US20040214277A1 - Methods and compositions for polypeptide engineering - Google Patents

Methods and compositions for polypeptide engineering Download PDF

Info

Publication number
US20040214277A1
US20040214277A1 US10/667,772 US66777203A US2004214277A1 US 20040214277 A1 US20040214277 A1 US 20040214277A1 US 66777203 A US66777203 A US 66777203A US 2004214277 A1 US2004214277 A1 US 2004214277A1
Authority
US
United States
Prior art keywords
dna
protein
dna substrate
products
library
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
US10/667,772
Inventor
Phillip Patten
Willem Stemmer
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.)
Codexis Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=25084330&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040214277(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US10/667,772 priority Critical patent/US20040214277A1/en
Publication of US20040214277A1 publication Critical patent/US20040214277A1/en
Priority to US11/136,115 priority patent/US20060166225A1/en
Priority to US11/198,765 priority patent/US20060084091A1/en
Priority to US11/286,271 priority patent/US20060223143A1/en
Assigned to CODEXIS, INC. reassignment CODEXIS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CODEXIS MAYFLOWER HOLDINGS, LLC
Abandoned legal-status Critical Current

Links

Images

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
    • C12N15/102Mutagenizing nucleic acids
    • C12N15/1031Mutagenizing nucleic acids mutagenesis by gene assembly, e.g. assembly by oligonucleotide extension PCR
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43595Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from coelenteratae, e.g. medusae
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C12N15/102Mutagenizing nucleic acids
    • C12N15/1027Mutagenizing nucleic acids by DNA shuffling, e.g. RSR, STEP, RPR
    • 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
    • C12N15/1037Screening libraries presented on the surface of microorganisms, e.g. phage display, E. coli display
    • 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
    • C12N15/1058Directional evolution of libraries, e.g. evolution of libraries is achieved by mutagenesis and screening or selection of mixed population of organisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • 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
    • C12N15/67General methods for enhancing the expression
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0069Oxidoreductases (1.) acting on single donors with incorporation of molecular oxygen, i.e. oxygenases (1.13)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1003Transferases (2.) transferring one-carbon groups (2.1)
    • C12N9/1007Methyltransferases (general) (2.1.1.)
    • 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)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • 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)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • C12N9/86Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5) acting on amide bonds in cyclic amides, e.g. penicillinase (3.5.2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6811Selection methods for production or design of target specific oligonucleotides or binding molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6853Nucleic acid amplification reactions using modified primers or templates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/02Libraries contained in or displayed by microorganisms, e.g. bacteria or animal cells; Libraries contained in or displayed by vectors, e.g. plasmids; Libraries containing only microorganisms or vectors
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • C40B40/08Libraries containing RNA or DNA which encodes proteins, e.g. gene libraries
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • 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
    • C12N2795/00Bacteriophages
    • C12N2795/00011Details
    • C12N2795/14011Details ssDNA Bacteriophages
    • C12N2795/14041Use of virus, viral particle or viral elements as a vector
    • C12N2795/14043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic 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
    • C12N2795/00Bacteriophages
    • C12N2795/00011Details
    • C12N2795/14011Details ssDNA Bacteriophages
    • C12N2795/14111Inoviridae
    • C12N2795/14141Use of virus, viral particle or viral elements as a vector
    • C12N2795/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B50/00Methods of creating libraries, e.g. combinatorial synthesis
    • C40B50/06Biochemical methods, e.g. using enzymes or whole viable microorganisms

Definitions

  • Recursive sequence recombination entails performing iterative cycles of recombination and screening or selection to “evolve” individual genes, whole plasmids or viruses, multigene clusters, or even whole genomes (Stemmer, Bio/Technology 13:549-553 (1995)). Such techniques do not require the extensive analysis and computation required by conventional methods for polypeptide engineering. Recursive sequence recombination allows the recombination of large numbers of mutations in a minimum number of selection cycles, in contrast to traditional, pairwise recombination events.
  • recursive sequence recombination (RSR) techniques provide particular advantages in that they provide recombination between mutations in any or all of these, thereby providing a very fast way of exploring the manner in which different combinations of mutations can affect a desired result.
  • One aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule comprising:
  • a further aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide and comprise defined segments, the method comprising:
  • step (b) amplifying the segments of the at least a first and second DNA substrate molecules with the primers of step (a) in a polymerase chain reaction;
  • step (c) assembling the products of step (b) to generate a library of recombinant DNA substrate molecules
  • a further aspect of the invention is a method of enriching a population of DNA fragments for mutant sequences comprising:
  • a further aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule, by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules share a region of sequence homology of about 10 to 100 base pairs and comprise defined segments, the method comprising:
  • a further aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide and comprise defined segments, the method comprising:
  • a further aspect of the invention is a method for optimizing expression of a protein by evolving the protein, wherein the protein is encoded by a DNA substrate molecule, comprising:
  • each oligonucleotide comprises at least two regions complementary to the DNA molecule and at least one degenerate region, each degenerate region encoding a region of an amino acid sequence of the protein;
  • a further aspect of the invention is a method for optimizing expression of a protein encoded by a DNA substrate molecule by evolving the protein, wherein the DNA substrate molecule comprises at least one lac operator and a fusion of a DNA sequence encoding the protein with a DNA sequence encoding a lac headpiece dimer, the method comprising:
  • a further aspect of the invention is a method for evolving functional expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA sequence encoding filamentous phage protein to generate a fusion protein, the method comprising:
  • a further aspect of the invention is a method for optimizing expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a lac headpiece dimer, wherein the DNA substrate molecule is present on a first plasmid vector, the method comprising:
  • a further aspect of the invention is a method for optimizing expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a filamentous phage gene, wherein the fusion is carried on a phagemid comprising a library of chaperonin gene mutants, the method comprising:
  • a further aspect of the invention is a method for optimizing secretion of a protein in a host by evolving a gene encoding a secretory function, comprising:
  • a further aspect of the invention is a method for evolving an improved DNA polymerase comprising:
  • a further aspect of the invention is a method for evolving a DNA polymerase with an error rate greater than that of wild type DNA polymerase comprising:
  • a further aspect of the invention is a method for evolving a DNA polymerase, comprising:
  • a further aspect of the invention is a method for evolving a p-nitrophenol phosphonatase from a phosphonatase encoded by a DNA substrate molecule, comprising:
  • a further aspect of the invention is a method for evolving a protease encoded by a DNA substrate molecule comprising:
  • a further aspect of the invention is a method for screening a library of protease mutants displayed on a phage to obtain an improved protease, wherein a DNA substrate molecule encoding the protease is fused to DNA encoding a filamentous phage protein to generate a fusion protein, comprising:
  • a further aspect of the invention is a method for screening a library of protease mutants to obtain an improved protease, the method comprising:
  • a further aspect of the invention is a method for evolving an alpha interferon gene comprising:
  • a further aspect of the invention is a method for screening a library of mutants of a DNA substrate encoding a protein for an evolved DNA substrate, comprising:
  • a further aspect of the invention is a method for evolving a DNA substrate molecule encoding an interferon alpha, comprising:
  • a further aspect of the invention is a method for evolving the stability of a protein encoded by a DNA substrate molecule, the DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA sequence encoding a filamentous phage protein to generate a fusion protein, the method comprising:
  • a further aspect of the invention is a method for evolving a protein having at least two subunits, comprising:
  • a further aspect of the invention is a method for evolving the coupling of a mammalian 7-transmembrane receptor to a yeast signal transduction pathway, comprising:
  • a further aspect of the invention is a method for recombining at least a first and second DNA substrate molecule, comprising:
  • a further aspect of the invention is a method for evolving a DNA substrate sequence encoding a protein of interest, wherein the DNA substrate comprises a vector, the vector comprising single-stranded DNA, the method comprising:
  • FIG. 1 depicts the alignment of oligo PCR primers for evolution of bovine calf intestinal alkaline phosphatase.
  • FIG. 2 depicts the alignment of alpha interferon amino acid and nucleic acid sequences.
  • the invention provides a number of strategies for evolving polypeptides through recursive recombination methods.
  • the strategies of the invention can generally be classified as “coarse grain shuffling” and “fine grain shuffling.” As described in detail below, these strategies are especially applicable in situations where some structural or functional information is available regarding the polypeptides of interest, where the nucleic acid to be manipulated is large, when selection or screening of many recombinants is cumbersome, and so on.
  • “Coarse grain shuffling” generally involves the exchange or recombination of segments of nucleic acids, whether defined as functional domains, exons, restriction endonuclease fragments, or otherwise arbitrarily defined segments.
  • “Fine grain shuffling” generally involves the introduction of sequence variation within a segment, such as within codons.
  • Coarse grain and fine grain shuffling allow analysis of variation occuring within a nucleic acid sequence, also termed “searching of sequence space.” Although both techniques are meritorious, the results are qualitatively different. For example, coarse grain searches are often better suited for optimizing multigene clusters such as polyketide operons, whereas fine grain searches are often optimal for optimizing a property such as protein expression using codon usage libraries.
  • the strategies generally entail evolution of gene(s) or segment(s) thereof to allow retention of function in a heterologous cell or improvement of function in a homologous or heterologous cell. Evolution is effected generally by a process termed recursive sequence recombination. Recursive sequence recombination can be achieved in many different formats and permutations of formats, as described in further detail below. These formats share some common principles. Recursive sequence recombination entails successive cycles of recombination to generate molecular diversity, i.e., the creation of a family of nucleic acid molecules showing substantial sequence identity to each other but differing in the presence of mutations.
  • Each recombination cycle is followed by at least one cycle of screening or selection for molecules having a desired characteristic.
  • the molecule(s) selected in one round form the starting materials for generating diversity in the next round.
  • recombination can occur in vivo or in vitro.
  • diversity resulting from recombination can be augmented in any cycle by applying prior methods of mutagenesis (e.g., error-prone PCR or cassette mutagenesis, passage through bacterial mutator strains, treatment with chemical mutagens) to either the substrates for or products of recombination.
  • the term “gene” is used herein broadly to refer to any segment or sequence of DNA associated with a biological function. Genes can be obtained from a variety of sources, including cloning from a source of interest or synthesizing from known or predicted sequence information, and may include sequences designed to have desired parameters.
  • Cells of particular interest include many bacterial cell types, both gram-negative and gram-positive, such as Rhodococcus, Streptomycetes, Actinomycetes, Corynebacteria, Penicillium, Bacillus, Escherichia coli, Pseudomonas, Salmonella, and Erwinia.
  • Cells of interest also include eukaryotic cells, particularly mammalian cells (e.g., mouse, hamster, primate, human), both cell lines and primary cultures.
  • Such cells include stem cells, including embryonic stem cells, zygotes, fibroblasts, lymphocytes, Chinese hamster ovary (CHO), mouse fibroblasts (NIH3T3), kidney, liver, muscle, and skin cells.
  • Other eukaryotic cells of interest include plant cells, such as maize, rice, wheat, cotton, soybean, sugarcane, tobacco, and arabidopsis; fish, algae, fungi ( Penicillium, Fusarium, Aspergillus, Podospora, Neurospora ), insects, yeasts ( Picchia and Saccharomyces ).
  • a preferred host has the ability to replicate vector DNA, express proteins of interest, and properly traffic proteins of interest.
  • Particularly advantageous hosts are E. coli, lactobacilli, Streptomycetes, Actinomycetes, fungi such as Saccaromyces cerivisiae or Pischia pastoris, Schneider cells, L-cells, COS cells, CHO cells, and transformed B cell lines such as SP2/0, J558, NS-1 and AG8-653.
  • the breeding procedure starts with at least two substrates that generally show substantial sequence identity to each other (i.e., at least about 50%, 70%, 80% or 90% sequence identity), but differ from each other at certain positions.
  • the difference can be any type of mutation, for example, substitutions, insertions and deletions.
  • different segments differ from each other in perhaps 5-20 positions.
  • the starting materials must differ from each other in at least two nucleotide positions. That is, if there are only two substrates, there should be at least two divergent positions. If there are three substrates, for example, one substrate can differ from the second as a single position, and the second can differ from the third at a different single position.
  • the starting DNA segments can be natural variants of each other, for example, allelic or species variants.
  • the segments can also be from nonallelic genes showing some degree of structural and usually functional relatedness (e.g., different genes within a superfamily such as the immunoglobulin superfamily).
  • the starting DNA segments can also be induced variants of each other.
  • one DNA segment can be produced by error-prone PCR replication of the other, or by substitution of a mutagenic cassette. Induced mutants can also be prepared by propagating one (or both) of the segments in a mutagenic strain.
  • the second DNA segment is not a single segment but a large family of related segments.
  • the different segments forming the starting materials are often the same length or substantially the same length. However, this need not be the case; for example; one segment can be a subsequence of another.
  • the segments can be present as part of larger molecules, such as vectors, or can be in isolated form.
  • the starting DNA segments are recombined by any of the recursive sequence recombination formats provded herein to generate a diverse library of recombinant DNA segments.
  • a library can vary widely in size from having fewer than 10 to more than 10 5 , 10 9 , or 10 12 members.
  • the starting segments and the recombinant libraries generated include full-length coding sequences and any essential regulatory sequences, such as a promoter and polyadenylation sequence, required for expression. However, if this is not the case, the recombinant DNA segments in the library can be inserted into a common vector providing the missing sequences before performing screening/selection.
  • the library of recombinant DNA segments generated already exists in a cell which is usually the cell type in which expression of the enzyme with altered substrate specificity is desired. If recursive sequence recombination is performed in vitro, the recombinant library is preferably introduced into the desired cell type before screening/selection. The members of the recombinant library can be linked to an episome or virus before introduction or can be introduced directly. In some embodiments of the invention, the library is amplified in a first host, and is then recovered from that host and introduced to a second host more amenable to expression, selection, or screening, or any other desirable parameter.
  • the manner in which the library is introduced into the cell type depends on the DNA-uptake characteristics of the cell type, e.g., having viral receptors, being capable of conjugation, or being naturally competent. If the cell type is insusceptible to natural and chemical-induced competence, but susceptible to electroporation, one would usually employ electroporation. If the cell type is insusceptible to electroporation as well, one can employ biolistics.
  • the biolistic PDS-1000 Gene Gun uses helium pressure to accelerate DNA-coated gold or tungsten microcarriers toward target cells. The process is applicable to a wide range of tissues, including plants, bacteria, fungi, algae, intact animal tissues, tissue culture cells, and animal embryos.
  • sequences can be DNA or RNA and can be of various lengths depending on the size of the gene or DNA fragment to be recombined or reassembled. Preferably the sequences are from 50 bp to 100 kb.
  • the pool of related substrates can be fragmented, usually at random, into fragments of from about 5 bp to 5 kb or more.
  • the size of the random fragments is from about 10 bp to 1000 bp, more preferably the size of the DNA fragments is from about 20 bp to 500 bp.
  • the substrates can be digested by a number of different methods, such as DNAseI or RNAse digestion, random shearing or restriction enzyme digestion.
  • the concentration of nucleic acid fragments of a particular length is often less than 0.1% or 1% by weight of the total nucleic acid.
  • the number of different specific nucleic acid fragments in the mixture is usually at least about 100, 500 or 1000.
  • the mixed population of nucleic acid fragments are denatured by heating to about 80° C. to 100° C., more preferably from 90° C. to 96° C., to form single-stranded nucleic acid fragments.
  • Single-stranded nucleic acid fragments having regions of sequence identity with other single-stranded nucleic acid fragments can then be reannealed by cooling to 20° C. to 75° C., and preferably from 40° C. to 65° C. Renaturation can be accelerated by the addition of polyethylene glycol (“PEG”) or salt.
  • the salt concentration is preferably from 0 mM to 600 mM, more preferably the salt concentration is from 10 mM to 100 mM.
  • the salt may be such salts as (NH 4 ) 2 SO 4 , KCl, or NaCl.
  • concentration of PEG is preferably from 0% to 20%, more preferably from 5% to 10%.
  • the fragments that reanneal can be from different substrates.
  • the annealed nucleic acid fragments are incubated in the presence of a nucleic acid polymerase, such as Taq or Klenow, Mg ++ at 1 mM-20 mM, and dNTP's (i.e. dATP, dCTP, dGTP and dTTP). If regions of sequence identity are large, Taq or other high-temperature polymerase can be used with an annealing temperature of between 45-65° C. If the areas of identity are small, Klenow or other low-temperature polymerases can be used with an annealing temperature of between 20-30° C.
  • the polymerase can be added to the random nucleic acid fragments prior to annealing, simultaneously with annealing or after annealing.
  • the cycle of denaturation, renaturation and incubation of random nucleic acid fragments in the presence of polymerase is sometimes referred to as “shuffling” of the nucleic acid in vitro.
  • This cycle is repeated for a desired number of times. Preferably the cycle is repeated from 2 to 100 times, more preferably the sequence is repeated from 10 to 40 times.
  • the resulting nucleic acids are a family of double-stranded polynucleotides of from about 50 bp to about 100 kb, preferably from 500 bp to 50 kb.
  • the population represents variants of the starting substrates showing substantial sequence identity thereto but also diverging at several positions. The population has many more members than the starting substrates.
  • the population of fragments resulting from recombination is preferably first amplified by PCR, then cloned into an appropriate vector and the ligation mixture used to transform host cells.
  • subsequences of recombination substrates can be generated by amplifying the full-length sequences under conditions which produce a substantial fraction, typically at least 20 percent or more, of incompletely extended amplification products.
  • the amplification products, including the incompletely extended amplification products are denatured and subjected to at least one additional cycle of reannealing and amplification.
  • This variation wherein at least one cycle of reannealing and amplification provides a substantial fraction of incompletely extended products, is termed “stuttering.”
  • the incompletely extended products anneal to and prime extension on different sequence-related template species.
  • At least one cycle of amplification can be conducted using a collection of overlapping single-stranded DNA fragments of related sequence, and different lengths. Each fragment can hybridize to and prime polynucleotide chain extension of a second fragment from the collection, thus forming sequence-recombined polynucleotides.
  • single-stranded DNA fragments of variable length can be generated from a single primer by Vent DNA polymerase on a first DNA template. The single stranded DNA fragments are used as primers for a second, Kunkel-type template, consisting of a uracil-containing circular single-stranded DNA. This results in multiple substitutions of the first template into the second (see Levichkin et al., Mol. Biology 29:572-577 (1995)).
  • Nucleic acid sequences can be recombined by recursive sequence recombination even if they lack sequence homology. Homology can be introduced using synthetic oligonucleotides as PCR primers.
  • all of the primers used to amplify one particular segment are synthesized to contain an additional sequence of 20-40 bases 5′ to the gene (sequence A) and a different 20-40 base sequence 3′ to the segment (sequence B).
  • An adjacent segment is amplified using a 5′ primer which contains the complementary strand of sequence B (sequence B′), and a 3′ primer containing a different 20-40 base sequence (C).
  • primers for the next adjacent segment contain sequences C′ (complementary to C) and D. In this way, small regions of homology are introduced, making the segments into site-specific recombination cassettes. Subsequent to the initial amplification of individual segments, the amplified segments can then be mixed and subjected to primeness PCR.
  • domains within a polypeptide are shuffled, it may not be possible to introduce additional flanking sequences to the domains, due to the constraint of maintaining a continuous open reading frame. Instead, groups of oligonucleotides are synthesized that are homologous to the 3′ end of the first domain encoded by one of the genes to be shuffled, and the 5′ ends of the second domains encoded by all of the other genes to be shuffled together. This is repeated with all domains, thus providing sequences that allow recombination between protein domains while maintaining their order.
  • the initial substrates for recombination are a collection of polynucleotides comprising variant forms of a gene.
  • the variant forms usually show substantial sequence identity to each other sufficient to allow homologous recombination between substrates.
  • the diversity between the polynucleotides can be natural (e.g., allelic or species variants), induced (e.g., error-prone PCR or error-prone recursive sequence recombination), or the result of in vitro recombination. Diversity can also result from resynthesizing genes encoding natural proteins with alternative codon usage. There should be at least sufficient diversity between substrates that recombination can generate more diverse products than there are starting materials.
  • the diverse substrates are incorporated into plasmids.
  • the plasmids are often standard cloning vectors, e.g., bacterial multicopy plasmids. However, in some methods to be described below, the plasmids include mobilization (MOB) functions.
  • the substrates can be incorporated into the same or different plasmids.
  • plasmids having different types of selectable markers are used to allow selection for cells containing at least two types of vector.
  • the different plasmids can come from two distinct incompatibility groups to allow stable co-existence of two different plasmids within the cell. Nevertheless, plasmids from the same incompatibility group can still co-exist within the same cell for sufficient time to allow homologous recombination to occur.
  • Plasmids containing diverse substrates are initially introduced into cells by any method (e.g., chemical transformation, natural competence, electroporation, biolistics, packaging into phage or viral systems). Often, the plasmids are present at or near saturating concentration (with respect to maximum transfection capacity) to increase the probability of more than one plasmid entering the same cell.
  • the plasmids containing the various substrates can be transfected simultaneously or in multiple rounds. For example, in the latter approach cells can be transfected with a first aliquot of plasmid, transfectants selected and propagated, and then infected with a second aliquot of plasmid.
  • the conditions for electroporation are the same as those conventionally used for introducing exogenous DNA into cells (e.g., 1,000-2,500 volts, 400 ⁇ F and a 1-2 mM gap). Under these conditions, plasmids are exchanged between cells allowing all substrates to participate in recombination. In addition the products of recombination can undergo further rounds of recombination with each other or with the original substrate. The rate of evolution can also be increased by use of conjugative transfer. To exploit conjugative transfer, substrates can be cloned into plasmids having MOB genes, and tra genes are also provided in cis or in trans to the MOB genes.
  • the effect of conjugative transfer is very similar to electroporation in that it allows plasmids to move between cells and allows recombination between any substrate and the products of previous recombination to occur, merely by propagating the culture.
  • the rate of evolution can also be increased by fusing cells to induce exchange of plasmids or chromosomes. Fusion can be induced by chemical agents, such as PEG, or viral proteins, such as influenza virus hemagglutinin, HSV-1 gB and gD.
  • the rate of evolution can also be increased by use of mutator host cells (e.g., Mut L, S, D, T, H in bacteria and Ataxia telangiectasia human cell lines).
  • the time for which cells are propagated and recombination is allowed to occur varies with the cell type but is generally not critical, because even a small degree of recombination can substantially increase diversity relative to the starting materials.
  • Cells bearing plasmids containing recombined genes are subject to screening or selection for a desired function. For example, if the substrate being evolved contains a drug resistance gene, one would select for drug resistance. Cells surviving screening or selection can be subjected to one or more rounds of screening/selection followed by recombination or can be subjected directly to an additional round of recombination. “Screening” as used herein is intended to include “selection” as a type of screen.
  • the next round of recombination can be achieved by several different formats independently of the previous round.
  • a further round of recombination can be effected simply by resuming the electroporation or conjugation-mediated intercellular transfer of plasmids described above.
  • a fresh substrate or substrates the same or different from previous substrates, can be transfected into cells surviving selection/screening.
  • the new substrates are included in plasmid vectors bearing a different selective marker and/or from a different incompatibility group than the original plasmids.
  • cells surviving selection/screening can be subdivided into two subpopulations, and plasmid DNA from one subpopulation transfected into the other, where the substrates from the plasmids from the two subpopulations undergo a further round of recombination.
  • the rate of evolution can be increased by employing DNA extraction, electroporation, conjugation or mutator cells, as described above.
  • DNA from cells surviving screening/selection can be extracted and subjected to in vitro recursive sequence recombination.
  • a second round of screening/selection is performed, preferably under conditions of increased stringency. If desired, further rounds of recombination and selection/screening can be performed using the same strategy as for the second round. With successive rounds of recombination and selection/screening, the surviving recombined substrates evolve toward acquisition of a desired phenotype.
  • the final product of recombination that has acquired the desired phenotype differs from starting substrates at 0.1%-25% of positions and has evolved at a rate orders of magnitude in excess (e.g., by at least 10-fold, 100-fold, 1000-fold, or 10,000 fold) of the rate of evolution driven by naturally acquired mutation of about 1 mutation per 10 ⁇ 9 positions per generation (see Anderson et al., Proc. Natl. Acad. Sci. U.S.A. 93:906-907 (1996)).
  • the “final product” may be transferred to another host more desirable for utilization of the “shuffled” DNA. This is particularly advantageous in situations where the more desirable host is less efficient as a host for the many cycles of mutation/ recombination due to the lack of molecular biology or genetic tools available for other organisms such as E. coli.
  • the strategy used for plasmid-plasmid recombination can also be used for virus-plasmid recombination; usually, phage-plasmid recombination.
  • virus-plasmid recombination usually, phage-plasmid recombination.
  • the initial substrates for recombination are cloned into both plasmid and viral vectors. It is usually not critical which substrate(s) is/are inserted into the viral vector and which into the plasmid, although usually the viral vector should contain different substrate(s) from the plasmid.
  • the plasmid (and the virus) typically contains a selective marker.
  • the plasmid and viral vectors can both be introduced into cells by transfection as described above.
  • Homologous recombination occurs between plasmid and virus generating both recombined plasmids and recombined virus.
  • viruses such as filamentous phage, in which intracellular DNA exists in both double-stranded and single-stranded forms, both can participate in recombination.
  • recombination can be augmented by use of electroporation or conjugation to transfer plasmids between cells.
  • Recombination can also be augmented for some types of virus by allowing the progeny virus from one cell to reinfect other cells.
  • virus infected-cells show resistance to superinfection. However, such resistance can be overcome by infecting at high multiplicity and/or using mutant strains of the virus in which resistance to superinfection is reduced.
  • viruses such as filamentous phage, stably exist with a plasmid in the cell and also extrude progeny phage from the cell.
  • Other viruses such as lambda having a cosmid genome, stably exist in a cell like plasmids without producing progeny virions.
  • Other viruses such as the T-phage and lytic lambda, undergo recombination with the plasmid but ultimately kill the host cell and destroy plasmid DNA.
  • cells containing recombinant plasmids and virus can be screened/selected using the same approach as for plasmid-plasmid recombination. Progeny virus extruded by cells surviving selection/screening can also be collected and used as substrates in subsequent rounds of recombination. For viruses that kill their host cells, recombinant genes resulting from recombination reside only in the progeny virus. If the screening or selective assay requires expression of recombinant genes in a cell, the recombinant genes should be transferred from the progeny virus to another vector, e.g., a plasmid vector, and retransfected into cells before selection/screening is performed.
  • another vector e.g., a plasmid vector
  • filamentous phage the products of recombination are present in both cells surviving recombination and in phage extruded from these cells.
  • the dual source of recombinant products provides some additional options relative to the plasmid-plasmid recombination.
  • DNA can be isolated from phage particles for use in a round of in vitro recombination.
  • the progeny phage can be used to transfect or infect cells surviving a previous round of screening/selection, or fresh cells transfected with fresh substrates for recombination.
  • the principles described for plasmid-plasmid and plasmid-viral recombination can be applied to virus-virus recombination with a few modifications.
  • the initial substrates for recombination are cloned into a viral vector.
  • the same vector is used for all substrates.
  • the virus is one that, naturally or as a result of mutation, does not kill cells.
  • some viral genomes can be packaged in vitro or using a packaging cell line. The packaged viruses are used to infect cells at high multiplicity such that there is a high probability that a cell will receive multiple viruses bearing different substrates.
  • cosmids containing recombinant genes can be recovered from surviving cells, e.g., by heat induction of a cos ⁇ lysogenic host cell, or extraction of DNA by standard procedures, followed by repackaging cosmid DNA in vitro.
  • viruses are filamentous phage
  • recombination of replicating form DNA occurs by propagating the culture of infected cells. Selection/screening identifies colonies of cells containing viral vectors having recombinant genes with improved properties, together with infectious particles (i.e., phage or packaged phagemids) extruded from such cells. Subsequent options are essentially the same as for plasmid-viral recombination.
  • This format can be used to especially evolve chromosomal substrates.
  • the format is particularly preferred in situations in which many chromosomal genes contribute to a phenotype or one does not know the exact location of the chromosomal gene(s) to be evolved.
  • the initial substrates for recombination are cloned into a plasmid vector. If the chromosomal gene(s) to be evolved are known, the substrates constitute a family of sequences showing a high degree of sequence identity but some divergence from the chromosomal gene. If the chromosomal genes to be evolved have not been located, the initial substrates usually constitute a library of DNA segments of which only a small number show sequence identity to the gene or gene(s) to be evolved. Divergence between plasmid-borne substrate and the chromosomal gene(s) can be induced by mutagenesis or by obtaining the plasmid-borne substrates from a different species than that of the cells bearing the chromosome.
  • the plasmids bearing substrates for recombination are transfected into cells having chromosomal gene(s) to be evolved. Evolution can occur simply by propagating the culture, and can be accelerated by transferring plasmids between cells by conjugation or electroporation. Evolution can be further accelerated by use of mutator host cells or by seeding a culture of nonmutator host cells being evolved with mutator host cells and inducing intercellular transfer of plasmids by electroporation or conjugation.
  • mutator host cells used for seeding contain a negative selectable marker to facilitate isolation of a pure culture of the nonmutator cells being evolved. Selection/screening identifies cells bearing chromosomes and/or plasmids that have evolved toward acquisition of a desired function.
  • plasmid-plasmid recombination proceeds in similar fashion to those described for plasmid-plasmid recombination.
  • further recombination can be effected by propagating cells surviving recombination in combination with electroporation or conjugative transfer of plasmids.
  • plasmids bearing additional substrates for recombination can be introduced into the surviving cells.
  • such plasmids are from a different incompatibility group and bear a different selective marker than the original plasmids to allow selection for cells containing at least two different plasmids.
  • plasmid and/or chromosomal DNA can be isolated from a subpopulation of surviving cells and transfected into a second subpopulation. Chromosomal DNA can be cloned into a plasmid vector before transfection.
  • the virus is usually one that does not kill the cells, and is often a phage or phagemid.
  • the procedure is substantially the same as for plasmid-chromosome recombination.
  • Substrates for recombination are cloned into the vector.
  • Vectors including the substrates can then be transfected into cells or in vitro packaged and introduced into cells by infection.
  • Viral genomes recombine with host chromosomes merely by propagating a culture. Evolution can be accelerated by allowing intercellular transfer of viral genomes by electroporation, or reinfection of cells by progeny virions. Screening/selection identifies cells having chromosomes and/or viral genomes that have evolved toward acquisition of a desired function.
  • viral genomes can be transferred between cells surviving selection/recombination by electroporation.
  • viruses extruded from cells surviving selection/screening can be pooled and used to superinfect the cells at high multiplicity.
  • fresh substrates for recombination can be introduced into the cells, either on plasmid or viral vectors.
  • fine grain and “coarse grain” shuffling.
  • coarse grain methods allow one to exchange chunks of genetic material between substrate nucleic acids, thereby limiting diversity in the resulting recombinants to exchanges or substitutions of domains, restriction fragments, oligo-encoded blocks of mutations, or other arbitrarily defined segments, rather than introducing diversity more randomly across the substrate.
  • fine grain shuffling methods allow the generation of all possible recombinations, or permutations, of a given set of very closely linked mutations, including multiple permutations, within a single segment, such as a codon.
  • coarse grain or fine grain shuffling techniques are not performed as exhaustive searches of all possible mutations within a nucleic acid sequence. Rather, these techniques are utilized to provide a sampling of variation possible within a gene based on known sequence or structural information.
  • the size of the sample is typically determined by the nature of the screen or selection process. For example, when a screen is performed in a 96-well microtiter format, it may be preferable to limit the size of the recombinant library to about 100 such microtiter plates for convenience in screening.
  • restriction enzyme sites in nucleic acids to direct the recombination of mutations in a nucleic acid sequence of interest.
  • These techniques are particularly preferred in the evolution of fragments that cannot readily be shuffled by existing methods due to the presence of repeated DNA or other problematic primary sequence motifs. They are also preferred for shuffling large fragments (typically greater than 10 kb), such as gene clusters that cannot be readily shuffled and “PCR-amplified” because of their size. Although fragments up to 50 kb have been reported to be amplified by PCR (Barnes, Proc. Natl. Acad. Sci. ( U.S.A.
  • the restriction endonucleases used are of the Class II type (Sambrook et al., Molecular Cloning, CSH Press, 1987) and of these, preferably those which generate nonpalindromic sticky end overhangs such as Alwn I, Sfi I or BstX1. These enzymes generate nonpalindromic ends that allow for efficient ordered reassembly with DNA ligase.
  • restriction enzyme (or endonuclease) sites are identified by conventional restriction enzyme mapping techniques (Sambrook et al., Molecular Cloning, CSH Press, 1987), by analysis of sequence information for that gene, or by introduction of desired restriction sites into a nucleic acid sequence by synthesis (i.e. by incorporation of silent mutations).
  • the DNA substrate molecules to be digested can either be from in vivo replicated DNA, such as a plasmid preparation, or from PCR amplified nucleic acid fragments harboring the restriction enzyme recognition sites of interest, preferably near the ends of the fragment.
  • at least two variants of a gene of interest, each having one or more mutations are digested with at least one restriction enzyme determined to cut within the nucleic acid sequence of interest.
  • the restriction fragments are then joined with DNA ligase to generate full length genes having shuffled regions. The number of regions shuffled will depend on the number of cuts within the nucleic acid sequence of interest.
  • the shuffled molecules can be introduced into cells as described above and screened or selected for a desired property. Nucleic acid can then be isolated from pools (libraries) or clones having desired properties and subjected to the same procedure until a desired degree of improvement is obtained.
  • At least one DNA substrate molecule or fragment thereof is isolated and subjected to mutagenesis.
  • the pool or library of religated restriction fragments are subjected to mutagenesis before the digestion-ligation process is repeated.
  • “Mutagenesis” as used herein comprises such techniques known in the art as PCR mutagenesis, oligonucleotide-directed mutagenesis, site-directed mutagenesis, etc., and recursive sequence recombination by any of the techniques described herein.
  • Polyketide clusters (Khosla et al., TIBTECH 14, September 1996) are typically 10 to 100 kb in length, specifying multiple large polypeptides which assemble into very large multienzyme complexes. Due to the modular nature of these complexes and the modular nature of the biosynthetic pathway, nucleic acids encoding protein modules can be exchanged between different polyketide clusters to generate novel and functional chimeric polyketides.
  • a further technique for recursively recombining mutations in a nucleic acid sequence utilizes “reassembly PCR”. This method can be used to assemble multiple segments that have been separately evolved into a full length nucleic acid template such as a gene. This technique is performed when a pool of advantageous mutants is known from previous work or has been identified by screening mutants that may have been created by any mutagenesis technique known in the art, such as PCR mutagenesis, cassette mutagenesis, doped oligo mutagenesis, chemical mutagenesis, or propagation of the DNA template in vivo in mutator strains.
  • Boundaries defining segments of a nucleic acid sequence of interest preferably lie in intergenic regions, introns, or areas of a gene not likely to have mutations of interest.
  • oligonucleotide primers are synthesized for PCR amplification of segments of the nucleic acid sequence of interest, such that the sequences of the oligonucleotides overlap the junctions of two segments.
  • the overlap region is typically about 10 to 100 nucleotides in length.
  • Each of the segments is amplified with a set of such primers.
  • the PCR products are then “reassembled” according to assembly protocols such as those used in Sections IA-B above to assemble randomly fragmented genes.
  • PCR products are first purified away from the primers, by, for example, gel electrophoresis or size exclusion chromatography. Purified products are mixed together and subjected to about 1-10 cycles of denaturing, reannealing, and extension in the presence of polymerase and deoxynucleoside triphosphates (dNTP's) and appropriate buffer salts in the absence of additional primers (“self-priming”). Subsequent PCR with primers flanking the gene are used to amplify the yield of the fully reassembled and shuffled genes. This method is necessarily “coarse grain” and hence only recombines mutations in a blockwise fashion, an advantage for some searches such as when recombining allelic variants of multiple genes within an operon.
  • dNTP's polymerase and deoxynucleoside triphosphates
  • self-priming self-priming
  • the resulting reassembled genes are subjected to mutagenesis before the process is repeated.
  • oligonucleotides that incorporate uracil into the primers are used for PCR amplification.
  • uracil is incorporated at one site in the oligonucleotide.
  • the products are treated with uracil glycosylase, thereby generating a single-stranded overhang, and are reassembled in an ordered fashion by a method such as disclosed by Rashtchian ( Current Biology, 3:1-36 (1995)).
  • the PCR primers for amplification of segments of the nucleic acid sequence of interest are used to introduce variation into the gene of interest as follows. Mutations at sites of interest in a nucleic acid sequence are identified by screening or selection, by sequencing homologues of the nucleic acid sequence, and so on. Oligonucleotide PCR primers are then synthesized which encode wild type or mutant information at sites of interest. These primers are then used in PCR mutagenesis to generate libraries of full length genes encoding permutations of wild type and mutant information at the designated positions. This technique is typically advantagous in cases where the screening or selection process is expensive, cumbersome, or impractical relative to the cost of sequencing the genes of mutants of interest and synthesizing mutagenic oligonucleotides.
  • An example of this method is the evolution of an improved Taq polymerase, as described in detail below. Mutant proteins resulting from application of the method are identified and assayed in a sequencing reaction to identify mutants with improved sequencing properties. This is typically done in a high throughput format (see, for example, Broach et al. Nature 384 (Supp): 14-16 (1996)) to yield, after screening, a small number, e.g., about 2 to 100, of candidate recombinants for further evaluation. The mutant genes can then be sequenced to provide information regarding the location of the mutation.
  • the corresponding mutagenic oligonucleotide primers can be synthesized from this information, and used in a reassembly reaction as described above to efficiently generate a library with an average of many mutations per gene.
  • multiple rounds of this protocol allows the efficient search for improved variants of the Taq polymerase.
  • recombination reactions are enriched for mutant sequences so that the multiple mutant spectrum, i.e. possible combinations of mutations, is more efficiently sampled.
  • the rationale for this is as follows. Assume that a number, n, of mutant clones with improved activity is obtained, wherein each clone has a single point mutation at a different position in the nucleic acid sequence. If this population of mutant clones with an average of one mutation of interest per nucleic acid sequence is then put into a recombination reaction, the resulting population will still have an average of one mutation of interest per nucleic acid sequence as defined by a Poisson distribution, leaving the multiple mutation spectrum relatively unpopulated.
  • the amount of screening required to identify recombinants having two or more mutations can be dramatically reduced by the following technique.
  • the nucleic acid sequences of interest are obtained from a pool of mutant clones and prepared as fragments, typically by digestion with a restriction endonuclease, sonication, or by PCR amplification.
  • the fragments are denatured, then allowed to reanneal, thereby generating mismatched hybrids where one strand of a mutant has hybridized with a complementary strand from a different mutant or wild-type clone.
  • the reannealed products are then fragmented into fragments of about 20-100 bp, for example, by the use of DNAseI.
  • This fragmentation reaction has the effect of segregating regions of the template containing mismatches (mutant information) from those encoding wild type sequence.
  • the mismatched hybrids can then be affinity purified using aptamers, dyes, or other agents which bind to mismatched DNA.
  • a preferred embodiment is the use of mutS protein affinity matrix (Wagner et al., Nucleic Acids Res. 23(19):3944-3948 (1995); Su et al., Proc. Natl. Acad. Sci. ( U.S.A. ), 83:5057-5061(1986)) with a preferred step of amplifying the affinity-purified material in vitro prior to an assembly reaction.
  • This amplified material is then put into a assembly PCR reaction as decribed above.
  • this material can be titrated against the original mutant pool (e.g., from about 100% to 10% of the mutS enriched pool) to control the average number of mutations per clone in the next round of recombination.
  • Another application of this method is in the assembly of gene constructs that are enriched for polymorphic bases occurring as natural or selected allelic variants or as differences between homologous genes of related species. For example, one may have several varieties of a plant that are believed to have heritable variation in a trait of interest (e.g., drought resistance). It then is of interest to construct a library of these variant genes containing many mutations per gene. MutS selection can be applied in combination with the assembly techniques described herein to generate such a pool of recombinants that are highly enriched for polymorphic (“mutant”) information. In some embodiments, the pool of recombinant genes is provided in a transgenic host. Recombinants can be further evolved by PCR amplification of the transgene from transgenic organisms that are determined to have an improved phenotype and applying the formats described in this invention to further evolve them.
  • a trait of interest e.g., drought resistance
  • the substrate molecules for recombination have uniformly low homology, sporadically distributed regions of homology, or the region of homology is relatively small (for example, about 10-100 bp), such as phage displayed peptide ligands. These factors can reduce the efficiency and randomness of recombination in RSR.
  • this problem is addressed by the introduction of introns between coding exons in sequences encoding protein homologues.
  • introns can be used (Chong et al., J. Biol. Chem., 271:22159-22168 (1996)).
  • a nucleic acid sequence such as a gene or gene family
  • the segments are preferably exons.
  • Introns are engineered between the segments.
  • the intron inserted between the first and second segments is at least about 10% divergent from the intron inserted between second and third segments
  • the intron inserted between second and third segments is at least about 10% divergent from the introns inserted between any of the previous segment pairs, and so on through segments n and n+1.
  • the introns between any given set of exons will thus initially be identical between all clones in the library, whereas the exons can be arbitrarily divergent in sequence.
  • the introns therefore provide homologous DNA sequences that will permit application of any of the described methods for RSR while the exons can be arbitrarily small or divergent in sequence, and can evolve to achieve an arbitrarily large degree of sequence divergence without a significant loss in efficiency in recombination. Restriction sites can also be engineered into the intronic nucleic acid sequence of interest so as to allow a directed reassemmbly of restriction fragments.
  • the starting exon DNA may be synthesized de novo from sequence information, or may be present in any nucleic acid preparation (e.g., genomic, cDNA, libraries, and so on).
  • 1 to 10 nonhomologous introns can be designed to direct recombination of the nucleic acid sequences of interest by placing them between exons.
  • the sequence of the introns can be all or partly obtained from known intron sequence.
  • the introns are self-splicing. Ordered sets of introns and exon libraries are assembled into functional genes by standard methods (Sambrook et al., Molecular Cloning, CSH Press (1987)).
  • Any of the formats for in vitro or in vivo recombination described herein can be applied for recursive exon shuffling.
  • a preferred format is to use nonpalindromic restriction sites such as Sfi I placed into the intronic sequences to promote shuffling. Pools of selected clones are digested with Sfi I and religated. The nonpalindromic overhangs promote ordered reassembly of the shuffled exons.
  • These libraries of genes can be expressed and screened for desired properties, then subjected to further recursive rounds of recombination by this process. In some embodiments, the libraries are subjected to mutagenesis before the process is repeated.
  • An example of how the introduction of an intron into a mammalian library format would be used advantageously is as follows.
  • An intron containing a lox (Sauer et al., Proc. Natl. Acad. Sci. ( U.S.A. ), 85:5166-5170 (1988)) site is arbitrarily introduced between amino acids 92 and 93 in each alpha interferon parental substrate.
  • a library of 10 4 chimeric interferon genes is made for each of the two exons (residues 1-92 and residues 93-167), cloned into a replicating plasmid vector, and introduced into target cells.
  • the number 10 4 is arbitrarily chosen for convenience in screening.
  • An exemplary vector for expression in mammalian cells would contain an SV40 origin, with the host cells expressing SV40 large T antigen, so as to allow transient expression of the interferon constructs.
  • the cells are challenged with a cytopathic virus such as vesicular stomatitis virus (VSV) in an interferon protection assay (e.g., Meister et al., J. Gen. Virol. 67:1633-1643, (1986)).
  • VSV vesicular stomatitis virus
  • the amplified plasmids are then transfected at high multiplicity (e.g. 10 micrograms of plasmid per 10 6 cells) into a cre expressing host that can support replication of that vector.
  • the presence of cre in the host cells promotes efficient recombination at the lox site in the interferon intron, thus shuffling the selected sets of exons.
  • This population of cells is then used in a second round of selection by viral challenge and the process is applied recursively.
  • the cre recombinase is preferrably expressed transiently on a cotransfected molecule that cannot replicate in the host.
  • the method can be used with more than one intron, with recombination enhancing sequences other than cre/lox (e.g., int/xis, etc.), and with other vector systems such as but not limited to retroviruses, adenovirus or adeno- associated virus.
  • a search of a region of sequence space defined by a set of substrates, such as members of a gene family, having less than about 80%, more typically, less than about 50% homology is desired.
  • This region which can be part or all of a gene or a gene is arbitrarily delineated into segments.
  • the segment borders can be chosen randomly, based on correspondence with natural exons, based on structural considerations (loops, alpha helices, subdomains, whole domains, hydrophobic core, surface, dynamic simulations), and based on correlations with genetic mapping data.
  • the segments are then amplified by PCR with a pool of “bridge” oligonucleotides at each junction.
  • bridge oligonucleotides
  • the set of five genes is broken into three segments A, B and C, and if there are five versions of each segment (A 1 , A 2 , . . . C 4 , C 5 )
  • twenty five oligonucleotides are made for each strand of the A-B junctions where each bridge oligo has 20 bases of homology to one of the A and one of the B segments.
  • the number of required oligonucleotides can be reduced by choosing segment boundaries that are identical in some or all of the gene family members. Oligonucleotides are similarly synthesized for the B-C junction.
  • the family of A domains is amplified by PCR with an outside generic A primer and the pool of A-B junction oligonucleotides; the B domains with the A-B plus the B-C bridge oligonucleotides, and the C domains with the B-C bridge oligonucleotides plus a generic outside primer.
  • Full length genes are made then made by assembly PCR or by the dUTP/uracil glycosylase methods described above.
  • products from this step are subjected to mutagenesis before the process of selection and recombination is repeated, until a desired level of improvement or the evolution of a desired property is obtained. This is typically determined using a screening or selection as appropriate for the protein and property of interest.
  • sequence information from one or more substrate sequences is added to a given “parental” sequence of interest, with subsequent recombination between rounds of screening or selection.
  • this is done with site-directed mutagenesis performed by techniques well known in the art (Sambrook et al., Molecular Cloning, CSH Press (1987)) with one substrate as template and oligonucleotides encoding single or multiple mutations from other substrate sequences, e.g. homologous genes.
  • the selected recombinant(s) can be further evolved using RSR techniques described herein.
  • site-directed mutagenesis can be done again with another collection of oligonucleotides encoding homologue mutations, and the above process repeated until the desired properties are obtained.
  • degenerate oligonucleotides can be used that encode the sequences in both homologues.
  • One oligo may include many such degenerate codons and still allow one to exhaustively search all permutations over that block of sequence. An example of this is provided below for the evolution of alpha interferon genes.
  • DNA substrate molecules are introduced into cells, wherein the cellular machinery directs their recombination.
  • a library of mutants is constructed and screened or selected for mutants with improved phenotypes by any of the techniques described herein.
  • the DNA substrate molecules encoding the best candidates are recovered by any of the techniques described herein, then fragmented and used to transfect a mammalian host and screened or selected for improved function.
  • the DNA substrate molecules are recovered from the mammalian host, such as by PCR, and the process is repeated until a desired level of improvement is obtained.
  • the fragments are denatured and reannealed prior to transfection, coated with recombination stimulating proteins such as recA, or co-transfected with a selectable marker such as Neo R to allow the positive selection for cells receiving recombined versions of the gene of interest.
  • this format is preferred for the in vivo affinity maturation of an antibody by RSR.
  • a library of mutant antibodies is generated, as described herein for the 48G7 affinity maturation.
  • This library is FACS purified with ligand to enrich for antibodies with the highest 0.1-10% affinity.
  • the V regions genes are recovered by PCR, fragmented, and cotransfected or electorporated with a vector into which reassembled V region genes can recombine. DNA substrate molecules are recovered from the cotranfected cells, and the process is repeated until the desired level of improvment is obtained.
  • Other embodiments include reassembling the V regions prior to the electroporation so that an intact V region exon can recombine into an antibody expression cassette. Further embodiments include the use of this format for other eukaryotic genes or for the evolution of whole viruses.
  • a gene of interest is cloned into a vector that generates single stranded DNA, such as a phagemid.
  • the resulting DNA substrate is mutagenzied by RSR in any method known in the art, transfected into host cells, and subjected to a screen or selection for a desired property or improved phenotype.
  • DNA from the selected or screened phagemids is amplified, by, for example, PCR or plasmid preparation. This DNA preparation contains the various mutant sequences that one wishes to permute.
  • This DNA is fragmented and denatured, and annealed with single-stranded DNA (ssDNA) phagemid template (ssDNA encoding the wild-type gene and vector sequences).
  • ssDNA single-stranded DNA
  • a preferred embodiment is the use of dut( ⁇ ) ung( ⁇ ) host strains such as CJ236 (Sambrook et al., Molecular Cloning CSH Press (1987)) for the preparation of ssDNA.
  • Gaps in annealed template are filled with DNA polymerase and ligated to form closed relaxed circles. Since multiple fragments can anneal to the phagemid, the newly synthesized strand now consists of shuffled sequences. These products are transformed into a mutS strain of E. coli which is dut+ ung+. Phagemid DNA is recovered from the transfected host and subjected again to this protocol until the desired level of improvement is obtained. The gene encoding the protein of interest in this library of recovered phagemid DNA can be mutagenzied by any technique, including RSR, before the process is repeated.
  • the degenerate oligonucleotides have the general structure of about 20 nucleotides of identity to a DNA substrate molecule encoding a protein of interest, followed by a region of about 20 degenerate nucleotides which encode a region of the protein, followed by another region of about 20 nucleotides of identity.
  • the region of identity utilizes preferred codons for the host.
  • the oligonucleotides are identical to the DNA substrate at least one 5′ and one 3′ nucleotide, but have at least 85% sequence homology to the DNA substrate molecule, with the difference due to the use of degenerate codons.
  • a set of such degenerate oligonucleotides is used in which each oligonucleotide overlaps with another by the general formula n ⁇ 10, wherein n is the length of the oligonucleotide.
  • n is the length of the oligonucleotide.
  • Such oligonucleotides are typically about 20-1000 nucleotides in length.
  • the assembled genes are then cloned, expressed, and screened or selected for improved expression.
  • the assembled genes can be subjected to recursive recombination methods as descibed above until the desired improvement is achieved.
  • this technique can be used to evolve bovine intestinal alkaline phosphatase (BIAP) for active expression in E. coli.
  • BIAP bovine intestinal alkaline phosphatase
  • This enzyme is commonly used as a reporter gene in assay formats such as ELISA.
  • the cloned gene cannot be expressed in active form in a prokaryotic host such as E. coli in good yield.
  • Development of such an expression system would allow one to access inexpensive expression technology for BIAP and, importantly, for engineered variants with improved activity or chemical coupling properties (such as chemical coupling to antibodies).
  • a detailed example is provide in the Experimental Examples section.
  • proteins of interest when overexpressed or expressed in heterologous hosts form inclusion bodies, with the majority of the expressed protein being found in insoluble aggregates.
  • Recursive sequence recombination techniques can be used to optimize folding of such target proteins. There are several ways to improve folding, including mutating evolving the target protein of interest and evolving chaperonin proteins.
  • the lac repressor “headpiece dimer” is a small protein containing two headpiece domains connected by a short peptide linker which binds the lac operator with sufficient affinity that polypeptide fusions to this headpiece dimer will remain bound to the plasmid that encodes them throughout an affinity purification process (Gates et al., J. Mol. Biol. 255:373-386 (1995)). This property can be exploited, as follows, to evolve mutant proteins of interest with improved folding properties.
  • the protein of interest can be mammalian, yeast, bacterial, etc.
  • a fusion protein between the lac headpiece dimer and a target protein sequence is constructed, for example, as disclosed by Gates (supra).
  • This construct containing at least one lac operator, is mutagenized by technologies common in the arts such as PCR mutagenesis, chemical mutagenesis, oligo directed mutagenesis (Sambrook et al., Molecular Cloning CSH Press (1987)).
  • the resulting library is transformed into a host cell, and expression of the fusion protein is induced, preferably with arabinose.
  • An extract or lysate is generated from a culture of the library expressing the construct.
  • Insoluble protein is fractionated from soluble protein/DNA complexes by centrifugation or affinity chromatography, and the yield of soluble protein/DNA complexes is quantitated by quantitative PCR (Sambrook et al., Molecular Cloning, CSH Press, 1987) of the plasmid.
  • a reagent that is specific for properly folded protein such as a monoclonal antibody or a natural ligand, is used to purify soluble protein/DNA complexes.
  • the plasmid DNA from this step is isolated, subjected to RSR and again expressed. These steps are repeated until the yield of soluble protein/DNA complexes has reached a desired level of improvement. Individual clones are then screened for retention of functional properties of the protein of interest, such as enzymatic activity, etc.
  • This technique is generically useful for evolving solubility and other properties such as cellular trafficking of proteins heterologously expressed in a host cell of interest. For example, one could select for efficient folding and nuclear localization of a protein fused to the lac repressor headpiece dimer by encoding the protein on a plasmid encoding an SV40 origin of replication and a lac operator, and transiently expressing the fusion protein in a mammalian host expressing T antigen. Purification of protein/DNA complexes from nuclear HIRT extracts (Seed and Aruffo, Proc. Natl. Acad. Sci. ( U.S.A. ), 84:3365-3369 (1987)) would allow one to select for efficient folding and nuclear localization proteins.
  • a problem often encountered in phage display methods such as those disclosed by O'Neil et al. ( Current Biology, 5:443-449 (1995)) is the inability to functionally express a protein of interest on phage. Without being limited to any one theory, improper folding of the protein of interest can be responsible for this problem.
  • RSR can be used to evolve a protein of interest for functional expression on phage.
  • a fusion protein is constructed between gene III or gene VIII and the target protein and then mutagenized, for example by PCR mutagenesis.
  • the mutagenzied library is then expressed in a phage display format, a phage lysate is made, and these phage are affinity selected for those bearing functionally displayed fusion proteins using an affinity matrix containing a known ligand for the target protein.
  • DNA from the functionally selected phage is purified, and the displayed genes of interest are shuffled and recloned into the phage display format.
  • the selection, shuffling and recloning steps are repeated until the yield of phage with functional displayed protein has reached desired levels as defined, for example, by the fraction of phage that are retained on a ligand affinity matrix or the biological activity associated with the displayed phage.
  • Individual clones are then screened to identify candidate mutants with improved display properties, desired level of expression, and functional properties of interest (e.g., ability to bind a ligand or receptor, lymphokine activity, enzymatic activity, etc.).
  • a functional screen or selection is used to identify an evolved protein not expressed on a phage.
  • the target protein which cannot initially be efficiently expressed in a host of interest, is mutagenized and a functional screen or selection is used to identify cells expressing functional protein.
  • the protein of interest may complement a function in the host cell, cleave a colorimetric substrate, etc. Recursive sequence recombination is then used to rapidly evolve improved functional expression from such a pool of improved mutants.
  • AMV reverse transcriptase is of particular commercial importance because it is active at a higher temperature (42° C.) and is more robust than many other reverse transcriptases.
  • it is difficult to express in prokaryotic hosts such as E. coli, and is consequently expensive because it has to be purified from chicken cells.
  • an evolved AMV reverse transcriptase that can be expressed efficiently in E. coli is highly desirable.
  • the AMV reverse transcriptase gene (Papas et al., J. Cellular Biochem 20:95-103 (1982)) is mutagenized by any method common in the art.
  • the library of mutant genes is cloned into a colE1 plasmid (Amp resistant) under control of the lac promoter in a polA12 (Ts) recA718 (Sweasy et al. Proc. Natl. Acad. Sci. U.S.A. 90:4626-4630 (1993)) E. coli host.
  • the library is induced with IPTG, and shifted to the nonpermissive temperature.
  • AMV RTX genes are recovered by PCR by using oligonucleotides flanking the cloned gene. The resulting PCR products are subjected to in vitro RSR, selected as described above, and the process is repeated until the level of functional expression is acceptable. Individual clones are then screened for RNA-dependent DNA polymerization and other properties of interest (e.g. half life at room temperature, error rate). The candidate clones are subjected to mutagenesis, and then tested again to yield an AMV RT that can be expressed in E. coli at high levels.
  • overexpression of a protein can lead to the accumulation of folding intermediates which have a tendency to aggregate.
  • the role of chaperonins is thought to be to stabilize such folding intermediates against aggregration; thus, overexpression of a protein of interest can lead to overwhelming the capacity of chaperoning.
  • Chaperonin genes can be evolved using the techniques of the invention, either alone or in combination with the genes encoding the protein of interest, to overcome this problem.
  • proteins of interest which are especially suited to this approach include but are not limited to: cytokines; malarial coat proteins; T cell receptors; antibodies; industrial enzymes (e.g., detergent proteases and detergent lipases); viral proteins for use in vaccines; and plant seed storage proteins.
  • Sources of chaperonin genes include but are limited to E. coli chaperonin genes encoding such proteins as thioredoxin, Gro ES/Gro EL, PapD, ClpB, DsbA, DsbB, DnaJ, DnaK, and GrpE; mammalian chaperoning such as Hsp70, Hsp72, Hsp73, Hsp40,Hsp60, Hsp10, Hdj1, TCP-1, Cpn60, BiP; and the homologues of these chaperonin genes in other species such as yeast (J. G. Wall and A. Pluckthun, Current Biology, 6:507-516 (1995); Hartl, Nature, 381:571-580 (1996)). Additionally, heterologous genomic or cDNA libraries can be used as libraries to select or screen for novel chaperoning.
  • E. coli chaperonin genes encoding such proteins as thioredoxin, Gro ES/Gro EL,
  • the chaperonin genes are evolved independently of the gene(s) for the protein of interest.
  • the improvement in the evolved chaperonin can be assayed, for example, by screening for enhancement of the activity of the target protein itself or for the activity of a fusion protein comprising the target protein and a selectable or screenable protein (e.g., GFP, alkaline phosphatase or beta-galactosidase).
  • a selectable or screenable protein e.g., GFP, alkaline phosphatase or beta-galactosidase.
  • the chaperonin genes and the target protein genes are encoded on the same plasmid, but not necessarily evolved together.
  • a lac headpiece dimer can be fused to the protein target to allow for selection of plasmids which encode soluble protein.
  • Chaperonin genes are provided on this same plasmid (“cis”) and are shuffled and evolved rather than the target protein.
  • the chaperonin genes can be cloned onto a phagemid plasmid that encodes a gene III or gene VIII fusion with a protein of interest.
  • the cloned chaperonins are mutagenized and, as with the selection described above, phage expressing functionally displayed fusion protein are isolated on an affinity matrix.
  • the chaperonin genes from these phage are shuffled and the cycle of selection, mutation and recombination are applied recursively until fusion proteins are efficiently displayed in functional form.
  • coli for example, is known to be influenced by many proteins including: a secretory ATPase (SecA), a translocase complex (SecB, SecD, SecE, SecF, and SecY), chaperonins (DnaK, DnaJ, GroES, GroEL), signal peptidases (LepB, LspA, Ppp), specific folding catalysts (DsbA) and other proteins of less well defined function (e.g., Ffh, FtsY) (Sandkvist et al., Curr. Op. Biotechnol. 7:505-511 (1996)). Overproduction of wild type or mutant copies of these genes for these proteins can significantly increase the yield of mature secreted protein.
  • RSR provides a route to evolution of one or more of the above named components of the secretory pathway.
  • the following strategy is employed to optimize protein secretion in E. coli. Variations on this method, suitable for application to Bacillus subtilis, Pseudomonas, Saccaromyces cerevisi ⁇ , Pichia pastoris, mammalian cells and other hosts are also described.
  • the general protocol is as follows.
  • genes named above are obtained by PCR amplification from E. coli genomic DNA using known flanking sequence, and cloned in an ordered array into a plasmid or cosmid vector. These genes do not in general occur naturally in clusters, and hence these will comprise artificial gene clusters.
  • the genes may be cloned under the control of their natural promoter or under the control of another promoter such as the lac, tac, arabinose, or trp promoters.
  • rare restriction sites such as Sfi I are placed between the genes to facilitate ordered reassembly of shuffled genes as described in the methods of the invention.
  • the gene cluster is mutagenized and introduced into a host cell in which the gene of interest can be inducibly expressed.
  • Expression of the target gene to be secreted and of the cloned genes is induced by standard methods for the promoter of interest (e.g., addition of 1 mM IPTG for the lac promoter).
  • the efficiency of protein secretion by a library of mutants is measured, for example by the method of colony blotting (Skerra et al., Anal. Biochem. 196:151-155 (1991)). Those colonies expressing the highest levels of secreted protein (the top 0.1-10%; preferably the top 1%) are picked. Plasmid DNA is prepared from these colonies and shuffled according to any of the methods of the invention.
  • each individual gene is amplified from the population and subjected to RSR.
  • the fragments are digested with Sfi I (introduced between each gene with nonpalindromic overhangs designed to promote ordered reassembly by DNA ligase) and ligated together, preferably at low dilution to promote formation of covalently closed relaxed circles ( ⁇ 1 ng/microliter).
  • Sfi I introduced between each gene with nonpalindromic overhangs designed to promote ordered reassembly by DNA ligase
  • Each of the PCR amplified gene populations may be shuffled prior to reassembly into the final gene cluster.
  • the ligation products are transformed back into the host of interest and the cycle of selection and RSR is repeated.
  • Analogous strategies can be employed in other hosts such as Pseudomonas, Bacillus subtilis, yeast and mammalian cells.
  • the homologs of the E. coli genes listed above are targets for optimization, and indeed many of these homologs have been identified in other species (Pugsley, Microb. Rev. 57:50-108 (1993)).
  • other components such as the six polypeptides of the signal recognition particle, the trans-locating chain-associating membrane protein (TRAM), BiP, the Ssa proteins and other hsp70 homologs, and prsA ( B. subtilis ) (Simonen and Pulva, Microb. Rev.
  • replicating episomal vectors such as SV40-neo (Sambrook et al., Molecular Cloning, CSH Press (1987), Northrup et al., J. Biol. Chem. 268(4):2917-2923 (1993)) for mammalian cells or 2 micron or ars plasmids for yeast (Strathern et al., The Molecular Biology of the Yeast Saccaromyces, CSH Press (1982)) are used. Integrative vectors such as pJM 103, pJM 113 or pSGMU2 are preferred for B. subtilis (Perego, Chap. 42, pp. 615-624 in: Bacillus subtilis and Other Gram - Positive Bacteria, A. Sonenshein, J. Hoch, and R. Losick, eds., 1993).
  • thermostable DNA polymerase can be evolved, thus allowing the performance of DNA polymerization assays with little or no purification of the expressed DNA polymerase.
  • Initial constructs are made by fusing a signal peptide such as that from STII or OmpA to the amino terminus of the protein to be secreted.
  • a gene cluster of cloned genes believed to act in the secretory pathway of interest are mutagenized and coexpressed with the target construct. Individual clones are screened for expresion of the gene product.
  • the secretory gene clusters from improved clones are recovered and recloned and introduced back into the original host. Preferably, they are first subjected to mutagenesis before the process is repeated. This cycle is repeated until the desired improvement in expression of secreted protein is achieved.
  • Phage display O'Neil et al., Current Biology 5:443-449 (1995) and the other display formats (Gates et al., J. Mol. Biol. 255:373-386 (1995); Mattheakis et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 91:9022-9026 (1994)) described herein represent general methodologies for applying affinity-based selections to proteins of interest. For example, Matthews and Wells ( Science 260:1113-1117 (1993)) have used phage display of a protease substrate to select improved substrates.
  • an enzyme used in antibiotic biosynthesis can be evolved for new substrate specificity and activity under desired conditions using phage display selections.
  • Some antibiotics are currently made by chemical modifications of biologically produced starting compounds. Complete biosynthesis of the desired molecules is currently impractical because of the lack of an enzyme with the required enzymatic activity and substrate specificity (Skatrud, TIBTECH 10:324-329, September 1992).
  • 7-aminodeacetooxycephalosporanic acid (7-ADCA) is a precursor for semi-synthetically produced cephalosporins. 7-ADCA is made by a chemical ring expansion of penicillin G followed by enzymatic deacylation of the phenoxyacetal group.
  • 7-ADCA can be made enzymatically from deacetylcephalosporin C (DAOC V), which could in turn be derived from penicillin V by enzymatic ring expansion if a suitably modified penicillin expandase could be evolved (Cantwell et al., Curr. Genet. 17:213-221 (1990)).
  • DAOC V deacetylcephalosporin C
  • 7-ADCA could in principle 35 be produced enzymatically from penicillin V using a modified penicillin N expandase, such as mutant forms of the S. clavuligerus cefE gene (Skatrud, TIBTECH 10:324-329, September 1992).
  • penicillin V is not accepted as a substrate by any known expandase with sufficient efficiency to be commercially useful.
  • RSR techniques of the invention can be used to evolve the penicillin expandase encoded by cefE or other expandases so that they will use penicillin V as a substrate.
  • Phage display or other display format selections are applied to this problem by expressing libraries of cefE penicillin expandase mutants in a display format, selecting for binding to substrates or transition state analogs, and applying RSR to rapidly evolve high affinity binders. Candidates are further screened to identify mutants with improved enzymatic activity on penicillin V under desired reaction conditions, such as pH, temperature, solvent concentration, etc. RSR is applied to further evolve mutants with the desired expandase activity. A number of transition state analogs (TSA's) are suitable for this reaction. The following structure is the initial TSA that is used for selection of the display library of cefE mutants:
  • candidate mutants are expressed in a high throughput format and specific activity for expanding penicillin V to DAOC V is quantitatively measured. Recombinants with improved enzymatic activity are mutagenized and the process repeated to further evolve them.
  • Retention of TSA binding by a displayed enzyme is a good selection for retention of the overall integrity of the active site and hence can be exploited to select for mutants which retain activity under conditions of interest.
  • Such conditions include but are not limited to: different pH optima, broader pH optima, activity in altered solvents such as DMSO (Seto et al., DNA Sequence 5:131-140 (1995)) or formamide (Chen et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 90:5618-5622, (1993)) altered temperature, improved shelf life, altered or broadened substrate specificity, or protease resistance.
  • DMSO Seto et al., DNA Sequence 5:131-140 (1995)
  • formamide Choen et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 90:5618-5622, (1993)
  • altered temperature improved shelf life
  • altered or broadened substrate specificity or proteas
  • thermostable DNA polymerase for more efficient mutagenesis or as a useful correlate for acceptance of base analogs for the purposes described above; higher fidelity polymerase for PCR (Lundberg et al., Gene 108:1-6 (1991)); higher fidelity reverse transcriptase for retroviral gene therapy vehicles to reduce mutation of the therapeutic construct and of the retrovirus; improved PCR of GC rich DNA and PCR with modified bases (S. Turner and F. J. Jenkins, BioTechniques 19(1):48-52 (1995)).
  • mutant libraries of mutant polymerase genes are screened by direct high throughput screening for improved sequencing properties. The best candidates are then subjected to RSR.
  • mutant libraries of candidate polymerases such as Taq polymerase are constructed using standard methods such as PCR mutagenesis (Caldwell et al., PCR Meth. App. 2:28-33 (1992)) and/or cassette mutagenesis (Sambrook et al., Molecular Cloning, CSH Press (1987)). Incorporation of mutations into Taq DNA polymerase such as the active site residue from T7 polymerase that improves acceptance of dideoxy nucleotides (Tabor and Richardson, J. Biol. Chem.
  • a high throughput, small scale simple purification for polymerase expressed in each well is performed.
  • simple single-step purifications of His-tagged Taq expressed in E. coli have been described (Smirnov et al., Russian J. Bioorganic Chem. 21(5):341-342 (1995)), and could readily be adapted for a 96-well expression and purification format.
  • a high throughput sequencing assay is used to perform sequencing reactions with the purified samples.
  • the data is analyzed to identify mutants with improved sequencing properties, according to any of these criteria: higher quality ladders on GC-rich templates, especially greater than 60% GC, including such points as fewer artifactual termination products and stronger signals than given with the wild-type enzyme; less termination of reactions by inosine in primer labelled reactions, e.g., fluorescent labelled primers; less variation in incorporation of signals in reactions with fluorescent dideoxy nucleotides at any given position; longer sequencing ladders than obtained with the wild-type enzyme, such as about 20 to 100 nucleotides; improved acceptance of other known base analogs such as 7-deaza purines; improved acceptance of new base analogs from combinatorial chemistry libraries (See, for example, Hogan, Nature 384(Supp):17-1996).
  • a screen or selection is performed as follows.
  • the replication of a plasmid can be placed under obligate control of a polymerase expressed in E. coli or another microorganism.
  • the effectiveness of this system has been demonstrated for making plasmid replication dependent on mammalian polymerase beta (Sweasy et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 90:4626-4630, (1993)), Taq polymerase (Suzuki et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 93:9670-9675 (1996)), or HIV reverse transcriptase (Kim et al., Proc. Natl.
  • the mutant polymerase gene is placed on a plasmid bearing a colE1 origin and expressed under the control of an arabinose promoter.
  • the library is enriched for active polymerases essentially as described by Suzuki et al., (supra), with polymerase expression being induced by the presence of arabinose in the culture.
  • a further quantitative screen utilizes the presence of GFP (green fluorescence protein) on the same plasmid, replica plating onto arabinose at the nonpermissive temperature in the absence of a selective antibiotic, and using a fluorimeter to quantitatively measure fluorescence of each culture.
  • GFP activity correlates with plasmid stability and copy number which is in turn dependent on expression of active polymerase.
  • a polymerase with a very high error rate would be a superior sequencing enzyme, as it would have a more normalized signal for incorporation of base analogs such as the currently used fluorescently labelled dideoxies because it will have reduced specificity and selectivity.
  • the error rates of currently used polymerases are on the order of 10 ⁇ 5 to 10 ⁇ 6 , orders of magnitude lower than what can be detected given the resolving power of the gel systems.
  • An error rate of 1%, and possibly as high as 10%, could not be detected by current gel systems, and thus there is a large window of opportunity to increase the “sloppiness” of the enzyme.
  • An error-prone cycling polymerase would have other uses such as for hypermutagenesis of genes by PCR.
  • the system described by Suzuki is used to make replication of a reporter plasmid dependent on the expressed polymerase.
  • This system puts replication of the first 200-300 bases next to the ColE1 origin directly under the control of the expressed polymerase (Sweasy and Loeb, J. Bact. 177:2923-2925 (1995); Sweasy et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 90:4626-4630 (1993)).
  • a screenable or selectable reporter gene containing stop codons is positioned in this region, such as LacZ alpha containing one, two or three stop codons.
  • the constructs are grown on arabinose at the nonpermissible temperature, allowed to recover, and plated on selective lactose minimal media that demands reversion of the stop codons in the reporter cassette.
  • Mutant polymerases are recovered from the survivors by PCR. The survivors are enriched for mutators because their mutator phenotype increases the rate of reversion of stop codons in the reporter lacZ alpha fragment.
  • the polymerase genes from the survivors are subjected to RSR, then the polymerase mutants are retransformed into the indicator strain.
  • Mutators can be visually screened by plating on arabinose/Xgal plates at the nonpermissive temperature. Mutator polymerases will give rise to colonies with a high frequency of blue papillae due to reversion of the stop codon(s).
  • Candidate papillators can be rescreened by picking a non-papillating region of the most heavily papillated colonies (i.e, “best” colonies) and replating on the arabinose/Xgal indicator medium to further screen for colonies with increased papillation rates. These steps are repeated until a desired reversion rate is achieved (e.g., 10 ⁇ 2 to 10 ⁇ 3 mutations per base pair per replication).
  • Colonies which exhibit high frequency papillation are candidates for encoding an error prone polymerase. These candidates are screened for improved sequencing properties: essentially as for the high throughput screen described above. Briefly, mutant Taq proteins are expressed and purified in a 96-well format. The purified proteins are used in sequencing reactions and the sequence data are analyzed to identify mutants that exhibit the improvements outlined herein. Mutants with improved properties are subjected to RSR and rescreened for further improvements in function.
  • GFP containing stop codons instead of lacZ alpha with stop codons is used for the construction.
  • Cells with reverted stop codons in GFP are selected by fluorescence activated cell sorter (FACS).
  • FACS selection is performed by gating the brightest about 0.1-10%, preferably the top 0.1 to 1%, and collected according to a protocol similar to that of Dangl et al., ( Cytometry 2(6):395-401 (1982)).
  • the polA gene is flanked with lox sites or other targets of a site specific recombinase.
  • the recombinase is induced, thus allowing one to inducibly delete the polA gene (Mulbery et al., Nucleic Acid Res. 23:485-490 (1995)) This would allow one to perform “Loeb-type” selections at any temperature and in any host. For example, one could set up such a selection in a recA deficient mesophile or thermophile by placing the polA homologue in an inducibly deletable format and thus apply the selection for active polymerase under more general conditions.
  • this general system is preferred for directed in vivo mutagenesis of genes.
  • the target gene is cloned into the region near a plasmid origin of replication that puts its replication obligately under control of the error prone polymerase.
  • the construct is passaged through a polA(ts) recA strain and grown at the nonpermissive temperature, thus specifically mutagenizing the target gene while replicating the rest of the plasmid with high fidelity.
  • selection is based on the ability of mutant DNA polymerases to PCR amplify DNA under altered conditions or by utilizing base analogs.
  • the mutant polymerases act on the template that encodes them in a PCR amplification, thus differentially replicating those polymerases.
  • an initial library of mutants is replica plated.
  • Polymerase preparations are done in a 96-well format. Crude plasmid preparations are made of the same set.
  • Each plasmid prep is PCR-amplified using the polymerase prep derived from that plasmid under the conditions for which one wishes to optimize the polymerase (e.g., added DMSO or formamide, altered temperature of denaturation or extension, altered buffer salts, PCR with base analogs such a-thiol dNTP's for use with mass spectroscopy sequencing, PCR of GC rich DNA (>60% GC),PCR with novel base analogs such as 7-deaza purines, 2′ fluoro dNTP's, rNTP's, PCR with inosine, etc.).
  • the amplified genes are pooled, cloned,and subjected to mutagenesis, and the process repeated until an improvement is achieved.
  • Alkaline phosphatase is a widely used reporter enzyme for ELISA assays, protein fusion assays, and in a secreted form as a reporter gene for mammalian cells.
  • the chemical lability of p-nitrophenyl phosphate (pNPP) substrates and the existence of cellular phosphatases that cross-react with pNPP is an important limitation on the sensitivity of assays using this reporter gene.
  • a reporter gene with superior signal to noise properties can be developed based on hydrolysis of p-nitrophenyl phosphonates, which are far more stable to base catalyzed hydrolysis than the corresponding phosphates.
  • Proteases and lipases are added in large quantities to detergents to enzymatically degrade protein and lipid stains on clothes.
  • the incorporation of these enzymes into detergents has significantly reduced the need for surfactants in detergents with a consequent reduction in the cost of formulation of detergents and improvement in stain removal properties.
  • Proteases with improved specific activity, improved range of protein substrate specificity, improved shelf life, improved stability at elevated temperature, and reduced requirements for surfactants would add value to these products.
  • subtilisin can be evolved as follows.
  • the cloned subtilisin gene (von der Osten et al., J. Biotechnol. 28:55-68 (1993)) can be subjected to RSR using growth selections on complex protein media by virtue of secreted subtilisin degrading the complex protein mixture.
  • libraries of subtilisin mutants are constructed in an expression vector which directs the mutant protein to be secreted by Bacillus subtilus.
  • Bacillus hosts transformed with the libraries are grown in minimal media with complex protein formulation as carbon and/or nitrogen source.
  • Subtilisin genes are recovered from fast growers and subjected to RSR, then screened for improvement in a desired property.
  • selection for improved proteases is performed as follows.
  • a library of mutant protease genes is constructed on a display phage and the phage grown in a multiwell format or on plates.
  • the phage are overlayed with a “protein net” which ensnares the phage.
  • the net can consist of a protein or proteins engineered with surface disulphides and then crosslinked with a library of peptide linkers.
  • a further embodiment employs an auxiliary matrix to further trap the phage.
  • the phage are further incubated, then washed to collect liberated phage wherein the displayed protease was able to liberate the phage from the protein net.
  • the protease genes are then subjected to RSR for further evolution.
  • a further embodiment employs a library of proteases encoded by but not displayed on a phagemid wherein streptavidin is fused to pIII by a peptide linker.
  • the library of protease mutants is evolved to cleave the linker by selecting phagemids on a biotin column between rounds of amplification.
  • the protease is not necessarily provided in a display format.
  • the host cells secrete the protease encoded by but not surface diplayed by a phagemid, while constrained to a well, for example, in a microtiter plate.
  • Phage display format is preferred where an entire high titre lysate is encased in a protein net matrix, and the phage expressing active and broad specificity proteases digesting the matrix to be liberated for the next round of amplification, mutagenesis, and selection.
  • the phage are not constrained to a well but, rather, protein binding filters are used to make a colony of plaque lifts and are screened for activity with chromogenic or fluorogenic substrates. Colonies or plaques corresponding to positive spots on the filters are picked and the encoded protease genes are recovered by, for example, PCR. The protease genes are then subjected to RSR for further evolution.
  • Peptide substrates containing fluoropores attached to the carboxy terminus and fluorescence quenching moities on the amino terminus such as those described by Holskin, et al, ( Anal. Biochem. 227:148-55 (1995)) (e.g., (4-4′-dimethylaminophenazo)benzoyl-arg-gly-val-val-asn-ala-ser-ser-arg-leu-ala-5-(2′-aminoethyl)-amino]-naphthalene-1-sulfonic acid) are used to screen protease mutants for broadened or altered specificity.
  • a library of peptide substrates is designed with a flourophore on the amino terminus and a potent fluorescence quencher on the carboxy terminus, or vice versa.
  • Supernatants containing secreted proteases are incubated either separately with various members of the library or with a complex cocktail. Those proteases which are highly active and have broad specificity will cleave the majority of the peptides, thus releasing the fluorophore from the quencher and giving a positive signal on a fluorimeter. This technique is amenable to a high density multiwell format.
  • Table I lists proteins that are of particular commercial interest to the pharmaceutical industry. These proteins are all candidates for RSR evolution to improve function, such as ligand binding, shelf life, reduction of side effects through enhanced specificity, etc. All are well-suited to manipulation by the techniques of the invention. Additional embodiments especially applicable to this list are described below.
  • mutants of Table I are screened for activity in a functional assay.
  • mutants of IL2 are screened for resistance to plasma or tissue proteases with retention of activity for the low affinity IL2 receptor but with loss of activity on the high affinity IL2 receptor.
  • the genes from mutants with improved activity relative to wild-type are recovered, and subjected to RSR to improve the phenotype further.
  • the libraries are generated in a display format such that the mature folded protein is physically linked to the genetic information that encodes it.
  • Examples include phage display using filamentous phage (O'Neil et al., Current Biology 5:443-449 (1995)) or bacteriophage lambda gene V display (Dunn, J. Mol. Biol. 248:497-506 (1995)), peptides on plasmids (Gates et al., J. Mol. Biol.
  • the following method can be used for selection.
  • a lysate of phage encoding IFN alpha mutants for example, can be used directly at suitable dilution to stimulate cells with a GFP reporter construct (Crameri et al., Nat. Med. 14:315-319 (1996)) under the control of an IFN responsive promoter, such as an MHC class I promoter. Phage remaining attached after stimulation, expression and FACS purification of the responsive cells, can be purified by FACS. Preferably, the brightest cells are collected. The phage are collected and their DNA subjected to RSR until the level of desired improvement is achieved.
  • IL-3 is prepared in one of these display formats and subjected to RSR to evolve an agonist with a desired level of activity.
  • a library of IL3 mutants on a filamentous phage vector is created and affinity selected (“panned”) against purified IL3 receptor to obtain mutants with improved affinity.
  • the mutant IL-3 genes are recovered by PCR, subjected to RSR, and recloned into the display vector. The cycle is repeated until the desired affinity or agonist activity is achieved.
  • the display formats descibed above preferentially are applied to a single chain version of the protein. Mutagenesis, such as RSR, can be used in these display formats to evolve improved single chain derivatives of multimeric factors which initially have low but detectable activity. This strategy is described in more detail below.
  • the eukaryotic cell is the unit of biological selection.
  • the following general protocol can be used to apply RSR to the improvement of proteins using eukaryotic cells as the unit of selection: (1) transfection of libraries of mutants into a suitable host cell, (2) expression of the encoded gene product(s) either transiently or stably, (3) functional selection for cells with an improved phenotype (expression of a receptor with improved affinity for a target ligand; viral resistance, etc., (4) recovery of the mutant genes by, for example, PCR followed by preparation of HIRT supernatants with subsequent tranformation of E. coli, (5) RSR and (6) repetition of steps (1)-(5) until the desired degree of improvement is achieved.
  • the affinity of an antibody for a ligand can be improved using mammalian surface display and RSR.
  • Antibodies with higher affinity for their cognate ligands are then screened for improvement of one or more of the following properties: (1) improved therapeutic properties (increased cell killing, neutralization of ligands, activation of signal transduction pathways by crosslinking receptors), (2) improved in vivo imaging applications (detection of the antibody by covalent/noncovalent binding of a radionuclide or any agent detectable outside of the body by noninvasive means, such as NMR), (3) improved analytical applications (ELISA detection of proteins or small molecules), and (4) improved catalysts (catalytic antibodies).
  • the methods described are general and can be extended to any receptor-ligand pair of interest. A specific example is provided in the experimental section.
  • Retroviral vectors provide a second alternative, but they are limited in the size of acceptable insert ( ⁇ 10 kb) and consistent, high expression levels are sometimes difficult to achieve. Random integration results in varying expression levels, thus introducing noise and limiting one's ability to distinguish between improvements in the affinity of the mutant protein vs. increased expression.
  • a related class of strategies that can be used effectively to achieve “one gene-one cell” DNA transfer and consistent expression levels for RSR is to use a viral vector which contains a lox site and to introduce this into a host that expresses cre recombinase, preferably transiently, and contains one or more lox sites integrated into its genome, thus limiting the variability of integration sites (Rohlman et al. Nature Biotech. 14:1562-1565 (1996)).
  • An alternative strategy is to transfect with limiting concentrations of plasmid (i.e., about one copy per cell) using a vector that can replicate in the target cells, such as is the case with plasmids bearing SV40 origins transfected into COS cells.
  • This strategy requires that either the host cell or the vector supply a replication factor such as SV40 large T antigen.
  • Northrup et al. J. Biol. Chem. 268:2917-2923 (1993) describe a strategy wherein a stable transfectant expressing SV40 large T antigen is then transfected with vectors bearing SV40 origins.
  • Transient expression i.e, non-integrating plasmids is a preferred format for cellular display selections because it reduces the cycle time and increases the number of mutants that can be screened.
  • the target cell is transfected with GFP cloned onto a vector containing SV40 large T antigen, an SV40 origin, and a reporter gene such as GFP; a related format is cotransfection with limiting amounts of the SV40 large T antigen expression vector and an excess of a reporter such as GFP cloned onto an SV40 origin containing plasmid.
  • a reporter gene such as GFP
  • the brightest cells are purified by FACS.
  • SV40 large T antigen mutants are recovered by PCR, and subjected to mutagenesis. The cycle is repeated until the desired level of improvement is obtained.
  • mutant proteins are selected or screened based on their ability to exert a biological effect in an autocrine fashion on the cell expressing the mutant protein.
  • a library of alpha interferon genes can be selected for induction of more potent or more specific antiviral activity as follows.
  • a library of interferon alpha mutants is generated in a vector which allows for induction of expression (i.e. under control of a metallothionein promoter) and efficient secretion in a multiwell format (96-well for example) with one or a few independent clones per well.
  • the promoter is not inducible,and may be constitutive.
  • cloned interferon genes are induced.
  • the cells are challenged with a cytotoxic virus against which one wishes to evolve an optimized interferon (for example vesicular stomatitus virus or HIV).
  • an optimized interferon for example vesicular stomatitus virus or HIV.
  • Surviving cells are recovered.
  • the cloned interferon genes are recovered by PCR amplification, subjected to RSR, and cloned back into the transfection vector and retransfected into the host cells. These steps are repeated until the desired level of antiviral activity is evolved.
  • the virus of interest is not strongly cytotoxic.
  • a conditionally lethal gene such as herpes simplex virus thymidine kinase
  • thymidine kinase is cloned into the virus and after challenge with virus and recovery, conditionally lethal selective conditions are applied to kill cells that are infected with virus.
  • An example of a conditionally lethal gene is herpes TK, which becomes lethal upon treating cells expressing this gene with the thymidine analog acyclovir.
  • the antiproliferative activity of the cloned interferons is selected by treating the cells with agents that kill dividing cells (for example, DNA alkylating agents).
  • potent cytokines are selected by expressing and secreting a library of cytokines in cells that have GFP or another reporter under control of a promoter that is induced by the cytokine, such as the MHC class I promoter being induced by evolved variants of alpha interferon.
  • the signal transduction pathway is configured such that the wild type cytokine to be evolved gives a weak but detectable signal.
  • proteins are evolved by RSR to have improved half life in serum.
  • a preferred method for improving half-life is evolving the affinity of a protein of interest for a long lived serum protein, such as an antibody or other abundant serum protein. Examples of how affinity for an antibody can enhance serum half life include the co-administration of IL2 and anti-IL2 antibodies which increases serum half-life and anti-tumor activity of human recombinant IL2 (Courtney et al., Immunopharmacology 28:223-232 (1994)).
  • the eight most abundant human serum proteins are serum albumin, immunoglobulins, lipoproteins, haptoglobin, fibrinogen, transferrin, alpha-1 antitrypsin, and alpha-2 macroglobulin (Doolittle, chapter 6, The Plasma Proteins F. Putnam, ed.; Academic Press, 1984). These and other abundant serum proteins such as ceruloplasmin and fibronectin are the primary targets against which to evolve binding sites on therapeutic proteins such as in Table I for the purpose of extending half-life. In the case of antibodies, the preferred strategy is to evolve affinity for constant regions rather than variable regions in order to minimize individual variation in the concentration of the relevant target epitope (antibody V region usage between different individuals is significantly variable).
  • Binding sites of the desired affinity are evolved by applying phage display, peptides on plasmid display or polysome display selections to the protein of interest.
  • half life is improved by derivatization with PEG, other polymer conjugates or half-life extending chemical moieties.
  • PEG polymer conjugates or half-life extending chemical moieties.
  • derivatization can also result in reduced affinity of the therapeutic protein for its receptor or ligand.
  • RSR is used to discover alternative sites in the primary sequence that can be substituted with lysine or other appropriate residues for chemical or enzymatic conjugation with half-life extending chemical moieties, and which result in proteins with maximal retention of biological activity.
  • a preferred strategy is to express a library of mutants of the protein in a display format, derivatize the library with the agent of interest (i.e. PEG) using chemistry that does not biologically inactivate the display system, select based on affinity for the cognate receptor, PCR amplify the genes encoding the selected mutants, shuffle, reassemble, reclone into the display format, and iterate until a mutant with the desired activity post modification is obtained.
  • An alternative format is to express, purify and derivatize the mutants in a high throughput format, screen for mutants with optimized activity, recover the corresponding genes, subject the genes to RSR and repeat.
  • binding sites for target human proteins that are localized in particular tissues of interest are evolved by RSR.
  • an interferon that localizes efficiently to the liver can be engineered to contain a binding site for a liver surface protein such as hepatocyte growth factor receptor.
  • a liver surface protein such as hepatocyte growth factor receptor.
  • affinity for abundant epitopes on erythrocytes such as ABO blood antigens to localize a given protein to the blood stream.
  • the protein of interest is evolved to have increased stability to proteases.
  • the clinical use of IL2 is limited by serious side effects that are related to the need to administer high doses.
  • High doses are required due to the short half life (3-5 min, Lotze et al., JAMA 256(22):3117-3124 (1986)) and the consequent need for high doses to maintain a therapeutic level of IL2.
  • One of the factors contributing to short half-lives of therapeutic proteins is proteolysis by serum proteases.
  • Cathepsin D a major renal acid protease, is responsible for the degradation of IL2 in Balb/c mice (Ohnishi et al., Cancer Res. 50:1107-1112 (1990)).
  • a preferred protocol is as follows.
  • a library of the mutagenized protein of interest is expressed in a display system with a gene-distal epitope tag (i.e. on the N-terminus of a phage display construct such that if it is cleaved off by proteases, the epitope tag is lost).
  • the expressed proteins are treated with defined proteases or with complex cocktails such as whole human serum.
  • Affinity selection with an antibody to the gene distal tag is performed.
  • a second selection demanding biological function e.g., binding to cognate receptor
  • Phage retaining the epitope tag and hence protease resistant
  • the process is repeated until the desired level of resistance is attained.
  • the procedure is performed in a screening format wherein mutant proteins are expressed and purified in a high throughput format and screened for protease resistance with retention of biological activity.
  • the protein of interest is evolved to have increased shelf life.
  • a library of the mutagenized nucleic acid squence encoding the protein of interest is expressed in a display format or high throughput expression format, and exposed for various lengths of time to conditions for which one wants to evolve stability (heat, metal ions, nonphysiological pH of, for example, ⁇ 6 or >8, lyophilization, freeze-thawing).
  • Genes are recovered from from survivors, for example, by PCR.
  • the DNA is subjected to mutagenesis, such as RSR, and the process repeated until the desired level of improvement is achieved.
  • the substrate for evolution by RSR is preferably a single chain contruction.
  • the possibility of performing asymetric mutagenesis on constructs of homomultimeric proteins provides important new pathways for further evolution of such constructs that is not open to the proteins in their natural homomultimeric states.
  • a given mutation in a homomultimer will result in that change being present in each identical subunit.
  • the domains can mutate independently of each other.
  • a single chain version of urokinase-type plasminogen activator has been made, and it has been shown that the single chain construct is more resistant to plasminogen activator inhibitor type 1 than the native homodimer (Higazi et al., Blood 87:3545-3549 (1996)).
  • a linker is constructed which joins the amino terminus of one subunit of a protein of interest to the carboxyl terminus of another subunit in the complex.
  • These fusion proteins can consist of linked versions of homodimers, homomultimers, heterodimers or higher order heteromultimers.
  • one adds polypeptide linkers between the native termini to be joined. Two significant variations can be made. First, one can construct diverse libraries of variations of the wild type sequence in and around the junctions and in the linkers to facilitate the construction of active fusion proteins.
  • a population of selected mutant single chain constructs is PCR amplified in two seprate PCR reactions such that each of the two domains is amplified separately.
  • Oligonucleotides are derived from the 5′ and 3′ ends of the gene and from both strands of the linker.
  • the separately amplified domains are shuffled in separate reactions, then the two populations are recombined using PCR reassembly to generate intact single chain constructs for further rounds of selection and evolution.
  • Other examples include evolved interferon alpha variants that arrest tumor cell proliferation but do not stimulate NK cells, IL2 variants that stimulate the low affinity IL2 receptor complex but not the high affinity receptor (or vice versa), superantigens that stimulate only a subset of the V beta proteins recognized by the wild type protein (preferably a single V beta), antagonistic forms of chemokines that specifically antagonize only a receptor of interest, antibodies with reduced cross-reactivity, and chimeric factors that specifically activate a particular receptor complex.
  • chemokines that specifically antagonize only a receptor of interest
  • antibodies with reduced cross-reactivity chimeric factors that specifically activate a particular receptor complex.
  • chimeric factors that specifically activate a particular receptor complex.
  • a preferred strategy is the selection or screening for mutants with increased agonist activity using the whole cell formats described above, combined with RSR.
  • a library of mutants of IL3 is expressed in active form on phage as described by Gram et al. ( J. Immun. Meth. 161:169-176 (1993)).
  • Clonal lysates resulting from infection with plaque-purified phage are prepared in a high through-put format such as a 96-well microtiter format.
  • An IL3-dependent cell line expressing a reporter gene such as GFP is stimulated with the phage lysates in a high throughput 96-well.
  • Phage that result in positive signals at the greatest dilution of phage supernatants are recovered; alternatively, DNA encoding the mutant IL3 can be recovered by PCR.
  • single cells expressing GFP under control of an IL3 responsive promoter can stimulated with the IL3 phage library, and the positive FACS sorted. The nucleic acid is then subjected to PCR, and the process repeated until the desired level of improvement is obtained.
  • Atrial natriuretic factor [0354] Atrial natriuretic factor
  • Atrial peptides [0356] Atrial peptides
  • C-X-C chemokines e.g., T39765, NAP-2, ENA-78, Gro-a, Gro-b, Gro-c, IP-10, GCP-2, NAP-4, SDF-1, PF4, MIG
  • CC chemokines e.g., Monocyte chemoattractant protein-1, Monocyte chemoattractant protein-2, Monocyte chemoattractant protein-3, Monocyte inflammatory protein-1 alpha, Monocyte inflammatory protein-1 beta, RANTES, I309, R83915, R91733, HCC1, T58847, D31065, T64262
  • CSF Colony stimulating factor
  • Hedgehog proteins e.g., Sonic, Indian, Desert
  • Hemoglobin for blood substitute; for radiosensitization
  • NAF Neutrophil inhibitory factor
  • Soluble interleukin receptors (IL-1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15)
  • Superantigens i.e., Staphylococcal enterotoxins (SEA, SEB, SEC1, SEC2, SEC3, SED, SEE), Toxic shock syndrome toxin (TSST-1), Exfoliating toxins A and B, Pyrogenic exotoxins A, B, and C, and M. arthritidis mitogen
  • TNF beta Tumor necrosis factor beta
  • TNFR Tumor necrosis factor receptor
  • TNF alpha Tumor necrosis factor-alpha
  • RSR can be used in several ways to modify eukaryotic signal transduction or transcriptional pathways. Any component of a signal transduction pathway of interest, of the regulatory regions and transcriptional activators that interact with this region and with chemicals that induce transcription can be evolved. This generates regulatory systems in which transcription is activated more potently by the natural inducer or by analogues of the normal inducer. This technology is preferred for the development and optimization of diverse assays of biotechnological interest.
  • dozens of 7 transmembrane receptors (7-TM) are validated targets for drug discovery (see, for example, Siderovski et al., Curr Biol., 6(2):211-212 (1996); An et al., FEBS Lett., 375(1-2):121-124 (1995); Raport et al., Gene, 163(2):295-299 (1995); Song et al., Genomics, 28(2):347-349 (1995); Strader et al. FASEB J., 9(9) :745-754 (1995); Benka et al., FEBS Lett., 363(1-2):49-52 (1995); Spiegel, J. Clin Endocrinol.
  • biodetectors or biosensors for different chemicals can be developed by evolving 7-TM's to respond agonistically to novel chemicals or proteins of interest. In this case, selection would be for contructs that are activated by the new chemical or polypeptide to be detected. Screening could be done simply with fluorescence or light activated cell sorting, since the desired improvement is coupled to light production.
  • biosensors can be developed that will respond to any chemical for which there are receptors, or for which receptors can be evolved by recursive sequence recombination, such as hormones, growth factors, metals and drugs.
  • the receptors may be intracellular and direct activators of transcription, or they may be membrane bound receptors that activate transcription of the signal indirectly, for example by a phosphorylation cascade. They may also not act on transcription at all, but may produce a signal by some post-transcriptional modification of a component of the signal generating pathway. These receptors may also be generated by fusing domains responsible for binding different ligands with different signalling domains.
  • recursive sequence recombination can be used to increase the amplitude of the signal generated to optimize expression and functioning of chimeric receptors, and to alter the specificity of the chemicals detected by the receptor.
  • G proteins can be evolved to efficiently couple mammalian 7-TM receptors to yeast signal transduction pathways.
  • G alpha protein loci in mammals which can be grouped by sequence and functional similarity into four groups, Gs (Gna, Gna1), Gi (Gnai-2, Gnai-3, Gnai-1, Gnao, Gnat-1, Gnat-2, Gnaz), Gq (Gnaq, Gna-11, Gna-14, Gna-15) and G12 (Gna-12, Gna-13) (B. Nurnberg et al., J. Mol. Med., 73:123-132 (1995)).
  • G alpha proteins are complexed noncovalently with G beta and G gamma proteins as well as to their cognate 7-TM receptor(s). Receptor and signal specificity are controlled by the particular combination of G alpha, G beta (of which there are five known loci) and G gamma (seven known loci) subunits. Activation of the heterotrimeric complex by ligand bound receptor results in dissociation of the complex into G alpha monomers and G beta, gamma dimers which then transmit signals by associating with downstream effector proteins.
  • the G alpha subunit is believed to be the subunit that contacts the 7-TM, and thus it is a focal point for the evolution of chimeric or evolved G alpha subunits that can transmit signals from mammalian 7-TM's to yeast downstream genes.
  • Yeast based bioassays for mammalian receptors will greatly facilitate the discovery of novel ligands.
  • Kang et al. Mol. Cell Biol. 10:2582-2590 (1990)
  • SCG1 SCG1
  • yeast/mammalian G alpha proteins These hybrids have partial function, such as complementing the growth defect in scg1 strains, but do not allow mating and hence do not fully complement function in the pheromone signal transduction pathway.
  • Price et al. Mol. Cell Biol.
  • Such strains are useful as reporter strains for mammalian receptors, but suffer from important limitations as exemplified by the study of Kang et al., where there appears to be a block in the transmission of signals from the yeast pheromone receptors to the mammalian G proteins.
  • a mammalian 7-TM receptor to yeast signal transduction pathways one couples the mammalian receptor to yeast, mammalian, or chimeric G alpha proteins, and these will in turn productively interact with downstream components in the pathway to induce expression of a pheromone responsive promoter such as FUS-1.
  • Such functional reconstitution is commonly referred to as “coupling”.
  • the methods described herein can be used to evolve the coupling of mammalian 7-TM receptors to yeast signal transduction pathways.
  • a typical approach is as follows: (1) clone a 7-TM of interest into a yeast strain with a modified pheromone response pathway similar to that described by Price (e.g., strains deficient in FAR1, a negative regulator of G 1 cyclins, and deficient in SST2 which causes the cells to be hypersensitive to the presence of pheromone), (2) construct libraries of chimeras between the mammalian G alpha protein(s) known or thought to interact with the GPA1 or homologous yeast G alpha proteins, (3) place a selectable reporter gene such as HIS3 under control of the pheromone responsive promoter FUS1 (Price et al., Mol.
  • a screenable gene such as luciferase may be placed under the control of the FUS1 promoter; (4) transform library (2) into strain (3) (HIS( ⁇ )), (5) screen or select for expression of the reporter in response to the ligand of interest, for example by growing the library of transformants on minimal plates in the presence of ligand to demand HIS3 expression, (6) recover the selected cells, and and apply RSR to evolve improved expression of the reporter under the control of the pheromone responsive promoter FUS1.
  • a second important consideration in evolving strains with optimized reporter constructs for signal transduction pathways of interest is optimizing the signal to noise ratio (the ratio of gene expression under inducing vs noninducing conditions).
  • Many 7-TM pathways are leaky such that the maximal induction of a typical reporter gene is 5 to 10-fold over background. This range of signal to noise may be insufficient to detect small effects in many high through put assays. Therefore, it is of interest to couple the 7-TM pathway to a second nonlinear amplification system that is tuned to be below but near the threshold of activation in the uninduced state.
  • An example of a nonlinear amplification system is expression of genes driven by the lambda PL promoter.
  • GAL4 a limiting component of a transcription factor for such a promoter
  • An example of such a coupled system is to place GAL4 under control of the FUS-1 pheromone responsive promoter and to have the intracellular GAL4 (itself a transcriptional enhancer) level positively feedback on itself by placing a GAL4 binding site upstream of the FUS-1 promoter.
  • a reporter gene is also put under the control of a GAL4 activated promoter. This system is designed so that GAL4 expression will nonlinearly self-amplify and co-amplify expression of a reporter gene such as luciferase upon reaching a certain threshold in the cell.
  • RSR can be used to great advantage to evolve reporter constructs with the desired signaling properties, as follows: (1) A single plasmid construct is made which contains both the GAL4/pheromone pathway regulated GAL4 gene and the GAL4 regulated reporter gene. (2) This construct is mutagenized and transformed into the appropriately engineered yeast strain expressing a 7-TM and chimeric yeast/mammalian protein of interest. (3) Cells are stimulated with agonists and screened (or selected) based on the activity of the reporter gene. In a preferred format, luciferase is the reporter gene and activity is quantitated before and after stimulation with the agonist, thus allowing for a quantitative measurement of signal to noise for each colony. (4) Cells with improved reporter properties are recovered, the constructs are shuffled, and RSR is applied to further evolve the plasmid to give optimal signal noise characteristics.
  • a preferred strategy to evolve BIAP is as follows.
  • a codon usage libary is constructed from 60-mer oligonucleotides such that the central 20 bases of each oligo specifies the wild type protein, but encodes the wild-type protein sequence with degenerate codons.
  • very rare codons for the prokaryotic host of choice such as E. coli, are not used.
  • the 20 bases at each end of the oligo use non-degenerate, but preferred, codons in E. coli.
  • the oligonucleotides are assembled into full-length genes as described above. The assembled products are cloned into an expression vector by techniques well known in the art.
  • the codon usage library is expressed with a library of secretory leader sequences, each of which directs the encoded BIAP protein to the E. coli periplasm.
  • a library of leader sequences is used to optimize the combination of leader sequence and mutant. Examples of leader sequences are reviewed by Schatz et al. ( Ann Rev. Genet. 24:215-248 (1990)).
  • the cloned BIAP genes are expressed under the control of an inducible promoter such as the arabinose promoter. Arabinose-induced colonies are screened by spraying with a substrate for BIAP, bromo-chloro-indolyl phosphate (BCIP). The bluest colonies are picked visually and subjected to the RSR procedures described herein.
  • oligonucleotides for construction of the codon usage library are listed in Table II. The corresponding locations of these promoters is provided in FIG. 1. TABLE II 1. AACCCTCCAG TTCCGAACCC CATATGATGA TCACCCTGCG TAAACTGCCG 2. AACCCTCCAG TTCCGAACCC CATATGAAAA AAACCGCT 3. AACCCTCCAG TTCCGAACCC ATATACATAT GCGTGCTAAA 4. AACCCTCCAG TTCCGAACCC CATATGAAAT ACCTGCTGCC GACC 5. AACCCTCCAG TTCCGAACCC GATATACATA TGAAACAGTC 6.
  • TGGTGTTATG TCTGCTCAGG CDATGGCDGT DGAYTTYCAY CTGGTTCCGG TTGAAGAGGA 7.
  • GGCTGGTTTC GCTACCGTTG CDCARGCDGC DCCDAARGAY CTGGTTCCGG TTGAAGAGGA 8.
  • CACCCCGATC GCTATCTCTT CYTTYGCDTC YACYGGYTCY CTGGTTCCGG TTGAAGAGGA 9.
  • TGCCGCTGCT GTTCACCCCG GTDACYAARG CDGCDCARGT DCTGGTTCCG GTTGAAGAGG A 11.
  • GGGTTTCCGG ACCCAGTTTA CCRTTCATYT GRCCYTTCAG GATACGGGTA GCGGTAACGG 48.
  • affinity maturation of this antibody can be recapitulated and improved upon using cassette mutagenesis of the CDR's (or random mutagenesis such as with PCR), mammalian display, FACS selection for improved binding, and RSR to rapidly evolve improved affinity by recombining mutations encoding improved binding.
  • Genomic antibody expression shuttle vectors similar to those described by Gascoigne et al. are constructed such that libraries of mutant V region exons can be readily cloned into the shuttle vectors.
  • the kappa construct is cloned onto a plasmid encoding puromycin resistance and the heavy chain is cloned onto a neomycin resistance encoding vector.
  • the cDNA derived variable region sequences encoding the mature and germline heavy and light chain V regions are reconfigured by PCR mutagenesis into genomic exons flanked by Sfi I sites with complementary Sfi I sites placed at the appropriate locations in the genomic shuttle vectors.
  • the oligonucleotides used to create the intronic Sfi I sites flanking the VDJ exon are: 5′ Sfi I: 5′-TTCCATTTCA TACATGGCCG AAGGGGCCGT GCCATGAGGA TTTT-3′; 3′ Sfi I: 5′-TTCTAAATG CATGTTGGCC TCCTTGGCCG GATTCTGAGC CTTCAGGACC A-3′.
  • Stable transfectant lines are made for each of the two light and heavy chain constructs (mature and germline) using the B cell myeloma AG8-653 (a gift from J. Kearney) as a host using standard electroporation protocols.
  • Libraries of mutant plasmids encoding the indicated libraries of V-L mutants are transfected into the stable transformant expressing the germline V-H; and the V-H mutants are transfected into the germline V-L stable transfectant line.
  • the libraries are introduced by protoplast fusion (Sambrook et al., Molecular Cloning, CSH Press (1987)) to ensure that the majority of transfected cells receive one and only one mutant plasmid sequence (which would not be the case for electroporation where the majority of the transfected cells would receive many plasmids, each expressing a different mutant sequence).
  • JWJ-1 The p-nitrophenylphosphonate hapten (JWJ-1) recognized by this antibody is synthesized as described by Patten et al. ( Science 271:1086-1091 (1996)). JWJ-1 is coupled directly to 5-(((2-aminoethyl)thio)acetyl) fluorescein (Molecular Probes, Inc.) by formation of an amide bond using a standard coupling chemistry such as EDAC (March, Advanced Organic Chemistry, Third edition, John Wiley and Sons, 1985) to give a monomeric JWJ-1-FITC probe.
  • EDAC March, Advanced Organic Chemistry, Third edition, John Wiley and Sons, 1985
  • a “dimeric” conjugate (two molecules of JWJ-1 coupled to a FACS marker) is made in order to get a higher avidity probe, thus making low affinity interactions (such as with the germline antibody) more readily detected by FACS. This is generated by staining with Texas Red conjugated to an anti-fluorescein antibody in the presence of two equivalents of JWJ-1-FITC.
  • the bivalent structure of IgG then provides a homogeneous bivalent reagent.
  • a spin column is used to remove excess JWJ-1-FITC molecules that are not bound to the anti-FITC reagent.
  • a tetravalent reagent is made as follows.
  • biotin is coupled with EDAC to two equivalents of ethylenediamine, and this is then be coupled to the free carboxylate on JWJ-1.
  • the biotiylated JWJ-1 product is purified by ion exchange chromatography and characterized by mass spectrometry.
  • FITC labelled avidin is incubated with the biotinylated JWJ-1 in order to generate a tetravalent probe.
  • the FACS selection is performed as follows, according to a protocol similar to that of Panka et al. ( Proc. Natl. Acad. Sci. ( U.S.A. ) 85:3080-3084 (1988)). After transfection of libraries of mutant antibody genes by the method of protoplast fusion (with recovery for 36-72 hours), the cells are incubated on ice with fluorescently labelled hapten. The incubation is done on ice to minimize pinocytosis of the FITC conjugate which may contribute to nonspecific background. The cells are then sorted on the FACS either with or without a washing step.
  • FACSing without a washing step is preferable because the off rate for the germline antibody prior to affinity maturation is expected to be very fast (>0.1 sec-1; Patten et al., Science 271:1086-1091 (1996)); a washing step adds a complicating variable. The brightest 0.1-10% of the cells are collected.
  • Plasmids are recovered from the FACS selected cells by the transformation of an E. coli host with Hirt supernatants.
  • the mutant V gene exons are PCR-amplified from the FACS selected cells.
  • the recovered V gene exons are subjected to RSR, recloned into the corresponding genomic shuttle vector, and the procedure recursively applied until the mean fluorescence intensity has increased.
  • a relevant positive control for improved binding is transfection with the affinity matured 48G7 exons (Patten et al., op. cit.).
  • This methodology can be applied to evolve any receptor-ligand or binding partner interaction.
  • Natural expression formats can be used to express libraries of mutants of any receptor for which one wants to improve the affinity for the natural or novel ligands. Typical examples would be improvement of the affinity of T cell receptors for ligands of interest (i.e. MHC/tumor peptide antigen complexes) or TNF receptor for TNF (soluble forms of TNF receptors are used therapeutically to neutralize TNF activity).
  • This format can also be used to select for mutant forms of ligands by expressing the ligand in a membrane bound form with an engineered membrane anchor by a strategy analogous to that of Wettstein et al.( J. Exp. Med. 174:219-28 (1991)). FACS selection is then performed with fluorescently labelled receptor.
  • this format one could, for example, evolve improved receptor antagonists from naturally occurring receptor antagonists (IL1 receptor antagonist, for example). Mutant forms of agonists with improved affinity for their cognate receptors could also be evolved in this format. These mutants would be candidates for improved agonists or potent receptor antagonists, analogous to reported antagonistic mutant forms of IL3.
  • IFN- ⁇ non-allelic human interferon-alpha genes
  • IFN-Con1 A consensus human alpha interferon, IFN-Con1
  • IFN-Con1 A consensus human alpha interferon, IFN-Con1
  • IFN-Con1 A consensus human alpha interferon, IFN-Con1
  • IFN-Con1 A consensus human alpha interferon, IFN-Con1
  • IFN-Con1 A consensus human alpha interferon, IFN-Con1
  • IFN-Con1 has been constructed synthetically wherein the most common residue in fourteen known IFN- ⁇ 's has been put at each position, and it compares favorably with the naturally occurring interferons (Ozes et al., J. Interferon Res. 12:55-59 (1992)).
  • IFN-Con1 contains.20 amino acid changes relative to IFN- ⁇ 2a, the INF- ⁇ to which it is most closely related.
  • IFN-Con1 has 10-fold higher specific antiviral activity than any known natural IFN subtype.
  • IFN- ⁇ Con1 has in vitro activities 10 to 20 fold higher than that of recombinant IFN ⁇ -2a (the major IFN used clinically) in antiviral, antiproliferative and NK cell activation.
  • interferon hybrids which combine the most desirable traits from two or more interferons.
  • IFN- ⁇ 's The biological effects of IFN- ⁇ 's are diverse, and include such properties as induction of antiviral state (induction of factors that arrest translation and degrade mRNA); inhibition of cell growth; induction of Class I and Class II MHC; activation of monocytes and macrophages; activation of natural killer cells; activation of cytotoxic T cells; modulation of Ig synthesis in B cells; and pyrogenic activity.
  • the various IFN- ⁇ 's subtypes have unique spectra of activities on different target cells and unique side effect profiles (Ortaldo et al., Proc. Natl. Acad. Sci. ( U.S.A. ) 81:4926-4929 (1984); Overall et al., J. Interferon Res. 12:281-288 (1992); Fish and Stebbing, Biochem. Biophys. Res. Comm. 112:537-546 (1983); Weck et al., J. Gen. Virol. 57:233-237 (1981)).
  • human IFN ⁇ has very mild side effects but low antiviral activity.
  • Human IFN ⁇ 8 has very high antiviral activity, but relatively severe side effects.
  • Human IFN ⁇ 7 lacks NK activity and blocks NK stimulation by other INF ⁇ 's.
  • Human IFN- ⁇ J lacks the ability to stimulate NK cells, but it can bind to the IFN- ⁇ receptor on NK cells and block the stimulatory activity of IFN- ⁇ A (Langer et al., J. Interferon Res. 6:97-105 (1986)).
  • interferons are limited by diverse and severe side effect profiles which include flu-like symptoms, fatigue, neurological disorders including hallucination, fever, hepatic enzyme elevation, and leukopenia.
  • the multiplicity of effects of IFN- ⁇ 's has stimulated the hypothesis that there may be more than one receptor or a multicomponent receptor for the IFN- ⁇ family (R. Hu et al., J. Biol. Chem. 268:12591-12595 (1993)).
  • the existence of abundant naturally occurring diversity within the human alpha IFN's (and hence a large sequence space of recombinants) along with the complexity of the IFN- ⁇ receptors and activities creates an opportunity for the construction of superior hybrids.
  • 171 of the 175 changes can be incorporated into homologue libraries using single degenerate codons at the corresponding positions.
  • Arg, Trp and Gly can all be encoded by the degenerate codon [A,T,G]GG.
  • 1.3 ⁇ 10 25 hybrids can be captured with a single set of degenerate oligonucleotides.
  • 27 oligonucleotides is sufficient to shuffle all eleven human alpha interferons. Virtually all of the natural diversity is thereby encoded and fully permuted due to degeneracies in the nine “block” oligonucleotides in Table V.
  • oligo directed shuffling i.e. bridge oligonucleotides
  • the general methods described above are employed to screen or select these mutants for improved function.
  • exemplary descriptions of IFN based assays are described below.
  • libraries are constructed wherein the 11 homologous forms of the nine segments are permuted (note that in many cases two homologues are identical over a given segment). All nine segments are PCR- amplified out of all eleven IFN alpha genes with the eighteen oligonucleotides listed in Table IV, and reassembled into full length genes with oligo directed recombination. An arbitrary number, e.g., 1000, clones from the library are prepared in a 96-well expression/purification format. Hybrids with the most potent antiviral activities are screened. Nucleic acid is recovered by PCR amplification, and subjected to recombination using bridge oligonucleotides. These steps are repeated until candidates with desired properties are obtained.
  • each of the nine segments is synthesized with one degenerate oligo per segment.
  • Degeneracies are chosen to capture all of the IFN-alpha diversity that can be captured with a single degenerate codon without adding any non-natural sequence.
  • a second set of degenerate oligonucleotides encoding the nine segments is generated wherein all of the natural diversity is captured, but additional non-natural mutations are included at positions where necessitated by the constraints of the genetic code.
  • One way to make this approach more “gentle” would be to obtain a candidate starting point and to gently search from there.
  • This starting point could be either one of the natural IFN-alpha's (such as IFN alpha-2a which is the one that is being used most widely therapeutically), the characterized IFN-Con1 consensus interferon, or a hit from screening the shuffled IFN-alpha's described above.
  • IFN alpha-2a which is the one that is being used most widely therapeutically
  • the characterized IFN-Con1 consensus interferon or a hit from screening the shuffled IFN-alpha's described above.
  • Given a starting point one would make separate libraries wherein one breeds the degenerate segment libraries one at a time into the founder sequence. Improved hits from each library would then be bred together to gently build up mutations all throughout the molecule.
  • the following assays are used to screen IFN alpha mutants: inhibition of viral killing; standard error of 30-50%; inhibition of plaque forming units; very low standard error (can measure small effects); reduced viral yield (useful for nonlethal, nonplaque forming viruses); inhibition of cell growth (3H-thymidine uptake assay; activation of NK cells to kill tumor cells; suppression of tumor formation by human INF administered to nude mice engrafted with human tumors (skin tumors for example).
  • IFN alpha mutants are expressed and purified in high throughput formats such as expression, lysis and purification in a 96-well format using anti-IFN antibodies or an epitope tag and affinity resin.
  • the purified IFN preparations are screened in a high throughput format, scored, and the mutants encoding the highest activities of interest are subjected to further mutagenesis, such as RSR, and the process repeated until a desired level of activity is obtained.
  • Standard phage display formats are used to display biologically active IFN. Libraries of chimeric IFN genes are expressed in this format and are selected (positively or negatively) for binding (or reduced binding) to one or more purified IFN receptor preparations or to one or more IFN receptor expressing cell types.
  • Protein expressed by mutants can be screened in high throughput format on a reporter cell line which expresses GFP or luciferase under the control of an IFN alpha responsive promoter, such as an MHC Class I promoter driving GFP expression.
  • the range of concentration used in biological assays is: 1 ng/ml for NK activation, 0.1-10 ng/ml for antiproliferative activity on Eskol cells, and 0.1-1 ng/ml on Daudi cells (Ozes et al., J. Interferon Res. 12:55-59 (1992)).
  • interferon alpha2a and consensus interferon are expressed in active recombinant form in E. coli, so at least these two do not require glycosylation for activity.
  • IFN alpha expressed on filamentous phage is likely to be biologically active as phage lysates without further purification.
  • Libraries of IFN chimeras are expressed in phage display formats and scored in the assays described above and below to identify mutants with improved properties to be put into further rounds of RSR.
  • a phage lysate can be used directly at suitable dilution to stimulate cells with a GFP reporter construct under the control of an IFN responsive promoter. Assuming that the phage remain attached after stimulation, expression and FACS purification of the responsive cells, one could then directly FACS purify hybrids with improved activity from very large libraries (up to and perhaps larger than 10 7 phage per FACS run).
  • a second way in which FACS is used to advantage in this format is the following.
  • Cells can be stimulated in a multiwell format with one lysate per well and a GFP type reporter construct. All stimulated cells are FACS purified to collect the brightest cells, and the IFN genes recovered and subjected to RSR, with iteration of the protocol until the desired level of improvement is obtained. In this protocol the stimulation is performed with individual concentrated lysates and hence the requirement that a single phage be sufficient to stimulate the cell is relaxed. Furthermore, one can gate to collect the brightest cells which, in turn, should have the most potent phage attached to them.
  • a sample protocol follows for the cell surface display of IFN alpha mutants.
  • This form of display has at least two advantages over phage display.
  • the protein is displayed by a eukaryotic cell and hence can be expressed in a properly glycosylated form which may be necessary for some IFN alphas (and other growth factors).
  • it is a very high valency display format and is preferred in detecting activity from very weakly active mutants.
  • a library of mutant IFN's is constructed wherein a polypeptide signal for addition of a phosphoinositol tail has been fused to the carboxyl terminus, thus targeting the protein for surface expression (Wettstein et al., J. Exp. Med. 174:219-28 (1991)).
  • the library is used to transfect reporter cells described above (luciferase reporter gene) in a microtiter format. Positives are detected with a charge coupling device(CCD) camera. Nucleic acids are recovered either by HIRT and retransformation of the host or by PCR, and are subjected to RSR for further evolution.
  • a sample protocol follows for the autocrine display of IFN alpha mutants.
  • a library of IFN mutants is generated in a vector which allows for induction of expression (i.e. metallothionein promoter) and efficient secretion.
  • the recipient cell line carrying an IFN responsive reporter cassette [GFP or luciferase] is induced by transfection with the mutant IFN constructs. Mutants which stimulate the IFN responsive promoter are detected by by FACS or CCD camera.
  • Brackets indicate degeneracy with equal mixture of the specified bases at those positions.
  • the purpose of the degeneracy is to allow this one set of primers to prime all members of the IFN family with similar efficiency.
  • the choice of the oligo driven recombination points is important because they will get “overwritten” in each cycle of breeding and hence cannot coevolve with the rest of the sequence over many cycles of selection.
  • TABLE V Oligonucleotides needed for “fine grain” recombination of natural diversity over each of the nine blocks #Length of Block oligo required 1 76 2 95 3 65 4 56 5 51 6 93 7 50 8 62 9 80

Abstract

Methods are provided for the evolution of proteins of industrial and pharmaceutical interest, including methods for effecting recombination and selection. Compositions produced by these methods are also disclosed.

Description

  • This application is a continuation-in-part of U.S. patent application Ser. No. 08/198,431, filed Feb. 17, 1994, Ser. No. PCT/US95/02126, filed, Feb. 17, 1995, Ser. No. 08/425,684, filed Apr. 18, 1995, Ser. No. 08/537,874, filed Oct. 30, 1995, Ser. No. 08/564,955, filed Nov. 30, 1995, Ser. No. 08/621,859, filed Mar. 25, 1996, Ser. No. 08/621,430, filed Mar. 25, 1996, Ser. No. PCT/US96/05480, filed Apr. 18, 1996, Ser. No. 08/650,400, filed May 20, 1996, Ser. No. 08/675,502, filed Jul. 3, 1996, Ser. No. 08/721,824, filed Sep. 27, 1996, and Ser. No. 08/722,660 filed Sep. 27, 1996 the specifications of which are herein incorporated by reference in their entirety for all purposes.[0001]
  • BACKGROUND OF THE INVENTION
  • Recursive sequence recombination entails performing iterative cycles of recombination and screening or selection to “evolve” individual genes, whole plasmids or viruses, multigene clusters, or even whole genomes (Stemmer, [0002] Bio/Technology 13:549-553 (1995)). Such techniques do not require the extensive analysis and computation required by conventional methods for polypeptide engineering. Recursive sequence recombination allows the recombination of large numbers of mutations in a minimum number of selection cycles, in contrast to traditional, pairwise recombination events.
  • Thus, recursive sequence recombination (RSR) techniques provide particular advantages in that they provide recombination between mutations in any or all of these, thereby providing a very fast way of exploring the manner in which different combinations of mutations can affect a desired result. [0003]
  • In some instances, however, structural and/or functional information is available which, although not required for recursive sequence recombination, provides opportunities for modification of the technique. In other instances, selection and/or screening of a large number of recombinants can be costly or time-consuming. A further problem can be the manipulation of large nucleic acid molecules. The instant invention addresses these issues and others. [0004]
  • SUMMARY OF THE INVENTION
  • One aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule comprising: [0005]
  • (a) digesting at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide, with a restriction endonuclease; [0006]
  • (b) ligating the mixture to generate a library of recombinant DNA molecules; [0007]
  • (c) screening or selecting the products of (b) for a desired property; and [0008]
  • (d) recovering a recombinant DNA substrate molecule encoding an evolved protein. [0009]
  • A further aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide and comprise defined segments, the method comprising: [0010]
  • (a) providing a set of oligonucleotide PCR primers, comprising at least one primer for each segment, wherein the primer sequence is complementary to at least one junction with another segment; [0011]
  • (b) amplifying the segments of the at least a first and second DNA substrate molecules with the primers of step (a) in a polymerase chain reaction; [0012]
  • (c) assembling the products of step (b) to generate a library of recombinant DNA substrate molecules; [0013]
  • (d) screening or selecting the products of (c) for a desired property; and [0014]
  • (e) recovering a recombinant DNA substrate molecule from (d) encoding an evolved protein. [0015]
  • A further aspect of the invention is a method of enriching a population of DNA fragments for mutant sequences comprising: [0016]
  • (a) denaturing and renaturing the population of fragments to generate a population of hybrid double-stranded fragments in which at least one double-stranded fragment comprises at least one base pair mismatch; [0017]
  • (b) fragmenting the products of (a) into fragments of about 20-100 bp; [0018]
  • (c) affinity-purifying fragments having a mismatch on an affinity matrix to generate a pool of DNA fragments enriched for mutant sequences; and [0019]
  • (d) assembling the products of (c) to generate a library of recombinant DNA substrate molecules. [0020]
  • A further aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule, by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules share a region of sequence homology of about 10 to 100 base pairs and comprise defined segments, the method comprising: [0021]
  • (a) providing regions of homology in the at least a first and second DNA substrate molecules by inserting an intron sequence between at least two defined segments; [0022]
  • (b) fragmenting and recombining DNA substrate molecules of (a), wherein regions of homology are provided by the introns; [0023]
  • (c) screening or selecting the products of (b) for a desired property; and [0024]
  • (d) recovering a recombinant DNA substrate molecule from the products of (c) encoding an evolved protein. [0025]
  • A further aspect of the invention is a method for evolving a protein encoded by a DNA substrate molecule by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide and comprise defined segments, the method comprising: [0026]
  • (a) providing a set of oligonucleotide PCR primers, wherein for each strand of each segment a pair of primers is provided, one member of each pair bridging the junction at one end of the segment and the other bridging the junction at the other end of the segment, with the terminal ends of the DNA molecule having as one member of the pair a generic primer, and wherein a set of primers is provided for each of the at least a first and second substrate molecules; [0027]
  • (b) amplifying the segments of the at least a first and second DNA substrate molecules with the primers of (a) in a polymerase chain reaction; [0028]
  • (c) assembling the products of (b) to generate a pool of recombinant DNA molecules; [0029]
  • (d) selecting or screening the products of (c) for a desired property; and [0030]
  • (e) recovering a recombinant DNA substrate molecule from the products of (d) encoding an evolved protein. [0031]
  • A further aspect of the invention is a method for optimizing expression of a protein by evolving the protein, wherein the protein is encoded by a DNA substrate molecule, comprising: [0032]
  • (a) providing a set of oligonucleotides, wherein each oligonucleotide comprises at least two regions complementary to the DNA molecule and at least one degenerate region, each degenerate region encoding a region of an amino acid sequence of the protein; [0033]
  • (b) assembling the set of oligonucleotides into a library of full length genes; [0034]
  • (c) expressing the products of (b) in a host cell; [0035]
  • (d) screening the products of (c) for improved expression of the protein; and [0036]
  • (e) recovering a recombinant DNA substrate molecule encoding an evolved protein from (d). [0037]
  • A further aspect of the invention is a method for optimizing expression of a protein encoded by a DNA substrate molecule by evolving the protein, wherein the DNA substrate molecule comprises at least one lac operator and a fusion of a DNA sequence encoding the protein with a DNA sequence encoding a lac headpiece dimer, the method comprising: [0038]
  • (a) transforming a host cell with a library of mutagenized DNA substrate molecules; [0039]
  • (b) inducing expression of the protein encoded by the library of (a); [0040]
  • (c) preparing an extract of the product of (b); [0041]
  • (d) fractionating insoluble protein from complexes of soluble protein and DNA; and [0042]
  • (e) recovering a DNA substrate molecule encoding an evolved protein from (d). [0043]
  • A further aspect of the invention is a method for evolving functional expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA sequence encoding filamentous phage protein to generate a fusion protein, the method comprising: [0044]
  • (a) providing a host cell producing infectious particles expressing a fusion protein encoded by a library of mutagenized DNA substrate molecules; [0045]
  • (b) recovering from (a) infectious particles displaying the fusion protein; [0046]
  • (c) affinity purifying particles displaying the mutant protein using a ligand for the protein; and [0047]
  • (d) recovering a DNA substrate molecule encoding an evolved protein from affinity purified particles of (c). [0048]
  • A further aspect of the invention is a method for optimizing expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a lac headpiece dimer, wherein the DNA substrate molecule is present on a first plasmid vector, the method comprising: [0049]
  • (a) providing a host cell transformed with the first vector and a second vector comprising a library of mutants of at least one chaperonin gene, and at least one lac operator; [0050]
  • (b) preparing an extract of the product of (a); [0051]
  • (c) fractionating insoluble protein from complexes of soluble protein and DNA; and [0052]
  • (d) recovering DNA encoding a chaperonin gene from (c). [0053]
  • A further aspect of the invention is a method for optimizing expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a filamentous phage gene, wherein the fusion is carried on a phagemid comprising a library of chaperonin gene mutants, the method comprising: [0054]
  • (a) providing a host cell producing infectious particles expressing a fusion protein encoded by a library of mutagenized DNA substrate molecules; [0055]
  • (b) recovering from (a) infectious particles displaying the fusion protein; [0056]
  • (c) affinity purifying particles displaying the protein using a ligand for the protein; and [0057]
  • (d) recovering DNA encoding the mutant chaperonin from affinity purified particles of (c). [0058]
  • A further aspect of the invention is a method for optimizing secretion of a protein in a host by evolving a gene encoding a secretory function, comprising: [0059]
  • (a) providing a cluster of genes encoding secretory functions; [0060]
  • (b) recombining at least a first and second sequence in the gene cluster of (a) encoding a secretory function, the at least a first and second sequences differing from each other in at least one nucleotide, to generate a library of recombinant sequences; [0061]
  • (c) transforming a host cell culture with the products of (b), wherein the host cell comprises a DNA sequence encoding the protein; [0062]
  • (d) subjecting the product of (c) to screening or selection for secretion of the protein; and [0063]
  • (e) recovering DNA encoding an evolved gene encoding a secretory function from the product of (d). [0064]
  • A further aspect of the invention is a method for evolving an improved DNA polymerase comprising: [0065]
  • (a) providing a library of mutant DNA substrate molecules encoding mutant DNA polymerase; [0066]
  • (b) screening extracts of cells transfected with (a) and comparing activity with wild type DNA polymerase; [0067]
  • (c) recovering mutant DNA substrate molecules from cells in (b) expressing mutant DNA polymerase having improved activity over wild-type DNA polymerase; and [0068]
  • (d) recovering a DNA substrate molecule encoding an evolved polymerase from the products of (c). [0069]
  • A further aspect of the invention is a method for evolving a DNA polymerase with an error rate greater than that of wild type DNA polymerase comprising: [0070]
  • (a) providing a library of mutant DNA substrate molecules encoding mutant DNA polymerase in a host cell comprising an indicator gene having a revertible mutation, wherein the indicator gene is replicated by the mutant DNA polymerase; [0071]
  • (b) screening the products of (a) for revertants of the indicator gene; [0072]
  • (c) recovering mutant DNA substrate molecules from revertants; and [0073]
  • (d) recovering a DNA substrate molecule encoding an evolved polymerase from the products of (c). [0074]
  • A further aspect of the invention is a method for evolving a DNA polymerase, comprising: [0075]
  • (a) providing a library of mutant DNA substrate molecules encoding mutant DNA polymerase, the library comprising a plasmid vector; [0076]
  • (b) preparing plasmid preparations and extracts of host cells transfected with the products of (a); [0077]
  • (c) amplifying each plasmid preparation in a PCR reaction using the mutant polymerase encoded by that plasmid, the polymerase being present in the host cell extract; [0078]
  • (d) recovering the PCR products of (c); and [0079]
  • (e) recovering a DNA substrate molecule encoding an evolved polymerase from the products of (d). [0080]
  • A further aspect of the invention is a method for evolving a p-nitrophenol phosphonatase from a phosphonatase encoded by a DNA substrate molecule, comprising: [0081]
  • (a) providing library of mutants of the DNA substrate molecule, the library comprising a plasmid expression vector; [0082]
  • (b) transfecting a host, wherein the host phn operon is deleted; [0083]
  • (c) selecting for growth of the transfectants of (b) using a p-nitrophenol phosphonatase as a substrate; [0084]
  • (d) recovering the DNA substrate molecules from transfectants selected from (c); and [0085]
  • (e) recovering a DNA substrate molecule from (d) encoding an evolved phosphonatase. [0086]
  • A further aspect of the invention is a method for evolving a protease encoded by a DNA substrate molecule comprising: [0087]
  • (a) providing library of mutants of the DNA substrate molecule, the library comprising a plasmid expression vector, wherein the DNA substrate molecule is linked to a secretory leader; [0088]
  • (b) transfecting a host; [0089]
  • (c) selecting for growth of the transfectants of (b) on a complex protein medium; and [0090]
  • (d) recovering a DNA substrate molecule from (c) encoding an evolved protease. [0091]
  • A further aspect of the invention is a method for screening a library of protease mutants displayed on a phage to obtain an improved protease, wherein a DNA substrate molecule encoding the protease is fused to DNA encoding a filamentous phage protein to generate a fusion protein, comprising: [0092]
  • (a) providing host cells expressing the fusion protein; [0093]
  • (b) overlaying host cells with a protein net to entrap the phage; [0094]
  • (c) washing the product of (b) to recover phage liberated by digestion of the protein net; [0095]
  • (d) recovering DNA from the product of (c); and [0096]
  • (e) recovering a DNA substrate from (d) encoding an improved protease. [0097]
  • A further aspect of the invention is a method for screening a library of protease mutants to obtain an improved protease, the method comprising: [0098]
  • (a) providing a library of peptide substrates, the peptide substrate comprising a fluorophore and a fluorescence quencher; [0099]
  • (b) screening the library of protease mutants for ability to cleave the peptide substrates, wherein fluorescence is measured; and [0100]
  • (c) recovering DNA encoding at least one protease mutant from (b). [0101]
  • A further aspect of the invention is a method for evolving an alpha interferon gene comprising: [0102]
  • (a) providing a library of mutant alpha interferon genes, the library comprising a filamentous phage vector; [0103]
  • (b) stimulating cells comprising a reporter construct, the reporter construct comprising a reporter gene under control of an interferon responsive promoter, and wherein the reporter gene is GFP; [0104]
  • (c) separating the cells expressing GFP by FACS; [0105]
  • (d) recovering phage from the product of (c); and [0106]
  • (e) recovering an evolved interferon gene from the product of (d). [0107]
  • A further aspect of the invention is a method for screening a library of mutants of a DNA substrate encoding a protein for an evolved DNA substrate, comprising: [0108]
  • (a) providing a library of mutants, the library comprising an expression vector; [0109]
  • (b) transfecting a mammalian host cell with the library of (a) wherein mutant protein is expressed on the surface of the cell; [0110]
  • (c) screening or selecting the products of (b) with a ligand for the protein; [0111]
  • (d) recovering DNA encoding mutant protein from the products of (c); and [0112]
  • (e) recovering an evolved DNA substrate from the products of (d). [0113]
  • A further aspect of the invention is a method for evolving a DNA substrate molecule encoding an interferon alpha, comprising: [0114]
  • (a) providing a library of mutant alpha interferon genes, the library comprising an expression vector wherein the alpha interferon genes are expressed under the control of an inducible promoter; [0115]
  • (b) transfecting host cells with the library of (a); [0116]
  • (c) contacting the product of (b) with a virus; [0117]
  • (d) recovering DNA encoding a mutant alpha interferon from host cells surviving step (c); and [0118]
  • (e) recovering an evolved interferon gene from the product of (d). [0119]
  • A further aspect of the invention is a method for evolving the stability of a protein encoded by a DNA substrate molecule, the DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA sequence encoding a filamentous phage protein to generate a fusion protein, the method comprising: [0120]
  • (a) providing a host cell expressing a library of mutants of the fusion protein; [0121]
  • (b) affinity purifying the mutants with a ligand for the protein, wherein the ligand is a human serum protein, tissue specific protein, or receptor; [0122]
  • (c) recovering DNA encoding a mutant protein from the affinity selected mutants of (b); and [0123]
  • (d) recovering an evolved gene encoding the protein from the product of (c). [0124]
  • A further aspect of the invention is a method for evolving a protein having at least two subunits, comprising: [0125]
  • (a) providing a library of mutant DNA substrate molecules for each subunit; [0126]
  • (b) recombining the libraries into a library of single chain constructs of the protein, the single chain construct comprising a DNA substrate molecule encoding each subunit sequence, the subunit sequence being linked by a linker at a nucleic acid sequence encoding the amino terminus of one subunit to a nucleic acid sequence encoding the carboxy terminus of a second subunit; [0127]
  • (c) screening or selecting the products of (B), [0128]
  • (d) recovering recombinant single chain construct DNA substrate molecules from the products of (c); [0129]
  • (e) subjecting the products of (d) to mutagenesis; and [0130]
  • (f) recovering an evolved single chain construct DNA substrate molecule from (e). [0131]
  • A further aspect of the invention is a method for evolving the coupling of a mammalian 7-transmembrane receptor to a yeast signal transduction pathway, comprising: [0132]
  • (a) expressing a library of mammalian G alpha protein mutants in a host cell, wherein the host cell expresses the mammalian 7-transmembrane receptor and a reporter gene, the receptor gene geing expressed under control of a pheromone responsive promoter; [0133]
  • (b) screening or selecting the products of (a) for expression of the reporter gene in the presence of a ligand for the 7-transmembrance receptor; and [0134]
  • (c) recovering DNA encoding an evolved G alpha protein mutant from screened or selected products of (b). [0135]
  • A further aspect of the invention is a method for recombining at least a first and second DNA substrate molecule, comprising: [0136]
  • (a) transfecting a host cell with at least a first and second DNA substrate molecule wherein the at least a first and second DNA substrate molecules are recombined in the host cell; [0137]
  • (b) screening or selecting the products of (a) for a desired property; and [0138]
  • (c) recovering recombinant DNA substrate molecules from (b). [0139]
  • A further aspect of the invention is a method for evolving a DNA substrate sequence encoding a protein of interest, wherein the DNA substrate comprises a vector, the vector comprising single-stranded DNA, the method comprising: [0140]
  • (a) providing single-stranded vector DNA and a library of mutants of the DNA substrate sequence; [0141]
  • (b) annealing single stranded DNA from the library of (a) to the single stranded vector DNA of (a); [0142]
  • (c) transforming the products of (b) into a host; [0143]
  • (d) screening the product of (c) for a desired property; and [0144]
  • (e) recovering evolved DNA substrate DNA from the products of (d).[0145]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts the alignment of oligo PCR primers for evolution of bovine calf intestinal alkaline phosphatase. [0146]
  • FIG. 2 depicts the alignment of alpha interferon amino acid and nucleic acid sequences.[0147]
  • DESCRIPTION OF THE SPECIFIC EMBODIMENTS
  • The invention provides a number of strategies for evolving polypeptides through recursive recombination methods. In some embodiments, the strategies of the invention can generally be classified as “coarse grain shuffling” and “fine grain shuffling.” As described in detail below, these strategies are especially applicable in situations where some structural or functional information is available regarding the polypeptides of interest, where the nucleic acid to be manipulated is large, when selection or screening of many recombinants is cumbersome, and so on. “Coarse grain shuffling” generally involves the exchange or recombination of segments of nucleic acids, whether defined as functional domains, exons, restriction endonuclease fragments, or otherwise arbitrarily defined segments. “Fine grain shuffling” generally involves the introduction of sequence variation within a segment, such as within codons. [0148]
  • Coarse grain and fine grain shuffling allow analysis of variation occuring within a nucleic acid sequence, also termed “searching of sequence space.” Although both techniques are meritorious, the results are qualitatively different. For example, coarse grain searches are often better suited for optimizing multigene clusters such as polyketide operons, whereas fine grain searches are often optimal for optimizing a property such as protein expression using codon usage libraries. [0149]
  • The strategies generally entail evolution of gene(s) or segment(s) thereof to allow retention of function in a heterologous cell or improvement of function in a homologous or heterologous cell. Evolution is effected generally by a process termed recursive sequence recombination. Recursive sequence recombination can be achieved in many different formats and permutations of formats, as described in further detail below. These formats share some common principles. Recursive sequence recombination entails successive cycles of recombination to generate molecular diversity, i.e., the creation of a family of nucleic acid molecules showing substantial sequence identity to each other but differing in the presence of mutations. Each recombination cycle is followed by at least one cycle of screening or selection for molecules having a desired characteristic. The molecule(s) selected in one round form the starting materials for generating diversity in the next round. In any given cycle, recombination can occur in vivo or in vitro. Furthermore, diversity resulting from recombination can be augmented in any cycle by applying prior methods of mutagenesis (e.g., error-prone PCR or cassette mutagenesis, passage through bacterial mutator strains, treatment with chemical mutagens) to either the substrates for or products of recombination. [0150]
  • I. Formats for Recursive Sequence Recombination [0151]
  • Some formats and examples for recursive sequence recombination, sometimes referred to as DNA shuffling, evolution, or molecular breeding, have been described by the present inventors and co-workers in co-pending applications U.S. patent application Ser. No. 08/198,431, filed Feb. 17, 1994, Ser. No. PCT/US95/02126, filed, Feb. 17, 1995, Ser. No. 08/425,684, filed Apr. 18, 1995, Ser. No. 08/537,874, filed Oct. 30, 1995, Ser. No. 08/564,955, filed Nov. 30, 1995, Ser. No. 08/621,859, filed Mar. 25, 1996, Ser. No. 08/621,430, filed Mar. 25, 1996, Ser. No. PCT/US96/05480, filed Apr. 18, 1996, Ser. No. 08/650,400, filed May 20, 1996, Ser. No. 08/675,502, filed Jul. 3, 1996, Ser. No. 08/721,824, filed Sep. 27, 1996, and Ser. No. 08/722,660 filed Sep. 27, 1996; Stemmer, [0152] Science 270:1510 (1995); Stemmer et al., Gene 164:49-53 (1995); Stemmer, Bio/Technology 13:549-553 (1995); Stemmer, Proc. Natl. Acad. Sci. U.S.A. 91:10747-10751 (1994); Stemmer, Nature 370:389-391 (1994); Crameri et al., Nature Medicine 2(1):1-3 (1996); Crameri et al., Nature Biotechnology 14:315-319 (1996), each of which is incorporated by reference in its entirety for all purposes.
  • In general, the term “gene” is used herein broadly to refer to any segment or sequence of DNA associated with a biological function. Genes can be obtained from a variety of sources, including cloning from a source of interest or synthesizing from known or predicted sequence information, and may include sequences designed to have desired parameters. [0153]
  • A wide variety of cell types can be used as a recipient of evolved genes. Cells of particular interest include many bacterial cell types, both gram-negative and gram-positive, such as [0154] Rhodococcus, Streptomycetes, Actinomycetes, Corynebacteria, Penicillium, Bacillus, Escherichia coli, Pseudomonas, Salmonella, and Erwinia. Cells of interest also include eukaryotic cells, particularly mammalian cells (e.g., mouse, hamster, primate, human), both cell lines and primary cultures. Such cells include stem cells, including embryonic stem cells, zygotes, fibroblasts, lymphocytes, Chinese hamster ovary (CHO), mouse fibroblasts (NIH3T3), kidney, liver, muscle, and skin cells. Other eukaryotic cells of interest include plant cells, such as maize, rice, wheat, cotton, soybean, sugarcane, tobacco, and arabidopsis; fish, algae, fungi (Penicillium, Fusarium, Aspergillus, Podospora, Neurospora), insects, yeasts (Picchia and Saccharomyces).
  • The choice of host will depend on a number of factors, depending on the intended use of the engineered host, including pathogenicity, substrate range, environmental hardiness, presence of key intermediates, ease of genetic manipulation, and likelihood of promiscuous transfer of genetic information to other organisms. A preferred host has the ability to replicate vector DNA, express proteins of interest, and properly traffic proteins of interest. Particularly advantageous hosts are [0155] E. coli, lactobacilli, Streptomycetes, Actinomycetes, fungi such as Saccaromyces cerivisiae or Pischia pastoris, Schneider cells, L-cells, COS cells, CHO cells, and transformed B cell lines such as SP2/0, J558, NS-1 and AG8-653.
  • The breeding procedure starts with at least two substrates that generally show substantial sequence identity to each other (i.e., at least about 50%, 70%, 80% or 90% sequence identity), but differ from each other at certain positions. The difference can be any type of mutation, for example, substitutions, insertions and deletions. Often, different segments differ from each other in perhaps 5-20 positions. For recombination to generate increased diversity relative to the starting materials, the starting materials must differ from each other in at least two nucleotide positions. That is, if there are only two substrates, there should be at least two divergent positions. If there are three substrates, for example, one substrate can differ from the second as a single position, and the second can differ from the third at a different single position. The starting DNA segments can be natural variants of each other, for example, allelic or species variants. The segments can also be from nonallelic genes showing some degree of structural and usually functional relatedness (e.g., different genes within a superfamily such as the immunoglobulin superfamily). The starting DNA segments can also be induced variants of each other. For example, one DNA segment can be produced by error-prone PCR replication of the other, or by substitution of a mutagenic cassette. Induced mutants can also be prepared by propagating one (or both) of the segments in a mutagenic strain. In these situations, strictly speaking, the second DNA segment is not a single segment but a large family of related segments. The different segments forming the starting materials are often the same length or substantially the same length. However, this need not be the case; for example; one segment can be a subsequence of another. The segments can be present as part of larger molecules, such as vectors, or can be in isolated form. [0156]
  • The starting DNA segments are recombined by any of the recursive sequence recombination formats provded herein to generate a diverse library of recombinant DNA segments. Such a library can vary widely in size from having fewer than 10 to more than 10[0157] 5, 109, or 1012 members. In general, the starting segments and the recombinant libraries generated include full-length coding sequences and any essential regulatory sequences, such as a promoter and polyadenylation sequence, required for expression. However, if this is not the case, the recombinant DNA segments in the library can be inserted into a common vector providing the missing sequences before performing screening/selection.
  • If the recursive sequence recombination format employed is an in vivo format, the library of recombinant DNA segments generated already exists in a cell, which is usually the cell type in which expression of the enzyme with altered substrate specificity is desired. If recursive sequence recombination is performed in vitro, the recombinant library is preferably introduced into the desired cell type before screening/selection. The members of the recombinant library can be linked to an episome or virus before introduction or can be introduced directly. In some embodiments of the invention, the library is amplified in a first host, and is then recovered from that host and introduced to a second host more amenable to expression, selection, or screening, or any other desirable parameter. The manner in which the library is introduced into the cell type depends on the DNA-uptake characteristics of the cell type, e.g., having viral receptors, being capable of conjugation, or being naturally competent. If the cell type is insusceptible to natural and chemical-induced competence, but susceptible to electroporation, one would usually employ electroporation. If the cell type is insusceptible to electroporation as well, one can employ biolistics. The biolistic PDS-1000 Gene Gun (Biorad, Hercules, Calif.) uses helium pressure to accelerate DNA-coated gold or tungsten microcarriers toward target cells. The process is applicable to a wide range of tissues, including plants, bacteria, fungi, algae, intact animal tissues, tissue culture cells, and animal embryos. One can employ electronic pulse delivery, which is essentially a mild electroporation format for live tissues in animals and patients. Zhao, [0158] Advanced Drug Delivery Reviews 17:257-262 (1995). Novel methods for making cells competent are described in co-pending application U.S. patent application Ser. No. 08/621,430, filed Mar. 25, 1996. After introduction of the library of recombinant DNA genes, the cells are optionally propagated to allow expression of genes to occur.
  • A. In Vitro Formats [0159]
  • One format for recursive sequence recombination utilizes a pool of related sequences. The sequences can be DNA or RNA and can be of various lengths depending on the size of the gene or DNA fragment to be recombined or reassembled. Preferably the sequences are from 50 bp to 100 kb. [0160]
  • The pool of related substrates can be fragmented, usually at random, into fragments of from about 5 bp to 5 kb or more. Preferably the size of the random fragments is from about 10 bp to 1000 bp, more preferably the size of the DNA fragments is from about 20 bp to 500 bp. The substrates can be digested by a number of different methods, such as DNAseI or RNAse digestion, random shearing or restriction enzyme digestion. The concentration of nucleic acid fragments of a particular length is often less than 0.1% or 1% by weight of the total nucleic acid. The number of different specific nucleic acid fragments in the mixture is usually at least about 100, 500 or 1000. [0161]
  • The mixed population of nucleic acid fragments are denatured by heating to about 80° C. to 100° C., more preferably from 90° C. to 96° C., to form single-stranded nucleic acid fragments. Single-stranded nucleic acid fragments having regions of sequence identity with other single-stranded nucleic acid fragments can then be reannealed by cooling to 20° C. to 75° C., and preferably from 40° C. to 65° C. Renaturation can be accelerated by the addition of polyethylene glycol (“PEG”) or salt. The salt concentration is preferably from 0 mM to 600 mM, more preferably the salt concentration is from 10 mM to 100 mM. The salt may be such salts as (NH[0162] 4)2SO4, KCl, or NaCl. The concentration of PEG is preferably from 0% to 20%, more preferably from 5% to 10%. The fragments that reanneal can be from different substrates.
  • The annealed nucleic acid fragments are incubated in the presence of a nucleic acid polymerase, such as Taq or Klenow, Mg[0163] ++ at 1 mM-20 mM, and dNTP's (i.e. dATP, dCTP, dGTP and dTTP). If regions of sequence identity are large, Taq or other high-temperature polymerase can be used with an annealing temperature of between 45-65° C. If the areas of identity are small, Klenow or other low-temperature polymerases can be used with an annealing temperature of between 20-30° C. The polymerase can be added to the random nucleic acid fragments prior to annealing, simultaneously with annealing or after annealing.
  • The cycle of denaturation, renaturation and incubation of random nucleic acid fragments in the presence of polymerase is sometimes referred to as “shuffling” of the nucleic acid in vitro. This cycle is repeated for a desired number of times. Preferably the cycle is repeated from 2 to 100 times, more preferably the sequence is repeated from 10 to 40 times. The resulting nucleic acids are a family of double-stranded polynucleotides of from about 50 bp to about 100 kb, preferably from 500 bp to 50 kb. The population represents variants of the starting substrates showing substantial sequence identity thereto but also diverging at several positions. The population has many more members than the starting substrates. The population of fragments resulting from recombination is preferably first amplified by PCR, then cloned into an appropriate vector and the ligation mixture used to transform host cells. [0164]
  • In a variation of in vitro shuffling, subsequences of recombination substrates can be generated by amplifying the full-length sequences under conditions which produce a substantial fraction, typically at least 20 percent or more, of incompletely extended amplification products. The amplification products, including the incompletely extended amplification products are denatured and subjected to at least one additional cycle of reannealing and amplification. This variation, wherein at least one cycle of reannealing and amplification provides a substantial fraction of incompletely extended products, is termed “stuttering.” In the subsequent amplification round, the incompletely extended products anneal to and prime extension on different sequence-related template species. [0165]
  • In a further variation, at least one cycle of amplification can be conducted using a collection of overlapping single-stranded DNA fragments of related sequence, and different lengths. Each fragment can hybridize to and prime polynucleotide chain extension of a second fragment from the collection, thus forming sequence-recombined polynucleotides. In a further variation, single-stranded DNA fragments of variable length can be generated from a single primer by Vent DNA polymerase on a first DNA template. The single stranded DNA fragments are used as primers for a second, Kunkel-type template, consisting of a uracil-containing circular single-stranded DNA. This results in multiple substitutions of the first template into the second (see Levichkin et al., [0166] Mol. Biology 29:572-577 (1995)).
  • Nucleic acid sequences can be recombined by recursive sequence recombination even if they lack sequence homology. Homology can be introduced using synthetic oligonucleotides as PCR primers. In addition to the specific sequences for the nucleic acid segment being amplified, all of the primers used to amplify one particular segment are synthesized to contain an additional sequence of 20-40 [0167] bases 5′ to the gene (sequence A) and a different 20-40 base sequence 3′ to the segment (sequence B). An adjacent segment is amplified using a 5′ primer which contains the complementary strand of sequence B (sequence B′), and a 3′ primer containing a different 20-40 base sequence (C). Similarly, primers for the next adjacent segment contain sequences C′ (complementary to C) and D. In this way, small regions of homology are introduced, making the segments into site-specific recombination cassettes. Subsequent to the initial amplification of individual segments, the amplified segments can then be mixed and subjected to primeness PCR.
  • When domains within a polypeptide are shuffled, it may not be possible to introduce additional flanking sequences to the domains, due to the constraint of maintaining a continuous open reading frame. Instead, groups of oligonucleotides are synthesized that are homologous to the 3′ end of the first domain encoded by one of the genes to be shuffled, and the 5′ ends of the second domains encoded by all of the other genes to be shuffled together. This is repeated with all domains, thus providing sequences that allow recombination between protein domains while maintaining their order. [0168]
  • B. In Vivo Formats [0169]
  • 1. Plasmid-Plasmid Recombination [0170]
  • The initial substrates for recombination are a collection of polynucleotides comprising variant forms of a gene. The variant forms usually show substantial sequence identity to each other sufficient to allow homologous recombination between substrates. The diversity between the polynucleotides can be natural (e.g., allelic or species variants), induced (e.g., error-prone PCR or error-prone recursive sequence recombination), or the result of in vitro recombination. Diversity can also result from resynthesizing genes encoding natural proteins with alternative codon usage. There should be at least sufficient diversity between substrates that recombination can generate more diverse products than there are starting materials. There must be at least two substrates differing in at least two positions. However, commonly a library of substrates of 10[0171] 3-108 members is employed. The degree of diversity depends on the length of the substrate being recombined and the extent of the functional change to be evolved. Diversity at between 0.1-25% of positions is typical. The diverse substrates are incorporated into plasmids. The plasmids are often standard cloning vectors, e.g., bacterial multicopy plasmids. However, in some methods to be described below, the plasmids include mobilization (MOB) functions. The substrates can be incorporated into the same or different plasmids. Often at least two different types of plasmid having different types of selectable markers are used to allow selection for cells containing at least two types of vector. Also, where different types of plasmid are employed, the different plasmids can come from two distinct incompatibility groups to allow stable co-existence of two different plasmids within the cell. Nevertheless, plasmids from the same incompatibility group can still co-exist within the same cell for sufficient time to allow homologous recombination to occur.
  • Plasmids containing diverse substrates are initially introduced into cells by any method (e.g., chemical transformation, natural competence, electroporation, biolistics, packaging into phage or viral systems). Often, the plasmids are present at or near saturating concentration (with respect to maximum transfection capacity) to increase the probability of more than one plasmid entering the same cell. The plasmids containing the various substrates can be transfected simultaneously or in multiple rounds. For example, in the latter approach cells can be transfected with a first aliquot of plasmid, transfectants selected and propagated, and then infected with a second aliquot of plasmid. [0172]
  • Having introduced the plasmids into cells, recombination between substrates to generate recombinant genes occurs within cells containing multiple different plasmids merely by propagating the cells. However, cells that receive only one plasmid are unable to participate in recombination and the potential contribution of substrates on such plasmids to evolution is not fully exploited (although these plasmids may contribute to some extent if they are progagated in mutator cells). The rate of evolution can be increased by allowing all substrates to participate in recombination. Such can be achieved by subjecting transfected cells to electroporation. The conditions for electroporation are the same as those conventionally used for introducing exogenous DNA into cells (e.g., 1,000-2,500 volts, 400 μF and a 1-2 mM gap). Under these conditions, plasmids are exchanged between cells allowing all substrates to participate in recombination. In addition the products of recombination can undergo further rounds of recombination with each other or with the original substrate. The rate of evolution can also be increased by use of conjugative transfer. To exploit conjugative transfer, substrates can be cloned into plasmids having MOB genes, and tra genes are also provided in cis or in trans to the MOB genes. The effect of conjugative transfer is very similar to electroporation in that it allows plasmids to move between cells and allows recombination between any substrate and the products of previous recombination to occur, merely by propagating the culture. The rate of evolution can also be increased by fusing cells to induce exchange of plasmids or chromosomes. Fusion can be induced by chemical agents, such as PEG, or viral proteins, such as influenza virus hemagglutinin, HSV-1 gB and gD. The rate of evolution can also be increased by use of mutator host cells (e.g., Mut L, S, D, T, H in bacteria and Ataxia telangiectasia human cell lines). [0173]
  • The time for which cells are propagated and recombination is allowed to occur, of course, varies with the cell type but is generally not critical, because even a small degree of recombination can substantially increase diversity relative to the starting materials. Cells bearing plasmids containing recombined genes are subject to screening or selection for a desired function. For example, if the substrate being evolved contains a drug resistance gene, one would select for drug resistance. Cells surviving screening or selection can be subjected to one or more rounds of screening/selection followed by recombination or can be subjected directly to an additional round of recombination. “Screening” as used herein is intended to include “selection” as a type of screen. [0174]
  • The next round of recombination can be achieved by several different formats independently of the previous round. For example, a further round of recombination can be effected simply by resuming the electroporation or conjugation-mediated intercellular transfer of plasmids described above. Alternatively, a fresh substrate or substrates, the same or different from previous substrates, can be transfected into cells surviving selection/screening. Optionally, the new substrates are included in plasmid vectors bearing a different selective marker and/or from a different incompatibility group than the original plasmids. As a further alternative, cells surviving selection/screening can be subdivided into two subpopulations, and plasmid DNA from one subpopulation transfected into the other, where the substrates from the plasmids from the two subpopulations undergo a further round of recombination. In either of the latter two options, the rate of evolution can be increased by employing DNA extraction, electroporation, conjugation or mutator cells, as described above. In a still further variation, DNA from cells surviving screening/selection can be extracted and subjected to in vitro recursive sequence recombination. [0175]
  • After the second round of recombination, a second round of screening/selection is performed, preferably under conditions of increased stringency. If desired, further rounds of recombination and selection/screening can be performed using the same strategy as for the second round. With successive rounds of recombination and selection/screening, the surviving recombined substrates evolve toward acquisition of a desired phenotype. Typically, in this and other methods of recursive recombination, the final product of recombination that has acquired the desired phenotype differs from starting substrates at 0.1%-25% of positions and has evolved at a rate orders of magnitude in excess (e.g., by at least 10-fold, 100-fold, 1000-fold, or 10,000 fold) of the rate of evolution driven by naturally acquired mutation of about 1 mutation per 10[0176] −9 positions per generation (see Anderson et al., Proc. Natl. Acad. Sci. U.S.A. 93:906-907 (1996)). The “final product” may be transferred to another host more desirable for utilization of the “shuffled” DNA. This is particularly advantageous in situations where the more desirable host is less efficient as a host for the many cycles of mutation/ recombination due to the lack of molecular biology or genetic tools available for other organisms such as E. coli.
  • 2. Virus-Plasmid Recombination [0177]
  • The strategy used for plasmid-plasmid recombination can also be used for virus-plasmid recombination; usually, phage-plasmid recombination. However, some additional comments particular to the use of viruses are appropriate. The initial substrates for recombination are cloned into both plasmid and viral vectors. It is usually not critical which substrate(s) is/are inserted into the viral vector and which into the plasmid, although usually the viral vector should contain different substrate(s) from the plasmid. As before, the plasmid (and the virus) typically contains a selective marker. The plasmid and viral vectors can both be introduced into cells by transfection as described above. However, a more efficient procedure is to transfect the cells with plasmid, select transfectants and infect the transfectants with virus. Because the efficiency of infection of many viruses approaches 100% of cells, most cells transfected and infected by this route contain both a plasmid and virus bearing different substrates. [0178]
  • Homologous recombination occurs between plasmid and virus generating both recombined plasmids and recombined virus. For some viruses, such as filamentous phage, in which intracellular DNA exists in both double-stranded and single-stranded forms, both can participate in recombination. Provided that the virus is not one that rapidly kills cells, recombination can be augmented by use of electroporation or conjugation to transfer plasmids between cells. Recombination can also be augmented for some types of virus by allowing the progeny virus from one cell to reinfect other cells. For some types of virus, virus infected-cells show resistance to superinfection. However, such resistance can be overcome by infecting at high multiplicity and/or using mutant strains of the virus in which resistance to superinfection is reduced. [0179]
  • The result of infecting plasmid-containing cells with virus depends on the nature of the virus. Some viruses, such as filamentous phage, stably exist with a plasmid in the cell and also extrude progeny phage from the cell. Other viruses, such as lambda having a cosmid genome, stably exist in a cell like plasmids without producing progeny virions. Other viruses, such as the T-phage and lytic lambda, undergo recombination with the plasmid but ultimately kill the host cell and destroy plasmid DNA. For viruses that infect cells without killing the host, cells containing recombinant plasmids and virus can be screened/selected using the same approach as for plasmid-plasmid recombination. Progeny virus extruded by cells surviving selection/screening can also be collected and used as substrates in subsequent rounds of recombination. For viruses that kill their host cells, recombinant genes resulting from recombination reside only in the progeny virus. If the screening or selective assay requires expression of recombinant genes in a cell, the recombinant genes should be transferred from the progeny virus to another vector, e.g., a plasmid vector, and retransfected into cells before selection/screening is performed. [0180]
  • For filamentous phage, the products of recombination are present in both cells surviving recombination and in phage extruded from these cells. The dual source of recombinant products provides some additional options relative to the plasmid-plasmid recombination. For example, DNA can be isolated from phage particles for use in a round of in vitro recombination. Alternatively, the progeny phage can be used to transfect or infect cells surviving a previous round of screening/selection, or fresh cells transfected with fresh substrates for recombination. [0181]
  • 3. Virus-Virus Recombination [0182]
  • The principles described for plasmid-plasmid and plasmid-viral recombination can be applied to virus-virus recombination with a few modifications. The initial substrates for recombination are cloned into a viral vector. Usually, the same vector is used for all substrates. Preferably, the virus is one that, naturally or as a result of mutation, does not kill cells. After insertion, some viral genomes can be packaged in vitro or using a packaging cell line. The packaged viruses are used to infect cells at high multiplicity such that there is a high probability that a cell will receive multiple viruses bearing different substrates. [0183]
  • After the initial round of infection, subsequent steps depend on the nature of infection as discussed in the previous section. For example, if the viruses have phagemid (Sambrook et al., [0184] Molecular Cloning, CSH Press, 1987) genomes such as lambda cosmids or M13, F1 or Fd phagemids, the phagemids behave as plasmids within the cell and undergo recombination simply by propagating the cells. Recombination is particularly efficient between single-stranded forms of intracellular DNA. Recombination can be augmented by electroporation of cells.
  • Following selection/screening, cosmids containing recombinant genes can be recovered from surviving cells, e.g., by heat induction of a cos[0185] lysogenic host cell, or extraction of DNA by standard procedures, followed by repackaging cosmid DNA in vitro.
  • If the viruses are filamentous phage, recombination of replicating form DNA occurs by propagating the culture of infected cells. Selection/screening identifies colonies of cells containing viral vectors having recombinant genes with improved properties, together with infectious particles (i.e., phage or packaged phagemids) extruded from such cells. Subsequent options are essentially the same as for plasmid-viral recombination. [0186]
  • 4. Chromosome Recombination [0187]
  • This format can be used to especially evolve chromosomal substrates. The format is particularly preferred in situations in which many chromosomal genes contribute to a phenotype or one does not know the exact location of the chromosomal gene(s) to be evolved. The initial substrates for recombination are cloned into a plasmid vector. If the chromosomal gene(s) to be evolved are known, the substrates constitute a family of sequences showing a high degree of sequence identity but some divergence from the chromosomal gene. If the chromosomal genes to be evolved have not been located, the initial substrates usually constitute a library of DNA segments of which only a small number show sequence identity to the gene or gene(s) to be evolved. Divergence between plasmid-borne substrate and the chromosomal gene(s) can be induced by mutagenesis or by obtaining the plasmid-borne substrates from a different species than that of the cells bearing the chromosome. [0188]
  • The plasmids bearing substrates for recombination are transfected into cells having chromosomal gene(s) to be evolved. Evolution can occur simply by propagating the culture, and can be accelerated by transferring plasmids between cells by conjugation or electroporation. Evolution can be further accelerated by use of mutator host cells or by seeding a culture of nonmutator host cells being evolved with mutator host cells and inducing intercellular transfer of plasmids by electroporation or conjugation. Preferably, mutator host cells used for seeding contain a negative selectable marker to facilitate isolation of a pure culture of the nonmutator cells being evolved. Selection/screening identifies cells bearing chromosomes and/or plasmids that have evolved toward acquisition of a desired function. [0189]
  • Subsequent rounds of recombination and selection/screening proceed in similar fashion to those described for plasmid-plasmid recombination. For example, further recombination can be effected by propagating cells surviving recombination in combination with electroporation or conjugative transfer of plasmids. Alternatively, plasmids bearing additional substrates for recombination can be introduced into the surviving cells. Preferably, such plasmids are from a different incompatibility group and bear a different selective marker than the original plasmids to allow selection for cells containing at least two different plasmids. As a further alternative, plasmid and/or chromosomal DNA can be isolated from a subpopulation of surviving cells and transfected into a second subpopulation. Chromosomal DNA can be cloned into a plasmid vector before transfection. [0190]
  • 5. Virus-Chromosome Recombination [0191]
  • As in the other methods described above, the virus is usually one that does not kill the cells, and is often a phage or phagemid. The procedure is substantially the same as for plasmid-chromosome recombination. Substrates for recombination are cloned into the vector. Vectors including the substrates can then be transfected into cells or in vitro packaged and introduced into cells by infection. Viral genomes recombine with host chromosomes merely by propagating a culture. Evolution can be accelerated by allowing intercellular transfer of viral genomes by electroporation, or reinfection of cells by progeny virions. Screening/selection identifies cells having chromosomes and/or viral genomes that have evolved toward acquisition of a desired function. [0192]
  • There are several options for subsequent rounds of recombination. For example, viral genomes can be transferred between cells surviving selection/recombination by electroporation. Alternatively, viruses extruded from cells surviving selection/screening can be pooled and used to superinfect the cells at high multiplicity. Alternatively, fresh substrates for recombination can be introduced into the cells, either on plasmid or viral vectors. [0193]
  • II. Application of Recursive Sequence Recombination to Evolution of Polypeptides [0194]
  • In addition to the techniques described above, some additionally advantageous modifications of these techniques for the evolution of polypeptides are described below. These methods are referred to as “fine grain” and “coarse grain” shuffling. The coarse grain methods allow one to exchange chunks of genetic material between substrate nucleic acids, thereby limiting diversity in the resulting recombinants to exchanges or substitutions of domains, restriction fragments, oligo-encoded blocks of mutations, or other arbitrarily defined segments, rather than introducing diversity more randomly across the substrate. In contrast to coarse grain shuffling, fine grain shuffling methods allow the generation of all possible recombinations, or permutations, of a given set of very closely linked mutations, including multiple permutations, within a single segment, such as a codon. [0195]
  • In some embodiments, coarse grain or fine grain shuffling techniques are not performed as exhaustive searches of all possible mutations within a nucleic acid sequence. Rather, these techniques are utilized to provide a sampling of variation possible within a gene based on known sequence or structural information. The size of the sample is typically determined by the nature of the screen or selection process. For example, when a screen is performed in a 96-well microtiter format, it may be preferable to limit the size of the recombinant library to about 100 such microtiter plates for convenience in screening. [0196]
  • A. Use of Restriction Enzyme Sites to Recombine Mutations [0197]
  • In some situations it is advantageous to use restriction enzyme sites in nucleic acids to direct the recombination of mutations in a nucleic acid sequence of interest. These techniques are particularly preferred in the evolution of fragments that cannot readily be shuffled by existing methods due to the presence of repeated DNA or other problematic primary sequence motifs. They are also preferred for shuffling large fragments (typically greater than 10 kb), such as gene clusters that cannot be readily shuffled and “PCR-amplified” because of their size. Although fragments up to 50 kb have been reported to be amplified by PCR (Barnes, [0198] Proc. Natl. Acad. Sci. (U.S.A.) 91:2216-2220 (1994)), it can be problematic for fragments over 10 kb, and thus alternative methods for shuffling in the range of 10-50 kb and beyond are preferred. Preferably, the restriction endonucleases used are of the Class II type (Sambrook et al., Molecular Cloning, CSH Press, 1987) and of these, preferably those which generate nonpalindromic sticky end overhangs such as Alwn I, Sfi I or BstX1. These enzymes generate nonpalindromic ends that allow for efficient ordered reassembly with DNA ligase. Typically, restriction enzyme (or endonuclease) sites are identified by conventional restriction enzyme mapping techniques (Sambrook et al., Molecular Cloning, CSH Press, 1987), by analysis of sequence information for that gene, or by introduction of desired restriction sites into a nucleic acid sequence by synthesis (i.e. by incorporation of silent mutations).
  • The DNA substrate molecules to be digested can either be from in vivo replicated DNA, such as a plasmid preparation, or from PCR amplified nucleic acid fragments harboring the restriction enzyme recognition sites of interest, preferably near the ends of the fragment. Typically, at least two variants of a gene of interest, each having one or more mutations, are digested with at least one restriction enzyme determined to cut within the nucleic acid sequence of interest. The restriction fragments are then joined with DNA ligase to generate full length genes having shuffled regions. The number of regions shuffled will depend on the number of cuts within the nucleic acid sequence of interest. The shuffled molecules can be introduced into cells as described above and screened or selected for a desired property. Nucleic acid can then be isolated from pools (libraries) or clones having desired properties and subjected to the same procedure until a desired degree of improvement is obtained. [0199]
  • In some embodiments, at least one DNA substrate molecule or fragment thereof is isolated and subjected to mutagenesis. In some embodiments, the pool or library of religated restriction fragments are subjected to mutagenesis before the digestion-ligation process is repeated. “Mutagenesis” as used herein comprises such techniques known in the art as PCR mutagenesis, oligonucleotide-directed mutagenesis, site-directed mutagenesis, etc., and recursive sequence recombination by any of the techniques described herein. [0200]
  • An example of the use of this format is in the manipulation of polyketide clusters. Polyketide clusters (Khosla et al., [0201] TIBTECH 14, September 1996) are typically 10 to 100 kb in length, specifying multiple large polypeptides which assemble into very large multienzyme complexes. Due to the modular nature of these complexes and the modular nature of the biosynthetic pathway, nucleic acids encoding protein modules can be exchanged between different polyketide clusters to generate novel and functional chimeric polyketides. The introduction of rare restriction endonuclease sites such as SfiI (eight base recognition, nonpalindromic overhangs) at nonessential sites between polypeptides or in introns engineered within polypeptides would provide “handles” with which to manipulate exchange of nucleic acid segments using the technique described above.
  • B. Reassembly PCR [0202]
  • A further technique for recursively recombining mutations in a nucleic acid sequence utilizes “reassembly PCR”. This method can be used to assemble multiple segments that have been separately evolved into a full length nucleic acid template such as a gene. This technique is performed when a pool of advantageous mutants is known from previous work or has been identified by screening mutants that may have been created by any mutagenesis technique known in the art, such as PCR mutagenesis, cassette mutagenesis, doped oligo mutagenesis, chemical mutagenesis, or propagation of the DNA template in vivo in mutator strains. Boundaries defining segments of a nucleic acid sequence of interest preferably lie in intergenic regions, introns, or areas of a gene not likely to have mutations of interest. Preferably, oligonucleotide primers (oligos) are synthesized for PCR amplification of segments of the nucleic acid sequence of interest, such that the sequences of the oligonucleotides overlap the junctions of two segments. The overlap region is typically about 10 to 100 nucleotides in length. Each of the segments is amplified with a set of such primers. The PCR products are then “reassembled” according to assembly protocols such as those used in Sections IA-B above to assemble randomly fragmented genes. In brief, in an assembly protocol the PCR products are first purified away from the primers, by, for example, gel electrophoresis or size exclusion chromatography. Purified products are mixed together and subjected to about 1-10 cycles of denaturing, reannealing, and extension in the presence of polymerase and deoxynucleoside triphosphates (dNTP's) and appropriate buffer salts in the absence of additional primers (“self-priming”). Subsequent PCR with primers flanking the gene are used to amplify the yield of the fully reassembled and shuffled genes. This method is necessarily “coarse grain” and hence only recombines mutations in a blockwise fashion, an advantage for some searches such as when recombining allelic variants of multiple genes within an operon. [0203]
  • In some embodiments, the resulting reassembled genes are subjected to mutagenesis before the process is repeated. [0204]
  • In some embodiments, oligonucleotides that incorporate uracil into the primers are used for PCR amplification. Typically uracil is incorporated at one site in the oligonucleotide. The products are treated with uracil glycosylase, thereby generating a single-stranded overhang, and are reassembled in an ordered fashion by a method such as disclosed by Rashtchian ([0205] Current Biology, 6:30-36 (1995)).
  • In a further embodiment, the PCR primers for amplification of segments of the nucleic acid sequence of interest are used to introduce variation into the gene of interest as follows. Mutations at sites of interest in a nucleic acid sequence are identified by screening or selection, by sequencing homologues of the nucleic acid sequence, and so on. Oligonucleotide PCR primers are then synthesized which encode wild type or mutant information at sites of interest. These primers are then used in PCR mutagenesis to generate libraries of full length genes encoding permutations of wild type and mutant information at the designated positions. This technique is typically advantagous in cases where the screening or selection process is expensive, cumbersome, or impractical relative to the cost of sequencing the genes of mutants of interest and synthesizing mutagenic oligonucleotides. [0206]
  • An example of this method is the evolution of an improved Taq polymerase, as described in detail below. Mutant proteins resulting from application of the method are identified and assayed in a sequencing reaction to identify mutants with improved sequencing properties. This is typically done in a high throughput format (see, for example, Broach et al. [0207] Nature 384 (Supp): 14-16 (1996)) to yield, after screening, a small number, e.g., about 2 to 100, of candidate recombinants for further evaluation. The mutant genes can then be sequenced to provide information regarding the location of the mutation. The corresponding mutagenic oligonucleotide primers can be synthesized from this information, and used in a reassembly reaction as described above to efficiently generate a library with an average of many mutations per gene. Thus, multiple rounds of this protocol allows the efficient search for improved variants of the Taq polymerase.
  • C. Enrichment for Mutant Sequence Information [0208]
  • In some embodiments of the invention, recombination reactions, such as those discussed above, are enriched for mutant sequences so that the multiple mutant spectrum, i.e. possible combinations of mutations, is more efficiently sampled. The rationale for this is as follows. Assume that a number, n, of mutant clones with improved activity is obtained, wherein each clone has a single point mutation at a different position in the nucleic acid sequence. If this population of mutant clones with an average of one mutation of interest per nucleic acid sequence is then put into a recombination reaction, the resulting population will still have an average of one mutation of interest per nucleic acid sequence as defined by a Poisson distribution, leaving the multiple mutation spectrum relatively unpopulated. [0209]
  • The amount of screening required to identify recombinants having two or more mutations can be dramatically reduced by the following technique. The nucleic acid sequences of interest are obtained from a pool of mutant clones and prepared as fragments, typically by digestion with a restriction endonuclease, sonication, or by PCR amplification. The fragments are denatured, then allowed to reanneal, thereby generating mismatched hybrids where one strand of a mutant has hybridized with a complementary strand from a different mutant or wild-type clone. The reannealed products are then fragmented into fragments of about 20-100 bp, for example, by the use of DNAseI. This fragmentation reaction has the effect of segregating regions of the template containing mismatches (mutant information) from those encoding wild type sequence. The mismatched hybrids can then be affinity purified using aptamers, dyes, or other agents which bind to mismatched DNA. A preferred embodiment is the use of mutS protein affinity matrix (Wagner et al., [0210] Nucleic Acids Res. 23(19):3944-3948 (1995); Su et al., Proc. Natl. Acad. Sci. (U.S.A.), 83:5057-5061(1986)) with a preferred step of amplifying the affinity-purified material in vitro prior to an assembly reaction. This amplified material is then put into a assembly PCR reaction as decribed above. Optionally, this material can be titrated against the original mutant pool (e.g., from about 100% to 10% of the mutS enriched pool) to control the average number of mutations per clone in the next round of recombination.
  • Another application of this method is in the assembly of gene constructs that are enriched for polymorphic bases occurring as natural or selected allelic variants or as differences between homologous genes of related species. For example, one may have several varieties of a plant that are believed to have heritable variation in a trait of interest (e.g., drought resistance). It then is of interest to construct a library of these variant genes containing many mutations per gene. MutS selection can be applied in combination with the assembly techniques described herein to generate such a pool of recombinants that are highly enriched for polymorphic (“mutant”) information. In some embodiments, the pool of recombinant genes is provided in a transgenic host. Recombinants can be further evolved by PCR amplification of the transgene from transgenic organisms that are determined to have an improved phenotype and applying the formats described in this invention to further evolve them. [0211]
  • D. Intron-driven Recombination [0212]
  • In some instances, the substrate molecules for recombination have uniformly low homology, sporadically distributed regions of homology, or the region of homology is relatively small (for example, about 10-100 bp), such as phage displayed peptide ligands. These factors can reduce the efficiency and randomness of recombination in RSR. In some embodiments of the invention, this problem is addressed by the introduction of introns between coding exons in sequences encoding protein homologues. In further embodiments of the invention, introns can be used (Chong et al., [0213] J. Biol. Chem., 271:22159-22168 (1996)).
  • In this method, a nucleic acid sequence, such as a gene or gene family, is arbitrarily defined to have segments. The segments are preferably exons. Introns are engineered between the segments. Preferably, the intron inserted between the first and second segments is at least about 10% divergent from the intron inserted between second and third segments, the intron inserted between second and third segments is at least about 10% divergent from the introns inserted between any of the previous segment pairs, and so on through segments n and n+1. The introns between any given set of exons will thus initially be identical between all clones in the library, whereas the exons can be arbitrarily divergent in sequence. The introns therefore provide homologous DNA sequences that will permit application of any of the described methods for RSR while the exons can be arbitrarily small or divergent in sequence, and can evolve to achieve an arbitrarily large degree of sequence divergence without a significant loss in efficiency in recombination. Restriction sites can also be engineered into the intronic nucleic acid sequence of interest so as to allow a directed reassemmbly of restriction fragments. The starting exon DNA may be synthesized de novo from sequence information, or may be present in any nucleic acid preparation (e.g., genomic, cDNA, libraries, and so on). For example, 1 to 10 nonhomologous introns can be designed to direct recombination of the nucleic acid sequences of interest by placing them between exons. The sequence of the introns can be all or partly obtained from known intron sequence. Preferably, the introns are self-splicing. Ordered sets of introns and exon libraries are assembled into functional genes by standard methods (Sambrook et al., [0214] Molecular Cloning, CSH Press (1987)).
  • Any of the formats for in vitro or in vivo recombination described herein can be applied for recursive exon shuffling. A preferred format is to use nonpalindromic restriction sites such as Sfi I placed into the intronic sequences to promote shuffling. Pools of selected clones are digested with Sfi I and religated. The nonpalindromic overhangs promote ordered reassembly of the shuffled exons. These libraries of genes can be expressed and screened for desired properties, then subjected to further recursive rounds of recombination by this process. In some embodiments, the libraries are subjected to mutagenesis before the process is repeated. [0215]
  • An example of how the introduction of an intron into a mammalian library format would be used advantageously is as follows. An intron containing a lox (Sauer et al., [0216] Proc. Natl. Acad. Sci. (U.S.A.), 85:5166-5170 (1988)) site is arbitrarily introduced between amino acids 92 and 93 in each alpha interferon parental substrate. A library of 104 chimeric interferon genes is made for each of the two exons (residues 1-92 and residues 93-167), cloned into a replicating plasmid vector, and introduced into target cells. The number 104 is arbitrarily chosen for convenience in screening. An exemplary vector for expression in mammalian cells would contain an SV40 origin, with the host cells expressing SV40 large T antigen, so as to allow transient expression of the interferon constructs. The cells are challenged with a cytopathic virus such as vesicular stomatitis virus (VSV) in an interferon protection assay (e.g., Meister et al., J. Gen. Virol. 67:1633-1643, (1986)). Cells surviving due to expression of interferon are recovered, the two libraries of interferon genes are PCR amplified, and recloned into a vector that can be amplified in E. coli. The amplified plasmids are then transfected at high multiplicity (e.g. 10 micrograms of plasmid per 106 cells) into a cre expressing host that can support replication of that vector. The presence of cre in the host cells promotes efficient recombination at the lox site in the interferon intron, thus shuffling the selected sets of exons. This population of cells is then used in a second round of selection by viral challenge and the process is applied recursively. In this format, the cre recombinase is preferrably expressed transiently on a cotransfected molecule that cannot replicate in the host. Thus, after segregation of recombinants from the cre expressing plasmid, no further recombination will occur and selection can be performed on genetically stable exon permutations. The method can be used with more than one intron, with recombination enhancing sequences other than cre/lox (e.g., int/xis, etc.), and with other vector systems such as but not limited to retroviruses, adenovirus or adeno- associated virus.
  • 5. Synthetic Oligonucleotide Mediated Recombination [0217]
  • 1. Oligo Bridge Across Sequence Space [0218]
  • In some embodiments of the invention, a search of a region of sequence space defined by a set of substrates, such as members of a gene family, having less than about 80%, more typically, less than about 50% homology, is desired. This region, which can be part or all of a gene or a gene is arbitrarily delineated into segments. The segment borders can be chosen randomly, based on correspondence with natural exons, based on structural considerations (loops, alpha helices, subdomains, whole domains, hydrophobic core, surface, dynamic simulations), and based on correlations with genetic mapping data. [0219]
  • Typically, the segments are then amplified by PCR with a pool of “bridge” oligonucleotides at each junction. Thus, if the set of five genes is broken into three segments A, B and C, and if there are five versions of each segment (A[0220] 1, A2, . . . C4, C5), twenty five oligonucleotides are made for each strand of the A-B junctions where each bridge oligo has 20 bases of homology to one of the A and one of the B segments. In some cases, the number of required oligonucleotides can be reduced by choosing segment boundaries that are identical in some or all of the gene family members. Oligonucleotides are similarly synthesized for the B-C junction. The family of A domains is amplified by PCR with an outside generic A primer and the pool of A-B junction oligonucleotides; the B domains with the A-B plus the B-C bridge oligonucleotides, and the C domains with the B-C bridge oligonucleotides plus a generic outside primer. Full length genes are made then made by assembly PCR or by the dUTP/uracil glycosylase methods described above. Preferably, products from this step are subjected to mutagenesis before the process of selection and recombination is repeated, until a desired level of improvement or the evolution of a desired property is obtained. This is typically determined using a screening or selection as appropriate for the protein and property of interest.
  • An illustration of this method is illustrated below for the recombination of eleven homologous human alpha interferon genes. [0221]
  • 2. Site Directed Mutagenesis (SDM) with Oligonucleotides Encoding Homologue Mutations Followed by Shuffling [0222]
  • In some embodiments of the invention, sequence information from one or more substrate sequences is added to a given “parental” sequence of interest, with subsequent recombination between rounds of screening or selection. Typically, this is done with site-directed mutagenesis performed by techniques well known in the art (Sambrook et al., [0223] Molecular Cloning, CSH Press (1987)) with one substrate as template and oligonucleotides encoding single or multiple mutations from other substrate sequences, e.g. homologous genes. After screening or selection for an improved phenotype of interest, the selected recombinant(s) can be further evolved using RSR techniques described herein. After screening or selection, site-directed mutagenesis can be done again with another collection of oligonucleotides encoding homologue mutations, and the above process repeated until the desired properties are obtained.
  • When the difference between two homologues is one or more single point mutations in a codon, degenerate oligonucleotides can be used that encode the sequences in both homologues. One oligo may include many such degenerate codons and still allow one to exhaustively search all permutations over that block of sequence. An example of this is provided below for the evolution of alpha interferon genes. [0224]
  • When the homologue sequence space is very large, it can be advantageous to restrict the search to certain variants. Thus, for example, computer modelling tools (Lathrop et al., [0225] J. Mol. Biol., 255:641-665 (1996)) can be used to model each homologue mutation onto the target protein and discard any mutations that are predicted to grossly disrupt structure and function.
  • F. Recombination Directed by Host Machinery [0226]
  • In some embodiments of the invention, DNA substrate molecules are introduced into cells, wherein the cellular machinery directs their recombination. For example, a library of mutants is constructed and screened or selected for mutants with improved phenotypes by any of the techniques described herein. The DNA substrate molecules encoding the best candidates are recovered by any of the techniques described herein, then fragmented and used to transfect a mammalian host and screened or selected for improved function. The DNA substrate molecules are recovered from the mammalian host, such as by PCR, and the process is repeated until a desired level of improvement is obtained. In some embodiments, the fragments are denatured and reannealed prior to transfection, coated with recombination stimulating proteins such as recA, or co-transfected with a selectable marker such as Neo[0227] R to allow the positive selection for cells receiving recombined versions of the gene of interest.
  • For example, this format is preferred for the in vivo affinity maturation of an antibody by RSR. In brief, a library of mutant antibodies is generated, as described herein for the 48G7 affinity maturation. This library is FACS purified with ligand to enrich for antibodies with the highest 0.1-10% affinity. The V regions genes are recovered by PCR, fragmented, and cotransfected or electorporated with a vector into which reassembled V region genes can recombine. DNA substrate molecules are recovered from the cotranfected cells, and the process is repeated until the desired level of improvment is obtained. Other embodiments include reassembling the V regions prior to the electroporation so that an intact V region exon can recombine into an antibody expression cassette. Further embodiments include the use of this format for other eukaryotic genes or for the evolution of whole viruses. [0228]
  • G. Phagemid-Based Assembly [0229]
  • In some embodiments of the invention, a gene of interest is cloned into a vector that generates single stranded DNA, such as a phagemid. The resulting DNA substrate is mutagenzied by RSR in any method known in the art, transfected into host cells, and subjected to a screen or selection for a desired property or improved phenotype. DNA from the selected or screened phagemids is amplified, by, for example, PCR or plasmid preparation. This DNA preparation contains the various mutant sequences that one wishes to permute. This DNA is fragmented and denatured, and annealed with single-stranded DNA (ssDNA) phagemid template (ssDNA encoding the wild-type gene and vector sequences). A preferred embodiment is the use of dut(−) ung(−) host strains such as CJ236 (Sambrook et al., [0230] Molecular Cloning CSH Press (1987)) for the preparation of ssDNA.
  • Gaps in annealed template are filled with DNA polymerase and ligated to form closed relaxed circles. Since multiple fragments can anneal to the phagemid, the newly synthesized strand now consists of shuffled sequences. These products are transformed into a mutS strain of [0231] E. coli which is dut+ ung+. Phagemid DNA is recovered from the transfected host and subjected again to this protocol until the desired level of improvement is obtained. The gene encoding the protein of interest in this library of recovered phagemid DNA can be mutagenzied by any technique, including RSR, before the process is repeated.
  • III. Improved Protein Expression [0232]
  • While recombinant DNA technology has proved to be a very general method for obtaining large, pure, and homogeneous quantities of almost all nucleic acid sequences of interest, similar generality has not yet been achieved for the production of large amounts of pure, homogeneous protein in recombinant form. A likely explanation is that protein expression, folding, localization and stability is intrinsically more complex and unpredictable than for DNA. The yield of expressed protein is a complex function of transcription rates, translation rates, interactions with the ribosome, interaction of the nascent polypeptide with chaperonins and other proteins in the cell, efficiency of oligomerization, interaction with components of secretion and other protein trafficking pathways, protease sensitivity, and the intrinsic stability of the final folded state. Optimization of such complex processes is well suited for the application of RSR. The following methods detail strategies for application of RSR to the optimization of protein expression. [0233]
  • A. Evolution of Mutant Genes with Improved Expression Using RSR on Codon Usage Libraries [0234]
  • The negative effect of rare [0235] E. coli codons on expression of recombinant proteins in this host has been clearly demonstrated (Rosenberg, et al., J. Bact. 175:716-722 (1993)). However, general rules for the choice of codon usage patterns to optimize expression of functional protein have been elusive. In some embodiments of the invention, protein expression is optimized by changing codons used in the gene of interest, based on the degeneracy of the genetic code. Typically, this is accomplished by synthesizing the gene using degenerate oligonucleotides. In some embodiments the degenerate oligonucleotides have the general structure of about 20 nucleotides of identity to a DNA substrate molecule encoding a protein of interest, followed by a region of about 20 degenerate nucleotides which encode a region of the protein, followed by another region of about 20 nucleotides of identity. In a preferred embodiment, the region of identity utilizes preferred codons for the host. In a further embodiment, the oligonucleotides are identical to the DNA substrate at least one 5′ and one 3′ nucleotide, but have at least 85% sequence homology to the DNA substrate molecule, with the difference due to the use of degenerate codons. In some embodiments, a set of such degenerate oligonucleotides is used in which each oligonucleotide overlaps with another by the general formula n−10, wherein n is the length of the oligonucleotide. Such oligonucleotides are typically about 20-1000 nucleotides in length. The assembled genes are then cloned, expressed, and screened or selected for improved expression. The assembled genes can be subjected to recursive recombination methods as descibed above until the desired improvement is achieved.
  • For example, this technique can be used to evolve bovine intestinal alkaline phosphatase (BIAP) for active expression in [0236] E. coli. This enzyme is commonly used as a reporter gene in assay formats such as ELISA. The cloned gene cannot be expressed in active form in a prokaryotic host such as E. coli in good yield. Development of such an expression system would allow one to access inexpensive expression technology for BIAP and, importantly, for engineered variants with improved activity or chemical coupling properties (such as chemical coupling to antibodies). A detailed example is provide in the Experimental Examples section.
  • B. Improved Folding [0237]
  • In some embodiments of the invention, proteins of interest when overexpressed or expressed in heterologous hosts form inclusion bodies, with the majority of the expressed protein being found in insoluble aggregates. Recursive sequence recombination techniques can be used to optimize folding of such target proteins. There are several ways to improve folding, including mutating evolving the target protein of interest and evolving chaperonin proteins. [0238]
  • 1. Evolving A Target Protein [0239]
  • a. Inclusion Body Fractionation Selection Using lac Headpiece Dimer Fusion Protein [0240]
  • The lac repressor “headpiece dimer” is a small protein containing two headpiece domains connected by a short peptide linker which binds the lac operator with sufficient affinity that polypeptide fusions to this headpiece dimer will remain bound to the plasmid that encodes them throughout an affinity purification process (Gates et al., [0241] J. Mol. Biol. 255:373-386 (1995)). This property can be exploited, as follows, to evolve mutant proteins of interest with improved folding properties. The protein of interest can be mammalian, yeast, bacterial, etc.
  • A fusion protein between the lac headpiece dimer and a target protein sequence is constructed, for example, as disclosed by Gates (supra). This construct, containing at least one lac operator, is mutagenized by technologies common in the arts such as PCR mutagenesis, chemical mutagenesis, oligo directed mutagenesis (Sambrook et al., [0242] Molecular Cloning CSH Press (1987)). The resulting library is transformed into a host cell, and expression of the fusion protein is induced, preferably with arabinose. An extract or lysate is generated from a culture of the library expressing the construct. Insoluble protein is fractionated from soluble protein/DNA complexes by centrifugation or affinity chromatography, and the yield of soluble protein/DNA complexes is quantitated by quantitative PCR (Sambrook et al., Molecular Cloning, CSH Press, 1987) of the plasmid. Preferably, a reagent that is specific for properly folded protein, such as a monoclonal antibody or a natural ligand, is used to purify soluble protein/DNA complexes. The plasmid DNA from this step is isolated, subjected to RSR and again expressed. These steps are repeated until the yield of soluble protein/DNA complexes has reached a desired level of improvement. Individual clones are then screened for retention of functional properties of the protein of interest, such as enzymatic activity, etc.
  • This technique is generically useful for evolving solubility and other properties such as cellular trafficking of proteins heterologously expressed in a host cell of interest. For example, one could select for efficient folding and nuclear localization of a protein fused to the lac repressor headpiece dimer by encoding the protein on a plasmid encoding an SV40 origin of replication and a lac operator, and transiently expressing the fusion protein in a mammalian host expressing T antigen. Purification of protein/DNA complexes from nuclear HIRT extracts (Seed and Aruffo, [0243] Proc. Natl. Acad. Sci. (U.S.A.), 84:3365-3369 (1987)) would allow one to select for efficient folding and nuclear localization proteins.
  • b. Functional Expression of Protein Using Phase Display [0244]
  • A problem often encountered in phage display methods such as those disclosed by O'Neil et al. ([0245] Current Biology, 5:443-449 (1995)) is the inability to functionally express a protein of interest on phage. Without being limited to any one theory, improper folding of the protein of interest can be responsible for this problem. RSR can be used to evolve a protein of interest for functional expression on phage. Typically, a fusion protein is constructed between gene III or gene VIII and the target protein and then mutagenized, for example by PCR mutagenesis. The mutagenzied library is then expressed in a phage display format, a phage lysate is made, and these phage are affinity selected for those bearing functionally displayed fusion proteins using an affinity matrix containing a known ligand for the target protein. DNA from the functionally selected phage is purified, and the displayed genes of interest are shuffled and recloned into the phage display format. The selection, shuffling and recloning steps are repeated until the yield of phage with functional displayed protein has reached desired levels as defined, for example, by the fraction of phage that are retained on a ligand affinity matrix or the biological activity associated with the displayed phage. Individual clones are then screened to identify candidate mutants with improved display properties, desired level of expression, and functional properties of interest (e.g., ability to bind a ligand or receptor, lymphokine activity, enzymatic activity, etc.).
  • In some embodiments of the invention, a functional screen or selection is used to identify an evolved protein not expressed on a phage. The target protein, which cannot initially be efficiently expressed in a host of interest, is mutagenized and a functional screen or selection is used to identify cells expressing functional protein. For example, the protein of interest may complement a function in the host cell, cleave a colorimetric substrate, etc. Recursive sequence recombination is then used to rapidly evolve improved functional expression from such a pool of improved mutants. [0246]
  • For example, AMV reverse transcriptase is of particular commercial importance because it is active at a higher temperature (42° C.) and is more robust than many other reverse transcriptases. However, it is difficult to express in prokaryotic hosts such as [0247] E. coli, and is consequently expensive because it has to be purified from chicken cells. Thus an evolved AMV reverse transcriptase that can be expressed efficiently in E. coli is highly desirable.
  • In brief, the AMV reverse transcriptase gene (Papas et al., [0248] J. Cellular Biochem 20:95-103 (1982)) is mutagenized by any method common in the art. The library of mutant genes is cloned into a colE1 plasmid (Amp resistant) under control of the lac promoter in a polA12 (Ts) recA718 (Sweasy et al. Proc. Natl. Acad. Sci. U.S.A. 90:4626-4630 (1993)) E. coli host. The library is induced with IPTG, and shifted to the nonpermissive temperature. This selects for functionally expressed reverse transcriptase genes under the selective conditions reported for selection of active HIV reverse transcriptase mutants reported by Kim et al. (Proc. Natl. Acad. Sci. (U.S.A.), 92:684-688 (1995)). The selected AMV RTX genes are recovered by PCR by using oligonucleotides flanking the cloned gene. The resulting PCR products are subjected to in vitro RSR, selected as described above, and the process is repeated until the level of functional expression is acceptable. Individual clones are then screened for RNA-dependent DNA polymerization and other properties of interest (e.g. half life at room temperature, error rate). The candidate clones are subjected to mutagenesis, and then tested again to yield an AMV RT that can be expressed in E. coli at high levels.
  • 2. Evolved Chaperoning [0249]
  • In some embodiments of the invention, overexpression of a protein can lead to the accumulation of folding intermediates which have a tendency to aggregate. Without being limited to any one theory, the role of chaperonins is thought to be to stabilize such folding intermediates against aggregration; thus, overexpression of a protein of interest can lead to overwhelming the capacity of chaperoning. Chaperonin genes can be evolved using the techniques of the invention, either alone or in combination with the genes encoding the protein of interest, to overcome this problem. [0250]
  • Examples of proteins of interest which are especially suited to this approach include but are not limited to: cytokines; malarial coat proteins; T cell receptors; antibodies; industrial enzymes (e.g., detergent proteases and detergent lipases); viral proteins for use in vaccines; and plant seed storage proteins. [0251]
  • Sources of chaperonin genes include but are limited to [0252] E. coli chaperonin genes encoding such proteins as thioredoxin, Gro ES/Gro EL, PapD, ClpB, DsbA, DsbB, DnaJ, DnaK, and GrpE; mammalian chaperoning such as Hsp70, Hsp72, Hsp73, Hsp40,Hsp60, Hsp10, Hdj1, TCP-1, Cpn60, BiP; and the homologues of these chaperonin genes in other species such as yeast (J. G. Wall and A. Pluckthun, Current Biology, 6:507-516 (1995); Hartl, Nature, 381:571-580 (1996)). Additionally, heterologous genomic or cDNA libraries can be used as libraries to select or screen for novel chaperoning.
  • In general, evolution of chaperonins is accomplished by first mutagenizing chaperonin genes, screening or selecting for improved expression of the target protein of interest, subjecting the mutated chaperonin genes to RSR, and repeating selection or screening. As with all RSR techniques, this is repeated until the desired improvement of expression of the protein of interest is obtained. Two exemplary approaches are provide below. [0253]
  • a. Chaperonin Evolution in Trans to the Protein of Interest With a Screen or Selection for Improved Function [0254]
  • In some embodiments the chaperonin genes are evolved independently of the gene(s) for the protein of interest. The improvement in the evolved chaperonin can be assayed, for example, by screening for enhancement of the activity of the target protein itself or for the activity of a fusion protein comprising the target protein and a selectable or screenable protein (e.g., GFP, alkaline phosphatase or beta-galactosidase). [0255]
  • b. Chaperonin Operon in cis [0256]
  • In some embodiments, the chaperonin genes and the target protein genes are encoded on the same plasmid, but not necessarily evolved together. For example, a lac headpiece dimer can be fused to the protein target to allow for selection of plasmids which encode soluble protein. Chaperonin genes are provided on this same plasmid (“cis”) and are shuffled and evolved rather than the target protein. Similarly, the chaperonin genes can be cloned onto a phagemid plasmid that encodes a gene III or gene VIII fusion with a protein of interest. The cloned chaperonins are mutagenized and, as with the selection described above, phage expressing functionally displayed fusion protein are isolated on an affinity matrix. The chaperonin genes from these phage are shuffled and the cycle of selection, mutation and recombination are applied recursively until fusion proteins are efficiently displayed in functional form. [0257]
  • 3. Improved Intracellular Localization [0258]
  • Many overexpressed proteins of biotechnological interest are secreted into the periplasm or media to give advantages in purification or activity assays. Optimization for high level secretion is difficult because the process is controlled by many genes and hence optimization may require multiple mutations affecting the expression level and structure of several of these components. Protein secretion in [0259] E. coli, for example, is known to be influenced by many proteins including: a secretory ATPase (SecA), a translocase complex (SecB, SecD, SecE, SecF, and SecY), chaperonins (DnaK, DnaJ, GroES, GroEL), signal peptidases (LepB, LspA, Ppp), specific folding catalysts (DsbA) and other proteins of less well defined function (e.g., Ffh, FtsY) (Sandkvist et al., Curr. Op. Biotechnol. 7:505-511 (1996)). Overproduction of wild type or mutant copies of these genes for these proteins can significantly increase the yield of mature secreted protein. For example, overexpression of secY or secY4 significantly increased the periplasmic yield of mature human IL6 from a hIL6-pre-OmpA fusion (Perez-Perez et al., Bio-Technology 12:178-180 (1994)). Analogously, overexpression of DnaK/DnaJ in E. coli improved the yield of secreted human granulocyte colony stimulating factor (Perez-Perez et al., Biochem. Biophys. Res. Commun. 210:254-259 (1995)).
  • RSR provides a route to evolution of one or more of the above named components of the secretory pathway. The following strategy is employed to optimize protein secretion in [0260] E. coli. Variations on this method, suitable for application to Bacillus subtilis, Pseudomonas, Saccaromyces cerevisiæ, Pichia pastoris, mammalian cells and other hosts are also described. The general protocol is as follows.
  • One or more of the genes named above are obtained by PCR amplification from [0261] E. coli genomic DNA using known flanking sequence, and cloned in an ordered array into a plasmid or cosmid vector. These genes do not in general occur naturally in clusters, and hence these will comprise artificial gene clusters. The genes may be cloned under the control of their natural promoter or under the control of another promoter such as the lac, tac, arabinose, or trp promoters. Typically, rare restriction sites such as Sfi I are placed between the genes to facilitate ordered reassembly of shuffled genes as described in the methods of the invention.
  • The gene cluster is mutagenized and introduced into a host cell in which the gene of interest can be inducibly expressed. Expression of the target gene to be secreted and of the cloned genes is induced by standard methods for the promoter of interest (e.g., addition of 1 mM IPTG for the lac promoter). The efficiency of protein secretion by a library of mutants is measured, for example by the method of colony blotting (Skerra et al., [0262] Anal. Biochem. 196:151-155 (1991)). Those colonies expressing the highest levels of secreted protein (the top 0.1-10%; preferably the top 1%) are picked. Plasmid DNA is prepared from these colonies and shuffled according to any of the methods of the invention.
  • Preferably, each individual gene is amplified from the population and subjected to RSR. The fragments are digested with Sfi I (introduced between each gene with nonpalindromic overhangs designed to promote ordered reassembly by DNA ligase) and ligated together, preferably at low dilution to promote formation of covalently closed relaxed circles (<1 ng/microliter). Each of the PCR amplified gene populations may be shuffled prior to reassembly into the final gene cluster. The ligation products are transformed back into the host of interest and the cycle of selection and RSR is repeated. [0263]
  • Analogous strategies can be employed in other hosts such as [0264] Pseudomonas, Bacillus subtilis, yeast and mammalian cells. The homologs of the E. coli genes listed above are targets for optimization, and indeed many of these homologs have been identified in other species (Pugsley, Microb. Rev. 57:50-108 (1993)). In addition to these homologs, other components such as the six polypeptides of the signal recognition particle, the trans-locating chain-associating membrane protein (TRAM), BiP, the Ssa proteins and other hsp70 homologs, and prsA (B. subtilis) (Simonen and Pulva, Microb. Rev. 57:109-137 (1993)) are targets for optimization by RSR. In general, replicating episomal vectors such as SV40-neo (Sambrook et al., Molecular Cloning, CSH Press (1987), Northrup et al., J. Biol. Chem. 268(4):2917-2923 (1993)) for mammalian cells or 2 micron or ars plasmids for yeast (Strathern et al., The Molecular Biology of the Yeast Saccaromyces, CSH Press (1982)) are used. Integrative vectors such as pJM 103, pJM 113 or pSGMU2 are preferred for B. subtilis (Perego, Chap. 42, pp. 615-624 in: Bacillus subtilis and Other Gram-Positive Bacteria, A. Sonenshein, J. Hoch, and R. Losick, eds., 1993).
  • For example, an efficiently secreted thermostable DNA polymerase can be evolved, thus allowing the performance of DNA polymerization assays with little or no purification of the expressed DNA polymerase. Such a procedure would be preferred for the expression of libraries of mutants of any protein that one wished to test in a high throughput assay, for example any of the pharmaceutical proteins listed in Table I, or any industrial enzyme. Initial constructs are made by fusing a signal peptide such as that from STII or OmpA to the amino terminus of the protein to be secreted. A gene cluster of cloned genes believed to act in the secretory pathway of interest are mutagenized and coexpressed with the target construct. Individual clones are screened for expresion of the gene product. The secretory gene clusters from improved clones are recovered and recloned and introduced back into the original host. Preferably, they are first subjected to mutagenesis before the process is repeated. This cycle is repeated until the desired improvement in expression of secreted protein is achieved. [0265]
  • IV. Evolved Polypeptide Properties [0266]
  • A. Evolved Transition State Analog and Substrate Binding [0267]
  • There are many enzymes of industrial interest that have substantially suboptimal activity on the substrate of interest. In many of these cases, the enzyme obtained from nature is required to work either under conditions that are very different from the conditions under which it evolved or to have activity towards a substrate that is different from the natural substrate. [0268]
  • The application of evolutionary technologies to industrial enzymes is often significantly limited by the types of selections that can be applied and the modest numbers of mutants that can be surveyed in screens. Selection of enzymes or catalytic antibodies, expressed in a display format, for binding to transition state analogs (McCafferty et al., [0269] Appl. Biochem. Biotechnol. 47:157-171 (1994)) or substrate analogs (Janda et al., Proc. Natl. Acad. Sci. (U.S.A.) 91:2532-2536, (1994)) represents a general strategy for selecting for mutants with with improved catalytic efficiency.
  • Phage display (O'Neil et al., [0270] Current Biology 5:443-449 (1995) and the other display formats (Gates et al., J. Mol. Biol. 255:373-386 (1995); Mattheakis et al., Proc. Natl. Acad. Sci. (U.S.A.) 91:9022-9026 (1994)) described herein represent general methodologies for applying affinity-based selections to proteins of interest. For example, Matthews and Wells (Science 260:1113-1117 (1993)) have used phage display of a protease substrate to select improved substrates. Display of active enzymes on the surface of phage, on the other hand, allows selection of mutant proteins with improved transition state analog binding. Improvements in affinity for transition state analogs correlate with improvements in catalytic efficiency. For example, Patten et al., Science 271:1086-1091 (1996) have shown that improvements in affinity of a catalytic antibody for its hapten are well correlated with improvements in catalytic efficiency, with an 80-fold improvement in kcat/Km being achieved for an esterolytic antibody.
  • For example, an enzyme used in antibiotic biosynthesis can be evolved for new substrate specificity and activity under desired conditions using phage display selections. Some antibiotics are currently made by chemical modifications of biologically produced starting compounds. Complete biosynthesis of the desired molecules is currently impractical because of the lack of an enzyme with the required enzymatic activity and substrate specificity (Skatrud, [0271] TIBTECH 10:324-329, September 1992). For example, 7-aminodeacetooxycephalosporanic acid (7-ADCA) is a precursor for semi-synthetically produced cephalosporins. 7-ADCA is made by a chemical ring expansion of penicillin G followed by enzymatic deacylation of the phenoxyacetal group. 7-ADCA can be made enzymatically from deacetylcephalosporin C (DAOC V), which could in turn be derived from penicillin V by enzymatic ring expansion if a suitably modified penicillin expandase could be evolved (Cantwell et al., Curr. Genet. 17:213-221 (1990)). Thus, 7-ADCA could in principle 35 be produced enzymatically from penicillin V using a modified penicillin N expandase, such as mutant forms of the S. clavuligerus cefE gene (Skatrud, TIBTECH 10:324-329, September 1992). However, penicillin V is not accepted as a substrate by any known expandase with sufficient efficiency to be commercially useful. As outlined below, RSR techniques of the invention can be used to evolve the penicillin expandase encoded by cefE or other expandases so that they will use penicillin V as a substrate.
  • Phage display or other display format selections are applied to this problem by expressing libraries of cefE penicillin expandase mutants in a display format, selecting for binding to substrates or transition state analogs, and applying RSR to rapidly evolve high affinity binders. Candidates are further screened to identify mutants with improved enzymatic activity on penicillin V under desired reaction conditions, such as pH, temperature, solvent concentration, etc. RSR is applied to further evolve mutants with the desired expandase activity. A number of transition state analogs (TSA's) are suitable for this reaction. The following structure is the initial TSA that is used for selection of the display library of cefE mutants: [0272]
    Figure US20040214277A1-20041028-C00001
  • Libraries of the known penicillin expandases (Skatrud, [0273] TIBTECH 10:324-329(1992); Cantwell et al., Curr. Genet. 17:13-221 (1990)) are made as described herein. The display library is subjected to selection for binding to penicillin V and/or to transition state analog given above for the conversion of penicillin V to DAOC V. These binding selections may be performed under non-physiological reaction conditions, such as elevated temperature, to obtain mutants that are active under the new conditions. RSR is applied to evolve mutants with 2-105 fold improvement in binding affinity for the selecting ligand. When the desired level of improved binding has been obtained, candidate mutants are expressed in a high throughput format and specific activity for expanding penicillin V to DAOC V is quantitatively measured. Recombinants with improved enzymatic activity are mutagenized and the process repeated to further evolve them.
  • Retention of TSA binding by a displayed enzyme (e.g., phage display, lac headpiece dimer, polysome display, etc.) is a good selection for retention of the overall integrity of the active site and hence can be exploited to select for mutants which retain activity under conditions of interest. Such conditions include but are not limited to: different pH optima, broader pH optima, activity in altered solvents such as DMSO (Seto et al., [0274] DNA Sequence 5:131-140 (1995)) or formamide (Chen et al., Proc. Natl. Acad. Sci. (U.S.A.) 90:5618-5622, (1993)) altered temperature, improved shelf life, altered or broadened substrate specificity, or protease resistance. A further example, the evolution of a p-nitrophenyl esterase, using a mammalian display format, is provided below.
  • B. Improvement of DNA and RNA Polymerases [0275]
  • Of particular commercial importance are improved polymerases for use in nucleic acid sequencing and polymerase chain reactions. The following properties are attractive candidates for improvement of a DNA sequencing polymerase: (1) suppression of termination by inosine in labelled primer format (H. Dierick et al., [0276] Nucleic Acids Res. 21:4427-4428 (1993)) (2) more normalized peak heights, especially with fluorescently labelled dideoxy terminators (Parker et al., BioTechniques 19:116-121 (1995)), (3) better sequencing of high GC content DNA (>60% GC) by, for example, tolerating >10% DMSO (D. Seto et al., DNA Sequence 5:131-140 (1995); Scheidl et al., BioTechniques 19(5):691-694 (1995)), or (4) improved acceptance of novel base analogs such as inosine, 7-deaza dGTP (Dierick et al., Nucleic Acids Res. 21:4427-4428 (1993)) or other novel base analogs that improve the above properties.
  • Novel sequencing formats have been described which use matrix assisted laser desorption ionization time of flight (MALDT-TOF) mass spectroscopy to resolve dideoxy ladders. (Smith, [0277] Nature Biotechnology 14:1084-1085 (1996)). It is noted in Smith's recent review that fragmentation of the DNA is the singular feature limiting the development of this method as a viable alternative to standard gel electrophoresis for DNA sequencing. Base analogs which stabilize the N-glycosidic bond by modifications of the purine bases to 7-deaza analogs (Kirpekar et al., Rapid Comm. in Mass Spec. 9:525-531 (1995)) or of the 2′ hydroxyl (such as 2′-H or 2′-F) “relieve greatly the mass range limitation” of this technique (Smith, 1996). Thus, evolved polymerases that can efficiently incorporate these and other base analogs conferring resistance to fragmentation under MALDI-TOF conditions are valuable innovations.
  • Other polymerase properties of interest for improvement by RSR are low fidelity thermostable DNA polymerase for more efficient mutagenesis or as a useful correlate for acceptance of base analogs for the purposes described above; higher fidelity polymerase for PCR (Lundberg et al., [0278] Gene 108:1-6 (1991)); higher fidelity reverse transcriptase for retroviral gene therapy vehicles to reduce mutation of the therapeutic construct and of the retrovirus; improved PCR of GC rich DNA and PCR with modified bases (S. Turner and F. J. Jenkins, BioTechniques 19(1):48-52 (1995)).
  • Thus, in some embodiments of the invention, libraries of mutant polymerase genes are screened by direct high throughput screening for improved sequencing properties. The best candidates are then subjected to RSR. Briefly, mutant libraries of candidate polymerases such as Taq polymerase are constructed using standard methods such as PCR mutagenesis (Caldwell et al., [0279] PCR Meth. App. 2:28-33 (1992)) and/or cassette mutagenesis (Sambrook et al., Molecular Cloning, CSH Press (1987)). Incorporation of mutations into Taq DNA polymerase such as the active site residue from T7 polymerase that improves acceptance of dideoxy nucleotides (Tabor and Richardson, J. Biol. Chem. 265:8322-8328 (1990)) and mutations that inactivate the 5′-3′ exonuclease activity (R. S. Rano, BioTechniques 18:390-396 (1995)) are incorporated into these libraries. The reassembly PCR technique, for example, as described above is especially suitable for this problem. Similarly, chimeric polymerase libraries are made by breeding existing thermophilic polymerases, sequenase, and E. coli polI with each other using the bridge oligonucleotide methods described above. The libraries are expressed in formats wherein human or robotic colony picking is used to replica pick individual colonies into 96 well plates where small cultures are grown, and polymerase expression is induced.
  • A high throughput, small scale simple purification for polymerase expressed in each well is performed. For example, simple single-step purifications of His-tagged Taq expressed in [0280] E. coli have been described (Smirnov et al., Russian J. Bioorganic Chem. 21(5):341-342 (1995)), and could readily be adapted for a 96-well expression and purification format.
  • A high throughput sequencing assay is used to perform sequencing reactions with the purified samples. The data is analyzed to identify mutants with improved sequencing properties, according to any of these criteria: higher quality ladders on GC-rich templates, especially greater than 60% GC, including such points as fewer artifactual termination products and stronger signals than given with the wild-type enzyme; less termination of reactions by inosine in primer labelled reactions, e.g., fluorescent labelled primers; less variation in incorporation of signals in reactions with fluorescent dideoxy nucleotides at any given position; longer sequencing ladders than obtained with the wild-type enzyme, such as about 20 to 100 nucleotides; improved acceptance of other known base analogs such as 7-deaza purines; improved acceptance of new base analogs from combinatorial chemistry libraries (See, for example, Hogan, [0281] Nature 384(Supp):17-1996).
  • The best candidates are then subjected to mutagenesis, and then selected or screened for the improved sequencing properties decribed above. [0282]
  • In another embodiment, a screen or selection is performed as follows. The replication of a plasmid can be placed under obligate control of a polymerase expressed in [0283] E. coli or another microorganism. The effectiveness of this system has been demonstrated for making plasmid replication dependent on mammalian polymerase beta (Sweasy et al., Proc. Natl. Acad. Sci. (U.S.A.) 90:4626-4630, (1993)), Taq polymerase (Suzuki et al., Proc. Natl. Acad. Sci. (U.S.A.) 93:9670-9675 (1996)), or HIV reverse transcriptase (Kim et al., Proc. Natl. Acad. Sci. (U.S.A.) 92:684-688 (1995)). The mutant polymerase gene is placed on a plasmid bearing a colE1 origin and expressed under the control of an arabinose promoter. The library is enriched for active polymerases essentially as described by Suzuki et al., (supra), with polymerase expression being induced by the presence of arabinose in the culture.
  • A further quantitative screen utilizes the presence of GFP (green fluorescence protein) on the same plasmid, replica plating onto arabinose at the nonpermissive temperature in the absence of a selective antibiotic, and using a fluorimeter to quantitatively measure fluorescence of each culture. GFP activity correlates with plasmid stability and copy number which is in turn dependent on expression of active polymerase. [0284]
  • A polymerase with a very high error rate would be a superior sequencing enzyme, as it would have a more normalized signal for incorporation of base analogs such as the currently used fluorescently labelled dideoxies because it will have reduced specificity and selectivity. The error rates of currently used polymerases are on the order of 10[0285] −5 to 10−6, orders of magnitude lower than what can be detected given the resolving power of the gel systems. An error rate of 1%, and possibly as high as 10%, could not be detected by current gel systems, and thus there is a large window of opportunity to increase the “sloppiness” of the enzyme. An error-prone cycling polymerase would have other uses such as for hypermutagenesis of genes by PCR.
  • In some embodiments, the system described by Suzuki (Suzuki et al., [0286] Proc. Natl. Acad. Sci. (U.S.A.) 96:9670-9675 (1996)) is used to make replication of a reporter plasmid dependent on the expressed polymerase. This system puts replication of the first 200-300 bases next to the ColE1 origin directly under the control of the expressed polymerase (Sweasy and Loeb, J. Bact. 177:2923-2925 (1995); Sweasy et al., Proc. Natl. Acad. Sci. (U.S.A.) 90:4626-4630 (1993)). A screenable or selectable reporter gene containing stop codons is positioned in this region, such as LacZ alpha containing one, two or three stop codons. The constructs are grown on arabinose at the nonpermissible temperature, allowed to recover, and plated on selective lactose minimal media that demands reversion of the stop codons in the reporter cassette. Mutant polymerases are recovered from the survivors by PCR. The survivors are enriched for mutators because their mutator phenotype increases the rate of reversion of stop codons in the reporter lacZ alpha fragment.
  • The polymerase genes from the survivors are subjected to RSR, then the polymerase mutants are retransformed into the indicator strain. Mutators can be visually screened by plating on arabinose/Xgal plates at the nonpermissive temperature. Mutator polymerases will give rise to colonies with a high frequency of blue papillae due to reversion of the stop codon(s). Candidate papillators can be rescreened by picking a non-papillating region of the most heavily papillated colonies (i.e, “best” colonies) and replating on the arabinose/Xgal indicator medium to further screen for colonies with increased papillation rates. These steps are repeated until a desired reversion rate is achieved (e.g., 10[0287] −2 to 10−3 mutations per base pair per replication).
  • Colonies which exhibit high frequency papillation are candidates for encoding an error prone polymerase. These candidates are screened for improved sequencing properties: essentially as for the high throughput screen described above. Briefly, mutant Taq proteins are expressed and purified in a 96-well format. The purified proteins are used in sequencing reactions and the sequence data are analyzed to identify mutants that exhibit the improvements outlined herein. Mutants with improved properties are subjected to RSR and rescreened for further improvements in function. [0288]
  • In some embodiments, GFP containing stop codons instead of lacZ alpha with stop codons is used for the construction. Cells with reverted stop codons in GFP are selected by fluorescence activated cell sorter (FACS). In general, FACS selection is performed by gating the brightest about 0.1-10%, preferably the top 0.1 to 1%, and collected according to a protocol similar to that of Dangl et al., ([0289] Cytometry 2(6):395-401 (1982)). In other embodiments, the polA gene is flanked with lox sites or other targets of a site specific recombinase. The recombinase is induced, thus allowing one to inducibly delete the polA gene (Mulbery et al., Nucleic Acid Res. 23:485-490 (1995)) This would allow one to perform “Loeb-type” selections at any temperature and in any host. For example, one could set up such a selection in a recA deficient mesophile or thermophile by placing the polA homologue in an inducibly deletable format and thus apply the selection for active polymerase under more general conditions.
  • In further embodiments, this general system is preferred for directed in vivo mutagenesis of genes. The target gene is cloned into the region near a plasmid origin of replication that puts its replication obligately under control of the error prone polymerase. The construct is passaged through a polA(ts) recA strain and grown at the nonpermissive temperature, thus specifically mutagenizing the target gene while replicating the rest of the plasmid with high fidelity. [0290]
  • In other embodiments, selection is based on the ability of mutant DNA polymerases to PCR amplify DNA under altered conditions or by utilizing base analogs. The mutant polymerases act on the template that encodes them in a PCR amplification, thus differentially replicating those polymerases. [0291]
  • In brief, an initial library of mutants is replica plated. Polymerase preparations are done in a 96-well format. Crude plasmid preparations are made of the same set. Each plasmid prep is PCR-amplified using the polymerase prep derived from that plasmid under the conditions for which one wishes to optimize the polymerase (e.g., added DMSO or formamide, altered temperature of denaturation or extension, altered buffer salts, PCR with base analogs such a-thiol dNTP's for use with mass spectroscopy sequencing, PCR of GC rich DNA (>60% GC),PCR with novel base analogs such as 7-deaza purines, 2′ fluoro dNTP's, rNTP's, PCR with inosine, etc.). The amplified genes are pooled, cloned,and subjected to mutagenesis, and the process repeated until an improvement is achieved. [0292]
  • C. Evolved Phosphonatase [0293]
  • Alkaline phosphatase is a widely used reporter enzyme for ELISA assays, protein fusion assays, and in a secreted form as a reporter gene for mammalian cells. The chemical lability of p-nitrophenyl phosphate (pNPP) substrates and the existence of cellular phosphatases that cross-react with pNPP is an important limitation on the sensitivity of assays using this reporter gene. A reporter gene with superior signal to noise properties can be developed based on hydrolysis of p-nitrophenyl phosphonates, which are far more stable to base catalyzed hydrolysis than the corresponding phosphates. Additionally, there are far fewer naturally occurring cellular phosphonatases than alkaline phosphatases. Thus a p-nitrophenyl phosphonatase is an attractive replacement for alkaline phosphatase because the background due to chemical and enzymatic hydrolysis is much lower. This will allow one to make ELISA's more sensitive for detecting very small concentrations of antigen. [0294]
  • Chen et al. ([0295] J. Mol. Biol. 234:165-178 (1993)) have shown that a Staph. aureus beta-lactamase can hydrolyze p-nitrophenyl phosphonate esters with single turnover kinetics. The active site Ser70 (the active site nucleophile for beta lactam hydrolysis) forms a covalent intermediate with the substrate. This is analogous to the first step in hydrolysis of beta lactams, and this enzyme can be evolved by RSR to hydrolyze phosphonates by a mechanism analogous to beta lactam hydrolysis. Metcalf and Wanner have described a cryptic phosphonate utilizing operon (phn) in E. coli, and have constructed strains bearing deletions of the phn operon (J. Bact. 175:3430-3442 (1993)). This paper discloses selections for growth of E. coli on phosphate free minimal media where the phosphorous is derived from hydrolysis of alkyl phosphonates by genes in the phn operon. Thus, one could select for evolved p-nitrophenyl phosphonatases that are active using biochemical selections on defined minimal media. Specifically, an efficient phosphonatase is evolved as follows. A library of mutants of the Staph. aureus beta lactamase or of one of the E. coli phn enzymes is constructed. The library is transformed into E. coli mutants wherein the phn operon has been deleted, and selected for growth on phosphate free MOPS minimal media containing p-nitrophenyl phosphonate. RSR is applied to selected mutants to further evolve the enzyme for improved hydrolysis of p-nitrophenyl phosphonates.
  • D. Evolved Detergent Proteases [0296]
  • Proteases and lipases are added in large quantities to detergents to enzymatically degrade protein and lipid stains on clothes. The incorporation of these enzymes into detergents has significantly reduced the need for surfactants in detergents with a consequent reduction in the cost of formulation of detergents and improvement in stain removal properties. Proteases with improved specific activity, improved range of protein substrate specificity, improved shelf life, improved stability at elevated temperature, and reduced requirements for surfactants would add value to these products. [0297]
  • As an example, subtilisin can be evolved as follows. The cloned subtilisin gene (von der Osten et al., [0298] J. Biotechnol. 28:55-68 (1993)) can be subjected to RSR using growth selections on complex protein media by virtue of secreted subtilisin degrading the complex protein mixture. More specifically, libraries of subtilisin mutants are constructed in an expression vector which directs the mutant protein to be secreted by Bacillus subtilus. Bacillus hosts transformed with the libraries are grown in minimal media with complex protein formulation as carbon and/or nitrogen source. Subtilisin genes are recovered from fast growers and subjected to RSR, then screened for improvement in a desired property.
  • E. Escape of Phage from a “Protein Net”[0299]
  • In some embodiments, selection for improved proteases is performed as follows. A library of mutant protease genes is constructed on a display phage and the phage grown in a multiwell format or on plates. The phage are overlayed with a “protein net” which ensnares the phage. The net can consist of a protein or proteins engineered with surface disulphides and then crosslinked with a library of peptide linkers. A further embodiment employs an auxiliary matrix to further trap the phage. The phage are further incubated, then washed to collect liberated phage wherein the displayed protease was able to liberate the phage from the protein net. The protease genes are then subjected to RSR for further evolution. A further embodiment employs a library of proteases encoded by but not displayed on a phagemid wherein streptavidin is fused to pIII by a peptide linker. The library of protease mutants is evolved to cleave the linker by selecting phagemids on a biotin column between rounds of amplification. [0300]
  • In a further embodiment, the protease is not necessarily provided in a display format. The host cells secrete the protease encoded by but not surface diplayed by a phagemid, while constrained to a well, for example, in a microtiter plate. Phage display format is preferred where an entire high titre lysate is encased in a protein net matrix, and the phage expressing active and broad specificity proteases digesting the matrix to be liberated for the next round of amplification, mutagenesis, and selection. [0301]
  • In a further embodiment, the phage are not constrained to a well but, rather, protein binding filters are used to make a colony of plaque lifts and are screened for activity with chromogenic or fluorogenic substrates. Colonies or plaques corresponding to positive spots on the filters are picked and the encoded protease genes are recovered by, for example, PCR. The protease genes are then subjected to RSR for further evolution. [0302]
  • F. Screens for Improved Protease Activity [0303]
  • Peptide substrates containing fluoropores attached to the carboxy terminus and fluorescence quenching moities on the amino terminus, such as those described by Holskin, et al, ([0304] Anal. Biochem. 227:148-55 (1995)) (e.g., (4-4′-dimethylaminophenazo)benzoyl-arg-gly-val-val-asn-ala-ser-ser-arg-leu-ala-5-(2′-aminoethyl)-amino]-naphthalene-1-sulfonic acid) are used to screen protease mutants for broadened or altered specificity. In brief, a library of peptide substrates is designed with a flourophore on the amino terminus and a potent fluorescence quencher on the carboxy terminus, or vice versa. Supernatants containing secreted proteases are incubated either separately with various members of the library or with a complex cocktail. Those proteases which are highly active and have broad specificity will cleave the majority of the peptides, thus releasing the fluorophore from the quencher and giving a positive signal on a fluorimeter. This technique is amenable to a high density multiwell format.
  • G. Improving Pharmaceutical Proteins Using RSR [0305]
  • Table I lists proteins that are of particular commercial interest to the pharmaceutical industry. These proteins are all candidates for RSR evolution to improve function, such as ligand binding, shelf life, reduction of side effects through enhanced specificity, etc. All are well-suited to manipulation by the techniques of the invention. Additional embodiments especially applicable to this list are described below. [0306]
  • First, high throughput methods for expressing and purifying libraries of mutant proteins, similar to the methods described above for Taq polymerase, are applied to the proteins of Table I. These mutants are screened for activity in a functional assay. For example, mutants of IL2 are screened for resistance to plasma or tissue proteases with retention of activity for the low affinity IL2 receptor but with loss of activity on the high affinity IL2 receptor. The genes from mutants with improved activity relative to wild-type are recovered, and subjected to RSR to improve the phenotype further. [0307]
  • Preferably, the libraries are generated in a display format such that the mature folded protein is physically linked to the genetic information that encodes it. Examples include phage display using filamentous phage (O'Neil et al., [0308] Current Biology 5:443-449 (1995)) or bacteriophage lambda gene V display (Dunn, J. Mol. Biol. 248:497-506 (1995)), peptides on plasmids (Gates et al., J. Mol. Biol. 255:373-386 (1995)) where the polypeptide of interest is fused to a lac headpiece dimer and the nascent translation product binds to a lac operator site encoded on the plasmid or PCR product, and polysome display (Mattheakis et al., Proc. Natl. Acad. Sci. (U.S.A.) 91:9022-9026 (1994)) where ribosomes are stalled on mRNA molecules such that the nascent polypeptide is exposed for interaction with cognate ligands without disrupting the stalled ribosome/mRNA complex. Selected complexes are subjected to RT-PCR to recover the genes.
  • When so displayed, affinity binding of the recombinant phage is often done using a receptor for the protein of interest. In some cases it is impractical to obtain purified receptor with retention of all desired biological characteristics (for example, 7-transmembrane (7-TM) receptors). In such cases, one could use cells expressing the receptor as the panning substrate. For example, Barry et al. ([0309] Nat. Med. 2:299-305 (1996)) have described successful panning of M13 libraries against whole cells to obtain phage that bind to the cells expressing a receptor of interest. This format could be generally applied to any of the proteins listed in Table I.
  • In some embodiments, the following method can be used for selection. A lysate of phage encoding IFN alpha mutants, for example, can be used directly at suitable dilution to stimulate cells with a GFP reporter construct (Crameri et al., [0310] Nat. Med. 14:315-319 (1996)) under the control of an IFN responsive promoter, such as an MHC class I promoter. Phage remaining attached after stimulation, expression and FACS purification of the responsive cells, can be purified by FACS. Preferably, the brightest cells are collected. The phage are collected and their DNA subjected to RSR until the level of desired improvement is achieved.
  • Thus, for example, IL-3 is prepared in one of these display formats and subjected to RSR to evolve an agonist with a desired level of activity. A library of IL3 mutants on a filamentous phage vector is created and affinity selected (“panned”) against purified IL3 receptor to obtain mutants with improved affinity. The mutant IL-3 genes are recovered by PCR, subjected to RSR, and recloned into the display vector. The cycle is repeated until the desired affinity or agonist activity is achieved. [0311]
  • Many proteins of interest are expressed as dimers or higher order multimeric forms. In some embodiments, the display formats descibed above preferentially are applied to a single chain version of the protein. Mutagenesis, such as RSR, can be used in these display formats to evolve improved single chain derivatives of multimeric factors which initially have low but detectable activity. This strategy is described in more detail below. [0312]
  • H. Whole Cell Selections [0313]
  • In some embodiments, the eukaryotic cell is the unit of biological selection. The following general protocol can be used to apply RSR to the improvement of proteins using eukaryotic cells as the unit of selection: (1) transfection of libraries of mutants into a suitable host cell, (2) expression of the encoded gene product(s) either transiently or stably, (3) functional selection for cells with an improved phenotype (expression of a receptor with improved affinity for a target ligand; viral resistance, etc., (4) recovery of the mutant genes by, for example, PCR followed by preparation of HIRT supernatants with subsequent tranformation of [0314] E. coli, (5) RSR and (6) repetition of steps (1)-(5) until the desired degree of improvement is achieved.
  • For example, previous work has shown that one can use mammalian surface display to functionally select cells expressing cloned genes, such as using an antibody to clone the gene for an expressed surface protein (Reviewed by Seed, [0315] Curr. Opin. Biotechnol. 6:567-573 (1995)). Briefly, cells are transiently transfected with libraries of cloned genes residing on replicating episomal vectors. An antibody directed against the protein of interest (whose gene one wishes to clone) is immobilized on a solid surface such as a plastic dish, and the transfected cells expressing the protein of interest are affinity selected.
  • For example, the affinity of an antibody for a ligand can be improved using mammalian surface display and RSR. Antibodies with higher affinity for their cognate ligands are then screened for improvement of one or more of the following properties: (1) improved therapeutic properties (increased cell killing, neutralization of ligands, activation of signal transduction pathways by crosslinking receptors), (2) improved in vivo imaging applications (detection of the antibody by covalent/noncovalent binding of a radionuclide or any agent detectable outside of the body by noninvasive means, such as NMR), (3) improved analytical applications (ELISA detection of proteins or small molecules), and (4) improved catalysts (catalytic antibodies). The methods described are general and can be extended to any receptor-ligand pair of interest. A specific example is provided in the experimental section. [0316]
  • The use of a one mutant sequence-one transfected cell protocol is a preferred design feature for RSR based protocols because the point is to use functional selection to identify mutants with improved phenotypes and, if the transfection is not done in a “clonal” fashion, the functional phenotype of any given cell is the result of the sum of many transfected sequences. Protoplast fusion is one method to achieve this end, since each protoplast contains typically greater than 50 copies each of a single plasmid variant. However, it is a relatively low efficiency process (about 10[0317] 3-104 transfectants), and it does not work well on some non-adherent cell lines such as B cell lines. Retroviral vectors provide a second alternative, but they are limited in the size of acceptable insert (<10 kb) and consistent, high expression levels are sometimes difficult to achieve. Random integration results in varying expression levels, thus introducing noise and limiting one's ability to distinguish between improvements in the affinity of the mutant protein vs. increased expression. A related class of strategies that can be used effectively to achieve “one gene-one cell” DNA transfer and consistent expression levels for RSR is to use a viral vector which contains a lox site and to introduce this into a host that expresses cre recombinase, preferably transiently, and contains one or more lox sites integrated into its genome, thus limiting the variability of integration sites (Rohlman et al. Nature Biotech. 14:1562-1565 (1996)).
  • An alternative strategy is to transfect with limiting concentrations of plasmid (i.e., about one copy per cell) using a vector that can replicate in the target cells, such as is the case with plasmids bearing SV40 origins transfected into COS cells. This strategy requires that either the host cell or the vector supply a replication factor such as SV40 large T antigen. Northrup et al. ([0318] J. Biol. Chem. 268:2917-2923 (1993)) describe a strategy wherein a stable transfectant expressing SV40 large T antigen is then transfected with vectors bearing SV40 origins. This format gave consistently higher transient expression and demonstrable plasmid replication, as assayed by sensitivity to digestion by Dpn I. Transient expression (i.e, non-integrating plasmids) is a preferred format for cellular display selections because it reduces the cycle time and increases the number of mutants that can be screened.
  • The expression of SV40 large T antigen or other replication factors may have deleterious effects on or may work inefficiently in some cells. In such cases, RSR is applied to the replication factor itself to evolve mutants with improved activity in the cell type of interest. A generic protocol for evolving such a factor is as follows: [0319]
  • The target cell is transfected with GFP cloned onto a vector containing SV40 large T antigen, an SV40 origin, and a reporter gene such as GFP; a related format is cotransfection with limiting amounts of the SV40 large T antigen expression vector and an excess of a reporter such as GFP cloned onto an SV40 origin containing plasmid. Typically after 1-10 days of transient expression, the brightest cells are purified by FACS. SV40 large T antigen mutants are recovered by PCR, and subjected to mutagenesis. The cycle is repeated until the desired level of improvement is obtained. [0320]
  • I. Autocrine Selection [0321]
  • In some embodiments, mutant proteins are selected or screened based on their ability to exert a biological effect in an autocrine fashion on the cell expressing the mutant protein. For example, a library of alpha interferon genes can be selected for induction of more potent or more specific antiviral activity as follows. A library of interferon alpha mutants is generated in a vector which allows for induction of expression (i.e. under control of a metallothionein promoter) and efficient secretion in a multiwell format (96-well for example) with one or a few independent clones per well. In some embodiments, the promoter is not inducible,and may be constitutive. [0322]
  • Expression of the cloned interferon genes is induced. The cells are challenged with a cytotoxic virus against which one wishes to evolve an optimized interferon (for example vesicular stomatitus virus or HIV). Surviving cells are recovered. The cloned interferon genes are recovered by PCR amplification, subjected to RSR, and cloned back into the transfection vector and retransfected into the host cells. These steps are repeated until the desired level of antiviral activity is evolved. [0323]
  • In some embodiments, the virus of interest is not strongly cytotoxic. In this case a conditionally lethal gene, such as herpes simplex virus thymidine kinase, is cloned into the virus and after challenge with virus and recovery, conditionally lethal selective conditions are applied to kill cells that are infected with virus. An example of a conditionally lethal gene is herpes TK, which becomes lethal upon treating cells expressing this gene with the thymidine analog acyclovir. In some embodiments, the antiproliferative activity of the cloned interferons is selected by treating the cells with agents that kill dividing cells (for example, DNA alkylating agents). [0324]
  • In some embodiments, potent cytokines are selected by expressing and secreting a library of cytokines in cells that have GFP or another reporter under control of a promoter that is induced by the cytokine, such as the MHC class I promoter being induced by evolved variants of alpha interferon. The signal transduction pathway is configured such that the wild type cytokine to be evolved gives a weak but detectable signal. [0325]
  • J. Half Life in Serum [0326]
  • In some embodiments of the invention, proteins are evolved by RSR to have improved half life in serum. A preferred method for improving half-life is evolving the affinity of a protein of interest for a long lived serum protein, such as an antibody or other abundant serum protein. Examples of how affinity for an antibody can enhance serum half life include the co-administration of IL2 and anti-IL2 antibodies which increases serum half-life and anti-tumor activity of human recombinant IL2 (Courtney et al., [0327] Immunopharmacology 28:223-232 (1994)).
  • The eight most abundant human serum proteins are serum albumin, immunoglobulins, lipoproteins, haptoglobin, fibrinogen, transferrin, alpha-1 antitrypsin, and alpha-2 macroglobulin (Doolittle, [0328] chapter 6, The Plasma Proteins F. Putnam, ed.; Academic Press, 1984). These and other abundant serum proteins such as ceruloplasmin and fibronectin are the primary targets against which to evolve binding sites on therapeutic proteins such as in Table I for the purpose of extending half-life. In the case of antibodies, the preferred strategy is to evolve affinity for constant regions rather than variable regions in order to minimize individual variation in the concentration of the relevant target epitope (antibody V region usage between different individuals is significantly variable).
  • Binding sites of the desired affinity are evolved by applying phage display, peptides on plasmid display or polysome display selections to the protein of interest. One could either mutagenize an existing binding site or otherwise defined region of the target protein, or append a peptide library to the N terminus, C terminus, or internally as a functionally nondisruptive loop. [0329]
  • In other embodiments of the invention, half life is improved by derivatization with PEG, other polymer conjugates or half-life extending chemical moieties. These are established methods for extending half-life of therapeutic proteins (R. Duncan, [0330] Clin. Pharmacokinet 27:290-306 (1994); Smith et al., TIBTECH 11 397-403 (1993)) and can have the added benefit of reducing immunogenicity (R. Duncan, Clin. Pharmacokinet 27:290-306 (1994)). However, derivatization can also result in reduced affinity of the therapeutic protein for its receptor or ligand. RSR is used to discover alternative sites in the primary sequence that can be substituted with lysine or other appropriate residues for chemical or enzymatic conjugation with half-life extending chemical moieties, and which result in proteins with maximal retention of biological activity.
  • A preferred strategy is to express a library of mutants of the protein in a display format, derivatize the library with the agent of interest (i.e. PEG) using chemistry that does not biologically inactivate the display system, select based on affinity for the cognate receptor, PCR amplify the genes encoding the selected mutants, shuffle, reassemble, reclone into the display format, and iterate until a mutant with the desired activity post modification is obtained. An alternative format is to express, purify and derivatize the mutants in a high throughput format, screen for mutants with optimized activity, recover the corresponding genes, subject the genes to RSR and repeat. [0331]
  • In further embodiments of the invention, binding sites for target human proteins that are localized in particular tissues of interest are evolved by RSR. For example, an interferon that localizes efficiently to the liver can be engineered to contain a binding site for a liver surface protein such as hepatocyte growth factor receptor. Analogously, one could evolve affinity for abundant epitopes on erythrocytes such as ABO blood antigens to localize a given protein to the blood stream. [0332]
  • In further embodiments of the invention, the protein of interest is evolved to have increased stability to proteases. For example, the clinical use of IL2 is limited by serious side effects that are related to the need to administer high doses. High doses are required due to the short half life (3-5 min, Lotze et al., [0333] JAMA 256(22):3117-3124 (1986)) and the consequent need for high doses to maintain a therapeutic level of IL2. One of the factors contributing to short half-lives of therapeutic proteins is proteolysis by serum proteases. Cathepsin D, a major renal acid protease, is responsible for the degradation of IL2 in Balb/c mice (Ohnishi et al., Cancer Res. 50:1107-1112 (1990)). Furthermore, Ohnishi showed that treatment of Balb/c mice with pepstatin, a potent inhibitor of this protease, prolongs the half life of recombinant human IL2 and augments lymphokine-activated killer cell activity in this mouse model.
  • Thus, evolution of protease resistant variants of IL2 or any of the proteins listed in Table I that are resistant to serum or kidney proteases is a preferred strategy for obtaining variants with extended serum half lives. [0334]
  • A preferred protocol is as follows. A library of the mutagenized protein of interest is expressed in a display system with a gene-distal epitope tag (i.e. on the N-terminus of a phage display construct such that if it is cleaved off by proteases, the epitope tag is lost). The expressed proteins are treated with defined proteases or with complex cocktails such as whole human serum. Affinity selection with an antibody to the gene distal tag is performed. A second selection demanding biological function (e.g., binding to cognate receptor) is performed. Phage retaining the epitope tag (and hence protease resistant) are recovered and subjected to RSR. The process is repeated until the desired level of resistance is attained. [0335]
  • In other embodiments, the procedure is performed in a screening format wherein mutant proteins are expressed and purified in a high throughput format and screened for protease resistance with retention of biological activity. [0336]
  • In further embodiments of the invention, the protein of interest is evolved to have increased shelf life. A library of the mutagenized nucleic acid squence encoding the protein of interest is expressed in a display format or high throughput expression format, and exposed for various lengths of time to conditions for which one wants to evolve stability (heat, metal ions, nonphysiological pH of, for example, <6 or >8, lyophilization, freeze-thawing). Genes are recovered from from survivors, for example, by PCR. The DNA is subjected to mutagenesis, such as RSR, and the process repeated until the desired level of improvement is achieved. [0337]
  • K. Evolved Single Chain Versions of Multisubunit Factors [0338]
  • As discussed above, in some embodiments of the invention, the substrate for evolution by RSR is preferably a single chain contruction. The possibility of performing asymetric mutagenesis on constructs of homomultimeric proteins provides important new pathways for further evolution of such constructs that is not open to the proteins in their natural homomultimeric states. In particular, a given mutation in a homomultimer will result in that change being present in each identical subunit. In single chain constructs, however, the domains can mutate independently of each other. [0339]
  • Conversion of multisubunit proteins to single chain constructs with new and useful properties has been demonstrated for a number of proteins. Most notably, antibody heavy and light chain variable domains have been linked into single chain Fv's (Bird et al., [0340] Science 242:423-426 (1988)), and this strategy has resulted in antibodies with improved thermal stability (Young et al., FEBS Lett 377:135-139 (1995)), or sensitivity to proteolysis (Solar et al., Prot. Eng. 8:717-723 (1995)). A functional single chain version of IL5, a homodimer, has been constructed, shown to have affinity for the IL5 receptor similar to that of wild type protein, and this construct has been used to perform assymetric mutagenesis of the dimer (Li et al., J. Biol. Chem. 271:1817-1820 (1996)). A single chain version of urokinase-type plasminogen activator has been made, and it has been shown that the single chain construct is more resistant to plasminogen activator inhibitor type 1 than the native homodimer (Higazi et al., Blood 87:3545-3549 (1996)). Finally, a single-chain insulin-like growth factor I/insulin hybrid has been constructed and shown to have higher affinity for chimeric insulin/IGF-1 receptors than that of either natural ligand (Kristensen et al., Biochem. J. 305:981-986 (1995)).
  • In general, a linker is constructed which joins the amino terminus of one subunit of a protein of interest to the carboxyl terminus of another subunit in the complex. These fusion proteins can consist of linked versions of homodimers, homomultimers, heterodimers or higher order heteromultimers. In the simplest case, one adds polypeptide linkers between the native termini to be joined. Two significant variations can be made. First, one can construct diverse libraries of variations of the wild type sequence in and around the junctions and in the linkers to facilitate the construction of active fusion proteins. Secondly, Zhang et al., ([0341] Biochemistry 32:12311-12318 (1993)) have described circular permutations of T4 lysozyme in which the native amino and carboxyl termini have been joined and novel amino and carboxyl termini have been engineered into the protein. The methods of circular permutation, libraries of linkers, and libraries of junctional sequences flanking the linkers allow one to construct libraries that are diverse in topological linkage strategies and in primary sequence. These libraries are expressed and selected for activity. Any of the above mentioned strategies for screening or selection can be used, with phage display being preferable in most cases. Genes encoding active fusion proteins are recovered, mutagenized, reselected, and subjected to standard RSR protocols to optimize their function. Preferably, a population of selected mutant single chain constructs is PCR amplified in two seprate PCR reactions such that each of the two domains is amplified separately. Oligonucleotides are derived from the 5′ and 3′ ends of the gene and from both strands of the linker. The separately amplified domains are shuffled in separate reactions, then the two populations are recombined using PCR reassembly to generate intact single chain constructs for further rounds of selection and evolution.
  • V. Improved Properties of Pharmaceutical Proteins [0342]
  • A. Evolved Specificity for Receptor or Cell Type of Interest [0343]
  • The majority of the proteins listed in Table I are either receptors or ligands of pharmaceutical interest. Many agonists such as chemokines or interleukins agonize more than one receptor. Evolved mutants with improved specificity may have reduced side effects due to their loss of activity on receptors which are implicated in a particular side effect profile. For most of these ligand/receptors, mutant forms with improved affinity would have improved pharmaceutical properties. For example, an antagonistic form of RANTES with improved affinity for CKR5 should be an improved inhibitor of HIV infection by virtue of achieving greater receptor occupancy for a given dose of the drug. Using the selections and screens outlined above in combination with RSR, the affinities and specificities of any of the proteins listed in Table I can be improved. For example, the mammalian display format could be used to evolve TNF receptors with improved affinity for TNF. [0344]
  • Other examples include evolved interferon alpha variants that arrest tumor cell proliferation but do not stimulate NK cells, IL2 variants that stimulate the low affinity IL2 receptor complex but not the high affinity receptor (or vice versa), superantigens that stimulate only a subset of the V beta proteins recognized by the wild type protein (preferably a single V beta), antagonistic forms of chemokines that specifically antagonize only a receptor of interest, antibodies with reduced cross-reactivity, and chimeric factors that specifically activate a particular receptor complex. As an example of this latter case, one could make chimeras between IL2 and IL4, 7, 9, or 15 that also can bind the IL2 receptor alpha, beta and gamma chains (Theze et al., [0345] Imm. Today 17:481-486 (1996)), and select for chimeras that retain binding for the intermediate affinity IL2 receptor complex on monocytes but have reduced affinity for the high affinity IL2 alpha, beta, gamma receptor complex on activated T cells.
  • B. Evolved Agonists with Increased Potency [0346]
  • In some embodiments of the invention, a preferred strategy is the selection or screening for mutants with increased agonist activity using the whole cell formats described above, combined with RSR. For example, a library of mutants of IL3 is expressed in active form on phage as described by Gram et al. ([0347] J. Immun. Meth. 161:169-176 (1993)). Clonal lysates resulting from infection with plaque-purified phage are prepared in a high through-put format such as a 96-well microtiter format. An IL3-dependent cell line expressing a reporter gene such as GFP is stimulated with the phage lysates in a high throughput 96-well. Phage that result in positive signals at the greatest dilution of phage supernatants are recovered; alternatively, DNA encoding the mutant IL3 can be recovered by PCR. In some embodiments, single cells expressing GFP under control of an IL3 responsive promoter can stimulated with the IL3 phage library, and the positive FACS sorted. The nucleic acid is then subjected to PCR, and the process repeated until the desired level of improvement is obtained.
  • TABLE I POLYPEPTIDE CANDIDATES FOR EVOLUTION
  • Name [0348]
  • Alpha-1 antitrypsin [0349]
  • Angiostatin [0350]
  • Antihemolytic factor [0351]
  • Apolipoprotein [0352]
  • Apoprotein [0353]
  • Atrial natriuretic factor [0354]
  • Atrial natriuretic polypeptide [0355]
  • Atrial peptides [0356]
  • C-X-C chemokines (e.g., T39765, NAP-2, ENA-78, Gro-a, Gro-b, Gro-c, IP-10, GCP-2, NAP-4, SDF-1, PF4, MIG) [0357]
  • Calcitonin [0358]
  • CC chemokines (e.g., Monocyte chemoattractant protein-1, Monocyte chemoattractant protein-2, Monocyte chemoattractant protein-3, Monocyte inflammatory protein-1 alpha, Monocyte inflammatory protein-1 beta, RANTES, I309, R83915, R91733, HCC1, T58847, D31065, T64262) [0359]
  • CD40 ligand [0360]
  • Collagen [0361]
  • Colony stimulating factor (CSF) [0362]
  • Complement factor Sa [0363]
  • Complement inhibitor [0364]
  • [0365] Complement receptor 1
  • Factor IX [0366]
  • Factor VII [0367]
  • Factor VIII [0368]
  • Factor X [0369]
  • Fibrinogen [0370]
  • Fibronectin [0371]
  • Glucocerebrosidase [0372]
  • Gonadotropin [0373]
  • Hedgehog proteins (e.g., Sonic, Indian, Desert) [0374]
  • Hemoglobin (for blood substitute; for radiosensitization) [0375]
  • Hirudin [0376]
  • Human serum albumin [0377]
  • Lactoferrin [0378]
  • Luciferase [0379]
  • Neurturin [0380]
  • Neutrophil inhibitory factor (NIF) [0381]
  • Osteogenic protein [0382]
  • Parathyroid hormone [0383]
  • Protein A [0384]
  • Protein G [0385]
  • Relaxin [0386]
  • Renin [0387]
  • Salmon calcitonin [0388]
  • Salmon growth hormone [0389]
  • Soluble complement receptor I [0390]
  • Soluble I-[0391] CAM 1
  • Soluble interleukin receptors (IL-1, 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15) [0392]
  • Soluble TNF receptor [0393]
  • Somatomedin [0394]
  • Somatostatin [0395]
  • Somatotropin [0396]
  • Streptokinase [0397]
  • Superantigens, i.e., Staphylococcal enterotoxins (SEA, SEB, SEC1, SEC2, SEC3, SED, SEE), Toxic shock syndrome toxin (TSST-1), Exfoliating toxins A and B, Pyrogenic exotoxins A, B, and C, and M. arthritidis mitogen [0398]
  • Superoxide dismutase [0399]
  • [0400] Thymosin alpha 1
  • Tissue plasminogen activator [0401]
  • Tumor necrosis factor beta (TNF beta) [0402]
  • Tumor necrosis factor receptor (TNFR) [0403]
  • Tumor necrosis factor-alpha (TNF alpha) [0404]
  • Urokinase [0405]
  • C. Evolution of Components of Eukaryotic Signal Transduction or Transcriptional Pathways [0406]
  • Using the screens and selections listed above, RSR can be used in several ways to modify eukaryotic signal transduction or transcriptional pathways. Any component of a signal transduction pathway of interest, of the regulatory regions and transcriptional activators that interact with this region and with chemicals that induce transcription can be evolved. This generates regulatory systems in which transcription is activated more potently by the natural inducer or by analogues of the normal inducer. This technology is preferred for the development and optimization of diverse assays of biotechnological interest. For example, dozens of 7 transmembrane receptors (7-TM) are validated targets for drug discovery (see, for example, Siderovski et al., [0407] Curr Biol., 6(2):211-212 (1996); An et al., FEBS Lett., 375(1-2):121-124 (1995); Raport et al., Gene, 163(2):295-299 (1995); Song et al., Genomics, 28(2):347-349 (1995); Strader et al. FASEB J., 9(9) :745-754 (1995); Benka et al., FEBS Lett., 363(1-2):49-52 (1995); Spiegel, J. Clin Endocrinol. Metab., 81(7):2434-2442 (1996); Post et al., FASEB J., 10(7):741-749 (1996); Reisine et al., Ann NY Acad. Sci., 780:168-175 (1996); Spiegel, Annu. Ref. Physiol., 58:143-170 (1996); Barak et al., Biochemistry, 34(47):15407-15414 (1995); and Shenker, Baillieres Clin. Endocrinol. Metab., 9(3):427-451 (1995)). The development of sensitive high throughput assays for agonists and antagonists of these receptors is essential for exploiting the full potential of combinatorial chemistry in discovering such ligands. Additionally, biodetectors or biosensors for different chemicals can be developed by evolving 7-TM's to respond agonistically to novel chemicals or proteins of interest. In this case, selection would be for contructs that are activated by the new chemical or polypeptide to be detected. Screening could be done simply with fluorescence or light activated cell sorting, since the desired improvement is coupled to light production.
  • In addition to detection of small molecules such as pharmaceutical drugs and environmental pollutants, biosensors can be developed that will respond to any chemical for which there are receptors, or for which receptors can be evolved by recursive sequence recombination, such as hormones, growth factors, metals and drugs. The receptors may be intracellular and direct activators of transcription, or they may be membrane bound receptors that activate transcription of the signal indirectly, for example by a phosphorylation cascade. They may also not act on transcription at all, but may produce a signal by some post-transcriptional modification of a component of the signal generating pathway. These receptors may also be generated by fusing domains responsible for binding different ligands with different signalling domains. Again, recursive sequence recombination can be used to increase the amplitude of the signal generated to optimize expression and functioning of chimeric receptors, and to alter the specificity of the chemicals detected by the receptor. [0408]
  • For example, G proteins can be evolved to efficiently couple mammalian 7-TM receptors to yeast signal transduction pathways. There are 23 presently known G alpha protein loci in mammals which can be grouped by sequence and functional similarity into four groups, Gs (Gna, Gna1), Gi (Gnai-2, Gnai-3, Gnai-1, Gnao, Gnat-1, Gnat-2, Gnaz), Gq (Gnaq, Gna-11, Gna-14, Gna-15) and G12 (Gna-12, Gna-13) (B. Nurnberg et al., [0409] J. Mol. Med., 73:123-132 (1995)). They possess an endogenous GTP-ase activity allowing reversible functional coupling between ligand-bound receptors and downstream effectors such as enzymes and ion channels. G alpha proteins are complexed noncovalently with G beta and G gamma proteins as well as to their cognate 7-TM receptor(s). Receptor and signal specificity are controlled by the particular combination of G alpha, G beta (of which there are five known loci) and G gamma (seven known loci) subunits. Activation of the heterotrimeric complex by ligand bound receptor results in dissociation of the complex into G alpha monomers and G beta, gamma dimers which then transmit signals by associating with downstream effector proteins. The G alpha subunit is believed to be the subunit that contacts the 7-TM, and thus it is a focal point for the evolution of chimeric or evolved G alpha subunits that can transmit signals from mammalian 7-TM's to yeast downstream genes.
  • Yeast based bioassays for mammalian receptors will greatly facilitate the discovery of novel ligands. Kang et al. ([0410] Mol. Cell Biol. 10:2582-2590 (1990)) have described the partial complementation of yeast strains bearing mutations in SCG1 (GPA1), a homologue of the alpha subunits of G proteins involved in signal transduction in mammalian cells, by mammalian and hybrid yeast/mammalian G alpha proteins. These hybrids have partial function, such as complementing the growth defect in scg1 strains, but do not allow mating and hence do not fully complement function in the pheromone signal transduction pathway. Price et al. (Mol. Cell Biol. 15:6188-6195 (1995)) have expressed rat somatostatin receptor subtype 2 (SSTR2) in yeast and demonstrated transmission of ligand binding signals by this 7-TM receptor through yeast and chimeric mammalian/yeast G alpha subunits (“coupling”) to a HIS3 reporter gene, under control of the pheromone responsive promoter FUS-1 enabling otherwise HIS3(−) cells to grow on minimal medium lacking histidine.
  • Such strains are useful as reporter strains for mammalian receptors, but suffer from important limitations as exemplified by the study of Kang et al., where there appears to be a block in the transmission of signals from the yeast pheromone receptors to the mammalian G proteins. In general, to couple a mammalian 7-TM receptor to yeast signal transduction pathways one couples the mammalian receptor to yeast, mammalian, or chimeric G alpha proteins, and these will in turn productively interact with downstream components in the pathway to induce expression of a pheromone responsive promoter such as FUS-1. Such functional reconstitution is commonly referred to as “coupling”. [0411]
  • The methods described herein can be used to evolve the coupling of mammalian 7-TM receptors to yeast signal transduction pathways. A typical approach is as follows: (1) clone a 7-TM of interest into a yeast strain with a modified pheromone response pathway similar to that described by Price (e.g., strains deficient in FAR1, a negative regulator of G[0412] 1 cyclins, and deficient in SST2 which causes the cells to be hypersensitive to the presence of pheromone), (2) construct libraries of chimeras between the mammalian G alpha protein(s) known or thought to interact with the GPA1 or homologous yeast G alpha proteins, (3) place a selectable reporter gene such as HIS3 under control of the pheromone responsive promoter FUS1 (Price et al., Mol. Cell Biol. 15:6188-6195 (1995)). Alternatively, a screenable gene such as luciferase may be placed under the control of the FUS1 promoter; (4) transform library (2) into strain (3) (HIS(−)), (5) screen or select for expression of the reporter in response to the ligand of interest, for example by growing the library of transformants on minimal plates in the presence of ligand to demand HIS3 expression, (6) recover the selected cells, and and apply RSR to evolve improved expression of the reporter under the control of the pheromone responsive promoter FUS1.
  • A second important consideration in evolving strains with optimized reporter constructs for signal transduction pathways of interest is optimizing the signal to noise ratio (the ratio of gene expression under inducing vs noninducing conditions). Many 7-TM pathways are leaky such that the maximal induction of a typical reporter gene is 5 to 10-fold over background. This range of signal to noise may be insufficient to detect small effects in many high through put assays. Therefore, it is of interest to couple the 7-TM pathway to a second nonlinear amplification system that is tuned to be below but near the threshold of activation in the uninduced state. An example of a nonlinear amplification system is expression of genes driven by the lambda PL promoter. Complex cooperative interactions between lambda repressor bound at three adjacent sites in the cI promoter result in very efficient repression above a certain concentration of repressor. Below a critical threshold dramatic induction is seen and there is a window within which a small decrease in repressor concentration leads to a large increase in gene expression (Ptashne, [0413] A Genetic Switch:Phage Lambda and Higher Organisms, Blackwell Scientific Publ. Cambridge, Mass., 1992). Analogous effects are seen for some eukaryotic promoters such as those regulated by GAL4. Placing the expression of a limiting component of a transcription factor for such a promoter (GAL4) under the control of a GAL4 enhanced 7-TM responsive promoter results in small levels of induction of the 7-TM pathway signal being amplified to a much larger change in the expression of a reporter construct also under the control of a GAL4 dependent promoter.
  • An example of such a coupled system is to place GAL4 under control of the FUS-1 pheromone responsive promoter and to have the intracellular GAL4 (itself a transcriptional enhancer) level positively feedback on itself by placing a GAL4 binding site upstream of the FUS-1 promoter. A reporter gene is also put under the control of a GAL4 activated promoter. This system is designed so that GAL4 expression will nonlinearly self-amplify and co-amplify expression of a reporter gene such as luciferase upon reaching a certain threshold in the cell. RSR can be used to great advantage to evolve reporter constructs with the desired signaling properties, as follows: (1) A single plasmid construct is made which contains both the GAL4/pheromone pathway regulated GAL4 gene and the GAL4 regulated reporter gene. (2) This construct is mutagenized and transformed into the appropriately engineered yeast strain expressing a 7-TM and chimeric yeast/mammalian protein of interest. (3) Cells are stimulated with agonists and screened (or selected) based on the activity of the reporter gene. In a preferred format, luciferase is the reporter gene and activity is quantitated before and after stimulation with the agonist, thus allowing for a quantitative measurement of signal to noise for each colony. (4) Cells with improved reporter properties are recovered, the constructs are shuffled, and RSR is applied to further evolve the plasmid to give optimal signal noise characteristics. [0414]
  • These approaches are general and illustrate how any component of a signal transduction pathway or transcription factor could be evolved using RSR and the screens and selections described above. For example, these specific methods could be used to evolve 7-TM receptors with specificity for novel ligands, specificity of nuclear receptors for novel ligands (for example to obtain herbicide or other small molecule-inducible expression of genes of interest in transgenic plants, such that a given set of genes can be induced upon treatment with a given chemical agent), specificity of transcription factors to be responsive to viral factors (thus inducing antiviral or lethal genes in cells expressing this transcription factor [transgenics or cells treated with gene therapy constructs]), or specificity of transcription factors for activity in cancer cells (for example p53 deficient cells, thus allowing one to infect with gene therapy constructs expressing conditionally lethal genes in a tumor specific fashion). [0415]
  • The following examples are offered by way of illustration, not by way of limitation. [0416]
  • EXPERIMENTAL EXAMPLES
  • I. Evolution of BIAP [0417]
  • A preferred strategy to evolve BIAP is as follows. A codon usage libary is constructed from 60-mer oligonucleotides such that the central 20 bases of each oligo specifies the wild type protein, but encodes the wild-type protein sequence with degenerate codons. Preferably, very rare codons for the prokaryotic host of choice, such as [0418] E. coli, are not used. The 20 bases at each end of the oligo use non-degenerate, but preferred, codons in E. coli. The oligonucleotides are assembled into full-length genes as described above. The assembled products are cloned into an expression vector by techniques well known in the art. In some embodiments, the codon usage library is expressed with a library of secretory leader sequences, each of which directs the encoded BIAP protein to the E. coli periplasm. A library of leader sequences is used to optimize the combination of leader sequence and mutant. Examples of leader sequences are reviewed by Schatz et al. (Ann Rev. Genet. 24:215-248 (1990)). The cloned BIAP genes are expressed under the control of an inducible promoter such as the arabinose promoter. Arabinose-induced colonies are screened by spraying with a substrate for BIAP, bromo-chloro-indolyl phosphate (BCIP). The bluest colonies are picked visually and subjected to the RSR procedures described herein.
  • The oligonucleotides for construction of the codon usage library are listed in Table II. The corresponding locations of these promoters is provided in FIG. 1. [0419]
    TABLE II
    1. AACCCTCCAG TTCCGAACCC CATATGATGA TCACCCTGCG TAAACTGCCG
    2. AACCCTCCAG TTCCGAACCC CATATGAAAA AAACCGCT
    3. AACCCTCCAG TTCCGAACCC ATATACATAT GCGTGCTAAA
    4. AACCCTCCAG TTCCGAACCC CATATGAAAT ACCTGCTGCC GACC
    5. AACCCTCCAG TTCCGAACCC GATATACATA TGAAACAGTC
    6. TGGTGTTATG TCTGCTCAGG CDATGGCDGT DGAYTTYCAY CTGGTTCCGG
    TTGAAGAGGA
    7. GGCTGGTTTC GCTACCGTTG CDCARGCDGC DCCDAARGAY CTGGTTCCGG
    TTGAAGAGGA
    8. CACCCCGATC GCTATCTCTT CYTTYGCDTC YACYGGYTCY CTGGTTCCGG
    TTGAAGAGGA
    9. GCTGCTGGCT GCTCAGCCGG CDATGGCDAT GGAYATYGGY CTGGTTCCGG
    TTGAAGAGGA
    10. TGCCGCTGCT GTTCACCCCG GTDACYAARG CDGCDCARGT DCTGGTTCCG
    GTTGAAGAGG A
    11. CCCGGCTTTC TGGAACCGTC ARGCDGCDCA RGCDCTGGAC GTTGCTAAAA
    AACTGCAGCC
    12. ACGTTATCCT GTTCCTGGGT GAYGGYATGG GYGTDCCDAC CGTTACCGCT
    ACCCGTATCC
    13. AAACTGGGTC CGGAAACCCC DCTGGCDATG GAYCARTTYC CGTACGTTGC
    TCTGTCTAAA
    14. GGTTCCGGAC TCTGCTGGTA CYGCDACYGC DTAYCTGTGC GGTGTTAAAG
    GTAACTACCG
    15. CTGCTCGTTA CAACCAGTGC AARACYACYC GYGGYAAYGA AGTTACCTCT
    GTTATGAACC
    16. TCTGTTGGTG TTGTTACCAC YACYCGYGTD CARCAYGCDT CTCCGGCTGG
    TGCTTACGCT
    17. GTACTCTGAC GCTGACCTGC CDGCDGAYGC DCARATGAAC GGTTGCCAGG
    ACATCGCTGC
    18. ACATCGACGT TATCCTGGGT GGYGGYCGYA ARTAYATGTT CCCGGTTGGT
    ACCCCGGACC
    19. TCTGTTAACG GTGTTCGTAA RCGYAARCAR AAYCTGGTDC AGGCTTGGCA
    GGCTAAACAC
    20. GAACCGTACC GCTCTGCTGC ARGCDGCDGA YGAYTCYTCT GTTACCCACC
    TGATGGGTCT
    21. AATACAACGT TCAGCAGGAC CAYACYAARG AYCCDACYCT GCAGGAAATG
    ACCGAAGTTG
    22. AACCCGCGTG GTTTCTACCT GTTYGTDGAR GGYGGYCGYA TCGACCACGG
    TCACCACGAC
    23. GACCGAAGCT GGTATGTTCG AYAAYGCDAT YGCDAARGCT AACGAACTGA
    CCTCTGAACT
    24. CCGCTGACCA CTCTCACGTT TTYTCYTTYG GYGGYTAYAC CCTGCGTGGT
    ACCTCTATCT
    25. GCTCTGGACT CTAAATCTTA YACYTCYATY CTGTAYGGYA ACGGTCCGGG
    TTACGCTCTG
    26. CGTTAACGAC TCTACCTCTG ARGAYCCDTC YTAYCARCAG CAGGCTGCTG
    TTCCGCAGGC
    27. AAGACGTTGC TGTTTTCGCT CGYGGYCCDC ARGCDCAYCT GGTTCACGGT
    GTTGAAGAAG
    28. ATGGCTTTCG CTGGTTGCGT DGARCCDTAY ACYGAYTGYA ACCTGCCGGC
    TCCGACCACC
    29. TGCTCACCTG GCTGCTTMAC CDCCDCCDCT GGCDCTGCTG GCTGGTGCTA
    TGCTGCTCCT C
    30. TTCCGCCTCT AGAGAATTCT TARTACAGRG THGGHGCCAG GAGGAGCAGC
    ATAGCACCAG CC
    31. AAGCAGCCAG GTGAGCAGCG TCHGGRATRG ARGTHGCGGT GGTCGGAGCC
    GGCAGGTT
    32. CGCAACCAGC GAAAGCCATG ATRTGHGCHA CRAARGTYTC TTCTTCAACA
    CCGTGAACCA
    33. GCGAAAACAG CAACGTCTTC RCCRCCRTGR GTYTCRGAHG CCTGCGGAAC
    AGCAGCCTGC
    34. AGAGGTAGAG TCGTTAACGT CHGGRCGRGA RCCRCCRCCC AGAGCGTAAC
    CCGGACCGTT
    35. AAGATTTAGA GTCCAGAGCT TTRGAHGGHG CCAGRCCRAA GATAGAGGTA
    CCACGCAGGG
    36. ACGTGAGAGT GGTCAGCGGT HACCAGRATC AGRGTRTCCA GTTCAGAGGT
    CAGTTCGTTA
    37. GAACATACCA GCTTCGGTCA GHGCCATRTA HGCYTTRTCG TCGTGGTGAC
    CGTGGTCGAT
    38. GGTAGAAACC ACGCGGGTTA CGRGAHACHA CRCGCAGHGC AACTTCGGTC
    ATTTCCTGCA
    39. TCCTGCTGAA CGTTGTATTT CATRTCHGCH GGYTCRAACA GACCCATCAG
    GTGGGTAACA
    40. CAGCAGAGCG GTACGGTTCC AHACRTAYTG HGCRCCYTGG TGTTTAGCCT
    GCCAAGCCTG
    41. TACGAACACC GTTAACAGAA GCRTCRTCHG GRTAYTCHGG GTCCGGGGTA
    CCAACCGGGA
    42. CCCAGGATAA CGTCGATGTC CATRTTRTTH ACCAGYTGHG CAGCGATGTC
    CTGGCAACCG
    43. CAGGTCAGCG TCAGAGTACC ARTTRCGRTT HACRGTRTGA GCGTAAGCAC
    CAGCCGGAGA
    44. TGGTAACAAC ACCAACAGAT TTRCCHGCYT TYTTHGCRCG GTTCATAACA
    GAGGTAACTT
    45. CACTGGTTGT AACGAGCAGC HGCRGAHACR CCRATRGTRC GGTAGTTACC
    TTTAACACCG
    46. ACCAGCAGAG TCCGGAACCT GRCGRTCHAC RTTRTARGTT TTAGACAGAG
    CAACGTACGG
    47. GGGTTTCCGG ACCCAGTTTA CCRTTCATYT GRCCYTTCAG GATACGGGTA
    GCGGTAACGG
    48. CCCAGGAACA GGATAACGTT YTTHGCHGCR GTYTGRATHG GCTGCAGTTT
    TTTAGCAACG
    49. ACGGTTCCAG AAAGCCGGGT CTTCCTCTTC AACCGGAACC AG
    50. CCTGAGCAGA CATAACACCA GCHGCHACHG CHACHGCCAG CGGCAGTTTA
    CGCAGGGTGA
    51. ACCGGGGTGA ACAGCAGCGG CAGCAGHGCC AGHGCRATRG TRGACTGTTT
    CATATGTATA TC
    52. GCCGGCTGAG CAGCCAGCAG CAGCAGRCCH GCHGCHGCGG TCGGCAGCAG
    GTAGTTTCA
    53. AAGAGATAGC GATCGGGGTG GTCAGHACRA TRCCCAGCAG TTTAGCACGC
    ATATGTATAT
    54. CAACGGTAGC GAAACCAGCC AGHGCHACHG CRATHGCRAT AGCGGTTTTT
    TTCATATG
    55  AGAATTCTCT AGAGGCGGAA ACTCTCCAAC TCCCAGGTT
    56. TGAGAGGTTG AGGGTCCAAT TGGGAGGTCA AGGCTTGGG
  • II. Mammalian Surface Display [0420]
  • During an immune response antibodies naturally undergo a process of affinity maturation resulting in mutant antibodies with improved affinities for their cognate antigens. This process is driven by somatic hypermutation of antibody genes coupled with clonal selection (Berek and Milstein, [0421] Immun. Rev. 96:23-41 (1987)). Patten et al. (Science 271:1086-1091 (1996)) have reconstructed the progression of a catalytic antibody from the germline sequence, which binds a p-nitrophenylphosphonate hapten with an affinity of 135 micromolar, to the affinity matured sequence which has acquired nine somatic mutations and binds with an affinity of 10 nanomolar. The affinity maturation of this antibody can be recapitulated and improved upon using cassette mutagenesis of the CDR's (or random mutagenesis such as with PCR), mammalian display, FACS selection for improved binding, and RSR to rapidly evolve improved affinity by recombining mutations encoding improved binding.
  • Genomic antibody expression shuttle vectors similar to those described by Gascoigne et al. ([0422] Proc. Natl. Acad. Sci. (U.S.A.) 84:2936-2940 (1987)) are constructed such that libraries of mutant V region exons can be readily cloned into the shuttle vectors. The kappa construct is cloned onto a plasmid encoding puromycin resistance and the heavy chain is cloned onto a neomycin resistance encoding vector. The cDNA derived variable region sequences encoding the mature and germline heavy and light chain V regions are reconfigured by PCR mutagenesis into genomic exons flanked by Sfi I sites with complementary Sfi I sites placed at the appropriate locations in the genomic shuttle vectors. The oligonucleotides used to create the intronic Sfi I sites flanking the VDJ exon are: 5′ Sfi I: 5′-TTCCATTTCA TACATGGCCG AAGGGGCCGT GCCATGAGGA TTTT-3′; 3′ Sfi I: 5′-TTCTAAATG CATGTTGGCC TCCTTGGCCG GATTCTGAGC CTTCAGGACC A-3′. Standard PCR mutagenesis protocols are applied to produce libraries of mutants wherein the following sets of residues (numbered according to Kabat, Sequences of Proteins of Immunological Interest, U.S. Dept of Health and Human Services, 1991) are randomized to NNK codons (GATC,GATC,GC):
    Chain CDR Mutated residues
    V-L 1 30, 31, 34
    V-L 2 52, 53, 55
    V-H 2 55, 56, 65
    V-H “4” 74, 76, 78
  • Stable transfectant lines are made for each of the two light and heavy chain constructs (mature and germline) using the B cell myeloma AG8-653 (a gift from J. Kearney) as a host using standard electroporation protocols. Libraries of mutant plasmids encoding the indicated libraries of V-L mutants are transfected into the stable transformant expressing the germline V-H; and the V-H mutants are transfected into the germline V-L stable transfectant line. In both cases, the libraries are introduced by protoplast fusion (Sambrook et al., [0423] Molecular Cloning, CSH Press (1987)) to ensure that the majority of transfected cells receive one and only one mutant plasmid sequence (which would not be the case for electroporation where the majority of the transfected cells would receive many plasmids, each expressing a different mutant sequence).
  • The p-nitrophenylphosphonate hapten (JWJ-1) recognized by this antibody is synthesized as described by Patten et al. ([0424] Science 271:1086-1091 (1996)). JWJ-1 is coupled directly to 5-(((2-aminoethyl)thio)acetyl) fluorescein (Molecular Probes, Inc.) by formation of an amide bond using a standard coupling chemistry such as EDAC (March, Advanced Organic Chemistry, Third edition, John Wiley and Sons, 1985) to give a monomeric JWJ-1-FITC probe. A “dimeric” conjugate (two molecules of JWJ-1 coupled to a FACS marker) is made in order to get a higher avidity probe, thus making low affinity interactions (such as with the germline antibody) more readily detected by FACS. This is generated by staining with Texas Red conjugated to an anti-fluorescein antibody in the presence of two equivalents of JWJ-1-FITC. The bivalent structure of IgG then provides a homogeneous bivalent reagent. A spin column is used to remove excess JWJ-1-FITC molecules that are not bound to the anti-FITC reagent. A tetravalent reagent is made as follows. One equivalent of biotin is coupled with EDAC to two equivalents of ethylenediamine, and this is then be coupled to the free carboxylate on JWJ-1. The biotiylated JWJ-1 product is purified by ion exchange chromatography and characterized by mass spectrometry. FITC labelled avidin is incubated with the biotinylated JWJ-1 in order to generate a tetravalent probe.
  • The FACS selection is performed as follows, according to a protocol similar to that of Panka et al. ([0425] Proc. Natl. Acad. Sci. (U.S.A.) 85:3080-3084 (1988)). After transfection of libraries of mutant antibody genes by the method of protoplast fusion (with recovery for 36-72 hours), the cells are incubated on ice with fluorescently labelled hapten. The incubation is done on ice to minimize pinocytosis of the FITC conjugate which may contribute to nonspecific background. The cells are then sorted on the FACS either with or without a washing step. FACSing without a washing step is preferable because the off rate for the germline antibody prior to affinity maturation is expected to be very fast (>0.1 sec-1; Patten et al., Science 271:1086-1091 (1996)); a washing step adds a complicating variable. The brightest 0.1-10% of the cells are collected.
  • Four parameters are manipulated to optimize the selection for increased binding: monomeric vs dimeric vs tetrameric hapten, concentration of hapten used in the staining reaction (low concentration selects for high affinity Kd's), time between washing and FACS (longer time selects for low off rates), and selectivity in the gating (i.e. take the top 0.1% to 10%, more preferably the top 0.1%). The constructs expressing the germline, mature, and both combinations of half germline are used as controls to optimize this selectivity. [0426]
  • Plasmids are recovered from the FACS selected cells by the transformation of an [0427] E. coli host with Hirt supernatants. Alternatively, the mutant V gene exons are PCR-amplified from the FACS selected cells. The recovered V gene exons are subjected to RSR, recloned into the corresponding genomic shuttle vector, and the procedure recursively applied until the mean fluorescence intensity has increased. A relevant positive control for improved binding is transfection with the affinity matured 48G7 exons (Patten et al., op. cit.).
  • In a further experiment, equal numbers of germline and each of the two half germline transfectants are mixed. The brightest cells are selected under conditions described above. The V genes are recovered by PCR, recloned into expression vectors, and co-transfected, either two plasmids per [0428] E. coli followed by protoplast fusion, or by bulk electroporation. The mean fluorescent intensity of the transfectants should increase due to enrichment of mature relative to germline V regions.
  • This methodology can be applied to evolve any receptor-ligand or binding partner interaction. Natural expression formats can be used to express libraries of mutants of any receptor for which one wants to improve the affinity for the natural or novel ligands. Typical examples would be improvement of the affinity of T cell receptors for ligands of interest (i.e. MHC/tumor peptide antigen complexes) or TNF receptor for TNF (soluble forms of TNF receptors are used therapeutically to neutralize TNF activity). [0429]
  • This format can also be used to select for mutant forms of ligands by expressing the ligand in a membrane bound form with an engineered membrane anchor by a strategy analogous to that of Wettstein et al.([0430] J. Exp. Med. 174:219-28 (1991)). FACS selection is then performed with fluorescently labelled receptor. In this format one could, for example, evolve improved receptor antagonists from naturally occurring receptor antagonists (IL1 receptor antagonist, for example). Mutant forms of agonists with improved affinity for their cognate receptors could also be evolved in this format. These mutants would be candidates for improved agonists or potent receptor antagonists, analogous to reported antagonistic mutant forms of IL3.
  • III. Evolution of Alpha Interferon [0431]
  • There are at [0432] hand 18 known non-allelic human interferon-alpha (INF-α) genes, with highly related primary structures (78-95% identical) and with a broad range of biological activities. Many hybrid interferons with interesting biological activities differing from the parental molecules have been described (reviewed by Horisberger and Di Marco, Pharm. Ther. 66:507-534 (1995)). A consensus human alpha interferon, IFN-Con1, has been constructed synthetically wherein the most common residue in fourteen known IFN-α's has been put at each position, and it compares favorably with the naturally occurring interferons (Ozes et al., J. Interferon Res. 12:55-59 (1992)). This IFN contains.20 amino acid changes relative to IFN-α2a, the INF-α to which it is most closely related. IFN-Con1 has 10-fold higher specific antiviral activity than any known natural IFN subtype. IFN-α Con1 has in vitro activities 10 to 20 fold higher than that of recombinant IFN α-2a (the major IFN used clinically) in antiviral, antiproliferative and NK cell activation. Thus, there is considerable interest in producing interferon hybrids which combine the most desirable traits from two or more interferons. However, given the enormous number of potential hybrids and the lack of a crystal structure of IFN-α or of the IFN-α receptor, there is a perceived impasse in the development of novel hybrids (Horisberger and Di Marco, Pharm. Ther. 66:507-534 (1995)).
  • The biological effects of IFN-α's are diverse, and include such properties as induction of antiviral state (induction of factors that arrest translation and degrade mRNA); inhibition of cell growth; induction of Class I and Class II MHC; activation of monocytes and macrophages; activation of natural killer cells; activation of cytotoxic T cells; modulation of Ig synthesis in B cells; and pyrogenic activity. [0433]
  • The various IFN-α's subtypes have unique spectra of activities on different target cells and unique side effect profiles (Ortaldo et al., [0434] Proc. Natl. Acad. Sci. (U.S.A.) 81:4926-4929 (1984); Overall et al., J. Interferon Res. 12:281-288 (1992); Fish and Stebbing, Biochem. Biophys. Res. Comm. 112:537-546 (1983); Weck et al., J. Gen. Virol. 57:233-237 (1981)). For example, human IFNα has very mild side effects but low antiviral activity. Human IFNα8 has very high antiviral activity, but relatively severe side effects. Human IFNα7 lacks NK activity and blocks NK stimulation by other INFα's. Human IFN-α J lacks the ability to stimulate NK cells, but it can bind to the IFN-α receptor on NK cells and block the stimulatory activity of IFN-αA (Langer et al., J. Interferon Res. 6:97-105 (1986)).
  • The therapeutic applications of interferons are limited by diverse and severe side effect profiles which include flu-like symptoms, fatigue, neurological disorders including hallucination, fever, hepatic enzyme elevation, and leukopenia. The multiplicity of effects of IFN-α's has stimulated the hypothesis that there may be more than one receptor or a multicomponent receptor for the IFN-α family (R. Hu et al., [0435] J. Biol. Chem. 268:12591-12595 (1993)). Thus, the existence of abundant naturally occurring diversity within the human alpha IFN's (and hence a large sequence space of recombinants) along with the complexity of the IFN-α receptors and activities creates an opportunity for the construction of superior hybrids.
  • A. Complexity of the Sequence Space [0436]
  • FIG. 2 shows the protein sequences of 11 human IFN-α's. The differences from consensus are indicated. Those positions where a degenerate codon can capture all of the diversity are indicated with an asterisk. Examination of the aligned sequences reveals that there are 57 positions with two, 15 positions with three, and 4 positions with four possible amino acids encoded in this group of alpha interferon genes. Thus, the potential diversity encoded by permutation of all of this naturally occurring diversity is: 2[0437] 57×315×44=5.3×1026. Among these hybrids, of the 76 polymorphisms spread over a total of 175 sites in the 11 interferon genes, 171 of the 175 changes can be incorporated into homologue libraries using single degenerate codons at the corresponding positions. For example, Arg, Trp and Gly can all be encoded by the degenerate codon [A,T,G]GG. Using such a strategy, 1.3×1025 hybrids can be captured with a single set of degenerate oligonucleotides. As is evident from Tables III to VI, 27 oligonucleotides is sufficient to shuffle all eleven human alpha interferons. Virtually all of the natural diversity is thereby encoded and fully permuted due to degeneracies in the nine “block” oligonucleotides in Table V.
  • B. Properties of a “Coarse Grain” Search of Homologue Sequence Space [0438]
  • The modelled structure of IFN alpha (Kontsek, [0439] Acta Vir. 38:345-360 (1994)) has been divided into nine segments based on a combination of criteria of maintaining secondary structure elements as single units and placing/choosing placement of the segment boundaries in regions of high identity. Hence, one can capture the whole family with a single set of mildly degenerate oligonucleotides. Table III and FIG. 2 give the precise locations of these boundaries at the protein and DNA levels respectively. It should be emphasized that this particular segmentation scheme is arbitrary and that other segmentation schemes could also be pursued. The general strategy does not depend on placement of recombination boundaries at regions of high identity between the family members or on any particular algorithm for breaking the structure into segments.
    TABLE III
    Segmentation Scheme for Alpha Interferon
    Amino # Permutations of all
    Segment Acids # Alleles Sequence Variations
    1  1-21 5 1024
    2 22-51 10  6.2 × 104
    3 52-67 6  96
    4 68-80 7 1024
    5 81-92 7  192
    6  93-115 10  2.5 × 105
    7 116-131 4   8
    8 132-138 4   8
    9 139-167 9 9216
  • Many of the IFN's are identical over some of the segments, and thus there are less than eleven different “alleles” of each segment. Thus, a library consisting of the permutations of the segment “alleles” would have a potential complexity of 2.1×10[0440] 7 (5 segment #1's times 10 segment #2's× . . . ×9 segment #9's). This is far more than can be examined in most of the screening procedures described, and thus this is a good problem for using RSR to search the sequence space.
  • C. Detailed Strategies for Using RSR to Search the IFN-alpha Homologue Sequence Space [0441]
  • The methods described herein for oligo directed shuffling (i.e. bridge oligonucleotides) are employed to construct libraries of interferon alpha hybrids, and the general methods described above are employed to screen or select these mutants for improved function. As there are numerous formats in which to screen or select for improved interferon activity, many of which depend on the unique properties of interferons, exemplary descriptions of IFN based assays are described below. [0442]
  • D. A Protocol for a Coarse Grain Search of Hybrid IFN Alpha Sequence Space [0443]
  • In brief, libraries are constructed wherein the 11 homologous forms of the nine segments are permuted (note that in many cases two homologues are identical over a given segment). All nine segments are PCR- amplified out of all eleven IFN alpha genes with the eighteen oligonucleotides listed in Table IV, and reassembled into full length genes with oligo directed recombination. An arbitrary number, e.g., 1000, clones from the library are prepared in a 96-well expression/purification format. Hybrids with the most potent antiviral activities are screened. Nucleic acid is recovered by PCR amplification, and subjected to recombination using bridge oligonucleotides. These steps are repeated until candidates with desired properties are obtained. [0444]
  • E. Strategies for Examining the Space of >10[0445] 26 Fine Grain Hybrids
  • In brief, each of the nine segments is synthesized with one degenerate oligo per segment. Degeneracies are chosen to capture all of the IFN-alpha diversity that can be captured with a single degenerate codon without adding any non-natural sequence. A second set of degenerate oligonucleotides encoding the nine segments is generated wherein all of the natural diversity is captured, but additional non-natural mutations are included at positions where necessitated by the constraints of the genetic code. In most cases all of the diversity can be captured with a single degenerate codon; in some cases a degenerate codon will capture all of the natural diversity but will add one non-natural mutation; at a few postions it is not possible to capture the natural diversity without putting in a highly degenerate codon which will create more than one non-natural mutation. It is at these positions that this second set of oligonucleotides will differ from the first set by being more inclusive. Each of the nine synthetic segments is then amplified by PCR with the 18 PCR oligonucleotides. Full length genes using the oligo directed recombination method are generated, transfected into a host, and assayed for hybrids with desired properties. The best hybrids from (e.g, the top 10%, 1% or 0.1%; preferably the top 1%) are subjected to RSR and the process repeated until a candidate with the desired properties is obtained. [0446]
  • F. “Non-gentle” Fine Grain Search [0447]
  • On the one hand, one could make libraries wherein each segment is derived from the degenerate synthetic oligonucleotides which will encode random permutations of the homologue diversity. In this case, the initial library will very sparsely search the space of >10[0448] 25 possible fine grain hybrids that are possible with this family of genes. One could proceed by breeding positives together from this search. However, there would be a large number of differences between independent members of such libraries, and consequently the breeding process would not be very “gentle” because pools of relatively divergent genes would be recombined at each step.
  • G. “Gentle” Fine Grain Search [0449]
  • One way to make this approach more “gentle” would be to obtain a candidate starting point and to gently search from there. This starting point could be either one of the natural IFN-alpha's (such as IFN alpha-2a which is the one that is being used most widely therapeutically), the characterized IFN-Con1 consensus interferon, or a hit from screening the shuffled IFN-alpha's described above. Given a starting point, one would make separate libraries wherein one breeds the degenerate segment libraries one at a time into the founder sequence. Improved hits from each library would then be bred together to gently build up mutations all throughout the molecule. [0450]
  • H. Functional Cellular Assays [0451]
  • The following assays, well known in the art, are used to screen IFN alpha mutants: inhibition of viral killing; standard error of 30-50%; inhibition of plaque forming units; very low standard error (can measure small effects); reduced viral yield (useful for nonlethal, nonplaque forming viruses); inhibition of cell growth (3H-thymidine uptake assay; activation of NK cells to kill tumor cells; suppression of tumor formation by human INF administered to nude mice engrafted with human tumors (skin tumors for example). [0452]
  • Most of these assays are amenable to high throughput screening. Libraries of recombinant IFN alpha mutants are expressed and purified in high throughput formats such as expression, lysis and purification in a 96-well format using anti-IFN antibodies or an epitope tag and affinity resin. The purified IFN preparations are screened in a high throughput format, scored, and the mutants encoding the highest activities of interest are subjected to further mutagenesis, such as RSR, and the process repeated until a desired level of activity is obtained. [0453]
  • I. Phage Display [0454]
  • Standard phage display formats are used to display biologically active IFN. Libraries of chimeric IFN genes are expressed in this format and are selected (positively or negatively) for binding (or reduced binding) to one or more purified IFN receptor preparations or to one or more IFN receptor expressing cell types. [0455]
  • J. GFP or Luciferase Under Control of IFN-Alpha Dependent Promoter [0456]
  • Protein expressed by mutants can be screened in high throughput format on a reporter cell line which expresses GFP or luciferase under the control of an IFN alpha responsive promoter, such as an MHC Class I promoter driving GFP expression. [0457]
  • K. Stimulation of Target Cells with Intact Infections Particles [0458]
  • Purification of active IFN will limit the throughput of the assays described above. Expression of active IFN alpha on filamentous phage M13 would allow one to obtain homogenous preparations of IFN mutants in a format where thousands or tens of thousands of mutants could readily be handled. Gram et al. ([0459] J. Imm. Meth. 161:169-176 (1993)) have demonstrated that human IL3, a cytokine with a protein fold similar in topology to IFN alpha, can be expressed on the surface of M13 and that the resultant phage can present active IL3 to IL3 dependent cell lines. Similarly, Saggio et al. (Gene 152:35-39 (1995)) have shown that human ciliary neurotrophic factor, a four helix bundle cytokine, is biologically active when expressed on phage at concentrations similar to those of the soluble cytokine. Analogously, libraries of IFN alpha mutants on M13 can be expressed and lysates of defined titre used to present biologically active IFN in the high throughput assays and selections described herein.
  • The following calculation supports the feasibility of applying this technology to IFN alpha. Assuming (1) titres of 1×10[0460] 10 phage/ml with five active copies of interferon displayed per phage, and (2) that the displayed interferon is equivalently active to soluble recombinant interferon (it may well be more potent due to multi-valency), the question then is whether one can reasonably expect to see biological activity.
  • (1×1010 phage/ml)×(5 IFN molecules/phage)×(1 mole/6×1023 molecules)×(26,000 gm/mole)×(109 ng/gm)=2.2 ng/ml
  • The range of concentration used in biological assays is: 1 ng/ml for NK activation, 0.1-10 ng/ml for antiproliferative activity on Eskol cells, and 0.1-1 ng/ml on Daudi cells (Ozes et al., [0461] J. Interferon Res. 12:55-59 (1992)). Although some subtypes are glycosylated, interferon alpha2a and consensus interferon are expressed in active recombinant form in E. coli, so at least these two do not require glycosylation for activity. Thus, IFN alpha expressed on filamentous phage is likely to be biologically active as phage lysates without further purification. Libraries of IFN chimeras are expressed in phage display formats and scored in the assays described above and below to identify mutants with improved properties to be put into further rounds of RSR.
  • When one phage is sufficient to activate one cell due to the high valency state of the displayed protein (five per phage in the gene III format; hundreds per phage in the gene VIII format; tens in the lambda gene V format), then a phage lysate can be used directly at suitable dilution to stimulate cells with a GFP reporter construct under the control of an IFN responsive promoter. Assuming that the phage remain attached after stimulation, expression and FACS purification of the responsive cells, one could then directly FACS purify hybrids with improved activity from very large libraries (up to and perhaps larger than 10[0462] 7 phage per FACS run).
  • A second way in which FACS is used to advantage in this format is the following. Cells can be stimulated in a multiwell format with one lysate per well and a GFP type reporter construct. All stimulated cells are FACS purified to collect the brightest cells, and the IFN genes recovered and subjected to RSR, with iteration of the protocol until the desired level of improvement is obtained. In this protocol the stimulation is performed with individual concentrated lysates and hence the requirement that a single phage be sufficient to stimulate the cell is relaxed. Furthermore, one can gate to collect the brightest cells which, in turn, should have the most potent phage attached to them. [0463]
  • L. Cell Surface Display Protocol for IFN Alpha Mutants [0464]
  • A sample protocol follows for the cell surface display of IFN alpha mutants. This form of display has at least two advantages over phage display. First, the protein is displayed by a eukaryotic cell and hence can be expressed in a properly glycosylated form which may be necessary for some IFN alphas (and other growth factors). Secondly, it is a very high valency display format and is preferred in detecting activity from very weakly active mutants. [0465]
  • In brief, a library of mutant IFN's is constructed wherein a polypeptide signal for addition of a phosphoinositol tail has been fused to the carboxyl terminus, thus targeting the protein for surface expression (Wettstein et al., [0466] J. Exp. Med. 174:219-28 (1991)). The library is used to transfect reporter cells described above (luciferase reporter gene) in a microtiter format. Positives are detected with a charge coupling device(CCD) camera. Nucleic acids are recovered either by HIRT and retransformation of the host or by PCR, and are subjected to RSR for further evolution.
  • M. Autocrine Display Protocol for Viral Resistance [0467]
  • A sample protocol follows for the autocrine display of IFN alpha mutants. In brief, a library of IFN mutants is generated in a vector which allows for induction of expression (i.e. metallothionein promoter) and efficient secretion. The recipient cell line carrying an IFN responsive reporter cassette [GFP or luciferase] is induced by transfection with the mutant IFN constructs. Mutants which stimulate the IFN responsive promoter are detected by by FACS or CCD camera. [0468]
  • A variation on this format is to challenge transfectants with virus and select for survivors. One could do multiple rounds of viral challenge and outgrowth on each set of transfectants prior to retrieving the genes. Multiple rounds of killing and outgrowth allow an exponential amplification of a small advantage and hence provide an advantage in detecting small improvements in viral killing. [0469]
    TABLE IV
    Oligonucleotides needed for blockwise
    recombination: 18
    Oligonucleotides for alpha interferon
    shuffling
    1. 5′-TGT[G/A]ATCTG[C/T]CT[C/G]AGACC
    2. 5′-GGCACAAATG[G/A/C]G[A/C]AGAATCTCTC
    3. 5′-AGAGATTCT[G/T]C[C/T/G]CATTTGTGCC
    4. 5′-CAGTTCCAGAAG[A/G]CT[G/C][C/A]AGCCATC
    5. 5′-GATGGCT[T/G][G/C]AG[T/C]CTTCTGGAACTG
    6. 5′-CTTCAATCTCTTCA[G/C]CACA
    7. 5′-TGTG[G/C]TGAAGAGATTGAAG
    8. 5′-GGA[T/A][G/C]AGA[C/G][C/G]CTCCTAGA
    9. 5′-TCTAGGAG[G/C][G/C]TCT[G/C][T/A]TCC
    10. 5′-GAACTT[T/G/A][T/A]CCAGCAA[A/C]TGAAT
    11. 5′-ATTCA[T/G]TTGCTGG[A/T][A/T/C]AAGTTC
    12. 5′-GGACT[T/C]CATCCTGGCTGTG
    13. 5′-CACAGCCAGGATG[G/A]AGTCC
    14. 5′-AAGAATCACTCTTTATCT
    15. 5′-AGATAAAGAGTGATTCTT
    16. 5′-TGGGAGGTTGTCAGAGCAG
    17. 5′-CTGCTCTGACAACCTCCCA
    18. 5′-TCA[A/T]TCCTT[C/A]CTC[T/C]TTAA
  • Brackets indicate degeneracy with equal mixture of the specified bases at those positions. The purpose of the degeneracy is to allow this one set of primers to prime all members of the IFN family with similar efficiency. The choice of the oligo driven recombination points is important because they will get “overwritten” in each cycle of breeding and hence cannot coevolve with the rest of the sequence over many cycles of selection. [0470]
    TABLE V
    Oligonucleotides needed for “fine grain” recombination
    of natural diversity over each of the nine blocks
    #Length of
    Block oligo required
    1 76
    2 95
    3 65
    4 56
    5 51
    6 93
    7 50
    8 62
    9 80
  • [0471]
    TABLE VI
    Amino acids that can be reached by a single
    step mutation in the codon of interest.
    Wild-Type Amino Amino acids reachable
    Acid by one mutation
    W C, R, G, L
    Y F, S. C, H, N, D
    F L, I, V, S, Y, C
    L S, W, F, I, M, V, P
    V F, L, I, M, A, D, E, G
    I F, L, M, V, T, N, K, S, R
    A S, P, T, V, D, E, G
    G V, A, D, E, R, S, C, W
    M L, I, V, T, K, R
    S F, L, Y, C, W, P, T, A, R, G, N, T,
    I
    T S, P, A, I, M, N, K, S, R
    P S, T, A, L, H, Q, R
    C F, S, Y, R, G, W
    N Y, H, K, D, S, T, I
    Q Y, H, K, E, L, P, R
    H Y, Q, N, D, L, P, R
    D Y, H, N, E, V, A, G
    E Q, K, D, V, A, G
    R L, P, H, Q, C, W, S, G, K, T, I, M
    K Q, N, E, R, T, I, M
  • Based on this Table, the polymorphic positions in IFN alpha where all of the diversity can be captured by a degenerate codon have been identified. Oligonucleotides of the length indicated in Table V above with the degeneracies inferred from Table VI are synthesized. [0472]
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims. [0473]
  • All references cited herein are expressly incorporated in their entirety for all purposes. [0474]
  • 1 101 1 50 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 1 aaccctccag ttccgaaccc catatgatga tcaccctgcg taaactgccg 50 2 38 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 2 aaccctccag ttccgaaccc catatgaaaa aaaccgct 38 3 40 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 3 aaccctccag ttccgaaccc atatacatat gcgtgctaaa 40 4 44 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 4 aaccctccag ttccgaaccc catatgaaat acctgctgcc gacc 44 5 40 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 5 aaccctccag ttccgaaccc gatatacata tgaaacagtc 40 6 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 6 tggtgttatg tctgctcagg cdatggcdgt dgayttycay ctggttccgg ttgaagagga 60 7 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 7 ggctggtttc gctaccgttg cdcargcdgc dccdaargay ctggttccgg ttgaagagga 60 8 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 8 caccccgatc gctatctctt cyttygcdtc yacyggytcy ctggttccgg ttgaagagga 60 9 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 9 gctgctggct gctcagccgg cdatggcdat ggayatyggy ctggttccgg ttgaagagga 60 10 61 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 10 tgccgctgct gttcaccccg gtdacyaarg cdgcdcargt dctggttccg gttgaagagg 60 a 61 11 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 11 cccggctttc tggaaccgtc argcdgcdca rgcdctggac gttgctaaaa aactgcagcc 60 12 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 12 acgttatcct gttcctgggt gayggyatgg gygtdccdac cgttaccgct acccgtatcc 60 13 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 13 aaactgggtc cggaaacccc dctggcdatg gaycarttyc cgtacgttgc tctgtctaaa 60 14 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 14 ggttccggac tctgctggta cygcdacygc dtayctgtgc ggtgttaaag gtaactaccg 60 15 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 15 ctgctcgtta caaccagtgc aaracyacyc gyggyaayga agttacctct gttatgaacc 60 16 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 16 tctgttggtg ttgttaccac yacycgygtd carcaygcdt ctccggctgg tgcttacgct 60 17 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 17 gtactctgac gctgacctgc cdgcdgaygc dcaratgaac ggttgccagg acatcgctgc 60 18 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 18 acatcgacgt tatcctgggt ggyggycgya artayatgtt cccggttggt accccggacc 60 19 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 19 tctgttaacg gtgttcgtaa rcgyaarcar aayctggtdc aggcttggca ggctaaacac 60 20 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 20 gaaccgtacc gctctgctgc argcdgcdga ygaytcytct gttacccacc tgatgggtct 60 21 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 21 aatacaacgt tcagcaggac cayacyaarg ayccdacyct gcaggaaatg accgaagttg 60 22 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 22 aacccgcgtg gtttctacct gttygtdgar ggyggycgya tcgaccacgg tcaccacgac 60 23 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 23 gaccgaagct ggtatgttcg ayaaygcdat ygcdaargct aacgaactga cctctgaact 60 24 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 24 ccgctgacca ctctcacgtt ttytcyttyg gyggytayac cctgcgtggt acctctatct 60 25 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 25 gctctggact ctaaatctta yacytcyaty ctgtayggya acggtccggg ttacgctctg 60 26 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 26 cgttaacgac tctacctctg argayccdtc ytaycarcag caggctgctg ttccgcaggc 60 27 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 27 aagacgttgc tgttttcgct cgyggyccdc argcdcayct ggttcacggt gttgaagaag 60 28 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 28 atggctttcg ctggttgcgt dgarccdtay acygaytgya acctgccggc tccgaccacc 60 29 61 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 29 tgctcacctg gctgcttmac cdccdccdct ggcdctgctg gctggtgcta tgctgctcct 60 c 61 30 62 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 30 ttccgcctct agagaattct tartacagrg thgghgccag gaggagcagc atagcaccag 60 cc 62 31 58 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 31 aagcagccag gtgagcagcg tchggratrg argthgcggt ggtcggagcc ggcaggtt 58 32 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 32 cgcaaccagc gaaagccatg atrtghgcha craargtytc ttcttcaaca ccgtgaacca 60 33 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 33 gcgaaaacag caacgtcttc rccrccrtgr gtytcrgahg cctgcggaac agcagcctgc 60 34 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 34 agaggtagag tcgttaacgt chggrcgrga rccrccrccc agagcgtaac ccggaccgtt 60 35 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 35 aagatttaga gtccagagct ttrgahgghg ccagrccraa gatagaggta ccacgcaggg 60 36 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 36 acgtgagagt ggtcagcggt haccagratc agrgtrtcca gttcagaggt cagttcgtta 60 37 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 37 gaacatacca gcttcggtca ghgccatrta hgcyttrtcg tcgtggtgac cgtggtcgat 60 38 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 38 ggtagaaacc acgcgggtta cgrgahacha crcgcaghgc aacttcggtc atttcctgca 60 39 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 39 tcctgctgaa cgttgtattt catrtchgch ggytcraaca gacccatcag gtgggtaaca 60 40 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 40 cagcagagcg gtacggttcc ahacrtaytg hgcrccytgg tgtttagcct gccaagcctg 60 41 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 41 tacgaacacc gttaacagaa gcrtcrtchg grtaytchgg gtccggggta ccaaccggga 60 42 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 42 cccaggataa cgtcgatgtc catrttrtth accagytghg cagcgatgtc ctggcaaccg 60 43 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 43 caggtcagcg tcagagtacc arttrcgrtt hacrgtrtga gcgtaagcac cagccggaga 60 44 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 44 tggtaacaac accaacagat ttrcchgcyt tytthgcrcg gttcataaca gaggtaactt 60 45 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 45 cactggttgt aacgagcagc hgcrgahacr ccratrgtrc ggtagttacc tttaacaccg 60 46 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 46 accagcagag tccggaacct grcgrtchac rttrtargtt ttagacagag caacgtacgg 60 47 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 47 gggtttccgg acccagttta ccrttcatyt grccyttcag gatacgggta gcggtaacgg 60 48 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 48 cccaggaaca ggataacgtt ytthgchgcr gtytgrathg gctgcagttt tttagcaacg 60 49 42 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 49 acggttccag aaagccgggt cttcctcttc aaccggaacc ag 42 50 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 50 cctgagcaga cataacacca gchgchachg chachgccag cggcagttta cgcagggtga 60 51 62 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 51 accggggtga acagcagcgg cagcaghgcc aghgcratrg trgactgttt catatgtata 60 tc 62 52 59 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 52 gccggctgag cagccagcag cagcagrcch gchgchgcgg tcggcagcag gtagtttca 59 53 60 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 53 aagagatagc gatcggggtg gtcaghacra trcccagcag tttagcacgc atatgtatat 60 54 58 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 54 caacggtagc gaaaccagcc aghgchachg crathgcrat agcggttttt ttcatatg 58 55 39 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 55 agaattctct agaggcggaa actctccaac tcccaggtt 39 56 39 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for codon usage library 56 tgagaggttg agggtccaat tgggaggtca aggcttggg 39 57 18 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 57 tgtratctgy ctsagacc 18 58 23 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 58 ggcacaaatg vgmagaatct ctc 23 59 22 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 59 agagattctk cbcatttgtg cc 22 60 24 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 60 cagttccaga agrctsmagc catc 24 61 24 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 61 gatggctksa gycttctgga actg 24 62 19 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 62 cttcaatctc ttcascaca 19 63 19 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 63 tgtgstgaag agattgaag 19 64 18 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 64 ggawsagass ctcctaga 18 65 18 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 65 tctaggagss tctswtcc 18 66 21 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 66 gaacttdwcc agcaamtgaa t 21 67 21 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 67 attcakttgc tggwhaagtt c 21 68 19 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 68 ggactycatc ctggctgtg 19 69 19 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 69 cacagccagg atgragtcc 19 70 18 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 70 aagaatcact ctttatct 18 71 18 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 71 agataaagag tgattctt 18 72 19 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 72 tgggaggttg tcagagcag 19 73 19 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 73 ctgctctgac aacctccca 19 74 18 DNA Artificial Sequence Description of Artificial Sequence degenerate oligonucleotide used for alpha interferon shuffling 74 tcawtccttm ctcyttaa 18 75 166 PRT consensus alpha interferon 75 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Gly Phe Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Lys Asp Ser Ser Ala Ala Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Val Glu Glu Thr Pro Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Arg Leu Arg Arg Lys Asp 165 76 166 PRT human alpha interferon 76 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Gly Leu Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Thr Gln Ala Ile Pro Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Glu Asp Ser Ser Ala Ala Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Tyr Gln Gln Leu Asn Asn Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Met Glu Glu Thr Pro Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Arg Leu Arg Arg Lys Asp 165 77 166 PRT human alpha interferon 77 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg Pro Asp Phe Gly Leu Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Thr Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Glu Asp Ser Ser Ala Ala Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Tyr Gln Gln Leu Asn Asn Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Met Glu Glu Thr Pro Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Ile Leu Arg Arg Lys Asp 165 78 166 PRT human alpha interferon 78 Cys Asn Leu Ser Gln Thr His Ser Leu Asn Asn Arg Arg Thr Leu Met 1 5 10 15 Leu Leu Ala Gln Met Arg Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Glu Phe Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Gln Ala Ile Ser Val Leu His Glu Met Met Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Lys Asn Ser Ser Ala Ala Trp Asp Glu Thr 65 70 75 80 Leu Leu Glu Lys Phe Tyr Ile Glu Leu Phe Gln Gln Met Asn Asp Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Val Glu Glu Thr Pro Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Lys Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Met Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Arg Leu Arg Arg Lys Asp 165 79 166 PRT human alpha interferon 79 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser His Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Gly Phe Pro Glu Glu Glu Phe Asp Gly His Gln Phe 35 40 45 Gln Lys Thr Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Glu Asp Ser Ser Ala Ala Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Val Glu Glu Thr Pro Leu Met 100 105 110 Asn Val Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Arg Leu Arg Arg Lys Asp 165 80 166 PRT human alpha interferon 80 Cys Asp Leu Pro Gln Thr His Ser Leu Gly His Arg Arg Thr Met Met 1 5 10 15 Leu Leu Ala Gln Met Arg Arg Ile Ser Leu Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Arg Phe Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Glu Ala Ile Ser Val Leu His Glu Val Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Lys Asp Ser Ser Val Ala Trp Asp Glu Arg 65 70 75 80 Leu Leu Asp Lys Leu Tyr Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Met Gln Glu Val Trp Val Gly Gly Thr Pro Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Phe Ser Ser Ser Arg Asn Leu Gln Glu 145 150 155 160 Arg Leu Arg Arg Lys Glu 165 81 166 PRT human alpha interferon 81 Cys Asp Leu Pro Gln Thr His Ser Leu Arg Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Glu Phe Arg Phe Pro Glu Glu Glu Phe Asp Gly His Gln Phe 35 40 45 Gln Lys Thr Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Glu Asp Ser Ser Ala Ala Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Val Glu Glu Thr Pro Leu Met 100 105 110 Asn Glu Asp Phe Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Met Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Phe Ser Phe Ser Thr Asn Leu Lys Lys 145 150 155 160 Gly Leu Arg Arg Lys Asp 165 82 166 PRT human alpha interferon 82 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Arg Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Glu Phe Pro Gln Glu Glu Phe Asp Asp Lys Gln Phe 35 40 45 Gln Lys Ala Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Lys Asp Ser Ser Ala Ala Leu Asp Glu Thr 65 70 75 80 Leu Leu Asp Glu Phe Tyr Ile Glu Leu Asp Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ser Cys Val Met Gln Glu Val Gly Val Ile Glu Ser Pro Leu Met 100 105 110 Tyr Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Ser Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Phe Ser Leu Ser Ile Asn Leu Gln Lys 145 150 155 160 Arg Leu Lys Ser Lys Glu 165 83 166 PRT human alpha interferon 83 Cys Asp Leu Pro Glu Thr His Ser Leu Asp Asn Arg Arg Thr Leu Met 1 5 10 15 Leu Leu Ala Gln Met Ser Arg Ile Ser Pro Ser Ser Cys Leu Met Asp 20 25 30 Arg His Asp Phe Gly Phe Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Pro Ala Ile Ser Val Leu His Glu Leu Ile Gln Gln Ile 50 55 60 Phe Asn Leu Phe Thr Thr Lys Asp Ser Ser Ala Ala Trp Asp Glu Asp 65 70 75 80 Leu Leu Asp Lys Phe Cys Thr Glu Leu Tyr Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Met Gln Glu Glu Arg Val Gly Glu Thr Pro Leu Met 100 105 110 Asn Ala Asp Ser Ile Leu Ala Val Lys Lys Tyr Phe Arg Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Leu Ser Thr Asn Leu Gln Glu 145 150 155 160 Arg Leu Arg Arg Lys Glu 165 84 166 PRT human alpha interferon 84 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg His Asp Phe Gly Phe Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Lys Asp Ser Ser Ala Ile Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Asn Gln Gln Leu Asn Asp Met 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Val Glu Glu Thr Pro Leu Met 100 105 110 Asn Val Asp Ser Ile Leu Ala Val Lys Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Phe Ser Leu Ser Lys Ile Phe Gln Glu 145 150 155 160 Arg Leu Arg Arg Lys Ser 165 85 166 PRT human alpha interferon 85 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser Pro Phe Ser Cys Leu Lys Asp 20 25 30 Arg Pro Asp Phe Gly Leu Pro Gln Glu Glu Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Thr Gln Ala Ile Ser Val Leu His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Glu Asp Ser Ser Ala Ala Trp Glu Gln Ser 65 70 75 80 Leu Leu Glu Lys Phe Ser Thr Glu Leu Tyr Gln Gln Leu Asn Asn Leu 85 90 95 Glu Ala Cys Val Ile Gln Glu Val Gly Met Glu Glu Thr Pro Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Thr Glu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Leu Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Ile Leu Arg Arg Lys Asp 165 86 166 PRT human alpha interferon 86 Cys Asp Leu Pro Gln Thr His Ser Leu Gly Asn Arg Arg Ala Leu Ile 1 5 10 15 Leu Leu Ala Gln Met Gly Arg Ile Ser His Phe Ser Cys Leu Lys Asp 20 25 30 Arg Tyr Asp Phe Gly Phe Pro Gln Glu Val Phe Asp Gly Asn Gln Phe 35 40 45 Gln Lys Ala Gln Ala Ile Ser Ala Phe His Glu Met Ile Gln Gln Thr 50 55 60 Phe Asn Leu Phe Ser Thr Lys Asp Ser Ser Ala Ala Trp Asp Glu Thr 65 70 75 80 Leu Leu Asp Lys Phe Tyr Ile Glu Leu Phe Gln Gln Leu Asn Asp Leu 85 90 95 Glu Ala Cys Val Thr Gln Glu Val Gly Val Glu Glu Ile Ala Leu Met 100 105 110 Asn Glu Asp Ser Ile Leu Ala Val Arg Lys Tyr Phe Gln Arg Ile Thr 115 120 125 Leu Tyr Leu Met Gly Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Glu Ile Met Arg Ser Phe Ser Phe Ser Thr Asn Leu Gln Lys 145 150 155 160 Gly Leu Arg Arg Lys Asp 165 87 501 DNA consensus alpha interferon 87 tgtgatctgc ctcagaccca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcctgcctg aaggacagac atgactttgg atttccccag 120 gaggagtttg atggcaacca gttccagaag gctcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca aaggactcat ctgctgcttg ggatgagagc 240 ctcctagaaa aattttccac tgaactttac cagcaactga atgacctgga agcctgtgtg 300 atacaggagg ttggggtgga agagactccc ctgatgaatg aggactccat cctggctgtg 360 aggaaatact tccaaagaat cactctttat ctgacagaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccttctctt tttcaacaaa cttgcaaaaa 480 agattaagga ggaaggattg a 501 88 501 DNA human alpha interferon 88 tgtgatctgc ctcagaccca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcctgcctg aaggacagac atgactttgg acttccccag 120 gaggagtttg atggcaacca gttccagaag actcaagcca tccctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca gaggactcat ctgctgcttg ggaacagagc 240 ctcctagaaa aattttccac tgaactttac cagcaactga ataacctgga agcatgtgtg 300 atagaggagg ttgggatgga agagactccc ctgatgaatg aggactccat cctggctgtg 360 aggaaatact tccaaagaat cactctttat ctaacagaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccctctctt tttcaacaaa cttgcaaaaa 480 agattaagga ggaaggattg a 501 89 501 DNA human alpha interferon 89 tgtgatctgc ctcagaccca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcctgcctg aaggacagac ctgactttgg acttccccag 120 gaggagtttg atggcaacca gttccagaag actcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca gaggactcat ctgctgcttg ggaacagagc 240 ctcctagaaa aattttccac tgaactttac cagcaactga ataacctgga agcatgtgtg 300 atacaggagg ttgggatgga agagactccc ctgatgaatg aggactccat cctggctgtg 360 aggaaatact tccaaagaat cactctttat ctaacagaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tctctctctt tttcaacaaa cttgcaaaaa 480 atattaagga ggaaggattg a 501 90 501 DNA human alpha interferon 90 tgtaatctgt ctcaaaccca cagcctgaat aacaggagga ctttgatgct catggcacaa 60 atgaggagaa tctctccttt ctcctgcctg aaggacagac atgactttga atttccccag 120 gaggaatttg atggcaacca gttccagaaa gctcaagcca tctctgtcct ccatgagatg 180 atgcagcaga ccttcaatct cttcagcaca aagaactcat ctgctgcttg ggatgagacc 240 ctcctagaaa aattctacat tgaacttttc cagcaaatga atgacctgga agcctgtgtg 300 atacaggagg ttggggtgga agagactccc ctgatgaatg aggactccat cctggctgtg 360 aagaaatact tccaaagaat cactctttat ctgatggaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccctctctt tttcaacaaa cttgcaaaaa 480 agattaagga ggaaggattg a 501 91 501 DNA human alpha interferon 91 tgtgatctgc ctcagaccca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcatgcctg aaggacagac atgatttcgg attccccgag 120 gaggagtttg atggccacca gttccagaag actcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca gaggactcat ctgctgcttg ggaacagagc 240 ctcctagaaa aattttccac tgaactttac cagcaactga atgacctgga agcatgtgtg 300 atacaggagg ttggggtgga agagactccc ctgatgaatg tggactccat cctggctgtg 360 aggaaatact tccaaagaat cactctttat ctaacagaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccctctcgt tttcaacaaa cttgcaaaaa 480 agattaagga ggaaggattg a 501 92 501 DNA human alpha interferon 92 tgtgatctgc ctcagaccca cagcctgggt cacaggagga ccatgatgct cctggcacaa 60 atgaggagaa tctctctttt ctcctgtctg aaggacagac atgacttcag atttccccag 120 gaggagtttg atggcaacca gttccagaag gctgaagcca tctctgtcct ccatgaggtg 180 attcagcaga ccttcaatct cttcagcaca aaggactcat ctgttgcttg ggatgagagg 240 cttctagaca aactctatac tgaactttac cagcagctga atgacctgga agcctgtgtg 300 atgcaggagg tgtgggtggg agggactccc ctgatgaatg aggactccat cctggctgtg 360 agaaaatact tccaaagaat cactctctac ctgacagaga aaaagtacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccttctctt catcaagaaa cttgcaagaa 480 aggttaagga ggaaggaata a 501 93 501 DNA human alpha interferon 93 tgtgatctgc ctcagaccca cagcctgcgt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcctgcttg aaggacagac atgaattcag attcccagag 120 gaggagtttg atggccacca gttccagaag actcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca gaggactcat ctgctgcttg ggaacagagc 240 ctcctagaaa aattttccac tgaactttac cagcaactga atgacctgga agcatgtgtg 300 atacaggagg ttggggtgga agagactccc ctgatgaatg aggactccat cctggctgtg 360 aggaaatact tccaaagaat cactctttat ctaatggaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccttctctt tttcaacaaa cttgaaaaaa 480 ggattaagga ggaaggattg a 501 94 501 DNA human alpha interferon 94 tgtgatctgc ctcagactca cagcctgggt aacaggaggg ccttgatact cctggcacaa 60 atgcgaagaa tctctccttt ctcctgcctg aaggacagac atgactttga attcccccag 120 gaggagtttg atgataaaca gttccagaag gctcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaacct cttcagcaca aaggactcat ctgctgcttt ggatgagacc 240 cttctagatg aattctacat cgaacttgac cagcagctga atgacctgga gtcctgtgtg 300 atgcaggaag tgggggtgat agagtctccc ctgatgaatg aggacttcat cctggctgtg 360 aggaaatact tccaaagaat cactctatat ctgacagaga agaaatacag ctcttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccttctctt tatcaatcaa cttgcaaaaa 480 agattgaaga gtaaggaatg a 501 95 501 DNA human alpha interferon 95 tgtgatctcc ctgagaccca cagcctggat aacaggagga ccttgatgct cctggcacaa 60 atgagcagaa tctctccttc ctcctgtctg atggacagac atgactttgg atttccccag 120 gaggagtttg atggcaacca gttccagaag gctccagcca tctctgtcct ccatgagctg 180 atccagcaga tcttcaacct cttctccaca aaagattcat ctgctgcttg ggatgaggac 240 ctcctagaca aattctgcac cgaactctac cagcagctga atgacttgga agcctgtgtg 300 atgcaggagg agagggtggg agaaactccc ctgatgtacg cggactccat cctggctgtg 360 aagaaatact tccaaagaat cactctctat ctgacagaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccctctctt tatcaacaaa cttgcaagaa 480 agattaagga ggaaggaata a 501 96 501 DNA human alpha interferon 96 tgtgatctgc ctcagaccca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcctgcctg aaggacagac atgactttgg attcccccaa 120 gaggagtttg atggcaacca gttccagaag gctcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca aaggactcat ctgctacttg ggaacagagc 240 ctcctagaaa aattttccac tgaacttaac cagcagctga atgacatgga agcctgcgtg 300 atacaggagg ttggggtgga agagactccc ctgatgaatg tggactctat cctggctgtg 360 aagaaatact tccaaagaat cactctttat ctgacagaga agaaatacag cccttgtgct 420 tgggaggttg tcagagcaga aatcatgaga tccttctctt tatcaaaaat ttttcaagaa 480 agattaagga ggaaggaatg a 501 97 501 DNA human alpha interferon 97 tgtgatctgc ctcagaccca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctccttt ctcctgcctg aaggacagac ctgactttgg acttccccag 120 gaggagtttg atggcaacca gttccagaag actcaagcca tctctgtcct ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca gaggactcat ctgctgcttg ggaacagagc 240 ctcctagaaa aattttccac tgaactttac cagcaactga ataacctgga agcatgtgtg 300 atacaggagg ttgggatgga agagactccc ctgatgaatg aggactccat cttggctgtg 360 aggaaatact tccaaagaat cactctttat ctaacagaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tctctctctt tttcaacaaa cttgcaaaaa 480 agattaagga ggaaggattg a 501 98 501 DNA human alpha interferon 98 tgtgatctgc ctcagactca cagcctgggt aataggaggg ccttgatact cctggcacaa 60 atgggaagaa tctctcattt ctcctgcctg aaggacagat atgatttcgg attcccccag 120 gaggtgtttg atggcaacca gttccagaag gctcaagcca tctctgcctt ccatgagatg 180 atccagcaga ccttcaatct cttcagcaca aaggattcat ctgctgcttg ggatgagacc 240 ctcctagaca aattctacat tgaacttttc cagcaactga atgacctaga agcctgtgtg 300 acacaggagg ttggggtgga agagattgcc ctgatgaatg aggactccat cctggctgtg 360 aggaaatact ttcaaagaat cactctttat ctgatggaga agaaatacag cccttgtgcc 420 tgggaggttg tcagagcaga aatcatgaga tccttctctt tttcaacaaa cttgcaaaaa 480 ggattaagaa ggaaggattg a 501 99 11 PRT Artificial Sequence Description of Artificial Sequence Protease peptide substrate 99 Arg Gly Val Val Asn Ala Ser Ser Arg Leu Ala 1 5 10 100 44 DNA Artificial Sequence Description of Artificial Sequence Introduced Sfi I site 100 ttccatttca tacatggccg aaggggccgt gccatgagga tttt 44 101 50 DNA Artificial Sequence Description of Artificial Sequence Introduced sfi I site 101 ttctaaatgc atgttggcct ccttggccgg attctgagcc ttcaggacca 50

Claims (273)

What is claimed is:
1. A method for evolving a protein encoded by a DNA substrate molecule comprising:
(a) digesting at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide, with a restriction endonuclease;
(b) ligating the mixture to generate a library of recombinant DNA molecules;
(c) screening or selecting the products of (b) for a desired property; and
(d) recovering a recombinant DNA substrate molecule encoding an evolved protein.
2. The method of claim 1, wherein the restriction endonuclease generates non-palindromic ends at cleavage sites.
3. The method of claim 1, wherein the substrate molecules have been engineered to contain at least one recognition site for a restriction endonuclease having non-palindromic ends at cleavage sites.
4. The method of claim 1, wherein (a)-(d) are repeated.
5. The method of claim 1, wherein the DNA substrate molecule comprises a gene cluster.
6. The method of claim 1, wherein at least one restriction endonuclease fragment from a DNA substrate molecule is isolated and subjected to mutagenesis to generate a library of mutant fragments.
7. The method of step 6, wherein the library of mutant fragments is used in the ligation of (b).
8. The method of claim 7, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
9. The method of claim 6, wherein mutagenesis comprises recursive sequence recombination.
10. The method of claim 1, wherein the products of (d) are subjected to mutagenesis.
11. The method of claim 10, wherein mutagenesis comprises recursive sequence recombination.
12. The method of claim 1, wherein the products of (d) are used as a DNA substrate molecule in (b).
13. The method of claim 10, wherein the products of claim 10 are used in (d).
14. The method of claim 1, wherein the recombinant DNA substrate molecule of (d) comprises a library of recombinant DNA substrate molecules.
15. An evolved protein produced by the method of claim 1.
16. A method for evolving a protein encoded by a DNA substrate molecule by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide and comprise defined segments, the method comprising:
(a) providing a set of oligonucleotide PCR primers, comprising at least one primer for each strand of each segment, wherein the primer sequence is complementary to at least one junction with another segment;
(b) amplifying the segments of the at least a first and second DNA substrate molecules with the primers of step (a) in a polymerase chain reaction;
(c) assembling the products of step (b) to generate a library of recombinant DNA substrate molecules;
(d) screening or selecting the products of (c) for a desired property; and
(e) recovering a recombinant DNA substrate molecule from (d) encoding an evolved protein.
17. The method of claim 16, wherein the at least a first and second DNA substrate molecules are subjected to mutagenesis prior to step (a).
18. The method of claim 16, wherein the at least a first and second DNA substrate molecules comprise alleles of a gene.
19. The method of claim 16, wherein the at least a first and second DNA substrate molecules comprise a library of mutants.
20. The method of claim 16, wherein the segments are defined by sites within intergenic regions.
21. The method of claim 16, wherein the segments are defined by sites within introns.
22. The method of claim 16, wherein the primers comprise a uracil substitution at one or more thymidine residues.
23. The method of claim 22, wherein the products of (b) are treated with uracil glycosylase.
24. The method of claim 16, wherein (a)-(e) are repeated.
25. The method of claim 16, wherein the at least a first and second DNA substrate molecule comprises a gene cluster.
26. The method of claim 16, wherein the at least first and second DNA substrate molecule encodes all or part of a DNA polymerase.
27. The method of claim 16, wherein at least one PCR primer differs from the at least a first and second DNA substrate molecules in at least one nucleotide.
28. The method of claim 27, wherein the PCR primer comprises a nucleotide sequence of a known mutant or polymorphism of the at least a first or second DNA substrate molecule.
29. The method of claim 28, wherein the PCR primer is degenerate and encodes the nucleotide sequences of more than one known mutant or polymorphism of the at least a first or second DNA substrate molecule.
30. The method of claim 29, wherein the at least a first and second DNA substrate molecule encodes all or part of a protein selected from Table I.
31. The method of claim 17, wherein mutagenesis comprises recursive sequence recombination.
32. The method of claim 16, wherein the products of (e) are subjected to mutagenesis.
33. The method of claim 32, wherein mutagenesis comprises recursive sequence recombination.
34. The method of claim 32, wherein the products of claim 32 are used in (b).
35. The method of claim 16, wherein the products of (e) are used as a DNA substrate molecule in (b).
36. The method of claim 16, wherein the recombinant DNA substrate molecule of (e) comprises a library of recombinant DNA substrate molecules.
37. An evolved protein produced by the method of claim 16.
38. A method of enriching a population of DNA fragments for mutant sequences comprising:
(a) denaturing and renaturing the population of fragments to generate a population of hybrid double-stranded fragments in which at least one double-stranded fragment comprises at least one base pair mismatch;
(b) fragmenting the products of (a) into fragments of about 20-100 bp;
(c) affinity-purifying fragments having a mismatch on an affinity matrix to generate a pool of DNA fragments enriched for mutant sequences; and
(d) assembling the products of (c) to generate a library of recombinant DNA substrate molecules.
39. The method of claim 38, wherein the population of DNA fragments is derived from at least a first and second DNA substrate molecule, the at least a first and second DNA substrate molecule differing from each other in at least one nucleotide.
40. The method of claim 39, wherein the at least a first and second DNA substrate molecules are obtained by mutagenesis of a DNA substrate molecule.
41. The method of claim 39, wherein the at least a first and second DNA substrate molecules comprise alleles of a gene.
42. The method of claim 39, wherein the at least a first and second DNA substrate molecules comprise polymorphic variants of a gene.
43. The method of claim 38, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
44. The method of claim 38, wherein the products of (c) are mixed with the products of (a) prior to (d).
45. A method for evolving a protein encoded by a DNA substrate molecule, by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules share a region of sequence homology of about 10 to 100 base pairs and comprise defined segments, the method comprising:
(a) providing regions of homology in the at least a first and second DNA substrate molecules by inserting an intron sequence between at least two defined segments;
(b) fragmenting and recombining DNA substrate molecules of (a), wherein regions of homology are provided by the introns;
(c) screening or selecting the products of (b) for a desired property; and
(d) recovering a recombinant DNA substrate molecule from the products of (c) encoding an evolved protein.
46. The method of claim 45, wherein the introns are self-splicing.
47. The method of claim 45, wherein the inserted introns comprise from about 1 to about 10 nonhomologous introns.
48. The method of claim 45, wherein the intron comprises a recognition site for a restriction endonucleases having non-palindromic ends at cleavage sites.
49. The method of claim 45, wherein (b)-(d) are repeated.
50. The method of claim 45, wherein the DNA substrate molecule comprises a gene cluster.
51. The method of claim 45, wherein at least one segment from a DNA substrate molecule is isolated and subjected to mutagenesis to generate a library of mutant fragments.
52. The method of claim 51, wherein the library of mutant segments is used in the recombination of (b).
53. The method of claim 45, wherein the segments are defined by exons.
54. The method of claim 45, wherein the segments are defined by intergenic regions.
55. The method of claim 45, wherein the at least a first and second DNA substrate molecules encode protein homologues.
56. The method of claim 45, wherein the intron contains a lox site, and wherein the products of (b) are used to transfect a Cre+ host.
57. The method of claim 45, wherein the at least a first and second DNA substrate molecule encodes all or part of a protein selected from Table I.
58. The method of claim 45, wherein the at least a first and second DNA substrate molecule are subjected to mutagenesis prior to step (a).
59. The method of claim 58, wherein mutagenesis comprises recursive sequence recombination.
60. The method of claim 45, wherein the products of (d) are subjected to mutagenesis.
61. The method of claim 58, wherein mutagenesis comprises recursive sequence recombination.
62. The method of claim 45, wherein the products of (d) are used as a DNA substrate molecule in (b).
63. The method of claim 45, wherein the recombinant DNA substrate molecule of (d) comprises a library of recombinant DNA substrate molecules.
64. An evolved protein produced by the method of claim 45.
65. A method for evolving a protein encoded by a DNA substrate molecule by recombining at least a first and second DNA substrate molecule, wherein the at least a first and second substrate molecules differ from each other in at least one nucleotide and comprise defined segments, the method comprising:
(a) providing a set of oligonucleotide PCR primers, wherein for each junction of segments a pair of primers is provided, one member of each pair bridging the junction at one end of a segment and the other bridging the junction at the other end of the segment, with the terminal ends of the DNA molecule having as one member of the pair a generic primer, and wherein a set of primers is provided for each of the at least a first and second substrate molecules;
(b) amplifying the segments of the at least a first and second DNA substrate molecules with the primers of (a) in a polymerase chain reaction;
(c) assembling the products of (b) to generate a pool of recombinant DNA molecules;
(d) selecting or screening the products of (c) for a desired property; and
(e) recovering a recombinant DNA substrate molecule from the products of (d) encoding an evolved protein.
66. The method of claim 65, wherein (a)-(e) is repeated.
67. The method of claim 65, wherein the at least a first and second DNA substrate molecule are subjected to mutagenesis prior to (a).
68. The method of claim 65, wherein the at least a first and second DNA substrate molecule comprise sequences encoding protein homologues.
69. The method of claim 65, wherein the primers comprise a uracil substitution at one or more thymidine residues.
70. The method of claim 69, wherein the products of (b) are treated with uracil glycosylase.
71. The method of claim 65, wherein the at least a first and second DNA substrate molecule encodes all or part of a protein selected from Table I.
72. The method of claim 65, wherein the at least a first and second DNA substrate molecule comprises a gene cluster.
73. An evolved protein produced by the method of claim 65.
74. The method of claim 65, wherein at least one PCR primer differs from the at least a first and second substrate molecules in at least one nucleotide.
75. The method of claim 74, wherein the PCR primer comprises a nucleotide sequence of a known mutant or polymorphism of the at least a first or second substrate molecule.
76. The method of claim 75, wherein the PCR primer is degenerate and encodes the nucleotide sequences of more than one known mutant or polymorphism of the at least a first or second substrate molecule.
77. The method of claim 67, wherein mutagenesis comprises recursive sequence recombination.
78. The method of claim 65, wherein the products of (e) are subjected to mutagenesis.
79. The method of claim 78, wherein mutagenesis comprises recursive sequence recombination.
80. The method of claim 65, wherein the products of (e) are used as a DNA substrate molecule in (b).
81. The method of claim 65, wherein the recombinant DNA substrate molecule of (e) comprises a library of recombinant DNA substrate molecules.
82. A method for optimizing expression of a protein by evolving the protein, wherein the protein is encoded by a DNA substrate molecule, comprising:
(a) providing a set of oligonucleotides, wherein each oligonucleotide comprises at least two regions complementary to the DNA molecule and at least one degenerate region, each degenerate region encoding a region of an amino acid sequence of the protein;
(b) assembling the set of oligonucleotides into a library of full length genes;
(c) expressing the products of (b) in a host cell;
(d) screening the products of (c) for improved expression of the protein; and
(e) recovering a recombinant DNA substrate molecule encoding an evolved protein from (d).
83. The method of claim 82, wherein the primers comprise about 20 nucleotides complementary to the DNA substrate molecule followed by a second region of about 20 degenerate nucleotides of homology with the DNA substrate molecules followed by about 20 nucleotides complementary to the DNA substrate.
84. The method of claim 82, wherein the protein is bovine intestinal alkaline phosphatase.
85. The method of claim 84, wherein the oligonucleotides comprise one or more primers from Table II.
86. The method of claim 82, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
87. The method of claim 82, wherein the DNA molecule comprises a gene cluster.
88. The method of claim 82, wherein (a)-(e) are repeated.
89. The method of claim 82, wherein the oligonucleotides comprise at least 5′ and 3′ nucleotide complementary to the DNA substrate molecule and about 20-300 nucleotides having up to about 85% sequence homology with a region of the DNA substrate molecule.
90. The method of claim 89, wherein the oligonucleotides comprise a set of oligonucleotides in which each oligonucleotide overlaps with a second oligonucleotide.
91. The method of claim 82, wherein the products of (e) are subjected to mutagenesis.
92. The method of claim 91, wherein mutagenesis comprises recursive sequence recombination.
93. The method of claim 82, wherein the recombinant DNA substrate molecule of (e) comprises a library of recombinant DNA substrate molecules.
94. An evolved protein produced by the method of claim 82.
95. A method for optimizing expression of a protein encoded by a DNA substrate molecule by evolving the protein, wherein the DNA substrate molecule comprises at least one lac operator and a fusion of a DNA sequence encoding the protein with a DNA sequence encoding a lac headpiece dimer, the method comprising:
(a) transforming a host cell with a library of mutagenized DNA substrate molecules;
(b) inducing expression of the protein encoded by the library of (a);
(c) preparing an extract of the product of (b);
(d) fractionating insoluble protein from complexes of soluble protein and DNA; and
(e) recovering a DNA substrate molecule encoding an evolved protein from (d).
96. The method of claim 95, wherein (a)-(e) are repeated.
97. The method of claim 95, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
98. An evolved protein produced by the method of claim 95.
99. The method of claim 95, wherein the products of (e) are subjected to mutagenesis.
100. The method of claim 99, wherein mutagenesis comprises recursive sequence recombination.
101. The method of claim 95, wherein the products of (e) are used as a DNA substrate molecule in (a).
102. The method of claim 95, wherein the recombinant DNA substrate molecule of (e) comprises a library of recombinant DNA substrate molecules.
103. A method for evolving functional expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA sequence encoding filamentous phage protein to generate a fusion protein, the method comprising:
(a) providing a host cell producing infectious particles expressing a fusion protein encoded by a library of mutagenized DNA substrate molecules;
(b) recovering from (a) infectious particles displaying the fusion protein;
(c) affinity purifying particles displaying the mutant protein using a ligand for the protein; and
(d) recovering a DNA substrate molecule encoding an evolved protein from affinity purified particles of (c).
104. The method of claim 103, wherein (a)-(d) are repeated.
105. The method of claim 103, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
106. An evolved protein produced by the method of claim 103.
107. The method of claim 103, wherein the products of (d) are subjected to mutagenesis.
108. The method of claim 107, wherein mutagenesis comprises recursive sequence recombination.
109. The method of claim 107, wherein the products of claim 107 are used as a DNA substrate molecule in (a).
110. The method of claim 103, wherein the DNA substrate molecule of (e) comprises a library of DNA substrate molecules.
111. The method of claim 103, wherein DNA sequence encoding the filamentous phage protein comprises a phagemid.
112. The method of claim 103, wherein DNA sequence encoding the filamentous phage protein comprises a phage.
113. A method for optimizing expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA substrate encoding a lac headpiece dimer, wherein the DNA substrate molecule is present on a first plasmid vector, the method comprising:
(a) providing a host cell transformed with the first vector and a second vector comprising a library of mutants of at least one chaperonin geneand at least one lac operator;
(b) preparing an extract of the product of (a);
(c) fractionating insoluble protein from complexes of soluble protein and DNA; and
(d) recovering DNA encoding a chaperonin gene from (c).
114. The method of claim 113, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
115. The method of claim 113, wherein the DNA substrate is subjected to mutagenesis independently of the chaperonin gene prior to (a).
116. The method of claim 113, wherein the DNA of (d) comprises a library of mutants.
117. The method of claim 113, wherein the first and second vectors are the same vector.
118. The method of claim 113, wherein (d) further comprises recovering an evolved DNA substrate molecule from the products of (c).
119. An evolved chaperonin produced by the method of claim 113.
120. An evolved protein produced by the method of claim 113.
121. The method of claim 113, wherein (a)-(d) are repeated.
122. The method of claim 113, wherein the products of (d) are subjected to mutagenesis.
123. The method of claim 122, wherein mutagenesis comprises recursive sequence recombination.
124. The method of claim 122, wherein the products of claim 122 are used in (a).
125. A method for optimizing expression of a protein encoded by a DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a filamentous phage gene, wherein the fusion is carried on a phagemid comprising a library of chaperonin gene mutants, the method comprising:
(a) providing a host cell producing infectious particles expressing a fusion protein encoded by a library of mutagenized DNA substrate molecules;
(b) recovering from (a) infectious particles displaying the fusion protein;
(c) affinity purifying particles displaying the protein using a ligand for the protein; and
(d) recovering DNA encoding the mutant chaperonin from affinity purified particles of (c).
126. The method of claim 125, wherein (a)-(d) are repeated.
127. The method of claim 125, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
128. An evolved chaperonin produced by the method of claim 125.
129. An evolved protein produced by the method of claim 125.
130. The method of claim 125, wherein the products of (d) are subjected to mutagenesis.
131. The method of claim 130, wherein mutagenesis comprises recursive sequence recombination.
132. The method of claim 130, wherein the products of claim 130 are used in (a).
133. The method of claim 125, wherein the DNA of (d) comprises a library of DNA substrate molecules.
134. The method of claim 125, wherein the DNA substrate molecule comprises a library of mutagenized DNA sequences encoding the protein of interest.
135. The method of claim 125, wherein (d) further comprises recovering DNA encoding the protein from affinity purified particles of (c).
136. A method for optimizing secretion of a protein in a host by evolving a gene encoding a secretory function, comprising:
(a) providing a cluster of genes encoding secretory functions;
(b) recombining at least a first and second sequence in the gene cluster of (a) encoding a secretory function, the at least a first and second sequences differing from each other in at least one nucleotide, to generate a library of recombinant sequences;
(c) transforming a host cell culture with the products of (b), wherein the host cell comprises a DNA sequence encoding the protein;
(d) subjecting the product of (c) to screening or selection for secretion of the protein; and
(e) recovering DNA encoding an evolved gene encoding a secretory function from the product of (d).
137. The method of claim 136, wherein the gene cluster comprises at least one recognition site for a restriction endonuclease having nonpalindromic ends at the cleavage site.
138. The method of claim 136, wherein the host is E. coli., yeast, Bacillus, Pseudomonas, or a mammalian cell.
139. The method of claim 136, wherein the protein is a thermostable DNA polymerase.
140. The method of claim 136, wherein protein is inducibly expressed.
141. The method of claim 136, wherein the protein is linked to a secretory leader sequence.
142. A secretory gene evolved by the method of claim 136.
143. The method of claim 136, wherein (a)-(e) are repeated.
144. The method of claim 136, wherein the DNA sequence of (c) encodes all or part of a protein selected from Table I.
145. The method of claim 136, wherein the DNA sequence of (c) comprises a library of mutant sequences.
146. The method of claim 136, wherein the products of (e) are subjected to mutagenesis.
147. The method of claim 146, wherein mutagenesis comprises recursive sequence recombination.
148. The method of claim 146, wherein the products of claim 146 are used in (a).
149. The method of claim 136, wherein the DNA of (e) comprises a library of evolved genes.
150. A method for evolving an improved DNA polymerase comprising:
(a) providing a library of mutant DNA substrate molecules encoding mutant DNA polymerase;
(b) screening extracts of cells transfected with (a) and comparing activity with wild type DNA polymerase;
(c) recovering mutant DNA substrate molecules from cells in (b) expressing mutant DNA polymerase having improved activity over wild-type DNA polymerase; and
(d) recovering a DNA substrate molecule encoding an evolved polymerase from the products of (c).
151. The method of claim 150, wherein the improved activity is at least one of the group of higher quality sequencing ladder, less termination of reactions with inosine, improve acceptance of base analogs, improved acceptance of dideoxy nucleotides, and longer sequencing ladders.
152. The method of claim 150, wherein the products of (a) are expressed under control of arabinose promoter in an E. coli host having a mutant host DNA polymerase.
153. The method of claim 150, wherein (a)-(d) are repeated.
154. An evolved DNA polymerase produced by the method of claim 150.
155. The method of claim 150, wherein the products of (d) are subjected to mutagenesis.
156. The method of claim 155, wherein mutagenesis comprises recursive sequence recombination.
157. The method of claim 155, wherein the products of claim 155 are used in (a).
158. The method of claim 150, wherein the DNA substrate molecule of (d) comprises a library of DNA substrate molecules.
159. A method for evolving a DNA polymerase with an error rate greater than that of wild type DNA polymerase comprising:
(a) providing a library of mutant DNA substrate molecules encoding mutant DNA polymerase in a host cell comprising an indicator gene having a revertible mutation, wherein the indicator gene is replicated by the mutant DNA polymerase;
(b) screening the products of (a) for revertants of the indicator gene;
(c) recovering mutant DNA substrate molecules from revertants; and
(d) recovering a DNA substrate molecule encoding an evolved polymerase from the products of (c).
160. The method of claim 159, wherein the indicator gene is LacZalpha or GFP.
161. The method of claim 159 wherein the revertible mutation is a stop codon.
162. The method of claim 159, wherein the host cell comprises a mutant host DNA polymerase.
163. A method for evolving a DNA polymerase, comprising:
(a) providing a library of mutant DNA substrate molecules encoding mutant DNA polymerase, the library comprising a plasmid vector;
(b) preparing plasmid preparations and extracts of host cells transfected with the products of (a);
(c) amplifying each plasmid preparation in a PCR reaction using the mutant polymerase encoded by that plasmid, the polymerase being present in the host cell extract;
(d) recovering the PCR products of (c); and
(e) recovering a DNA substrate molecule encoding an evolved polymerase from the products of (d).
164. The method of claim 163, wherein the reaction of (c) is carried out in the presence of an organic solvent, a base analog, or inosine.
165. The method of claim 163, wherein (a)-(e) are repeated.
166. An evolved polymerase produced by the method of claim 163.
167. The method of claim 163, wherein the products of (e) are subjected to mutagenesis.
168. The method of claim 167, wherein mutagenesis comprises recursive sequence recombination.
169. The method of claim 167, wherein the products of claim 167 are used in (a).
170. The method of claim 163, wherein the DNA substrate molecule of (e) comprises a library of DNA substrate molecules.
171. A method for evolving a p-nitrophenol phosphonatase from a phosphonatase encoded by a DNA substrate molecule, comprising:
(a) providing library of mutants of the DNA substrate molecule, the library comprising a plasmid expression vector;
(b) transfecting a host, wherein the host phn operon is deleted;
(c) selecting for growth of the transfectants of (b) using a p-nitrophenol phosphonatase as a substrate;
(d) recovering the DNA substrate molecules from transfectants selected from (c); and
(e) recovering a DNA substrate molecule from (d) encoding an evolved phosphonatase.
172. The method of claim 171, wherein (a)-(e) are repeated.
173. The method of claim 171, wherein the phosphonatase is selected from the group consisting of beta-lactamase and alkyl phosphonatase.
174. An evolved p-nitrophenol phosphonatase produced by the method of claim 173.
175. The method of claim 171, wherein the products of (e) are subjected to mutagenesis.
176. The method of claim 175, wherein mutagenesis comprises recursive sequence recombination.
177. The method of claim 175, wherein the products of claim 175 are used in (a).
178. The method of claim 171, wherein the DNA substrate molecule of (e) comprises a library of DNA substrate molecules.
179. A method for evolving a protease encoded by a DNA substrate molecule comprising:
(a) providing library of mutants of the DNA substrate molecule, the library comprising a plasmid expression vector, wherein the DNA substrate molecule is linked to a secretory leader;
(b) transfecting a host;
(c) selecting for growth of the transfectants of (b) on a complex protein medium; and
(d) recovering a DNA substrate molecule from (c) encoding an evolved protease.
180. The method of claim 179, wherein (a)-(d) are repeated.
181. An evolved subtilisin produced by the method of claim 179.
182. The method of claim 179, wherein the products of (d) are subjected to mutagenesis.
183. The method of claim 182, wherein mutagenesis comprises recursive sequence recombination.
184. The method of claim 182, wherein the products of claim 184 are used in (a).
185. The method of claim 179, wherein the DNA substrate molecule of (d) comprises a library of DNA substrate molecules.
186. The method of claim 179, wherein the protease is a subtilisin.
187. A method for screening a library of protease mutants displayed on a phage to obtain an improved protease, wherein a DNA substrate molecule encoding the protease is fused to DNA encoding a filamentous phage protein to generate a fusion protein, comprising:
(a) providing host cells expressing the fusion protein;
(b) overlaying host cells with a protein net to entrap the phage;
(c) washing the product of (b) to recover phage liberated by digestion of the protein net;
(d) recovering DNA from the product of (c); and
(e) recovering a DNA substrate from (d) encoding an improved protease.
188. The method of claim 187, wherein (a)-(e) are repeated.
189. An evolved protease produced by the method of claim 187.
190. The method of claim 187, wherein the products of (e) are subjected to mutagenesis.
191. The method of claim 190, wherein mutagenesis comprises recursive sequence recombination.
192. The method of claim 190, wherein the products of claim 190 are used in (a).
193. The method of claim 187, wherein the DNA substrate molecule of (e) comprises a library of DNA substrate molecules.
194. A method for screening a library of protease mutants to obtain an improved protease, the method comprising:
(a) providing a library of peptide substrates, the peptide substrate comprising a fluorophore and a fluorescence quencher;
(b) screening the library of protease mutants for ability to cleave the peptide substrates, wherein fluorescence is measured; and
(c) recovering DNA encoding at least one protease mutant from (b).
195. A method for evolving an alpha interferon gene comprising:
(a) providing a library of mutant alpha interferon genes, the library comprising a filamentous phage vector;
(b) stimulating cells comprising a reporter construct, the reporter construct comprising a reporter gene under control of an interferon responsive promoter, and wherein the reporter gene is GFP;
(c) separating the cells expressing GFP by FACS;
(d) recovering phage from the product of (c); and
(e) recovering an evolved interferon gene from the product of (d).
196. The method of claim 195, wherein the interferon responsive promoter is an MHC I promoter.
197. The method of claim 195, wherein (a)-(e) are repeated.
198. An evolved interferon produced by the method of claim 195.
199. The method of claim 195, wherein the products of (e) are subjected to mutagenesis.
200. The method of claim 199, wherein mutagenesis comprises recursive sequence recombination.
201. The method of claim 199, wherein the products of claim 199 are used in (a).
202. The method of claim 195, wherein the evolved interferon gene of (e) comprises a library of genes.
203. A method for screening a library of mutants of a DNA substrate encoding a protein for an evolved DNA substrate, comprising:
(a) providing a library of mutants, the library comprising an expression vector;
(b) transfecting a mammalian host cell with the library of (a), wherein mutant protein is expressed on the surface of the cell;
(c) screening or selecting the products of (b) with a ligand for the protein;
(d) recovering DNA encoding mutant protein from the products of (c); and
(e) recovering an evolved DNA substrate from the products of (d).
204. The method of claim 203, wherein the ligand is an antibody.
205. The method of claim 203, wherein the ligand is a substrate and the protein is an enzyme.
206. The method of claim 203, wherein the expression vector comprises an SV40 origin and the host cell is a Cos cell.
207. The method of claim 203, wherein the mutant protein is expressed transiently.
208. The method of claim 203, wherein the host cell further comprises SV40 large T antigen.
209. The method of claim 203, wherein the protein is an antibody.
210. The method of claim 203, wherein (a)-(e) are repeated.
211. The method of claim 203, wherein the DNA substrate molecule encodes all or part of a protein selected from Table I.
212. An evolved protein produced by the method of claim 203.
213. The method of claim 203, wherein the products of (e) are subjected to mutagenesis.
214. The method of claim 213, wherein mutagenesis comprises recursive sequence recombination.
215. The method of claim 213, wherein the products of claim 213 are used in (a).
216. The method of claim 203, wherein the DNA substrate molecule of (e) comprises a library of DNA substrate molecules.
217. A method for evolving a DNA substrate molecule encoding an interferon alpha, comprising:
(a) providing a library of mutant alpha interferon genes, the library comprising an expression vector wherein the alpha interferon genes are expressed under the control of an inducible promoter;
(b) transfecting host cells with the library of (a);
(c) contacting the product of (b) with a virus;
(d) recovering DNA encoding a mutant alpha interferon from host cells surviving step (c); and
(e) recovering an evolved interferon gene from the product of (d).
218. The method of claim 217, wherein the promoter is a metallothionein promoter.
219. The method of claim 217, wherein the virus is HIV.
220. The method of claim 217, wherein the virus further comprises a conditionally lethal gene.
221. The method of claim 217, wherein the conditionally lethal gene is thymidine kinase.
222. The method of claim 217, wherein the transfected cells are exposed to conditionally lethal selective conditions.
223. The method of claim 217, wherein (a)-(e) are repeated.
224. An evolved IFNα polymerase produced by the method of claim 217.
225. The method of claim 217, wherein the products of (e) are subjected to mutagenesis.
226. The method of claim 225, wherein mutagenesis comprises recursive sequence recombination.
227. The method of claim 225, wherein the products of claim 218 are used in (a).
228. The method of claim 217, wherein the DNA substrate molecule of (e) comprises a library of DNA substrate molecules.
229. A method for evolving the stability of a protein encoded by a DNA substrate molecule, the DNA substrate molecule comprising a fusion of a DNA sequence encoding the protein with a DNA sequence encoding a filamentous phage protein to generate a fusion protein, the method comprising:
(a) providing a host cell expressing a library of mutants of the fusion protein;
(b) affinity purifying the mutants with a ligand for the protein, wherein the ligand is a human serum protein, tissue specific protein, or receptor;
(c) recovering DNA encoding a mutant protein from the affinity selected mutants of (b); and
(d) recovering an evolved gene encoding the protein from the product of (c).
230. The method of claim 229, wherein the serum protein is serum albumin, immunoglobulin, lipoprotein, haptoglobin, fibrinogen, transferrin, alpha-1 anti-trypsin, or alpha -2 macroglobulin.
231. The method of claim 229, wherein the DNA sequence encoding the filamentous phage protein comprises a phage.
232. The method of claim 229, wherein the DNA sequence encoding the filamentous phage protein comprises a phagemid.
233. The method of claim 229, wherein the products of step (a) are derivitized with a half-life extending moiety.
234. The method of claim 229, wherein the moiety is polyethylene glycol.
235. The method of claim 229, wherein the DNA substrate molecule comprises a fusion of nucleic acid encoding the protein with nucleic acid encoding an epitope tag.
236. The method of claim 235, wherein the products of (a) are contacted with a protease prior to (b).
237. The method of claim 235, wherein the ligand is an antibody specific for the epitope tag.
238. The method of claim 229, wherein the protein is selected from Table I.
239. The method of claim 229, wherein the products of (a) are subjected to heat, metal ions, non-physiological pH, lyophilization, or freeze-thawing before (b).
240. The method of claim 229, wherein (a)-(e) are repeated.
241. An evolved polymerase produced by the method of claim 229.
242. The method of claim 229, wherein the products of (d) are subjected to mutagenesis.
243. The method of claim 242, wherein mutagenesis comprises recursive sequence recombination.
244. The method of claim 242, wherein the products of claim 242 are used in (a).
245. The method of claim 229, wherein the evolved gene of (d) comprises a library of DNA substrate molecules.
246. A method for evolving a protein having at least two subunits, comprising:
(a) providing a library of mutant DNA substrate molecules for each subunit;
(b) recombining the libraries into a library of single chain constructs of the protein, the single chain construct comprising a DNA substrate molecule encoding each subunit sequence, the subunit sequence being linked by a linker at a nucleic acid sequence encoding the amino terminus of one subunit to a nucleic acid sequence encoding the carboxy terminus of a second subunit;
(c) screening or selecting the products of (b),
(d) recovering recombinant single chain construct DNA substrate molecules from the products of (c);
(e) subjecting the products of (d) to mutagenesis; and
(f) recovering an evolved single chain construct DNA substrate molecule from (e).
247. The method of claim 246, wherein the products of (b) are displayed on a phage.
248. The method of claim 246, wherein the protein is selected from Table I.
249. The method of claim 246, wherein (a)-(f) are repeated.
250. An evolved protein produced by the method of claim 246.
251. The method of claim 246, wherein the products of (f) are subjected to mutagenesis.
252. The method of claim 246, wherein mutagenesis comprises recursive sequence recombination.
253. The method of claim 246, wherein the products of claim 246 are used in (a).
254. The method of claim 246, wherein the evolved DNA substrate molecule of (f) comprises a library of DNA substrate molecules.
255. A method for evolving the coupling of a mammalian 7-transmembrane receptor to a yeast signal transduction pathway, comprising:
(a) expressing a library of mammalian G alpha protein mutants in a host yeast cell, wherein the host cell expresses the mammalian 7-transmembrane receptor and a reporter gene, the receptor gene geing expressed under control of a yeast pheromone responsive promoter;
(b) screening or selecting the products of (a) for expression of the reporter gene in the presence of a ligand for the 7-transmembrance receptor; and
(c) recovering DNA encoding an evolved G alpha protein mutant from screened or selected products of (b).
256. The method of claim 255, wherein the products of (c) are subjected to mutagenesis.
257. The method of claim 256, wherein mutagenesis comprises recursive sequence recombination.
258. The method of claim 255, wherein the products of claim 255 are used in (a).
259. The method of claim 255, wherein (a)-(c) are repeated.
260. An evolved G alpha protein produced by the method of claim 255.
261. The method of claim 255, wherein the reporter gene is luciferase.
262. The method of claim 255, wherein the pheromone responsive promoter is positively regulated by GA L4 and wherein GAL4 is expressed under the control of a pheromone sensitive, GAL4 enhanced promoter.
263. A method for recombining at least a first and second DNA substrate molecule, comprising:
(a) transfecting a host cell with at least a first and second DNA substrate molecule wherein the at least a first and second DNA substrate molecules are recombined in the host cell;
(b) screening or selecting the products of (a) for a desired property; and
(c) recovering recombinant DNA substrate molecules from (b).
264. The method of claim 263, wherein the products of (c) are subjected to mutagenesis.
265. The method of claim 264, wherein the mutagenesis comprises recursive sequence recombination.
266. The method of claim 263, wherein (a)-(c) are repeated.
267. The method of claim 263, wherein the products of claim 263 are used in (a).
268. A method for evolving a DNA substrate sequence encoding a protein of interest, wherein the DNA substrate comprises a vector, the vector comprising single-stranded DNA, the method comprising:
(a) providing single-stranded vector DNA and a library of mutants of the DNA substrate sequence;
(b) annealing denatured double-stranded DNA from the library of (a) to the single stranded vector DNA of (a);
(c) transforming the products of (b) into a host;
(d) screening the product of (c) for a desired property; and
(e) recovering evolved DNA substrate DNA from the products of (d).
269. The method of claim 268, wherein the product of (e) is subjected to mutagenesis.
270. The method of claim 269, wherein mutagenesis comprises recursive sequence recombination.
271. The method of claim 269, wherein the product of claim 269 is used in (a).
272. The method of claim 268, wherein the host is a mutS host.
273. The method of claim 268, wherein the vector is a phagemid.
US10/667,772 1995-02-17 2003-09-22 Methods and compositions for polypeptide engineering Abandoned US20040214277A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/667,772 US20040214277A1 (en) 1996-12-18 2003-09-22 Methods and compositions for polypeptide engineering
US11/136,115 US20060166225A1 (en) 1996-12-18 2005-05-24 Methods and compositions for polypeptide engineering
US11/198,765 US20060084091A1 (en) 1996-12-18 2005-08-05 Methods and compositions for polypeptide engineering
US11/286,271 US20060223143A1 (en) 1995-02-17 2005-11-23 Methods and compositions for polypeptide engineering

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/769,062 US6335160B1 (en) 1995-02-17 1996-12-18 Methods and compositions for polypeptide engineering
US09/339,926 US6653072B1 (en) 1996-12-18 1999-06-24 Methods and compositions for polypeptide engineering
US10/667,772 US20040214277A1 (en) 1996-12-18 2003-09-22 Methods and compositions for polypeptide engineering

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/339,926 Continuation US6653072B1 (en) 1994-02-17 1999-06-24 Methods and compositions for polypeptide engineering

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US11/136,115 Continuation US20060166225A1 (en) 1996-12-18 2005-05-24 Methods and compositions for polypeptide engineering
US11/198,765 Continuation US20060084091A1 (en) 1996-12-18 2005-08-05 Methods and compositions for polypeptide engineering
US11/286,271 Continuation US20060223143A1 (en) 1995-02-17 2005-11-23 Methods and compositions for polypeptide engineering

Publications (1)

Publication Number Publication Date
US20040214277A1 true US20040214277A1 (en) 2004-10-28

Family

ID=25084330

Family Applications (20)

Application Number Title Priority Date Filing Date
US08/769,062 Expired - Lifetime US6335160B1 (en) 1994-02-17 1996-12-18 Methods and compositions for polypeptide engineering
US09/339,913 Expired - Lifetime US6303344B1 (en) 1996-12-18 1999-06-24 Methods and compositions for polypeptide engineering
US09/344,002 Expired - Lifetime US6355484B1 (en) 1994-02-17 1999-06-24 Methods and compositions for polypeptides engineering
US09/339,926 Expired - Fee Related US6653072B1 (en) 1994-02-17 1999-06-24 Methods and compositions for polypeptide engineering
US09/339,904 Expired - Lifetime US6319713B1 (en) 1994-02-17 1999-06-25 Methods and compositions for polypeptide engineering
US09/559,565 Expired - Fee Related US6455253B1 (en) 1996-12-18 2000-04-27 Methods and compositions for polypeptide engineering
US09/559,671 Expired - Fee Related US6613514B2 (en) 1994-02-17 2000-04-27 Methods and compositions for polypeptide engineering
US09/693,350 Expired - Fee Related US6579678B1 (en) 1996-12-18 2000-10-20 Methods and compositions for polypeptide engineering
US09/693,389 Expired - Fee Related US6586182B1 (en) 1996-12-18 2000-10-20 Methods and compositions for polypeptide engineering
US09/954,692 Expired - Fee Related US6946296B2 (en) 1995-11-30 2001-09-12 Methods and compositions for polypeptide engineering
US10/646,221 Expired - Fee Related US7534564B2 (en) 1995-02-17 2003-08-22 Methods and compositions for polypeptide engineering
US10/667,868 Abandoned US20050059109A1 (en) 1994-02-17 2003-09-22 Methods and compositions for polypeptide engineering
US10/667,772 Abandoned US20040214277A1 (en) 1995-02-17 2003-09-22 Methods and compositions for polypeptide engineering
US11/136,115 Abandoned US20060166225A1 (en) 1996-12-18 2005-05-24 Methods and compositions for polypeptide engineering
US11/198,765 Abandoned US20060084091A1 (en) 1996-12-18 2005-08-05 Methods and compositions for polypeptide engineering
US11/286,271 Abandoned US20060223143A1 (en) 1995-02-17 2005-11-23 Methods and compositions for polypeptide engineering
US11/515,879 Abandoned US20070031878A1 (en) 1995-02-17 2006-09-06 Methods and compositions for polypeptide engineering
US12/069,011 Expired - Fee Related US7776598B2 (en) 1996-12-18 2008-02-05 Methods and compositions for polypeptide engineering
US12/557,983 Abandoned US20110053781A1 (en) 1996-12-18 2009-09-11 Methods and compositions for polypeptide engineering
US13/224,145 Abandoned US20120015820A1 (en) 1996-12-18 2011-09-01 Methods and compositions for polypeptide engineering

Family Applications Before (12)

Application Number Title Priority Date Filing Date
US08/769,062 Expired - Lifetime US6335160B1 (en) 1994-02-17 1996-12-18 Methods and compositions for polypeptide engineering
US09/339,913 Expired - Lifetime US6303344B1 (en) 1996-12-18 1999-06-24 Methods and compositions for polypeptide engineering
US09/344,002 Expired - Lifetime US6355484B1 (en) 1994-02-17 1999-06-24 Methods and compositions for polypeptides engineering
US09/339,926 Expired - Fee Related US6653072B1 (en) 1994-02-17 1999-06-24 Methods and compositions for polypeptide engineering
US09/339,904 Expired - Lifetime US6319713B1 (en) 1994-02-17 1999-06-25 Methods and compositions for polypeptide engineering
US09/559,565 Expired - Fee Related US6455253B1 (en) 1996-12-18 2000-04-27 Methods and compositions for polypeptide engineering
US09/559,671 Expired - Fee Related US6613514B2 (en) 1994-02-17 2000-04-27 Methods and compositions for polypeptide engineering
US09/693,350 Expired - Fee Related US6579678B1 (en) 1996-12-18 2000-10-20 Methods and compositions for polypeptide engineering
US09/693,389 Expired - Fee Related US6586182B1 (en) 1996-12-18 2000-10-20 Methods and compositions for polypeptide engineering
US09/954,692 Expired - Fee Related US6946296B2 (en) 1995-11-30 2001-09-12 Methods and compositions for polypeptide engineering
US10/646,221 Expired - Fee Related US7534564B2 (en) 1995-02-17 2003-08-22 Methods and compositions for polypeptide engineering
US10/667,868 Abandoned US20050059109A1 (en) 1994-02-17 2003-09-22 Methods and compositions for polypeptide engineering

Family Applications After (7)

Application Number Title Priority Date Filing Date
US11/136,115 Abandoned US20060166225A1 (en) 1996-12-18 2005-05-24 Methods and compositions for polypeptide engineering
US11/198,765 Abandoned US20060084091A1 (en) 1996-12-18 2005-08-05 Methods and compositions for polypeptide engineering
US11/286,271 Abandoned US20060223143A1 (en) 1995-02-17 2005-11-23 Methods and compositions for polypeptide engineering
US11/515,879 Abandoned US20070031878A1 (en) 1995-02-17 2006-09-06 Methods and compositions for polypeptide engineering
US12/069,011 Expired - Fee Related US7776598B2 (en) 1996-12-18 2008-02-05 Methods and compositions for polypeptide engineering
US12/557,983 Abandoned US20110053781A1 (en) 1996-12-18 2009-09-11 Methods and compositions for polypeptide engineering
US13/224,145 Abandoned US20120015820A1 (en) 1996-12-18 2011-09-01 Methods and compositions for polypeptide engineering

Country Status (12)

Country Link
US (20) US6335160B1 (en)
EP (5) EP0946755A4 (en)
JP (2) JP5008784B2 (en)
KR (1) KR20000069591A (en)
AT (1) ATE481480T1 (en)
AU (1) AU732146B2 (en)
CA (2) CA2274319C (en)
DE (1) DE69739996D1 (en)
DK (2) DK1149905T3 (en)
ES (1) ES2351673T3 (en)
IL (1) IL130190A0 (en)
WO (1) WO1998027230A1 (en)

Families Citing this family (428)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7045289B2 (en) 1991-09-09 2006-05-16 Third Wave Technologies, Inc. Detection of RNA Sequences
US6759226B1 (en) 2000-05-24 2004-07-06 Third Wave Technologies, Inc. Enzymes for the detection of specific nucleic acid sequences
US7150982B2 (en) 1991-09-09 2006-12-19 Third Wave Technologies, Inc. RNA detection assays
US20060257890A1 (en) 1996-05-20 2006-11-16 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US6309883B1 (en) 1994-02-17 2001-10-30 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
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
US6335160B1 (en) 1995-02-17 2002-01-01 Maxygen, Inc. Methods and compositions for polypeptide engineering
US6238884B1 (en) 1995-12-07 2001-05-29 Diversa Corporation End selection in directed evolution
US6358709B1 (en) 1995-12-07 2002-03-19 Diversa Corporation End selection in directed evolution
US6939689B2 (en) 1995-12-07 2005-09-06 Diversa Corporation Exonuclease-mediated nucleic acid reassembly in directed evolution
US6537776B1 (en) * 1999-06-14 2003-03-25 Diversa Corporation Synthetic ligation reassembly in directed evolution
US6740506B2 (en) 1995-12-07 2004-05-25 Diversa Corporation End selection in directed evolution
US6764835B2 (en) 1995-12-07 2004-07-20 Diversa Corporation Saturation mutageneis in directed evolution
US6713279B1 (en) 1995-12-07 2004-03-30 Diversa Corporation Non-stochastic generation of genetic vaccines and enzymes
US5830696A (en) 1996-12-05 1998-11-03 Diversa Corporation Directed evolution of thermophilic enzymes
US6242211B1 (en) 1996-04-24 2001-06-05 Terragen Discovery, Inc. Methods for generating and screening novel metabolic pathways
AU743305C (en) 1997-01-17 2006-03-30 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
GB9701425D0 (en) * 1997-01-24 1997-03-12 Bioinvent Int Ab A method for in vitro molecular evolution of protein function
US6159687A (en) 1997-03-18 2000-12-12 Novo Nordisk A/S Methods for generating recombined polynucleotides
US6153410A (en) * 1997-03-25 2000-11-28 California Institute Of Technology Recombination of polynucleotide sequences using random or defined primers
GB9722131D0 (en) * 1997-10-20 1997-12-17 Medical Res Council Method
AU1124499A (en) 1997-10-28 1999-05-17 Maxygen, Inc. Human papillomavirus vectors
EP1030861A4 (en) 1997-10-31 2001-09-05 Maxygen Inc Modification of virus tropism and host range by viral genome shuffling
CA2313380C (en) 1997-12-08 2008-12-30 California Institute Of Technology Method for creating polynucleotide and polypeptide sequences
US6468745B1 (en) 1998-01-16 2002-10-22 Large Scale Biology Corporation Method for expressing a library of nucleic acid sequence variants and selecting desired traits
US6303848B1 (en) 1998-01-16 2001-10-16 Large Scale Biology Corporation Method for conferring herbicide, pest, or disease resistance in plant hosts
US6426185B1 (en) 1998-01-16 2002-07-30 Large Scale Biology Corporation Method of compiling a functional gene profile in a plant by transfecting a nucleic acid sequence of a donor plant into a different host plant in an anti-sense orientation
US6759243B2 (en) * 1998-01-20 2004-07-06 Board Of Trustees Of The University Of Illinois High affinity TCR proteins and methods
EP2189791A3 (en) 1998-02-04 2011-03-09 Life Technologies Corporation Microarrays and uses therefor
US7390619B1 (en) * 1998-02-11 2008-06-24 Maxygen, Inc. Optimization of immunomodulatory properties of genetic vaccines
DE1073770T1 (en) * 1998-04-30 2002-07-04 Max Planck Gesellschaft Novel method for the identification of clones with a desired biological property, starting from an expression gene bank (2001/06)
EP1073670A1 (en) 1998-05-01 2001-02-07 Maxygen, Inc. Optimization of pest resistance genes using dna shuffling
US6902918B1 (en) 1998-05-21 2005-06-07 California Institute Of Technology Oxygenase enzymes and screening method
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
CA2335269A1 (en) 1998-06-17 1999-12-23 Maxygen, Inc. Method for producing polynucleotides with desired properties
US6846655B1 (en) 1998-06-29 2005-01-25 Phylos, Inc. Methods for generating highly diverse libraries
US20030104417A1 (en) * 1998-08-12 2003-06-05 Proteus S.A. Template-mediated, ligation-oriented method of nonrandomly shuffling polynucleotides
US20030092023A1 (en) * 1998-08-12 2003-05-15 Daniel Dupret Method of shuffling polynucleotides using templates
WO2000009682A1 (en) 1998-08-12 2000-02-24 Maxygen, Inc. Dna shuffling of monooxygenase genes for production of industrial chemicals
US6991922B2 (en) * 1998-08-12 2006-01-31 Proteus S.A. Process for in vitro creation of recombinant polynucleotide sequences by oriented ligation
FR2782323B1 (en) * 1998-08-12 2002-01-11 Proteus PROCESS FOR THE IN VITRO PRODUCTION OF RECOMBINANT POLYNUCLEOTIDE SEQUENCES, SEQUENCE BANKS AND SEQUENCES THUS OBTAINED
IL140442A0 (en) * 1998-08-12 2002-02-10 Maxygen Inc Dna shuffling to produce herbicide selective crops
US20040191772A1 (en) * 1998-08-12 2004-09-30 Dupret Daniel Marc Method of shuffling polynucleotides using templates
US6951719B1 (en) 1999-08-11 2005-10-04 Proteus S.A. Process for obtaining recombined nucleotide sequences in vitro, libraries of sequences and sequences thus obtained
AU6510799A (en) * 1998-10-07 2000-04-26 Maxygen, Inc. Dna shuffling to produce nucleic acids for mycotoxin detoxification
EP1129184A1 (en) 1998-11-10 2001-09-05 Maxygen, Inc. Modified adp-glucose pyrophosphorylase for improvement and optimation of plant phenotypes
US20040005673A1 (en) * 2001-06-29 2004-01-08 Kevin Jarrell System for manipulating nucleic acids
JP2002537762A (en) * 1999-01-05 2002-11-12 トラスティーズ オブ ボストン ユニバーシティ Improved nucleic acid cloning
US6358712B1 (en) 1999-01-05 2002-03-19 Trustee Of Boston University Ordered gene assembly
AU2415200A (en) * 1999-01-18 2000-08-01 Maxygen, Inc. Methods of populating data structures for use in evolutionary simulations
US6436675B1 (en) 1999-09-28 2002-08-20 Maxygen, Inc. Use of codon-varied oligonucleotide synthesis for synthetic shuffling
US6917882B2 (en) 1999-01-19 2005-07-12 Maxygen, Inc. Methods for making character strings, polynucleotides and polypeptides having desired characteristics
US6376246B1 (en) 1999-02-05 2002-04-23 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
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
US7024312B1 (en) 1999-01-19 2006-04-04 Maxygen, Inc. Methods for making character strings, polynucleotides and polypeptides having desired characteristics
IL137868A0 (en) 1999-01-19 2001-10-31 Maxygen Inc Oligonucleotide mediated nucleic acid recombination
US6961664B2 (en) 1999-01-19 2005-11-01 Maxygen Methods of populating data structures for use in evolutionary simulations
US20090130718A1 (en) * 1999-02-04 2009-05-21 Diversa Corporation Gene site saturation mutagenesis
IL138206A (en) * 1999-02-04 2011-06-30 Verenium Corp Methods for non-stochastic generation of progeny polypeptides and hybrid polynucleotides
CA2361384A1 (en) 1999-02-11 2000-08-17 Sun Ai Raillard High throughput mass spectrometry
US6531316B1 (en) 1999-03-05 2003-03-11 Maxyag, Inc. Encryption of traits using split gene sequences and engineered genetic elements
AU3879300A (en) * 1999-03-09 2000-09-28 Diversa Corporation End selection in directed evolution
DE60041642D1 (en) * 1999-03-18 2009-04-09 Complete Genomics As METHODS OF CLONING AND MANUFACTURING CHAIN FRAGMENTS WITH SAFE CONTENTS OF INFORMATION
AU2005225057A1 (en) * 1999-03-26 2005-12-01 Bp Corporation North America Inc. Synthetic ligation reassembly in directed evolution
AU4211600A (en) * 1999-04-10 2000-11-14 Maxygen, Inc. Modified lipid production
WO2001004287A1 (en) * 1999-07-07 2001-01-18 Maxygen Aps A method for preparing modified polypeptides
WO2001004306A1 (en) * 1999-07-12 2001-01-18 Genentech, Inc. Expression vectors and methods
EP1208193A1 (en) * 1999-08-12 2002-05-29 Maxygen, Inc. Dna shuffling of dioxygenase genes for production of industrial chemicals
SK288086B6 (en) 1999-08-12 2013-06-03 Pfizer Products Inc. Polynucleotide molecule comprising mutated nucleotide sequence of Streptomyces avermitilis aveC allele, vector and host cell, method of making strain of S. avermitilis and process for producing avermectins and final composition
US20040002474A1 (en) * 1999-10-07 2004-01-01 Maxygen Inc. IFN-alpha homologues
US7430477B2 (en) 1999-10-12 2008-09-30 Maxygen, Inc. Methods of populating data structures for use in evolutionary simulations
WO2001029212A1 (en) * 1999-10-19 2001-04-26 Enchira Biotechnology Corporation Methods for chimeragenesis of whole genomes or large polynucleotides
AU1098001A (en) * 1999-10-19 2001-04-30 Enchira Biotechnology Corporation Method for directed evolution by random chimeragenesis on transient templates
AU1456101A (en) * 1999-11-03 2001-05-14 Maxygen, Inc. Antibody diversity generation
US6686515B1 (en) 1999-11-23 2004-02-03 Maxygen, Inc. Homologous recombination in plants
US7115712B1 (en) 1999-12-02 2006-10-03 Maxygen, Inc. Cytokine polypeptides
WO2001048478A1 (en) * 1999-12-27 2001-07-05 Toray Industries, Inc. Cytokine-like peptide
AU2788101A (en) * 2000-01-11 2001-07-24 Maxygen, Inc. Integrated systems and methods for diversity generation and screening
AU2001241939A1 (en) * 2000-02-28 2001-09-12 Maxygen, Inc. Single-stranded nucleic acid template-mediated recombination and nucleic acid fragment isolation
KR20040010041A (en) 2000-05-04 2004-01-31 예일 유니버시티 High density protein arrays for screening of protein activity
US6479262B1 (en) 2000-05-16 2002-11-12 Hercules, Incorporated Solid phase enzymatic assembly of polynucleotides
JP2004513878A (en) 2000-06-23 2004-05-13 マキシジェン, インコーポレイテッド Novel costimulatory molecules
US7074590B2 (en) 2000-06-23 2006-07-11 Maxygen, Inc. Chimeric promoters
US7846733B2 (en) * 2000-06-26 2010-12-07 Nugen Technologies, Inc. Methods and compositions for transcription-based nucleic acid amplification
US6994963B1 (en) 2000-07-10 2006-02-07 Ambion, Inc. Methods for recombinatorial nucleic acid synthesis
US6858422B2 (en) 2000-07-13 2005-02-22 Codexis, Inc. Lipase genes
AU2001273559A1 (en) * 2000-07-18 2002-01-30 Enchira Biotechnology Corporation Methods of ligation mediated chimeragenesis utilizing populations of scaffold and donor nucleic acids
US7435562B2 (en) * 2000-07-21 2008-10-14 Modular Genetics, Inc. Modular vector systems
US20030017552A1 (en) * 2000-07-21 2003-01-23 Jarrell Kevin A. Modular vector systems
WO2002010183A1 (en) 2000-07-31 2002-02-07 Menzel, Rolf Compositions and methods for directed gene assembly
AUPQ974900A0 (en) * 2000-08-29 2000-09-21 Macquarie Research Limited Degenerate oligonucleotide gene-shuffling
US7408041B2 (en) 2000-12-08 2008-08-05 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
AU2002246632B2 (en) 2000-12-08 2007-04-05 Alexion Pharmaceuticals, Inc. Chronic lymphocytic leukemia cell line and its use for producing an antibody
US20060057651A1 (en) 2000-12-08 2006-03-16 Bowdish Katherine S Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
US6958213B2 (en) * 2000-12-12 2005-10-25 Alligator Bioscience Ab Method for in vitro molecular evolution of protein function
EP1427847B1 (en) * 2000-12-13 2010-07-28 Nugen Technologies, Inc. Methods and compositions for generation of multiple copies of nucleic acid sequences and methods of detection thereof
US20020086292A1 (en) * 2000-12-22 2002-07-04 Shigeaki Harayama Synthesis of hybrid polynucleotide molecules using single-stranded polynucleotide molecules
US20030036641A1 (en) * 2001-01-31 2003-02-20 Padgett Hal S. Methods for homology-driven reassembly of nucleic acid sequences
US20110111413A1 (en) * 2001-02-02 2011-05-12 Padgett Hal S Method of optimizing codon usage through dna shuffling
CA2440560A1 (en) * 2001-03-07 2002-09-19 Xenogen Corporation Methods of screening for introduction of dna into a target cell
WO2002072772A2 (en) 2001-03-09 2002-09-19 Nugen Technologies, Inc. Methods and compositions for amplification of rna sequences
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
WO2002083868A2 (en) 2001-04-16 2002-10-24 California Institute Of Technology Peroxide-driven cytochrome p450 oxygenase variants
AU2002254773B2 (en) * 2001-05-03 2005-12-08 Rensselaer Polytechnic Institute Novel methods of directed evolution
EP1412495A2 (en) * 2001-05-10 2004-04-28 Novozymes A/S A method for producing recombined polynucleotides
EP1421203A4 (en) * 2001-05-17 2005-06-01 Diversa Corp Novel antigen binding molecules for therapeutic, diagnostic, prophylactic, enzymatic, industrial, and agricultural applications, and methods for generating and screening thereof
WO2002099080A2 (en) * 2001-06-05 2002-12-12 Gorilla Genomics, Inc. Methods for low background cloning of dna using long oligonucleotides
WO2003008563A2 (en) 2001-07-20 2003-01-30 California Institute Of Technology Improved cytochrome p450 oxygenases
EP1409666B1 (en) * 2001-07-23 2006-03-29 DSM IP Assets B.V. Process for preparing variant polynucleotides
ATE365211T1 (en) * 2001-08-20 2007-07-15 Regenesis Bioremediation Produ BIOSENSOR FOR SMALL MOLECULAR ANALYTES
US20040005709A1 (en) * 2001-10-24 2004-01-08 Hoogenboom Henricus Renerus Jacobus Mattheus Hybridization control of sequence variation
US20040009498A1 (en) * 2002-01-14 2004-01-15 Diversa Corporation Chimeric antigen binding molecules and methods for making and using them
US20050075298A1 (en) * 2002-01-31 2005-04-07 Wei Chen Methods and composition for delivering nucleic acids and/or proteins to the intestinal mucosa
US20040043003A1 (en) * 2002-01-31 2004-03-04 Wei Chen Clinical grade vectors based on natural microflora for use in delivering therapeutic compositions
US20040009937A1 (en) * 2002-01-31 2004-01-15 Wei Chen Methods and composition for delivering nucleic acids and/or proteins to the respiratory system
WO2003072054A2 (en) * 2002-02-25 2003-09-04 Cabot Corporation Custom ligand design for biomolecular filtration and purification for bioseperation
ES2564570T3 (en) 2002-03-01 2016-03-23 Codexis Mayflower Holdings, Llc Methods, systems and software for the identification of functional biomolecules
US7747391B2 (en) * 2002-03-01 2010-06-29 Maxygen, Inc. Methods, systems, and software for identifying functional biomolecules
US20050084907A1 (en) 2002-03-01 2005-04-21 Maxygen, Inc. Methods, systems, and software for identifying functional biomolecules
US7620500B2 (en) 2002-03-09 2009-11-17 Maxygen, Inc. Optimization of crossover points for directed evolution
DE60323067D1 (en) * 2002-03-11 2008-10-02 Nugen Technologies Inc METHOD FOR GENERATING DOUBLE-STRENGTHED DNA WITH A 3'-SINGLE STRING PART AND USE THESE COMPLEXES FOR RECOMBINATION
BR0309391A (en) 2002-04-19 2005-10-25 Diversa Corp Phospholipases, nucleic acids encoding them and methods for preparing 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
ATE356871T1 (en) * 2002-05-17 2007-04-15 Alligator Bioscience Ab A METHOD FOR IN VITRO MOLECULAR EVOLUTION OF PROTEIN FUNCTION
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
CA2494798A1 (en) 2002-08-06 2005-01-13 Verdia, Inc. Ap1 amine oxidase variants
US7611700B2 (en) * 2002-09-09 2009-11-03 Hanall Pharmaceuticals, Co., Ltd. Protease resistant modified interferon alpha polypeptides
US7563600B2 (en) 2002-09-12 2009-07-21 Combimatrix Corporation Microarray synthesis and assembly of gene-length polynucleotides
US20060024289A1 (en) * 2002-10-02 2006-02-02 Ruggles Sandra W Cleavage of VEGF and VEGF receptor by wild-type and mutant proteases
AU2003282724B2 (en) * 2002-10-02 2010-03-04 Catalyst Biosciences, Inc. Methods of generating and screening for proteases with altered specificity
DK1551876T3 (en) 2002-10-16 2011-06-14 Purdue Pharma Lp Antibodies that bind to cell associates approximately 125 / 0722P and methods for their use
WO2004042003A2 (en) * 2002-11-01 2004-05-21 Promega Corporation Cell lysis compositions, methods of use, apparatus, and kit
NZ540043A (en) * 2002-11-18 2007-11-30 Maxygen Inc Isolated or recombinant Interferon-alpha polypeptides and conjugates
US7314613B2 (en) * 2002-11-18 2008-01-01 Maxygen, Inc. Interferon-alpha polypeptides and conjugates
US20040121305A1 (en) * 2002-12-18 2004-06-24 Wiegand Roger Charles Generation of efficacy, toxicity and disease signatures and methods of use thereof
CN102618564B (en) 2003-03-06 2014-10-29 维莱尼姆公司 Amylases, nucleic acids encoding them and methods for making and using them
CA2889013C (en) 2003-03-07 2018-07-17 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.
US7402386B2 (en) 2003-04-14 2008-07-22 Nugen Technologies, Inc. Global amplification using random priming by a composite primer
CN1863914B (en) 2003-04-29 2011-03-09 先锋高级育种国际公司 Novel glyphosate-n-acetyltransferase (GAT) genes
WO2005017106A2 (en) 2003-06-17 2005-02-24 California Institute Of Technology Libraries of optimized cytochrome p450 enzymes and the optimized p450 enzymes
US7524664B2 (en) 2003-06-17 2009-04-28 California Institute Of Technology Regio- and enantioselective alkane hydroxylation with modified cytochrome P450
DK1641910T3 (en) 2003-07-02 2013-05-21 Verenium Corp Glucanases, nucleic acids encoding them, and methods of preparing and utilizing them
DK1660646T3 (en) 2003-08-11 2015-03-09 California Inst Of Techn Thermostable peroxide-driven cytochrome P450 oxygenase variants and methods of use
CA2535526C (en) 2003-08-11 2015-09-29 Diversa Corporation Laccases, nucleic acids encoding them and methods for making and using them
JP4809767B2 (en) 2003-08-18 2011-11-09 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Expression vectors, polypeptide display libraries, and methods for their production and use
PL1673457T3 (en) 2003-08-25 2011-12-30 Amyra Biotech Ag Novel fungal proteins and nucleic acids encoding same
EP2261332A3 (en) * 2003-08-27 2012-05-09 Proterec Ltd Libraries of recombinant chimeric proteins
CN101068934A (en) * 2003-10-29 2007-11-07 里伯米德生物技术公司 Compositions, methods and detection technologies for reiterative oligonucleotide synthesis
CN101671681B (en) 2004-02-25 2012-11-28 先锋高级育种国际公司 Novel bacillus thuringiensis crystal polypeptides, polynucleotides, and compositions thereof
JP5629423B2 (en) 2004-04-12 2014-11-19 カタリスト バイオサイエンシーズ, インコーポレイテッド Cleavage of VEGF and VEGF receptors by wild type and mutant MT-SP1
BRPI0511196A (en) * 2004-05-19 2007-12-04 Maxygen Inc isolated or recombinant polypeptide, conjugate, composition, host cell, vector, methods for making a polypeptide, for making a conjugate, for reducing the copy number of a virus in virus-infected cells, for reducing the level of rna hcv, dna hbv and rna hiv in the serum of a patient infected with hcv, hbv and hiv, and use of the polypeptide, conjugate or composition
ES2352172T3 (en) 2004-06-09 2011-02-16 Pioneer-Hi-Bred International, Inc. PLASTIDE TRANSIT PEPTIDES.
CN101432292B (en) 2004-06-16 2013-03-13 维莱尼姆公司 Compositions and methods for enzymatic decolorization of chlorophyll
US20060029573A1 (en) * 2004-06-30 2006-02-09 Chun Shen Pegylated interferon alpha-1b
GB0422052D0 (en) 2004-10-04 2004-11-03 Dansico As Enzymes
GB0423139D0 (en) 2004-10-18 2004-11-17 Danisco Enzymes
AU2005295351A1 (en) * 2004-10-18 2006-04-27 Codon Devices, Inc. Methods for assembly of high fidelity synthetic polynucleotides
US20070122817A1 (en) * 2005-02-28 2007-05-31 George Church Methods for assembly of high fidelity synthetic polynucleotides
CN102225966B (en) 2004-10-19 2012-12-26 隆萨股份公司 Method for solid phase peptide synthesis
CA2590245A1 (en) 2004-11-11 2006-05-18 Modular Genetics, Inc. Ladder assembly and system for generating diversity
US7329797B2 (en) * 2004-12-08 2008-02-12 Ceres, Inc. Modulating plant carbon levels
WO2007087601A2 (en) * 2006-01-25 2007-08-02 Ceres, Inc. Modulating plant protein levels
WO2007089610A1 (en) * 2006-01-26 2007-08-09 Ceres, Inc. Modulating plant oil levels
BRPI0519657A2 (en) * 2004-12-16 2009-03-03 Ceres Inc modulation of nitrogen levels in plants
CA2594832A1 (en) * 2005-01-13 2006-07-20 Codon Devices, Inc. Compositions and methods for protein design
WO2006099207A2 (en) 2005-03-10 2006-09-21 Diversa Corporation Lyase enzymes, nucleic acids encoding them and methods for making and using them
US8426184B2 (en) 2005-03-15 2013-04-23 Bp Corporation North America Cellulases, nucleic acids encoding them and methods for making and using them
US8715988B2 (en) 2005-03-28 2014-05-06 California Institute Of Technology Alkane oxidation by modified hydroxylases
US11214817B2 (en) 2005-03-28 2022-01-04 California Institute Of Technology Alkane oxidation by modified hydroxylases
WO2006127423A2 (en) * 2005-05-18 2006-11-30 Codon Devices, Inc. Methods of producing polynucleotide libraries using scarless ligation
AU2006299901A1 (en) * 2005-05-18 2007-04-19 Maxygen, Inc. Evolved interferon-alpha polypeptides
DK2314623T3 (en) 2005-06-21 2012-09-17 Xoma Technology Ltd IL-1 BETA-BINDING ANTIBODIES AND FRAGMENTS THEREOF
WO2007008951A1 (en) * 2005-07-12 2007-01-18 Codon Devices, Inc. Compositions and methods for design of non-immunogenic proteins
EP1902126A1 (en) * 2005-07-12 2008-03-26 Codon Devices, Inc Compositions and methods for biocatalytic engineering
US7666817B2 (en) 2005-08-31 2010-02-23 The Regents Of The University Of California Cellular libraries of peptide sequences (CLiPS) and methods of using the same
EP1929046B1 (en) * 2005-09-07 2012-07-11 Nugen Technologies, Inc. Improved nucleic acid amplification procedure
GB2432366B (en) * 2005-11-19 2007-11-21 Alligator Bioscience Ab A method for in vitro molecular evolution of protein function
KR100759817B1 (en) * 2005-12-08 2007-09-20 한국전자통신연구원 Method and device for predicting regulation of multiple transcription factors
CN103690947A (en) * 2006-01-12 2014-04-02 阿莱克申药物公司 Antibodies to ox-2/cd200 and uses thereof
JP2009525319A (en) 2006-02-02 2009-07-09 ライナット ニューロサイエンス コーポレイション Method of treating obesity by administering a trkB antagonist
WO2007092314A2 (en) 2006-02-02 2007-08-16 Verenium Corporation Esterases and related nucleic acids and methods
NZ595498A (en) 2006-02-10 2013-05-31 Verenium Corp Cellulolytic enzymes, nucleic acids encoding them and methods for making and using them
CA2638801C (en) 2006-02-14 2016-12-13 Verenium Corporation Xylanases, nucleic acids encoding them and methods for making and using them
EP2112227B1 (en) 2006-03-07 2013-05-15 Cargill, Incorporated Aldolases, nucleic acids encoding them and methods for making and using them
MX2008011442A (en) 2006-03-07 2008-11-18 Verenium Corp Aldolases, nucleic acids encoding them and methods for making and using them.
US20070231805A1 (en) * 2006-03-31 2007-10-04 Baynes Brian M Nucleic acid assembly optimization using clamped mismatch binding proteins
US20100024070A1 (en) * 2006-05-15 2010-01-28 Steven Craig Bobzin Modulation of oil levels in plants
WO2007136834A2 (en) * 2006-05-19 2007-11-29 Codon Devices, Inc. Combined extension and ligation for nucleic acid assembly
AR061365A1 (en) * 2006-06-13 2008-08-20 Athenix Corp METHODS TO GENERATE GENETIC DIVERSITY THROUGH MUTAGENESIS WITH PERMUTATIONS
WO2007149570A2 (en) * 2006-06-21 2007-12-27 Ceres, Inc. Modulation of protein levels in plants
US7572618B2 (en) 2006-06-30 2009-08-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
WO2008016709A2 (en) 2006-08-04 2008-02-07 California Institute Of Technology Methods and systems for selective fluorination of organic molecules
US8252559B2 (en) 2006-08-04 2012-08-28 The California Institute Of Technology Methods and systems for selective fluorination of organic molecules
CN101528766A (en) 2006-08-04 2009-09-09 维莱尼姆公司 Glucanases, nucleic acids encoding them and methods for making and using them
US7727745B2 (en) * 2006-08-14 2010-06-01 Bio-Id Diagnostic Inc. Synthesis of single-stranded DNA
CA2660705A1 (en) * 2006-08-15 2008-02-21 Commonwealth Scientific And Industrial Research Organisation Reassortment by fragment ligation
US8158411B2 (en) 2006-08-21 2012-04-17 Samsung Electronics Co., Ltd. Method of separating microorganism using nonplanar solid substrate and device for separating microorganism using the same
US8053191B2 (en) * 2006-08-31 2011-11-08 Westend Asset Clearinghouse Company, Llc Iterative nucleic acid assembly using activation of vector-encoded traits
CN101558154B (en) 2006-09-21 2016-02-24 帝斯曼知识产权资产管理有限公司 Phospholipid hydrolase, encode their nucleic acid and methods for making and using same thereof
US8877478B2 (en) 2006-09-21 2014-11-04 Verenium Corporation Phytases, nucleic acids encoding them and methods for making and using them
US20100151109A1 (en) * 2006-12-15 2010-06-17 Amr Saad Ragab Modulation of plant protein levels
EP2069490B2 (en) 2006-12-21 2018-02-21 BASF Enzymes LLC Amylases and glucoamylases, nucleic acids encoding them and methods for making and using them
DE102007003143A1 (en) 2007-01-16 2008-07-17 Henkel Kgaa New alkaline protease from Bacillus gibsonii and detergents and cleaners containing this novel alkaline protease
US20080175668A1 (en) * 2007-01-18 2008-07-24 Larry Wayne Haese Graywater recycling system including rainwater recovery
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
WO2008103248A1 (en) 2007-02-08 2008-08-28 Codexis, Inc. Ketoreductases and uses thereof
WO2009008908A2 (en) 2007-02-12 2009-01-15 Codexis, Inc. Structure-activity relationships
WO2008103474A1 (en) 2007-02-20 2008-08-28 Anaptysbio, Inc. Methods of generating libraries and uses thereof
US7625555B2 (en) 2007-06-18 2009-12-01 Novagen Holding Corporation Recombinant human interferon-like proteins
MX2010000979A (en) 2007-07-25 2010-03-26 Alexion Pharma Inc Methods and compositions for treating autoimmune disease.
WO2009014726A1 (en) 2007-07-26 2009-01-29 The Regents Of The University Of California Methods for enhancing bacterial cell display of proteins and peptides
DK3031919T3 (en) 2007-07-31 2019-01-07 Basf Enzymes Llc Tailored multi-site combination device
EP2185930A4 (en) * 2007-08-21 2010-11-24 Affomix Corp Interaction screening methods, systems and devices
WO2009029554A2 (en) 2007-08-24 2009-03-05 Codexis, Inc. Improved ketoreductase polypeptides for the stereoselective production of (r)-3-hydroxythiolane
EP2205727B1 (en) 2007-10-01 2015-06-24 Codexis, Inc. Ketoreductase polypeptides for the production of azetidinone
CN110577945A (en) 2007-10-03 2019-12-17 维莱尼姆公司 xylanases, nucleic acids encoding them, and methods for making and using them
DE102007051092A1 (en) 2007-10-24 2009-04-30 Henkel Ag & Co. Kgaa Subtilisin from Becillus pumilus and detergents and cleaners containing this new subtilisin
PL2612868T3 (en) 2007-11-01 2018-12-31 Astellas Pharma Inc. Immunosuppressive polypeptides and nucleic acids
EP2706122A3 (en) 2008-01-03 2014-06-18 Verenium Corporation Isomerases, nucleic acids encoding them and methods for making and using them
EP2865750A3 (en) 2008-01-03 2015-08-05 BASF Enzymes LLC Transferases and oxidoreductases, nucleic acids encoding them and methods for making and using them
WO2009097673A1 (en) * 2008-02-07 2009-08-13 Bio-Id Diagnostic Inc. Synthesis of single-stranded dna
US8768871B2 (en) 2008-02-12 2014-07-01 Codexis, Inc. Method of generating an optimized, diverse population of variants
EP2250595B1 (en) 2008-02-12 2017-06-14 Codexis, Inc. Method of selecting an optimized diverse population of variants
US8034568B2 (en) * 2008-02-12 2011-10-11 Nugen Technologies, Inc. Isothermal nucleic acid amplification methods and compositions
PL2255003T3 (en) * 2008-02-15 2015-08-31 Ceres Inc Drought and heat tolerance in plants
US7846666B2 (en) 2008-03-21 2010-12-07 Nugen Technologies, Inc. Methods of RNA amplification in the presence of DNA
EP3115459A3 (en) 2008-05-23 2017-04-26 E. I. du Pont de Nemours and Company Novel dgat genes for increased seed storage lipid production and altered fatty acid profiles in oilseed plants
US8383346B2 (en) 2008-06-13 2013-02-26 Codexis, Inc. Combined automated parallel synthesis of polynucleotide variants
US20090312196A1 (en) 2008-06-13 2009-12-17 Codexis, Inc. Method of synthesizing polynucleotide variants
EP2285958B1 (en) 2008-06-13 2016-03-09 Codexis, Inc. Method of synthesizing polynucleotide variants
EP2307419B1 (en) 2008-06-24 2013-11-06 Codexis, Inc. Biocatalytic processes for the preparation of substantially stereomerically pure fused bicyclic proline compounds
EP2329013B1 (en) 2008-08-27 2015-10-28 Codexis, Inc. Ketoreductase polypeptides for the production of a 3-aryl-3-hydroxypropanamine from a 3-aryl-3-ketopropanamine
US8288131B2 (en) 2008-08-27 2012-10-16 Codexis, Inc. Ketoreductase polypeptides and uses thereof
US8288141B2 (en) 2008-08-27 2012-10-16 Codexis, Inc. Ketoreductase polypeptides for the production of 3-aryl-3-hydroxypropanamine from a 3-aryl-3-ketopropanamine
CN102186972B (en) 2008-08-29 2014-08-20 科德克希思公司 Ketoreductase polypeptides for the stereoselective production of (4s)-3[(5s)-5(4-fluorophenyl)-5-hydroxypentanoyl]-4-phenyl-1,3-oxazolidin-2-one
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
EP3460061A1 (en) 2008-09-26 2019-03-27 Tocagen Inc. Recombinant replication competent murine leukemia gamma-retrovirus (mlv) for the expression of heat-stabilized cytosine deaminase
US8247192B2 (en) * 2008-11-10 2012-08-21 Codexis, Inc. Penicillin-G acylases
US8329438B2 (en) * 2008-12-25 2012-12-11 Codexis, Inc. Enone reductases
DK3354727T3 (en) 2009-01-08 2020-11-16 Codexis Inc TRANSAMINASE POLYPEPTIDES
EP2394164A4 (en) * 2009-02-03 2014-01-08 Complete Genomics Inc Oligomer sequences mapping
US8738296B2 (en) * 2009-02-03 2014-05-27 Complete Genomics, Inc. Indexing a reference sequence for oligomer sequence mapping
WO2010091024A1 (en) * 2009-02-03 2010-08-12 Complete Genomics, Inc. Oligomer sequences mapping
US8715996B2 (en) 2009-02-26 2014-05-06 Codexis, Inc. Beta-glucosidase variant enzymes and related polynucleotides
US8293507B2 (en) 2009-02-26 2012-10-23 Codexis, Inc. Transaminase biocatalysts
EP3282021A1 (en) * 2009-03-09 2018-02-14 Bioatla, LLC Mirac proteins
BRPI1013886A2 (en) 2009-03-17 2016-10-11 Codexis Inc endoglucanase variants, polynucleotides and related uses
BRPI1010239A2 (en) 2009-03-31 2016-10-11 Codexis Inc improved endoglucanases, derivatives and their uses
US20120100997A1 (en) 2009-04-24 2012-04-26 University Of Southern California Cd133 polymorphisms and expression predict clinical outcome in patients with cancer
CN102460155B (en) * 2009-04-29 2015-03-25 考利达基因组股份有限公司 Method and system for calling variations in a sample polynucleotide sequence with respect to a reference polynucleotide sequence
BRPI1011160A8 (en) 2009-05-21 2018-01-02 Verenium Corp PHYTASES, PROTEIN PREPARATION INCLUDING THEM, AND THEIR USES
WO2010148148A2 (en) 2009-06-16 2010-12-23 Codexis, Inc. β-GLUCOSIDASE VARIANTS
EP2443139B1 (en) 2009-06-16 2016-08-10 Codexis, Inc. Beta-glucosidase variant enzymes and related polynucleotides
EP3409765B1 (en) 2009-06-22 2021-08-04 Codexis, Inc. Ketoreductase-mediated stereoselective route to alpha chloroalcohols
US8921079B2 (en) 2009-06-22 2014-12-30 Codexis, Inc. Transaminase reactions
US8614081B2 (en) 2009-07-23 2013-12-24 Codexis, Inc. Nitrilase biocatalysts
EP2467473B1 (en) 2009-08-19 2016-03-23 Codexis, Inc. Ketoreductase polypeptides for the preparation of phenylephrine
EP2467395A1 (en) 2009-08-20 2012-06-27 Pioneer Hi-Bred International Inc. Functional expression of shuffled yeast nitrate transporter (ynti) in maize to improve nitrate uptake under low nitrate environment
EP2473604B1 (en) 2009-09-04 2017-01-11 Codexis, Inc. Variant cbh2 cellulases and related polynucleotides
EP2478137B1 (en) 2009-09-18 2017-09-13 Codexis, Inc. Reduced codon mutagenesis
WO2011041594A1 (en) 2009-09-30 2011-04-07 Codexis, Inc. Recombinant c1 b-glucosidase for production of sugars from cellulosic biomass
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
UA111708C2 (en) 2009-10-16 2016-06-10 Бандж Ойлз, Інк. METHOD OF OIL REFINING
US8916366B2 (en) 2009-11-20 2014-12-23 Codexis, Inc. Multi-cellulase enzyme compositions for hydrolysis of cellulosic biomass
ES2455269T3 (en) * 2009-11-25 2014-04-15 Codexis, Inc. Variants of recombinant beta-glucosidase for the production of soluble sugars from cellulosic biomass
BR112012012192A2 (en) 2009-11-25 2017-07-04 Codexis Inc b-glycosities recombinant thermoascus aurantiacus variants for production of fermentable sugars from cellulosic biomass
SG181535A1 (en) 2009-12-08 2012-07-30 Codexis Inc Synthesis of prazole compounds
WO2011084757A1 (en) 2009-12-21 2011-07-14 University Of Southern California Germline polymorphisms in the sparc gene associated with clinical outcome in gastric cancer
US20110191903A1 (en) 2009-12-31 2011-08-04 Pioneer Hi-Bred International, Inc. Engineering plant resistance to diseases caused by pathogens
WO2011085334A1 (en) 2010-01-11 2011-07-14 University Of Southern California Cd44 polymorphisms predict clinical outcome in patients with gastric cancer
US9080192B2 (en) 2010-02-10 2015-07-14 Codexis, Inc. Processes using amino acid dehydrogenases and ketoreductase-based cofactor regenerating system
US8715962B2 (en) 2010-03-31 2014-05-06 Codexis, Inc. Production of geranyl diphosphate
US9040262B2 (en) 2010-05-04 2015-05-26 Codexis, Inc. Biocatalysts for ezetimibe synthesis
EP2569331A1 (en) 2010-05-10 2013-03-20 Perseid Therapeutics LLC Polypeptide inhibitors of vla4
WO2011143632A2 (en) 2010-05-14 2011-11-17 Codexis, Inc. Cellobiohydrolase variants
US20110294170A1 (en) 2010-05-28 2011-12-01 Codexis, Inc. Pentose Fermentation By a Recombinant Microorganism
WO2011159910A2 (en) 2010-06-17 2011-12-22 Codexis, Inc. Biocatalysts and methods for the synthesis of (s)-3-(1-aminoethyl)-phenol
CA2804019A1 (en) 2010-06-28 2012-01-12 Codexis, Inc. Fatty alcohol forming acyl reductases (fars) and methods of use thereof
AU2011272878B2 (en) 2010-06-30 2015-04-23 Codexis, Inc. Highly stable beta-class carbonic anhydrases useful in carbon capture systems
WO2012021797A1 (en) 2010-08-13 2012-02-16 Pioneer Hi-Bred International, Inc. Methods and compositions for targeting sequences of interest to the chloroplast
US8932836B2 (en) 2010-08-16 2015-01-13 Codexis, Inc. Biocatalysts and methods for the synthesis of (1R,2R)-2-(3,4-dimethoxyphenethoxy)cyclohexanamine
US9006460B2 (en) 2010-08-16 2015-04-14 Cardiome International Ag Process for preparing aminocyclohexyl ether compounds
WO2012024662A2 (en) 2010-08-20 2012-02-23 Codexis, Inc. Expression constructs comprising fungal promoters
EP2606131B1 (en) 2010-08-20 2017-03-08 Codexis, Inc. Use of glycoside hydrolase 61 family proteins in processing of cellulose
US9068235B2 (en) 2010-11-02 2015-06-30 Codexis, Inc. Fungal strains
WO2012061432A1 (en) 2010-11-02 2012-05-10 Codexis, Inc. Compositions and methods for production of fermentable sugars
US10457935B2 (en) 2010-11-12 2019-10-29 Gen9, Inc. Protein arrays and methods of using and making the same
US9295965B2 (en) 2010-11-12 2016-03-29 Gen9, Inc. Methods and devices for nucleic acid synthesis
HUE037616T2 (en) 2010-12-08 2018-09-28 Codexis Inc Biocatalysts and methods for the synthesis of armodafinil
BR112013015829A2 (en) 2010-12-21 2018-05-29 Codexis Inc endoglucanase variants
US8663962B2 (en) 2011-03-30 2014-03-04 Codexis, Inc. Pentose fermentation by a recombinant microorganism
US20120258866A1 (en) * 2011-04-05 2012-10-11 Full Spectrum Genetics, Inc. Multi-dimensional selection of protein mutants using high throughput sequence analysis
MX358864B (en) 2011-04-12 2018-09-06 Gangagen Inc Chimeric antibacterial polypeptides.
WO2012142302A2 (en) 2011-04-13 2012-10-18 Codexis, Inc. Biocatalytic process for preparing eslicarbazepine and analogs thereof
HUE042817T2 (en) 2011-06-28 2019-07-29 Codexis Inc Protein variant generation by region shuffling
BR112013033614A2 (en) 2011-06-30 2018-05-29 Codexis Inc pentose fermentation by recombinant microorganism
WO2013016207A2 (en) 2011-07-22 2013-01-31 California Institute Of Technology Stable fungal cel6 enzyme variants
EP2748317B1 (en) 2011-08-22 2017-04-19 Codexis, Inc. Gh61 glycoside hydrolase protein variants and cofactors that enhance gh61 activity
BR112014003740A2 (en) 2011-08-23 2018-08-14 Codexis Inc cellobiohydrolase variants
EP3954770A1 (en) 2011-08-26 2022-02-16 Gen9, Inc. Compositions and methods for high fidelity assembly of nucleic acids
US9109209B2 (en) 2011-09-08 2015-08-18 Codexis, Inc. Biocatalysts and methods for the synthesis of substituted lactams
US20130084608A1 (en) 2011-09-30 2013-04-04 Codexis, Inc. Fungal proteases
WO2013074650A1 (en) 2011-11-18 2013-05-23 Codexis, Inc. Biocatalysts for the preparation of hydroxy substituted carbamates
US9611462B2 (en) 2011-12-20 2017-04-04 Codexis, Inc. Endoglucanase 1B (EG1B) variants
US20140357727A1 (en) 2011-12-20 2014-12-04 Codexis, Inc. Fatty alcohol forming acyl reductase (far) variants and methods of use
US9150853B2 (en) 2012-03-21 2015-10-06 Gen9, Inc. Methods for screening proteins using DNA encoded chemical libraries as templates for enzyme catalysis
SG11201405990PA (en) 2012-03-23 2014-11-27 Codexis Inc Biocatalysts and methods for synthesizing derivatives of tryptamine and tryptamine analogs
WO2013159055A1 (en) 2012-04-20 2013-10-24 Codexis, Inc. Production of fatty alcohols from engineered microorganisms
EP3543350B1 (en) 2012-04-24 2021-11-10 Gen9, Inc. Methods for sorting nucleic acids and multiplexed preparative in vitro cloning
CN104411823A (en) 2012-05-04 2015-03-11 纳幕尔杜邦公司 Compositions and methods comprising sequences having meganuclease activity
ES2712682T3 (en) 2012-05-08 2019-05-14 Codexis Inc Biocatalysts and methods for the hydroxylation of chemical compounds
CN107904216B (en) 2012-05-11 2021-06-29 科德克希思公司 Engineered imine reductases and methods for reductive amination of ketone and amine compounds
US9238676B2 (en) 2012-05-17 2016-01-19 Ra Pharmaceuticals, Inc. Peptide and peptidomimetic inhibitors
DK2859099T3 (en) 2012-06-11 2019-07-29 Codexis Inc MUSHROOMS-BETA-xylosidase VERSIONS
LT2864531T (en) 2012-06-25 2019-03-12 Gen9, Inc. Methods for nucleic acid assembly and high throughput sequencing
US9650655B2 (en) 2012-07-20 2017-05-16 Codexis, Inc. Production of fatty alcohols from engineered microorganisms
US9506087B2 (en) 2012-08-07 2016-11-29 Codexis, Inc. Glucose and xylose co-utilization in E. coli
US9663532B2 (en) 2012-10-29 2017-05-30 University Of Rochester Artemisinin derivatives, methods for their preparation and their use as antimalarial agents
US9611515B2 (en) 2012-11-20 2017-04-04 Codexis, Inc. Pentose fermentation by a recombinant microorganism
EP2928865B1 (en) 2012-12-07 2018-04-11 Merck Sharp & Dohme Corp. Biocatalytic transamination process
KR101600899B1 (en) * 2012-12-11 2016-03-09 주식회사 셀레믹스 Method of simultaneous synthesis of DNA library using high-throughput parallel DNA synthesis method
US9512447B2 (en) 2012-12-14 2016-12-06 Codexis, Inc. Modified native beta-ketoacyl-ACP synthases and engineered microorganisms
CA2895752C (en) 2012-12-21 2021-09-21 Codexis, Inc. Engineered biocatalysts and methods for synthesizing chiral amines
HUE044110T2 (en) 2013-01-18 2019-09-30 Codexis Inc Engineered biocatalysts useful for carbapenem synthesis
RU2695146C2 (en) 2013-01-31 2019-07-22 Кодексис, Инк. Methods, systems and software for identification of biomolecules with interacting components
CA2902824C (en) 2013-02-28 2021-06-22 Codexis, Inc. Engineered transaminase polypeptides for industrial biocatalysis
WO2014153242A1 (en) 2013-03-14 2014-09-25 Pioneer Hi-Bred International, Inc. Compositions having dicamba decarboxylase activity and methods of use
US9670493B2 (en) 2013-03-14 2017-06-06 Codexis, Inc. Low-phosphate repressible promoter
CA2905595A1 (en) 2013-03-14 2014-09-25 Pioneer Hi-Bred International, Inc. Compositions having dicamba decarboxylase activity and methods of use
WO2014150914A2 (en) 2013-03-15 2014-09-25 Pioneer Hi-Bred International, Inc. Phi-4 polypeptides and methods for their use
ES2890955T3 (en) 2013-04-18 2022-01-25 Codexis Inc Ammonia lyase modified phenylalanine polypeptides
BR112016003225B1 (en) 2013-08-16 2022-10-25 Pioneer Hi-Bred International, Inc. PIP-47 POLYPEPTIDE, CHIMERIC PIP-47 POLYPEPTIDE, COMPOSITION, FUSION PROTEIN, METHOD FOR CONTROLLING A PEST INSECT POPULATION, METHOD FOR INHIBITING THE GROWTH OR KILLING A PEST INSECT, DNA CONSTRUCTION, ISOLATED POLYNUCLEOTIDE, EXPRESSION CASSETTE, METHOD OF OBTAINING A TRANSGENIC PLANT AND METHOD TO CONTROL INSECT INFESTATION
CN105637099B (en) 2013-08-23 2020-05-19 深圳华大智造科技有限公司 Long fragment de novo assembly using short reads
CA3175967A1 (en) 2013-09-13 2015-03-19 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
CN105765592B (en) 2013-09-27 2019-12-17 科德克希思公司 Methods, devices and systems for automated screening of enzyme variants
HUE053379T2 (en) 2013-11-13 2021-06-28 Codexis Inc Engineered imine reductases and methods for the reductive amination of ketone and amine compounds
EP4122945A1 (en) 2013-12-23 2023-01-25 University of Rochester Methods and compositions for ribosomal synthesis of macrocyclic peptides
WO2015120270A1 (en) 2014-02-07 2015-08-13 Pioneer Hi Bred International, Inc. Insecticidal proteins and methods for their use
UA120608C2 (en) 2014-02-07 2020-01-10 Піонір Хай-Бред Інтернешнл, Інк. Insecticidal proteins and methods for their use
WO2015145463A1 (en) 2014-03-27 2015-10-01 Gangagen, Inc. Phage-derived compositions for improved mycobacterial therapy
US9605252B2 (en) 2014-04-16 2017-03-28 Codexis, Inc. Engineered tyrosine ammonia lyase
US9708587B2 (en) 2014-07-09 2017-07-18 Codexis, Inc. P450-BM3 variants with improved activity
WO2016014725A1 (en) 2014-07-22 2016-01-28 The University Of Notre Dame Du Lac Molecular constructs and uses thereof
US10273229B2 (en) 2014-08-28 2019-04-30 Codexis, Inc. N-substituted 4-aminoquinazoline derivatives and methods of use
US10045987B2 (en) 2014-08-28 2018-08-14 Codexis, Inc. Imidazoyl anilide derivatives and methods of use
MX2017004745A (en) 2014-10-16 2017-10-20 Pioner Hi-Bred Int Inc Insecticidal proteins and methods for their use.
DK3224354T3 (en) 2014-11-25 2021-03-01 Codexis Inc MANIPULATED IMINREDUCTASES AND PROCEDURES FOR THE REDUCTIVE AMINATION OF KETONE AND AMIN COMPOUNDS
SI3237621T1 (en) 2014-12-22 2023-11-30 Codexis, Inc. Human alpha-galactosidase variants
WO2016130412A1 (en) 2015-02-10 2016-08-18 Codexis, Inc. Ketoreductase polypeptides for the synthesis of chiral compounds
EA038923B1 (en) 2015-03-11 2021-11-10 Пайонир Хай-Бред Интернэшнл, Инк. Insecticidal dna construct and methods of use thereof
JP6755886B2 (en) 2015-05-07 2020-09-16 コデクシス, インコーポレイテッド Penicillin G acylase
EP3294757B1 (en) 2015-05-13 2023-12-27 Synlogic Operating Company, Inc. Bacteria engineered to treat a disease or disorder
CN108064233B (en) 2015-05-19 2022-07-15 先锋国际良种公司 Insecticidal proteins and methods of use thereof
WO2016210384A2 (en) 2015-06-25 2016-12-29 Synlogic, Inc. Bacteria engineered to treat metabolic diseases
CA2991195C (en) 2015-07-07 2021-06-29 Codexis, Inc. Novel p450-bm3 variants with improved activity
WO2017023486A1 (en) 2015-08-06 2017-02-09 Pioneer Hi-Bred International, Inc. Plant derived insecticidal proteins and methods for their use
US9988624B2 (en) 2015-12-07 2018-06-05 Zymergen Inc. Microbial strain improvement by a HTP genomic engineering platform
US11208649B2 (en) 2015-12-07 2021-12-28 Zymergen Inc. HTP genomic engineering platform
JP6821598B2 (en) 2015-12-07 2021-01-27 ザイマージェン インコーポレイテッド Promoter derived from Corynebacterium glutamicum
US20180325119A1 (en) 2015-12-18 2018-11-15 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
WO2017123676A1 (en) 2016-01-11 2017-07-20 Synlogic, Inc. Recombinant bacteria engineered to treat diseases and disorders associated with amino acid metabolism and methods of use thereof
EP3402497A1 (en) 2016-01-11 2018-11-21 Synlogic Operating Company, Inc. Bacteria engineered to treat metabolic diseases
WO2017123592A1 (en) 2016-01-11 2017-07-20 Synlogic, Inc. Bacteria engineered to treat disorders associated with bile salts
WO2017139708A1 (en) 2016-02-10 2017-08-17 Synlogic, Inc. Bacteria engineered to treat nonalcoholic steatohepatitis (nash)
BR112018072417B1 (en) 2016-05-04 2023-03-14 E. I. Du Pont De Nemours And Company RECOMBINANT INSECTICIDAL POLYPEPTIDE, CHIMERIC POLYPEPTIDE, COMPOSITION, RECOMBINANT POLYNUCLEOTIDE, DNA CONSTRUCTS, METHODS FOR OBTAINING A TRANSGENIC PLANT, METHODS FOR INHIBITING THE GROWTH OR EXTERMINATION OF AN INSECT PEST OR PEST POPULATION, METHOD FOR OBTAINING A TRANSFORMED PROKARYOTIC CELL TRANSFORMED AND METHOD TO GENETICALLY MODIFY THE INSECTICIDAL POLYPEPTIDE
KR102382489B1 (en) 2016-05-05 2022-04-01 코덱시스, 인코포레이티드 Penicillin G acylase
US10184117B2 (en) 2016-06-09 2019-01-22 Codexis, Inc. Biocatalysts and methods for hydroxylation of chemical compounds
JP2019517801A (en) 2016-06-15 2019-06-27 コデクシス, インコーポレイテッド Engineered .beta.-glucosidase and glucosylation method
WO2018005655A2 (en) 2016-06-30 2018-01-04 Zymergen Inc. Methods for generating a bacterial hemoglobin library and uses thereof
JP2019519241A (en) 2016-06-30 2019-07-11 ザイマージェン インコーポレイテッド Method for producing a glucose permease library and its use
BR112018077339A2 (en) 2016-07-01 2019-04-24 Pioneer Hi-Bred International, Inc. recombinant insecticide polypeptide, composition, recombinant polynucleotide, dna construct, transgenic plant and methods
JP7316662B2 (en) 2016-08-26 2023-07-28 コデクシス, インコーポレイテッド Engineered imine reductases and methods for reductive amination of ketone and amine compounds
MX2019005044A (en) 2016-11-01 2019-08-05 Pioneer Hi Bred Int Insecticidal proteins and methods for their use.
CA3044404A1 (en) 2016-12-14 2018-06-21 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
US11213028B2 (en) 2016-12-22 2022-01-04 Pioneer Hi-Bred International, Inc. Insecticidal proteins and methods for their use
KR20190102063A (en) 2017-01-05 2019-09-02 코덱시스, 인코포레이티드 Penicillin G acylase
US20180223264A1 (en) 2017-02-03 2018-08-09 Codexis, Inc. Engineered glycosyltransferases and steviol glycoside glucosylation methods
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.
MX2019009615A (en) 2017-02-13 2019-10-14 Codexis Inc Engineered phenylalanine ammonia lyase polypeptides.
SG11201909807TA (en) 2017-04-24 2019-11-28 Tesaro Inc Methods of manufacturing of niraparib
SG11201909712TA (en) 2017-04-27 2019-11-28 Codexis Inc Ketoreductase polypeptides and polynucleotides
AU2018266606A1 (en) 2017-05-08 2019-11-14 Codexis, Inc. Engineered ligase variants
BR112019023628A2 (en) 2017-05-11 2020-06-02 Pioneer Hi-Bred International, Inc. RECOMBINANT INSECTICIDE POLYPEPIDE, CHEMICAL INSECTICIDE PROTEIN, FUSION PROTEIN, AGRICULTURAL COMPOSITION, RECOMBINANT POLYNUCLEOTIDE, DNA BUILDING, TRANSGENIC PLANT, METHOD OF INHIBITING THE AGGREGATION OR EXERCISING AGAINST EXERCISE OR EXERCISE , METHOD TO CONTROL PEST INFESTATION AND METHOD TO IMPROVE THE PERFORMANCE OF A CULTURE
WO2018231462A1 (en) 2017-06-14 2018-12-20 Codexis, Inc. Engineered transaminase polypeptides for industrial biocatalysis
EP3645713A4 (en) 2017-06-27 2021-06-30 Codexis, Inc. Penicillin-g acylases
RU2020103727A (en) 2017-06-30 2021-07-30 Кодексис, Инк. VARIANTS OF RNA POLYMERASE T7
JP2020530266A (en) 2017-06-30 2020-10-22 コデクシス, インコーポレイテッド T7 RNA polymerase variant
US11634400B2 (en) 2017-08-19 2023-04-25 University Of Rochester Micheliolide derivatives, methods for their preparation and their use as anticancer and antiinflammatory agents
WO2019067499A1 (en) 2017-09-27 2019-04-04 Alexion Pharmaceuticals, Inc. Biomarker signature for predicting tumor response to anti-cd200 therapy
CA3080512A1 (en) 2017-11-07 2019-05-16 Codexis, Inc. Transglutaminase variants
US11015180B2 (en) 2017-12-13 2021-05-25 Codexis, Inc. Carboxyesterase polypeptides for amide coupling
EP3724324A1 (en) 2017-12-13 2020-10-21 GlaxoSmithKline Intellectual Property Development Ltd Carboxyesterase biocatalysts
US10900055B2 (en) 2018-06-12 2021-01-26 Codexis, Inc. Engineered tyrosine ammonia lyase
AU2019300838A1 (en) 2018-07-09 2021-01-07 Codexis, Inc. Engineered pantothenate kinase variant enzymes
CA3108728A1 (en) 2018-07-09 2020-01-16 Codexis, Inc. Engineered phosphopentomutase variant enzymes
JP2021530982A (en) 2018-07-09 2021-11-18 コデクシス, インコーポレイテッド Manipulated galactose oxidase variant enzyme
WO2020014048A1 (en) 2018-07-09 2020-01-16 Codexis, Inc. Engineered deoxyribose-phosphate aldolases
KR20210031926A (en) 2018-07-09 2021-03-23 코덱시스, 인코포레이티드 Engineered Purine Nucleoside Phosphorylase Variant Enzyme
WO2020013951A1 (en) 2018-07-12 2020-01-16 Codexis, Inc. Engineered phenylalanine ammonia lyase polypeptides
BR112021001661A8 (en) 2018-07-30 2022-08-09 Codexis Inc Modified glyclytransferase, modified polynucleotide, vector, host cell, methods to produce at least one modified glyclytransferase, to produce at least one variant of sucrose synthase, to glycosillation of a substrate, to produce refudiosido, refudiosidade A and/or regaudiosido I, and regradiosido D, COMPOSITION, MODIFIED SUCROSE SYNTHASE, AT LEAST ONE REBAUDIOSIDE, REBAUDIOSIDE M, REBAUDIOSIDE A, REBAUDIOSIDE I, E, REBAUDIOSIDE D.
JP2022512847A (en) 2018-10-29 2022-02-07 コデクシス, インコーポレイテッド Manipulated DNA polymerase variant
US11473077B2 (en) 2018-12-14 2022-10-18 Codexis, Inc. Engineered tyrosine ammonia lyase
JP2022515742A (en) 2018-12-20 2022-02-22 コデクシス, インコーポレイテッド Alpha-galactosidase variant
EP4042149A1 (en) 2019-10-02 2022-08-17 Abbott Diabetes Care Inc. Detection of analytes by protein switches
EP4133064A2 (en) 2020-04-10 2023-02-15 Codexis, Inc. Engineered transaminase polypeptides
CN115867564A (en) 2020-07-14 2023-03-28 先锋国际良种公司 Insecticidal proteins and methods of use thereof
AU2021331493A1 (en) 2020-08-28 2023-03-09 Codexis, Inc. Engineered amylase variants
WO2022125960A1 (en) 2020-12-11 2022-06-16 Willow Biosciences, Inc. Recombinant acyl activating enzyme (aae) genes for enhanced biosynthesis of cannabinoids and cannabinoid precursors
US11898174B2 (en) 2020-12-18 2024-02-13 Codexis, Inc. Engineered uridine phosphorylase variant enzymes
EP4314262A1 (en) 2021-04-02 2024-02-07 Codexis, Inc. Engineered guanylate kinase variant enzymes
CA3215105A1 (en) 2021-04-02 2022-10-06 Codexis, Inc. Engineered acetate kinase variant enzymes
WO2022212824A1 (en) 2021-04-02 2022-10-06 Codexis, Inc. Engineered adenylate kinase variant enzymes
IL305924A (en) 2021-04-02 2023-11-01 Codexis Inc Engineered cyclic gmp-amp synthase (cgas) variant enzymes
WO2023010083A2 (en) 2021-07-30 2023-02-02 Willow Biosciences, Inc. Recombinant prenyltransferase polypeptides engineered for enhanced biosynthesis of cannabinoids
WO2023023621A1 (en) 2021-08-19 2023-02-23 Willow Biosciences, Inc. Recombinant olivetolic acid cyclase polypeptides engineered for enhanced biosynthesis of cannabinoids
WO2023069921A1 (en) 2021-10-19 2023-04-27 Epimeron Usa, Inc. Recombinant thca synthase polypeptides engineered for enhanced biosynthesis of cannabinoids
WO2023173084A1 (en) 2022-03-11 2023-09-14 University Of Rochester Cyclopeptibodies and uses thereof
WO2023245168A1 (en) 2022-06-17 2023-12-21 Synlogic Operating Company, Inc. Bacteria engineered to treat diseases associated with bile acid metabolism and methods of use thereof
WO2023245171A1 (en) 2022-06-17 2023-12-21 Synlogic Operating Company, Inc. Bacteria engineered to treat diseases associated with bile acid metabolism and methods of use thereof
WO2023250478A1 (en) 2022-06-23 2023-12-28 Synlogic Operating Company, Inc. Recombinant bacteria engineered to treat diseases associated with methionine metabolism and methods of use thereof
WO2024040020A1 (en) 2022-08-15 2024-02-22 Absci Corporation Quantitative affinity activity specific cell enrichment

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6335160B1 (en) * 1995-02-17 2002-01-01 Maxygen, Inc. Methods and compositions for polypeptide engineering

Family Cites Families (188)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US3996945A (en) * 1975-04-07 1976-12-14 Mcdowell James A Extinguisher for cigarettes or cigars
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4351901A (en) * 1980-03-24 1982-09-28 Cetus Corporation Method for single nucleotide alteration
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4695543A (en) * 1982-03-23 1987-09-22 Bristol-Myers Company Alpha Interferon GX-1
US5618697A (en) 1982-12-10 1997-04-08 Novo Nordisk A/S Process for preparing a desired protein
US4994379A (en) 1983-03-09 1991-02-19 Cetus Corporation Modified signal peptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5538868A (en) 1984-02-08 1996-07-23 Cetus Oncology Corporation Recombinant ricin toxin fragments
US5422266A (en) * 1984-12-31 1995-06-06 University Of Georgia Research Foundation, Inc. Recombinant DNA vectors capable of expressing apoaequorin
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US5176995A (en) 1985-03-28 1993-01-05 Hoffmann-La Roche Inc. Detection of viruses by amplification and hybridization
EP0316018A3 (en) * 1985-03-29 1989-07-26 Cetus Oncology Corporation Modification of dna sequences
EP0590689B2 (en) 1985-03-30 2006-08-16 KAUFFMAN, Stuart A. Method for obtaining DNA, RNA, peptides, polypeptides or proteins by means of a DNA-recombinant technique
US6492107B1 (en) 1986-11-20 2002-12-10 Stuart Kauffman Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US4959312A (en) 1985-05-31 1990-09-25 The University Of Tennessee Research Corporation Full spectrum mutagenesis
US4766106A (en) 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
DE3688920T4 (en) 1985-07-03 1995-08-31 Genencor Int Hybrid polypeptides and process for their preparation.
US5866363A (en) 1985-08-28 1999-02-02 Pieczenik; George Method and means for sorting and identifying biological information
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
FI72765C (en) 1986-02-13 1987-07-10 Valmet Oy UTBREDNINGSVALS ELLER MOTSVARANDE FOER VAEVNADEN I EN PAPPERSMASKIN SAMT FOERFARANDE FOER FRAMSTAELLNING AV DENNA.
DK311186D0 (en) 1986-06-30 1986-06-30 Novo Industri As ENZYMES
US5824469A (en) 1986-07-17 1998-10-20 University Of Washington Method for producing novel DNA sequences with biological activity
US4795699A (en) 1987-01-14 1989-01-03 President And Fellows Of Harvard College T7 DNA polymerase
US4994368A (en) 1987-07-23 1991-02-19 Syntex (U.S.A.) Inc. Amplification method for polynucleotide assays
DK6488D0 (en) 1988-01-07 1988-01-07 Novo Industri As ENZYMES
WO1989006694A1 (en) * 1988-01-15 1989-07-27 Trustees Of The University Of Pennsylvania Process for selection of proteinaceous substances which mimic growth-inducing molecules
US5206152A (en) * 1988-04-08 1993-04-27 Arch Development Corporation Cloning and expression of early growth regulatory protein genes
US4994366A (en) * 1988-04-28 1991-02-19 Jac Creative Foods, Inc. Shrimp analog forming process
AU3869289A (en) * 1988-07-14 1990-02-05 Baylor College Of Medicine Solid phase assembly and reconstruction of biopolymers
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
FR2641793B1 (en) 1988-12-26 1993-10-01 Setratech METHOD OF IN VIVO RECOMBINATION OF DNA SEQUENCES HAVING BASIC MATCHING
US4902502A (en) 1989-01-23 1990-02-20 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
DD282028A5 (en) 1989-02-16 1990-08-29 Akad Wissenschaften Ddr PROCESS FOR PREPARING THERMOSTABILES, HYBRIDEN BACILLUS BETA-1,3-1,4-GLUCANASE
DE69028725T2 (en) * 1989-02-28 1997-03-13 Canon Kk Partially double-stranded oligonucleotide and method for its formation
DE3909710A1 (en) * 1989-03-23 1990-09-27 Boehringer Mannheim Gmbh METHOD FOR EXPRESSING A RECOMBINANT GENE
US5106727A (en) 1989-04-27 1992-04-21 Life Technologies, Inc. Amplification of nucleic acid sequences using oligonucleotides of random sequences as primers
US5043272A (en) 1989-04-27 1991-08-27 Life Technologies, Incorporated Amplification of nucleic acid sequences using oligonucleotides of random sequence as primers
EP0397463A3 (en) 1989-05-10 1991-03-06 Eastman Kodak Company Construction of synthetic double-stranded dna sequences
US5556750A (en) 1989-05-12 1996-09-17 Duke University Methods and kits for fractionating a population of DNA molecules based on the presence or absence of a base-pair mismatch utilizing mismatch repair systems
US6291161B1 (en) 1989-05-16 2001-09-18 Scripps Research Institute Method for tapping the immunological repertiore
US6291158B1 (en) 1989-05-16 2001-09-18 Scripps Research Institute Method for tapping the immunological repertoire
CA2016842A1 (en) 1989-05-16 1990-11-16 Richard A. Lerner Method for tapping the immunological repertoire
CA2016841C (en) 1989-05-16 1999-09-21 William D. Huse A method for producing polymers having a preselected activity
DK169306B1 (en) 1989-06-14 1994-10-10 Af 18 Juni 1990 As Means for removing or inactivating unwanted components in blood or other extracellular body fluids and method of preparing dextran-O-carbonyl-benzo-18-crown-6 and of dextran-hydroxyethylbenzo-18-crown-6
CA2019696A1 (en) 1989-06-26 1990-12-26 William L. Muth Control of aberrant expression vector accumulation during fermentation procedures
US5240845A (en) * 1989-07-11 1993-08-31 Otsuka Pharmaceutical Factory, Ltd. Mutated streptokinase proteins
US5045450A (en) * 1989-07-13 1991-09-03 Massachusetts Institute Of Technology Determination of a mutational spectrum
IT1231157B (en) 1989-07-20 1991-11-19 Crc Ricerca Chim NEW FUNCTIONAL HYBRID GENES OF BACILLUS THURINGIENSIS OBTAINED THROUGH IN VIVO RECOMBINATION.
US5514787A (en) * 1989-07-21 1996-05-07 Washington University DNA sequences encoding human membrane cofactor protein (MCP)
US5574205A (en) 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
US5023171A (en) 1989-08-10 1991-06-11 Mayo Foundation For Medical Education And Research Method for gene splicing by overlap extension using the polymerase chain reaction
US5360726A (en) 1989-09-12 1994-11-01 Board Of Trustees Operating Michigan State University Polypeptides enabling sorting of proteins to vacuoles in plants
US5030568A (en) * 1989-09-29 1991-07-09 Minnesota Mining And Manufacturing Company Bioconversion of naphthalene monomers
WO1991006570A1 (en) 1989-10-25 1991-05-16 The University Of Melbourne HYBRID Fc RECEPTOR MOLECULES
AU6886991A (en) 1989-11-08 1991-06-13 United States of America, as represented by the Secretary, U.S. Department of Commerce, The A method of synthesizing double-stranded dna molecules
DE4000939A1 (en) * 1990-01-15 1991-07-18 Brem Gottfried Prof Dr Dr METHOD FOR OBTAINING ANTIBODIES
ES2089038T3 (en) 1990-01-26 1996-10-01 Abbott Lab IMPROVED PROCEDURE TO AMPLIFY WHITE NUCLEIC ACIDS APPLICABLE FOR THE REACTION IN THE POLYMERASE AND LIGASE CHAIN.
US5427930A (en) 1990-01-26 1995-06-27 Abbott Laboratories Amplification of target nucleic acids using gap filling ligase chain reaction
ATE126535T1 (en) 1990-04-05 1995-09-15 Roberto Crea ''WALK-THROUGH'' MUTAGENesis.
AU7791991A (en) 1990-04-24 1991-11-11 Stratagene Methods for phenotype creation from multiple gene populations
US5877402A (en) 1990-05-01 1999-03-02 Rutgers, The State University Of New Jersey DNA constructs and methods for stably transforming plastids of multicellular plants and expressing recombinant proteins therein
US5427908A (en) * 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5851813A (en) 1990-07-12 1998-12-22 President And Fellows Of Harvard College Primate lentivirus antigenic compositions
ES2128323T3 (en) * 1990-07-24 1999-05-16 Hoffmann La Roche REDUCTION OF NON-SPECIFIC AMPLIFICATION DURING IN VITRO AMPLIFICATION OF NUCLEIC ACID USING MODIFIED NUCLEIC ACID BASES.
US5169764A (en) 1990-08-08 1992-12-08 Regeneron Pharmaceuticals, Inc. Multitrophic and multifunctional chimeric neurotrophic factors, and nucleic acids and plasmids encoding the chimeras
CA2089497A1 (en) * 1990-08-15 1992-02-16 Lendon Payne Self-assembling replication defective hybrid virus particles
US5264563A (en) 1990-08-24 1993-11-23 Ixsys Inc. Process for synthesizing oligonucleotides with random codons
ATE174598T1 (en) 1990-08-24 1999-01-15 Ixsys Inc METHOD FOR PRODUCING OLIGONUCLEOTIDES WITH RANDOM CODONS
CA2049879A1 (en) 1990-08-30 1992-03-01 Keith C. Backman Controlled initial target-dependent production of templates for ligase chain reaction
US5871974A (en) 1990-09-28 1999-02-16 Ixsys Inc. Surface expression libraries of heteromeric receptors
IL99553A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing oligonucleotides linked to expression elements,a kit for the preparation of vectors useful for the expression of a diverse population of random peptides and methods utilizing the same
US5770434A (en) 1990-09-28 1998-06-23 Ixsys Incorporated Soluble peptides having constrained, secondary conformation in solution and method of making same
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
US5858725A (en) 1990-10-10 1999-01-12 Glaxo Wellcome Inc. Preparation of chimaeric antibodies using the recombinant PCR strategy
US5187083A (en) 1990-11-13 1993-02-16 Specialty Laboratories, Inc. Rapid purification of DNA
US5234824A (en) 1990-11-13 1993-08-10 Specialty Laboratories, Inc. Rapid purification of DNA
US5489523A (en) 1990-12-03 1996-02-06 Stratagene Exonuclease-deficient thermostable Pyrococcus furiosus DNA polymerase I
CA2095633C (en) 1990-12-03 2003-02-04 Lisa J. Garrard Enrichment method for variant proteins with altered binding properties
IL100460A (en) 1990-12-20 1997-06-10 Ixsys Method for optimization of binding proteins and nucleic acids encoding a binding protein produced thereby
US5795747A (en) 1991-04-16 1998-08-18 Evotec Biosystems Gmbh Process for manufacturing new biopolymers
DE4112440C1 (en) 1991-04-16 1992-10-22 Diagen Institut Fuer Molekularbiologische Diagnostik Gmbh, 4000 Duesseldorf, De
US5514568A (en) 1991-04-26 1996-05-07 Eli Lilly And Company Enzymatic inverse polymerase chain reaction
US5512463A (en) 1991-04-26 1996-04-30 Eli Lilly And Company Enzymatic inverse polymerase chain reaction library mutagenesis
DE69213112T2 (en) 1991-06-20 1997-04-03 Hoffmann La Roche Improved methods for nucleic acid amplification
WO1993000103A1 (en) 1991-06-21 1993-01-07 The Wistar Institute Of Anatomy And Biology Chimeric envelope proteins for viral targeting
WO1993001282A1 (en) * 1991-07-01 1993-01-21 Berlex Laboratories, Inc. Novel mutagenesis methods and compositions
US6071889A (en) 1991-07-08 2000-06-06 Neurospheres Holdings Ltd. In vivo genetic modification of growth factor-responsive neural precursor cells
US5223408A (en) 1991-07-11 1993-06-29 Genentech, Inc. Method for making variant secreted proteins with altered properties
GB9115364D0 (en) 1991-07-16 1991-08-28 Wellcome Found Antibody
US5279952A (en) 1991-08-09 1994-01-18 Board Of Regents, The University Of Texas System PCR-based strategy of constructing chimeric DNA molecules
ATE148889T1 (en) 1991-09-18 1997-02-15 Affymax Tech Nv METHOD FOR SYNTHESIS OF VARIOUS COLLECTIONS OF OLIGOMERS
DE69229477T2 (en) 1991-09-23 1999-12-09 Cambridge Antibody Tech Methods for the production of humanized antibodies
ES2136092T3 (en) * 1991-09-23 1999-11-16 Medical Res Council PROCEDURES FOR THE PRODUCTION OF HUMANIZED ANTIBODIES.
US5872215A (en) * 1991-12-02 1999-02-16 Medical Research Council Specific binding members, materials and methods
GB9125979D0 (en) 1991-12-06 1992-02-05 Wellcome Found Antibody
EP0625205A1 (en) 1992-01-30 1994-11-23 Genzyme Limited Chiral synthesis with modified enzymes
US5773267A (en) * 1992-02-07 1998-06-30 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University D29 shuttle phasmids and uses thereof
GB9202796D0 (en) 1992-02-11 1992-03-25 Wellcome Found Antiviral antibody
US5843643A (en) 1992-02-21 1998-12-01 Ratner; Paul L. Site-specific transfection of eukaryotic cells using polypeptide-linked recombinant nucleic acid
EP0632829A1 (en) 1992-03-02 1995-01-11 Biogen, Inc. Thrombin receptor antagonists
WO1993018175A1 (en) * 1992-03-10 1993-09-16 Life Technologies, Inc. Udg-facilitated mutagenesis
EP1477558A1 (en) 1992-03-16 2004-11-17 WOHLSTADTER, Jacob Nathaniel Selection methods
JP3507073B2 (en) 1992-03-24 2004-03-15 ケンブリッジ アンティボディー テクノロジー リミティド Methods for producing members of a specific binding pair
DE69329641T2 (en) 1992-03-31 2001-06-21 Abbott Lab METHOD FOR MULTIPLE LIGASE CHAIN REACTION
US5316935A (en) 1992-04-06 1994-05-31 California Institute Of Technology Subtilisin variants suitable for hydrolysis and synthesis in organic media
US5474896A (en) * 1992-05-05 1995-12-12 Institut Pasteur Nucleotide sequence encoding the enzyme I-SceI and the uses thereof
AU4387193A (en) 1992-05-29 1993-12-30 Abbott Laboratories Ligase chain reaction starting with rna sequences
JPH06509947A (en) 1992-06-15 1994-11-10 シティ・オブ・ホープ Chimeric anti-CEA antibody
CA2139876A1 (en) 1992-07-10 1994-01-20 John Rambosek Recombinant dna encoding a desulfurization biocatalyst
US6107062A (en) 1992-07-30 2000-08-22 Inpax, Inc. Antisense viruses and antisense-ribozyme viruses
EP0627932B1 (en) 1992-11-04 2002-05-08 City Of Hope Antibody construct
DE4239311C2 (en) 1992-11-23 1996-04-18 Guehring Joerg Dr Drills, especially pointed drilling tools with exchangeable cutting inserts
WO1994012632A1 (en) * 1992-11-27 1994-06-09 University College London Improvements in nucleic acid synthesis by pcr
US5360728A (en) 1992-12-01 1994-11-01 Woods Hole Oceanographic Institution (W.H.O.I.) Modified apoaequorin having increased bioluminescent activity
EP0672142B1 (en) 1992-12-04 2001-02-28 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
US5541311A (en) 1992-12-07 1996-07-30 Third Wave Technologies, Inc. Nucleic acid encoding synthesis-deficient thermostable DNA polymerase
JP2879303B2 (en) * 1993-01-14 1999-04-05 佑 本庶 Method for preparing cDNA library, novel polypeptide and DNA encoding the same
US5571708A (en) 1993-04-19 1996-11-05 Bristol-Myers Squibb Company Thrombin-activatable plasminogen activator
US5501962A (en) 1993-06-21 1996-03-26 G. D. Searle & Co. Interleuken-3 (IL-3) human/murine hybrid polypeptides and recombinant production of the same
IT1264712B1 (en) 1993-07-13 1996-10-04 Eniricerche Spa BIOLOGICAL SYNTHESIS METHOD OF PEPTIDES
US5547859A (en) 1993-08-02 1996-08-20 Goodman; Myron F. Chain-terminating nucleotides for DNA sequencing methods
US5498531A (en) 1993-09-10 1996-03-12 President And Fellows Of Harvard College Intron-mediated recombinant techniques and reagents
US6015686A (en) 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
IL107268A0 (en) 1993-10-12 1994-01-25 Yeda Res & Dev Molecules influencing the shedding of the tnf receptor, their preparation and their use
JPH09507121A (en) 1993-10-26 1997-07-22 アフィマックス テクノロジーズ ナームロゼ ベノートスハップ Nucleic acid probe array on biological chip
US6027877A (en) 1993-11-04 2000-02-22 Gene Check, Inc. Use of immobilized mismatch binding protein for detection of mutations and polymorphisms, purification of amplified DNA samples and allele identification
US5556772A (en) 1993-12-08 1996-09-17 Stratagene Polymerase compositions and uses thereof
DE4343591A1 (en) 1993-12-21 1995-06-22 Evotec Biosystems Gmbh Process for the evolutionary design and synthesis of functional polymers based on shape elements and shape codes
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5928905A (en) 1995-04-18 1999-07-27 Glaxo Group Limited End-complementary polymerase reaction
US6165793A (en) 1996-03-25 2000-12-26 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6117679A (en) * 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6406855B1 (en) * 1994-02-17 2002-06-18 Maxygen, Inc. Methods and compositions for polypeptide engineering
US6395547B1 (en) * 1994-02-17 2002-05-28 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US5837458A (en) * 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US6309883B1 (en) 1994-02-17 2001-10-30 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5521077A (en) 1994-04-28 1996-05-28 The Leland Stanford Junior University Method of generating multiple protein variants and populations of protein variants prepared thereby
US5728821A (en) * 1994-08-04 1998-03-17 Bristol-Myers Squibb Company Mutant BR96 antibodies reactive with human carcinomas
WO1996017056A1 (en) * 1994-12-02 1996-06-06 Institut Pasteur Hypermutagenesis
US5514588A (en) 1994-12-13 1996-05-07 Exxon Research And Engineering Company Surfactant-nutrients for bioremediation of hydrocarbon contaminated soils and water
US5629179A (en) 1995-03-17 1997-05-13 Novagen, Inc. Method and kit for making CDNA library
KR19990008000A (en) 1995-04-24 1999-01-25 로버트 에스. 화이트 헤드 How to create a new metabolic pathway and screen it
US5885813A (en) 1995-05-31 1999-03-23 Amersham Life Science, Inc. Thermostable DNA polymerases
CA2151183C (en) * 1995-06-07 2005-05-17 Slawomir Jonasz Composite compression molded article, composition therefor and process for manufacture thereof, and use
US5667974A (en) 1995-06-07 1997-09-16 Abbott Laboratories Method for detecting nucleic acid sequences using competitive amplification
US5958672A (en) 1995-07-18 1999-09-28 Diversa Corporation Protein activity screening of clones having DNA from uncultivated microorganisms
US6057103A (en) 1995-07-18 2000-05-02 Diversa Corporation Screening for novel bioactivities
US6168919B1 (en) 1996-07-17 2001-01-02 Diversa Corporation Screening methods for enzymes and enzyme kits
US6004788A (en) 1995-07-18 1999-12-21 Diversa Corporation Enzyme kits and libraries
US6030779A (en) 1995-07-18 2000-02-29 Diversa Corporation Screening for novel bioactivities
EP0843725B1 (en) 1995-08-11 2002-04-17 Novozymes A/S Method for preparing polypeptide variants
US5962258A (en) 1995-08-23 1999-10-05 Diversa Corporation Carboxymethyl cellulase fromthermotoga maritima
US6051409A (en) 1995-09-25 2000-04-18 Novartis Finance Corporation Method for achieving integration of exogenous DNA delivered by non-biological means to plant cells
US5756316A (en) 1995-11-02 1998-05-26 Genencor International, Inc. Molecular cloning by multimerization of plasmids
US5939250A (en) 1995-12-07 1999-08-17 Diversa Corporation Production of enzymes having desired activities by mutagenesis
US6352842B1 (en) 1995-12-07 2002-03-05 Diversa Corporation Exonucease-mediated gene assembly in directed evolution
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
US5965408A (en) 1996-07-09 1999-10-12 Diversa Corporation Method of DNA reassembly by interrupting synthesis
US6238884B1 (en) 1995-12-07 2001-05-29 Diversa Corporation End selection in directed evolution
US20030215798A1 (en) 1997-06-16 2003-11-20 Diversa Corporation High throughput fluorescence-based screening for novel enzymes
US6358709B1 (en) 1995-12-07 2002-03-19 Diversa Corporation End selection in directed evolution
US6171820B1 (en) 1995-12-07 2001-01-09 Diversa Corporation Saturation mutagenesis in directed evolution
US5871969A (en) * 1996-02-12 1999-02-16 Human Genome Sciences, Inc. Nucleic acids encoding human neuronal attachment factor-1
US6096548A (en) 1996-03-25 2000-08-01 Maxygen, Inc. Method for directing evolution of a virus
US5783431A (en) 1996-04-24 1998-07-21 Chromaxome Corporation Methods for generating and screening novel metabolic pathways
US6242222B1 (en) * 1996-06-07 2001-06-05 Massachusetts Institute Of Technology Programmed sequential mutagenesis
US5763239A (en) 1996-06-18 1998-06-09 Diversa Corporation Production and use of normalized DNA libraries
US6093873A (en) 1996-08-19 2000-07-25 Institut National De La Sante Et De La Recherche Medicale Genetically engineered mice containing alterations in the gene encoding RXR
US6326204B1 (en) * 1997-01-17 2001-12-04 Maxygen, Inc. Evolution of whole cells and organisms by recursive sequence recombination
US6159687A (en) 1997-03-18 2000-12-12 Novo Nordisk A/S Methods for generating recombined polynucleotides
US5948653A (en) 1997-03-21 1999-09-07 Pati; Sushma Sequence alterations using homologous recombination
US6153410A (en) 1997-03-25 2000-11-28 California Institute Of Technology Recombination of polynucleotide sequences using random or defined primers
US6051049A (en) 1997-05-29 2000-04-18 Exothermic Distribution Corporation Utilization of strontium aluminate in steelmaking
US6087341A (en) 1998-02-12 2000-07-11 The Board Of Trustees Of The Leland Standford Junior University Introduction of nucleic acid into skin cells by topical application
US6365408B1 (en) 1998-06-19 2002-04-02 Maxygen, Inc. Methods of evolving a polynucleotides by mutagenesis and recombination
US6436675B1 (en) * 1999-09-28 2002-08-20 Maxygen, Inc. Use of codon-varied oligonucleotide synthesis for synthetic shuffling
US6376246B1 (en) * 1999-02-05 2002-04-23 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
EP1152058A1 (en) * 2000-05-03 2001-11-07 Institut Curie Methods and compositions for effecting homologous recombination
US6355160B1 (en) 2000-07-21 2002-03-12 Cecil A. Wiseman Gray-water recycling system

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6335160B1 (en) * 1995-02-17 2002-01-01 Maxygen, Inc. Methods and compositions for polypeptide engineering

Also Published As

Publication number Publication date
US7776598B2 (en) 2010-08-17
DK2202308T3 (en) 2013-03-25
JP5008784B2 (en) 2012-08-22
US20110053781A1 (en) 2011-03-03
US20060223143A1 (en) 2006-10-05
US20050059109A1 (en) 2005-03-17
US7534564B2 (en) 2009-05-19
US6455253B1 (en) 2002-09-24
JP2001506855A (en) 2001-05-29
JP2009089710A (en) 2009-04-30
US6946296B2 (en) 2005-09-20
ATE481480T1 (en) 2010-10-15
US6653072B1 (en) 2003-11-25
EP2202308B1 (en) 2013-02-13
EP0946755A4 (en) 2001-03-07
EP1149905B1 (en) 2010-09-15
EP0946755A1 (en) 1999-10-06
US20040248253A1 (en) 2004-12-09
WO1998027230A1 (en) 1998-06-25
EP1149904A1 (en) 2001-10-31
CA2274319C (en) 2013-04-09
AU732146B2 (en) 2001-04-12
DE69739996D1 (en) 2010-10-28
ES2351673T3 (en) 2011-02-09
CA2274319A1 (en) 1998-06-25
AU5729298A (en) 1998-07-15
US20060084091A1 (en) 2006-04-20
US20090149331A1 (en) 2009-06-11
US20070031878A1 (en) 2007-02-08
IL130190A0 (en) 2000-06-01
EP2202308A3 (en) 2011-11-30
US20020051976A1 (en) 2002-05-02
EP1231269A1 (en) 2002-08-14
US6586182B1 (en) 2003-07-01
US6319713B1 (en) 2001-11-20
US6579678B1 (en) 2003-06-17
US20060166225A1 (en) 2006-07-27
KR20000069591A (en) 2000-11-25
US6613514B2 (en) 2003-09-02
US6355484B1 (en) 2002-03-12
CA2589337A1 (en) 1998-06-25
EP2202308A2 (en) 2010-06-30
US6303344B1 (en) 2001-10-16
EP1149905A1 (en) 2001-10-31
DK1149905T3 (en) 2010-11-01
US20030027156A1 (en) 2003-02-06
US20120015820A1 (en) 2012-01-19
US6335160B1 (en) 2002-01-01

Similar Documents

Publication Publication Date Title
US6653072B1 (en) Methods and compositions for polypeptide engineering
US6406855B1 (en) Methods and compositions for polypeptide engineering
US20070009930A1 (en) Methods and compositions for polypeptide engineering
AU2011202018B2 (en) Methods and compositions for polypeptide engineering
AU2761602A (en) Methods and compositions for polypeptide engineering
AU5433601A (en) Methods and compositions for polypeptide engineering
MXPA99005653A (en) Methods and compositions for polypeptide engineering

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: CODEXIS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CODEXIS MAYFLOWER HOLDINGS, LLC;REEL/FRAME:066528/0932

Effective date: 20240206