US20100291066A1 - Donor specific antibody libraries - Google Patents

Donor specific antibody libraries Download PDF

Info

Publication number
US20100291066A1
US20100291066A1 US12/676,737 US67673708A US2010291066A1 US 20100291066 A1 US20100291066 A1 US 20100291066A1 US 67673708 A US67673708 A US 67673708A US 2010291066 A1 US2010291066 A1 US 2010291066A1
Authority
US
United States
Prior art keywords
donor
antibody
library
specific antibody
fragments
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
US12/676,737
Inventor
Lawrence Horowitz
Ramesh Bhatt
Arun K. Kashyap
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.)
I2 Pharmaceuticals Inc
Original Assignee
Sea Lane Biotechnologies LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sea Lane Biotechnologies LLC filed Critical Sea Lane Biotechnologies LLC
Priority to US12/676,737 priority Critical patent/US20100291066A1/en
Assigned to SEA LANE BIOTECHNOLOGIES, LLC reassignment SEA LANE BIOTECHNOLOGIES, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BHATT, RAMESH, HOROWITZ, LAWRENCE, KASHYAP, ARUN K.
Publication of US20100291066A1 publication Critical patent/US20100291066A1/en
Assigned to ALTUS BIOPHARMA INC. reassignment ALTUS BIOPHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SEA LANE BIOTECHNOLOGIES, LLC.
Assigned to I2 PHARMACEUTICALS, INC. reassignment I2 PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALTUS BIOPHARMA INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/005Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies constructed by phage libraries
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1018Orthomyxoviridae, e.g. influenza virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • C07K16/1045Lentiviridae, e.g. HIV, FIV, SIV
    • C07K16/1063Lentiviridae, e.g. HIV, FIV, SIV env, e.g. gp41, gp110/120, gp160, V3, PND, CD4 binding site
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B20/00Methods specially adapted for identifying library members
    • C40B20/04Identifying library members by means of a tag, label, or other readable or detectable entity associated with the library members, e.g. decoding processes
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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)

Definitions

  • the present invention concerns donor-specific antibody libraries and methods of making and using thereof.
  • the present invention also concerns neutralizing antibodies obtained from such donor-specific antibody libraries and methods of using the antibodies obtained for the prevention and/or treatment of various human diseases and conditions.
  • human antibody-based drug candidates are typically identified by screening of human antibody libraries comprising a random collection of antibody sequences from human repertoires that are typically unrelated to their intended application or applications.
  • Each antibody library created from a specific human donor potentially contains antibodies to every component, physiology, and metabolic alteration stemming from, or creating, every unique challenge that the donor has encountered, challenged, and surmounted over the course of that individual's lifetime.
  • typical human antibody libraries made with the current approaches are constructed without the knowledge of the health history of donors, little is known of what would be expected in the resulting immunoglobulin repertoires.
  • the present invention addresses these needs by providing methods and means for the creation, screening and handling of donor-specific antibody libraries from individuals who have been exposed to and survived or are surviving an encounter with a specific target disease.
  • the invention concerns a vector collection comprising a repertoire of nucleic acid molecules encoding antibody light or heavy chains or fragments thereof, derived from a human patient donor who has suffered from, or is suffering from, a disease evoking antibody production to a target antigen, wherein the collection is identified with a unique barcode.
  • the vector collection comprises a repertoire of nucleic acid molecules encoding antibody light chains or fragments thereof, such as antibody ⁇ light chains, or antibody ⁇ light chains, or fragments thereof.
  • the vector collection comprises a repertoire of nucleic acid molecules encoding antibody heavy chains or fragments thereof.
  • the barcode is a nucleotide sequence linked to or incorporated in the vectors present in the collection, and/or linked to or incorporated in the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of the nucleic acid molecules.
  • the barcode may be contiguous non-coding nucleotide sequence of one to about 24 nucleotides, which may, for example, be linked to the 3′ or 5′ non-coding region of the nucleic acid molecules.
  • the barcode is a nucleotide sequence that is a coding sequence of one or more silent mutations incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof.
  • the barcode is a non-contiguous nucleotide sequence. At least part of the non-contiguous nucleotide sequence may be linked to or incorporated in the vectors present in the collection. Alternatively, at least part of the non-contiguous nucleotide sequence may be incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of said nucleic acid molecules.
  • the barcode is a peptide or polypeptide sequence.
  • the vectors present in the vector collection are phagemid vectors, which may, for example, contain a bacteriophage gene III and a stop codon between the nucleic acid molecules encoding antibody light or heavy chains or fragments thereof and the bacteriophage III gene, and may have a barcode, such as a non-coding contiguous nucleotide sequence inserted in the untranslated region following the stop codon.
  • the invention concerns host cells comprising the vector collection of the present invention.
  • the host cells may by eukaryotic or prokaryotic host cells, such as, for example, E. coli cells.
  • the invention concerns a donor-specific antibody library comprising library members expressing a collection of antibodies or antibody fragments to a target antigen wherein the antibodies or antibody fragments are derived from a human donor who has suffered from, or is suffering from, a disease evoking antibody production to said target antigen, wherein said antibody library is identified with at least one unique barcode.
  • the antibody heavy and light chains are separately identified each with a barcode unique to the human donor from whom it derived.
  • the donor-specific antibody library is identified with one unique barcode.
  • the antibodies or antibody fragments are composed of antibody heavy and light chains or fragments thereof encoded by nucleic acid molecules present in a vector.
  • the barcode is a nucleotide sequence linked to or incorporated in the vectors present in the library, and/or linked to or incorporated in the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of the nucleic acid molecules.
  • the barcode is a contiguous non-coding nucleotide sequence of one to about 24 nucleotides, which may, for example, be linked to the 3′ or 5′ non-coding region of the nucleic acid molecules.
  • the barcode is encoded by a coding sequence of one or more silent mutations incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof.
  • the barcode is encoded by a non-contiguous nucleotide sequence.
  • a least part of the non-contiguous sequence encoding the barcode is linked to or incorporated in the vectors present in the library.
  • At least part of the non-contiguous sequence encoding the barcode is incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of such nucleic acid molecules.
  • the barcode is a peptide or polypeptide sequence.
  • vectors are phagemid vectors, may, for example, contain a bacteriophage gene III and a stop codon between the nucleic acid molecules encoding antibody light or heavy chains or fragments thereof and the bacteriophage II gene.
  • the medical history of the human patient donor shows that the donor has suffered from, or is suffering from said disease. In some embodiments, it is independently confirmed that the human donor suffered from, or is suffering from the disease.
  • the donor-specific antibody library is substantially devoid of antibodies and antibody fragments specifically binding antigens different from said target antigen.
  • the target antigen is an influenza A virus, such as an isolate of influenza A virus H1, H2, H3, H5, H7, or H9 subtype.
  • the library expresses at least one antibody or antibody fragment specifically binding to more than one influenza A virus subtype.
  • the library expresses at least one antibody or antibody fragment binding to and neutralizing the H5N1 subtype of influenza virus A.
  • the human donor has suffered from, or is suffering from a disease selected from the group consisting of the diseases listed in Table 1 below.
  • the antibody library expresses at least one antibody or antibody fragment binding to an antigen associated with the target disease.
  • the antibody library expresses at least one antibody or antibody fragment binding to and neutralizing an antigen associated with the target disease.
  • the donor-specific antibody library may, for example, be a phage library, in an embodiment, it may contain sequences encoding more than 10 6 different members of antibodies or antibody fragments, or more than 10 9 different members of antibodies or antibody fragments.
  • the donor-specific antibody library is, without limitation, a spore-display library, a ribosome display library, a mRNA display library, a microbial cell display library, a yeast display library, or a mammalian display library.
  • the nucleic acid encoding the antibodies or antibody fragments present in the library is reverse-transcribed from mRNA extracted from lymphocytes of the human patient donor, where the lymphocytes may, for example, originate from bone marrow, blood, spleen, or lymph node.
  • a serological profile of said human donor may be generated prior to extraction of said mRNA.
  • the medical history of the human donor, and optionally the donor's family, is examined prior to or following extraction of the mRNA.
  • the invention concerns a method of making a donor-specific library expressing a collection of antibodies or antibody fragments to a target antigen, comprising the steps of:
  • the method may further comprise steps of generating a serological profile of said patient and/or examining medical history of the patient prior or subsequent to step a).
  • the method may further comprise the steps of d) inserting said nucleic acids into expression vector: e) expressing the immunoglobulin repertoire; and f) displaying the immunoglobulin repertoire in a display system.
  • the method further comprises the step of selecting members of the library based their ability to neutralize or activate the target antigen.
  • the method yields at least one neutralizing antibody.
  • the method further comprises the step of creating one or more sub-libraries comprising library members that were found to neutralize or activate the target antigen.
  • the method comprises the step of sequencing at least one library member identified.
  • the invention concerns a method of treating or preventing a disease associated with a target antigen neutralized or activated by an antibody selected by the method described above, comprising administering to a human patient in need an effective amount of the antibody selected.
  • the antibody may, for example, be a neutralizing antibody, such as a neutralizing antibody to at least one influenza A virus subtype.
  • the disease is selected from the group consisting of the diseases listed in Table 1 below.
  • FIG. 1 is a flow chart schematically illustrating a typical method for the creatin of the human antibody libraries of the present invention.
  • FIG. 2 illustrates a typical panning enrichment scheme for increasing the reactive strength towards two different targets, A and B. Each round of enrichment increases the reactive strength of the pool towards the individual target(s).
  • FIG. 3 illustrates a strategy for the selection of clones cross-reactive with targets A and B, in which each successive round reinforces the reactive strength of the resulting pool towards both targets.
  • FIG. 4 illustrates a strategy for increasing the reactive strengths towards two different targets (targets A and B), by recombining parallel discovery pools to generate/increase cross-reactivity. Each round of selection of the recombined antibody library increases the reactive strength of the resulting pool towards both targets.
  • FIG. 5 illustrates a strategy for increasing cross-reactivity to a target B while maintaining reactivity to a target A.
  • a clone reactive with target A is selected, then a mutagenic library of the clones reactive with target A is prepared, and selection is performed as shown, yielding one or more antibody clones that show strong reactivity with both target A and target B.
  • FIG. 6 illustrates a representative mutagenesis method for generating a diverse multifunctional antibody collection by the “destinational mutagenesis” method.
  • FIG. 7 shows the amino acid sequences of 15 known hemagglutinin (H) protein subtypes.
  • FIG. 8 shows the H5 hemagglutinin (HA) serology results for blood samples obtained from six human survivors of a Turkish H5N1 bird flu outbreak. The data demonstrate the presence of antibodies to the HA antigen.
  • HA hemagglutinin
  • FIG. 9 shows serology results obtained with serum samples of twelve local donors, tested on H5 antigen (A/Vietnam/1203/2004) and H1N1 (A/New Caledonia/20/99) and H3N2 (A/Panama/2007/99) viruses.
  • FIG. 10 illustrates the unique barcoding approach used in the construction of antibody phage libraries.
  • FIG. 11 shows the analysis of antibody binding to hemagglutinins from different influenza A subtypes.
  • FIG. 12 shows the positions of H5 hemagglutinin binding Group 1 required and dominant mutations on the crystal structure at Fab 47e.
  • FIGS. 13 and 14 illustrate the use of destinational mutagenesis to create diverse antibody heavy and light chain libraries using the antibody heavy and light chain sequences identified by analysis of sera and bone marrow of Turkish bird flu survivors.
  • amino acid residues are used to refer to amino acid residues that are identical between two or more amino acid sequences aligned with each other.
  • disease refers to any disruption of normal body function, or the appearance of any type of pathology.
  • the etiological agent causing the disruption of normal physiology may or may not be known.
  • two patients may be diagnosed with the same disorder, the particular symptoms displayed by those individuals may or may not be identical.
  • an “effective amount” is an amount sufficient to effect beneficial or desired therapeutic (including preventative) results.
  • An effective amount can be administered in one or more administrations.
  • composition is defined as comprising an active ingredient, such as a neutralizing antibody generated from the present invention, and at least one additive, such as a pharmaceutically acceptable carrier, including, without limitation, water, minerals, proteins, and/or other excipients known to one skilled in the art.
  • active ingredient such as a neutralizing antibody generated from the present invention
  • additive such as a pharmaceutically acceptable carrier, including, without limitation, water, minerals, proteins, and/or other excipients known to one skilled in the art.
  • treating or “treatment” is intended to mean an amelioration of a clinical symptom indicative of a disease.
  • preventing or “prevention” is intended to mean a forestalling of a clinical symptom indicative of a disease
  • subject and “patient,” as used herein, are used interchangeably, and can refer to any to animal, and preferably a mammal, that is the subject of an examination, treatment, analysis, test or diagnosis.
  • subjects or patients include humans, non-human primates and other mammals, who may or may not have a disease or other pathological condition.
  • amino acid typically refers to an amino acid having its art recognized definition such as an amino acid selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gln); glutamic acid (Glu); glycine (Gly); histidine (His); isoleucine (Ile): leucine (Leu): lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val) although modified, synthetic, or rare amino acids may be used as desired.
  • amino acids can be subdivided into various sub-groups.
  • amino acids can be grouped as having a nonpolar side chain (e.g., Ala, Cys, Ile, Leu, Met, Phe, Pro, Val); a negatively charged side chain (e.g., Asp, Glu); a positively charged side chain (e.g., Arg, His, Lys); or an uncharged polar side chain (e.g., Asn, Cys, Gln, Gly, His, Met, Phe, Ser, Thr, Trp, and Tyr).
  • a nonpolar side chain e.g., Ala, Cys, Ile, Leu, Met, Phe, Pro, Val
  • a negatively charged side chain e.g., Asp, Glu
  • a positively charged side chain e.g., Arg, His, Lys
  • an uncharged polar side chain e.g., Asn, Cys, Gln, Gly, His, Met, Phe, Ser,
  • Amino acids can also be grouped as small amino acids (Gly, Ala), nucleophilic amino acids (Ser, His, Thr, Cys), hydrophobic amino acids (Val, Leu. Ile. Met, Pro), aromatic amino acids (Phe, Tyr, Trp, Asp, Glu), amides (Asp, Glu), and basic amino acids (Lys, Arg).
  • variant refers to a polypeptide that possesses at least one amino acid mutation or modification (i.e., alteration) as compared to a native polypeptide.
  • variants generated by “amino acid modifications” can be produced, for example, by substituting, deleting, inserting and/or chemically modifying at least one amino acid in the native amino acid sequence.
  • amino acid modification refers to a change in the amino acid sequence of a predetermined amino acid sequence.
  • exemplary modifications include an amino acid substitution, insertion and/or deletion.
  • amino acid modification at refers to the substitution or deletion of the specified residue, or the insertion of at least one amino acid residue adjacent to the specified residue.
  • insertion “adjacent” to a specified residue is meant insertion within one to two residues thereof. The insertion may be N-terminal or C-terminal to the specified residue.
  • amino acid substitution refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence with another different “replacement” amino acid residue.
  • the replacement residue or residues may be “naturally occurring amino acid residues” (i.e. encoded by the genetic code) and selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gln); glutamic acid (Glu); glycine (Gly); histidine (His): isoleucine (Ile): leucine (Leu); lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val). Substitution with one or more non-naturally occurring amino acid residues is also encompass
  • non-naturally occurring amino acid residue refers to a residue, other than those naturally occurring amino acid residues listed above, which is able to covalently bind adjacent amino acid residues(s) in a polypeptide chain.
  • non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine and other amino acid residue analogues such as those described in Ellman et al. Meth. Enzym. 202:301 336 (1991).
  • the procedures of Noren et al. Science 244:182 (1989) and Ellman et al., supra can be used. Briefly, these procedures involve chemically activating a suppressor tRNA with a non-naturally occurring amino acid residue followed by in vitro transcription and translation of the RNA.
  • amino acid insertion refers to the incorporation of at least one amino acid into a predetermined amino acid sequence. While the insertion will usually consist of the insertion of one or two amino acid residues, the present application contemplates larger “peptide insertions”, e.g. insertion of about three to about live or even up to about ten amino acid residues.
  • the inserted residue(s) may be naturally occurring or non-naturally occurring as disclosed above.
  • amino acid deletion refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.
  • polynucleotide(s) refers to nucleic acids such as DNA molecules and RNA molecules and analogs thereof (e.g., DNA or RNA generated using nucleotide analogs or using nucleic acid chemistry).
  • the polynucleotides may be made synthetically, e.g. using art-recognized nucleic acid chemistry or enzymatically using, e.g., a polymerase, and, if desired, be modified. Typical modifications include methylation, biotinylation, and other art-known modifications.
  • the nucleic acid molecule can be single-stranded or double-stranded and, where desired, linked to a detectable moiety.
  • mutagenesis refers to unless otherwise specified, any art recognized technique for altering a polynucleotide or polypeptide sequence. Preferred types of mutagenesis include error prone PCR mutagenesis, saturation mutagenesis, or other site directed mutagenesis.
  • Site-directed mutagenesis is a technique standard in the art, and is conducted using a synthetic oligonucleotide primer complementary to a single-stranded phage DNA to be mutagenized except for limited mismatching, representing the desired mutation. Briefly, the synthetic oligonucleotide is used as a primer to direct synthesis of a strand complementary to the single-stranded phage DNA, and the resulting double-stranded DNA is transformed into a phage-supporting host bacterium. Cultures of the transformed bacteria are plated in top agar, permitting plaque formation from single cells that harbor the phage.
  • Plaques of interest are selected by hybridizing with kinased synthetic primer at a temperature that permits hybridization of an exact match, but at which the mismatches with the original strand are sufficient to prevent hybridization. Plaques that hybridize with the probe are then selected, sequenced and cultured, and the DNA is recovered.
  • vector is used to refer to a rDNA molecule capable of autonomous replication in a cell and to which a DNA segment, e.g., gene or polynucleotide, can be operatively linked so as to bring about replication of the attached segment.
  • vectors capable of directing the expression of genes encoding for one or more polypeptides are referred to herein as “expression vectors.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • a nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • Percent amino acid sequence identity may be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al., Nucleic Acids Res. 25:3389-3402 (1997)).
  • NCBI-BLAST2 sequence comparison program may be downloaded from http://www.ncbi.nlm.nih.gov or otherwise obtained from the National Institute of Health. Bethesda, Md.
  • Antibody-dependent cell-mediated cytotoxicity and “ADCC” are used herein to refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express FcRs (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs e.g. Natural Killer (NK) cells, neutrophils, and macrophages
  • NK cells Natural Killer
  • neutrophils e.g., neutrophils, and macrophages
  • FcRs Fc receptors
  • influenza A subtype or “influenza A virus subtype” are used interchangeably, and refer to influenza A virus variants that are characterized by various combinations of the hemagglutinin (H) and neuraminidase (N) viral surface proteins, and thus are labeled by a combination of an H number and an N number, such as, for example, H1N1 and H3N2.
  • H hemagglutinin
  • N neuraminidase
  • the terms specifically include all strains (including extinct strains) within each subtype, which usually result from mutations and show different pathogenic profiles.
  • Such strains will also be referred to as various “isolates” of a viral subtype, including all past, present and future isolates. Accordingly, in this context, the terms “strain” and “isolate” are used interchangeably.
  • influenza is used to refer to a contagious disease caused by an influenza virus.
  • Mutagenesis can, for example, be performed using site-directed mutagenesis (Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)).
  • PCR amplification methods are described in U.S. Pat. Nos. 4,683,192, 4,683,202, 4,800,159, and 4,965,188, and in several textbooks including “PCR Technology: Principles and Applications for DNA Amplification”, H. Erlich, ed., Stockton Press, New York (1989); and PCR Protocols: A Guide to Methods and Applications , Innis et al., eds., Academic Press, San Diego, Calif. (1990).
  • antibodies of the present invention can also be identified by other display and enrichment technologies, such as, for example, ribosome or mRNA display (Mattheakis et al., Proc. Natl. Acad. Sci. USA 91:9022-9026 (1994); H-lanes and Pluckthun, Proc. Natl. Acad. Sci. USA 94:4937-4942 (1997)), microbial cell display, such as bacterial display (Georgiou et al., Nature Biotech. 15:29-34 (1997)), or yeast cell display (Kieke et al., Protein Eng.
  • ribosome or mRNA display Moattheakis et al., Proc. Natl. Acad. Sci. USA 91:9022-9026 (1994); H-lanes and Pluckthun, Proc. Natl. Acad. Sci. USA 94:4937-4942 (1997)
  • microbial cell display such as bacterial display (Georgiou e
  • ribosome display the antibody and the encoding mRNA are linked by the ribosome, which at the end of translating the mRNA is made to stop without releasing the polypeptide. Selection is based on the ternary complex as a whole.
  • a covalent bond between an antibody and the encoding mRNA is established via puromycin, used as an adaptor molecule (Wilson et al., Proc. Natl. Acad. Sci. USA 98:3750-3755 (2001)).
  • puromycin used as an adaptor molecule
  • Lipovsek and Pluckthun J. Immunol. Methods. 290:51-67 (2004).
  • Microbial cell display techniques include surface display on a yeast, such as Saccharomyces cerevisiae (Boder and Wittrup. Nat. Biotechnol. 15:553-557 (1997)).
  • a yeast such as Saccharomyces cerevisiae (Boder and Wittrup. Nat. Biotechnol. 15:553-557 (1997)).
  • antibodies can be displayed on the surface of S. cerevisiae via fusion to the ⁇ -agglutinin yeast adhesion receptor, which is located on the yeast cell wall.
  • This method provides the possibility of selecting repertoires by flow cytometry.
  • the yeast cells By staining the cells by fluorescently labeled antigen and an anti-epitope tag reagent, the yeast cells can be sorted according to the level of antigen binding and antibody expression on the cell surface.
  • Yeast display platforms can also be combined with phage (see, e.g., Van den Beucken et al., FEBS Leit. 546:288-2
  • the present invention concerns donor-specific antibody libraries from individuals who have successfully survived or are surviving an encounter with a specific disease.
  • the resulting antibody repertoires will include antibodies that were used by the donor to defend specifically against a relevant disease and thus are important tools, for example, for developing neutralizing antibodies for the prevention and/or treatment of a target disease.
  • infectious disorder Influenza viral infection, hepatitis C virus (HCV) infection, herpes simplex virus (HSV) infection, human immunodeficiency virus (HIV) infection, Methicillin-resistant Staphylococcus aureus (MRSA) infection, Epstein-Barr virus (EBV) infection, respiratory syncytial virus (RSV) infection, Pseudomonas.
  • HCV hepatitis C virus
  • HSV herpes simplex virus
  • HCV human immunodeficiency virus
  • MRSA Methicillin-resistant Staphylococcus aureus
  • EBV Epstein-Barr virus
  • RSV respiratory syncytial virus
  • Candida infections Respiratory disorder Asthma Allergies, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), adult respiratory distress syndrome (ARDS) metabolic disorder Frailty, cachexia, sarcopenia, Obesity, type II diabedyslipidemia, metabolic syndrome- associated myocardial infarction (MI), chronic renal failure (CRF), osteoporosis digestive disorder irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), Chron's disease, fatty liver disease, fibrosis, drug-induced liver disease Neurological disorder Alzheimer's disease, multiple sclerosis (MS), Parkinson's disease, bovine spongiform encephalopathy (BSE, mad cow disease) Cancer e.g., breast, renal, stomach, melanoma, lung, colon, glioma, lymphoma
  • FIG. 1 A method of creating the donor-specific libraries of the present invention is schematically illustrated in FIG. 1 .
  • potential donors are identified.
  • the patient donor may currently suffer from or may have recovered from and survived a target disease.
  • the donor-specific libraries herein may be created from the bone marrow of convalescent patients of prior influenza infections, including seasonal influenza outbreaks, epidemics, and pandemics.
  • the presence of antibodies to an influenza virus can be detected by the well-known hemagglutinin inhibition (HAI) assay (i, A. P., M. S. Pereira, and J. J. Skehel. 1982. Concepts and procedures for laboratory - based influenza surveillance . U.S. Department of Health and Human Services, Public Health Service, Centers for Disease Control, Atlanta. Ga.), or the microneutralization assay (Harmon et al., J. Clin. Microbiol. 26:333-337 (1988)).
  • HAI hemagglutinin inhibition
  • samples containing lymphocytes are collected from individuals (patient donors) known to have developed a target disease such as at least one disease from those listed in Table 1.
  • the sample may for example, derive from bone marrow blood, spleen, lymph nodes, tonsils, thymus and the like.
  • Bone marrow is a preferred source of the antibody libraries herein since it represents the complete “fossil archive” of individual donor's mature antibody repertoire.
  • lymphocytes are collected from a patient who has recovered from the targeted disease(s) at least for 1, 5, 10, 15, 20, 25 days, at least for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 months, or at least for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 years.
  • lymphocytes are collected from a patient who is having the targeted disease(s) at the time of collection, and has been diagnosed as having the disease(s) at least 1, 5, 10, 15, 20, 25 days or at least 1, 2, 3, 4, 5, 6, 8, 9, 10 months, or 1, 2, 3, 4, or 5 years prior to collection.
  • Kits for this purpose are well known and commercially available, such as, for example, BD Vacutainer® CPTTM cell preparation tubes can be used for centrifugal purification of lymphocytes, and guanidium. Trizol, or RNAlater used to stabilize the samples. Methods and kits for isolating lymphocytes from other sources, such as lymphoid organs are also well known and commercially available.
  • RNA is extracted and RT-PCR is performed to rescue antibody heavy and light chain repertoires, using immunoglobulin oligo primers known in the art.
  • RNA purification kits are available from commercial manufacturers, such as Qiagen, and can be used according to the manufacturer's instructions.
  • RNA Since RNA cannot serve as a template for PCR, it is first reverse transcribed into cDNA, which is subjected to PCR amplification.
  • the two most commonly used reverse transcriptases are avilo myeloblastosis virus reverse transcriptase (AMV-RT) and Moloney murine leukemia virus reverse transcriptase (MMLV-RT).
  • AMV-RT avilo myeloblastosis virus reverse transcriptase
  • MMLV-RT Moloney murine leukemia virus reverse transcriptase
  • the reverse transcription step is typically primed using specific primers, random hexamers, or oligo-dT primers, depending on the circumstances and the goal of expression profiling.
  • extracted RNA can be reverse-transcribed using a GeneAmp RNA PCR kit (Perkin Elmer. CA, USA), following the manufacturer's instructions.
  • the derived cDNA can then be used as a template in the subsequent PCR reaction.
  • the PCR repertoire products may be combined with linker oligos to generate scFv libraries to clone directly in frame with m13 pIII protein, following procedures known in the art.
  • Libraries using other display techniques, such as those discussed above, can be prepared by methods well known in the art.
  • RNA is extracted by Tri BD reagent (Sigma) from fresh or RNAlater stabilized tissue. Subsequently, the isolated donor total RNA is further purified to mRNA using Oligotex purification (Qiagen).
  • Oligotex purification Qiagen.
  • RNAse Block 100 mM DTT, Accuscript RT, and RNAse Block are added to each reaction and incubated at 42° C. for 1 h, and the reverse transcriptase is inactivated by heating at 70° C. for 15 minutes.
  • the cDNA obtained can be used as a template for RT-PCR amplification of the antibody heavy and light chain V genes, which can then be cloned into a vector, or, if phage display library is intended, into a phagemid vector. This procedure generates a repertoire of antibody heavy and light chain variable region clones (V H and V L libraries), which can be kept separate or combined for screening purposes.
  • the vector such as a phagemid vector
  • a host cell such as an E coli host
  • the vector collection may comprise a single or more than one antibody light chain or heavy chain subtype.
  • the vector collection may comprise sequences encoding antibody ⁇ and/or ⁇ light chains.
  • typically antibody light chains and antibody heavy chains are at first cloned separately, as discussed above, also separating the ⁇ and ⁇ light chain libraries.
  • the libraries can be archived, and, when needed, the heavy chain library can be combined with the segregated ⁇ and ⁇ light chain libraries and heavy and light chain pairings can be identified, e.g. by panning, in the case of phage display. It is possible to repeat these steps multiple times with various libraries or sub-libraries, depending on the goal to be attained.
  • the methods of the present invention provide great flexibility in including or excluding libraries, sub-libraries or clones, as needed during panning in order to maximize success.
  • sequences present in the vector collection harbor the coding sequences of the antibody heavy and light chains (or fragments) separately, the sequences may be excised and inserted into one or more expression vectors for expression of the antibody heavy and light chains, or fragments thereof.
  • the coding sequences of the antibody heavy and light chains, or fragments thereof are inserted into the same expression vector for coexpression of the heavy and light chains to produce the library of the antibodies or antibody fragments.
  • the expression vectors of the present invention contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 ⁇ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
  • Selection genes will typically contain a selection gene, also termed a selectable marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • Suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the antibodies-encoding nucleic acid, such as DHFR or thymidine kinase.
  • An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described by Urlaub et al. Proc. Natl. Acad. Sci. USA. 77:4216 (1980).
  • a suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7 [Stinchcomb et al., Nature, 282:39 (1979); Kingsman et al., Gene, 7:141 (1979); Tschemper et al., Gene, 10:157 (1980)].
  • the trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1 [Jones, Genetics, 85:12 (1977)].
  • Expression and cloning vectors usually contain a promoter operably linked to the antibody-encoding nucleic acid sequence to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the ⁇ -lactamase and lactose promoter systems [Chang et al., Nature, 275:615 (1978); Goeddel et al., Nature, 281:544 (1979)], alkaline phosphatase, a tryptophan (trp) promoter system [Goeddel, Nucleic Acids Res., 8:4057 (1980); EP 36,776], and hybrid promoters such as the tac promoter [deBoer et al., Proc. Natl. Acad. Sci. USA, 80:21-25 (1983)]. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding antibodies
  • Suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase [Hitzeman et al., J. Biol. Chem., 255:2073 (1980)] or other glycolytic enzymes [Hess et al., J. Adv.
  • yeast promoters which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73.657.
  • Transcription of the heavy chain or light chain genes in the expression vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 Jul. 1989), adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems.
  • viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 Jul. 1989), adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus,
  • Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription.
  • Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, ⁇ -fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus.
  • Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5′ or 3′ to the antibody coding sequence, but is preferably located at a site 5′ from the promoter.
  • Expression vectors used in eukaryotic host cells will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding antibody heavy and light chains.
  • the antibody library is produced in the form of a phage library, where the coding sequences of the antibody heavy and light chains, or fragments thereof, are cloned into a phagemid vector, such as a vector comprising the bacteriophage gene III.
  • a phagemid vector such as a vector comprising the bacteriophage gene III.
  • Phagemid vectors are well known and commercially available, including, for example, the pBluescript vector SKII+(Stratagene, Genbank Accession X52328), and other pBluscript vectors. Phage display technology enables the generation of large repertoires of human antibodies, and the biopanning procedure allows the selection of individual antibodies with any desired specificity or other properties.
  • immunoglobulin repertoires from peripheral lymphocytes of survivors of earlier epidemics and pandemics can be retrieved, stabilized, rescued and expressed in a manner similar to that described above.
  • repertoires can be recovered from properly timed vaccinated locally-sourced donors.
  • commercially available bone marrow total RNA or mRNA can be purchased from commercial sources to produce libraries suitable for H1 and H3, and, depending upon the background of donor, also suitable for H2 antibody screening.
  • antibodies can be isolated from biological samples obtained from immunized human donors as well. From immunized patients that have developed titers of antibody recognizing the particular antigen, bone marrow, blood, or another source of lymphocytes, is collected, and antibodies produced are isolated, amplified and expressed as described above.
  • antibody light and heavy chain libraries can be cloned separately.
  • various ⁇ and ⁇ light chain families can be separately pooled and cloned in equimolar amounts.
  • various heavy chain families can be pooled and cloned in equimolar amounts.
  • a typical screen can yield zero, one, or more than one target specific positive clone(s). If a particular combinatorial antibody library or libraries have been exhaustively screened and no further solutions seem attainable this may not be a failure of the heavy and light chain repertoire(s) ability to bind target, but rather the collection may have failed to bring together the necessary heavy and light chain pair required to bind target.
  • a typical rescued repertoire of light chains from any individual may contain between about 10 5 -10 6 unique light chains and between about 10 6 -10 8 unique heavy chains.
  • the possible combinatorial products of such pairings range from 10 11 to 10 14 . Such a collection exceeds the practical limits of most display systems, such as phage display, by several orders of magnitude.
  • a typical screen can yield any number of target specific positive clones.
  • the present invention enables the identification of the origins of any clone by their embedded barcode.
  • a typical antibody screen may combine phage antibodies from numerous donor specific libraries it is possible that some of the libraries and their combinatorial clones are not completely represented as antibody bearing phage particles.
  • a positive clone may have resulted from only a limited physical set of all the possible cloned solutions present in the sub-library phage population being screened. In such an instance it is of considerable interest to more fully interrogate the collection of donor specific phage.
  • the barcode from a positive clone guides one to the specific library responsible for the clone and allows to exclusively and more deeply screen the collection of interest.
  • donor profiles are logical guides for library selection and possible only in donor specific and segregated antibody libraries.
  • the libraries are described as heavy or light chain libraries, it will be apparent to those of ordinary skill in the art that the same description applies to the libraries of antibody fragments, fragments of antibody heavy and/or light chains, and libraries of antibody-like molecules.
  • antibodies with dual specificities such as, for example, showing reactivity with two different influenza A subtypes and/or with two strains (isolates) of the same subtype, and/or with human and non-human isolates, can be discovered and optimized through controlled cross-reactive selection and/or directed combinatorial and/or mutagenic engineering.
  • a library including antibodies showing cross-reactivity to two targets are subjected to multiple rounds of enrichment. If enrichment is based on reactivity with target A, each round of enrichment will increase the reactive strength of the pool towards target A. Similarly, if enrichment is based on reactivity with target B, each round of enrichment will increase the reactive strength of the pool towards target B.
  • FIG. 2 refers to panning, which is the selection method used when screening phage display libraries (see below), the approach is equally applicable to any type of library discussed above, or otherwise known in the art, and to any type of display technique.
  • Targets A and B include any targets to which antibodies bind, including but not limited to various isolates, types and sub-types of influenza viruses.
  • a cross-reactive discovery selection scheme can be used.
  • this scheme is illustrated in FIG. 3 showing the selection of antibodies with dual specificities.
  • an antibody library including antibodies showing reactivity with two targets, targets A and B is first selected for reactivity with one of the targets, e.g., target A, followed by selection for reactivity with the other target, e.g., target B.
  • Each successive selection round reinforces the reactive strength of the resulting pool towards both targets.
  • this method is particularly useful for identifying antibodies with dual specificity.
  • the method can be extended to identifying antibodies showing reactivity towards further targets, by including additional rounds of enrichment towards the additional target(s).
  • the library screened is a phage display library, selection is performed by cross-reactive panning, but other libraries and other selection methods can also be used.
  • a combination of the two methods discussed above includes two separate enrichment rounds for reactivity towards target A and target B, respectively, recombining the two pools obtained, and subsequent cross-reactive selection rounds, as described above. This approach is illustrated in FIG. 4 . Just as in the pure cross-reactive selection, each round of selection of the recombined library increases the reactive strength of the resulting pool towards both targets.
  • a clone showing strong reactivity with a target A, and having detectable cross-reactivity with target B is identified.
  • a mutagenic library is prepared, which is then selected, in alternating rounds, for reactivity with target B and target A respectively.
  • This scheme will result in antibodies that maintain strong reactivity with target A, and have increased reactivity with target B.
  • selection is performed by panning, if the libraries screened are phage display libraries, but other libraries, other display techniques, and other selection methods can also be used, following the same strategy.
  • targets A and B can, for example, be two different subtypes of the influenza A virus, two different strains (isolates) of the same influenza A virus, subtypes or isolates from two different species, where one species is preferably human.
  • target A may be an isolate of the 2004 Vietnam isolate of the H5N1 virus
  • target B may be a 1997 Hong Kong isolate of the H5N1 virus. It is emphasized that these examples are merely illustrative, and antibodies with dual and multiple specificities to any two or multiple targets can be identified, selected and optimized in an analogous manner.
  • antibody heavy and light chain libraries are cloned separately for each patient donor, where the individual libraries can be distinguished using unique barcodes.
  • the barcodes preferably are selected such that they are capable of propagating along with the clone(s) labeled, without interfering with the expression of the desired antibody chain or fragment thereof.
  • the barcode is inserted into the sequence of the expression vector, preferably, the 3′ untranslated region following the terminal pIII stop codon when a phagemaid vector is used.
  • a vector's unique sequence is determined and subsequently dedicated to a single defined library. This defined library can be derived not only from a single donor, but also from discrete pools of donors, or a synthetic repertoire or a semi-synthetic collection.
  • the barcode is inserted into the coding sequence of the antibody heavy and/or light chain or fragment thereof, at a position or in a form that does not interfere with the expression of the respective chains.
  • the barcodes can be non-coding DNA sequences of about 1-24 contiguous non-coding nucleotides in length that can be deconvoluted by sequencing or specific PCR primers. This way, a collection of nucleic acids, such as an antibody repertoire, can be linked at the cloning step.
  • the barcode is 3 or bases of randomly generated sequence.
  • the barcodes are coding sequences of silent mutations. If the libraries utilize restriction enzymes that recognize interrupted palidromes (e.g. Sfi GGCCNNNNNGGCC), distinct nucleotides can be incorporated in place of the “N's” to distinguish various collections of clones, such as antibody libraries. This barcoding approach has the advantage that the repertoire is linked at the amplification step.
  • the barcodes are non-contiguous nucleotide sequences, which may be present in the vector sequence and/or the coding sequence of the desired antibody chain.
  • a barcode with a non-contiguous sequence provides a great degree of flexibility in identifying the origins of the various individual sequences, and monitoring their subsequent handling.
  • the barcodes are coding sequences that encode immunologically distinct peptide or protein sequences fused to phage particles.
  • examples include, for example, epitope (e.g. Myc, HA, FLAG) fusions to pIII, pVIII, pVII, or pIX phages.
  • epitopes can be used singly or in various combinations, and can be provided in cis (on the library-encoding plasmid) or in trans (specifically modified helper phage) configuration.
  • barcodes include, without limitation, chemical and enzymatic phage modifications (for phage libraries) with haptens or fluorescent chromophores. Such tags are preferred for a single round of selection.
  • the individual heavy and light chain libraries obtained from individual donors, or other barcoded clone or collections, can be pooled, without losing the ability to distinguish the source of individual sequences.
  • cross-reactivity of the neutralizing antibodies with dual or multiple specificity can be further improved by methods known in the art, such as, for example, by Look Through Mutagenesis (LTM), as described in US. Patent Application Publication No. 20050136428, published Jun. 23, 2005, the entire disclosure of which is hereby expressly incorporated by reference.
  • LTM Look Through Mutagenesis
  • LTM Look-through mutagenesis
  • CDR complementarity determining region
  • combinatorial libraries (combinatorial beneficial mutations, CBMs) expressing all beneficial permutations can be produced by mixed DNA probes, positively selected, and analyzed to identify a panel of optimized scFv candidates. The procedure can be performed in a similar manner with Fv and other antibody libraries.
  • Mutagenesis can also be performed by walk-through mutagenesis (WTM), as described above.
  • WTM walk-through mutagenesis
  • Destinational mutagenesis can be used to rationally engineer a collection of antibodies based upon one or more antibody clones, preferably of differing reactivities.
  • destinational mutagenesis is used to encode single or multiple residues defined by analogous positions on like sequences such as those in the individual CDRs of antibodies. In this case, these collections are generated using oligo degeneracy to capture the range of residues found in the comparable positions.
  • the objective of destinational mutagenesis is to generate diverse multifunctional antibody collections, or libraries, between two or more discrete entities or collections.
  • To create a destinational mutagenesis library the CDR sequences for both antibodies are first attained and aligned. Next all positions of conserved identity are fixed with a single codon to the matched residue. At non-conserved positions a degenerate codon is incorporated to encode both residues. In some instances the degenerate codon will only encode the two parental residues at this position. However, in some instances additional co-products are produced. The level of co-product production can be dialed in to force co-product production or eliminate this production dependent upon size limits or goals.
  • the degenerate codon with A/G-C- in the first two positions would only encode threonine or alanine, irrespective of the base in the third position. If, for example, the next position residues are lysine and arginine the degenerate codon A-A/G-A/G will only encode lysine or arginine. However, if the degenerate codon A/C-A/G-A/G/C/T were used then asparagine, histidine, glutamine, and serine coproducts will be generated as well.
  • FIG. 6 is included herein to illustrate the use of the destinational mutagenesis method using CDRs of a TNF- ⁇ antibody and a CD11a antibody as the parental sequences mutagenized.
  • exemplary mutagenesis methods include saturation mutagenesis and error prone PCR.
  • Saturation mutagenesis (Hayashi et al., Biotechniques 17:310-315 (1994)) is a technique in which all 20 amino acids are substituted in a particular position in a protein and clones corresponding to each variant are assayed for a particular phenotype. (See, also U.S. Pat. Nos. 6,171,820; 6,358,709 and 6,361,974.)
  • Error prone PCR (Leung et al., Technique 1:11-15 (1989); Cadwell and Joyce, PCR Method Applic. 2:28-33 (1992)) is a modified polymerase chain reaction (PCR) technique introducing random point mutations into cloned genes.
  • PCR polymerase chain reaction
  • the resulting PCR products can be cloned to produce random mutant libraries or transcribed directly if a T7 promoter is incorporated within the appropriate PCR primer.
  • Optimization can be based on any of the libraries discussed above, or any other types of libraries known in the art, alone or in any combination.
  • antibodies with the desired neutralizing properties can be produced by methods well known in the art, including, for example, hybridoma techniques or recombinant DNA technology.
  • a mouse or other appropriate host animal such as a hamster
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor. J. Immunol. 133:3001 (1984); and Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • Recombinant monoclonal antibodies can, for example, be produced by isolating the DNA encoding the required antibody chains and co-transfecting a recombinant host cell with the coding sequences for co-expression, using well known recombinant expression vectors.
  • Recombinant host cells can be prokaryotic and eukaryotic cells, such as those described above.
  • human variable domains both light and heavy
  • the choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human framework region (FR) for the humanized antibody (Sims et al., J. Immunol. 151:2296 (1993); Chothia et al., J. Mol. Biol. 196:901 (1987)).
  • FR human framework region
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • human antibodies can be generated following methods known in the art.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • Jakobovits et al. Proc. Natl. Acad. Sci. USA 90:2551 (1993); Jakobovits et al. Nature 362:255-258 (1993); Bruggermann et al., Year in Immuno. 7:33 (1993); and U.S. Pat. Nos. 5,591,669, 5,589,369 and 5,545,807.
  • the neutralizing antibodies of the present invention can be used for the prevention and/or treatment of the targeted diseases.
  • the antibodies or other molecules, the delivery of which is facilitated by using the antibodies or antibody-based transport sequences, are usually used in the form of pharmaceutical compositions.
  • Antibodies are typically formulated in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride: hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben: catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol): low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyr
  • the antibodies also may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively), in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), or in macroemulsions.
  • coacervation techniques for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • the neutralizing antibodies disclosed herein may also be formulated as immunoliposomes.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang et al., Proc. Natl. Acad. Sci. USA 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and WO97/38731 published Oct. 23, 1997. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab′ fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al. J. Biol. Chem. 257:286-288 (1982) via a disulfide interchange reaction. A chemotherapeutic agent is optionally contained within the liposome. See Gabizon et al. J. National Cancer Inst. 81(19)1484 (1989).
  • the appropriate dosage of antibody will depend on the type of infection to be treated, the severity and course of the disease, and whether the antibody is administered for preventive or therapeutic purposes.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to about 15 mg/kg of antibody is a typical initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • Blood was obtained by standard venopuncture, allowed to clot, and processed to recover serum.
  • the serum was stored at ⁇ 20° C. for 3-4 days until they were shipped on dry ice.
  • Donors were anaesthetized with an injection of a local anesthetic and 5 ml of bone marrow was removed from the pelvic bone of each H5N1 survivor.
  • the 5 ml of bone marrow was placed into a sterile 50-ml tube containing 45 ml RNAlater (Ambion). The mixture was gently inverted approximately 8-20 times, until there were no visible clumps and the marrow and RNAlater were mixed well.
  • the specimen was refrigerated the between 2-10° C. overnight. Following the overnight refrigeration, the specimens were stored at ⁇ 20° C. for 3-4 days until they were shipped on dry ice. Upon receipt the RNAlater/marrow and sera containing tubes were stored at ⁇ 80° C. until processed.
  • ELISA plates (Thermo, Immulon 4HBX 96W) were coated with 100 ⁇ l of 100 ng/mL H5 hemagglutinin (Protein Sciences, A/Vietnam/1203/2004) in IX ELISA Plate Coating Solution (BiolFX) by overnight incubation at room temperature. The next day plates were washed three times with 300 ⁇ l PBS/0.05% Tween-20 (PBST). Following the wash, 300 ⁇ l of a blocking solution (4% Non-Fat dry Milk in PBS/0.05% Tween-20) was added and incubated for 1 hour at RTf. Following the blocking step, the plates were washed three times with 300 ⁇ l PBS/0.05% Tween-20.
  • Bone marrow ( ⁇ 2.5 ml in 20 ml RNA later), previously stored at ⁇ 80° C., was recovered by centrifugation to remove RNA later and then resuspended in 11.25 ml TRI BD reagent (Sigma) containing 300 ⁇ l Acetic Acid. The pellet was then vortexed vigorously. Next 1.5 ml BCP (1-bromo-3-chloropropane, Sigma) was added, mixed by vortexing, incubated at RT for 5 min, and then centrifuged at 12000 ⁇ g for 15 min at 4° C. The aqueous phase was carefully removed to not disturb the interface.
  • BCP 1-bromo-3-chloropropane
  • RNA from the aqueous phase was next precipitated by addition of 25 ml isopropanol, incubation at RT for 10 minutes, and centrifugation at 12000 ⁇ g for 10 min at 4° C.
  • isopropanol Following the addition of isopropanol, two phases were formed due to residual RNAlater, resulting in the precipitated RNA settling at the interface.
  • 5 ml aliquots of 50% isopropanol in H 2 0 were added and mixed until no phase separation was noticeable, at which point the RNA was pelleted by centrifugation at 12000 ⁇ g for 10 min at 4° C.
  • RNA pellet was washed with 75% EtOH, transferred to an RNAse-free 1.6 ml microcentrifuge tube, and again recovered by centrifugation. Finally the RNA pellet was resuspended in 100 ⁇ l 1 mM Na-phosphate, pH 8.2 and the A 260 and A 280 were read to assess RNA purity.
  • RNA Prior to reverse transcription mRNA was purified from total RNA according to Qiagen Oligotex mRNA purification kit. Briefly, 50-200 ⁇ g bone marrow RNA was brought to 250 ⁇ l with RNasc-free water and mixed with 250 ⁇ l of OBB buffer and Oligotex suspension followed by incubation for 3 min at 70° C. Hybridization between the oligo dT 30 of the Oligotex particle and the mRNA poly-A-tail was carried out at room temperature for min. The hybridized suspensions were then transferred to a spin column and centrifuged for 1 min. The spin column was washed twice with 400 ⁇ l Buffer OW2. Purified mRNA was then eluted twice by centrifugation with 20 ⁇ l hot (70° C.) Buffer OEB. Typical yields were 500 ng to 1.5 ⁇ g total RNA.
  • Reverse transcription (RT) reactions were accomplished by mixing together 75-100 ng mRNA with 2 ⁇ l 10 ⁇ Accuscript RT Buffer (Stratagene), 0.8 ⁇ l 100 mM dNTPs, and either N9 (300 ng) or oligo dT primer (100 ng) and then brought to a final volume of 17 ⁇ l with water. The mixtures were heated at 65° C. for 5 min, and then allowed to cool to room temperature. Next 2 ⁇ l DTT, 0.5 ⁇ l RNase Block (Stratagene), 0.5 ⁇ l AccuScript RT (Stratagene) were added to each reaction.
  • N9 primed reactions were incubated for 10 minutes at room temperature and the oligo-dT primed reactions were incubated on ice for 10 minutes. Finally, both reactions were incubated at 42° C. for 60 minutes followed by 70° C. for 15 minutes to kill the enzyme.
  • Antibody heavy and light chain repertoires were amplified from bone marrow cDNA essentially using previously described methods and degenerate primers (O'Brien, P. M., Aitken R. Standard protocols for the construction of scFv Libraries. Antibody Phage Display Methods and Protocols , vol. 178, 59-71, 2001, Humana Press) based upon human germline V and J regions.
  • PCR reactions using Oligo dT primed cDNA (from 75 ng mRNA) for lambda light chains and N9 primed cDNA (from 75 ng mRNA for kappa light chains, from 100 ng mRNA for heavy chains) were mixed together with 5 ⁇ l 10 ⁇ amplification buffer (Invitrogen), 1.5 ⁇ l dNTPs (10 mM), 1 ⁇ l MgSO4 (50 mM), 2.5 ⁇ l V region primers (10 uM) and 2.5 ⁇ l J region primers (10 uM)-10 uM for V H , 0.5 ⁇ l Platinum Pfx Polymerase (Invitrogen), and sterile dH 2 O to final volume of 50 ⁇ l.
  • PCR parameters were as follows: step 1-95° C. 5 minutes, step 2-95° C. 30 seconds, step 3-58° C. 30 seconds, step 4-68° C. 1 minute, step 5-cycle step 2-4 40 times, step 6-68° C. 5 minutes.
  • Light chain PCR products were cleaned up using Qiagen PCR Cleanup kit.
  • Heavy chains PCR products were gel purified from 1.5% agarose gel using Qiagen Gel Extraction Kit and then reamplified.
  • Heavy chain reamplification was carried out as follows: Mixed 10 ⁇ l 10 ⁇ amplification buffer (Invitrogen), 3 ⁇ l dNTPs (10 mM), 2 ⁇ l MgSO4 (50 mM), 5 ⁇ l each V H primers (10 uM) and J H primers (10 uM), 5 ⁇ l Heavy chain Primary PCR product, 1 ⁇ l Platinum Pfx, volume adjusted to 100 ⁇ l with water. Cycling parameters were as follows: step 1-95° C. 5 minutes, step 2-95° C. 30 seconds, step 3-58° C. 30 seconds, step 4-68° C. 1 minute, step 5-cycle step 2-4 20 times, step 6-68° C. 5 minutes. Re-amplified heavy chain PCR products were cleaned up from a 1.5% agarose-TAE gel using Qiagen Extraction Kit.
  • the library was transformed in five electroporations in 80 ⁇ l TG-1 or XL-1 Blue aliquots, each recovered in 1 ml SOC, pooled and outgrown for one hour at 37° C. Total number of transformants was determined following this outgrowth by plating an aliquot from each of the transformations. The remaining electroporation was amplified by growing overnight at 37° C. in 200 ml 2YT+50 ⁇ g/ml Ampicillin+2% glucose. The subsequent light chain library was recovered by plasmid purification from these overnight cultures using a Qiagen High Speed Maxiprep Kit.
  • each of the donor-specific heavy chains (V H 1, V H 2, 5, 6 pool. V H 3, and V H 4) were digested with a 40 Unit excess/ ⁇ g DNA with Sfil and Xhol and gel purified from a 1.5% agarose-TAE gel using Qiagen Gel Extraction Kit.
  • 15 ⁇ g of each light chain library vector was digested with 40 Unit/ ⁇ g DNA with Sfil and Xhol and gel purified from a 1% agarose-TAE gel using Qiagen Gel Extraction Kit.
  • Library ligations were set up by combining 1.2 ⁇ g Sfil/Xhol digested, gel purified heavy chain donor collections and 5 ⁇ g of each light chain library (kappa and lambda) overnight at 14° C.
  • the library ligations were then desalted with Edge BioSystem Pefroma spin columns and then transformed through 20 electroporations per library in 80 ⁇ l TG-1 aliquots, each recovered in 1 ml SOC, pooled and outgrown for one hour at 37° C. Again following this outgrowth an aliquot of each was used to determine the total number of transformants with the remainder transferred to 1L 2YT+50 ⁇ g/ml Ampicillin +2% glucose and grown at 37 C with vigorous aeration to an OD 600 of ⁇ 0.3. Next M13K07 helper phage was then added at a multiplicity of infection (MOI) of 5:1 and incubated for 1 hour at 37° C., with no agitation.
  • MOI multiplicity of infection
  • the cells were harvested by centrifugation and resuspended in 1L 2YT+50 ⁇ g/ml Ampicillin, 70 ⁇ g/ml Kanamycin and grown overnight at 37° C. with vigorous aeration to allow for scFv phagemid production.
  • the next morning the cells were collected by centrifugation and supernatant containing phagemid was collected.
  • the phagemids were precipitated from the supernatant by the addition of 0.2 volumes 20% PEG/5 M NaCl solution and incubation for 1 hour on ice.
  • the phagemid library stocks were then harvested by centrifugation and resuspended in 20 ml sterile PBS. Residual bacteria were removed by an additional centrifugation and the final phagemid libraries were stored at ⁇ 20° C. in PBS+50% glycerol.
  • ELISA plates (Immulon 4HBX flat bottom, Nunc) were coated with 100 ⁇ l of 100 ng/ml, H5 hemagglutinin protein(Protein Sciences, A/Vietnam/1203/2004) in ELISA Coating Solution (BioFX) by overnight incubation at room temperature. The next day plates were washed three times with 300 ⁇ l PBST. Following the wash, 300 ⁇ l of a blocking solution (4% Non-Fat dry Milk in PBS/0.05% Tween-20) was added and incubated for 30 mins on ice. Following the blocking step, the plates were washed three times with 300 ⁇ l PBST.
  • a blocking solution 4% Non-Fat dry Milk in PBS/0.05% Tween-20
  • glycerol was removed from the frozen phagemid stocks using Millipore Amicon Ultra columns and then blocked in 4% nonfat dry milk for 15 minutes.
  • 100 ⁇ l aliquots of phagemid were distributed into 8 wells (total phage ⁇ 1 ⁇ 10 12 CFU) and incubated for 2 hours at 4° C. followed by washing 6-8 times with 300 ⁇ l PBST.
  • Phagemid were collected following a 10 min at room temperature in 100 ⁇ l/well Elution buffer (0.2M glycine-HCl, pH 2.2, 1 mg/ml BSA). The eluate was then neutralized by the addition of 56.25 ⁇ l 2M Tris base per ml eluate. Following neutralization.
  • TGI cells 5 ml TGI cells (OD 6oo ⁇ 0.3) were infected with 0.5 ml neutralized phage at 37° C. for 30 minutes in 2-YT with no shaking. Following this step some cells were plated onto LB AMP Glucose plates to determine total phagemid recovery. The remaining inoculum was placed into 10 ml 2-YTAG (final concentration 2% glucose and 50 ⁇ g/ml ampicillin) and grown at 37° C. with vigorous aeration to OD 600 —0.3. Next the cultures were infected with M13K07 helper phage at an MOI of 5:1 and incubated at 37° C. for 30-60 minutes with no shaking.
  • the cells were collected by centrifugation and resuspended in 25 ml 2-YTAK (Ampicillin 50 ⁇ g/ml, Kanamycin 70 ⁇ g/ml), transferred to a fresh culture flask, and grown ON at 37° C. with shaking. Subsequent rounds were similarly recovered and amplified.
  • 2-YTAK Amicillin 50 ⁇ g/ml, Kanamycin 70 ⁇ g/ml
  • E. coli HB2151 transformed cells from biopanned phage were grown overnight at 37° C. in 1 ml of 2YT+100 ⁇ g/ml AMP. The following morning the cells were harvested by centrifugation and resuspended in 1.5 ml periplasmic lysis buffer (1 ml BBS (Teknova)+0.5 ml 10 mg/ml lysozyme+EDTA to 10 mM final concentration). The cells were again pelleted by centrifugation and the scFv containing periplasmic lysates were collected.
  • the scFv lysates were combined 1:1 with dilution buffer (PBS/0.05% BSA) and 100 ⁇ l was added to wells that had been previously antigen coated with and blocked with dilution buffer. The samples were incubated for 2 hours at room temperature and then washed three times with PBS/0.05% Tween. Next 100 ⁇ l of 1:5000 diluted Biotin Anti-Histidine mouse (Serotec) in dilution buffer was added to each well and incubated for 1 hr at room temperature.
  • HAI Hemagglutinin Inhibition
  • Hemagglutination Inhibition was performed essentially following the method of Rogers et al. Virology 131:394-408 (1983), in round bottom microliter plates (Corning) using 4 HAU (hemagglutinating units) of virus or protein/well.
  • HAU hemagglutinating units
  • 25 ⁇ l samples of purified single chain variable fragments (scFv) were mixed with 25 ⁇ l of PBS containing 4 HAU of the test virus in each microtiter well. Following a preincubation of 15 minutes at room temperature, 25 ⁇ l of 0.75% human erythrocytes were added, and mixed.
  • HAI antibody activity was determined by visual inspection following a 60 min incubation at room temperature.
  • IgG 1 proteins were generated from either scFv or Fabs by subcloning their coding regions into a pCI-based (Promega) full length mammalian protein expression and then transfecting them into 293 Freestyle cells (Invitrogen) according to manufacturers guidelines. Briefly 20 ⁇ g of light chain and 10 ⁇ g heavy chain encoding plasmid were combined with 1.0 ml 293fectin and incubated for 60 minutes. Following this preincubation the DNA mixture was combined with 3 ⁇ 10 7 cells in 30 ml media and the resulting cell suspension was grown according to manufacturers suggestion for 7 days. After seven days the secreted immunoglobulins were purified from the culture supernatants using protein A chromatography (Calbiochem). The resulting purified antibodies were buffer exchanged into sterile PBS using centrifugal size filtration (Centricon Plus-20) and their protein concentrations determined by colorimetric BCA assay (Pierce).
  • Microtiter plates were coated with 0.1 ml of the following antigens diluted in coating buffer and incubated overnight at room temperature: 100 ng/ml H5N1 Vietnam 1203/04, 250 ng/ml H5N1 Turkey/65596/06, 1 ⁇ g/ml H5N1 Indonesia/5/05, 700 ng/ml H1N1 New Caledonia/20/99, 1 ⁇ g/ml H1N1 North Carolina/1/18, 100 ng/ml and H3N2 Wisconsin/67/05. Blocking was done with 0.3 ml of blocking buffer (4% Non-fat dry milk in PBS/0.05% Tween-20).
  • Each transfection contained ambisense plasmids (for the expression of both vRNAs and mRNAs) for the A/Puerto Rico/8/34/PA, PB1, PB2, NP, M, and NS segments, in addition to vRNA (pPOL1 type) and protein expression plasmids (pCAGGS type) for A/Vietnam/1203/04 HA and NA (pCAGGS expression plasmid was kindly provided by J. Miyazaki, Osaka University, Osaka, Japan) (Miyazaki, J. et al. Gene 1989; 79(2):269-77). Twenty hours following transfection, 293T cells were resuspended in cell culture supernatant, and used to inoculate 10-day-old embryonated eggs.
  • Antibodies were screened for neutralizing activity against viruses as follows. Two fold serial dilutions of each Mab were incubated with 100 TCIDs 50 of virus in PBS at 37° for 1 h. Madin-Darby Canine Kidney cell monolayers in 24 well plates were washed once with PBS and inoculated with virus-antibody mixtures. Following incubation for 1 h at 37° C. in 5% CO 2 the inoculum was removed and monolayers were again washed once with PBS. Opti-MEM supplemented with 0.3% BSA, 0.01% FBS and 1 ug/ml TPCK-treated trypsin was added and cells were incubated for 72 h at 37° C. The presence of virus in cell culture supernatants was assessed by HA assays using 0.5% chicken red blood cells.
  • Bone marrow and blood samples were collected from six survivors of the H5N1 bird flu outbreak that had taken place in Turkey in January 2006, approximately four months after the outbreak. For all six survivors the initial diagnosis of bird flu was made following by physical examination, clinical laboratory testing, and molecular diagnostic determination, sanctioned by the Turkish Ministry of Health. Four of these survivors were additionally confirmed by the World Health Organization (WHO). Serum samples were analyzed to confirm the presence of antibodies to H5 hemagglutinin (A/Vietnam/1203/2004) using the serology protocol described above. As shown in FIG. 8 , the blood samples of all six patients (designated SLB H1-H6, respectively) demonstrated the presence of antibodies to the H5 antigen.
  • the antibody heavy and light chain repertoires were then amplified from the bone marrow cDNA as described above, and individual antibody heavy and light chain phage libraries were cloned separately for each survivor, using the above-described three-nucleotide bar coding to distinguish the individual libraries.
  • Table 2 shows the light chain and full library total transformants in both scFv and Fab formats. Total diversity represented by all libraries is 5.6 ⁇ 10 9
  • Bone marrow and blood samples were also collected from twelve local donors who were treated for flu symptoms in the year of 2006. Serology was performed as described above to confirm the presence of antibodies to H1, H3 and H5 hemagglutinin, respectively.
  • Serology was performed as described above to confirm the presence of antibodies to H1, H3 and H5 hemagglutinin, respectively.
  • FIG. 8 all serum samples tested positive for antibodies to H1 and/or H3 hemagglutinins, where the dominance of a certain subtype depended on the influenza A virus subtype to which the particular donor was exposed most throughout his or her lifetime.
  • donors SLB 1 and SLB 5 in FIG. 9 there were donors whose serum contained a significant level of antibodies of H5 hemagglutinin as well (donors SLB 1 and SLB 5 in FIG. 9 ).
  • the antibody heavy and light chain repertoires were then amplified from the bone marrow cDNA as described above, and individual antibody heavy and light chain phage libraries were cloned separately for each donor, using the above-described three-nucleotide bar coding to distinguish the individual libraries.
  • H5N1 survivor libraries summarized in Table 2, were panned against inactivated virus containing the Vietnam/1203/04 virus HA and NA proteins or recombinant purified hemagglutinin (Barbas, C. et al. (2001) Phage Display, A Laboratory Manual (Cold Spring Harbor Laboratory Press)). Following three to four rounds of phage panning, individual clones from enriched phage pools were analyzed by ELISA against H5N1 virus or purified hemagglutinin and the positive clones were sequenced to determine their heavy and light chain sequences and to read their survivor bar code (D. W. Coomber, Methods Mol Biol 178, 133 (2002)).
  • H5N1 antibodies from two survivors with hemagglutinins from H1N1 viruses.
  • Bars are H5N1 Vietnam 1203/04(dark grey), H5N1 Turkey/65596/06 (white), H5N1 Indonesia/5/05 (diagonal stripes), H1N1 New Caledonia/20/99 (vertical stripes), H1N1 South Carolina/1/18 (crosshatch stripes), and H3N2 Wisconsin/67/05 (light gray).
  • the antibodies were assayed for their ability to neutralize an H5 HA (Vietnam/1203/04) containing influenza virus.
  • One antibody derived from survivor 2 and 3 from survivor 5 that recognized a common epitope (epitope “A”) were all neutralizing whereas the two antibodies derived from survivor 1 that recognized a second epitope (epitope “13”) were not.
  • a collection of viruses bearing H5 subtype hemagglutinin was also tested (A/Vn/1203/04; A/Indo/5/05; A/Turkey/65596/06; A/Egypt/06).
  • the antibodies showed no activity against H3 subtype influenza.
  • three of the monoclonal antibodies (1-3) that neutralized H5 containing viruses also strongly neutralized all viruses bearing HA from subtypes H1 (Table 5).
  • antibody libraries when one obtains large numbers of antibodies, they can be grouped as to their relatedness. Thus, when a function for a given antibody in the collection is observed one can predict that other members of the group to which it belongs will have similar activity.
  • Table 4 shows example sequences displaying the immunochemical basis of neutralization discovered from Survivor 5 libraries following H5N1 Vietnam panning.
  • Antibody regions and Kabat numbering ranges are listed at the top of each sequence column.
  • the heavy chain/light chain pairing is indicated in the first column as follows: *—paired with 2 unique light chains, ⁇ —paired with 3 unique light chains, ⁇ —paired with 4 unique light chains, ⁇ —paired with 5 unique light chains, and ⁇ —paired with 12 unique light chains.
  • the group is comprised of 61 unique members that most closely resemble the V H 1e germ line heavy chain. Some heavy chains are paired with more than one light chain. In total these heavy chains have 114 unique pairings to both kappa and lambda light chains. Comparing these heavy chains to the highly related VH1e germline, we observe three types of point substitutions. Some changes appear to be required, others are dominant, and some residues have only been changed sporadically.
  • FIG. 12 shows the positions of the required mutations in the structure of the antibody superimposed on the crystal structure of a highly related anti-HIV Fab called 47e (lrzi.pdb) (Huang, C. C. et al. (2004) Proc. Nat. Acad. Sci. 101, 2706-2711).
  • FIG. 12 shows the positions of H5 hemagglutinin binding Group 1 required and dominant mutations on the crystal structure of Fab 47e.
  • the required mutations are shown as G52 (52A (Pro>Gly)), M53 (Ile>Met), T57 (Ala>Thr), E73 (Lys>Glu) and LM74 (Ser>Leu or Met).
  • the dominant mutations are shown as T24 (Ala>Thr), V34 (Ile>Val), T35 (Ser>Thr), and A50 (Gly>Ala).
  • the required and dominant Group 1 heavy chain sequences identified in H5 Vietnam/1203/2004H A biopanning are superimposed on the crystal structure of the highly related anti-HIV Fab 47e. Mutations are shown in both backbone (top) and space-filling (bottom) models. A tight cluster is formed by four of the required mutations in and adjacent to CDR2.
  • the required mutations 52A Pro>Gly
  • 53 Ile>Met
  • 73 Lys>Glu
  • 74 Ser>Leu or Met
  • the remaining required mutation 57 is partially buried at the base of the CDR2 loop.
  • the surface exposed changes in CDR 2 and framework 3 are likely to have a direct role in antigen binding while the less exposed required mutation and the non-essential dominant mutations may have indirect effects through stabilizing and/or positioning the CDR2 loop.
  • the antibodies from survivor 2 are comprised of 2 unique heavy chains that most closely resemble the V H 4-4b germ line heavy chain (Table 5).
  • the first heavy chain has been found paired with 5 unique lambda light chains, four of which are from the infrequently used lambda 6 light chain family and the other is paired with a single kappa light chain.
  • Antibody 4 whose neutralization profile was more restricted came from this group.
  • codon usage of individual clones shows independent origin of selected H5 HA binding clones.
  • Table 6A corresponds to CDR2 and Table 6B corresponds to Framework 3.
  • Germ line codons are shown as bolded codons.
  • a change from a germ line codon to the same amino acid is shown as a plain text codon.
  • a first change from a germ line amino acid is shown as a bolded, underlined codon.
  • a second change from a germ line amino acid is shown as an italicized, underlined codon.
  • a third change from a germ line amino acid is shown as an underlined,
  • Blood is obtained by standard venopuncture, allowed to clot, and processed to recover serum.
  • the serum is stored at ⁇ 20° C. for 3-4 days until they are shipped on dry ice.
  • Donors are anaesthetized with an injection of a local anesthetic and 5 ml of bone marrow is removed from the pelvic bone of each patient donor.
  • the 5 ml of bone marrow is placed into a sterile 50-ml tube containing 45 ml RNAlater (Ambion). The mixture is gently inverted approximately 8-20 times, until there are no visible clumps and the marrow and RNAlater are mixed well.
  • the specimen is refrigerated the between 2-10° C. overnight.
  • RNAlater/marrow and sera containing tubes are stored at ⁇ 80° C. until processed.
  • Candidate patient should be tested HIV positive prior to be selected as donors.

Abstract

The present application concerns donor-specific antibody libraries derived from a patient donor who has suffered from, or is suffering from one or more diseases discussed herein. The present application also concerns the method of making and using the donor-specific antibodies. The present application further concerns the neutralizing antibodies obtained from the donor-specific antibody libraries and the methods of using these antibodies for the prevention/treatment of human disease.

Description

    FIELD OF THE INVENTION
  • The present invention concerns donor-specific antibody libraries and methods of making and using thereof. The present invention also concerns neutralizing antibodies obtained from such donor-specific antibody libraries and methods of using the antibodies obtained for the prevention and/or treatment of various human diseases and conditions.
  • BACKGROUND OF THE INVENTION
  • The generation and identification of specific agents for the diagnosis, prevention, and treatment of human diseases requires access to vast collections of useful chemistries. With the advent and rapid development of a variety of techniques for the creation and screening of antibody libraries, monoclonal antibodies against disease targets have become one of the major categories of new drug candidates. Since for human use, in addition to specificity and efficacy, safety is of primary concern, libraries of human monoclonal antibodies have become of particular importance.
  • At present, the development of human antibody-based drug candidates are typically identified by screening of human antibody libraries comprising a random collection of antibody sequences from human repertoires that are typically unrelated to their intended application or applications. Each antibody library created from a specific human donor potentially contains antibodies to every component, physiology, and metabolic alteration stemming from, or creating, every unique challenge that the donor has encountered, challenged, and surmounted over the course of that individual's lifetime. As typical human antibody libraries made with the current approaches are constructed without the knowledge of the health history of donors, little is known of what would be expected in the resulting immunoglobulin repertoires.
  • Thus, it is of great interest to create antibody libraries from individuals who have successfully survived or are surviving an encounter with specific diseases because their resulting repertoires include antibodies that were used by the donor to defend specifically against a relevant disease. It is also important to provide methods for the efficient screening and handling of such libraries, including the ability to remove or isolate negative or positive elements, eliminate undesirable content, and produce human antibodies with improved properties.
  • The present invention addresses these needs by providing methods and means for the creation, screening and handling of donor-specific antibody libraries from individuals who have been exposed to and survived or are surviving an encounter with a specific target disease.
  • SUMMARY OF THE INVENTION
  • In one aspect, the invention concerns a vector collection comprising a repertoire of nucleic acid molecules encoding antibody light or heavy chains or fragments thereof, derived from a human patient donor who has suffered from, or is suffering from, a disease evoking antibody production to a target antigen, wherein the collection is identified with a unique barcode.
  • In one embodiment, the vector collection comprises a repertoire of nucleic acid molecules encoding antibody light chains or fragments thereof, such as antibody λ light chains, or antibody κ light chains, or fragments thereof.
  • In another embodiment, the vector collection comprises a repertoire of nucleic acid molecules encoding antibody heavy chains or fragments thereof.
  • In yet another embodiment, the barcode is a nucleotide sequence linked to or incorporated in the vectors present in the collection, and/or linked to or incorporated in the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of the nucleic acid molecules.
  • Thus, the barcode may be contiguous non-coding nucleotide sequence of one to about 24 nucleotides, which may, for example, be linked to the 3′ or 5′ non-coding region of the nucleic acid molecules.
  • In a further embodiment, the barcode is a nucleotide sequence that is a coding sequence of one or more silent mutations incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof.
  • In a still further embodiment, the barcode is a non-contiguous nucleotide sequence. At least part of the non-contiguous nucleotide sequence may be linked to or incorporated in the vectors present in the collection. Alternatively, at least part of the non-contiguous nucleotide sequence may be incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of said nucleic acid molecules.
  • In another embodiment, the barcode is a peptide or polypeptide sequence.
  • In a different embodiment, the vectors present in the vector collection are phagemid vectors, which may, for example, contain a bacteriophage gene III and a stop codon between the nucleic acid molecules encoding antibody light or heavy chains or fragments thereof and the bacteriophage III gene, and may have a barcode, such as a non-coding contiguous nucleotide sequence inserted in the untranslated region following the stop codon.
  • In another aspect, the invention concerns host cells comprising the vector collection of the present invention. The host cells may by eukaryotic or prokaryotic host cells, such as, for example, E. coli cells.
  • In a further aspect, the invention concerns a donor-specific antibody library comprising library members expressing a collection of antibodies or antibody fragments to a target antigen wherein the antibodies or antibody fragments are derived from a human donor who has suffered from, or is suffering from, a disease evoking antibody production to said target antigen, wherein said antibody library is identified with at least one unique barcode.
  • In one embodiment, the antibody heavy and light chains are separately identified each with a barcode unique to the human donor from whom it derived.
  • In another embodiment, the donor-specific antibody library is identified with one unique barcode.
  • In yet another embodiment, the antibodies or antibody fragments are composed of antibody heavy and light chains or fragments thereof encoded by nucleic acid molecules present in a vector.
  • In a further embodiment, the barcode is a nucleotide sequence linked to or incorporated in the vectors present in the library, and/or linked to or incorporated in the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of the nucleic acid molecules.
  • In a still further embodiment, the barcode is a contiguous non-coding nucleotide sequence of one to about 24 nucleotides, which may, for example, be linked to the 3′ or 5′ non-coding region of the nucleic acid molecules.
  • In a different embodiment, the barcode is encoded by a coding sequence of one or more silent mutations incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof.
  • In another embodiment, the barcode is encoded by a non-contiguous nucleotide sequence.
  • In a further embodiment, a least part of the non-contiguous sequence encoding the barcode is linked to or incorporated in the vectors present in the library.
  • In a still further embodiment, at least part of the non-contiguous sequence encoding the barcode is incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of such nucleic acid molecules.
  • In different embodiment, the barcode is a peptide or polypeptide sequence.
  • In another embodiment, vectors are phagemid vectors, may, for example, contain a bacteriophage gene III and a stop codon between the nucleic acid molecules encoding antibody light or heavy chains or fragments thereof and the bacteriophage II gene.
  • In yet another embodiment, the medical history of the human patient donor shows that the donor has suffered from, or is suffering from said disease. In some embodiments, it is independently confirmed that the human donor suffered from, or is suffering from the disease.
  • In an additional embodiment, the donor-specific antibody library is substantially devoid of antibodies and antibody fragments specifically binding antigens different from said target antigen.
  • In one embodiment, the target antigen is an influenza A virus, such as an isolate of influenza A virus H1, H2, H3, H5, H7, or H9 subtype.
  • In another embodiment, the library expresses at least one antibody or antibody fragment specifically binding to more than one influenza A virus subtype.
  • In yet another embodiment, the library expresses at least one antibody or antibody fragment binding to and neutralizing the H5N1 subtype of influenza virus A.
  • In a further embodiment, the human donor has suffered from, or is suffering from a disease selected from the group consisting of the diseases listed in Table 1 below.
  • In a still further embodiment, the antibody library expresses at least one antibody or antibody fragment binding to an antigen associated with the target disease.
  • In an additional embodiment, the antibody library expresses at least one antibody or antibody fragment binding to and neutralizing an antigen associated with the target disease.
  • The donor-specific antibody library may, for example, be a phage library, in an embodiment, it may contain sequences encoding more than 106 different members of antibodies or antibody fragments, or more than 109 different members of antibodies or antibody fragments.
  • In other embodiments, the donor-specific antibody library is, without limitation, a spore-display library, a ribosome display library, a mRNA display library, a microbial cell display library, a yeast display library, or a mammalian display library.
  • In an embodiment, the nucleic acid encoding the antibodies or antibody fragments present in the library is reverse-transcribed from mRNA extracted from lymphocytes of the human patient donor, where the lymphocytes may, for example, originate from bone marrow, blood, spleen, or lymph node.
  • If desired, a serological profile of said human donor may be generated prior to extraction of said mRNA.
  • Alternatively, or in addition, the medical history of the human donor, and optionally the donor's family, is examined prior to or following extraction of the mRNA.
  • In another aspect, the invention concerns a method of making a donor-specific library expressing a collection of antibodies or antibody fragments to a target antigen, comprising the steps of:
  • a) obtaining mRNA from lymphocytes of a human patient donor who has suffered from, or who is suffering from a disease evoking antibody production to said target antigen;
  • b) generating a collection of nucleic acids comprising sequences encoding an immunoglobulin repertoire of the patient by reverse transcription of said obtained mRNA; and
  • c) identifying the donor-specific library with an unique barcode labeling said nucleic acids.
  • The method may further comprise steps of generating a serological profile of said patient and/or examining medical history of the patient prior or subsequent to step a).
  • The method may further comprise the steps of d) inserting said nucleic acids into expression vector: e) expressing the immunoglobulin repertoire; and f) displaying the immunoglobulin repertoire in a display system.
  • In another embodiment, the method further comprises the step of selecting members of the library based their ability to neutralize or activate the target antigen.
  • In a still further embodiment, the method yields at least one neutralizing antibody.
  • In another embodiment, the method further comprises the step of creating one or more sub-libraries comprising library members that were found to neutralize or activate the target antigen.
  • In yet another embodiment, the method comprises the step of sequencing at least one library member identified.
  • In a further aspect, the invention concerns a method of treating or preventing a disease associated with a target antigen neutralized or activated by an antibody selected by the method described above, comprising administering to a human patient in need an effective amount of the antibody selected.
  • The antibody may, for example, be a neutralizing antibody, such as a neutralizing antibody to at least one influenza A virus subtype.
  • In another embodiment, the disease is selected from the group consisting of the diseases listed in Table 1 below.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a flow chart schematically illustrating a typical method for the creatin of the human antibody libraries of the present invention.
  • FIG. 2 illustrates a typical panning enrichment scheme for increasing the reactive strength towards two different targets, A and B. Each round of enrichment increases the reactive strength of the pool towards the individual target(s).
  • FIG. 3 illustrates a strategy for the selection of clones cross-reactive with targets A and B, in which each successive round reinforces the reactive strength of the resulting pool towards both targets.
  • FIG. 4 illustrates a strategy for increasing the reactive strengths towards two different targets (targets A and B), by recombining parallel discovery pools to generate/increase cross-reactivity. Each round of selection of the recombined antibody library increases the reactive strength of the resulting pool towards both targets.
  • FIG. 5 illustrates a strategy for increasing cross-reactivity to a target B while maintaining reactivity to a target A. First, a clone reactive with target A is selected, then a mutagenic library of the clones reactive with target A is prepared, and selection is performed as shown, yielding one or more antibody clones that show strong reactivity with both target A and target B.
  • FIG. 6 illustrates a representative mutagenesis method for generating a diverse multifunctional antibody collection by the “destinational mutagenesis” method.
  • FIG. 7 shows the amino acid sequences of 15 known hemagglutinin (H) protein subtypes.
  • FIG. 8 shows the H5 hemagglutinin (HA) serology results for blood samples obtained from six human survivors of a Turkish H5N1 bird flu outbreak. The data demonstrate the presence of antibodies to the HA antigen.
  • FIG. 9 shows serology results obtained with serum samples of twelve local donors, tested on H5 antigen (A/Vietnam/1203/2004) and H1N1 (A/New Caledonia/20/99) and H3N2 (A/Panama/2007/99) viruses.
  • FIG. 10 illustrates the unique barcoding approach used in the construction of antibody phage libraries.
  • FIG. 11 shows the analysis of antibody binding to hemagglutinins from different influenza A subtypes.
  • FIG. 12 shows the positions of H5 hemagglutinin binding Group 1 required and dominant mutations on the crystal structure at Fab 47e.
  • FIGS. 13 and 14 illustrate the use of destinational mutagenesis to create diverse antibody heavy and light chain libraries using the antibody heavy and light chain sequences identified by analysis of sera and bone marrow of Turkish bird flu survivors.
  • DETAILED DESCRIPTION A. Definitions
  • Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, N.Y. 1994), provides one skilled in the art with a general guide to many of the terms used in the present application.
  • One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below.
  • The phrase “conserved amino acid residues” is used to refer to amino acid residues that are identical between two or more amino acid sequences aligned with each other.
  • The terms “disease,” “disorder” and “condition” are used interchangeably herein, and refer to any disruption of normal body function, or the appearance of any type of pathology. The etiological agent causing the disruption of normal physiology may or may not be known. Furthermore, although two patients may be diagnosed with the same disorder, the particular symptoms displayed by those individuals may or may not be identical.
  • An “effective amount” is an amount sufficient to effect beneficial or desired therapeutic (including preventative) results. An effective amount can be administered in one or more administrations.
  • A “composition,” as used herein, is defined as comprising an active ingredient, such as a neutralizing antibody generated from the present invention, and at least one additive, such as a pharmaceutically acceptable carrier, including, without limitation, water, minerals, proteins, and/or other excipients known to one skilled in the art.
  • As used herein, the term “treating” or “treatment” is intended to mean an amelioration of a clinical symptom indicative of a disease.
  • As used herein, the term “preventing” or “prevention” is intended to mean a forestalling of a clinical symptom indicative of a disease
  • The terms “subject” and “patient,” as used herein, are used interchangeably, and can refer to any to animal, and preferably a mammal, that is the subject of an examination, treatment, analysis, test or diagnosis. Thus, subjects or patients include humans, non-human primates and other mammals, who may or may not have a disease or other pathological condition.
  • The term “amino acid” or “amino acid residue” typically refers to an amino acid having its art recognized definition such as an amino acid selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gln); glutamic acid (Glu); glycine (Gly); histidine (His); isoleucine (Ile): leucine (Leu): lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val) although modified, synthetic, or rare amino acids may be used as desired. Thus, modified and unusual amino acids listed in 37 CFR 1.822(b)(4) are included within this definition and expressly incorporated herein by reference. Amino acids can be subdivided into various sub-groups. Thus, amino acids can be grouped as having a nonpolar side chain (e.g., Ala, Cys, Ile, Leu, Met, Phe, Pro, Val); a negatively charged side chain (e.g., Asp, Glu); a positively charged side chain (e.g., Arg, His, Lys); or an uncharged polar side chain (e.g., Asn, Cys, Gln, Gly, His, Met, Phe, Ser, Thr, Trp, and Tyr). Amino acids can also be grouped as small amino acids (Gly, Ala), nucleophilic amino acids (Ser, His, Thr, Cys), hydrophobic amino acids (Val, Leu. Ile. Met, Pro), aromatic amino acids (Phe, Tyr, Trp, Asp, Glu), amides (Asp, Glu), and basic amino acids (Lys, Arg).
  • The term “variant” with respect to a reference polypeptide refers to a polypeptide that possesses at least one amino acid mutation or modification (i.e., alteration) as compared to a native polypeptide. Variants generated by “amino acid modifications” can be produced, for example, by substituting, deleting, inserting and/or chemically modifying at least one amino acid in the native amino acid sequence.
  • An “amino acid modification” refers to a change in the amino acid sequence of a predetermined amino acid sequence. Exemplary modifications include an amino acid substitution, insertion and/or deletion.
  • An “amino acid modification at” a specified position, refers to the substitution or deletion of the specified residue, or the insertion of at least one amino acid residue adjacent to the specified residue. By insertion “adjacent” to a specified residue is meant insertion within one to two residues thereof. The insertion may be N-terminal or C-terminal to the specified residue.
  • An “amino acid substitution” refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence with another different “replacement” amino acid residue. The replacement residue or residues may be “naturally occurring amino acid residues” (i.e. encoded by the genetic code) and selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gln); glutamic acid (Glu); glycine (Gly); histidine (His): isoleucine (Ile): leucine (Leu); lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val). Substitution with one or more non-naturally occurring amino acid residues is also encompassed by the definition of an amino acid substitution herein.
  • A “non-naturally occurring amino acid residue” refers to a residue, other than those naturally occurring amino acid residues listed above, which is able to covalently bind adjacent amino acid residues(s) in a polypeptide chain. Examples of non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine and other amino acid residue analogues such as those described in Ellman et al. Meth. Enzym. 202:301 336 (1991). To generate such non-naturally occurring amino acid residues, the procedures of Noren et al. Science 244:182 (1989) and Ellman et al., supra, can be used. Briefly, these procedures involve chemically activating a suppressor tRNA with a non-naturally occurring amino acid residue followed by in vitro transcription and translation of the RNA.
  • An “amino acid insertion” refers to the incorporation of at least one amino acid into a predetermined amino acid sequence. While the insertion will usually consist of the insertion of one or two amino acid residues, the present application contemplates larger “peptide insertions”, e.g. insertion of about three to about live or even up to about ten amino acid residues. The inserted residue(s) may be naturally occurring or non-naturally occurring as disclosed above.
  • An “amino acid deletion” refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.
  • The term “polynucleotide(s)” refers to nucleic acids such as DNA molecules and RNA molecules and analogs thereof (e.g., DNA or RNA generated using nucleotide analogs or using nucleic acid chemistry). As desired, the polynucleotides may be made synthetically, e.g. using art-recognized nucleic acid chemistry or enzymatically using, e.g., a polymerase, and, if desired, be modified. Typical modifications include methylation, biotinylation, and other art-known modifications. In addition, the nucleic acid molecule can be single-stranded or double-stranded and, where desired, linked to a detectable moiety.
  • The term “mutagenesis” refers to unless otherwise specified, any art recognized technique for altering a polynucleotide or polypeptide sequence. Preferred types of mutagenesis include error prone PCR mutagenesis, saturation mutagenesis, or other site directed mutagenesis.
  • “Site-directed mutagenesis” is a technique standard in the art, and is conducted using a synthetic oligonucleotide primer complementary to a single-stranded phage DNA to be mutagenized except for limited mismatching, representing the desired mutation. Briefly, the synthetic oligonucleotide is used as a primer to direct synthesis of a strand complementary to the single-stranded phage DNA, and the resulting double-stranded DNA is transformed into a phage-supporting host bacterium. Cultures of the transformed bacteria are plated in top agar, permitting plaque formation from single cells that harbor the phage. Theoretically, 50% of the new plaques will contain the phage having, as a single strand, the mutated form; 50% will have the original sequence. Plaques of interest are selected by hybridizing with kinased synthetic primer at a temperature that permits hybridization of an exact match, but at which the mismatches with the original strand are sufficient to prevent hybridization. Plaques that hybridize with the probe are then selected, sequenced and cultured, and the DNA is recovered.
  • The term “vector” is used to refer to a rDNA molecule capable of autonomous replication in a cell and to which a DNA segment, e.g., gene or polynucleotide, can be operatively linked so as to bring about replication of the attached segment. Vectors capable of directing the expression of genes encoding for one or more polypeptides are referred to herein as “expression vectors. The term “control sequences” refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • Percent amino acid sequence identity may be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al., Nucleic Acids Res. 25:3389-3402 (1997)). The NCBI-BLAST2 sequence comparison program may be downloaded from http://www.ncbi.nlm.nih.gov or otherwise obtained from the National Institute of Health. Bethesda, Md. NCBI-BLAST2 uses several search parameters, wherein all of those search parameters are set to default values including, for example, unmask=yes, strand=all, expected occurrences=10, minimum low complexity length=15/5, multi-pass e-value=0.01, constant for multi-pass=25, dropoff for final gapped alignment=25 and scoring matrix=BLOSUM62.
  • “Antibody-dependent cell-mediated cytotoxicity” and “ADCC” are used herein to refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express FcRs (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. Various immune cells express different Fc receptors (FcRs). Thus, the primary cells for mediating ADCC, NK cells, express FcγRIII only, whereas monocytes express FcγRI, FcγRII and FcγRIII.
  • The terms “influenza A subtype” or “influenza A virus subtype” are used interchangeably, and refer to influenza A virus variants that are characterized by various combinations of the hemagglutinin (H) and neuraminidase (N) viral surface proteins, and thus are labeled by a combination of an H number and an N number, such as, for example, H1N1 and H3N2. The terms specifically include all strains (including extinct strains) within each subtype, which usually result from mutations and show different pathogenic profiles. Such strains will also be referred to as various “isolates” of a viral subtype, including all past, present and future isolates. Accordingly, in this context, the terms “strain” and “isolate” are used interchangeably.
  • The term “influenza” is used to refer to a contagious disease caused by an influenza virus.
  • B. General Techniques
  • Techniques for performing the methods of the present invention are well known in the art and described in standard laboratory textbooks, including, for example, Ausubel et al., Current Protocols of Molecular Biology, John Wiley and Sons (1997); Molecular Cloning: A Laboratory Manual. Third Edition, J. Sambrook and D. W. Russell, eds., Cold Spring Harbor, N.Y., USA, Cold Spring Harbor Laboratory Press, 2001; O'Brian et al., Analytical Chemistry of Bacillus Thuringiensis, Hickle and Fitch, eds., Am. Chem. Soc. 1990; Bacillus thuringiensis: biology, ecology and safety, T. R. Glare and M. O'Callaghan, eds., John Wiley, 2000; Antibody Phage Display, Methods and Protocols, Humana Press, 2001; and Antibodies, G. Subramanian, ed., Kluwer Academic, 2004. Mutagenesis can, for example, be performed using site-directed mutagenesis (Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)). PCR amplification methods are described in U.S. Pat. Nos. 4,683,192, 4,683,202, 4,800,159, and 4,965,188, and in several textbooks including “PCR Technology: Principles and Applications for DNA Amplification”, H. Erlich, ed., Stockton Press, New York (1989); and PCR Protocols: A Guide to Methods and Applications, Innis et al., eds., Academic Press, San Diego, Calif. (1990).
  • The methods of the present invention are not limited by any particular technology used for the display of antibodies. Although the invention is illustrated with reference to phage display, antibodies of the present invention can also be identified by other display and enrichment technologies, such as, for example, ribosome or mRNA display (Mattheakis et al., Proc. Natl. Acad. Sci. USA 91:9022-9026 (1994); H-lanes and Pluckthun, Proc. Natl. Acad. Sci. USA 94:4937-4942 (1997)), microbial cell display, such as bacterial display (Georgiou et al., Nature Biotech. 15:29-34 (1997)), or yeast cell display (Kieke et al., Protein Eng. 10:1303-1310 (1997)), display on mammalian cells, spore display (Isticato et al., J. Bacteriol. 183:6294-6301 (2001); Cheng et al., Appl. Environ. Microbiol. 71:3337-3341 (2005) and co-pending provisional application Ser. No. 60/955,592, filed Aug. 13, 2007) viral display, such as retroviral display (Urban et al. Nucleic Acids Res. 33:e35 (2005), display based on protein-DNA linkage (Odegrip et al., Proc. Acad. Natl. Sci. USA 101:2806-2810 (2004); Reiersen et al., Nucleic Acids Res. 33:e10 (2005)), and microbead display (Sepp et al. FEBS Lett. 532:455-458 (2002)).
  • In ribosome display, the antibody and the encoding mRNA are linked by the ribosome, which at the end of translating the mRNA is made to stop without releasing the polypeptide. Selection is based on the ternary complex as a whole.
  • In a mRNA display library, a covalent bond between an antibody and the encoding mRNA is established via puromycin, used as an adaptor molecule (Wilson et al., Proc. Natl. Acad. Sci. USA 98:3750-3755 (2001)). For use of this technique to display antibodies, see, e.g. Lipovsek and Pluckthun, J. Immunol. Methods. 290:51-67 (2004).
  • Microbial cell display techniques include surface display on a yeast, such as Saccharomyces cerevisiae (Boder and Wittrup. Nat. Biotechnol. 15:553-557 (1997)). Thus, for example antibodies can be displayed on the surface of S. cerevisiae via fusion to the α-agglutinin yeast adhesion receptor, which is located on the yeast cell wall. This method provides the possibility of selecting repertoires by flow cytometry. By staining the cells by fluorescently labeled antigen and an anti-epitope tag reagent, the yeast cells can be sorted according to the level of antigen binding and antibody expression on the cell surface. Yeast display platforms can also be combined with phage (see, e.g., Van den Beucken et al., FEBS Leit. 546:288-294 (2003)).
  • For a review of techniques for selecting and screening antibody libraries see, e.g. Hoogenboom, Nature Biotechnol. 23(9): 1105-1116 (2005).
  • C. Detailed Description of Preferred Embodiments
  • I. Preparation of Donor-Specific Antibody Libraries
  • The present invention concerns donor-specific antibody libraries from individuals who have successfully survived or are surviving an encounter with a specific disease. The resulting antibody repertoires will include antibodies that were used by the donor to defend specifically against a relevant disease and thus are important tools, for example, for developing neutralizing antibodies for the prevention and/or treatment of a target disease.
  • While the present invention is applicable to any target disease that evokes antibody production in a human subject, representative, non-limiting, examples of such diseases are listed in Table 1.
  • TABLE 1
    Type of disorder Representative examples
    infectious disorder Influenza viral infection, hepatitis C virus
    (HCV) infection, herpes simplex virus (HSV)
    infection, human immunodeficiency virus
    (HIV) infection, Methicillin-resistant
    Staphylococcus aureus (MRSA) infection,
    Epstein-Barr virus (EBV) infection, respiratory
    syncytial virus (RSV) infection, Pseudomonas.
    Candida infections
    Respiratory disorder Asthma, Allergies, chronic obstructive
    pulmonary disease (COPD), idiopathic
    pulmonary fibrosis (IPF), adult respiratory
    distress syndrome (ARDS)
    metabolic disorder Frailty, cachexia, sarcopenia, Obesity, type II
    diabedyslipidemia, metabolic syndrome-
    associated myocardial infarction (MI), chronic
    renal failure (CRF), osteoporosis
    digestive disorder irritable bowel syndrome (IBS), inflammatory
    bowel disease (IBD), Chron's disease, fatty liver
    disease, fibrosis, drug-induced liver disease
    Neurological disorder Alzheimer's disease, multiple sclerosis (MS),
    Parkinson's disease, bovine spongiform
    encephalopathy (BSE, mad cow disease)
    Cancer e.g., breast, renal, stomach, melanoma, lung,
    colon, glioma, lymphoma
  • A method of creating the donor-specific libraries of the present invention is schematically illustrated in FIG. 1. As a first step, potential donors are identified. The patient donor may currently suffer from or may have recovered from and survived a target disease. Thus, for example, as illustrated in the Examples, the donor-specific libraries herein may be created from the bone marrow of convalescent patients of prior influenza infections, including seasonal influenza outbreaks, epidemics, and pandemics.
  • When selecting or identifying a patient donor, it is important to confirm that the patient indeed had or is having the target disease. Part of the confirmation is the examination of the medical history of the patient donor. In addition to the medical history, various other factors, such as the medical history of the patient's family, the patient's sex, weight, health state, etc., should be taken into consideration. If the patient history is not available or unreliable, or for any other reason such as a further confirmation measure the serological profile of the patient may be determined. Serological assays are well known in the art and can be performed in various formats, such as in the form of various ELISA assay formats. Thus, for example, the presence of antibodies to an influenza virus can be detected by the well-known hemagglutinin inhibition (HAI) assay (i, A. P., M. S. Pereira, and J. J. Skehel. 1982. Concepts and procedures for laboratory-based influenza surveillance. U.S. Department of Health and Human Services, Public Health Service, Centers for Disease Control, Atlanta. Ga.), or the microneutralization assay (Harmon et al., J. Clin. Microbiol. 26:333-337 (1988)). This step might not be necessary if the serum sample has already been confirmed to contain influenza neutralizing antibodies.
  • In order to prepare donor-specific human antibody libraries, samples containing lymphocytes are collected from individuals (patient donors) known to have developed a target disease such as at least one disease from those listed in Table 1. The sample may for example, derive from bone marrow blood, spleen, lymph nodes, tonsils, thymus and the like. Bone marrow is a preferred source of the antibody libraries herein since it represents the complete “fossil archive” of individual donor's mature antibody repertoire.
  • Samples containing lymphocytes can be collected from the patient donor at various time points. In one embodiment, lymphocytes are collected from a patient who has recovered from the targeted disease(s) at least for 1, 5, 10, 15, 20, 25 days, at least for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 months, or at least for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 years. In another embodiment, lymphocytes are collected from a patient who is having the targeted disease(s) at the time of collection, and has been diagnosed as having the disease(s) at least 1, 5, 10, 15, 20, 25 days or at least 1, 2, 3, 4, 5, 6, 8, 9, 10 months, or 1, 2, 3, 4, or 5 years prior to collection.
  • Peripheral blood samples, especially from geographically distant sources, may need to be stabilized prior to transportation and use. Kits for this purpose are well known and commercially available, such as, for example, BD Vacutainer® CPT™ cell preparation tubes can be used for centrifugal purification of lymphocytes, and guanidium. Trizol, or RNAlater used to stabilize the samples. Methods and kits for isolating lymphocytes from other sources, such as lymphoid organs are also well known and commercially available.
  • Upon receipt of the stabilized lymphocytes or whole bone marrow, RNA is extracted and RT-PCR is performed to rescue antibody heavy and light chain repertoires, using immunoglobulin oligo primers known in the art.
  • Methods for preparation of RNA from bone marrow lymphocytes, or lymphocytes from any other source are well known in the art. General methods for mRNA extraction are disclosed in standard textbooks of molecular biology, including Ausubel et al., Current Protocols of Molecular Biology, John Wiley and Sons (1997). RNA purification kits are available from commercial manufacturers, such as Qiagen, and can be used according to the manufacturer's instructions.
  • Since RNA cannot serve as a template for PCR, it is first reverse transcribed into cDNA, which is subjected to PCR amplification. The two most commonly used reverse transcriptases are avilo myeloblastosis virus reverse transcriptase (AMV-RT) and Moloney murine leukemia virus reverse transcriptase (MMLV-RT). The reverse transcription step is typically primed using specific primers, random hexamers, or oligo-dT primers, depending on the circumstances and the goal of expression profiling. For example, extracted RNA can be reverse-transcribed using a GeneAmp RNA PCR kit (Perkin Elmer. CA, USA), following the manufacturer's instructions. The derived cDNA can then be used as a template in the subsequent PCR reaction.
  • In order to create phage-display libraries, the PCR repertoire products may be combined with linker oligos to generate scFv libraries to clone directly in frame with m13 pIII protein, following procedures known in the art. Libraries using other display techniques, such as those discussed above, can be prepared by methods well known in the art.
  • In a typical protocol, whole RNA is extracted by Tri BD reagent (Sigma) from fresh or RNAlater stabilized tissue. Subsequently, the isolated donor total RNA is further purified to mRNA using Oligotex purification (Qiagen). Next first strand cDNA synthesis, is generated by using random nonamer oligonucleotides and or oligo (dT)18 primers according to the protocol of AccuScript reverse transcriptase (Stratagene). Briefly, 100 ng mRNA, 0.5 mM dNTPs and 300 ng random nonamers and or 500 ng oligo (dT)18 primers in Accuscript RT buffer (Stratagene) are incubated at 65° C. for 5 min, followed by rapid cooling to 4° C. Then, 100 mM DTT, Accuscript RT, and RNAse Block are added to each reaction and incubated at 42° C. for 1 h, and the reverse transcriptase is inactivated by heating at 70° C. for 15 minutes. The cDNA obtained can be used as a template for RT-PCR amplification of the antibody heavy and light chain V genes, which can then be cloned into a vector, or, if phage display library is intended, into a phagemid vector. This procedure generates a repertoire of antibody heavy and light chain variable region clones (VH and VL libraries), which can be kept separate or combined for screening purposes. The vector, such as a phagemid vector, can then be introduced into a host cell, such as an E coli host, to generate a vector collection comprising a repertoire of nucleic acid molecules encoding antibody light chains or heavy chains or fragments thereof. In each case, the vector collection may comprise a single or more than one antibody light chain or heavy chain subtype. Thus, the vector collection may comprise sequences encoding antibody κ and/or λ light chains.
  • In the methods of the present invention, typically antibody light chains and antibody heavy chains are at first cloned separately, as discussed above, also separating the κ and λ light chain libraries. The libraries can be archived, and, when needed, the heavy chain library can be combined with the segregated κ and λ light chain libraries and heavy and light chain pairings can be identified, e.g. by panning, in the case of phage display. It is possible to repeat these steps multiple times with various libraries or sub-libraries, depending on the goal to be attained. The methods of the present invention provide great flexibility in including or excluding libraries, sub-libraries or clones, as needed during panning in order to maximize success.
  • In particular, because the sequences present in the vector collection harbor the coding sequences of the antibody heavy and light chains (or fragments) separately, the sequences may be excised and inserted into one or more expression vectors for expression of the antibody heavy and light chains, or fragments thereof. Preferably, the coding sequences of the antibody heavy and light chains, or fragments thereof, are inserted into the same expression vector for coexpression of the heavy and light chains to produce the library of the antibodies or antibody fragments.
  • The expression vectors of the present invention contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2μ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
  • Expression vectors will typically contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the antibodies-encoding nucleic acid, such as DHFR or thymidine kinase. An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described by Urlaub et al. Proc. Natl. Acad. Sci. USA. 77:4216 (1980). A suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7 [Stinchcomb et al., Nature, 282:39 (1979); Kingsman et al., Gene, 7:141 (1979); Tschemper et al., Gene, 10:157 (1980)]. The trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1 [Jones, Genetics, 85:12 (1977)].
  • Expression and cloning vectors usually contain a promoter operably linked to the antibody-encoding nucleic acid sequence to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the β-lactamase and lactose promoter systems [Chang et al., Nature, 275:615 (1978); Goeddel et al., Nature, 281:544 (1979)], alkaline phosphatase, a tryptophan (trp) promoter system [Goeddel, Nucleic Acids Res., 8:4057 (1980); EP 36,776], and hybrid promoters such as the tac promoter [deBoer et al., Proc. Natl. Acad. Sci. USA, 80:21-25 (1983)]. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding antibodies
  • Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase [Hitzeman et al., J. Biol. Chem., 255:2073 (1980)] or other glycolytic enzymes [Hess et al., J. Adv. Enzyme Reg., 7:149 (1968); Holland, Biochemistry, 17:4900 (1978)], such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • Other yeast promoters, which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73.657.
  • Transcription of the heavy chain or light chain genes in the expression vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 Jul. 1989), adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems.
  • Transcription of a DNA encoding the antibody genes by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, α-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. The enhancer may be spliced into the vector at a position 5′ or 3′ to the antibody coding sequence, but is preferably located at a site 5′ from the promoter.
  • Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding antibody heavy and light chains.
  • Still other methods, vectors, and host cells suitable for adaptation to the synthesis of polypeptide, in recombinant vertebrate cell culture are described in Gething et al., Nature, 293:620-625 (1981); Mantei et al., Nature, 281:40-46 (1979); EP 117,060; and EP 117,058.
  • In a particular embodiment, the antibody library is produced in the form of a phage library, where the coding sequences of the antibody heavy and light chains, or fragments thereof, are cloned into a phagemid vector, such as a vector comprising the bacteriophage gene III. Phagemid vectors are well known and commercially available, including, for example, the pBluescript vector SKII+(Stratagene, Genbank Accession X52328), and other pBluscript vectors. Phage display technology enables the generation of large repertoires of human antibodies, and the biopanning procedure allows the selection of individual antibodies with any desired specificity or other properties.
  • For example, immunoglobulin repertoires from peripheral lymphocytes of survivors of earlier epidemics and pandemics, such as the 1918 Spanish Flu, can be retrieved, stabilized, rescued and expressed in a manner similar to that described above. For additional H1 and H3 libraries, repertoires can be recovered from properly timed vaccinated locally-sourced donors. As an additional option, commercially available bone marrow total RNA or mRNA can be purchased from commercial sources to produce libraries suitable for H1 and H3, and, depending upon the background of donor, also suitable for H2 antibody screening. In general, for target diseases where vaccination is an available treatment option, antibodies can be isolated from biological samples obtained from immunized human donors as well. From immunized patients that have developed titers of antibody recognizing the particular antigen, bone marrow, blood, or another source of lymphocytes, is collected, and antibodies produced are isolated, amplified and expressed as described above.
  • As discussed above, for each donor, antibody light and heavy chain libraries can be cloned separately. Thus, for each donor, various κ and λlight chain families can be separately pooled and cloned in equimolar amounts. Similarly, for each donor, various heavy chain families can be pooled and cloned in equimolar amounts. By enabling gene family specific rescue of antibodies, the methods of the present invention yield libraries more completely representing the antibody repertoire of the donor, including antibodies that are less abundant and, in the case of pooled antibody libraries, guaranteeing immunoglobulin contributions from any and all individuals. For example, as illustrated in Example 1 by the Examples, in preparing the influenza heavy and light chain libraries herein, 6 κ light chain families, 11λ light chain families and 4 heavy chain families were rescued.
  • A typical screen can yield zero, one, or more than one target specific positive clone(s). If a particular combinatorial antibody library or libraries have been exhaustively screened and no further solutions seem attainable this may not be a failure of the heavy and light chain repertoire(s) ability to bind target, but rather the collection may have failed to bring together the necessary heavy and light chain pair required to bind target. A typical rescued repertoire of light chains from any individual may contain between about 105-106 unique light chains and between about 106-108 unique heavy chains. The possible combinatorial products of such pairings range from 1011 to 1014. Such a collection exceeds the practical limits of most display systems, such as phage display, by several orders of magnitude. Consequently, with current display technologies (such as phage display), only a fraction of the combinatorial possibilities are captured and assessed in any single phage antibody library. Therefore, recloning the original set of heavy chains with the original collection of light chains will generate an entirely new set of shuffled heavy and light chain combinations with likely novel antibodies to a particular target. Such newly reshuffled collections were found to transform previously existing poorly performing donor specific libraries into highly productive collections. Specifically, for a collection from a single donor previously only 0.3×-fold enrichment could be achieved compared to background after three rounds of selection. However, when this collection was recloned and reshuffled it became capable of 15-fold enrichment following 3 rounds of panning resulting in 55 novel sequences from 92 selected clones.
  • As mentioned previously, a typical screen can yield any number of target specific positive clones. The present invention enables the identification of the origins of any clone by their embedded barcode. As a typical antibody screen may combine phage antibodies from numerous donor specific libraries it is possible that some of the libraries and their combinatorial clones are not completely represented as antibody bearing phage particles. In which case a positive clone may have resulted from only a limited physical set of all the possible cloned solutions present in the sub-library phage population being screened. In such an instance it is of considerable interest to more fully interrogate the collection of donor specific phage. In this case the barcode from a positive clone guides one to the specific library responsible for the clone and allows to exclusively and more deeply screen the collection of interest.
  • In instances where desired antibodies must have functional capabilities beyond those initially used as the basis for the initial library construction, such as neutralization, or activation, we can prospectively profile individual donor sera for evidence of such activities. If the desired activities are present at reasonable titers in any particular donor sera, one can select those corresponding libraries are selected to screen against the target of interest. In other instances the relevant selection criteria may be unrelated to serology, but related to donor characteristics such as age, gender, or medical histories. In any event, donor profiles are logical guides for library selection and possible only in donor specific and segregated antibody libraries.
  • It is not unusual to complete a phage panning screen and discover the presence of immunodominant clones. Furthermore, it is also not unusual to rediscover such clone upon repeated panning screening regimens. In the case of a dominant clone or clones, where either more or different clones are desirable, it is important to avoid the library material responsible for the presence of this clone. In typical phage antibody libraries the specific library or materials responsible for the clones origin are not separable from the collection, however in donor specific libraries it is possible to rescreen the libraries and simply omit the undesirable donor sublibrary or sublibraries, thereby forcing positive selection away from previously identified dominant clones.
  • Although, for simplicity, the libraries are described as heavy or light chain libraries, it will be apparent to those of ordinary skill in the art that the same description applies to the libraries of antibody fragments, fragments of antibody heavy and/or light chains, and libraries of antibody-like molecules.
  • In a particular embodiment, antibodies with dual specificities, such as, for example, showing reactivity with two different influenza A subtypes and/or with two strains (isolates) of the same subtype, and/or with human and non-human isolates, can be discovered and optimized through controlled cross-reactive selection and/or directed combinatorial and/or mutagenic engineering.
  • In a typical enrichment scheme, illustrated in FIG. 2, a library including antibodies showing cross-reactivity to two targets, designated as targets A and B, are subjected to multiple rounds of enrichment. If enrichment is based on reactivity with target A, each round of enrichment will increase the reactive strength of the pool towards target A. Similarly, if enrichment is based on reactivity with target B, each round of enrichment will increase the reactive strength of the pool towards target B. Although FIG. 2 refers to panning, which is the selection method used when screening phage display libraries (see below), the approach is equally applicable to any type of library discussed above, or otherwise known in the art, and to any type of display technique. Targets A and B include any targets to which antibodies bind, including but not limited to various isolates, types and sub-types of influenza viruses.
  • If the goal is to identify neutralizing antibodies with multiple specificities, a cross-reactive discovery selection scheme can be used. In the interest of simplicity, this scheme is illustrated in FIG. 3 showing the selection of antibodies with dual specificities. In this case, an antibody library including antibodies showing reactivity with two targets, targets A and B, is first selected for reactivity with one of the targets, e.g., target A, followed by selection for reactivity with the other target, e.g., target B. Each successive selection round reinforces the reactive strength of the resulting pool towards both targets. Accordingly, this method is particularly useful for identifying antibodies with dual specificity. Of course, the method can be extended to identifying antibodies showing reactivity towards further targets, by including additional rounds of enrichment towards the additional target(s). Again, if the library screened is a phage display library, selection is performed by cross-reactive panning, but other libraries and other selection methods can also be used.
  • A combination of the two methods discussed above includes two separate enrichment rounds for reactivity towards target A and target B, respectively, recombining the two pools obtained, and subsequent cross-reactive selection rounds, as described above. This approach is illustrated in FIG. 4. Just as in the pure cross-reactive selection, each round of selection of the recombined library increases the reactive strength of the resulting pool towards both targets.
  • In a further embodiment, illustrated in FIG. 5, first a clone showing strong reactivity with a target A, and having detectable cross-reactivity with target B is identified. Based on this clone, a mutagenic library is prepared, which is then selected, in alternating rounds, for reactivity with target B and target A respectively. This scheme will result in antibodies that maintain strong reactivity with target A, and have increased reactivity with target B. Just as before, selection is performed by panning, if the libraries screened are phage display libraries, but other libraries, other display techniques, and other selection methods can also be used, following the same strategy.
  • As discussed above, targets A and B can, for example, be two different subtypes of the influenza A virus, two different strains (isolates) of the same influenza A virus, subtypes or isolates from two different species, where one species is preferably human. Thus, for example, target A may be an isolate of the 2004 Vietnam isolate of the H5N1 virus, and target B may be a 1997 Hong Kong isolate of the H5N1 virus. It is emphasized that these examples are merely illustrative, and antibodies with dual and multiple specificities to any two or multiple targets can be identified, selected and optimized in an analogous manner.
  • Once neutralizing antibodies with the desired properties have been identified, it might be desirable to identify the dominant epitope or epitopes recognized by the majority of such antibodies. Methods for epitope mapping are well known in the art and are disclosed, for example, in Morris, Glenn E., Epitope Mapping Protocols, Totowa, N. J. ed., Humana Press, 1996: and Epitope Mapping: A Practical Approach, Westwood and Hay. eds. Oxford University Press, 2001.
  • II. Identifying Donor-Specific Antibody Library with Unique Barcoding
  • According to the present invention, following amplification of the antibody heavy and light chain repertoires from cDNA, such as bone marrow cDNA, prepared as described above, preferably antibody heavy and light chain libraries are cloned separately for each patient donor, where the individual libraries can be distinguished using unique barcodes.
  • The barcodes preferably are selected such that they are capable of propagating along with the clone(s) labeled, without interfering with the expression of the desired antibody chain or fragment thereof. In an exemplary embodiment of the present invention, the barcode is inserted into the sequence of the expression vector, preferably, the 3′ untranslated region following the terminal pIII stop codon when a phagemaid vector is used. Upon clonal isolation, a vector's unique sequence is determined and subsequently dedicated to a single defined library. This defined library can be derived not only from a single donor, but also from discrete pools of donors, or a synthetic repertoire or a semi-synthetic collection. In another embodiment, the barcode is inserted into the coding sequence of the antibody heavy and/or light chain or fragment thereof, at a position or in a form that does not interfere with the expression of the respective chains.
  • Thus the barcodes can be non-coding DNA sequences of about 1-24 contiguous non-coding nucleotides in length that can be deconvoluted by sequencing or specific PCR primers. This way, a collection of nucleic acids, such as an antibody repertoire, can be linked at the cloning step. In a exemplary embodiment of the present invention, the barcode is 3 or bases of randomly generated sequence.
  • In another example, the barcodes are coding sequences of silent mutations. If the libraries utilize restriction enzymes that recognize interrupted palidromes (e.g. Sfi GGCCNNNNNGGCC), distinct nucleotides can be incorporated in place of the “N's” to distinguish various collections of clones, such as antibody libraries. This barcoding approach has the advantage that the repertoire is linked at the amplification step.
  • In a further embodiment, the barcodes are non-contiguous nucleotide sequences, which may be present in the vector sequence and/or the coding sequence of the desired antibody chain. Thus, a barcode with a non-contiguous sequence provides a great degree of flexibility in identifying the origins of the various individual sequences, and monitoring their subsequent handling.
  • In a different example, the barcodes are coding sequences that encode immunologically distinct peptide or protein sequences fused to phage particles. Examples include, for example, epitope (e.g. Myc, HA, FLAG) fusions to pIII, pVIII, pVII, or pIX phages. The epitopes can be used singly or in various combinations, and can be provided in cis (on the library-encoding plasmid) or in trans (specifically modified helper phage) configuration.
  • Other examples of possible barcodes include, without limitation, chemical and enzymatic phage modifications (for phage libraries) with haptens or fluorescent chromophores. Such tags are preferred for a single round of selection.
  • The individual heavy and light chain libraries obtained from individual donors, or other barcoded clone or collections, can be pooled, without losing the ability to distinguish the source of individual sequences.
  • III. Optimizing Neutralizing Antibodies from the Donor-Specific Antibody Libraries
  • If desired, cross-reactivity of the neutralizing antibodies with dual or multiple specificity can be further improved by methods known in the art, such as, for example, by Look Through Mutagenesis (LTM), as described in US. Patent Application Publication No. 20050136428, published Jun. 23, 2005, the entire disclosure of which is hereby expressly incorporated by reference.
  • Look-through mutagenesis (LTM) is a multidimensional mutagenesis method that simultaneously assesses and optimizes combinatorial mutations of selected amino acids. The process focuses on a precise distribution within one or more complementarity determining region (CDR) domains and explores the synergistic contribution of amino acid side-chain chemistry. LTM generates a positional series of single mutations within a CDR where each wild type residue is systematically substituted by one of a number of selected amino acids. Mutated CDRs are combined to generate combinatorial single-chain variable fragment (scFv) libraries of increasing complexity and size without becoming prohibitive to the quantitative display of all variants. After positive selection, clones with improved properties are sequenced, and those beneficial mutations are mapped. To identify synergistic mutations for improved binding properties, combinatorial libraries (combinatorial beneficial mutations, CBMs) expressing all beneficial permutations can be produced by mixed DNA probes, positively selected, and analyzed to identify a panel of optimized scFv candidates. The procedure can be performed in a similar manner with Fv and other antibody libraries.
  • Mutagenesis can also be performed by walk-through mutagenesis (WTM), as described above.
  • Another useful mutagenic method to intentionally design cross-reactivity of the antibodies herein with more than one influenza A subtype and/or more than one isolate of the same subtype, is referred herein as “destinational” mutagenesis. Destinational mutagenesis can be used to rationally engineer a collection of antibodies based upon one or more antibody clones, preferably of differing reactivities. In the context of the present invention, destinational mutagenesis is used to encode single or multiple residues defined by analogous positions on like sequences such as those in the individual CDRs of antibodies. In this case, these collections are generated using oligo degeneracy to capture the range of residues found in the comparable positions. It is expected that within this collection a continuum of specificities will exist between or even beyond those of the parental clones. The objective of destinational mutagenesis is to generate diverse multifunctional antibody collections, or libraries, between two or more discrete entities or collections. To create a destinational mutagenesis library, the CDR sequences for both antibodies are first attained and aligned. Next all positions of conserved identity are fixed with a single codon to the matched residue. At non-conserved positions a degenerate codon is incorporated to encode both residues. In some instances the degenerate codon will only encode the two parental residues at this position. However, in some instances additional co-products are produced. The level of co-product production can be dialed in to force co-product production or eliminate this production dependent upon size limits or goals.
  • Thus, for example, if the first position of the two antibodies respectively are threonine and alanine, the degenerate codon with A/G-C- in the first two positions would only encode threonine or alanine, irrespective of the base in the third position. If, for example, the next position residues are lysine and arginine the degenerate codon A-A/G-A/G will only encode lysine or arginine. However, if the degenerate codon A/C-A/G-A/G/C/T were used then asparagine, histidine, glutamine, and serine coproducts will be generated as well.
  • As a convenience it is simpler to use only antibodies with matched CDR lengths. One way to force this is to screen a size restricted library for the second antigen, based on the CDR length and potentially even framework restrictions imparted by the initially discovered antibody. It is noted, however, that using CDRs of equal length is only a convenience and not a requirement. It is easy to see that, while this method will be useful to create large functionally diverse libraries of influenza A virus neutralizing antibodies, its applicability is much broader. This mutagenesis technique can be used to produce functionally diverse libraries or collections of any antibody. Thus, FIG. 6 is included herein to illustrate the use of the destinational mutagenesis method using CDRs of a TNF-α antibody and a CD11a antibody as the parental sequences mutagenized.
  • Other exemplary mutagenesis methods include saturation mutagenesis and error prone PCR.
  • Saturation mutagenesis (Hayashi et al., Biotechniques 17:310-315 (1994)) is a technique in which all 20 amino acids are substituted in a particular position in a protein and clones corresponding to each variant are assayed for a particular phenotype. (See, also U.S. Pat. Nos. 6,171,820; 6,358,709 and 6,361,974.)
  • Error prone PCR (Leung et al., Technique 1:11-15 (1989); Cadwell and Joyce, PCR Method Applic. 2:28-33 (1992)) is a modified polymerase chain reaction (PCR) technique introducing random point mutations into cloned genes. The resulting PCR products can be cloned to produce random mutant libraries or transcribed directly if a T7 promoter is incorporated within the appropriate PCR primer.
  • Other mutagenesis techniques are also well known and described, for example, in In Vitro Mutagenesis Protocols. J. Braman, Ed. Humana Press. 2001.
  • Optimization can be based on any of the libraries discussed above, or any other types of libraries known in the art, alone or in any combination.
  • IV. Production of Neutralizing Antibodies
  • Once antibodies with the desired neutralizing properties are identified, such antibodies, including antibody fragments can be produced by methods well known in the art, including, for example, hybridoma techniques or recombinant DNA technology.
  • In the hybridoma method, a mouse or other appropriate host animal, such as a hamster, is immunized to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • The hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor. J. Immunol. 133:3001 (1984); and Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • Recombinant monoclonal antibodies can, for example, be produced by isolating the DNA encoding the required antibody chains and co-transfecting a recombinant host cell with the coding sequences for co-expression, using well known recombinant expression vectors. Recombinant host cells can be prokaryotic and eukaryotic cells, such as those described above.
  • The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity. According to the so-called “best-fit” method, the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the rodent is then accepted as the human framework region (FR) for the humanized antibody (Sims et al., J. Immunol. 151:2296 (1993); Chothia et al., J. Mol. Biol. 196:901 (1987)). It is important that antibodies be humanized with retention of high affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • In addition, human antibodies can be generated following methods known in the art. For example, transgenic animals (e.g., mice) can be made that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA 90:2551 (1993); Jakobovits et al. Nature 362:255-258 (1993); Bruggermann et al., Year in Immuno. 7:33 (1993); and U.S. Pat. Nos. 5,591,669, 5,589,369 and 5,545,807.
  • V. Use of Neutralizing Antibodies
  • The neutralizing antibodies of the present invention can be used for the prevention and/or treatment of the targeted diseases. For therapeutic applications, the antibodies or other molecules, the delivery of which is facilitated by using the antibodies or antibody-based transport sequences, are usually used in the form of pharmaceutical compositions.
  • Techniques and formulations generally may be found in Remington's Pharmaceutical Sciences, 18th Edition. Mack Publishing Co. (Easton, Pa. 1990). See also, Wang and Hanson “Parenteral Formulations of Proteins and Peptides: Stability and Stabilizers,” Journal of Parenteral Science and Technology. Technical Report No. 10, Supp. 42-2S (1988).
  • Antibodies are typically formulated in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride: hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben: catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol): low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium: metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).
  • The antibodies also may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively), in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, supra.
  • The neutralizing antibodies disclosed herein may also be formulated as immunoliposomes. Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang et al., Proc. Natl. Acad. Sci. USA 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and WO97/38731 published Oct. 23, 1997. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. Fab′ fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al. J. Biol. Chem. 257:286-288 (1982) via a disulfide interchange reaction. A chemotherapeutic agent is optionally contained within the liposome. See Gabizon et al. J. National Cancer Inst. 81(19)1484 (1989).
  • For the prevention or treatment of disease, the appropriate dosage of antibody will depend on the type of infection to be treated, the severity and course of the disease, and whether the antibody is administered for preventive or therapeutic purposes. The antibody is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to about 15 mg/kg of antibody is a typical initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • Further details of the invention are illustrated by the following non-limiting Examples.
  • Example 1 Antibody Libraries from Survivors of Prior Bird Flu Outbreaks and Preparation of Neutralizing Antibodies
  • Materials and Bone Marrow Protocol and Sera Preparation
  • Blood was obtained by standard venopuncture, allowed to clot, and processed to recover serum. The serum was stored at −20° C. for 3-4 days until they were shipped on dry ice. Donors were anaesthetized with an injection of a local anesthetic and 5 ml of bone marrow was removed from the pelvic bone of each H5N1 survivor. Next the 5 ml of bone marrow was placed into a sterile 50-ml tube containing 45 ml RNAlater (Ambion). The mixture was gently inverted approximately 8-20 times, until there were no visible clumps and the marrow and RNAlater were mixed well. Next the specimen was refrigerated the between 2-10° C. overnight. Following the overnight refrigeration, the specimens were stored at −20° C. for 3-4 days until they were shipped on dry ice. Upon receipt the RNAlater/marrow and sera containing tubes were stored at −80° C. until processed.
  • Serology: H/A ELISA
  • ELISA plates (Thermo, Immulon 4HBX 96W) were coated with 100 μl of 100 ng/mL H5 hemagglutinin (Protein Sciences, A/Vietnam/1203/2004) in IX ELISA Plate Coating Solution (BiolFX) by overnight incubation at room temperature. The next day plates were washed three times with 300 μl PBS/0.05% Tween-20 (PBST). Following the wash, 300 μl of a blocking solution (4% Non-Fat dry Milk in PBS/0.05% Tween-20) was added and incubated for 1 hour at RTf. Following the blocking step, the plates were washed three times with 300 μl PBS/0.05% Tween-20. Next, 100 μl serum samples diluted 1:20,000 in PBS/0.05% Tween were incubated for 1-2 hours at RT and then washed three times with 300 μl PBS/0.05% Tween-20. 100 μl of an anti-human Fc-HRP conjugate diluted 1:5,000 in PBS/0.05% Tween was incubated for 1-2 hours at RT and then washed three times with 300 μl PBS/0.05% Tween-20. Following this final wash, 100 μl of chromogenic substrate solution was added (TMB1 Substrate, BioFx) and after sufficient amount of time terminated by the addition of 100 μl of STOP Solution (BioFx). Absorbances at 450 nm were read on a plate reader (Molecular Devices Thermomax microplate reader with Softmax Pro software), data recorded, and subsequently plotted using Excel (Microsoft).
  • Bone Marrow: RNA Extraction and mRNA Purification
  • Bone marrow (˜2.5 ml in 20 ml RNA later), previously stored at −80° C., was recovered by centrifugation to remove RNA later and then resuspended in 11.25 ml TRI BD reagent (Sigma) containing 300 μl Acetic Acid. The pellet was then vortexed vigorously. Next 1.5 ml BCP (1-bromo-3-chloropropane, Sigma) was added, mixed by vortexing, incubated at RT for 5 min, and then centrifuged at 12000×g for 15 min at 4° C. The aqueous phase was carefully removed to not disturb the interface. Total RNA from the aqueous phase was next precipitated by addition of 25 ml isopropanol, incubation at RT for 10 minutes, and centrifugation at 12000×g for 10 min at 4° C. Following the addition of isopropanol, two phases were formed due to residual RNAlater, resulting in the precipitated RNA settling at the interface. To eliminate the residual RNAlater and allow maximal recovery of RNA, 5 ml aliquots of 50% isopropanol in H 20 were added and mixed until no phase separation was noticeable, at which point the RNA was pelleted by centrifugation at 12000×g for 10 min at 4° C. The RNA pellet was washed with 75% EtOH, transferred to an RNAse-free 1.6 ml microcentrifuge tube, and again recovered by centrifugation. Finally the RNA pellet was resuspended in 100 μl 1 mM Na-phosphate, pH 8.2 and the A260 and A280 were read to assess RNA purity.
  • Prior to reverse transcription mRNA was purified from total RNA according to Qiagen Oligotex mRNA purification kit. Briefly, 50-200 μg bone marrow RNA was brought to 250 μl with RNasc-free water and mixed with 250 μl of OBB buffer and Oligotex suspension followed by incubation for 3 min at 70° C. Hybridization between the oligo dT30 of the Oligotex particle and the mRNA poly-A-tail was carried out at room temperature for min. The hybridized suspensions were then transferred to a spin column and centrifuged for 1 min. The spin column was washed twice with 400 μl Buffer OW2. Purified mRNA was then eluted twice by centrifugation with 20 μl hot (70° C.) Buffer OEB. Typical yields were 500 ng to 1.5 μg total RNA.
  • Reverse Transcription Using N9 and Oligo dT on Bone Marrow mRNA
  • Reverse transcription (RT) reactions were accomplished by mixing together 75-100 ng mRNA with 2 μl 10× Accuscript RT Buffer (Stratagene), 0.8 μl 100 mM dNTPs, and either N9 (300 ng) or oligo dT primer (100 ng) and then brought to a final volume of 17 μl with water. The mixtures were heated at 65° C. for 5 min, and then allowed to cool to room temperature. Next 2 μl DTT, 0.5 μl RNase Block (Stratagene), 0.5 μl AccuScript RT (Stratagene) were added to each reaction. Next, the N9 primed reactions were incubated for 10 minutes at room temperature and the oligo-dT primed reactions were incubated on ice for 10 minutes. Finally, both reactions were incubated at 42° C. for 60 minutes followed by 70° C. for 15 minutes to kill the enzyme.
  • PCR from bone marrow-derived cDNA
  • Antibody heavy and light chain repertoires were amplified from bone marrow cDNA essentially using previously described methods and degenerate primers (O'Brien, P. M., Aitken R. Standard protocols for the construction of scFv Libraries. Antibody Phage Display Methods and Protocols, vol. 178, 59-71, 2001, Humana Press) based upon human germline V and J regions.
  • Briefly, PCR reactions using Oligo dT primed cDNA (from 75 ng mRNA) for lambda light chains and N9 primed cDNA (from 75 ng mRNA for kappa light chains, from 100 ng mRNA for heavy chains) were mixed together with 5 μl 10× amplification buffer (Invitrogen), 1.5 μl dNTPs (10 mM), 1 μl MgSO4 (50 mM), 2.5 μl Vregion primers (10 uM) and 2.5 μl Jregion primers (10 uM)-10 uM for VH, 0.5 μl Platinum Pfx Polymerase (Invitrogen), and sterile dH2O to final volume of 50 μl. PCR parameters were as follows: step 1-95° C. 5 minutes, step 2-95° C. 30 seconds, step 3-58° C. 30 seconds, step 4-68° C. 1 minute, step 5-cycle step 2-4 40 times, step 6-68° C. 5 minutes. Light chain PCR products were cleaned up using Qiagen PCR Cleanup kit. Heavy chains PCR products were gel purified from 1.5% agarose gel using Qiagen Gel Extraction Kit and then reamplified. Heavy chain reamplification was carried out as follows: Mixed 10 μl 10× amplification buffer (Invitrogen), 3 μl dNTPs (10 mM), 2 μl MgSO4 (50 mM), 5 μl each VH primers (10 uM) and JH primers (10 uM), 5 μl Heavy chain Primary PCR product, 1 μl Platinum Pfx, volume adjusted to 100 μl with water. Cycling parameters were as follows: step 1-95° C. 5 minutes, step 2-95° C. 30 seconds, step 3-58° C. 30 seconds, step 4-68° C. 1 minute, step 5-cycle step 2-4 20 times, step 6-68° C. 5 minutes. Re-amplified heavy chain PCR products were cleaned up from a 1.5% agarose-TAE gel using Qiagen Extraction Kit.
  • Antibody Phage Library Construction
  • Separate antibody libraries for each individual bird flu survivor were constructed using unique identifying 3-nucleotide barcodes inserted in the untranslated region following the stop codon of the pIII gene of filamentous phage.
  • Light Chain Cloning:
  • 1 μg each of pooled kappa light chain and pooled lambda light chain per donor were digested with NotI and BamHI and gel purified from a 1.5% agarose-TAE gel using Qiagen Gel Extraction Kit. 5 μg of each vector (pAMPFab) was digested with NotI and BamHI and gel purified from a 1% agarose-TAE gel using Qiagen Gel Extraction Kit. Library ligations were performed with 200 ng of gel purified Kappa or Lambda inserts and 1 μg of gel purified vector in 60 μl for 1 hour at RT or overnight at 14° C. Ligations were desalted using Edge BioSystem Perfroma spin columns. The library was transformed in five electroporations in 80 μl TG-1 or XL-1 Blue aliquots, each recovered in 1 ml SOC, pooled and outgrown for one hour at 37° C. Total number of transformants was determined following this outgrowth by plating an aliquot from each of the transformations. The remaining electroporation was amplified by growing overnight at 37° C. in 200 ml 2YT+50 μg/ml Ampicillin+2% glucose. The subsequent light chain library was recovered by plasmid purification from these overnight cultures using a Qiagen High Speed Maxiprep Kit.
  • Heavy Chain Cloning:
  • 1.5-2 μg each of the donor-specific heavy chains (V H1, V H2, 5, 6 pool. V H3, and VH4) were digested with a 40 Unit excess/μg DNA with Sfil and Xhol and gel purified from a 1.5% agarose-TAE gel using Qiagen Gel Extraction Kit. 15 μg of each light chain library vector was digested with 40 Unit/μg DNA with Sfil and Xhol and gel purified from a 1% agarose-TAE gel using Qiagen Gel Extraction Kit. Library ligations were set up by combining 1.2 μg Sfil/Xhol digested, gel purified heavy chain donor collections and 5 μg of each light chain library (kappa and lambda) overnight at 14° C. The library ligations were then desalted with Edge BioSystem Pefroma spin columns and then transformed through 20 electroporations per library in 80 μl TG-1 aliquots, each recovered in 1 ml SOC, pooled and outgrown for one hour at 37° C. Again following this outgrowth an aliquot of each was used to determine the total number of transformants with the remainder transferred to 1L 2YT+50 μg/ml Ampicillin +2% glucose and grown at 37 C with vigorous aeration to an OD600 of ˜0.3. Next M13K07 helper phage was then added at a multiplicity of infection (MOI) of 5:1 and incubated for 1 hour at 37° C., with no agitation. Next the cells were harvested by centrifugation and resuspended in 1L 2YT+50 μg/ml Ampicillin, 70 μg/ml Kanamycin and grown overnight at 37° C. with vigorous aeration to allow for scFv phagemid production. The next morning the cells were collected by centrifugation and supernatant containing phagemid was collected. The phagemids were precipitated from the supernatant by the addition of 0.2 volumes 20% PEG/5 M NaCl solution and incubation for 1 hour on ice. The phagemid library stocks were then harvested by centrifugation and resuspended in 20 ml sterile PBS. Residual bacteria were removed by an additional centrifugation and the final phagemid libraries were stored at −20° C. in PBS+50% glycerol.
  • Phagemid Panning and Amplification
  • ELISA plates (Immulon 4HBX flat bottom, Nunc) were coated with 100 μl of 100 ng/ml, H5 hemagglutinin protein(Protein Sciences, A/Vietnam/1203/2004) in ELISA Coating Solution (BioFX) by overnight incubation at room temperature. The next day plates were washed three times with 300 μl PBST. Following the wash, 300 μl of a blocking solution (4% Non-Fat dry Milk in PBS/0.05% Tween-20) was added and incubated for 30 mins on ice. Following the blocking step, the plates were washed three times with 300 μl PBST. Just prior to phage panning, the glycerol was removed from the frozen phagemid stocks using Millipore Amicon Ultra columns and then blocked in 4% nonfat dry milk for 15 minutes. Next, 100 μl aliquots of phagemid were distributed into 8 wells (total phage ˜1×1012 CFU) and incubated for 2 hours at 4° C. followed by washing 6-8 times with 300 μl PBST. Phagemid were collected following a 10 min at room temperature in 100 μl/well Elution buffer (0.2M glycine-HCl, pH 2.2, 1 mg/ml BSA). The eluate was then neutralized by the addition of 56.25 μl 2M Tris base per ml eluate. Following neutralization. 5 ml TGI cells (OD6oo ˜0.3) were infected with 0.5 ml neutralized phage at 37° C. for 30 minutes in 2-YT with no shaking. Following this step some cells were plated onto LB AMP Glucose plates to determine total phagemid recovery. The remaining inoculum was placed into 10 ml 2-YTAG (final concentration 2% glucose and 50 μg/ml ampicillin) and grown at 37° C. with vigorous aeration to OD600—0.3. Next the cultures were infected with M13K07 helper phage at an MOI of 5:1 and incubated at 37° C. for 30-60 minutes with no shaking. The cells were collected by centrifugation and resuspended in 25 ml 2-YTAK (Ampicillin 50 μg/ml, Kanamycin 70 μg/ml), transferred to a fresh culture flask, and grown ON at 37° C. with shaking. Subsequent rounds were similarly recovered and amplified.
  • scFv ELISA
  • Individual colonies of E. coli HB2151 transformed cells from biopanned phage were grown overnight at 37° C. in 1 ml of 2YT+100 μg/ml AMP. The following morning the cells were harvested by centrifugation and resuspended in 1.5 ml periplasmic lysis buffer (1 ml BBS (Teknova)+0.5 ml 10 mg/ml lysozyme+EDTA to 10 mM final concentration). The cells were again pelleted by centrifugation and the scFv containing periplasmic lysates were collected. The scFv lysates were combined 1:1 with dilution buffer (PBS/0.05% BSA) and 100 μl was added to wells that had been previously antigen coated with and blocked with dilution buffer. The samples were incubated for 2 hours at room temperature and then washed three times with PBS/0.05% Tween. Next 100 μl of 1:5000 diluted Biotin Anti-Histidine mouse (Serotec) in dilution buffer was added to each well and incubated for 1 hr at room temperature. Following this incubation the wells were washed three times with PBS/0.05% Tween and then to each well 100 μl of 1:2500 Streptavidin: HRP (Serotec) was added and incubated for 1 hr at room temperature and then washed three times with PBS/0.05% Tween. Following this final wash, 100 μl of chromogenic substrate solution was added (TMB1 Substrate, BioFx) and after sufficient amount of time terminated by the addition of 100 μl of STOP Solution (BioFx). Absorbances at 450 nm were read on a plate reader (Molecular Devices Thermomax microplate reader with Softmax Pro software), data recorded, and subsequently plotted using Excel (Microsoft).
  • Sequencing
  • To deduce the heavy and light chain sequences, individual clones were grown and plasmid DNA extracted (Qiagen). The plasmid DNA was subjected to standard DNA sequencing.
  • Hemagglutinin Inhibition (HAI) Assays
  • Hemagglutination Inhibition was performed essentially following the method of Rogers et al. Virology 131:394-408 (1983), in round bottom microliter plates (Corning) using 4 HAU (hemagglutinating units) of virus or protein/well. For HAI determinations 25 μl samples of purified single chain variable fragments (scFv) were mixed with 25 μl of PBS containing 4 HAU of the test virus in each microtiter well. Following a preincubation of 15 minutes at room temperature, 25 μl of 0.75% human erythrocytes were added, and mixed. HAI antibody activity was determined by visual inspection following a 60 min incubation at room temperature.
  • In particular, the following protocols were used:
  • Antibodies, Proteins and Viruses.
  • IgG1 proteins were generated from either scFv or Fabs by subcloning their coding regions into a pCI-based (Promega) full length mammalian protein expression and then transfecting them into 293 Freestyle cells (Invitrogen) according to manufacturers guidelines. Briefly 20 μg of light chain and 10 μg heavy chain encoding plasmid were combined with 1.0 ml 293fectin and incubated for 60 minutes. Following this preincubation the DNA mixture was combined with 3×107 cells in 30 ml media and the resulting cell suspension was grown according to manufacturers suggestion for 7 days. After seven days the secreted immunoglobulins were purified from the culture supernatants using protein A chromatography (Calbiochem). The resulting purified antibodies were buffer exchanged into sterile PBS using centrifugal size filtration (Centricon Plus-20) and their protein concentrations determined by colorimetric BCA assay (Pierce).
  • Cross-reaction IgG ELISA.
  • Microtiter plates were coated with 0.1 ml of the following antigens diluted in coating buffer and incubated overnight at room temperature: 100 ng/ml H5N1 Vietnam 1203/04, 250 ng/ml H5N1 Turkey/65596/06, 1 μg/ml H5N1 Indonesia/5/05, 700 ng/ml H1N1 New Caledonia/20/99, 1 μg/ml H1N1 North Carolina/1/18, 100 ng/ml and H3N2 Wisconsin/67/05. Blocking was done with 0.3 ml of blocking buffer (4% Non-fat dry milk in PBS/0.05% Tween-20). Following blocking antibodies diluted to 0.5 μg/ml in 2% non-fat dry milk blocking buffer were incubated for two hours at 4 C, washed, and later detected using a 1:3000 dilution of peroxidase conjugated anti-human Fc antibody (Jackson ImmunoResearch) in 2% non-fat dry milk blocking buffer and standard TMB substrate detection (BioFX). Absorbance at 450 nm was read, data recorded, and reported herein
  • Viral Microneutralization.
  • Indonesia and Turkey hemagglutinin genes were synthetically assembled using human codon optimized sequences (DNA 2.0) and then used to generate recombinant engineered viruses. Recombinant influenza viruses were generated using reverse genetics as previously described (Fodor, E. et al. J Virol. 1999; 73(11): 9679-82). Briefly, 1 ug each of 10 plasmids was transfected into 293 T cells in monolayer. Each transfection contained ambisense plasmids (for the expression of both vRNAs and mRNAs) for the A/Puerto Rico/8/34/PA, PB1, PB2, NP, M, and NS segments, in addition to vRNA (pPOL1 type) and protein expression plasmids (pCAGGS type) for A/Vietnam/1203/04 HA and NA (pCAGGS expression plasmid was kindly provided by J. Miyazaki, Osaka University, Osaka, Japan) (Miyazaki, J. et al. Gene 1989; 79(2):269-77). Twenty hours following transfection, 293T cells were resuspended in cell culture supernatant, and used to inoculate 10-day-old embryonated eggs.
  • Antibodies were screened for neutralizing activity against viruses as follows. Two fold serial dilutions of each Mab were incubated with 100 TCIDs50 of virus in PBS at 37° for 1 h. Madin-Darby Canine Kidney cell monolayers in 24 well plates were washed once with PBS and inoculated with virus-antibody mixtures. Following incubation for 1 h at 37° C. in 5% CO2 the inoculum was removed and monolayers were again washed once with PBS. Opti-MEM supplemented with 0.3% BSA, 0.01% FBS and 1 ug/ml TPCK-treated trypsin was added and cells were incubated for 72 h at 37° C. The presence of virus in cell culture supernatants was assessed by HA assays using 0.5% chicken red blood cells.
  • Results
  • Bone marrow and blood samples were collected from six survivors of the H5N1 bird flu outbreak that had taken place in Turkey in January 2006, approximately four months after the outbreak. For all six survivors the initial diagnosis of bird flu was made following by physical examination, clinical laboratory testing, and molecular diagnostic determination, sanctioned by the Turkish Ministry of Health. Four of these survivors were additionally confirmed by the World Health Organization (WHO). Serum samples were analyzed to confirm the presence of antibodies to H5 hemagglutinin (A/Vietnam/1203/2004) using the serology protocol described above. As shown in FIG. 8, the blood samples of all six patients (designated SLB H1-H6, respectively) demonstrated the presence of antibodies to the H5 antigen. Following this confirmation, RNA was extracted from the bone marrow samples of these individuals, and bone marrow mRNA was purified and reverse transcribed using the protocols described above. The antibody heavy and light chain repertoires were then amplified from the bone marrow cDNA as described above, and individual antibody heavy and light chain phage libraries were cloned separately for each survivor, using the above-described three-nucleotide bar coding to distinguish the individual libraries.
  • Using this vector with its coding system, we successfully cloned repertoires from the bone marrow of five of the six survivors in both single chain (scFv) and Fab phagemid formats. Each collection from an individual survivor has a diversity of greater than 1.0×108 members. Furthermore, we created additional bar coded libraries comprised of mixed survivor light and heavy chains with a final diversity of 1.1×109. Collectively the 5 donor-specific collections and the pooled libraries from all donors have a total diversity of 1.0×109 as a scFv collection and 4.2×109 as a Fab-displayed collection (Table 4).
  • Table 2 shows the light chain and full library total transformants in both scFv and Fab formats. Total diversity represented by all libraries is 5.6×109
  • TABLE 2
    Light Chains Completed Libraries
    Kappa Lambda Kappa Lambda
    scFV
    H5-1 3.00E+06 4.00E+06 1.50E+08 1.20E+08
    H5-2 3.00E+06 3.00E+06 4.00E+07 1.60E+07
    H5-3 8.20E+05 1.70E+06 5.30E+07 1.50E+08
    H5-5 7.00E+06 5.60E+06 6.50E+08 5.60E+07
    H5-6 1.50E+06 5.00E+06 ND 1.00E+07
    H5Pool Not Determined 1.80E+08 5.70E+08
    Totals 1.10E+09 9.20E+08
    scFv Total 2.00E+09
    Fab
    H5-1 1.50E+06 2.90E+06 2.90E+08 4.60E+08
    H5-2 3.10E+06 9.40E+05 4.40E+08 4.30E+08
    H5-3 2.80E+06 2.30E+06 3.90E+08 3.90E+08
    H5-5 7.00E+06 5.60E+06 7.20E+08 1.70E+08
    H5-6 1.50E+06 5.00E+06 ND ND
    H5Pool 1.90E+07 2.60E+08
    Totals 2.10E+09 1.45E+09
    Fab Total 3.60E+09
  • Bone marrow and blood samples were also collected from twelve local donors who were treated for flu symptoms in the year of 2006. Serology was performed as described above to confirm the presence of antibodies to H1, H3 and H5 hemagglutinin, respectively. As shown in FIG. 8, all serum samples tested positive for antibodies to H1 and/or H3 hemagglutinins, where the dominance of a certain subtype depended on the influenza A virus subtype to which the particular donor was exposed most throughout his or her lifetime. Interestingly, there were donors whose serum contained a significant level of antibodies of H5 hemagglutinin as well (donors SLB1 and SLB5 in FIG. 9). Following this confirmation, RNA was extracted from the bone marrow samples of the donors, and bone marrow mRNA was purified and reverse transcribed using the protocols described above. The antibody heavy and light chain repertoires were then amplified from the bone marrow cDNA as described above, and individual antibody heavy and light chain phage libraries were cloned separately for each donor, using the above-described three-nucleotide bar coding to distinguish the individual libraries.
  • Selecting Binding Antibodies
  • The H5N1 survivor libraries summarized in Table 2, were panned against inactivated virus containing the Vietnam/1203/04 virus HA and NA proteins or recombinant purified hemagglutinin (Barbas, C. et al. (2001) Phage Display, A Laboratory Manual (Cold Spring Harbor Laboratory Press)). Following three to four rounds of phage panning, individual clones from enriched phage pools were analyzed by ELISA against H5N1 virus or purified hemagglutinin and the positive clones were sequenced to determine their heavy and light chain sequences and to read their survivor bar code (D. W. Coomber, Methods Mol Biol 178, 133 (2002)). From this, we isolated specific H5 hemagglutinin binding clones from all five of the individual survivor libraries. In total, more than 300 hundred different antiviral antibodies were recovered, of which 146 specifically bind the H5 hemagglutinin protein.
  • General Features of the Selected Clones
  • Overall, the individual donors use different germ lines for both heavy and light chains, demonstrating that individual patients have found different solutions to the same potentially lethal immunological challenge. The major features of combinatorial antibody libraries that can be used both to give confidence as to the quality of the obtained repertoire and to provide information as to the chemistry of antibody binding and/or neutralization are seen in these clones. These clones contain all the hallmarks of the previously described repeated clones (“jackpot solution”) to antigen binding that is found in the natural progression of affinity maturation, as well as in selected synthetic antibody libraries (Lerner, R. A. Angew Chem Int Ed Engl 45, 8106 (Dec. 11, 2006); A. Rajpal et al., Proc Natl Acad Sci USA 102, 8466 (Jun. 14, 2005)). The presence of “jackpots” in these large collections validates the screening procedure because, unless the phage were selected on the basis of activity, the chance of obtaining the same clone multiple times is highly improbable. Moreover, when one analyzes the heavy chain differences within groups, it was observed that many of the amino acid substitutions were chemically and structurally conservative (Table 4). As with repeated clones, the appearance of multiple amino acid substitutions that are chemically reasonable is unlikely to be a random event.
  • Binding Specificity of Recovered Antibodies
  • Six clones were selected from three survivors that recognized two different epitopes for conversion into full IgG1 proteins. The binding of four of these antibodies was mapped to the HA1 subunit of the hemagglutinin protein, while the remaining two did not. As the HA1 subunit has tremendous relevance in infection we further analyzed the activities of those four antibodies as described below.
  • One goal of these studies was to recover those rare antibodies that broadly neutralize divergent viral strains. There was a suggestion that some of our antibodies might be broadly reactive because the serum from the donors had high titer antibodies against a divergent subfamily of H5N1 viruses that extended beyond the virus with which they were infected. To determine the degree of cross reactivity at the level of individual antibodies, we first analyzed binding of our clones to different influenza hemagglutinin antigens (FIG. 11). Not surprisingly, these antibodies recognize hemagglutinin from the corresponding infecting Turkey/65596/06 strain, and in addition recognize the heterologous hemagglutinin from the Vietnam/1203/04 strain used for selection. Moreover, they recognize the antigenically divergent Indonesian/5/05 H5 hemagglutinin. Furthermore, we found that four prototype antibodies bound hemagglutinin from the closely related subtype H1N1 contemporary reference strain New Caledonia/20/99. Notably, the three neutralizing antibodies belonging to survivor 5 also bound hemagglutinin from the H1N1 South Carolina/1/18 isolate that emerged during the 1918 Spanish Flu pandemic. Conversely, none of these four antibodies bound hemagglutinin from the contemporary H3N2 Wisconsin/67/05 reference strain, indicating that even though the antibodies display broad spectrum binding amongst and between Influenza subtypes, the reactivity did not extend to all influenza subtypes. FIG. 11 shows the cross-reactivity of H5N1 antibodies from two survivors with hemagglutinins from H1N1 viruses. (A) Bars are H5N1 Vietnam 1203/04(dark grey), H5N1 Turkey/65596/06 (white), H5N1 Indonesia/5/05 (diagonal stripes), H1N1 New Caledonia/20/99 (vertical stripes), H1N1 South Carolina/1/18 (crosshatch stripes), and H3N2 Wisconsin/67/05 (light gray).
  • Neutralization Studies
  • Initially the antibodies were assayed for their ability to neutralize an H5 HA (Vietnam/1203/04) containing influenza virus. One antibody derived from survivor 2 and 3 from survivor 5 that recognized a common epitope (epitope “A”) were all neutralizing whereas the two antibodies derived from survivor 1 that recognized a second epitope (epitope “13”) were not.
  • Based on the striking sequence similarity of clones separately isolated from survivor 5 against either H5N1 or H1N1 hemagglutinin, we predicted that their cross reactivity would extend beyond simple binding and they would also have the highly unusual property of neutralizing both H5N1 and H1N1 virus. To test the cross neutralizing activity of the IgGs, we tested representative antibodies from the H5N1 screen in a neutralization assay to see if they would also neutralize H1N1 or H3N2 virus (Table 3). We studied the H1 bearing virus A/New Cal/20/99 and the H3 bearing virus A/Hong Kong/68. A collection of viruses bearing H5 subtype hemagglutinin was also tested (A/Vn/1203/04; A/Indo/5/05; A/Turkey/65596/06; A/Egypt/06). The antibodies showed no activity against H3 subtype influenza. However, three of the monoclonal antibodies (1-3) that neutralized H5 containing viruses also strongly neutralized all viruses bearing HA from subtypes H1 (Table 5).
  • TABLE 3
    Virus*
    H5 H1 H3
    A/Vietnam A/Vietnam A/Indonesia A/Turkey A/Egypt A/New A/Hong
    1203/04 ‡ 1203/04 ‡ 5/05 65596/06 14725/06 Cal 20/99 Kong/68
    Ab 1† 32-64  7-28 28 28 3.5-7   28 >1000
    Ab 2† 188 162-650 80 325 20-40 162-325 >1000
    Ab 3† 175 53 47 94 12-23 23-47 >1000
    Ab 4†  5-19 1.6-6.6 >1000 Not done Not done >1000 >1000
  • Immunochemical Basis of Neutralization
  • One advantage of antibody libraries is that when one obtains large numbers of antibodies, they can be grouped as to their relatedness. Thus, when a function for a given antibody in the collection is observed one can predict that other members of the group to which it belongs will have similar activity.
  • Table 4 shows example sequences displaying the immunochemical basis of neutralization discovered from Survivor 5 libraries following H5N1 Vietnam panning. The 61 unique heavy chain sequences aligned with their germline variable regions from the 114 unique heavy and light chain combinations. Requisite mutations are shown in bolded, underlined text (column 5—PI to GM and A to T; column 6—KS to EL or EM or XL) and predominant mutations are shown in italicized, underlined text (column 2—A to T; column 3—IS to VT; column 5—G to A; column 8—K to Q or R). Heavy chains sequences also discovered in H1N1 New Caledonia panning are highlighted in gray. Antibody regions and Kabat numbering ranges are listed at the top of each sequence column. The heavy chain/light chain pairing is indicated in the first column as follows: *—paired with 2 unique light chains, †—paired with 3 unique light chains, ‡—paired with 4 unique light chains, §—paired with 5 unique light chains, and ¶—paired with 12 unique light chains.
  • TABLE 4
    Figure US20100291066A1-20101118-C00001
    Figure US20100291066A1-20101118-C00002
  • Members of the group that contained the neutralizing antibody collection against epitope “A” from survivor 5, are shown in Table 4. The group is comprised of 61 unique members that most closely resemble the VH1e germ line heavy chain. Some heavy chains are paired with more than one light chain. In total these heavy chains have 114 unique pairings to both kappa and lambda light chains. Comparing these heavy chains to the highly related VH1e germline, we observe three types of point substitutions. Some changes appear to be required, others are dominant, and some residues have only been changed sporadically. The changes that are required occur in every clone in the group within CDR2 at position 52A (Pro>Gly), 53 (Ile>Met), and 57 (Ala>Thr), as well as in the framework 3 region at position 73 (Lys>Glu) and 74 (Ser>Leu or Met), all of which vary from the germline side chain chemistries, suggesting that these mutations are critical to antigen binding and neutralization. The second set of mutations is dominant and found in most clones. The first, in framework 1 at position 24 (Ala>Thr), represents a significant chemical change. The next three are conservative changes in CDR1 at positions 34(Ile>Val) and 35 (Ser>Thr) and also in CDR2 at position 50 (Gly>Ala). All four of these dominant substitutions, however, are dispensable, suggesting that, while beneficial, they are not essential. The sporadic changes found throughout framework regions 1, 3, and 4, as well as CDR3, are all conservative and likely represent minor optimization events.
  • FIG. 12 shows the positions of the required mutations in the structure of the antibody superimposed on the crystal structure of a highly related anti-HIV Fab called 47e (lrzi.pdb) (Huang, C. C. et al. (2004)Proc. Nat. Acad. Sci. 101, 2706-2711). FIG. 12 shows the positions of H5 hemagglutinin binding Group 1 required and dominant mutations on the crystal structure of Fab 47e. The required mutations are shown as G52 (52A (Pro>Gly)), M53 (Ile>Met), T57 (Ala>Thr), E73 (Lys>Glu) and LM74 (Ser>Leu or Met). The dominant mutations are shown as T24 (Ala>Thr), V34 (Ile>Val), T35 (Ser>Thr), and A50 (Gly>Ala). The required and dominant Group 1 heavy chain sequences identified in H5 Vietnam/1203/2004H A biopanning are superimposed on the crystal structure of the highly related anti-HIV Fab 47e. Mutations are shown in both backbone (top) and space-filling (bottom) models. A tight cluster is formed by four of the required mutations in and adjacent to CDR2. The required mutations 52A (Pro>Gly), 53 (Ile>Met), 73 (Lys>Glu) and 74 (Ser>Leu or Met) form a remarkably tight cluster on the exposed surface of the heavy chain variable domain where they form a ridge that prominently protrudes from the protein surface (FIG. 12). The remaining required mutation 57 (Ala>Thr) is partially buried at the base of the CDR2 loop. The surface exposed changes in CDR 2 and framework 3 are likely to have a direct role in antigen binding while the less exposed required mutation and the non-essential dominant mutations may have indirect effects through stabilizing and/or positioning the CDR2 loop.
  • The antibodies from survivor 2 are comprised of 2 unique heavy chains that most closely resemble the VH4-4b germ line heavy chain (Table 5). The first heavy chain has been found paired with 5 unique lambda light chains, four of which are from the infrequently used lambda 6 light chain family and the other is paired with a single kappa light chain. Antibody 4 whose neutralization profile was more restricted came from this group.
  • TABLE 5
    Group FR1 CDR1 FR2 CDR2
    2 (1-29) (30-35) (36-46) (47-58)
    Heavy QVQLQESGPGLVKPSETLSLTCTVSGYSF DSGYYWG   WLRQPPGKGLE WIGSIYHSRNTY
    chain
    Lambda FMLTQPHSVSESPGKTVTISCTGSGGN IARNYVQWY QQRPGSAPV TVILEDDKRP
    light FMLTQPHSVSESPGKTVTISCTGSSGS IASNYVQWY QQRPGSAPT TVIYEDYQRP
    chains SVLTQPPSASGTPGQRVTISCSGSSSN IGSNTVNWY KQLPGTAPR LLIYSNDQRP
    SVLTQPPSASGTPGQRVTLSCSGSSSN IGGNSVNWY QHVPGTAPK LLMHSDDQRP
    PELTQPHSVSESPGKTVTISCTGSGGR IATNHVQWY QQRPGSAPT IVIYENNQRP
    PELTQPPSASGTPGQRVTISCSGSSSN IGSNTVNWY QQLPGTAPK LLIYSNNQRP
    Kappa DIQMTQSPSSLSAFVGDRVTITCQASQDI SNYLNWY QQKPGKAPK LLIYDATNLE
    light
    chain
    Group FR3 CDR3 FR4
    2 (59-92) (93-101) (102-113)
    Heavy YNPSLKSRVTISVDTSKNQFSLQLSSVTAADTAVYYC ARGTWYSSNLRYWFD PWGKGTLVRVSS
    chain
    Lambda SGIPDRFSGSIDRSSNSASLTISGLRTEDEALYYC QSYDDSDLV VFGGGTKLT
    light SGVPDRFSGSIDSSSNSASLTISGLKTEDEADYYC QSYDDSDHL IFGGGTKLTVL
    chains SGVPDRFSGSKSGTSASLAISGLQSEDEANYYC AAWDDSLSGW VFGGGTKLTVL
    SGVPDRFSGSKSGTSXSLAISGLQSEDEADYYC AXWDDSLNAW VFGGXTKVTVL
    SGVPNRFSGSIDDSSNSASLTISALRTEDEADYYC QSADATNV FFGGGTKVTVL
    SGVPDRFSGSKSGTSASLAISGLQSEDEADYYC AAWDDSLNGW VFGGXTKLTVL
    Kappa TGVPSRFSGSGSGTDFTFTISSLQPEDIATYYC QQYDNLPL TFGGGTKVDIKR
    light
    chain
  • The probability that a given mutation is important to the activity of an antibody increases as a function of the number of times it was independently selected. To determine if the required mutations were selected during somatic mutation from independent clones or were from the progeny of a single clone that further mutated during subsequent replications, the codon usage of the dominant mutations were analyzed (Table 6A-6B). The data reveal that although different codons were used they resulted in the same amino acid changes, demonstrating that these mutations arose independently in different clones and were, thus, selected multiple times. This convergent outcome for independently selected events is strong evidence that these dominant mutations play a critical role in the binding to the virus and/or its neutralization.
  • As illustrated in Tables 6A-6B, codon usage of individual clones shows independent origin of selected H5 HA binding clones. DNA alignment and encoded amino acids for 6 representative Group 1 antibodies against VH1-e germline. The use of different codons for the same amino acids demonstrates that each unique sequence is of a distinct origin. Table 6A corresponds to CDR2 and Table 6B corresponds to Framework 3. Germ line codons are shown as bolded codons. A change from a germ line codon to the same amino acid is shown as a plain text codon. A first change from a germ line amino acid is shown as a bolded, underlined codon. A second change from a germ line amino acid is shown as an italicized, underlined codon. A third change from a germ line amino acid is shown as an underlined,
  • TABLE 6A
    Clone Kabat CDR2 
    Number Sequence 47 48 49 50 51 52 52A 52B 52C 53 54 55 56 57 58
    VH1-e  TGG ATG GGA AGG ATC ATC CCT ATC CTT  GGT ATA GCA AAC TAC GCA
    germline W M G R I I P I L G I A N Y A
    27 TGG ATG GGC GCG ATC ATC GGT ATG TTT GGT ACA ACA AAC TAC GCA
    W M G A I I G M F G T T N Y A
    30 TGG ATG GGA GGG ATC ATC GGT ATG TTT GGA ACA ACC AAC TAT GCA
    W M G G I I G M F G T T N Y A
    33 TGG ATG GGA GCG ATC ATC GGT ATG TTT GGT ACA ACA AAC TAC GCA
    W M G A I I G M F G T T N Y A
    41 TGG ATG GGC GCG ATC ATC GGT ATG TTT GGT ACA ACA AAC TAC GCA
    W M G A I I G M F G T T N Y A
    50 TGG ATG GGA GGG ATC ATC GGT ATG TTT GGT ACA ACG AAC TAT GCA
    W M G G I I G M F G T T N Y A
    17 TGG ATG GGA GGG ATC ATC GGT ATG TTT GGT ACA ACA AAC TAC GCA
    W M G G I I G M F G T T N Y A
    # Codons 1 1 2 2 1 1 1 1 1 2 1 3 1 2 1
    used
  • TABLE 6B
    Clone Kabat Framework  3 
    Number Sequence 67 68 69 70 71 72 73 74 75 76 77 78
    VH1-e  GTC ACG ATT ACC GCG GAC AAA TCC ACG AGC ACA GCC
    germline V T I T A D K S T S T A
    27 GTC ACG CTT ACC GCG GAC GAA TTA ACG TCC ACA GCC
    V T L T A D E L T S T A
    30 CTC ACA ATC ACC GCG GAC GAG ATG ACG TCC ACA GCC
    L T I T A D E M T S T A
    33 GTC ACA ATC ACC GCG GAC GAA TTA ACG TCC ACA GCC
    V T I T A D E L T S T A
    41 GTC ACG CTT ACC GCG GAC GAA TTA ACG TCC ACA GCC
    V T L T A D E L T S T A
    50 GTC ACG ATT ACC GCG GAC GAG ATG ACG TCC ACA GCC
    V T I T A D E M T S T A
    17 GTC ACG ATT ACC GCG GAC GAA TTA ACG TCC ACA GCC
    V T I T A D E L T S T A
    # Codons
    2 2 3 1 1 1 2 2 1 1 1 1
    used
  • Example 2 Constructing Donor-Specific Antibody Library for Patients Infected with HIV
  • Bone Marrow Protocol and Sera Preparation
  • Blood is obtained by standard venopuncture, allowed to clot, and processed to recover serum. The serum is stored at −20° C. for 3-4 days until they are shipped on dry ice. Donors are anaesthetized with an injection of a local anesthetic and 5 ml of bone marrow is removed from the pelvic bone of each patient donor. Next the 5 ml of bone marrow is placed into a sterile 50-ml tube containing 45 ml RNAlater (Ambion). The mixture is gently inverted approximately 8-20 times, until there are no visible clumps and the marrow and RNAlater are mixed well. Next the specimen is refrigerated the between 2-10° C. overnight. Following the overnight refrigeration, the specimens are stored at −20° C. for 3-4 days until they are shipped on dry ice. Upon receipt the RNAlater/marrow and sera containing tubes are stored at −80° C. until processed. Candidate patient should be tested HIV positive prior to be selected as donors.
  • Bone marrow extraction and mRNA purification, reverse transcription, PCR, antibody light and heavy chain construction, phagemid panning and amplification, ELISA and sequencing are performed essentially as described in Example 1.
  • Although in the foregoing description the invention is illustrated with reference to certain embodiments, it is not so limited. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims.
  • All references cited throughout the specification are hereby expressly incorporated by reference.

Claims (67)

1. A vector collection comprising a repertoire of nucleic acid molecules encoding antibody light or heavy chains or fragments thereof, derived from a human patient donor who has suffered from, or is suffering from, a disease evoking antibody production to a target antigen, wherein said collection is identified with a unique barcode.
2. The vector collection of claim 1 comprising a repertoire of nucleic acid molecules encoding antibody light chains or fragments thereof.
3. The vector collection of claim 2 wherein the antibody light chains are λ chains.
4. The vector collection of claim 2 wherein the antibody light chains are κ chains.
5. The vector collection of claim 1 comprising a repertoire of nucleic acid molecules encoding antibody heavy chains or fragments thereof.
6. The vector collection of claim 1 wherein the barcode is a nucleotide sequence linked to or incorporated in the vectors present in the collection, and/or linked to or incorporated in the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of said nucleic acid molecules.
7. The vector collection of claim 6 wherein said barcode is a contiguous non-coding nucleotide sequence of one to about 24 nucleotides.
8. The vector collection of claim 7 wherein said nucleotide sequence is linked to the 3′ or 5′ non-coding region of said nucleic acid molecules.
9. The vector collection of claim 6 wherein said nucleotide sequence is a coding sequence of one or more silent mutations incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof.
10. The vector collection of claim 6 wherein said nucleotide sequence in non-contiguous.
11. The vector collection of claim 10 wherein at least part of said non-contiguous nucleotide sequence is linked to or incorporated in the vectors present in the collection.
12. The vector collection of claim 10 wherein at least part of said non-contiguous sequence is incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of said nucleic acid molecules.
13. The vector collection of claim 1 wherein the barcode is a peptide or polypeptide sequence.
14. The vector collection of claim 1 wherein the vectors are phagemid vectors.
15. The vector collection of claim 15 wherein the phagemid vectors contain a bacteriophage gene III and a stop codon between the nucleic acid molecules encoding antibody light or heavy chains or fragments thereof and the bacteriophage III gene.
16. The vector collection of claim 15 wherein the barcode is a non-coding contiguous nucleotide sequence inserted in the untranslated region following said stop codon.
17. Host cells comprising the vector collection of claim 1.
18. The host cells of claim 16 which are E. coli host cells.
19. A donor-specific antibody library comprising library members expressing a collection of antibodies or antibody fragments to a target antigen wherein said antibodies or antibody fragments are derived from a human donor who has suffered from, or is suffering from, a disease evoking antibody production to said target antigen, wherein said antibody library is identified with at least one unique barcode.
20. The donor-specific antibody library of claim 19 wherein said antibody heavy and light chains are separately identified each with a barcode unique to the human donor from whom it derived.
21. The donor-specific antibody library of claim 19 which is identified with one unique barcode.
22. The donor-specific antibody library of claim 19 wherein said antibodies or antibody fragments are composed of antibody heavy and light chains or fragments thereof encoded by nucleic acid molecules present in a vector.
23. The donor-specific antibody library of claim 22 wherein the barcode is a nucleotide sequence linked to or incorporated in the vectors present in the library, and/or linked to or incorporated in the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of said nucleic acid molecules.
24. The donor-specific antibody library of claim 23 wherein said barcode is a contiguous non-coding nucleotide sequence of one to about 24 nucleotides.
25. The donor-specific antibody library of claim 24 wherein said nucleotide sequence is linked to the 3′ or 5′ non-coding region of said nucleic acid molecules.
26. The donor-specific antibody library of claim 23 wherein said nucleotide sequence is a coding sequence of one or more silent mutations incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof.
27. The donor-specific antibody library of claim 23 wherein said nucleotide sequence in non-contiguous.
28. The donor-specific antibody library of claim 27 wherein at least part of said non-contiguous sequence is linked to or incorporated in the vectors present in the library.
29. The donor-specific antibody library of claim 27 wherein at least part of said non-contiguous sequence is incorporated into the nucleic acid molecules encoding the antibody light or heavy chains or fragments thereof such that it does not interfere with the expression of said nucleic acid molecules.
30. The donor-specific antibody library of claim 19 wherein the barcode is a peptide or polypeptide sequence.
31. The donor-specific antibody library of claim 22 wherein the vectors are phagemid vectors.
32. The donor-specific antibody library of claim 31 wherein the phagemid vectors contain a bacteriophage gene III and a stop codon between the nucleic acid molecules encoding antibody light or heavy chains or fragments thereof and the bacteriophage III gene.
33. The donor-specific antibody library of claim 19 expressing a collection of antibody heavy chains or fragments thereof.
34. The donor specific antibody library of claim 33 comprising more than one antibody heavy chain family.
35. The donor-specific antibody library of claim 19 wherein the medical history of said human donor shows that the patient has suffered from, or is suffering from said disease.
36. The donor-specific antibody library of claim 35 wherein it is independently confirmed that said human donor suffered from, or is suffering from said disease.
37. The donor-specific antibody library of claim 19, which is substantially devoid of antibodies and antibody fragments specifically binding antigens different from said target antigen.
38. The donor-specific antibody library of claim 19 wherein said target antigen is an influenza A virus.
39. The donor-specific antibody library of claim 38 wherein said target antigen is an isolate of influenza A virus H1, H2 or H3 subtype.
40. The donor-specific antibody library of claim 38 wherein the target antigen is selected from the group comprising H5, H7 and H9 influenza A virus subtypes.
41. The donor-specific antibody library of claim 38 expressing at least one antibody or antibody fragment specifically binding to more than one influenza A virus subtype.
42. The donor-specific antibody library of claim 39 expressing at least one antibody or antibody fragment binding to and neutralizing the H5N1 subtype of influenza virus A.
43. The donor-specific antibody library of claim 19 wherein the human donor has suffered from, or is suffering from a disease selected from the group consisting of the diseases listed in Table 1.
44. The donor-specific antibody library of claim 43 expressing at least one antibody or antibody fragment binding to an antigen associated with said target disease.
45. The donor-specific antibody library of claim 43 expressing at least one antibody or antibody fragment binding to and neutralizing an antigen associated with said target disease.
46. The donor-specific antibody library of claim 19 expressing a least one neutralizing antibody.
47. The donor-specific antibody library of Claim 19, wherein said donor-specific antibody library is a phage library.
48. The donor-specific antibody library of Claim 47, wherein said collection contains sequences encoding more than 106 different members of antibodies or antibody fragments.
49. The donor-specific antibody library of Claim 47, wherein said collection contains sequences encoding more than 109 different members of antibodies or antibody fragments.
50. The donor-specific antibody library of claim 19, which is selected from the group consisting of a spore-display library, a ribosome display library, a mRNA display library, a microbial cell display library, a yeast display library, and a mammalian display library.
51. The donor-specific antibody library of Claim 50, wherein said nucleic acid is reverse-transcribed from mRNA extracted from lymphocytes of said human donor.
52. The donor-specific antibody library of Claim 51, wherein said lymphocytes are derived from bone marrow, blood, spleen, or lymph node.
53. The donor-specific antibody library of Claim 52, wherein a serological profile of said human donor is generated prior to extraction of said mRNA.
54. The donor-specific antibody library of Claim 53, wherein a medical history of said human donor is examined prior to or following extraction of said mRNA.
55. A method of making a donor-specific library expressing a collection of antibodies or antibody fragments to a target antigen, comprising the steps of:
a) obtaining mRNA from lymphocytes of a human patient donor who has suffered from, or who is suffering from a disease evoking antibody production to said target antigen;
b) generating a collection of nucleic acids comprising sequences encoding an immunoglobulin repertoire of said patient by reverse transcription of said obtained mRNA: and
c) identifying said donor-specific library with an unique barcode labeling said nucleic acids.
56. The method of Claim 55, further comprising steps of generating a serological profile of said patient and/or examining medical history of said patient prior or subsequent to step a).
57. The method of Claim 56, further comprises steps of:
d) inserting said nucleic acids into expression vector;
e) expressing said immunoglobulin repertoire; and
f) displaying said immunoglobulin repertoire in a display system.
58. The method of Claim 57, wherein the display system is a phagemid.
59. The method of claim 58 further comprising the step of selecting members of the library based their ability to neutralize or activate the target antigen.
60. The method of claim 59 that yields at least one neutralizing antibody.
61. The method of claim 59 that yields more than one neutralizing antibody.
62. The method of claim 61 further comprising the step of creating one or more sub-libraries comprising library members that were found to neutralize or activate the target antigen.
63. The method of claim 59 comprising the step of sequencing at least one library member identified.
64. A method of treating or preventing a disease associated with a target antigen neutralized or activated by an antibody selected by the method of claim 59, comprising administering to said human patient in need an effective amount of the antibody selected.
65. The method of claim 64 wherein the antibody is a neutralizing antibody.
66. The method of claim 65 wherein the disease is an influenza virus A infection.
67. The method of claim 64 wherein the disease is selected from the group consisting of the diseases listed in Table 1.
US12/676,737 2007-09-11 2008-09-11 Donor specific antibody libraries Abandoned US20100291066A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/676,737 US20100291066A1 (en) 2007-09-11 2008-09-11 Donor specific antibody libraries

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US11853795 2007-09-11
US11/853,795 US8148085B2 (en) 2006-05-15 2007-09-11 Donor specific antibody libraries
PCT/US2008/075998 WO2009036157A1 (en) 2007-09-11 2008-09-11 Donor specific antibody libraries
US12/676,737 US20100291066A1 (en) 2007-09-11 2008-09-11 Donor specific antibody libraries

Publications (1)

Publication Number Publication Date
US20100291066A1 true US20100291066A1 (en) 2010-11-18

Family

ID=40220152

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/853,795 Active US8148085B2 (en) 2006-05-15 2007-09-11 Donor specific antibody libraries
US12/676,737 Abandoned US20100291066A1 (en) 2007-09-11 2008-09-11 Donor specific antibody libraries
US13/422,313 Abandoned US20120264647A1 (en) 2006-05-15 2012-03-16 Donor specific antibody libraries

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/853,795 Active US8148085B2 (en) 2006-05-15 2007-09-11 Donor specific antibody libraries

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/422,313 Abandoned US20120264647A1 (en) 2006-05-15 2012-03-16 Donor specific antibody libraries

Country Status (6)

Country Link
US (3) US8148085B2 (en)
EP (1) EP2190477A1 (en)
JP (1) JP2010538618A (en)
CN (1) CN101854950A (en)
AU (1) AU2008298939A1 (en)
WO (1) WO2009036157A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015004633A1 (en) 2013-07-12 2015-01-15 Neotope Biosciences Limited Antibodies that recognize islet-amyloid polypeptide (iapp)
WO2015004632A1 (en) 2013-07-12 2015-01-15 Neotope Biosciences Limited Antibodies that recognize iapp
WO2015155694A1 (en) 2014-04-08 2015-10-15 Prothena Biosciences Limited Blood-brain barrier shuttles containing antibodies recognizing alpha-synuclein
EP3498296A1 (en) 2012-10-08 2019-06-19 Prothena Biosciences Limited Antibodies recognizing alpha-synuclein

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7584439B2 (en) * 2006-12-07 2009-09-01 Agere Systems Inc. Cell modeling for integrated circuit design with characterization of upstream driver strength
JP2012521786A (en) 2009-03-30 2012-09-20 モウント シナイ スクール オフ メディシネ Influenza virus vaccine and use thereof
WO2010130636A1 (en) 2009-05-11 2010-11-18 Crucell Holland B.V. Human binding molecules capable of neutralizing influenza virus h3n2 and uses thereof
WO2010132604A2 (en) * 2009-05-13 2010-11-18 Sea Lane Biotechnologies, Llc Neutralizing molecules to influenza viruses
EP2435475B1 (en) 2009-05-26 2018-10-17 Icahn School of Medicine at Mount Sinai Monoclonal antibodies against influenza virus generated by cyclical administration and uses thereof
US20110077966A1 (en) * 2009-09-28 2011-03-31 Augusta E.N.T., P.C. Allergy electronic medical record system and method
EP2536425B1 (en) 2010-02-18 2019-06-19 Icahn School of Medicine at Mount Sinai Vaccines for use in the prophylaxis and treatment of influenza virus disease
US9708373B2 (en) 2010-03-30 2017-07-18 Icahn School Of Medicine At Mount Sinai Influenza virus vaccine and uses thereof
WO2012058072A1 (en) * 2010-10-27 2012-05-03 Harrisvaccines, Inc Method of rapidly producing vaccines for animals
KR102112205B1 (en) * 2011-04-28 2020-06-12 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Identification of polynucleotides associated with a sample
US20230287392A1 (en) * 2011-04-28 2023-09-14 The Board Of Trustees Of The Leland Stanford Junior University Identification of Polynucleotides Associated with a Sample
BR112014006694A2 (en) 2011-09-20 2020-11-17 Mount Sinai School Of Medicine influenza vaccines and uses of these
CA2850720C (en) 2011-09-30 2016-03-29 Celltrion Inc. Binding molecule having influenza a virus-neutralizing activity produced from human b cell
US9977861B2 (en) 2012-07-18 2018-05-22 Illumina Cambridge Limited Methods and systems for determining haplotypes and phasing of haplotypes
CN105263516A (en) 2012-12-18 2016-01-20 西奈山伊坎医学院 Influenza virus vaccines and uses thereof
WO2014159960A1 (en) 2013-03-14 2014-10-02 Icahn School Of Medicine At Mount Sinai Antibodies against influenza virus hemagglutinin and uses thereof
AR096601A1 (en) 2013-06-21 2016-01-20 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
US9562101B2 (en) * 2013-06-21 2017-02-07 Novartis Ag Lectin-like oxidized LDL receptor 1 antibodies and methods of use
US20150225770A1 (en) * 2014-02-12 2015-08-13 U.S. Army Research Laboratory Attn: Rdrl-Loc-I Phenotype-neutral barcodes for digital analysis
KR101740030B1 (en) 2014-03-07 2017-05-29 사회복지법인 삼성생명공익재단 Single-chain Fv Antibody Library, Method for Preparing Thereof and Screening Method of Single-chain Fv Antibody by Using the Same
DK3126384T3 (en) * 2014-04-01 2021-01-18 Adimab Llc MULTISPECIFIC ANTIBODY ANALOGS INCLUDING A COMMON LIGHT CHAIN, AND METHODS FOR THEIR PREPARATION AND USE
US20180008702A1 (en) * 2014-12-05 2018-01-11 Celltrion Inc. Adjuvant composition containing at least one influenza virus neutralizing and binding molecule and vaccine composition containing same
CA2974699A1 (en) 2015-01-23 2016-07-28 Icahn School Of Medicine At Mount Sinai Influenza virus vaccination regimens
EP3514181A1 (en) * 2015-02-24 2019-07-24 Academia Sinica A phage-displayed single-chain variable fragment library and antibodies selected therefrom
AU2016315941B2 (en) * 2015-09-01 2022-09-08 The Regents Of The University Of California Modular polypeptide libraries and methods of making and using same
CA3023143A1 (en) 2016-06-15 2017-12-21 Icahn School Of Medicine At Mount Sinai Influenza virus hemagglutinin proteins and uses thereof
ES2926548T3 (en) * 2016-10-31 2022-10-26 Univ Zuerich Collection of tags and methods for protein detection, preferably by mass spectrometry
US11254733B2 (en) 2017-04-07 2022-02-22 Icahn School Of Medicine At Mount Sinai Anti-influenza B virus neuraminidase antibodies and uses thereof
US20220283161A1 (en) * 2019-07-25 2022-09-08 The Johns Hopkins University Methods and compositions for hepatitis c virus (hcv)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6607878B2 (en) * 1997-10-06 2003-08-19 Stratagene Collections of uniquely tagged molecules

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA836080B (en) 1982-08-23 1984-04-25 Scripps Clinic Res Broad spectrum influenza antisera
US5667988A (en) 1992-01-27 1997-09-16 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
US6337070B1 (en) 1993-04-29 2002-01-08 Takara Shuzo Co., Ltd. Polypeptides for use in generating anti-human influenza virus antibodies
EP0699750A1 (en) * 1994-06-07 1996-03-06 Gesellschaft für biotechnologische Forschung mbH (GBF) A collection of phagemids, a collection of Escherichia coli cells carrying the phagemids, a collection of phagemid particles produced from said collection and phagemid particles obtained according to the process
US6169175B1 (en) * 1997-08-06 2001-01-02 Centers For Disease Control And Prevention Preparation and use of recombinant influenza A virus M2 construct vaccines
HUP0200400A3 (en) 1999-03-23 2006-06-28 Biovation Ltd Aberdeen Protein isolation and analysis
FR2817869B1 (en) 2000-12-07 2005-01-14 Technopharm MONOCLONAL HUMAN ANTIBODY DIRECTED AGAINST INFLUENZA VIRUS OR A FRAGMENT THEREOF
AU2002249854B2 (en) 2000-12-18 2007-09-20 Dyax Corp. Focused libraries of genetic packages
WO2003052101A1 (en) 2001-12-14 2003-06-26 Rosetta Inpharmatics, Inc. Sample tracking using molecular barcodes
TW200407161A (en) 2002-03-13 2004-05-16 Gemini Science Inc Human monoclonal antibodies to influenza M2 protein and method of making and using same
AU2004286201B2 (en) 2003-09-10 2010-09-09 Altheadx, Inc. Expression profiling using microarrays
US20060073506A1 (en) 2004-09-17 2006-04-06 Affymetrix, Inc. Methods for identifying biological samples
WO2007031550A2 (en) 2005-09-15 2007-03-22 Crucell Holland B.V. Method for preparing immunoglobulin libraries
GB0522460D0 (en) 2005-11-03 2005-12-14 Prendergast Patrick T Composition and method for the treatment of avian influenza
RU2008134426A (en) 2006-01-26 2010-03-10 ЭйчИкс ДИАГНОСТИКС, ИНК. (US) MONOCLONAL ANTIBODIES CONNECTING TO AVIAN INFLUENZA VIRUS
US20080014205A1 (en) 2006-05-15 2008-01-17 Lawrence Horowitz Neutralizing Antibodies to Influenza Viruses
MX2009002174A (en) 2006-09-07 2009-03-12 Crucell Holland Bv Human binding molecules capable of neutralizing influenza virus h5n1 and uses thereof.
MX2009009912A (en) 2007-03-27 2010-01-18 Sea Lane Biotechnologies Llc Constructs and libraries comprising antibody surrogate light chain sequences.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6607878B2 (en) * 1997-10-06 2003-08-19 Stratagene Collections of uniquely tagged molecules

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3498296A1 (en) 2012-10-08 2019-06-19 Prothena Biosciences Limited Antibodies recognizing alpha-synuclein
WO2015004633A1 (en) 2013-07-12 2015-01-15 Neotope Biosciences Limited Antibodies that recognize islet-amyloid polypeptide (iapp)
WO2015004632A1 (en) 2013-07-12 2015-01-15 Neotope Biosciences Limited Antibodies that recognize iapp
WO2015155694A1 (en) 2014-04-08 2015-10-15 Prothena Biosciences Limited Blood-brain barrier shuttles containing antibodies recognizing alpha-synuclein
US10562973B2 (en) 2014-04-08 2020-02-18 Prothena Bioscience Limited Blood-brain barrier shuttles containing antibodies recognizing alpha-synuclein

Also Published As

Publication number Publication date
US8148085B2 (en) 2012-04-03
AU2008298939A1 (en) 2009-03-19
US20120264647A1 (en) 2012-10-18
WO2009036157A1 (en) 2009-03-19
CN101854950A (en) 2010-10-06
US20080152657A1 (en) 2008-06-26
EP2190477A1 (en) 2010-06-02
JP2010538618A (en) 2010-12-16

Similar Documents

Publication Publication Date Title
US20100291066A1 (en) Donor specific antibody libraries
AU2007249160B2 (en) Neutralizing antibodies to influenza viruses
US11685773B2 (en) Method for mass humanization of rabbit antibodies
US10017561B2 (en) Neutralizing molecules to influenza viruses
US9169318B2 (en) Neutralizing molecules to viral antigens
US8642513B2 (en) Method for preparing immunoglobulin libraries
KR101540822B1 (en) constructs and libraries comprising antibody surrogate light chain sequences
JP5797642B2 (en) Synthetic polypeptide libraries and methods for generating naturally diversified polypeptide variants
Singh et al. Construction of a single-chain variable-fragment antibody against the superantigen staphylococcal enterotoxin B
US20210032368A1 (en) Methods and compositions for the development of antibodies specific to epitope post-translational modification status
Leow et al. Monoclonal IgY Antibodies 13
Harvey Anchored periplasmic expression (APEx): a versatile technology for the flow cytometric selection of high affinity antibodies from Escherichia coli expressed libraries
Keklikian Construction of a Synthetic Human VL Phage Display Library and Isolation of Potential Neuropilin-1-specific VL Therapeutics from the Library

Legal Events

Date Code Title Description
AS Assignment

Owner name: SEA LANE BIOTECHNOLOGIES, LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HOROWITZ, LAWRENCE;BHATT, RAMESH;KASHYAP, ARUN K.;REEL/FRAME:024620/0841

Effective date: 20100309

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ALTUS BIOPHARMA INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SEA LANE BIOTECHNOLOGIES, LLC.;REEL/FRAME:041154/0070

Effective date: 20161214

Owner name: I2 PHARMACEUTICALS, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ALTUS BIOPHARMA INC.;REEL/FRAME:041154/0613

Effective date: 20161216