA molecules
US6506602B1 (en) 1996-03-25 2003-01-14 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6096548A (en) 1996-03-25 2000-08-01 Maxygen, Inc. Method for directing evolution of a virus
US6300065B1 (en) 1996-05-31 2001-10-09 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6696251B1 (en) 1996-05-31 2004-02-24 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6699658B1 (en) 1996-05-31 2004-03-02 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6838238B1 (en) 1996-10-17 2005-01-04 Invitrogen Corporation Morphatides: novel shape and structure libraries
US6426335B1 (en) * 1997-10-17 2002-07-30 Gilead Sciences, Inc. Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
EP0948632A1 (en) 1996-10-31 1999-10-13 Smithkline Beecham Corporation Methods for the characterization and selection of rna target motifs that bind compounds of pharmaceutical use
US20070009930A1 (en) * 1996-12-18 2007-01-11 Maxygen, Inc. Methods and compositions for polypeptide engineering
EP1007732B1 (en) 1997-01-17 2006-07-26 Maxygen, Inc. EVOLUTION OF procaryotic WHOLE CELLS BY RECURSIVE SEQUENCE RECOMBINATION
US7148054B2 (en) 1997-01-17 2006-12-12 Maxygen, Inc. Evolution of whole cells and organisms by recursive sequence recombination
US6326204B1 (en) 1997-01-17 2001-12-04 Maxygen, Inc. Evolution of whole cells and organisms by recursive sequence recombination
KR100566859B1 (en) 1997-01-21 2006-04-03 제너럴 하스피톨 코포레이션 Selection of proteins using rna-protein fusions
US6261804B1 (en) 1997-01-21 2001-07-17 The General Hospital Corporation Selection of proteins using RNA-protein fusions
US8207093B2 (en) * 1997-01-21 2012-06-26 The General Hospital Corporation Selection of proteins using RNA-protein fusions
US5871924A (en) * 1997-01-27 1999-02-16 Nexstar Pharmaceuticals, Inc. Method for the production of ligands capable of facilitating aminoacyl-RNA synthesis
US6153410A (en) * 1997-03-25 2000-11-28 California Institute Of Technology Recombination of polynucleotide sequences using random or defined primers
US6180341B1 (en) 1997-05-01 2001-01-30 Board Of Regents, The Universiry Of Texas System In vitro scanning saturation mutagenesis of proteins
US6391547B1 (en) * 1997-09-09 2002-05-21 Center For The Application Of Molecular Biology To International Agriculture Microbial β-glucuronidase genes, gene products and uses thereof
US7087420B1 (en) 1997-07-17 2006-08-08 Cambia Microbial β-glucuronidase genes, gene products and uses thereof
US6241701B1 (en) 1997-08-01 2001-06-05 Genetronics, Inc. Apparatus for electroporation mediated delivery of drugs and genes
US6670127B2 (en) 1997-09-16 2003-12-30 Egea Biosciences, Inc. Method for assembly of a polynucleotide encoding a target polypeptide
EP1030861A4 (en) 1997-10-31 2001-09-05 Maxygen Inc Modification of virus tropism and host range by viral genome shuffling
DK1036198T3 (en) * 1997-12-08 2013-01-02 California Inst Of Techn Method for Preparation of Polynucleotide and Polypeptide Sequences
US20070166741A1 (en) 1998-12-14 2007-07-19 Somalogic, Incorporated Multiplexed analyses of test samples
WO1999031276A1 (en) 1997-12-15 1999-06-24 Nexstar Pharmaceuticals, Inc. Homogeneous detection of a target through nucleic acid ligand-ligand beacon interaction
US20030219803A1 (en) * 1997-12-15 2003-11-27 Somalogic, Incorporated Homogeneous detection of a target through nucleic acid ligand-ligand beacon interaction
US5989823A (en) 1998-09-18 1999-11-23 Nexstar Pharmaceuticals, Inc. Homogeneous detection of a target through nucleic acid ligand-ligand beacon interaction
US6759243B2 (en) 1998-01-20 2004-07-06 Board Of Trustees Of The University Of Illinois High affinity TCR proteins and methods
US6541011B2 (en) 1998-02-11 2003-04-01 Maxygen, Inc. Antigen library immunization
US7390619B1 (en) * 1998-02-11 2008-06-24 Maxygen, Inc. Optimization of immunomodulatory properties of genetic vaccines
FR2782325B1 (en) * 1998-08-12 2002-05-24 Proteus METHOD OF IDENTIFYING POLYNUCLEOTIDE SEQUENCES AND / OR CORRESPONDING PROTEINS FROM A SAMPLE OF NUCLEIC ACIDS
US7157083B2 (en) * 1998-04-17 2007-01-02 Surrogate Pharmaceutical Pathways, Llc Compositions and methods for treating retroviral infections
US6500617B1 (en) 1998-05-01 2002-12-31 Maxygen, Inc. Optimization of pest resistance genes using DNA shuffling
US7153655B2 (en) * 1998-06-16 2006-12-26 Alligator Bioscience Ab Method for in vitro molecular evolution of protein function involving the use of exonuclease enzyme and two populations of parent polynucleotide sequence
US6365408B1 (en) 1998-06-19 2002-04-02 Maxygen, Inc. Methods of evolving a polynucleotides by mutagenesis and recombination
CA2334170A1 (en) 1998-07-27 2000-02-10 Genentech, Inc. Improved transformation efficiency in phage display through modification of a coat protein
WO2000009464A1 (en) 1998-08-17 2000-02-24 Phylos, Inc. Identification of compound-protein interactions using libraries of protein-nucleic acid fusion molecules
US6773911B1 (en) * 1998-11-23 2004-08-10 Amgen Canada Inc. Apoptosis-inducing factor
US20020055626A1 (en) * 2000-06-09 2002-05-09 Turner C. Alexander Novel human seven transmembrane proteins and polynucleotides encoding the same
US6570003B1 (en) * 2001-01-09 2003-05-27 Lexion Genetics Incorporated Human 7TM proteins and polynucleotides encoding the same
WO2000040715A2 (en) * 1999-01-05 2000-07-13 Trustees Of Boston University Improved nucleic acid cloning
WO2000042559A1 (en) * 1999-01-18 2000-07-20 Maxygen, Inc. Methods of populating data structures for use in evolutionary simulations
US20030054390A1 (en) * 1999-01-19 2003-03-20 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
US20070065838A1 (en) * 1999-01-19 2007-03-22 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
US20030054360A1 (en) * 1999-01-19 2003-03-20 Larry Gold Method and apparatus for the automated generation of nucleic acid ligands
US20090130718A1 (en) * 1999-02-04 2009-05-21 Diversa Corporation Gene site saturation mutagenesis
US6329145B1 (en) 1999-02-09 2001-12-11 Gilead Science, Inc. Determining non-nucleic acid molecule binding to target by competition with nucleic acid ligand
EP1165775A2 (en) 1999-03-05 2002-01-02 Maxygen, Inc. Recombination of insertion modified nucleic acids
US6703240B1 (en) 1999-04-13 2004-03-09 Maxygar, Inc. Modified starch metabolism enzymes and encoding genes for improvement and optimization of plant phenotypes
US7332308B1 (en) 1999-05-21 2008-02-19 The Penn State Research Foundation Incrementally truncated nucleic acids and methods of making same
US6280943B1 (en) 1999-06-17 2001-08-28 Gilead Sciences, Inc. 2′-fluoropyrimidine anti-calf intestinal phosphatase nucleic acid ligands
DK1860188T3 (en) 1999-07-07 2011-06-27 Zymogenetics Inc Human cytokine receptor
US6387620B1 (en) * 1999-07-28 2002-05-14 Gilead Sciences, Inc. Transcription-free selex
US6171795B1 (en) 1999-07-29 2001-01-09 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands to CD40ligand
DE60042021D1 (en) * 1999-07-29 2009-05-28 Gilead Sciences Inc NUCLEAR ACID SURFACES FOR THE HEPATOCYTIC GROWTH FACTOR / DISPERSION FACTOR (HGF / SF) AND ITS C-MET RECEPTOR
US7005260B1 (en) 2000-01-28 2006-02-28 Gilead Sciences, Inc. Tenascin-C nucleic acid ligands
US6506887B1 (en) 1999-07-29 2003-01-14 Somalogic, Incorporated Conditional-selex
US6593125B2 (en) 2000-11-20 2003-07-15 Lexicon Genetics Incorporated Human kinases and polynucleotides encoding the same
US20080050809A1 (en) * 1999-09-28 2008-02-28 Alejandro Abuin Novel human kinases and polynucleotides encoding the same
US6586230B1 (en) * 2000-10-27 2003-07-01 Lexicon Genetics Incorporated Human kinase and polynucleotides encoding the same
US6541252B1 (en) 2000-05-19 2003-04-01 Lexicon Genetics Incorporated Human kinases and polynucleotides encoding the same
US6797510B1 (en) 2001-05-24 2004-09-28 Lexicon Genetics Incorporated Human kinases and polynucleotides encoding the same
US6841377B1 (en) 2001-06-13 2005-01-11 Lexicon Genetics Incorporated Human kinase and polynucleotides encoding the same
US6849425B1 (en) 1999-10-14 2005-02-01 Ixsys, Inc. Methods of optimizing antibody variable region binding affinity
JP2003516755A (en) * 1999-12-15 2003-05-20 ジェネンテック・インコーポレーテッド Shotgun scanning, a combined method for mapping functional protein epitopes
WO2001053539A1 (en) 2000-01-24 2001-07-26 Phylos, Inc. Sensitive, multiplexed diagnostic assays for protein analysis
US7022479B2 (en) * 2000-01-24 2006-04-04 Compound Therapeutics, Inc. Sensitive, multiplexed diagnostic assays for protein analysis
CA2398483A1 (en) * 2000-01-26 2001-08-16 Lexicon Genetics Incorporated Human neurexin-like proteins and polynucleotides encoding the same
US20050239061A1 (en) * 2000-03-01 2005-10-27 Marshall William S Identification and use of effectors and allosteric molecules for the alteration of gene expression
US7514239B2 (en) 2000-03-28 2009-04-07 Amgen Inc. Nucleic acid molecules encoding beta-like glycoprotein hormone polypeptides and heterodimers thereof
AU2001289284A1 (en) * 2000-04-04 2001-10-15 Enanta Pharmaceuticals, Inc. Methods for identifying peptide aptamers capable of altering a cell phenotype
EP2199393B1 (en) * 2000-04-17 2012-10-31 Dyax Corp. Methods of constructing display libraries of genetic packages for members of a diverse family of peptides
US8288322B2 (en) 2000-04-17 2012-10-16 Dyax Corp. Methods of constructing libraries comprising displayed and/or expressed members of a diverse family of peptides, polypeptides or proteins and the novel libraries
AU2001257091A1 (en) * 2000-04-18 2001-10-30 Gilead Sciences, Inc. Aptamer based two-site binding assay
US6479262B1 (en) 2000-05-16 2002-11-12 Hercules, Incorporated Solid phase enzymatic assembly of polynucleotides
WO2001090179A2 (en) * 2000-05-23 2001-11-29 Lexicon Genetics Incorporated Novel human thrombospondin-like proteins and polynucleotides encoding the same
AU2001275337A1 (en) * 2000-06-07 2001-12-17 Lexicon Genetics Incorporated Novel human transporter proteins and polynucleotides encoding the same
US6465632B1 (en) * 2000-06-09 2002-10-15 Lexicon Genetics Incorporated Human phosphatases and polynucleotides encoding the same
EP2077279B1 (en) 2000-06-28 2012-09-05 Amgen Inc. Thymic stromal lymphopoietin receptor molecules and uses thereof
US6994963B1 (en) 2000-07-10 2006-02-07 Ambion, Inc. Methods for recombinatorial nucleic acid synthesis
AU2001273432A1 (en) * 2000-07-13 2002-01-30 Invitrogen Corporation Methods and compositions for rapid protein and peptide extraction and isolation using a lysis matrix
AU2001277014A1 (en) * 2000-07-21 2002-02-05 Trustees Of Boston University Modular vector systems
WO2002010183A1 (en) * 2000-07-31 2002-02-07 Menzel, Rolf Compositions and methods for directed gene assembly
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
US6902734B2 (en) 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
AU2001283094A1 (en) * 2000-08-07 2002-02-18 Sloan-Kettering Institute For Cancer Research Method and composition for immunization using mixed pools of mutated nucleic acids or peptides
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
CA2420036A1 (en) * 2000-08-22 2002-02-28 Lexicon Genetics Incorporated Human 7tm proteins and polynucleotides encoding the same
AU2001285326A1 (en) * 2000-08-31 2002-03-13 Lexicon Genetics Incorporated Human kinase proteins and polynucleotides encoding the same
US6376190B1 (en) 2000-09-22 2002-04-23 Somalogic, Inc. Modified SELEX processes without purified protein
EP2322648A1 (en) * 2000-09-26 2011-05-18 Duke University RNA aptamers and methods for identifying the same
EP1325457A4 (en) * 2000-10-10 2007-10-24 Genencor Int Information rich libraries
JP4176466B2 (en) 2000-10-16 2008-11-05 ギリード・サイエンシズ・インコーポレーテッド Nucleic acid ligands for prostate specific membrane antigen
WO2002070705A2 (en) * 2000-10-27 2002-09-12 Lexicon Genetics Incorporated Human 7tm proteins and polynucleotides encoding them
AU2002232936A1 (en) * 2000-10-30 2002-05-15 Lexicon Genetics Incorporated Human 7TM proteins and polynucleotides encoding the same
AU2002220275A1 (en) * 2000-12-08 2002-06-18 The Board Of Trustees Of The University Of Illinois Mutated class ii major histocompatibility proteins
EP1341906A2 (en) * 2000-12-11 2003-09-10 Lexicon Genetics Incorporated Novel human kinase and polynucleotides encoding the same
AU2002232642B2 (en) * 2000-12-12 2007-02-01 Lexicon Pharmaceuticals, Inc. Novel human kinases and uses thereof
US6958213B2 (en) 2000-12-12 2005-10-25 Alligator Bioscience Ab Method for in vitro molecular evolution of protein function
JP4860098B2 (en) 2000-12-18 2012-01-25 ダイアックス、コープ Library focused on genetic packaging
US6852844B1 (en) 2000-12-20 2005-02-08 Lexicon Genetics Incorporated Human protocadherin proteins and polynucleotides encoding the same
AU2002231736A1 (en) 2000-12-22 2002-07-08 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Use of repulsive guidance molecule (rgm) and its modulators
US20020086292A1 (en) 2000-12-22 2002-07-04 Shigeaki Harayama Synthesis of hybrid polynucleotide molecules using single-stranded polynucleotide molecules
WO2002059325A2 (en) * 2000-12-27 2002-08-01 Lexicon Genetics Incorporated Human kinases and polynucleotides encoding the same
JP4061043B2 (en) 2000-12-28 2008-03-12 株式会社ポストゲノム研究所 Method for producing peptide etc. by in vitro transcription / translation system
EP1407029A2 (en) * 2001-01-23 2004-04-14 Lexicon Genetics Incorporated Novel human kinases and polynucleotides encoding the same
US7582423B2 (en) * 2001-02-02 2009-09-01 Novici Biotech Llc Population of polynucleotide sequence variants
JP4689940B2 (en) * 2001-02-02 2011-06-01 ノビシ バイオテック,リミティド ライアビリティ カンパニー Method for increasing heteroduplex complementarity
US20040142433A1 (en) * 2001-02-02 2004-07-22 Padgett Hal S. Polynucleotide sequence variants
US7838219B2 (en) * 2001-02-02 2010-11-23 Novici Biotech Llc Method of increasing complementarity in a heteroduplex
WO2002065125A1 (en) * 2001-02-13 2002-08-22 Invitrogen Corporation Methods and compositions for isolation of biological macromolecules
EP1373496A4 (en) * 2001-02-22 2004-10-27 Praecis Pharm Inc Methods for identifying peptides which modulate a biological process
US6875845B2 (en) 2001-03-02 2005-04-05 Zymogenetics, Inc. Mouse cytokine receptor
AU2002257076A1 (en) 2001-03-19 2002-10-03 President And Fellows Of Harvard College Nucleic acid shuffling
US7807408B2 (en) * 2001-03-19 2010-10-05 President & Fellows Of Harvard College Directed evolution of proteins
WO2002074932A2 (en) 2001-03-20 2002-09-26 Lexicon Genetics Incorporated Novel human kinase and polynucleotides encoding the same
CA2443516A1 (en) * 2001-04-06 2002-10-17 Lexicon Genetics Incorporated Novel human kinases and polynucleotides encoding the same
WO2002081671A1 (en) * 2001-04-06 2002-10-17 Lexicon Genetics Incorporated Novel human kinase and polynucleotides encoding the same
US6644173B2 (en) * 2001-04-11 2003-11-11 Keuring, Incorporated Beverage filter cartridge holder
AU2002254773B2 (en) * 2001-05-03 2005-12-08 Rensselaer Polytechnic Institute Novel methods of directed evolution
CA2446967A1 (en) * 2001-05-09 2002-11-14 Lexicon Genetics Incorporated Novel human kinases and polynucleotides encoding the same
SG157961A1 (en) * 2001-05-25 2010-01-29 Univ Duke Modulators of pharmacological agents
WO2002097095A1 (en) * 2001-05-25 2002-12-05 Lexicon Genetics Incorporated Novel human transporter proteins and polynucleotides encoding the same
JP2005500829A (en) 2001-05-29 2005-01-13 レキシコン・ジェネティクス・インコーポレーテッド Novel human hydroxylase and polynucleotide encoding the enzyme
US20030148380A1 (en) * 2001-06-05 2003-08-07 Belcher Angela M. Molecular recognition of materials
US20050164515A9 (en) * 2001-06-05 2005-07-28 Belcher Angela M. Biological control of nanoparticle nucleation, shape and crystal phase
US20030113714A1 (en) * 2001-09-28 2003-06-19 Belcher Angela M. Biological control of nanoparticles
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
KR100576674B1 (en) 2001-06-20 2006-05-10 제넨테크, 인크. Compositions and Methods for the Diagnosis and Treatment of Tumor
US20030148408A1 (en) 2001-09-18 2003-08-07 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
DE10131441A1 (en) * 2001-06-29 2003-01-30 Henkel Kgaa A new group of alpha amylases and a method for identifying and obtaining new alpha amylases
US20040204669A1 (en) * 2001-07-05 2004-10-14 Hofmann Gunter A. Apparatus for electroporation mediated delivery for drugs and genes
US20030148264A1 (en) * 2001-07-06 2003-08-07 Genentech, Inc. Phage displayed PDZ domain ligands
US6979733B2 (en) * 2001-08-03 2005-12-27 Diversa Corporation Epoxide hydrolases, nucleic acids encoding them and methods of making and using them
US6943001B2 (en) * 2001-08-03 2005-09-13 Diversa Corporation Epoxide hydrolases, nucleic acids encoding them and methods for making and using them
DE10138753B4 (en) * 2001-08-07 2017-07-20 Henkel Ag & Co. Kgaa Detergents and cleaners with hybrid alpha-amylases
US7647184B2 (en) * 2001-08-27 2010-01-12 Hanall Pharmaceuticals, Co. Ltd High throughput directed evolution by rational mutagenesis
JP4578098B2 (en) 2001-10-01 2010-11-10 ダイアックス、コープ Multi-chain eukaryotic display vectors and their use
US20030073104A1 (en) * 2001-10-02 2003-04-17 Belcher Angela M. Nanoscaling ordering of hybrid materials using genetically engineered mesoscale virus
EP1840211A1 (en) 2001-10-31 2007-10-03 Danisco A/S Pyranosone dehydratase from phanerochaete chrysosporium
CN100374457C (en) 2001-11-14 2008-03-12 森托科尔公司 Anti-IL-6 antibodies, compositions, methods and uses
US20030185870A1 (en) * 2001-11-20 2003-10-02 Grinstaff Mark W. Interfacial biomaterials
US6768476B2 (en) * 2001-12-05 2004-07-27 Etenna Corporation Capacitively-loaded bent-wire monopole on an artificial magnetic conductor
WO2003051314A2 (en) 2001-12-18 2003-06-26 Medallion Biomedical, Llc Antibiotic compounds
US20030157534A1 (en) * 2001-12-26 2003-08-21 Manuel Engelhorn DNA immunization with libraries of minigenes encoding degenerate variants of major histocompatability class l restricted epitopes
KR100801388B1 (en) 2002-01-02 2008-02-05 제넨테크, 인크. Compositions and Methods for the Diagnosis and Treatment of Tumor
US20030224392A1 (en) * 2002-01-28 2003-12-04 Manuel Engelhorn Method for identification of mutant antigens with enhanced immunogenicity
US7078211B2 (en) * 2002-02-01 2006-07-18 Large Scale Biology Corporation Nucleic acid molecules encoding endonucleases and methods of use thereof
US20030157495A1 (en) * 2002-02-01 2003-08-21 Padgett Hal S. Nucleic acid molecules encoding CEL I endonuclease and methods of use thereof
AU2003217379A1 (en) 2002-02-15 2003-09-09 Somalogic, Inc. Methods and reagents for detecting target binding by nucleic acid ligands
AU2003217693A1 (en) * 2002-02-22 2003-09-09 Board Of Trustees Of The University Of Illinois Beta chain-associated regulator of apoptosis
US7662924B2 (en) * 2002-02-22 2010-02-16 The Board Of Trustees Of The University Of Illinois Beta chain-associated regulator of apoptosis
US20030224404A1 (en) * 2002-02-25 2003-12-04 Manuel Vega High throughput directed evolution of nucleic acids by rational mutagenesis
WO2003072054A2 (en) * 2002-02-25 2003-09-04 Cabot Corporation Custom ligand design for biomolecular filtration and purification for bioseperation
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
MXPA04010092A (en) 2002-04-16 2004-12-13 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
CN102174546A (en) 2002-04-19 2011-09-07 维莱尼姆公司 Phospholipases, nucleic acids encoding them and methods for making and using them
US7226771B2 (en) 2002-04-19 2007-06-05 Diversa Corporation Phospholipases, nucleic acids encoding them and methods for making and using them
US7262012B2 (en) * 2002-05-17 2007-08-28 Alligator Bioscience Ab Method for in vitro molecular evolution of protein function using varied exonuclease digestion in two polynucleotide populations
EP1513879B1 (en) * 2002-06-03 2018-08-22 Genentech, Inc. Synthetic antibody phage libraries
DE60319259T2 (en) * 2002-06-14 2009-03-05 Dyax Corp., Cambridge Recombination of nucleic acid library members
EP1552002A4 (en) * 2002-06-18 2006-02-08 Archemix Corp Aptamer-toxin molecules and methods for using same
ATE516366T1 (en) 2002-07-25 2011-07-15 Archemix Corp REGULATED APTAMER THERAPEUTICS
MXPA05001287A (en) 2002-08-06 2005-08-03 Verdia Llc Ap1 amine oxidase variants.
US20040067532A1 (en) 2002-08-12 2004-04-08 Genetastix Corporation High throughput generation and affinity maturation of humanized antibody
US20060020396A1 (en) * 2002-09-09 2006-01-26 Rene Gantier Rational directed protein evolution using two-dimensional rational mutagenesis scanning
AU2003263552A1 (en) * 2002-09-09 2004-03-29 Nautilus Biotech Rational evolution of cytokines for higher stability, the cytokines and encoding nucleic acid molecules
US20050202438A1 (en) * 2002-09-09 2005-09-15 Rene Gantier Rational directed protein evolution using two-dimensional rational mutagenesis scanning
US9303262B2 (en) 2002-09-17 2016-04-05 Archemix Llc Methods for identifying aptamer regulators
US20050064508A1 (en) * 2003-09-22 2005-03-24 Semzyme Peptide mediated synthesis of metallic and magnetic materials
WO2004029207A2 (en) 2002-09-27 2004-04-08 Xencor Inc. Optimized fc variants and methods for their generation
JP3447009B1 (en) * 2002-10-29 2003-09-16 實 平垣 Construct structure and method for producing the same
US20040180360A1 (en) * 2002-11-21 2004-09-16 Charles Wilson 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
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc 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
WO2004050899A2 (en) * 2002-12-03 2004-06-17 Archemix Corporation Method for in vitro selection of 2’-substituted nucleic acids
US20050037394A1 (en) * 2002-12-03 2005-02-17 Keefe Anthony D. Method for in vitro selection of 2'-substituted nucleic acids
AU2003303459A1 (en) * 2002-12-26 2004-07-22 Applied Research Systems Ars Holding N.V. Spliced variants of lgr6
CA2510003A1 (en) * 2003-01-16 2004-08-05 Genentech, Inc. Synthetic antibody phage libraries
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
CA2920416A1 (en) 2003-03-06 2004-10-28 Basf Enzymes Llc Amylases, nucleic acids encoding them and methods for making and using them
CA3007908A1 (en) 2003-03-07 2005-04-14 Dsm Ip Assets B.V. Hydrolases, nucleic acids encoding them and methods for making and using them
MXPA05010681A (en) 2003-04-04 2005-12-15 Diversa Corp Pectate lyases, nucleic acids encoding them and methods for making and using them.
US20050250106A1 (en) * 2003-04-24 2005-11-10 David Epstein Gene knock-down by intracellular expression of aptamers
AU2003241409A1 (en) * 2003-05-12 2005-01-21 Potomac Pharmaceuticals, Inc. Gene expression suppression agents
US9708410B2 (en) 2003-05-30 2017-07-18 Janssen Biotech, Inc. Anti-tissue factor antibodies and compositions
WO2005003319A2 (en) 2003-07-02 2005-01-13 Diversa Corporation Glucanases, nucleic acids encoding them and methods for making and using them
PL1641822T3 (en) 2003-07-08 2013-10-31 Genentech Inc Il-17 a/f heterologous polypeptides and therapeutic uses thereof
EP2311994A1 (en) * 2003-08-01 2011-04-20 Life Technologies Corporation Compositions and methods for preparing short RNA molecules and other nucleic acids
WO2005012531A2 (en) * 2003-08-01 2005-02-10 Genentech, Inc. Antibody cdr polypeptide sequences with restricted diversity
WO2005021714A2 (en) 2003-08-11 2005-03-10 Diversa Corporation Laccases, nucleic acids encoding them and methods for making and using them
JP2005065575A (en) * 2003-08-22 2005-03-17 Japan Science & Technology Agency Preparation of protein molecular-diversified population by frame shuffling
EP1660534A2 (en) * 2003-08-22 2006-05-31 MedImmune, Inc. Humanization of antibodies
US20060134105A1 (en) * 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
US8883147B2 (en) 2004-10-21 2014-11-11 Xencor, Inc. Immunoglobulins insertions, deletions, and substitutions
US8399618B2 (en) 2004-10-21 2013-03-19 Xencor, Inc. Immunoglobulin insertions, deletions, and substitutions
PT2161283E (en) 2003-11-17 2014-08-29 Genentech Inc Compositions comprising antibodies against cd79b conjugated to a growth inhibitory agent or cytotoxic agent and methods for the treatment of tumor of hematopoietic origin
US20050112585A1 (en) * 2003-11-21 2005-05-26 Dominic Zichi Method for adjusting the quantification range of individual analytes in a multiplexed assay
EP1697741A4 (en) * 2003-12-04 2008-02-13 Xencor Inc Methods of generating variant proteins with increased host string content and compositions thereof
CA2550431A1 (en) * 2003-12-19 2005-07-14 Osteotech, Inc. Tissue-derived mesh for orthopedic regeneration
US7803931B2 (en) 2004-02-12 2010-09-28 Archemix Corp. Aptamer therapeutics useful in the treatment of complement-related disorders
LT3385384T (en) 2004-02-12 2020-09-25 Archemix Llc Aptamer therapeutics useful in the treatment of complement-related disorders
AU2005220910A1 (en) * 2004-03-05 2005-09-22 Archemix Corp. Aptamers to the human IL-12 cytokine family and their use as autoimmune disease therapeutics
US20060193821A1 (en) * 2004-03-05 2006-08-31 Diener John L Aptamers to the human IL-12 cytokine family and their use as autoimmune disease therapeutics
US7569223B2 (en) * 2004-03-22 2009-08-04 The Rockefeller University Phage-associated lytic enzymes for treatment of Streptococcus pneumoniae and related conditions
EP2053062A1 (en) 2004-03-24 2009-04-29 Xencor, Inc. Immunoglobin variants outside the Fc region
US7785903B2 (en) * 2004-04-09 2010-08-31 Genentech, Inc. Variable domain library and uses
US20080214489A1 (en) * 2004-04-19 2008-09-04 Anthony Dominic Keefe Aptamer-mediated intracellular delivery of oligonucleotides
ES2494940T3 (en) * 2004-04-22 2014-09-16 Regado Biosciences, Inc. Enhanced coagulation factor modulators
US7579450B2 (en) 2004-04-26 2009-08-25 Archemix Corp. Nucleic acid ligands specific to immunoglobulin E and their use as atopic disease therapeutics
US20070286854A1 (en) 2004-04-28 2007-12-13 Bayer Healthcare Ag Diagnostics And Therapeutics For Diseases Associated With Dipeptidyl-Peptidase 1 (Dpp1)
AU2005250369A1 (en) * 2004-05-18 2005-12-15 Genentech, Inc. M13 virus major coat protein variants for C-terminal and BI-terminal display of a heterologous protein
EP1791853B1 (en) 2004-06-16 2014-02-26 DSM IP Assets B.V. Compositions and methods for enzymatic decolorization of chlorophyll
CA2578046A1 (en) * 2004-09-07 2006-03-16 Archemix Corp. Aptamer medicinal chemistry
US7566701B2 (en) * 2004-09-07 2009-07-28 Archemix Corp. Aptamers to von Willebrand Factor and their use as thrombotic disease therapeutics
BRPI0514984A (en) 2004-09-07 2008-07-01 Archemix Corp aptamers for von willebrand factor and their use as therapeutics for thrombotic disease
GB0422052D0 (en) 2004-10-04 2004-11-03 Dansico As Enzymes
GB0423139D0 (en) 2004-10-18 2004-11-17 Danisco Enzymes
EP2845865A1 (en) 2004-11-12 2015-03-11 Xencor Inc. Fc variants with altered binding to FcRn
CA2594356C (en) 2005-01-05 2018-07-17 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H. Synthetic immunoglobulin domains with binding properties engineered in regions of the molecule different from the complementarity determining regions
PA8660701A1 (en) 2005-02-04 2006-09-22 Pfizer Prod Inc SMALL AGONISTS AND THEIR USES
JP2008532931A (en) 2005-02-08 2008-08-21 ザイモジェネティクス, インコーポレイテッド Anti-IL-20, anti-IL-22 and anti-IL-22RA antibodies and binding partners and uses in inflammation
MX2007010922A (en) 2005-03-10 2007-10-12 Genentech Inc Methods and compositions for modulating vascular integrity.
US20090038023A1 (en) 2005-03-10 2009-02-05 Verenium Corporation Lyase Enzymes, Nucleic Acids Encoding Them and Methods For Making and Using Them
EP1861506B1 (en) 2005-03-15 2015-04-15 BP Corporation North America Inc. Cellulases, nucleic acids encoding them and methods for making and using them
PE20061324A1 (en) 2005-04-29 2007-01-15 Centocor Inc ANTI-IL-6 ANTIBODIES, COMPOSITIONS, METHODS AND USES
EP3683230A1 (en) 2005-05-12 2020-07-22 ZymoGenetics, Inc. Compositions and methods for modulating immune responses
US20060271262A1 (en) * 2005-05-24 2006-11-30 Mclain Harry P Iii Wireless agricultural network
US8389469B2 (en) * 2005-06-06 2013-03-05 The Rockefeller University Bacteriophage lysins for Bacillus anthracis
US7582291B2 (en) * 2005-06-30 2009-09-01 The Rockefeller University Bacteriophage lysins for Enterococcus faecalis, Enterococcus faecium and other bacteria
MX2007016401A (en) * 2005-06-30 2008-02-20 Abbott Lab Il-12/p40 binding proteins.
ES2710289T3 (en) 2005-06-30 2019-04-24 Janssen Biotech Inc Anti-IL-23 antibodies, compositions, procedures and uses
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500359A3 (en) 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500352A1 (en) 2005-08-19 2012-09-19 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
DE602006018746D1 (en) 2005-08-24 2011-01-20 Univ Rockefeller PLY-GBS MUTANT LYSINE
WO2007035527A2 (en) 2005-09-15 2007-03-29 Duke University Non-fouling polymeric surface modification and signal amplification method for biomolecular detection
ES2542501T3 (en) * 2005-09-30 2015-08-06 Abbvie Deutschland Gmbh & Co Kg Protein binding domains of the protein family of repulsive targeting molecules (RGM) and functional fragments thereof, as well as their use
EP2465870A1 (en) 2005-11-07 2012-06-20 Genentech, Inc. Binding polypeptides with diversified and consensus VH/VL hypervariable sequences
GB2432366B (en) * 2005-11-19 2007-11-21 Alligator Bioscience Ab A method for in vitro molecular evolution of protein function
KR20180058863A (en) 2005-11-30 2018-06-01 애브비 인코포레이티드 Monoclonal antibodies against amyloid beta protein and uses thereof
RU2442793C2 (en) * 2005-11-30 2012-02-20 Эбботт Лэборетриз ANTISUBSTANCES AGAINST GLOBULOMER Aβ, THEIR ANTIGEN-BINDING PARTS, CORRESPONDING HYBRIDOMAS, NUCLEIC ACIDS, VECTORS, HOST CELLS, WAYS OF PRODUCTION OF MENTIONED ANTISUBSTANCES, COMPOSITIONS CONTAINING MENTIONED ANTISUBSTANCES, APPLICATION OF MENTIONED ANTISUBSTANCES AND WAYS OF APPLICATION OF MENTIONED ANTISUBSTANCES
US20070237764A1 (en) * 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
RS60616B1 (en) 2005-12-29 2020-09-30 Janssen Biotech Inc Human anti-il-23 antibodies, compositions, method and uses
EP2216403A3 (en) 2006-02-02 2010-11-24 Verenium Corporation Esterases and related nucleic acids and methods
WO2007092313A2 (en) * 2006-02-03 2007-08-16 Indiana University Research And Technology Corporation Construction of open reading frame libraries and protein structures
EP1989301B1 (en) 2006-02-10 2014-09-24 BP Corporation North America Inc. Cellulolytic enzymes, nucleic acids encoding them and methods for making and using them
USRE45660E1 (en) 2006-02-14 2015-09-01 Bp Corporation North America Inc. Xylanases, nucleic acids encoding them and methods for making and using them
BRPI0708614A2 (en) 2006-03-07 2011-06-07 Verenium Corp aldolases, nucleic acids encoding the same and methods for producing and using the same
EP2316962B1 (en) 2006-03-07 2014-07-09 Cargill, Incorporated Aldolases, nucleic acids encoding them and methods for making and using them
MX2008011323A (en) * 2006-03-08 2008-11-18 Archemix Corp Complement binding aptamers and anti-c5 agents useful in the treatment of ocular disorders.
EP2614839A3 (en) 2006-04-05 2015-01-28 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
EP2016101A2 (en) * 2006-05-09 2009-01-21 Genentech, Inc. Binding polypeptides with optimized scaffolds
AT503861B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING T-CELL RECEPTORS
AT503889B1 (en) * 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F MULTIVALENT IMMUNE LOBULINE
AT503902B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING IMMUNE LOBULINS
ATE524493T1 (en) 2006-07-24 2011-09-15 Biorexis Pharmaceutical Corp EXENDIN FUSION PROTEINS
CA2669453C (en) 2006-08-04 2018-11-13 Verenium Corporation Glucanases, nucleic acids encoding them and methods for making and using them
WO2008021290A2 (en) 2006-08-09 2008-02-21 Homestead Clinical Corporation Organ-specific proteins and methods of their use
EP3339445B1 (en) * 2006-09-08 2020-07-15 AbbVie Bahamas Ltd. Interleukin -13 binding proteins
US20100297103A1 (en) 2006-09-14 2010-11-25 Medical & Biological Laboratories Co., Ltd. Antibody having enhanced adcc activity and method for production thereof
PT2617823E (en) 2006-09-21 2015-11-23 Basf Enzymes Llc Phytases, nucleic acids encoding them and methods for making and using them
ES2451266T3 (en) 2006-09-21 2014-03-26 Dsm Ip Assets B.V. Phospholipases, nucleic acids that encode them, and methods to obtain and use them
US9382327B2 (en) * 2006-10-10 2016-07-05 Vaccinex, Inc. Anti-CD20 antibodies and methods of use
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EP2479266B1 (en) 2006-12-21 2016-03-09 BASF Enzymes LLC Amylases and glucoamylases, nucleic acids encoding them and methods for making and using them
US7964356B2 (en) 2007-01-16 2011-06-21 Somalogic, Inc. Method for generating aptamers with improved off-rates
US8975026B2 (en) 2007-01-16 2015-03-10 Somalogic, Inc. Method for generating aptamers with improved off-rates
US7947447B2 (en) 2007-01-16 2011-05-24 Somalogic, Inc. Method for generating aptamers with improved off-rates
US7855054B2 (en) 2007-01-16 2010-12-21 Somalogic, Inc. Multiplexed analyses of test samples
US20110136099A1 (en) 2007-01-16 2011-06-09 Somalogic, Inc. Multiplexed Analyses of Test Samples
CA2675366A1 (en) * 2007-01-18 2008-07-24 University Of Southern California Gene polymorphisms predictive for dual tki therapy
AU2008210495B2 (en) 2007-01-30 2014-02-27 Bp Corporation North America, Inc. Enzymes for the treatment of lignocellulosics, nucleic acids encoding them and methods for making and using them
US8143046B2 (en) 2007-02-07 2012-03-27 Danisco Us Inc., Genencor Division Variant Buttiauxella sp. phytases having altered properties
US20090075378A1 (en) 2007-02-20 2009-03-19 Anaptysbio, Inc. Somatic hypermutation systems
US7875431B2 (en) 2007-02-22 2011-01-25 Genentech, Inc. Methods for detecting inflammatory bowel disease
WO2008104386A2 (en) * 2007-02-27 2008-09-04 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
PE20090329A1 (en) * 2007-05-30 2009-03-27 Abbott Lab HUMANIZED ANTIBODIES AGAINST GLOBULOMER AB (20-42) AND ITS USES
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
US20090203766A1 (en) * 2007-06-01 2009-08-13 Archemix Corp. vWF aptamer formulations and methods for use
WO2009000006A1 (en) 2007-06-26 2008-12-31 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H. Display of binding agents
CN103627792A (en) 2007-07-17 2014-03-12 私募蛋白质体公司 Multiplexed analyses of test samples
EP2033971A1 (en) * 2007-09-06 2009-03-11 Abbott GmbH & Co. KG Bone Morphogenetic Protein (BMP) binding domains of proteins of the Repulsive Guidance Molecule (RGM) protein family and functional fragments thereof and their application
US8877688B2 (en) 2007-09-14 2014-11-04 Adimab, Llc Rationally designed, synthetic antibody libraries and uses therefor
EP3753947A1 (en) 2007-09-14 2020-12-23 Adimab, LLC Rationally designed, synthetic antibody libraries and uses therefor
CA2697583C (en) 2007-09-28 2016-04-12 Portola Pharmaceuticals, Inc. Antidotes for factor xa inhibitors and methods of using the same
US8268783B2 (en) 2007-09-28 2012-09-18 Portola Pharmaceuticals, Inc. Antidotes for factor Xa inhibitors and methods of using the same
NZ584967A (en) 2007-10-03 2012-08-31 Verenium Corp Xylanases, nucleic acids encoding them and methods for making and using them
AU2008317374B2 (en) 2007-10-23 2015-03-19 The Regents Of The University Of Colorado Competitive inhibitors of invariant chain expression and/or ectopic clip binding
EP2214708A4 (en) 2007-10-26 2011-01-12 Centocor Ortho Biotech Inc Vectors, host cells, and methods of production and uses
US8906700B2 (en) 2007-11-06 2014-12-09 Ambergen, Inc. Methods and compositions for phototransfer
EP2242843B1 (en) * 2007-12-31 2015-05-27 XOMA Technology Ltd. Methods and materials for targeted mutagenesis
CN101965397B (en) 2008-01-03 2016-09-28 巴斯夫酶有限责任公司 Transferring enzyme and oxidoreductase, the nucleic acid encoding them and its methods for making and using same
WO2009088753A1 (en) 2008-01-03 2009-07-16 Verenium Corporation Isomerases, nucleic acids encoding them and methods for making and using them
US7682809B2 (en) 2008-01-11 2010-03-23 Agilent Technologies, Inc. Direct ATP release sequencing
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US9873957B2 (en) 2008-03-13 2018-01-23 Dyax Corp. Libraries of genetic packages comprising novel HC CDR3 designs
CA2968164C (en) 2008-04-24 2019-08-20 Dyax Corp. Libraries of genetic packages comprising novel hc cdr1, cdr2, and cdr3 and novel lc cdr1, cdr2, and cdr3 designs
US20100260668A1 (en) * 2008-04-29 2010-10-14 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
DK2281034T3 (en) 2008-04-30 2016-01-25 Dupont Nutrition Biosci Aps A method using alcohol dehydrogenase Pseudoglucanobacter saccharoketogenes
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
CA2723219A1 (en) 2008-05-09 2009-11-12 Abbott Gmbh & Co. Kg Antibodies to receptor of advanced glycation end products (rage) and uses thereof
CN102112494A (en) 2008-06-03 2011-06-29 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
EP3002299A1 (en) 2008-06-03 2016-04-06 AbbVie Inc. Dual variable domain immunoglobulins and uses thereof
AU2009259959A1 (en) 2008-06-20 2009-12-23 Wyeth Llc Compositions and methods of use of ORF1358 from beta-hemolytic streptococcal strains
CA2728473A1 (en) * 2008-06-20 2009-12-23 National University Corporation Okayama University Antibody against oxidized ldl/.beta.2gpi complex and use of the same
JP5674654B2 (en) * 2008-07-08 2015-02-25 アッヴィ・インコーポレイテッド Prostaglandin E2 double variable domain immunoglobulin and use thereof
BRPI0915825A2 (en) 2008-07-08 2015-11-03 Abbott Lab prostaglandin binding proteins and uses thereof
US8703416B2 (en) 2008-07-17 2014-04-22 Somalogic, Inc. Method for purification and identification of sperm cells
WO2010011315A2 (en) * 2008-07-25 2010-01-28 Viral Genetics, Inc. Proteins for use in diagnosing and treating infection and disease
US20100166782A1 (en) * 2008-07-25 2010-07-01 Martha Karen Newell Clip inhibitors and methods of modulating immune function
US9182406B2 (en) * 2008-08-04 2015-11-10 Biodesy, Inc. Nonlinear optical detection of molecules comprising an unnatural amino acid possessing a hyperpolarizability
AU2009281713B9 (en) 2008-08-14 2014-05-29 Teva Pharmaceuticals Australia Pty Ltd Anti-IL-12/IL-23 antibodies
US8153391B2 (en) 2008-08-29 2012-04-10 Bunge Oils, Inc. Hydrolases, nucleic acids encoding them and methods for making and using them
US8357503B2 (en) 2008-08-29 2013-01-22 Bunge Oils, Inc. Hydrolases, nucleic acids encoding them and methods for making and using them
US8198062B2 (en) 2008-08-29 2012-06-12 Dsm Ip Assets B.V. Hydrolases, nucleic acids encoding them and methods for making and using them
EP2360172B1 (en) 2008-10-20 2017-10-18 Gwangju Institute of Science and Technology Bipodal peptide binder
CA2745271A1 (en) * 2008-12-04 2010-06-10 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
US8492133B2 (en) 2009-01-20 2013-07-23 Ramot At Tel Aviv University, Ltd. MIR-21 promoter driven targeted cancer therapy
PE20120586A1 (en) * 2009-01-29 2012-06-17 Abbott Lab IL-1 BINDING PROTEINS
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
US8030026B2 (en) 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof
EP2810652A3 (en) 2009-03-05 2015-03-11 AbbVie Inc. IL-17 binding proteins
GB0908770D0 (en) 2009-04-24 2009-07-01 Danisco Method
EP2698374B1 (en) 2009-05-21 2016-09-14 BASF Enzymes LLC Phytases, nucleic acids encoding them and methods for making and using them
CA2767858C (en) 2009-07-15 2019-02-12 Portola Pharmaceuticals, Inc. Unit dose formulation of antidotes for factor xa inhibitors and methods of using the same
TW201109438A (en) * 2009-07-29 2011-03-16 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US8623358B2 (en) 2009-08-29 2014-01-07 Abbvie Inc. Therapeutic DLL4 binding proteins
EP2473524A4 (en) 2009-09-01 2013-05-22 Abbott Lab Dual variable domain immunoglobulins and uses thereof
AU2010291902A1 (en) * 2009-09-14 2012-04-05 Dyax Corp. Libraries of genetic packages comprising novel HC CDR3 designs
EP3187877A1 (en) 2009-09-25 2017-07-05 XOMA Technology Ltd. Screening methods
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
KR20140015139A (en) 2009-10-15 2014-02-06 애브비 인코포레이티드 Dual variable domain immunoglobulins and uses thereof
UA111708C2 (en) 2009-10-16 2016-06-10 Бандж Ойлз, Інк. METHOD OF OIL REFINING
UA109884C2 (en) 2009-10-16 2015-10-26 A POLYPEPTIDE THAT HAS THE ACTIVITY OF THE PHOSPHATIDYLINOSYTOL-SPECIFIC PHOSPHOLIPASE C, NUCLEIC ACID, AND METHOD OF METHOD
WO2011050194A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
UY32979A (en) * 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
WO2011053707A1 (en) * 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
JP5856065B2 (en) 2009-11-30 2016-02-09 ジェネンテック, インコーポレイテッド Compositions and methods for tumor diagnosis and treatment
SG181563A1 (en) 2009-12-08 2012-07-30 Abbott Gmbh & Co Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration
CN102933231B (en) 2010-02-10 2015-07-29 伊缪诺金公司 CD20 antibody and uses thereof
JP5981853B2 (en) 2010-02-18 2016-08-31 ジェネンテック, インコーポレイテッド Neuregulin antagonists and their use in the treatment of cancer
JP5988436B2 (en) 2010-02-23 2016-09-07 ジェネンテック, インコーポレイテッド Compositions and methods for tumor diagnosis and treatment
MY171234A (en) 2010-02-24 2019-10-04 Immunogen Inc Folate receptor 1 antibodies and immunoconjugates and uses thereof
EP2542582A4 (en) 2010-03-02 2013-12-04 Abbvie Inc Therapeutic dll4 binding proteins
AU2011223527B2 (en) 2010-03-03 2014-11-13 Somalogic Operating Co., Inc. Aptamers to 4-1BB and their use in treating diseases and disorders
DK2552961T3 (en) 2010-03-30 2018-03-05 Janssen Biotech Inc HUMANIZED IL-25 ANTIBODIES
WO2011130195A1 (en) 2010-04-12 2011-10-20 Somalogic, Inc. APTAMERS TO β-NGF AND THEIR USE IN TREATING β-NGF MEDIATED DISEASES AND DISORDERS
ES2684475T3 (en) 2010-04-15 2018-10-03 Abbvie Inc. Proteins that bind to beta amyloid
KR20130079384A (en) 2010-05-03 2013-07-10 제넨테크, 인크. Compositions and methods for the diagnosis and treatment of tumor
AU2011252883B2 (en) 2010-05-14 2015-09-10 Abbvie Inc. IL-1 binding proteins
GB201011513D0 (en) 2010-07-08 2010-08-25 Danisco Method
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
MX360336B (en) 2010-07-16 2018-10-30 Adimab Llc Star Antibody libraries.
CN103298834A (en) 2010-08-03 2013-09-11 Abbvie公司 Dual variable domain immunoglobulins and uses thereof
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
DK3333188T3 (en) 2010-08-19 2022-03-07 Zoetis Belgium S A Anti-NGF antibodies and their use
BR112013004581A2 (en) 2010-08-26 2017-06-27 Abbvie Inc dual variable domain immunoglobulins and their uses
CA2811789A1 (en) 2010-10-06 2012-04-12 Bp Corporation North America Inc. Variant cbh i polypeptides with reduced product inhibition
KR20130133247A (en) 2010-12-21 2013-12-06 애브비 인코포레이티드 Il-1-alpha and -beta bispecific dual variable domain immunoglobulins and their use
US20120275996A1 (en) 2010-12-21 2012-11-01 Abbott Laboratories IL-1 Binding Proteins
ES2899186T3 (en) 2010-12-31 2022-03-10 Bioatla Inc Rapid antibody humanization
CA2831136A1 (en) 2011-03-21 2012-09-27 Biodesy, Llc Classification of kinase inhibitors using nonlinear optical techniques
EP3252155A1 (en) 2011-04-21 2017-12-06 The Rockefeller University Streptococcus bacteriophage lysins for detection and treatment of gram positive bacteria
EP2734236A4 (en) 2011-07-13 2015-04-15 Abbvie Inc Methods and compositions for treating asthma using anti-il-13 antibodies
WO2013016449A2 (en) 2011-07-26 2013-01-31 Indicator Systems International, Inc. Assays for the detection of microbes
JP2014533927A (en) 2011-09-15 2014-12-18 ジェネンテック, インコーポレイテッド How to promote differentiation
CN104114695B (en) 2011-10-05 2018-04-17 洛克菲勒大学 Dimerization bacteriophage lysin
CA2850836A1 (en) 2011-10-15 2013-04-18 Genentech, Inc. Methods of using scd1 antagonists
WO2013063095A1 (en) 2011-10-24 2013-05-02 Abbvie Inc. Immunobinders directed against sclerostin
BR112014012539B1 (en) 2011-11-23 2022-12-20 Medimmune, Llc ANTIBODY SPECIFICALLY BINDING HER3, COMPOSITION COMPRISING THE SAME, AND USES OF THE ANTIBODY
CA2855840C (en) 2011-12-14 2023-08-29 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
EP2791175A2 (en) 2011-12-14 2014-10-22 Abbvie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
AR089529A1 (en) 2011-12-30 2014-08-27 Abbvie Inc UNION PROTEINS SPECIFIC DUALS DIRECTED AGAINST IL-13 AND / OR IL-17
CN104168920A (en) 2012-01-18 2014-11-26 霍夫曼-拉罗奇有限公司 Methods of using FGF19 modulators
PT2804878T (en) 2012-01-20 2018-11-29 Genzyme Corp Anti-cxcr3 antibodies
RU2644337C2 (en) 2012-01-27 2018-02-08 Эббви Дойчланд Гмбх Унд Ко. Кг Compositions and methods for diagnostics and treatment of diseases associated with neurites degeneration
US20130209473A1 (en) 2012-02-11 2013-08-15 Genentech, Inc. R-spondin translocations and methods using the same
NZ630020A (en) 2012-03-08 2016-08-26 Halozyme Inc Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
BR112014020173A8 (en) 2012-03-16 2017-07-11 Hoffmann La Roche METHODS FOR THE TREATMENT OF A MELANOMA, USES OF AN INHIBITOR, COMPOSITIONS, KIT, METHOD OF INHIBITION, METHOD OF IDENTIFICATION, METHOD OF ADJUSTING THE TREATMENT AND INVENTION
US9139863B2 (en) 2012-03-16 2015-09-22 Genentech, Inc. Engineered conformationally-stabilized proteins
WO2013137920A1 (en) 2012-03-16 2013-09-19 Genentech, Inc. Engineered conformationally-stabilized proteins
JP6200437B2 (en) 2012-03-16 2017-09-20 ユニバーシティ ヘルス ネットワーク Methods and compositions for modulating TOSO activity
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
EP2841951B1 (en) 2012-04-25 2019-12-11 Biodesy, Inc. Methods for detecting allosteric modulators of proteins
WO2013170191A1 (en) 2012-05-11 2013-11-14 Genentech, Inc. Methods of using antagonists of nad biosynthesis from nicotinamide
EP3508497A1 (en) 2012-05-24 2019-07-10 Mountgate Group Limited Compositions and methods related to prevention and treatment of rabies infection
US9617334B2 (en) 2012-06-06 2017-04-11 Zoetis Services Llc Caninized anti-NGF antibodies and methods thereof
WO2014011955A2 (en) 2012-07-12 2014-01-16 Abbvie, Inc. Il-1 binding proteins
NZ726258A (en) 2012-08-31 2019-07-26 Immunogen Inc Antibodies and uses thereof to detect folate receptor 1
PE20151179A1 (en) 2012-11-01 2015-09-12 Abbvie Inc DUAL ANTI-VEGF / DLL4 VARIABLE DOMAIN IMMUNOGLOBULINS AND USES OF THEM
US9550986B2 (en) 2012-12-21 2017-01-24 Abbvie Inc. High-throughput antibody humanization
WO2014128235A1 (en) 2013-02-22 2014-08-28 F. Hoffmann-La Roche Ag Methods of treating cancer and preventing drug resistance
RU2015137610A (en) 2013-03-06 2017-04-10 Дженентек, Инк. METHODS FOR TREATMENT AND PREVENTION OF DRUG RESISTANCE OF MALIGNANT TUMORS
GB201308828D0 (en) 2013-03-12 2013-07-03 Verenium Corp Phytase
GB201308843D0 (en) 2013-03-14 2013-07-03 Verenium Corp Phytase formulation
US9790478B2 (en) 2013-03-14 2017-10-17 Abbott Laboratories HCV NS3 recombinant antigens and mutants thereof for improved antibody detection
US9194873B2 (en) 2013-03-14 2015-11-24 Abbott Laboratories HCV antigen-antibody combination assay and methods and compositions for use therein
CN105378099B (en) 2013-03-14 2021-05-11 雅培制药有限公司 HCV core lipid binding domain monoclonal antibodies
KR20150127216A (en) 2013-03-14 2015-11-16 제넨테크, 인크. Methods of treating cancer and preventing cancer drug resistance
BR112015023797A2 (en) 2013-03-15 2017-10-24 Abbvie Inc dual specificity binding proteins directed against il-1b and / or il-17
BR112015023203A8 (en) 2013-03-15 2018-01-23 Constellation Pharmaceuticals Inc methods for treating cancer, method for increasing the efficiency of cancer treatment, method for delaying and / or preventing cancer development, method for treating an individual with cancer, method for increasing sensitivity for a cancer therapy agent, method for extending a sensitivity period and method for extending the duration of response to cancer therapy.
KR102158924B1 (en) 2013-03-15 2020-09-22 제넨테크, 인크. Biomarkers and methods of treating pd-1 and pd-l1 related conditions
BR112015023709A2 (en) 2013-03-15 2017-07-18 Pioneer Hi Bred Int phi-4 polypeptide, polynucleotide, composition, method for inhibiting growth, method for controlling a population, plant, seed, expression cassette, method for expressing a polynucleotide in a plant, method for protecting a plant, fusion protein
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
US10183988B2 (en) 2013-06-07 2019-01-22 Duke University Anti-Complement factor H antibodies
AR095173A1 (en) 2013-07-25 2015-09-30 Basf Enzymes Llc FITASA
MX359026B (en) 2013-08-16 2018-09-12 Pioneer Hi Bred Int Insecticidal proteins and methods for their use.
KR20230145514A (en) 2013-08-30 2023-10-17 이뮤노젠 아이엔씨 Antibodies and assays for detection of folate receptor 1
CN105764924A (en) 2013-09-12 2016-07-13 哈洛齐梅公司 Modified anti-epidermal growth factor receptor antibodies and methods of use thereof
BR112016005543B1 (en) 2013-09-13 2022-03-08 Pioneer Hi-Bred International, Inc RECOMBINANT PIP-72 POLYPEPTIDE, DNA CONSTRUCTION, METHOD FOR OBTAINING A TRANSGENIC PLANT, HOST CELL, COMPOSITION, FUSION PROTEIN, METHOD FOR CONTROLLING AN INSECT PEST POPULATION, METHOD FOR INHIBITING THE GROWTH OR KILLING AN INSECT PEST, METHOD FOR TO CONTROL AN INSECT INFESTATION IN A TRANSGENIC PLANT, METHOD FOR IDENTIFYING A NUCLEOTIDE SEQUENCE IN A BIOLOGICAL SAMPLE, METHOD FOR IDENTIFYING A PIP-72 POLYPEPTIDE IN A SAMPLE
WO2015097536A2 (en) 2013-12-24 2015-07-02 Janssen Pharmaceutical Nv Anti-vista antibodies and fragments
WO2015116902A1 (en) 2014-01-31 2015-08-06 Genentech, Inc. G-protein coupled receptors in hedgehog signaling
WO2015120270A1 (en) 2014-02-07 2015-08-13 Pioneer Hi Bred International, Inc. Insecticidal proteins and methods for their use
EP3102592B1 (en) 2014-02-07 2020-05-20 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
CA2941693A1 (en) 2014-03-07 2015-09-11 University Health Network Methods and compositions for detection of targets involved in cancer metastasis
DK3128997T3 (en) 2014-04-08 2020-08-24 Boston Pharmaceuticals Inc BINDING MOLECULES SPECIFIC TO IL-21 AND USES THEREOF
CA2943299A1 (en) 2014-04-11 2015-10-15 Medimmune, Llc Bispecific her2 antibodies
GB201406767D0 (en) 2014-04-15 2014-05-28 Cancer Rec Tech Ltd Humanized anti-Tn-MUC1 antibodies anf their conjugates
EP3636073B1 (en) 2014-05-05 2023-11-15 Regeneron Pharmaceuticals, Inc. Humanized c5 and c3 animals
KR20170005016A (en) 2014-05-23 2017-01-11 제넨테크, 인크. Mit biomarkers and methods using the same
US20230190750A1 (en) 2014-06-13 2023-06-22 Genentech, Inc. Methods of treating and preventing cancer drug resistance
CN114057857A (en) 2014-06-20 2022-02-18 豪夫迈·罗氏有限公司 CHAGASIN-based scaffold compositions, methods and uses
WO2016061206A1 (en) 2014-10-16 2016-04-21 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
EP3209773B1 (en) 2014-10-24 2020-03-18 DuPont Nutrition Biosciences ApS Proline tolerant tripeptidyl peptidases and uses thereof
EP3172329A1 (en) 2014-10-24 2017-05-31 Danisco US Inc. Method for producing alcohol by use of a tripeptidyl peptidase
DK3218406T3 (en) 2014-11-10 2021-06-21 Medimmune Ltd BINDING MOLECULES SPECIFIC TO CD73 AND USES THEREOF
US20160129108A1 (en) 2014-11-11 2016-05-12 Medimmune Limited Therapeutic combinations comprising anti-cd73 antibodies and uses thereof
WO2016094881A2 (en) 2014-12-11 2016-06-16 Abbvie Inc. Lrp-8 binding proteins
EP3237906B8 (en) 2014-12-23 2020-10-28 Bluelight Therapeutics, Inc. Attachment of proteins to interfaces for use in nonlinear optical detection
SG10201906471PA (en) 2015-01-14 2019-09-27 Brigham & Womens Hospital Inc Treatment of cancer with anti-lap monoclonal antibodies
EA038923B1 (en) 2015-03-11 2021-11-10 Пайонир Хай-Бред Интернэшнл, Инк. Insecticidal dna construct and methods of use thereof
JP6952605B2 (en) 2015-04-24 2021-10-20 ジェネンテック, インコーポレイテッド Multispecific antigen binding protein
CN108064233B (en) 2015-05-19 2022-07-15 先锋国际良种公司 Insecticidal proteins and methods of use thereof
SI3303395T1 (en) 2015-05-29 2020-03-31 Abbvie Inc. Anti-cd40 antibodies and uses thereof
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
EP3313882B1 (en) 2015-06-24 2020-03-11 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
US11473073B2 (en) 2015-06-26 2022-10-18 Dupont Nutrition Biosciences Aps Aminopeptidases for protein hydrolyzates
EP3313884B9 (en) 2015-06-29 2021-05-19 ImmunoGen, Inc. Anti-cd123 antibodies and conjugates and derivatives thereof
US10287338B2 (en) 2015-07-10 2019-05-14 Miran NERSISSIAN Factor VIII protein compositions and methods of treating of hemophilia A
WO2017023863A1 (en) 2015-07-31 2017-02-09 Research Institute At Nationwide Children's Hospital Peptides and antibodies for the removal of biofilms
EP3943602A1 (en) 2015-08-06 2022-01-26 Pioneer Hi-Bred International, Inc. Plant derived insecticidal proteins and methods for their use
EP3365027B1 (en) 2015-10-14 2022-03-30 Research Institute at Nationwide Children's Hospital Hu specific antibodies and their use in inhibiting biofilm
WO2017083451A1 (en) 2015-11-10 2017-05-18 Medimmune, Llc Binding molecules specific for asct2 and uses thereof
WO2017105987A1 (en) 2015-12-18 2017-06-22 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
EP3405587A4 (en) 2015-12-23 2019-10-30 Moonshot Pharma LLC Methods for inducing an immune response by promoting premature termination codon read-through
MX2018009800A (en) 2016-02-12 2018-11-09 Janssen Pharmaceutica Nv Anti-vista (b7h5) antibodies.
US20210120845A1 (en) 2016-02-25 2021-04-29 Dupont Nutrition Biosciences Aps Method for producing a protein hydrolysate employing an aspergillus fumigatus tripeptidyl peptidase
AU2017230091B2 (en) 2016-03-10 2022-04-07 Acceleron Pharma Inc. Activin type 2 receptor binding proteins and uses thereof
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
EA201892190A1 (en) 2016-03-29 2019-04-30 Янссен Байотек, Инк. TREATMENT OF PSORIASIS WITH ANTI-BODY TO IL-12 AND / OR IL-23 WITH THE GROWTH OF THE INTERVAL BETWEEN THE INTRODUCTION OF DOSE
WO2017175058A1 (en) 2016-04-07 2017-10-12 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
KR102460040B1 (en) 2016-04-27 2022-11-01 애브비 인코포레이티드 Method for treating diseases in which IL-13 activity is detrimental using anti-IL-13 antibody
WO2017192560A1 (en) 2016-05-04 2017-11-09 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
CA3025377A1 (en) 2016-06-02 2017-12-07 Abbvie Inc. Glucocorticoid receptor agonist and immunoconjugates thereof
WO2017214339A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
WO2017214322A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
CN116284404A (en) 2016-06-08 2023-06-23 艾伯维公司 anti-B7-H3 antibodies and antibody drug conjugates
GB201610198D0 (en) 2016-06-10 2016-07-27 Ucb Biopharma Sprl Anti-ige antibodies
WO2017215790A1 (en) 2016-06-17 2017-12-21 National Hellenic Research Foundation Systems for recombinant protein production
CA3026113A1 (en) 2016-07-01 2018-01-04 Pioneer Hi-Bred International, Inc. Insecticidal proteins from plants and methods for their use
WO2018049275A1 (en) 2016-09-09 2018-03-15 Genentech, Inc. Selective peptide inhibitors of frizzled
CA3037961A1 (en) 2016-09-30 2018-04-05 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il23 specific antibody
US20180106800A1 (en) 2016-10-03 2018-04-19 Abbott Laboratories Methods of assessing uch-l1 status in patient samples
WO2018084936A1 (en) 2016-11-01 2018-05-11 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
MA46861A (en) 2016-11-16 2019-09-25 Janssen Biotech Inc METHOD OF TREATING PSORIASIS WITH A SPECIFIC ANTI-IL-23 ANTIBODY
JP7274417B2 (en) 2016-11-23 2023-05-16 イミュノア・セラピューティクス・インコーポレイテッド 4-1BB binding protein and uses thereof
CN110662770A (en) 2016-11-23 2020-01-07 比奥维拉迪维治疗股份有限公司 Bispecific antibodies that bind factor IX and factor X
CN110088123B (en) 2016-12-14 2023-10-20 先锋国际良种公司 Insecticidal proteins and methods of use thereof
WO2018118811A1 (en) 2016-12-22 2018-06-28 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
US20180230218A1 (en) 2017-01-04 2018-08-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
CA3049114A1 (en) 2017-01-04 2018-07-12 Lauren O. Bakaletz Antibody fragments for the treatment of biofilm-related disorders
EP3565589A1 (en) 2017-01-04 2019-11-13 Research Institute at Nationwide Children's Hospital Dnabii vaccines and antibodies with enhanced activity
WO2018127791A2 (en) 2017-01-06 2018-07-12 Biosion, Inc. Erbb2 antibodies and uses therefore
MX2019008989A (en) 2017-01-30 2019-10-09 Janssen Biotech Inc Anti-tnf antibodies, compositions, and methods for the treatment of active psoriatic arthritis.
MX2019009377A (en) 2017-02-07 2019-12-11 Janssen Biotech Inc Anti-tnf antibodies, compositions, and methods for the treatment of active ankylosing spondylitis.
MX2019009371A (en) 2017-02-08 2019-09-23 Pionner Hi Bred Int Inc Insecticidal combinations of plant derived insecticidal proteins and methods for their use.
WO2018152496A1 (en) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions and methods for the diagnosis and treatment of zika virus infection
US10765762B2 (en) 2017-02-27 2020-09-08 Regeneron Pharmaceuticals, Inc. Humanized model of kidney and liver disorders
WO2018170178A1 (en) 2017-03-15 2018-09-20 Research Institute At Nationwide Children's Hospital Composition and methods for disruption of bacterial biofilms without accompanying inflammation
CA3052513A1 (en) 2017-03-23 2018-09-27 Abbott Laboratories Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in a human subject using the early biomarker ubiquitin carboxy-terminal hydrolase l1
EP3610268A1 (en) 2017-04-15 2020-02-19 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury in a human subject using early biomarkers
US20190078160A1 (en) 2017-04-21 2019-03-14 Genentech, Inc. Use of klk5 antagonists for treatment of a disease
US11236151B2 (en) 2017-04-25 2022-02-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies and methods for the diagnosis and treatment of Epstein Barr virus infection
JP7080899B2 (en) 2017-04-28 2022-06-06 アボット・ラボラトリーズ Methods to aid in the hyperacute diagnosis and determination of traumatic brain injury using early biomarkers on at least two samples from the same human subject
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
CA3063200A1 (en) 2017-05-11 2018-11-15 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
CA3078725A1 (en) 2017-05-25 2018-11-29 Abbott Laboratories Methods for aiding in the determination of whether to perform imaging on a human subject who has sustained or may have sustained an injury to the head using early biomarkers
AU2018275236A1 (en) 2017-05-30 2019-10-31 Abbott Laboratories Methods for aiding in diagnosing and evaluating a mild traumatic brain injury in a human subject using cardiac troponin I
US11654135B2 (en) 2017-06-22 2023-05-23 Moonshot Pharma Llc Methods for treating colon cancer with compositions comprising amlexanox and immune checkpoint inhibitors
US11169159B2 (en) 2017-07-03 2021-11-09 Abbott Laboratories Methods for measuring ubiquitin carboxy-terminal hydrolase L1 levels in blood
CN111094334A (en) 2017-07-19 2020-05-01 美国卫生与公众服务部 Antibodies and methods for diagnosis and treatment of hepatitis B virus infection
CN110945023B (en) 2017-07-31 2023-08-18 豪夫迈·罗氏有限公司 Humanization method based on three-dimensional structure
TW201922780A (en) 2017-09-25 2019-06-16 美商健生生物科技公司 Safe and effective method of treating Lupus with anti-IL12/IL23 antibody
WO2019070161A2 (en) 2017-10-04 2019-04-11 Opko Pharmaceuticals, Llc Articles and methods directed to personalized therapy of cancer
EP3694552A1 (en) 2017-10-10 2020-08-19 Tilos Therapeutics, Inc. Anti-lap antibodies and uses thereof
WO2019094595A2 (en) 2017-11-09 2019-05-16 Pinteon Therapeutics Inc. Methods and compositions for the generation and use of humanized conformation-specific phosphorylated tau antibodies
AU2018378084A1 (en) 2017-12-09 2020-05-14 Abbott Laboratories Methods for aiding in diagnosing and evaluating a traumatic brain injury in a human subject using a combination of GFAP and UCH-L1
BR112019028254A2 (en) 2017-12-09 2020-07-14 Abbott Laboratories methods to assist in the diagnosis and evaluation of a patient who has suffered an orthopedic injury and who has suffered or may have suffered a head injury, such as a mild traumatic brain injury (lct), using glial fibrillar acid protein (gfap) and / or the carboxy-terminal hydrolase of ubiquitin l1 (uch-l1)
WO2019126472A1 (en) 2017-12-22 2019-06-27 Genentech, Inc. Use of pilra binding agents for treatment of a disease
EP3732489A1 (en) 2017-12-29 2020-11-04 Abbott Laboratories Novel biomarkers and methods for diagnosing and evaluating traumatic brain injury
WO2019150309A1 (en) 2018-02-02 2019-08-08 Hammack Scott Modulators of gpr68 and uses thereof for treating and preventing diseases
MX2020008479A (en) 2018-02-14 2020-11-24 Viela Bio Inc Antibodies to feline mcdonough sarcoma (fms)-like tyrosine kinase 3 receptor ligand (flt3l) and uses thereof for treating autoimmune and inflammatory diseases.
MA54132A (en) 2018-03-05 2022-04-27 Janssen Biotech Inc METHODS OF TREATING CROHN'S DISEASE WITH SPECIFIC ANTI-IL23 ANTIBODY
CN111819197A (en) 2018-03-12 2020-10-23 硕腾服务有限责任公司 anti-NGF antibodies and methods thereof
WO2019213619A1 (en) 2018-05-04 2019-11-07 Abbott Laboratories Hbv diagnostic, prognostic, and therapeutic methods and products
BR112020023407A2 (en) 2018-05-17 2021-02-09 Bp Corporation North America Inc. production of 2-keto-3-deoxy-d-gluconic acid in filamentous fungi
EP3801010A4 (en) 2018-06-08 2022-02-23 Crystal Bioscience Inc. Transgenic animal for producing diversified antibodies that have the same light chain i
TW202016144A (en) 2018-06-21 2020-05-01 日商第一三共股份有限公司 Compositions including cd3 antigen binding fragments and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
JP2021530697A (en) 2018-07-18 2021-11-11 ヤンセン バイオテツク,インコーポレーテツド Sustained response predictor after treatment with anti-IL23 specific antibody
US11548938B2 (en) 2018-08-21 2023-01-10 Quidel Corporation DbpA antibodies and uses thereof
US20230183728A1 (en) 2018-09-07 2023-06-15 Basf Plant Science Company Gmbh Improved method for the production of high levels of pufa in plants
CN113412332A (en) 2018-09-07 2021-09-17 巴斯夫植物科学有限公司 Improved methods for producing high levels of PUFAs in plants
CA3110656A1 (en) 2018-09-07 2020-03-12 Basf Plant Science Company Gmbh Improved method for the production of high levels of pufa in plants
US10961307B2 (en) 2018-09-24 2021-03-30 Janssen Biotech, Inc. Methods of treating moderately to severely active ulcerative colitis by administering an anti-IL12/IL23 antibody
CA3114925A1 (en) 2018-10-05 2020-04-09 Research Institute At Nationwide Children's Hospital Compositions and methods for enzymatic disruption of bacterial biofilms
EP3863722A2 (en) 2018-10-10 2021-08-18 Tilos Theapeutics, Inc. Anti-lap antibody variants and uses thereof
AU2019383017A1 (en) 2018-11-20 2021-06-03 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-IL-23 specific antibody
WO2020106358A1 (en) 2018-11-20 2020-05-28 Takeda Vaccines, Inc. Novel anti-zika virus antibodies and uses thereof
US20200197517A1 (en) 2018-12-18 2020-06-25 Janssen Biotech, Inc. Safe and Effective Method of Treating Lupus with Anti-IL12/IL23 Antibody
EP3897117A1 (en) 2018-12-21 2021-10-27 Compass Therapeutics LLC Transgenic mouse expressing common human light chain
CN113574066A (en) 2019-01-15 2021-10-29 詹森生物科技公司 anti-TNF antibody compositions and methods for treating juvenile idiopathic arthritis
EA202192038A1 (en) 2019-01-23 2021-10-05 Янссен Байотек, Инк. ANTIBODY COMPOSITIONS TO TNF FOR USE IN METHODS FOR TREATMENT OF PSORIATIC ARTHRITIS
US20220144934A1 (en) 2019-03-14 2022-05-12 Janssen Biotech, Inc. Methods for Producing Anti-TNF Antibody Compositions
JP2022524860A (en) 2019-03-14 2022-05-10 ヤンセン バイオテツク,インコーポレーテツド Methods for Producing Anti-TNF Antibody Compositions
EP3938384A4 (en) 2019-03-14 2022-12-28 Janssen Biotech, Inc. Manufacturing methods for producing anti-il12/il23 antibody compositions
US20220153830A1 (en) 2019-03-14 2022-05-19 Janssen Biotech, Inc. Manufacturing Methods for Producing Anti-TNF Antibody Compositions
MA55383A (en) 2019-03-18 2022-01-26 Janssen Biotech Inc METHOD OF TREATING PSORIASIS IN PEDIATRIC SUBJECTS WITH AN ANTI-IL12/IL23 ANTIBODY
WO2020227554A1 (en) 2019-05-09 2020-11-12 Genentech, Inc. Methods of making antibodies
CN113874073A (en) 2019-05-23 2021-12-31 詹森生物科技公司 Methods of treating inflammatory bowel disease with combination therapy of antibodies to IL-23 and TNF α
WO2020245677A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active ankylosing spondylitis
CA3142665A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibody compositions, and methods for the treatment of psoriatic arthritis
MX2022000137A (en) 2019-07-08 2022-05-19 Res Inst Nationwide Childrens Hospital Antibody compositions for disrupting biofilms.
WO2021028752A1 (en) 2019-08-15 2021-02-18 Janssen Biotech, Inc. Anti-tfn antibodies for treating type i diabetes
CN115298213A (en) 2019-12-19 2022-11-04 奎多公司 Monoclonal antibody fusion
KR20220119408A (en) 2019-12-20 2022-08-29 바스프 에스이 Reduced toxicity of terpenes and increased production potential in microorganisms
CN115397848A (en) 2020-02-05 2022-11-25 拉利玛生物医药公司 TAT peptide binding proteins and uses thereof
US20230272056A1 (en) 2020-04-09 2023-08-31 Merck Sharp & Dohme Llc Affinity matured anti-lap antibodies and uses thereof
EP4136459A1 (en) 2020-04-13 2023-02-22 Abbott Laboratories Methods, complexes and kits for detecting or determining an amount of a ss-coronavirus antibody in a sample
US20210338833A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Chimeric antigen receptor-targeting ligands and uses thereof
US20210340524A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Methods for identifying chimeric antigen receptor-targeting ligands and uses thereof
BR112022024475A2 (en) 2020-06-04 2022-12-27 Isobionics B V SANTALENE BETA SYNTHETIC SYNTHASE, SYNTHETIC NUCLEIC ACID, EXPRESSION CASSETTE, METHOD FOR PRODUCING A COMPOSITION, NON-HUMAN HOST CELL, COMPOSITION, AND, USE OF ANY OF THE SYNTHETIC SANTALENE SYNTHESES
EP4182466A2 (en) 2020-07-14 2023-05-24 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
WO2022031804A1 (en) 2020-08-04 2022-02-10 Abbott Laboratories Improved methods and kits for detecting sars-cov-2 protein in a sample
US11988671B2 (en) 2020-08-04 2024-05-21 Abbott Rapid Diagnostics International Unlimited Company Assays for detecting SARS-CoV-2
EP4211166A1 (en) 2020-09-11 2023-07-19 MedImmune Limited Therapeutic b7-h4 binding molecules
KR20230066095A (en) 2020-09-12 2023-05-12 메디뮨 리미티드 Scoring Methods for Anti-B7H4 Antibody-Drug Conjugate Therapy
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
US20220170948A1 (en) 2020-12-01 2022-06-02 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a human subject having received a head computerized tomography scan that is negative for a tbi
EP4015626A1 (en) 2020-12-18 2022-06-22 Isobionics B.V. Enzymes and methods for fermentative production of monoterpene esters
WO2022147147A1 (en) 2020-12-30 2022-07-07 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
WO2022153194A1 (en) 2021-01-13 2022-07-21 Memorial Sloan Kettering Cancer Center Antibody-pyrrolobenzodiazepine derivative conjugate
TW202245844A (en) 2021-01-13 2022-12-01 紀念斯隆凱特琳癌症中心 Anti-dll3 antibody-drug conjugate
AR124681A1 (en) 2021-01-20 2023-04-26 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
BR112023018400A2 (en) 2021-03-12 2023-12-12 Janssen Biotech Inc METHOD FOR TREATMENT OF PSORIATIC ARTHRITIS PATIENTS WITH INADEQUATE RESPONSE TO TNF THERAPY WITH SPECIFIC ANTI-IL23 ANTIBODY
AU2022233792A1 (en) 2021-03-12 2023-10-26 Janssen Biotech, Inc. Safe and effective method of treating psoriatic arthritis with anti-il23 specific antibody
WO2022197782A1 (en) 2021-03-17 2022-09-22 Receptos Llc Methods of treating atopic dermatitis with anti il-13 antibodies
CN116997571A (en) 2021-03-18 2023-11-03 免疫医疗有限公司 Therapeutic binding molecules that bind CCR9
WO2022241057A1 (en) 2021-05-12 2022-11-17 Applied Biomedical Science Institute Binding polypeptides against sars cov-2 and uses thereof
BR112023024169A2 (en) 2021-05-18 2024-02-06 Abbott Lab METHODS FOR ASSESSING BRAIN INJURY IN A PEDIATRIC INDIVIDUAL
EP4340875A1 (en) 2021-05-20 2024-03-27 Janssen Biotech, Inc. Method of treating inflammatory bowel disease with a combination therapy of antibodies to il-23 and tnf alpha
EP4356129A1 (en) 2021-06-14 2024-04-24 Abbott Laboratories Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
KR20240032991A (en) 2021-07-09 2024-03-12 얀센 바이오테크 인코포레이티드 Manufacturing Methods for Producing Anti-TNF Antibody Compositions
CN117916260A (en) 2021-07-09 2024-04-19 詹森生物科技公司 Methods of manufacture for preparing anti-IL 12/IL23 antibody compositions
KR20240034218A (en) 2021-07-09 2024-03-13 얀센 바이오테크 인코포레이티드 Manufacturing Methods for Producing Anti-TNF Antibody Compositions
CN117795087A (en) 2021-08-02 2024-03-29 巴斯夫欧洲公司 Novel production of aromatic compounds with violet subunit ethane synthase
US11807685B2 (en) 2021-08-05 2023-11-07 The Uab Research Foundation Anti-CD47 antibody and uses thereof
WO2023028186A1 (en) 2021-08-27 2023-03-02 Abbott Laboratories Methods for detecting immunoglobulin g, subclass 4 (igg4) in a biological sample
WO2023034777A1 (en) 2021-08-31 2023-03-09 Abbott Laboratories Methods and systems of diagnosing brain injury
CA3232176A1 (en) 2021-09-30 2023-04-06 Beth MCQUISTON Methods and systems of diagnosing brain injury
CA3236779A1 (en) 2021-10-29 2023-05-04 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
WO2023084488A1 (en) 2021-11-15 2023-05-19 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
US20230159633A1 (en) 2021-11-23 2023-05-25 Janssen Biotech, Inc. Method of Treating Ulcerative Colitis with Anti-IL23 Specific Antibody
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
US20230213536A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
TW202348252A (en) 2022-02-16 2023-12-16 英商梅迪繆思有限公司 Combination therapies for treatment of cancer with therapeutic binding molecules
US20230312703A1 (en) 2022-03-30 2023-10-05 Janssen Biotech, Inc. Method of Treating Psoriasis with IL-23 Specific Antibody
WO2023223265A1 (en) 2022-05-18 2023-11-23 Janssen Biotech, Inc. Method for evaluating and treating psoriatic arthritis with il23 antibody
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury
WO2024059692A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Hbv diagnostic, prognostic, and therapeutic methods and products

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4366264A (en) * 1982-04-16 1982-12-28 Stanley Wawzonek Use of calcium metasilicate (wollastonite) as a formaldehyde suppressant for urea formaldehyde resins
US4490358A (en) * 1982-03-01 1984-12-25 President And Fellows Of Harvard College Screening vaccines and immunization process
US4634678A (en) * 1982-12-13 1987-01-06 Molecular Genetics Research And Development Limited Partnership Plasmid cloning and expression vectors for use in microorganisms
US4959312A (en) * 1985-05-31 1990-09-25 The University Of Tennessee Research Corporation Full spectrum mutagenesis

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4237224A (en) * 1974-11-04 1980-12-02 Board Of Trustees Of The Leland Stanford Jr. University Process for producing biologically functional molecular chimeras
US4321365A (en) * 1977-10-19 1982-03-23 Research Corporation Oligonucleotides useful as adaptors in DNA cloning, adapted DNA molecules, and methods of preparing adaptors and adapted molecules
US4366246A (en) * 1977-11-08 1982-12-28 Genentech, Inc. Method for microbial polypeptide expression
US4293652A (en) * 1979-05-25 1981-10-06 Cetus Corporation Method for synthesizing DNA sequentially
US4271145A (en) * 1979-10-22 1981-06-02 The Massachusetts General Hospital Process for producing antibodies to hepatitis virus and cell lines therefor
US4338397A (en) * 1980-04-11 1982-07-06 President And Fellows Of Harvard College Mature protein synthesis
US4879219A (en) * 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US4362867A (en) * 1980-12-10 1982-12-07 Research Corporation Recombinant cDNA construction method and hybrid nucleotides useful in cloning
US4394443A (en) * 1980-12-18 1983-07-19 Yale University Method for cloning genes
USRE32833E (en) * 1982-03-01 1989-01-17 President And Fellows Of Harvard College Screening vaccines and immunization process
DE3246071A1 (en) * 1982-12-13 1984-06-14 Christian Dipl.-Ing. 8900 Augsburg Strobel Method of genetic analysis and synthesis
DE3300632A1 (en) * 1982-12-13 1984-07-12 Christian Dipl.-Ing. 8900 Augsburg Strobel Method of genetic analysis and synthesis
DE3303173A1 (en) * 1982-12-13 1984-08-02 Christian Dipl.-Ing. 8900 Augsburg Strobel Method of genetic analysis and synthesis
US4719179A (en) * 1984-11-30 1988-01-12 Pharmacia P-L Biochemicals, Inc. Six base oligonucleotide linkers and methods for their use
US5723286A (en) * 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4490358A (en) * 1982-03-01 1984-12-25 President And Fellows Of Harvard College Screening vaccines and immunization process
US4366264A (en) * 1982-04-16 1982-12-28 Stanley Wawzonek Use of calcium metasilicate (wollastonite) as a formaldehyde suppressant for urea formaldehyde resins
US4634678A (en) * 1982-12-13 1987-01-06 Molecular Genetics Research And Development Limited Partnership Plasmid cloning and expression vectors for use in microorganisms
US4959312A (en) * 1985-05-31 1990-09-25 The University Of Tennessee Research Corporation Full spectrum mutagenesis

Also Published As

Publication number Publication date
WO1986005803A1 (en) 1986-10-09
US5824514A (en) 1998-10-20
EP0229046B1 (en) 1994-05-04
US5723323A (en) 1998-03-03
DE3587814T2 (en) 1994-11-10
AU4434585A (en) 1986-10-23
FR2579618B1 (en) 1987-12-24
DE3588239T2 (en) 2003-01-02
CA1339937C (en) 1998-06-30
GB2183661A (en) 1987-06-10
GB2183661B (en) 1989-06-28
EP0229046A1 (en) 1987-07-22
DE229046T1 (en) 1987-12-17
HK20292A (en) 1992-03-27
JPS62502235A (en) 1987-09-03
US5817483A (en) 1998-10-06
US5976862A (en) 1999-11-02
DE3590766C2 (en) 1991-01-10
EP0590689A2 (en) 1994-04-06
SG7992G (en) 1992-03-20
DE3546807C2 (en) 1991-03-28
GB8628313D0 (en) 1986-12-31
EP0590689B1 (en) 2002-05-22
DE3546806C2 (en) 1991-03-28
US5814476A (en) 1998-09-29
CN86102090A (en) 1986-11-05
DE3590766T (en) 1987-04-23
JP2584613B2 (en) 1997-02-26
EP0590689A3 (en) 1994-04-20
DE3588239T3 (en) 2007-03-08
EP1186660A3 (en) 2002-03-20
CA1341595C (en) 2009-04-28
DE3588239D1 (en) 2002-06-27
CH0229046H1 (en) 1998-07-15
EP0590689B2 (en) 2006-08-16
IN165561B (en) 1989-11-18
IN169027B (en) 1991-08-17
FR2579618A1 (en) 1986-10-03
JP2004089197A (en) 2004-03-25
JPH0856667A (en) 1996-03-05
JP2002325594A (en) 2002-11-12
US6569641B1 (en) 2003-05-27
DE3587814D1 (en) 1994-06-09
EP1186660A2 (en) 2002-03-13

Similar Documents

Publication Publication Date Title
US5824514A (en) Process for the production of expression vectors comprising at least one stochastic sequence of polynucleotides
US6492107B1 (en) Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
SU1644720A3 (en) Method for preparing antigen determinant of aids virus
US5750344A (en) Method for selection of biologically active peptide sequences
KR20190082318A (en) CRISPR / CPF1 system and method
KR860001515B1 (en) Process of replication and translation of human pre-growth hormone
JP2002517995A (en) Method for producing a polynucleotide having desired properties
JPH10304876A (en) Improved recombinant dna molecule
WO2002061097A1 (en) Cloning vectors and vector components
CN114672447B (en) Bacterial strain with self-flocculation capability and preparation method and application thereof
Böck et al. Location of the structural gene for glycyl ribonucleic acid synthetase by means of a strain of Escherichia coli possessing an unusual enzyme
CN114107176A (en) CHO cell line for stably expressing African swine fever CD2v protein and construction method and application thereof
CN109402096B (en) AID enzyme mutant and application thereof
RU2561466C2 (en) Expression plasmid for human recombinant coagulation factor viii with deleted b-domain in cho cells, cho cell producing human recombinant coagulation factor viii with deleted b-domain, and method for producing above factor
Glover et al. The principles of cloning DNA
US5382656A (en) Cellulose synthase associated proteins
US5736331A (en) Method for identifying nucleic acids encoding c-fos promoter activating proteins
CN117568369A (en) Protein system for rapidly screening binding with decoy RNA, method and application
CN117384864A (en) Recombinant adenovirus for expressing SARS-CoV-2Omicron Spike protein and its application
CN116970632A (en) Recombinant plasmid for expressing SARS-CoV-2S protein, its construction method and application
Zehring STRUCTURE OF BACTERIOPHAGE N4 ACTIVITY: TWO RNA POLYMERASE.
US20030060603A1 (en) Antibodies generated against polypeptide targets expressed from polynucleotide administration
Bigorra l LES DERNIERS BREVETS DlkPOS&: MORCEAUX CHOiSlS.
OR METHODS AND COMPOSITIONS FOR THE TREATMENT OF LUNG CONDITIONS
WO1990000610A1 (en) New expression vectors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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