WO2022204332A1 - Compositions comprising c-met agonist antibodies and methods for use in ocular treatment - Google Patents

Compositions comprising c-met agonist antibodies and methods for use in ocular treatment Download PDF

Info

Publication number
WO2022204332A1
WO2022204332A1 PCT/US2022/021618 US2022021618W WO2022204332A1 WO 2022204332 A1 WO2022204332 A1 WO 2022204332A1 US 2022021618 W US2022021618 W US 2022021618W WO 2022204332 A1 WO2022204332 A1 WO 2022204332A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
met antibody
seq
amino acid
met
Prior art date
Application number
PCT/US2022/021618
Other languages
French (fr)
Inventor
Spencer Alford
Original Assignee
Combangio, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Combangio, Inc. filed Critical Combangio, Inc.
Publication of WO2022204332A1 publication Critical patent/WO2022204332A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen

Definitions

  • Blast and blunt injuries to the eye can cause a series of mechanical disruptions to the ocular contents including commotio retinae, traumatic cataract, disruption of the zonular attachments to the lens, angle recession, iris dialysis, and rupture of the pupillary sphincter. Treatment of these injuries has been limited to mechanical repair (when possible) of the iris, replacement of the crystalline lens with plastic lens implants, and repair of retinal detachments. There has been no treatment to repair the cellular architecture of the retina or the anterior chamber.
  • traumatic optic neuropathy and optic nerve avulsion are among the six leading types of ocular injury that required specialized ophthalmic care during Operation Iraqi Freedom (Cho and Savitsky, “Ocular Trauma Chapter 7”, in Combat Casualty Care: Lessons learned from Oef and Oif, by Brian Eastbridge and Eric Savitsky, pp. 299-342, Ft. Detrick, Md.: Borden Institute (US) Government Printing Office, 2012), incorporated herein by reference in its entirety.
  • Sixty percent of traumatic head injuries result in neuro-ophthalmic abnormalities (Van Stavem, el al.
  • the cornea serves a protective role as the outermost tissue of the eye, however it is highly vulnerable to severe injury and disease. Its lack of blood vessels enables its transparency but also limits its ability to heal. Comeal injury, due to its potential to cause irreversible blindness, requires prompt intervention and aggressive treatment.
  • the critical need for improved ocular surface healing therapies is particularly apparent for chemical bums and in severe comeal diseases, such as ocular manifestations of acute Chronic Graft v. Host Disease (GvHD), Stevens-Johnson Syndrome, Ocular Mucous Membrane Pemphigoid and other conditions giving rise to persistent comeal epithelial defect, which collectively comprise an incidence of over 100,000 cases per year. (See, Dietrich-Ntoukas etal. Cornea.
  • topical ophthalmic drug development is impeded by many anatomical constraints including tear turnover and dilution, nasolacrimal drainage, and reflex blinking with often less than 5% of the topically administered dose reaching deeper ocular tissues (Gaudana et al, Pharm Res. 26(5): 1197-216 (2009)).
  • the initial insult causes rifts in the comeal epithelium thereby enabling the passage of topically applied MSC-S to penetrate the epithelial layers.
  • the provide invention provides a composition comprising an anti-Met antibody, wherein the antibody comprises: a) a vhCDRl, vhCDR2, and vhCDR3 from SEQ ID NO:5; and b) a vlCDRl, vlCDR2, and vlCDR3 from SEQ ID NO: 13.
  • the antibody comprises a heavy chain variable domain having at least 80% identity to SEQ ID NO:5 and a light chain variable domain having at least 80% identity to SEQ ID NO: 13.
  • the antibody comprises: a) a vhCDRl having 0-2 amino acid substitutions from the vhCDRl from SEQ ID NO:5; b) a vhCDR2 having 0-2 amino acid substitutions from the vhCDR2 from SEQ ID NO:5; c) a vhCDR3 having 0-2 amino acid substitutions from the vhCDR3 from SEQ ID
  • the antibody comprises a heavy chain variable domain comprising SEQ ID NO:5 and a light chain variable domain comprising SEQ ID NO: 13.
  • the antibody comprises a CHl-hinge-CH2-CH3 region from human IgGl, IgG2, IgG3, or IgG4.
  • the antibody comprises a CHl-hinge-CH2-CH3 region from human IgGl .
  • the antibody comprises a CL region of human kappa constant.
  • the heavy chain comprises SEQ ID NO: 1 and the light chain comprises SEQ ID NO:9. [0015] In some embodiments, the heavy chain further comprising a first signal peptide sequence of SEQ ID NO:3 and the light chain further comprising a second signal peptide sequence of SEQ ID NO: 11.
  • the present invention provides a nucleic composition comprising: a) a first nucleic acid encoding the heavy chain variable domain of the anti-Met antibody; and b) a second nucleic acid encoding the light chain variable domain of the same anti-Met antibody.
  • the present invention provides an expression vector composition comprising: a) a first expression vector comprising the first nucleic acid; and b) a second expression vector comprising the second nucleic acid.
  • the present invention provides an expression vector composition comprising an expression vector comprising the first and second nucleic acids.
  • the present invention provides a host cell comprising the expression vector composition.
  • the present invention provides a method of making an anti- Met antibody comprising: a) culturing the host cell under conditions wherein the antibody is expressed; and b) recovering the anti-Met antibody.
  • the present invention provides a method of treating an ocular condition in a subject in need thereof, wherein the method comprises administering to the subject an anti-MET antibody, the antibody comprising: a) a vhCDRl, vhCDR2, and vhCDR3 from SEQ ID NO:5; and b) a vlCDRl, vlCDR2, and vlCDR3 from SEQ ID NO: 13.
  • the antibody comprises a heavy chain variable domain having at least 80% identity to SEQ ID NO:5 and a light chain variable domain having at least 80% identity to SEQ ID NO: 13.
  • the antibody comprises: a) a vhCDRl having 0-2 amino acid substitutions from the vhCDRl from SEQ ID NO:5; b) a vhCDR2 having 0-2 amino acid substitutions from the vhCDR2 from SEQ ID NO:5; c) a vhCDR3 having 0-2 amino acid substitutions from the vhCDR3 from SEQ ID
  • the antibody used for such a treatment comprises a heavy chain variable domain comprising SEQ ID NO:5 and a light chain variable domain comprising SEQ ID NO: 13.
  • the antibody used for such a treatment comprises a CHl-hinge-CH2-CH3 region from human IgGl, IgG2, IgG3, or IgG4.
  • the antibody used for such a treatment comprises a CHl-hinge-CH2-CH3 region from human IgGl. [0027] In some embodiments, the antibody used for such a treatment comprises a CL region of human kappa constant.
  • the heavy chain of the antibody used for such a treatment comprises SEQ ID NO: 1 and the light chain comprises SEQ ID NO:9. [0029] In some embodiments, the heavy chain of the antibody used for such a treatment further comprising a first signal peptide sequence of SEQ ID NO:3 and the light chain further comprising a second signal peptide sequence of SEQ ID NO: 11.
  • the ocular condition is selected from the group consisting of Chronic Graft v. Host Disease (GvHD), Stevens-Johnson Syndrome, Ocular Mucous Membrane Pemphigoid, Persistent Comeal Epithelial Defect (PCED), dry eye, ocular nerve tissue damage, concussive injury to the eye (such as concussive injury, ocular contusion, or chemical bum), surgical debridement, and contact lens wear.
  • GvHD Chronic Graft v. Host Disease
  • Stevens-Johnson Syndrome Ocular Mucous Membrane Pemphigoid
  • PCED Persistent Comeal Epithelial Defect
  • dry eye ocular nerve tissue damage
  • concussive injury to the eye such as concussive injury, ocular contusion, or chemical bum
  • surgical debridement and contact lens wear.
  • the present disclosures provide for the use of the composition comprising the anti-Met antibody described herein for treating the ocular condition. In some embodiments, the present disclosures provide for the use of the composition for the manufacture of a medicament for treating the ocular condition in a subject in need thereof.
  • FIGURE 1 Schematic diagram of an exemplary embodiment of anti-Met antibody composition preparation, processing, and use.
  • FIGURE 2 SDS-PAGE and western blot analysis of HR11.
  • Lane Ml Protein Marker, TaKaRa, Cat. No. 3452.
  • Lane M2 Protein Marker, GenScript, Cat. No. M00521.
  • Lane 1 Reducing condition.
  • Lane 2 Non-reducing condition.
  • Lane P Human IgGl, Kappa (Sigma, Cat.No.15154) as a positive control.
  • Primary antibody Goat Anti-Human IgG-HRP (GenScript, Cat. No. A00166).
  • Primary antibody Goat Anti-Human Kappa-HRP (SouthemBiotech, Cat. No. 2060-05).
  • FIGURE 3A-3G Exemplary amino acid and nucleic acid sequences of HR11.
  • FIGURE 4 Mechanical wound efficacy of anti-Met monoclonal antibody (HR11) - depicted are representative images of eyes treated with HR11 or vehicle control.
  • FIGURE 5 Mechanical wound efficacy of anti-Met monoclonal antibody (HR11) - percentage of wounds completely closed.
  • FIGURE 6 Mechanical wound efficacy of anti-Met monoclonal antibody (HR11) - HR11 mAh promotes comeal wound healing. A 3.0 mm epithelial defect was created in mouse corneas. Wounds were treated with HR11 twice daily at 3.0 mg/mL for seven days.
  • c-Met is a typical receptor tyrosine kinase (RTK) present on a cell surface that induces intracellular signal transmission by binding with its ligand hepatocyte growth factor (HGF), thereby facilitating cell growth.
  • RTK receptor tyrosine kinase
  • HGF hepatocyte growth factor
  • c-Met may facilitate liver tissue regeneration after a hepatectomy or damage from liver cancer or cirrhosis; kidney tissue regeneration after simple or partial kidney resection from cancer, infection, renal stones, or renal artery stricture; skin tissue regeneration in patients with skin damage from bums, bedsores, or skin ulcers; and heart tissue regeneration after damage from cardiac infarction.
  • c-Met is involved in a variety of mechanisms, for example, cancer occurrence, metastasis, cancer cell migration and invasion and angiogenesis, and in the growth of a variety of cells. Further to the ability to facilitate regeneration and growth of normal tissue cells such as liver, kidney and heart cells, c-Met is known to facilitate growth and proliferation of stem cells when bound to a growth factor HGF.
  • c-Met refers to a receptor tyrosine kinase (RTK) that binds to a hepatocyte growth factor (HGF).
  • RTK receptor tyrosine kinase
  • HGF hepatocyte growth factor
  • the c-Met protein may include, for example, polypeptides encoded by nucleotide sequences of GenBank Accession Number NM.sub.— 000245, proteins encoded by polypeptide sequences of GenBank Accession Number NM.sub.— 000236, or extracellular domains thereof.
  • the RTK c-Met is involved in a variety of mechanisms, for example, cancer occurrence, metastasis, cancer cell migration and invasion, angiogenesis, cell migration, and cell proliferation.
  • antigen binding domain or “ABD” herein is meant a set of six Complementary Determining Regions (CDRs) that, when present as part of a polypeptide sequence, specifically binds a target antigen as discussed herein.
  • CDRs Complementary Determining Regions
  • a “Met antigen binding domain” binds Met antigen as outlined herein.
  • these CDRs are generally present as a first set of variable heavy CDRs (vhCDRs or VHCDRS) and a second set of variable light CDRs (vlCDRs or VLCDRS), each comprising three CDRs: vhCDRl, vhCDR2, vhCDR3 for the heavy chain and vlCDRl, vlCDR2 and vlCDR3 for the light.
  • the CDRs are present in the variable heavy and variable light domains, respectively, and together form an Fv region.
  • the six CDRs of the antigen binding domain are contributed by a variable heavy and variable light chain.
  • variable heavy domain containing the vhCDRl, vhCDR2 and vhCDR3
  • variable light domain vl or VL; containing the vlCDRl, vlCDR2 and vlCDR3
  • C-terminus of the vh domain being attached to the N-terminus of the CHI domain of the heavy chain
  • C-terminus of the vl domain being attached to the N-terminus of the constant light domain (and thus forming the light chain).
  • Antigen-binding protein refers to a protein that comprises an antigen binding domain.
  • human antibody includes all antibodies that have one or more variable and constant regions derived from human immunoglobulin sequences. In one embodiment, all of the variable and constant domains are derived from human immunoglobulin sequences (a fully human antibody).
  • Human antibodies may be prepared in a variety of ways, including immunization of a mouse that is genetically modified to express human antibodies. One can engineer mouse strains deficient in mouse antibody production with large fragments of the human Ig loci in anticipation that such mice would produce human antibodies in the absence of mouse antibodies. Large human Ig fragments may preserve the large variable gene diversity as well as the proper regulation of antibody production and expression.
  • human antibody repertoire in these mouse strains may yield high affinity fully human antibodies against any antigen of interest, including human antigens.
  • antigen-specific human MAbs with the desired specificity may be produced and selected.
  • Human antibodies can also be prepared by panning human antibody libraries expressed on phage, phagemids, ribosomes, or other particles.
  • a “humanized antibody” has a sequence that differs from the sequence of an antibody derived from a non-human species by one or more amino acid substitutions, deletions, and/or additions, such that the humanized antibody is less likely to induce an immune response, and/or induces a less severe immune response, as compared to the non human species antibody, when it is administered to a human subject.
  • certain amino acids in the framework and constant domains of the heavy and/or light chains of the non-human species antibody are mutated to produce the humanized antibody.
  • the constant domain(s) from a human antibody are fused to the variable domain(s) of anon-human species.
  • one or more amino acid residues in one or more CDR sequences of a non-human antibody are changed to reduce the likely immunogenicity of the non-human antibody when it is administered to a human subject, wherein the changed amino acid residues either are not critical for immunospecific binding of the antibody to its antigen, or the changes to the amino acid sequence that are made are conservative changes, such that the binding of the humanized antibody to the antigen is not significantly worse than the binding of the non-human antibody to the antigen.
  • Examples of methods for making humanized antibodies may be found in U.S. Pat. Nos. 6,054,297, 5,886,152 and 5,877,293, the disclosures of each of which are incorporated by reference herein.
  • chimeric antibody refers to an antibody that contains one or more regions from one antibody and one or more regions from one or more other antibodies.
  • a “CDR grafted antibody” is an antibody comprising one or more CDRs derived from an antibody of a particular species or isotype and the framework of another antibody of the same or different species or isotype.
  • modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence or an alteration to a moiety chemically linked to a protein.
  • a modification may be an altered carbohydrate or PEG structure attached to a protein.
  • amino acid modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
  • the amino acid modification is always to an amino acid coded for by DNA, e.g. the 20 amino acids that have codons in DNA and RNA.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with a different amino acid.
  • the substitution is to an amino acid that is not naturally occurring at the particular position, either not naturally occurring within the organism or in any organism.
  • the substitution N297A refers to a variant polypeptide, in this case an Fc variant, in which the asparagine at position 297 is replaced with alanine.
  • a protein which has been engineered to change the nucleic acid coding sequence but not change the starting amino acid is not an “amino acid substitution”; that is, despite the creation of a new gene encoding the same protein, if the protein has the same amino acid at the particular position that it started with, it is not an amino acid substitution.
  • amino acid insertion or "insertion” as used herein is meant the addition of an amino acid sequence at a particular position in a parent polypeptide sequence.
  • -233E or 233E designates an insertion of glutamic acid after position 233 and before position 234.
  • -233ADE or A233ADE designates an insertion of AlaAspGlu after position 233 and before position 234.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid sequence at a particular position in a parent polypeptide sequence.
  • E233- or E233#, E233() or E233del designates a deletion of glutamic acid at position 233.
  • EDA233- or EDA233# designates a deletion of the sequence GluAspAla that begins at position 233.
  • variant protein or “protein variant”, or “variant” as used herein is meant a protein that differs from that of a parent protein by virtue of at least one amino acid modification.
  • Protein variant may refer to the protein itself, a composition comprising the protein, or the amino sequence that encodes it.
  • the protein variant has at least one amino acid modification compared to the parent protein, e.g. from about one to about seventy amino acid modifications, in one exemplary embodiment from about one to about five amino acid modifications compared to the parent.
  • the parent polypeptide for example an Fc parent polypeptide, is a human wild type sequence, such as the Fc region from IgGl, IgG2, IgG3 or IgG4, although human sequences with variants can also serve as “parent polypeptides”.
  • Exemplary protein variant sequences herein will possess at least about 80% identity with a parent protein sequence, in some embodiments at least about 90% identity, in some embodiments at least about 95-98- 99% identity.
  • Variant protein can refer to the variant protein itself, compositions comprising the protein variant, or the DNA sequence that encodes it.
  • antibody variant or “variant antibody” as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification
  • IgG variant or “variant IgG” as used herein is meant an antibody that differs from a parent IgG (again, in many cases, from a human IgG sequence) by virtue of at least one amino acid modification
  • immunoglobulin variant or “variant immunoglobulin” as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification
  • Fc variant or “variant Fc” as used herein is meant a protein comprising an amino acid modification in an Fc domain.
  • the Fc variants of the present invention are defined according to the amino acid modifications that compose them.
  • the EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference.)
  • the modification can be an addition, deletion, or substitution.
  • Substitutions can include naturally occurring amino acids and, in some cases, synthetic amino acids. Examples include U.S. Pat. No. 6,586,207; WO 98/48032; WO 03/073238; US2004-0214988A1; WO 05/35727A2; WO 05/74524A2; J. W.
  • protein herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
  • the peptidyl group may comprise naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids (see Simon et al., PNAS USA 89(20):9367 (1992), entirely incorporated by reference).
  • the amino acids may either be naturally occurring or synthetic (e.g. not an amino acid that is coded for by DNA); as will be appreciated by those in the art.
  • homo-phenylalanine, citrulline, ornithine and noreleucine are considered synthetic amino acids for the purposes of the invention, and both D- and L-(R or S) configured amino acids may be utilized.
  • the variants of the present invention may comprise modifications that include the use of synthetic amino acids incorporated using, for example, the technologies developed by Schultz and colleagues, including but not limited to methods described by Cropp & Shultz, 2004, Trends Genet.
  • polypeptides may include synthetic derivatization of one or more side chains or termini, glycosylation, PEGylation, circular permutation, cyclization, linkers to other molecules, fusion to proteins or protein domains, and addition of peptide tags or labels.
  • residue as used herein is meant a position in a protein and its associated amino acid identity.
  • Asparagine 297 also referred to as Asn297 or N297
  • Asn297 is a residue at position 297 in the human antibody IgGl .
  • Fab or "Fab region” as used herein is meant the polypeptide that comprises the VH, CHI, VL, and CL immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody or antibody fragment.
  • Fv or “Fv fragment” or “Fv region” as used herein is meant a polypeptide that comprises the VL and VH domains of a single antibody. As will be appreciated by those in the art, these generally are made up of two chains.
  • single chain Fv or “scFv” herein is meant a variable heavy domain covalently attached to a variable light domain, generally using a scFv linker as discussed herein, to form a scFv or scFv domain.
  • a scFv domain can be in either orientation from N- to C-terminus (vh-linker-vl or vl-linker-vh).
  • the linker is a scFv linker as is generally known in the art, with the linker peptide predominantly including the following amino acid residues: Gly, Ser, Ala, or Thr.
  • the linker peptide should have a length that is adequate to link two molecules in such a way that they assume the correct conformation relative to one another so that they retain the desired activity.
  • the linker is from about 1 to 50 amino acids in length, in some embodiments about 1 to 30 amino acids in length. In one embodiment, linkers of 1 to 20 amino acids in length may be used, with from about 5 to about 10 amino acids finding use in some embodiments.
  • Useful linkers include glycine-serine polymers, including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least one (and generally from 3 to 4), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers.
  • glycine-serine polymers including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least one (and generally from 3 to 4), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers.
  • nonproteinaceous polymers including but not limited to polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol, may find use as linkers, that is may find use as linkers.
  • IgG subclass modification or “isotype modification” as used herein is meant an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype.
  • IgGl comprises a tyrosine and IgG2 a phenylalanine at EU position 296, a F296Y substitution in IgG2 is considered an IgG subclass modification.
  • IgGl has a proline at position 241 and IgG4 has a serine there, an IgG4 molecule with a S241P is considered an IgG subclass modification.
  • subclass modifications are considered amino acid substitutions herein.
  • amino acid and “amino acid identity” as used herein is meant one of the 20 naturally occurring amino acids that are coded for by DNA and RNA.
  • effector function as used herein is meant a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to ADCC, ADCP, and CDC. In many cases, it is desirable to ablate most or all effector functions using either different IgG isotypes (e.g. IgG4) or amino acid substitutions in the Fc domain; however, preserving binding to the FcRn receptor is desirable, as this contributes to the half-life of the antibodies in human serum.
  • IgG Fc ligand as used herein is meant a molecule, for example a polypeptide, from any organism that binds to the Fc region of an IgG antibody to form an Fc/Fc ligand complex.
  • Fc ligands include but are not limited to FcyRIs, FcyRIIs, FcyRIIIs, FcRn, Clq, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcyR.
  • Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcyRs (Davis et ak, 2002, Immunological Reviews 190:123-136, entirely incorporated by reference).
  • Fc ligands may include undiscovered molecules that bind Fc. Particular IgG Fc ligands are FcRn and Fc gamma receptors.
  • Fc ligand as used herein is meant a molecule, for example a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc/Fc ligand complex.
  • parent polypeptide as used herein is meant a starting polypeptide that is subsequently modified to generate a variant.
  • the parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it.
  • parent immunoglobulin as used herein is meant an unmodified immunoglobulin polypeptide that is modified to generate a variant
  • parent antibody as used herein is meant an unmodified antibody that is modified to generate a variant antibody. It should be noted that "parent antibody” includes known commercial, recombinantly produced antibodies as outlined below.
  • Fc or “Fc region” or “Fc domain” as used herein is meant the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain and in some cases, part of the hinge.
  • Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains.
  • Fc may include the J chain.
  • the Fc domain comprises immunoglobulin domains Cy2 and Cy3 (Cy2 and Cy3) and the lower hinge region between Cyl (Cyl) and Cy2 (Cy2).
  • the human IgG heavy chain Fc region is usually defined to include residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat.
  • amino acid modifications are made to the Fc region, for example to alter binding to one or more FcyR receptors or to the FcRn receptor.
  • “heavy constant region” herein is meant the CHl-hinge-CH2-CH3 portion of an antibody.
  • position as used herein is meant a location in the sequence of a protein. Positions may be numbered sequentially, or according to an established format, for example the EU index for antibody numbering.
  • target antigen as used herein is meant the molecule that is bound specifically by the variable region of a given antibody.
  • the target antigen of interest herein is Met, usually human Met and optionally cyno Met, the sequences of which are shown in .
  • target cell as used herein is meant a cell that expresses a target antigen.
  • variable region as used herein is meant the region of an immunoglobulin that comprises one or more Ig domains substantially encoded by any of the VK (V. kappa), nl (V.lamda), and/or VH genes that make up the kappa, lambda, and heavy chain immunoglobulin genetic loci respectively.
  • wild type or WT herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations.
  • a WT protein has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
  • the antibodies of the present invention are generally isolated or recombinant. “Isolated,” when used to describe the various polypeptides disclosed herein, means a polypeptide that has been identified and separated and/or recovered from a cell or cell culture from which it was expressed. Ordinarily, an isolated polypeptide will be prepared by at least one purification step. An “isolated antibody,” refers to an antibody which is substantially free of other antibodies having different antigenic specificities.
  • the term “recombinant,” as applied to a polynucleotide means the polynucleotide is the product of various combinations of cloning, restriction or ligation steps, and other procedures resulting in a construct distinct and/or different from a polynucleotide found in nature. The terms respectively include replicates of the original polynucleotide construct and progeny of the original virus construct.
  • the term in vivo refers to an event that takes place in a subject's body.
  • synthetic antibody as used herein includes an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage.
  • the term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using synthetic DNA or amino acid sequence technology which is available and well known in the art.
  • in vitro refers to an event that takes places outside of a subject's body.
  • in vitro assays encompass cell-based assays in which cells alive or dead are employed and may also encompass a cell-free assay in which no intact-cells are employed.
  • Specific binding or “specifically binds to” or is “specific for” a particular antigen or an epitope means binding that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. For example, specific binding can be determined by competition with a control molecule that is similar to the target.
  • Specific binding for a particular antigen or an epitope can be exhibited, for example, by an antibody having a KD for an antigen or epitope of at least about 10 9 M, at least about 10 10 M, at least about 10 11 M, at least about 10 12 M, at least about 10 13 M, at least about 10 14 M, at least about 10 15 M, where KD refers to a dissociation rate of a particular antibody-antigen interaction.
  • an antibody that specifically binds an antigen will have a KD that is 20-, 50-, 100-, 500-, 1000-, 5,000-, 10,000- or more times greater for a control molecule relative to the antigen or epitope.
  • binding for a particular antigen or an epitope can be exhibited, for example, by an antibody having a KA or Ka for an antigen or epitope of at least 20-, 50-, 100-, 500-, 1000-, 5,000-, 10,000- or more times greater for the epitope relative to a control, where KA or Ka refers to an association rate of a particular antibody-antigen interaction. Binding affinity is generally measured using surface plasmon resonance (e.g. Biacore assay) and flow cytometry with antigen-expressing cells.
  • surface plasmon resonance e.g. Biacore assay
  • polypeptide refers to any polymer formed from multiple amino acids, regardless of length or posttranslational modification (e.g., phosphorylation or glycosylation), associated, at least in part, by covalent bonding (e.g., “protein” as used herein refers both to linear polymers (chains) of amino acids associated by peptide bonds as well as proteins exhibiting secondary, tertiary, or quaternary structure, which can include other forms of intramolecular and intermolecular association, such as hydrogen and van der Waals bonds, within or between peptide chain(s)).
  • protein refers both to linear polymers (chains) of amino acids associated by peptide bonds as well as proteins exhibiting secondary, tertiary, or quaternary structure, which can include other forms of intramolecular and intermolecular association, such as hydrogen and van der Waals bonds, within or between peptide chain(s)).
  • polypeptides include, but are not limited to, proteins, peptides, oligopeptides, dimers, multimers, variants, and the like.
  • the polypeptide can be unmodified such that it lacks modifications such as phosphorylation and glycosylation.
  • a polypeptide can contain part or all of a single naturally-occurring polypeptide, or can be a fusion or chimeric polypeptide containing amino acid sequences from two or more naturally-occurring polypeptides.
  • isolated polypeptide or “purified polypeptide” is meant a polypeptide that is substantially free from the materials with which the polypeptide is normally associated in nature or in culture.
  • the polypeptides of the invention can be obtained, for example, by extraction from a natural source if available (for example, a mammalian cell), by expression of a recombinant nucleic acid encoding the polypeptide (for example, in a cell or in a cell-free translation system), or by chemically synthesizing the polypeptide.
  • polypeptide may be obtained by cleaving full length polypeptides. When the polypeptide is a fragment of a larger naturally occurring polypeptide, the isolated polypeptide is shorter than and excludes the full-length, naturally-occurring polypeptide of which it is a fragment.
  • sequence identity refers to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity.
  • percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences.
  • Suitable programs to determine percent sequence identity include for example the BLAST suite of programs available from the U.S. Government’s National Center for Biotechnology Information BLAST web site. Comparisons between two sequences can be carried using either the BLASTN or BLASTP algorithm. BLASTN is used to compare nucleic acid sequences, while BLASTP is used to compare amino acid sequences. ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or MegAlign, available from DNASTAR, are additional publicly available software programs that can be used to align sequences. One skilled in the art can determine appropriate parameters for maximal alignment by particular alignment software. In certain embodiments, the default parameters of the alignment software are used.
  • isolated refers to material removed from its original environment and is thus altered “by the hand of man” from its natural state.
  • enriched means to selectively concentrate or to increase the amount of one or more materials by elimination of the unwanted materials or selection and separation of desirable materials from a mixture (e.g., separate cells with specific cell markers from a heterogeneous cell population in which not all cells in the population express the marker).
  • substantially purified means a population of cells substantially homogeneous for a particular marker or combination of markers. By substantially homogeneous is meant at least 90%, and in some embodiments 95% homogeneous for a particular marker or combination of markers.
  • multipotent stem cells are true stem cells but can only differentiate into a limited number of types.
  • the bone marrow contains multipotent stem cells that give rise to all the cells of the blood but may not be able to differentiate into other cells types.
  • non-human animal-derived materials such as bovine serum, proteins, lipids, carbohydrates, nucleic acids, vitamins, etc.
  • non-human animal-derived materials such as bovine serum, proteins, lipids, carbohydrates, nucleic acids, vitamins, etc.
  • clinical grade materials such as recombinantly produced human proteins, are used in the preparation, growth, culturing, expansion, storage and/or formulation of such compositions and/or processes.
  • the term “expanded”, in reference to cell compositions, means that the cell population constitutes a significantly higher concentration of cells than is obtained using previous methods.
  • the level of cells per gram of amniotic tissue in expanded compositions of AMP cells is at least 50-fold and up to 150-fold higher than the number of cells in the primary culture after 5 passages, as compared to about a 20-fold increase in such cells using previous methods.
  • the level of cells per gram of amniotic tissue in expanded compositions of AMP cells is at least 30-fold and up to 100- fold higher than the number of cells in the primary culture after 3 passages.
  • an “expanded” population has at least a 2-fold, and up to a 10-fold, improvement in cell numbers per gram of amniotic tissue over previous methods.
  • the term “expanded” is meant to cover only those situations in which a person has intervened to elevate the number of the cells.
  • conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide support to or affect the behavior of other cells. Such factors include, but are not limited to, hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, chemokines, receptors, inhibitors and granules.
  • the medium containing the cellular factors is the conditioned medium. Examples of methods of preparing conditioned media have been described in U.S. Pat. No. 6,372,494 which is incorporated by reference in its entirety herein.
  • conditioned medium also refers to components, such as proteins, that are recovered and/or purified from conditioned medium or from for example, MSC cells.
  • the term “mesenchymal stem cell composition” or “MSC composition” means conditioned medium that has been derived from MSCs and in some instances has undergone further processing.
  • anti-Met antibody composition can refer to the crude conditioned media derived from the MSC.
  • anti-Met antibody composition can refer to the composition obtained from the crude conditioned media after it has been subjected to further processing as described herein.
  • the term “suspension” means a liquid containing dispersed components, e.g., cytokines.
  • the dispersed components may be fully solubilized, partially solubilized, suspended or otherwise dispersed in the liquid.
  • Suitable liquids include, but are not limited to, water, osmotic solutions such as salt and/or sugar solutions, cell culture media, and other aqueous or non-aqueous solutions.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxy glutamate, and O-phosphoserine.
  • Amino acid analogs refer to compounds that have the same basic chemical structure as a naturally occurring amino acid, e.g., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid.
  • amino acids can be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, can be referred to by their commonly accepted single-letter codes.
  • An “amino acid substitution” refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence (an amino acid sequence of a starting polypeptide) with a second, different “replacement” amino acid residue.
  • amino acid insertion refers to the incorporation of at least one additional amino acid into a predetermined amino acid sequence.
  • amino acid deletion refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.
  • Polypeptide “peptide”, and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • Nucleic acid refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double- stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences and as well as the sequence explicitly indicated.
  • degenerate codon substitutions can be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al, Nucleic Acid Res. 19:5081, 1991; Ohtsuka e/ a/. , Biol. Chem. 260:2605-2608, 1985; and Cassol et al, 1992; Rossolini et al, Mol. Cell. Probes 8:91-98, 1994).
  • modifications at the second base can also be conservative.
  • nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene.
  • Polynucleotides used herein can be composed of any polyribonucleotide or polydeoxribonucleotide, which can be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides can be composed of single- and double- stranded DNA, DNA that is a mixture of single- and double- stranded regions, single- and double- stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that can be single- stranded or, more typically, double- stranded or a mixture of single- and double- stranded regions.
  • polynucleotide can be composed of triple- stranded regions comprising RNA or DNA or both RNA and DNA.
  • a polynucleotide can also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons. “Modified” bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications can be made to DNA and RNA; thus, “polynucleotide” embraces chemically, enzymatically, or metabolically modified forms.
  • a “vector” is capable of transferring gene sequences to target-cells.
  • vector construct means any nucleic acid construct capable of directing the expression of a gene of interest and which can transfer gene sequences to target-cells, which can be accomplished by genomic integration of all or a portion of the vector, or transient or inheritable maintenance of the vector as an extrachromosomal element.
  • vector transfer vector mean any nucleic acid construct capable of directing the expression of a gene of interest and which can transfer gene sequences to target-cells, which can be accomplished by genomic integration of all or a portion of the vector, or transient or inheritable maintenance of the vector as an extrachromosomal element.
  • the term includes cloning, and expression vehicles, as well as integrating vectors.
  • regulatory element includes a nucleotide sequence which controls some aspect of the expression of nucleic acid sequences.
  • regulatory elements illustratively include an enhancer, an internal ribosome entry site (IRES), an intron, an origin of replication, a polyadenylation signal (pA), a promoter, an enhancer, a transcription termination sequence, and an upstream regulatory domain, which contribute to the replication, transcription, and/or post-transcriptional processing of a nucleic acid sequence.
  • regulatory elements can also include cv.v-regulatoiy DNA elements as well as transposable elements (TEs). Those of ordinary skill in the art are capable of selecting and using these and other regulatory elements in an expression construct with no more than routine experimentation. Expression constructs can be generated using a genetic recombinant approach or synthetically using well-known methodology.
  • control element or “control sequence” is a nucleotide sequence involved in an interaction of molecules contributing to the functional regulation of a polynucleotide, including replication, duplication, transcription, splicing, translation, or degradation of the polynucleotide. The regulation may affect the frequency, speed, or specificity of the process, and may be enhancing or inhibitory in nature.
  • Control elements known in the art include, for example, transcriptional regulatory sequences such as promoters and enhancers.
  • a promoter is a DNA region capable under certain conditions of binding RNA polymerase and initiating transcription of a coding region usually located downstream (in the 3’ direction) from the promoter.
  • secretome composition refers to a composition comprising one or more substances which are secreted from a cell.
  • a secretome composition may include one or more cytokines, one or more exosomes, and/or one or more microvesicles.
  • a secretome composition may be purified or unpurified.
  • a secretome composition may further comprise one or more substances that are not secreted from a cell (e.g., culture media, additives, nutrients, etc.).
  • a secretome composition does not comprise and or comprises only trace amounts of one or more substances that are not secreted from a cell (e.g., culture media, additives, nutrients, etc.).
  • treatment covers any treatment of a human or nonhuman mammal (e.g., rodent, cat, dog, horse, cattle, sheep, and primates etc.), and includes preventing the disease or condition from occurring in a subject who may be predisposed to the disease or condition but has not yet been diagnosed as having it. It also includes inhibiting (arresting development ol), relieving or ameliorating (causing regression ol), or curing (permanently stopping development or progression) the disease, condition and/or any related symptoms.
  • a human or nonhuman mammal e.g., rodent, cat, dog, horse, cattle, sheep, and primates etc.
  • treatment covers any treatment of a disease or condition of a mammal, particularly a human, and includes: (a) preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it; (b) inhibiting the disease or condition, e.g., arresting its development; (c) relieving and or ameliorating the disease or condition, e.g., causing regression of the disease or condition; or (d) curing the disease or condition, e.g., stopping its development or progression.
  • the population of subjects treated by the methods of the invention includes subjects suffering from the undesirable condition or disease, as well as subjects at risk for development of the condition or disease.
  • treatment refers to any administration of a therapy that partially or completely alleviates, ameliorates, relives, inhibits, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms, features, and/or causes of a particular disease, disorder, and/or condition.
  • such treatment may be of a subject who does not exhibit signs of the relevant disease, disorder, and/or condition, and/or of a subject who exhibits only early signs of the disease, disorder, and/or condition.
  • such treatment may be of a subject who exhibits one or more established signs of the relevant disease, disorder, and/or condition.
  • treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, and/or condition. In some embodiments, treatment may be of a subject known to have one or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, and/or condition.
  • a “wound” is any disruption, from whatever cause, of normal anatomy (internal and/or external anatomy) including but not limited to traumatic injuries such as mechanical ( e.g . contusion, penetrating), thermal, chemical, electrical, radiation, concussive and incisional injuries; elective injuries such as operative surgery and resultant incisional hernias, fistulas, etc. ; acute wounds, chronic wounds, infected wounds, and sterile wounds, as well as wounds associated with disease states (e.g. ocular contusion).
  • traumatic injuries such as mechanical (e.g . contusion, penetrating), thermal, chemical, electrical, radiation, concussive and incisional injuries
  • elective injuries such as operative surgery and resultant incisional hernias, fistulas, etc.
  • acute wounds, chronic wounds, infected wounds, and sterile wounds as well as wounds associated with disease states (e.g. ocular cont
  • wound healing refers to improving, by some form of intervention, the natural cellular processes and humoral substances of tissue repair such that healing is faster, and/or the resulting healed area has less scaring and/or the wounded area possesses tissue strength that is closer to that of uninjured tissue and/or the wounded tissue attains some degree of functional recovery.
  • a “therapeutically effective” or “effective” dosage or amount of a composition is an amount sufficient to have a positive effect on a given medical condition. If not immediate, the therapeutically effective or effective dosage or amount may, over period of time, provide a noticeable or measurable effect on a patient's health and well-being.
  • composition refers to an a mixture of at least one compound, such as the compound of the anti-Met antibody provided herein, with at least one and optionally more than one other pharmaceutically acceptable chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • pharmaceutically acceptable chemical components such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • pharmaceutically acceptable carrier refers to a carrier or a diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compounds.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound.
  • mixing As used herein, the terms “mix”, “mixing”, and the like describe a mechanical process or a mechanical treatment of the components. For example, mixing can be in the sense of carrying out repeated cycles of pressing and folding or comparable processing steps which lead to an intense compression and mixing of the provided hydrophobic matrices.
  • adult stem cells can be harvested from a variety of adult tissues, including bone marrow, fat, and dental pulp tissue. While all adult stem cells are cable of self-renewal and are considered multipotent, their therapeutic functions vary depending on their origin. As a result, each type of adult stem cell has unique characteristics that make them suitable for certain diseases.
  • MSCs Mesenchymal stem cells
  • osteoblasts e.g., bone cells
  • chondrocytes e.g., cartilage cells
  • myocytes e.g., muscle cells
  • adipocytes e.g., fat cells which give rise to marrow adipose tissue.
  • isolated refers to cells removed from their original environment. Stem cells produce factors, such as growth factors, that regulate or are important for regulating multiple biological processes.
  • a growth factor is an agent, such as a naturally occurring substance capable of stimulating cellular growth and/or proliferation and/or cellular differentiation.
  • growth factors are proteins or steroid hormones. While the terms “growth factor” and “factor” and the like are used interchangeably herein, the term “biological factor” is not limited to growth factors.
  • Human mesenchymal stem cells can be characterized by the surface marker profile of CD45-/CD31-/CD73+/CD90+/CD105+/CD44+ (or any suitable subset thereof). (See Bourin etal, Cytotherapy 15(6):641-648 (2013)). Further, appropriate stem cells display the CD34+ positive at the time of isolation, but lose this marker during culturing. Therefore, the full marker profile for one stem cell type that may be used according to the present application includes CD45-/CD31-/CD73+/CD90+/CD105+. In another embodiment utilizing mouse stem cells, the stem cells are characterized by the Sca- 1 marker, instead of CD34, to define what appears to be a homologue to the human cells described above, with the remaining markers remaining the same.
  • conditioned medium refers to media which includes biological factors secreted by MSCs. This can also be referred to herein as the “secretome”, “MSC-CM”, “anti-Met antibody composition” and/or “MSC derived secretome”. Also provided are processed “conditioned medium” which included biological factors secreted by MSCs and which has been further processed by, for example, filtration, purification, and/or concentration procedures.
  • the “conditioned medium” is obtained by culturing stem cells in media, as described herein in detail, and separating the resulting media, which contains stem cells and their secreted stem cell products (secretome) into conditioned medium that contains biological factors and fewer stem cells than were present prior to separation.
  • the conditioned medium may be used in the methods described herein and is substantially free of stem cells (may contain a small percentage of stem cells) or free of stem cells.
  • Biological factors that may be in the conditioned medium include, but are not limited to, proteins (e.g., cytokines, chemokines, growth factors, enzymes), nucleic acids (e.g, miRNA), lipids (e.g., phospholipids), polysaccharides, and/or combinations thereof. Any combination(s) of these biological factors may be either bound within or on the surface of extracellular vesicles (e.g., exosomes) or separate from extracellular vesicles.
  • antibody is used generally.
  • Traditional antibody structural units typically comprise a tetramer.
  • Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one “light” (typically having a molecular weight of about 25 kDa) and one “heavy” chain (typically having a molecular weight of about 50-70 kDa).
  • Human light chains are classified as kappa and lambda light chains.
  • the present invention is directed to antibodies that generally are based on the IgG class, which has several subclasses, including, but not limited to IgGl, IgG2, IgG3, and IgG4.
  • IgGl, IgG2 and IgG4 are used more frequently than IgG3. It should be noted that IgGl has different allotypes with polymorphisms at 356 (D or E) and 358 (L or M). The sequences depicted herein use the 356D/358M allotype, however the other allotype is included herein. That is, any sequence inclusive of an IgGl Fc domain included herein can have 356E/358L replacing the 356D/358M allotype.
  • each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition, generally referred to in the art and herein as the “Fv domain” or “Fv region”.
  • Fv domain or “Fv region”.
  • CDR complementarity determining region
  • Variable refers to the fact that certain segments of the variable region differ extensively in sequence among antibodies. Variability within the variable region is not evenly distributed. Instead, the V regions consist of relatively invariant stretches called framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called “hypervariable regions” that are each 9-15 amino acids long or longer.
  • Each VH and VL is composed of three hypervariable regions (“complementary determining regions,” “CDRs”) and four FRs, arranged from amino- terminus to carboxy -terminus in the following order: FR1-CDR1-FR2-CDR2-FR3-CDR3- FR4.
  • the hypervariable region generally encompasses amino acid residues from about amino acid residues 24-34 (LCDR1; “L” denotes light chain), 50-56 (LCDR2) and 89-97 (LCDR3) in the light chain variable region and around about 31-35B (HCDR1; “H” denotes heavy chain), 50-65 (HCDR2), and 95-102 (HCDR3) in the heavy chain variable region; Rabat et al., SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991) and/or those residues forming a hypervariable loop (e.g.
  • variable heavy and/or variable light sequence includes the disclosure of the associated (inherent) CDRs.
  • disclosure of each variable heavy region is a disclosure of the vhCDRs (e.g. vhCDRl, vhCDR2 and vhCDR3) and the disclosure of each variable light region is a disclosure of the vlCDRs (e.g. vlCDRl, vlCDR2 and vlCDR3).
  • vlCDRs e.g. vlCDRl, vlCDR2 and vlCDR3
  • the Rabat numbering system is generally used when referring to a residue in the variable domain (approximately, residues 1-107 of the light chain variable region and residues 1-113 of the heavy chain variable region) and the hinge and the EU numbering system for Fc regions (e.g, Rabat et al., supra (1991)).
  • a “full CDR set” comprises the three variable light and three variable heavy CDRs, e.g. a vlCDRl, vlCDR2, vlCDR3, vhCDRl, vhCDR2 and vhCDR3. These can be part of a larger variable light or variable heavy domain, respectfully.
  • the variable heavy and variable light domains can be on separate polypeptide chains, when a heavy and light chain is used, or on a single polypeptide chain in the case of scFv sequences.
  • the CDRs contribute to the formation of the antigen-binding, or more specifically, epitope binding site of antibodies.
  • Epitope refers to a determinant that interacts with a specific antigen binding site in the variable region of an antibody molecule known as a paratope. Epitopes are groupings of molecules such as amino acids or sugar side chains and usually have specific structural characteristics, as well as specific charge characteristics. A single antigen may have more than one epitope.
  • the epitope may comprise amino acid residues directly involved in the binding (also called immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically antigen binding peptide; in other words, the amino acid residue is within the footprint of the specifically antigen binding peptide.
  • Epitopes may be either conformational or linear.
  • a conformational epitope is produced by spatially juxtaposed amino acids from different segments of the linear polypeptide chain.
  • a linear epitope is one produced by adjacent amino acid residues in a polypeptide chain. Conformational and nonconformational epitopes may be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Antibodies that recognize the same epitope can be verified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen, for example “binning.” As outlined below, the invention not only includes the enumerated antigen binding domains and antibodies herein, but those that compete for binding with the epitopes bound by the enumerated antigen binding domains.
  • each chain defines a constant region primarily responsible for effector function.
  • Kabat et al. collected numerous primary sequences of the variable regions of heavy chains and light chains. Based on the degree of conservation of the sequences, they classified individual primary sequences into the CDR and the framework and made a list thereof (see SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, E.A. Kabat et al., entirely incorporated by reference).
  • immunoglobulin domains in the heavy chain.
  • immunoglobulin (Ig) domain herein is meant a region of an immunoglobulin having a distinct tertiary structure.
  • the heavy chain domains including, the constant heavy (CH) domains and the hinge domains.
  • the IgG isotypes each have three CH regions. Accordingly, “CH” domains in the context of IgG are as follows: “CHI” refers to positions 118-220 according to the EU index as in Kabat.
  • CH2 refers to positions 237-340 according to the EU index as in Kabat
  • CH3 refers to positions 341-447 according to the EU index as in Kabat.
  • the pi variants can be in one or more of the CH regions, as well as the hinge region, discussed below.
  • Ig domain of the heavy chain is the hinge region.
  • hinge region or “hinge region” or “antibody hinge region” or “immunoglobulin hinge region” herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.
  • the IgG CHI domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237.
  • the antibody hinge is herein defined to include positions 221 (D221 in IgGl) to 236 (G236 in IgGl), wherein the numbering is according to the EU index as in Kabat.
  • the lower hinge is included, with the “lower hinge” generally referring to positions 226 or 230.
  • the light chain generally comprises two domains, the variable light domain (containing the light chain CDRs and together with the variable heavy domains forming the Fv region), and a constant light chain region (often referred to as CL or CK).
  • Fc region Another region of interest for additional substitutions, outlined below, is the Fc region.
  • the antibodies herein can be derived from a mixture from different species, e.g. a chimeric antibody and/or a humanized antibody.
  • chimeric antibodies and “humanized antibodies” refer to antibodies that combine regions from more than one species.
  • chimeric antibodies traditionally comprise variable region(s) from a mouse (or rat, in some cases) and the constant region(s) from a human.
  • Humanized antibodies generally refer to non-human antibodies that have had the variable-domain framework regions swapped for sequences found in human antibodies.
  • a humanized antibody the entire antibody, except the CDRs, is encoded by a polynucleotide of human origin or is identical to such an antibody except within its CDRs.
  • the CDRs some or all of which are encoded by nucleic acids originating in a non-human organism, are grafted into the beta-sheet framework of a human antibody variable region to create an antibody, the specificity of which is determined by the engrafted CDRs.
  • the creation of such antibodies is described in, e.g., WO 92/11018, Jones, 1986, Nature 321:522-525, Verhoeyen et ak, 1988, Science 239:1534-1536, all entirely incorporated by reference.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin, and thus will typically comprise a human Fc region.
  • Humanized antibodies can also be generated using mice with a genetically engineered immune system. Roque et al., 2004, Biotechnol. Prog. 20:639-654, entirely incorporated by reference.
  • Humanization methods include but are not limited to methods described in Jones et al., 1986, Nature 321:522-525; Riechmann et al.,1988; Nature 332:323-329; Verhoeyen et al., 1988, Science, 239:1534-1536; Queen et al., 1989, Proc Natl Acad Sci, USA 86:10029- 33; He et al., 1998, J. Immunol. 160: 1029-1035; Carter et al., 1992, Proc Natl Acad Sci USA 89:4285-9, Presta et al., 1997, Cancer Res. 57(20):4593-9; Gorman et al., 1991, Proc. Natl. Acad.
  • Humanization or other methods of reducing the immunogenicity of nonhuman antibody variable regions may include resurfacing methods, as described for example in Roguska et al., 1994, Proc. Natl. Acad. Sci. USA 91:969-973, entirely incorporated by reference.
  • the antibodies of the invention comprise a heavy chain variable region from a particular germline heavy chain immunoglobulin gene and/or a light chain variable region from a particular germline light chain immunoglobulin gene (with optional mutations as is generally described herein).
  • such antibodies may comprise or consist of a human antibody comprising heavy or light chain variable regions that are "the product of or "derived from” a particular germline sequence.
  • a human antibody that is "the product of or "derived from” a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequences of human germline immunoglobulins and selecting the human germline immunoglobulin sequence that is closest in sequence (i.e., greatest % identity) to the sequence of the human antibody.
  • a human antibody that is "the product of or "derived from” a particular human germline immunoglobulin sequence may contain amino acid differences as compared to the germline sequence, due to, for example, naturally-occurring somatic mutations or intentional introduction of site-directed mutation.
  • a humanized antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the antibody as being derived from human sequences when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences).
  • a humanized antibody may be at least 95, 96, 97, 98 or 99%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene.
  • a humanized antibody derived from a particular human germline sequence will display no more than 10-20 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene.
  • the humanized antibody may display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene (again, prior to the introduction of any variants herein; that is, the number of variants is generally low, prior to the introduction of the variants of the invention).
  • the parent antibody has been affinity matured, as is known in the art.
  • Structure-based methods may be employed for humanization and affinity maturation, for example as described in USSN 11/004,590.
  • Selection based methods may be employed to humanize and/or affinity mature antibody variable regions, including but not limited to methods described in Wu et al., 1999, J. Mol. Biol. 294:151-162; Baca et al.,
  • the present invention provides anti-Met antibodies, or antigen binding proteins, pharmaceutical compositions for wound healing or tissue regeneration that includes the anti-Met antibodies or antigen-binding proteins.
  • the present invention provides antigen-binding proteins that bind to Met, including but not limited to full length anti-Met antibodies and ScFvs, pharmaceutical compositions for wound healing or tissue regeneration that includes antigen-binding proteins including but not limited to full length anti-Met antibodies and ScFvs that bind to Met.
  • the invention provides antigen binding domains, including but not limited to full length antibodies and ScFvs, which contain a number of specific, enumerated sets of 6 CDRs and defined variable heavy (vh, VH or VH) and variable light (vl, VL or VL), that bind to Met.
  • the antigen-binding protein of the present invention is an anti- Met antibody.
  • the anti-Met antibody is an antibody comprising a set of six CDRs (vhCDRl, vhCDR2, vhCDR3, vlCDRl, vlCDR2 and vlCDR3) fromHRll as depicted in Figure 3.
  • the anti-Met antibody is an antibody comprising a set of six CDRs (vhCDRl, vhCDR2, vhCDR3, vlCDRl, vlCDR2 and vlCDR3) from HR11, with the exception that at least one of the six CDRs comprises one or two conservative amino acid substitutions.
  • one CDR comprises one or two conservative amino acid substitutions. In another embodiment, two CDRs comprise one or two conservative amino acid substitutions. In another embodiment, three CDRs comprise one or two conservative amino acid substitutions. In another embodiment, four CDRs comprise one or two conservative amino acid substitutions. In another embodiment, five CDRs comprise one or two conservative amino acid substitutions. In another embodiment, six CDRs comprise one or two conservative amino acid substitutions.
  • variations are made in both the framework regions that retain at least 80%, 85%, 90% or 95% identity to germline gene sequences.
  • the CDRs can have amino acid modifications (e.g., from 1, 2, 3, 4 or 5 amino acid modifications in the set of CDRs (that is, the CDRs can be modified as long as the total number of changes in the set of 6 CDRs is less than 6 amino acid modifications, with any combination of CDRs being changed; e.g., there may be one change in vlCDRl, two in vhCDR2, none in vhCDR3, etc.).
  • CDR1 and/or CDR2 can have amino acid modifications (e.g., from 1, 2, 3, 4 or 5 amino acid modifications in either CDR1, CDR2, or both), while CDR3 does not contain modifications.
  • the anti-Met antibody is an antibody comprising the variable heavy (vh) domain (SEQ ID NO:5) and variable light (vl) domain (SEQ ID NO: 13) from HR11.
  • the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 80% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively.
  • the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 85% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively.
  • the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 90% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively.
  • the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 95% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively.
  • the antigen-binding domain of the anti-Met antibody is linked to a human IgG constant domain of IgGl, IgG2, IgG3, IgG4 or IgG4.
  • the human IgG domain of the anti-Met antibody is IgGl.
  • the IgGl domain of the anti-Met antibody comprises an amino acid sequence of SEQ ID NO:7.
  • the anti-Met antibody comprises a constant light chain region of human Kappa constant.
  • the constant light chain region of the anti- Met antibody comprises an amino acid sequence of SEQ ID NO: 15.
  • the anti-Met antibody is an antibody comprising the heavy chain (SEQ ID NO:l) and light chain (SEQ ID NO: 9) fromHRll. In one embodiment, the anti-Met antibody is HR11.
  • the antibodies of the invention can be modified, or engineered, to alter the amino acid sequences by amino acid substitutions.
  • amino acid substitutions can be made to alter the affinity of the CDRs for the antigen (including both increasing and decreasing binding), as well as to alter additional functional properties of the antibodies.
  • the antibodies may be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity.
  • an antibody according to at least some embodiments of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody. Such embodiments are described further below.
  • the numbering of residues in the Fc region is that of the EU index of Kabat.
  • the hinge region of CHI is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased.
  • This approach is described further in U.S. Pat. No. 5,677,425 by Bodmer et al.
  • the number of cysteine residues in the hinge region of CHI is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.
  • the antibody can be modified to abrogate in vivo Fab arm exchange, in particular when IgG4 constant domains are used.
  • this process involves the exchange of IgG4 half-molecules (one heavy chain plus one light chain) between other IgG4 antibodies that effectively results in antibodies which are functionally monovalent. Mutations to the hinge region and constant domains of the heavy chain can abrogate this exchange (see Aalberse, RC, Schuurman J., 2002, Immunology 105:9-19).
  • a mutation that finds particular use in the present invention is the S241P in the context of an IgG4 constant domain. IgG4 finds use in the present invention as it has no significant effector function, and is thus used to block the receptor-ligand binding without cell depletion.
  • amino acid substitutions can be made in the Fc region, in general for altering binding to FcyR receptors.
  • Fc gamma receptor By “Fc gamma receptor”, “FcyR” or “FcgammaR” as used herein is meant any member of the family of proteins that bind the IgG antibody Fc region and is encoded by an FcyR gene.
  • this family includes but is not limited to FcyR I (CD64), including isoforms FcyRIa, FcyRIb, and FcyRIc; FcyRII (CD32), including isoforms FcyRIIa (including allotypes H131 and R131), FcyRIIb (including FcyRIIb-l and FcyRIIb-2), and FcyRIIc; and FcyRIII (CD16), including isoforms FcyRIIIa (including allotypes V158 and F158) and FcyRIIIb (including allotypes FcyRIIIb-NAl and FcyRIIIb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference), as well as any undiscovered human FcyRs or FcyR isoforms or allotypes.
  • An FcyR may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Mouse FcyRs include but are not limited to FcyR I (CD64), FcyRII (CD32), FcyRIII-l (CD16), and FcyRIII-2 (CD16-2), as well as any undiscovered mouse FcyRs or FcyR isoforms or allotypes.
  • Fc substitutions that can be made to alter binding to one or more of the FcyR receptors.
  • Substitutions that result in increased binding as well as decreased binding can be useful.
  • ADCC antibody dependent cell-mediated cytotoxicity; the cell- mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcyRIIb an inhibitory receptor
  • Amino acid substitutions that find use in the present invention include those listed in U.S.
  • the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector functions of the antibody.
  • the effector ligand to which affinity is altered can be, for example, an Fc receptor or the Cl component of complement.
  • one or more amino acids selected from amino acid residues 329, 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered Clq binding and/or reduced or abolished complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • one or more amino acid residues within amino acid positions 231 and 239 are altered to thereby alter the ability of the antibody to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al.
  • the Fc region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fey receptor by modifying one or more amino acids at the following positions: 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305,
  • ADCC antibody dependent cellular cytotoxicity
  • the antibodies of the invention are modified to increase its biological half- life.
  • Various approaches are possible.
  • one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Pat. No. 6,277,375 to Ward.
  • the antibody can be altered within the Cm or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Pat. Nos. 5,869,046 and 6,121,022 by Presta et al.
  • Additional mutations to increase serum half-life are disclosed in U.S. Patent Nos. 8,883,973, 6,737,056 and 7,371,826 and include 428L, 434A, 434S, and 428L/434S.
  • the glycosylation of an antibody is modified.
  • an aglycosylated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen or reduce effector function such as ADCC.
  • Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence, for example N297.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site, with an alanine replacement finding use in some embodiments.
  • an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures.
  • altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies according to at least some embodiments of the invention to thereby produce an antibody with altered glycosylation.
  • the cell lines Ms704, Ms705, and Ms709 lack the fucosyltransferase gene, FUT8 (a (1,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their carbohydrates.
  • the Ms704, Ms705, and Ms709 FUT8 cell lines are created by the targeted disruption of the FUT8 gene in CHO/DG44 cells using two replacement vectors (see U.S. Patent Publication No. 20040110704 by Yamane et al. and Yamane-Ohnuki et al. (2004) Biotechnol Bioeng 87:614-22).
  • a cell line with a functionally disrupted FUT8 gene which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the a 1,6 bond-related enzyme.
  • Hanai et al. also describe cell lines which have a low enzyme activity for adding fucose to the N-acetylglucosamine that binds to the Fc region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662).
  • PCT Publication WO 03/035835 by Presta describes a variant CHO cell line, Led 3 cells, with reduced ability to attach fucose to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (see also Shields, R. L. et al. (2002) J Biol. Chem. 277:26733-26740).
  • PCT Publication WO 99/54342 by Umana et al.
  • glycoprotein-modifying glycosyl transferases e.g., b( 1.4)-N-acetylglucosaminyl transferase III (GnTIII)
  • GnTIII glycoprotein-modifying glycosyl transferases
  • the fucose residues of the antibody may be cleaved off using a fucosidase enzyme.
  • the fucosidase a-L-fucosidase removes fucosyl residues from antibodies (Tarentino, A. L. et al. (1975) Biochem. 14:5516-23).
  • Another modification of the antibodies herein that is contemplated by the invention is PEGylation or the addition of other water soluble moieties, typically polymers, e.g., in order to enhance half-life.
  • An antibody can be PEGylated to, for example, increase the biological (e.g., serum) half-life of the antibody.
  • PEG polyethylene glycol
  • the antibody, or fragment thereof typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment.
  • PEG polyethylene glycol
  • the PEGylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • a reactive PEG molecule or an analogous reactive water-soluble polymer.
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Ci-Cio) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide.
  • the antibody to be PEGylated is an aglycosylated antibody. Methods for PEGylating proteins are known in the art and can be applied to the antibodies according to at least some embodiments of the invention. See for example, EP 0 154316 by Nishimura et al. and EP 0401 384 by Ishikawa et al.
  • affinity maturation is done. Amino acid modifications in the CDRs are sometimes referred to as "affinity maturation".
  • An "affinity matured" antibody is one having one or more alteration(s) in one or more CDRs which results in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s). In some cases, it may be desirable to decrease the affinity of an antibody to its antigen.
  • one or more amino acid modifications are made in one or more of the CDRs of the antibodies of the invention.
  • 1 or 2 or 3-amino acids are substituted in any single CDR, and generally no more than from 1, 2, 3. 4, 5, 6, 7, 8 9 or 10 changes are made within a set of CDRs.
  • any combination of no substitutions, 1, 2 or 3 substitutions in any CDR can be independently and optionally combined with any other substitution.
  • Affinity maturation can be done to increase the binding affinity of the antibody for the antigen by at least about 10% to 50-100-150% or more, or from 1 to 5 fold as compared to the "parent" antibody.
  • Exemplary affinity matured antibodies will have nanomolar or even picomolar affinities for the antigen.
  • Affinity matured antibodies are produced by known procedures. See, for example, Marks et al., 1992, Biotechnology 10:779-783 that describes affinity maturation by variable heavy chain (VH) and variable light chain (VL) domain shuffling. Random mutagenesis of CDR and/or framework residues is described in: Barbas, et al. 1994, Proc. Nat. Acad.
  • amino acid modifications can be made in one or more of the CDRs of the antibodies of the invention that are "silent", e.g. that do not significantly alter the affinity of the antibody for the antigen. These can be made for a number of reasons, including optimizing expression (as can be done for the nucleic acids encoding the antibodies of the invention).
  • variant CDRs and antibodies of the invention can include amino acid modifications in one or more of the CDRs of the enumerated antibodies of the invention.
  • amino acid modifications can also independently and optionally be made in any region outside the CDRs, including framework and constant regions.
  • Nucleic acid compositions encoding the anti-Met antibodies of the invention are also provided, as well as expression vectors containing the nucleic acids and host cells transformed with the nucleic acid and/or expression vector compositions.
  • expression vectors containing the nucleic acids and host cells transformed with the nucleic acid and/or expression vector compositions are also provided, as well as expression vectors containing the nucleic acids and host cells transformed with the nucleic acid and/or expression vector compositions.
  • the protein sequences depicted herein can be encoded by any number of possible nucleic acid sequences, due to the degeneracy of the genetic code.
  • An exemplary embodiment of the nucleic acid sequences and expression vectors of the present invention is provided in Figure 3.
  • nucleic acid compositions that encode the antibodies will depend on the format of the antibody.
  • tetrameric antibodies containing two heavy chains and two light chains are encoded by two different nucleic acids, one encoding the heavy chain and one encoding the light chain.
  • These can be put into a single expression vector or two expression vectors, as is known in the art, transformed into host cells, where they are expressed to form the antibodies of the invention.
  • a single nucleic acid encoding the variable heavy chain-linker-variable light chain is generally used, which can be inserted into an expression vector for transformation into host cells.
  • the nucleic acids can be put into expression vectors that contain the appropriate transcriptional and translational control sequences, including, but not limited to, signal and secretion sequences, regulatory sequences, promoters, origins of replication, selection genes, etc.
  • Exemplary mammalian host cells for expressing the recombinant antibodies according to at least some embodiments of the invention include Chinese Hamster Ovary (CHO cells), PERC6, HEK293 and others as is known in the art.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • a nucleic acid is "isolated” or “rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art.
  • the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser)3, such that the VH and VL sequences can be expressed as a contiguous single chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242:423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al., (1990) Nature 348:552-554).
  • a flexible linker e.g., encoding the amino acid sequence (Gly4-Ser)3
  • compositions comprising antigen binding proteins, including but not limited to anti-Met antibodies such as full length anti- Met antibodies or ScFvs, are provided herein.
  • the anti-Met antibody in the composition of the present invention does not promote angiogenesis. In some embodiments, the anti-Met antibody exhibits anti-angiogenic properties. In some embodiments, the composition comprising the anti-Met antibody provides for reduced angiogenesis. In some embodiments, the composition comprising the anti-Met antibody provides for a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in angiogenesis. In some embodiments, the composition comprising anti-Met antibody provides for a 10%, 20%, 30%, 40%, 50%,
  • the composition comprising the anti-Met antibody provides for a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in angiogenesis.
  • the anti-Met antibody has low angiogenesis induction.
  • the anti-Met antibody has reduced angiogenic response.
  • the anti-Met antibody has reduced angiogenic capacity.
  • the anti-Met antibody impairs and/or reduces the normal formation of blood vessels in presence of media supportive of angiogenesis.
  • the anti-Met antibody has reduced angiogenic capacity when the anti- Met antibody is compared to untreated control.
  • the anti-Met antibody has reduced angiogenic capacity as compared to a sample treated with serum containing media. In some embodiments, the anti-Met antibody attenuates an angiogenic response. In some embodiments, the anti-Met antibody reduces the angiogenic response induce by serum containing media. In some embodiments, an angiogenic response is indicated by tube formation in a cell based assay. In some embodiments, an angiogenic response is indicated by tube formation in an endothelial cell tube formation assay. In some embodiments, an angiogenic response is indicated by blood vessel formation in a CAM (Chick Chorioallantoic membrane) assay. In some embodiments, an angiogenic response is indicated by blood vessel formation in any blood vessel formation assay known in the art.
  • the anti-Met antibody composition is formulated at a pH of about pH 4.5 to about pH 8. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 4.7 to about pH 7.8. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 5.0 to about pH 7.5.
  • the anti-Met antibody composition is formulated at a pH of about pH 5.5 to about pH 7.5. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 6 to about pH 7.5.
  • the anti-Met antibody composition is formulated at a pH of about pH 4.5, about pH 5.0, about pH 5.5, about pH 6.0, about pH 6.5, about pH 7.0, about pH 7.4, about pH 8.0. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8,
  • the anti-Met antibody composition does not comprise certain components. In some embodiments, the anti-Met antibody composition does not comprise certain components found in cellular media. In some embodiments, the anti-Met antibody composition does not comprise one or more components selected from the group consisting of xenobiotic components (for example, animal serum); Phenol red; peptides and biomolecules ⁇ 3kDa; antibiotics; protein aggregates (for example, protein aggregates >200nm); cells; cell debris (cell debris do not include exosomes/ Extracellular Vesicles (EVs); for example, non-exosome, non-EV cell debris); hormones (for example, hormones include, but are not limited to insulin and/or hydrocortisone); and/or L-glutamine.
  • xenobiotic components for example, animal serum
  • Phenol red peptides and biomolecules ⁇ 3kDa
  • antibiotics protein aggregates (for example, protein aggregates >200nm)
  • cells cell debris do not include exosomes/ Extracellular Ve
  • the anti-Met antibody composition does not comprise xenobiotic components. In some embodiments, the anti-Met antibody composition does not comprise Phenol red. In some embodiments, the anti-Met antibody composition does not comprise peptides and biomolecules ⁇ 3kDa. In some embodiments, the anti-Met antibody composition does not comprise antibiotics. In some embodiments, the anti-Met antibody composition does not comprise protein aggregates (for example, protein aggregates >200nm). In some embodiments, the anti-Met antibody composition does not comprise cells. In some embodiments, the anti-Met antibody composition does not comprise cell debris (cell debris do not include exosomes/EVs; for example, non-exosome, non-EV cell debris). In some embodiments, the anti-Met antibody composition does not comprise hormones (for example, hormones include, but are not limited to insulin and/or hydrocortisone. In some embodiments, the anti-Met antibody composition does not comprise L-glutamine.
  • the anti-Met antibody further comprises mannitol, lactose, sorbitol, xylitol, sucrose, trehalose, mannose, maltose, lactose, glucose, raffmose, cellobiose, gentiobiose, isomaltose, arabinose, glucosamine, fructose, dextrose, and/or combinations thereof.
  • the anti-Met antibody further comprises phosphate.
  • the phosphate source is sodium phosphate or potassium phosphate.
  • the phosphate source is sodium phosphate.
  • the phosphate source is potassium phosphate.
  • the anti-Met antibody further comprises mono/di-sodium phosphate, mannitol, and trehalose, wherein the composition has a pH of about pH 7.4.
  • the anti-Met antibody composition can comprise one or more additional agents including but not limited to glycine, glycerol, sodium chloride, potassium chloride, and/or dextrose.
  • the anti-Met antibody composition can comprise one or more additional agents selected from the group consisting of glycine, glycerol, sodium chloride, potassium chloride, and dextrose.
  • the anti-Met antibody composition can comprise one or more additional agents selected from the group consisting of glycine and glycerol, and dextrose. In some embodiments, the anti-Met antibody composition can comprise one or more additional agents selected from the group consisting of sodium chloride and potassium chloride.
  • the anti-Met antibody composition is formulated in a buffer system.
  • the anti-Met antibody composition is formulated in a buffer system including but not limited to di/mono sodium phosphate, sodium citrate/citric acid, boric acid/sodium citrate, boric acid/sodium tetraborate, and/or citric acid/disodium phosphate.
  • the anti-Met antibody composition is formulated in a buffer system selected from the group consisting of di/mono sodium phosphate, sodium citrate/citric acid, boric acid/sodium citrate, boric acid/sodium tetraborate, and/or citric acid/disodium phosphate.
  • the anti-Met antibody composition is formulated in a di/mono sodium phosphate buffer system. In some embodiments, the anti- Met antibody composition is formulated in sodium citrate/citric acid buffer system. In some embodiments, the anti-Met antibody composition is formulated in a boric acid/sodium citrate buffer system. In some embodiments, the anti-Met antibody composition is formulated in a boric acid/sodium tetraborate buffer system. In some embodiments, the anti-Met antibody composition is formulated in a citric acid/disodium phosphate buffer system.
  • the phosphate source is sodium phosphate or potassium phosphate. In some embodiments, the phosphate source is sodium phosphate. In some embodiments, the phosphate source is potassium phosphate. In some embodiments, the anti-Met antibody composition comprises di-sodium phosphate/citric acid, mannitol, and trehalose, wherein the composition has a pH of about pH 6.4.
  • the anti-Met antibody composition further comprises a tonicity adjusting or tonicity modifying agent.
  • tonicity adjusting or tonicity modifying agent includes but is not limited to NaCl, KC1, mannitol, dextrose, sucrose, sorbitol, and/or glycerin.
  • tonicity adjusting or tonicity modifying agent is selected from the group consisting of NaCl, KC1, mannitol, dextrose, sucrose, sorbitol, and/or glycerin.
  • the anti-Met antibody composition further comprises an adhesive agent.
  • the anti-Met antibody composition further comprises an adhesive agent including but not limited to hypromellose, Poloxamer 407, Poloxamer 188, Poloxomer 237, Poloxomer 338, Hypromellose, (HPMC), HEC, polycarbophil, polyvinylpyrrolidone (PVP), PVA (polyvinyl alcohol, polyimide, sodium hyaluronate, gellan gum, poly(lactic acid-co-gly colic acid) (PLGA), polysiloxane, polyimide, carboxymethylcellulose (CMC), or hydroxypropyl methylcellulose (HPMC), hydroxy methyl cellulose, hydroxy ethyl cellulose, sodium carboxy methyl cellulose, fibrin glue, polyethyelene glycol, and GelCORE.
  • PVP polyvinylpyrrolidone
  • PVA polyvinyl alcohol
  • polyimide sodium hyaluronate
  • gellan gum poly
  • the adhesive agent is hypromellose. In some embodiments, the adhesive agent is fibrin glue. In some embodiments, the adhesive agent is a polyethyelene glycol. In some embodiments, the adhesive agent is GelCORE (see, Sani, et cil, Science Advances, Vol. 5, no. 3 (2019)).
  • the anti-Met antibody composition comprises (a) the anti-Met antibody produced by any one of the methods described herein; and (b) a polymer.
  • the anti-Met antibody compositions provided herein are in the form of a therapeutic bandage (e.g., a polymer impregnated with anti-Met antibody composition).
  • the therapeutic bandage may be configured as needed, depending on the application.
  • the bandage is in the form or a patch or is configured as mesh.
  • the anti-Met antibody compositions exhibit bio penetrance, for example, ocular penetration, comeal penetration, and/or comeal permeation. In some embodiments, the anti-Met antibody composition exhibits the ability to be absorbed by the eye. In some embodiments, the anti-Met antibody composition exhibits inherent bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits excipient-enabled bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to upregulation of the smaller factors. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of a biopreservative. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of the biopreservative benzalkonium chloride.
  • the anti-Met antibody compositions exhibit long half- life and/or have increased stability as compared to other treatments.
  • the anti-Met antibody compositions as provided herein allow for an upregulation of proteins that are allow for increased stability of the anti-Met antibody.
  • the anti-Met antibody compositions as provided herein allow for upregulating chaperone proteins to improve stability of other proteins in the anti-Met antibody.
  • the anti-Met antibody compositions exhibit ultrapotency when administered to a subject in need thereof. In some embodiments, the anti-Met antibody compositions allow for therapeutic efficacy with one drop or one administration per day.
  • the anti-Met antibody composition comprises (a) anti- Met antibody composition produced by any one of the methods described herein; and (b) a polymer.
  • the anti-Met antibody composition comprises anti-Met antibody composition which is produced as described herein and a polymer.
  • the anti-Met antibody composition comprises the anti-Met antibody composition which is produced as described herein and a polymer.
  • the polymer can be a biodegradable polymer from which the anti-Met antibody composition and/or anti-Met antibody composition components can be released.
  • the polymer enables sustained (slow) release of the anti-Met antibody composition components.
  • the anti-Met antibody compositions provided herein are in the form of a therapeutic bandage (e.g., a polymer impregnated with anti-Met antibody composition).
  • the therapeutic bandage may be configured as needed, depending on the application.
  • the bandage is in the form or a patch or is configured as mesh.
  • the anti-Met antibody compositions exhibit bio penetrance, for example, ocular penetration, comeal penetration, and/or comeal permeation. In some embodiments, the anti-Met antibody composition exhibits the ability to be absorbed by the eye. In some embodiments, the anti-Met antibody composition exhibits inherent bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits excipient-enabled bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to upregulation of the smaller factors. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of a biopreservative. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of the biopreservative benzalkonium chloride.
  • the anti-Met antibody compositions exhibit long half- life and/or have increased stability as compared to other treatments.
  • the anti-Met antibody compositions as provided herein allow for an upregulation of proteins that are allow for increased stability of the anti-Met antibody composition.
  • the anti-Met antibody compositions as provided herein allow for upregulating chaperone proteins to improve stability of other proteins in the anti-Met antibody composition.
  • the anti-Met antibody compositions exhibit ultrapotency when administered to a subject in need thereof. In some embodiments, the anti-Met antibody compositions allow for therapeutic efficacy with one drop or one administration per day.
  • the anti-Met antibody is prepared in a pharmaceutical formulation. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.1 mg - 10 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.2 mg - 9 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.3 mg - 8 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.4 mg - 7 mg per 1 mL of anti-Met antibody.
  • the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.5 mg - 6 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.6 mg - 5 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.7 mg - 4 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.8 mg - 3 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.9 mg - 2 mg per 1 mL of anti-Met antibody.
  • the anti-Met antibody is prepared in a formulation comprising about 0.1 mg - 10 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.2 mg - 9 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.3 mg - 8 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.4 mg - 7 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.5 mg - 6 mg per 1 mL of anti-Met antibody.
  • the anti-Met antibody is prepared in a formulation comprising about 0.6 mg - 5 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.7 mg - 4 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.8 mg - 3 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.9 mg - 2 mg per 1 mL of anti-Met antibody.
  • the anti-Met antibody is prepared in a formulation comprising 2 mg - 3 mg per mL of monobasic sodium phosphate. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 4% to 5% per mL of monobasic sodium phosphate. [00105] In some embodiments, the anti-Met antibody is prepared in a formulation comprising 11 mg - 12 mg per mL of dibasic sodium phosphate. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 21.5% to 23% per mL of dibasic sodium phosphate.
  • the anti-Met antibody is prepared in a formulation comprising 11.5 mg - 13 mg per mL of mannitol. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 23% to 25% per mL of mannitol.
  • the anti-Met antibody is prepared in a formulation comprising 23 mg - 25 mg per mL of trehalose dihydrate. In some embodiments, the anti- Met antibody is prepared in a formulation comprising 46% to 48% per mL of trehalose dihydrate.
  • the anti-Met antibody is prepared in a formulation that does not comprise hypromellose. In some embodiments, the anti-Met antibody is prepared in a formulation that optionally comprises hypromellose. In some embodiments, the anti- Met antibody is prepared in a formulation comprising 0.5 mg-2 mg per mL of hypromellose. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 1% to 3% per mL of hypromellose. [00109] In some embodiments, the anti-Met antibody is prepared in a formulation comprising hydrochloric acid and/or sodium hydroxide. In some embodiments, the anti-Met antibody is prepared in a formulation comprising hydrochloric acid. In some embodiments, the anti-Met antibody is prepared in a formulation comprising sodium hydroxide. In some embodiments, the hydrochloric acid and/or sodium hydroxide is employed to obtain the desired pH.
  • the anti-Met antibody is prepared in a formulation comprising the components as provided in Table 1 below
  • Table 1 anti-Met antibody formulation embodiment.
  • the anti-Met antibody of the present disclosure exhibits a variety of therapeutic properties, including for example, anti-angiogenic properties (blood vessels and/or lymphatic vessels), anti-fibrotic properties, anti-inflammatory properties, properties promoting cell migration and proliferation, mitogenic promoting properties, anti-oxidative stress/damage properties,
  • the anti-Met antibody exhibits anti-inflammatory properties. In some embodiments, the anti-Met antibody inhibits inflammation. In some embodiments, the anti-Met antibody inhibits inflammation by 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%. 90%, or 100% (e.g., complete reduction in inflammation). In some embodiments, the anti-Met antibody prevents degranulation of mast cells.
  • the anti-Met antibody promotes cell migration and proliferation, including for example, mitogenic and motogenic activities. In some embodiments, the anti-Met antibody promotes mitogenic activities. In some embodiments, the anti-Met antibody promotes motogenic activities. In some embodiments, the anti-Met antibody comprises FGF7, which provides for the cell migration and proliferation activities of the anti-Met antibody.
  • the anti-Met antibody increases cell migration and proliferation by at least 5%, at least 10%, at least 15%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%. at least 90%, or at least 100%, or more. In some embodiments, the anti-Met antibody increases cell migration and proliferation by at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, or more.
  • the anti-Met antibody provides for anti-oxidative stress and or reduction in cellular damage.
  • the anti-Met antibody comprises anti-oxidative stress and reduction in cellular damage factors.
  • the anti-oxidative stress and reduction in cellular damage factors include but are not limited to SOD-1, SOD-2, SOD-3, HO-1.
  • the anti-oxidative stress and reduction in cellular damage factor is selected from the group consisting of SOD- 1, SOD-2, SOD-3, HO-1.
  • the anti-Met antibody accelerates wound healing (shortening wound healing period). In some embodiments, the anti-Met antibody accelerates wound healing by at least 10% to at least 100% or more. In some embodiments, the anti-Met antibody accelerates wound healing by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least or more. [00117] In some embodiments, the anti-Met antibody exhibits anti-scarring properties. In some embodiments, the anti-Met antibody inhibits scar formation.
  • the anti-Met antibody inhibits scar formation by 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% (e.g., complete prevention of scar formation).
  • the present invention provides methods for characterization of the anti-Met antibody.
  • the characterization will include measuring biophysical parameters.
  • various potency assays can be performed on the anti- Met antibody as described herein.
  • the anti-Met antibody can be subjected to measuring biophysical parameters.
  • characterization assays include but are not limited to biophysical assays, biochemical assays, and bioassays.
  • characterization assays can include but are not limited to physical component characterizations, oxidative stress assays, safety analysis, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays.
  • characterization assays are selected from the group consisting of physical component characterizations, oxidative stress assays, safety analysis, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays.
  • oxidative stress prevention assays can be performed on the anti-Met antibody.
  • the anti-Met antibody prevents comeal epithelium damage.
  • the anti-Met antibody reduces the presence of inflammation.
  • the anti-Met antibody reduces the presence of inflammation as determined by an increase in the present of anti-inflammation markers.
  • the anti-Met antibody reduces the presence of inflammation as determined by an increase in the present of anti-inflammation markers, such as, for example, IL-8.
  • the anti-Met antibody can be evaluated for blood compatibility and implementing tests for sterility as well as pyrogen and endotoxin levels.
  • the anti-Met antibody can be evaluated blood compatibility.
  • evaluating blood compatibility includes assays for hemolysis and hemagglutination.
  • the anti-Met antibody does not exhibit detrimental effects with systemic exposure.
  • the anti-Met antibody does not exhibit detrimental effects with systemic exposure, such as with severe ocular bums.
  • the anti-Met antibody does not exhibit hemagglutination activity.
  • the anti-Met antibody does not induce hemolysis.
  • the anti-Met antibody does not induce hemolytic activity.
  • the biophysical characteristics of the anti-Met antibody and the composition comprising the peptide can be evaluated and/or determined.
  • the following parameters can be measured to further characterize the anti-Met antibody and the composition comprising the peptide: thermal melting, thermal aggregation, Delta G, and/or viscosity.
  • a thermal melting assay is employed to determine anti-Met antibody stability.
  • a thermal aggregation assay is employed to determine anti- Met antibody stability.
  • delta G is employed as a measure for determining anti-Met antibody stability.
  • viscosity is measured as an anti-Met antibody characteristic.
  • viscosity is to determine anti-Met antibody stability
  • biophysical metrics can be employed to establish stability parameters for characterizing different anti-Met antibody formulations.
  • the anti-Met antibody compos is stable at -20°C, 4°C, and room temperature (20°C), for at least 7 days.
  • the anti-Met antibody is stable -20°C, 4°C, and room temperature (20°C), for at least 14 days.
  • the anti-Met antibody is stable for at least 7 days, at least 1 week, at least 2 weeks, at least 3 weeks, or at least 1 month.
  • the anti-Met antibody is stable for at least 7 days, at least 14 days, at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month, at least 2 months, or at least 3 months at about -20°C.
  • the anti-Met antibody is stable for at least 7 days, at least 14 days, at least 1 week, at least 2 weeks, at least 3 weeks, or at least 1 month at about 4°C. In some embodiments, the anti-Met antibody is stable for at least 7 days, at least 14 days, at least 1 week, at least 2 weeks, at least 3 weeks, or at least 1 month at about 20°C (or room temperature).
  • the anti-Met antibody is stable for at least 7 days at about -20°C. In some embodiments, the anti-Met antibody is stable for at least 7 days at about 4°C. In some embodiments, the anti-Met antibody is stable for at least 7 days at about 20°C. In some embodiments, the anti-Met antibody is stable for at least 7 days at about 25°C (room temperature).
  • the anti-Met antibody is stable for at least 14 days at about -20°C. In some embodiments, the anti-Met antibody is stable for at least 14 days at about 4°C. In some embodiments, the anti-Met antibody is stable for at least 14 days at about 20°C (or room temperature). In some embodiments, the anti-Met antibody is stable for at least 14 days at about 25°C (room temperature).
  • the comeal epithelium more precisely, the apical surface of the epithelium has a major contribution to the overall barrier properties of the cornea and change to the comeal barrier serves as a sensitive factor for biocompatibility analysis.
  • the biophysical characteristics of the anti-Met antibody can be evaluated and/or determined such as by an epithelial barrier integrity assay.
  • the epithelial barrier integrity assay is a transepithelial electrical resistance (TEER).
  • TEER transepithelial electrical resistance
  • 3D tissues can be transferred into 24-well plates containing 2 mL of TEER buffer and incubated for 10 min.
  • TEER can be measured using an epithelial volt-ohm meter EVOMO and the EndOhm-12 chamber (World Precision, Sarasota, FL).
  • tissues can be used for tissue viability assessment using the following formula:
  • TEER can be employed to evaluate the effect on barrier integrity after topical application of the anti-Met antibody.
  • TEER can be employed to evaluate the effect on barrier integrity after topical application of the anti-Met antibody following comeal epithelial damage caused by topical exposure to nitrogen mustard (NM) utilizing the EpiComeal tissue model (MatTek Corp).
  • anti-Met antibody can be applied topically, for example at 6 pg/ml (diluted in Placebo solution), as described in Example 6.
  • EpiComeal tissues were cultured in 5 ml medium at standard culture conditions for 24h.
  • bioassays can be employed to characterize the anti-Met antibody.
  • bioassays can be related to comeal wound healing: epithelial cell migration and proliferation, stromal cell differentiation (e.g, scarring); neovascularization, and inflammation.
  • bioassays can be employed to evaluate the ability of the anti-Met antibody to mediate comeal wound healing: epithelial cell migration and proliferation, stromal cell differentiation (scarring); neovascularization; and inflammation.
  • the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to promote proliferation and migration. In some embodiments, the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to promote proliferation. In some embodiments, the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to promote migration. In some embodiments, the anti-Met antibody promotes proliferation and/or migration. In some embodiments, the anti-Met antibody promotes proliferation. In some embodiments, the anti-Met antibody promotes migration. In some embodiments, the anti-Met antibody can be evaluated use a transwell migration assay to determined proliferation promoting ability.
  • a migration assay can be employed to evaluate for the ability of the anti-Met antibody to promote migration and proliferation.
  • a migration assay can be employed to evaluate for the ability of the anti-Met antibody to promote migration and proliferation, wherein the migration assay is an in vitro wound closure assay.
  • the migration assay can include a “scratch assay” (also referred to as a “scratch wound assay”).
  • the anti-Met antibody promotes migration and this promotion of migration and proliferation is determined and/or examined utilizing a “scratch assay”.
  • a scratch assay method is based on when artificial gap, also referred to as a “scratch”, occurs on a confluent cell monolayer.
  • the artificial gap or scratch is a linear gap.
  • the artificial gap or scratch is a horizontal linear gap.
  • the artificial gap or scratch is a circular gap.
  • the artificial gap or scratch is a crosshatched gap.
  • the “scratch” can be monitored for the cells on the edge of the newly created gap migrating and/or proliferating toward the opening to close/cover the “scratch”. See, for example, Liang, C., Park, A. & Guan, J. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc 2, 329-333 (2007).)
  • the wound closure in a scratch assay is characterized as total cells migrated into the wound gap. In some embodiments, the wound closure in a scratch assay is characterized as wound closed, as a percentage. In some embodiments, the wound closure in a scratch assay is characterized as wound remaining, expressed as a percentage. In some embodiments, the wound closure in a scratch assay is characterized as size of gap. In some embodiments, the wound closure in a scratch assay is characterized as surface area of wound. In some embodiments, the wound closure in a scratch assay is characterized as time required for wound closure. In some embodiments, the wound closure in a scratch assay is characterized as rate of wound closure.
  • the wound closure in a scratch assay is characterized as EC50 from a curve generated from plotting wound closure relative to concentration of the anti-Met antibody at a given time point.
  • the migration assay can include a transwell migration assay employing comeal epithelial cells (or other cell surrogate once validation) — (e.g., wound closure) can be performed on the anti-Met antibody.
  • the anti-Met antibody promotes wound closure as determined using a transwell migration assay.
  • in vitro wound closure assays include but are not limited to a “scratch assay” (also referred to as a “scratch wound assay”) or a circular scratch wound method or circular scratch wound assay or circular wound closure assay.
  • human comeal epithelial cell proliferation assays can be performed on the anti-Met antibody. In some embodiments, human comeal epithelial cell proliferation assays are indicative of a test for wound closure properties of the anti-Met antibody. In some embodiments, the anti-Met antibody promotes wound closure as determined using a human comeal epithelial cell proliferation assay.
  • a circular scratch wound method or circular scratch wound assay or circular wound closure assay can be employed.
  • the OrisTM Cell Migration Assay platform can be employed (see. also, as described herein in Example 6).
  • an endothelial cell tube formation assay can be performed on the anti-Met antibody. In some embodiments, an endothelial cell tube formation assays can be indicative that the anti-Met antibody is not pro-angiogenic. In some embodiments, an endothelial cell tube formation assay provides a measure of the angiogenic potential of the anti-Met antibody. In some embodiments, the anti-Met antibody exhibits anti-angiogenic properties. In some embodiments, the anti-Met antibody is anti- angiogenic properties. In some embodiments, an endothelial cell tube formation assay provides the ratio of anti- angiogenesis signals and pro-angiogenesis signals.
  • an endothelial cell tube formation assay a negative result will confirm the anthpro ratio is high and will ensure the anti-Met antibody will not promote neovascularization. In some embodiments, an endothelial cell tube formation assay a negative result will confirm the anti: pro ratio is high and will ensure the anti-Met antibody will not promote CNV (choroidal neovascularization) or neovascularization in general. In some embodiments, an inhibition of TGFb induced myofibroblast differentiation assay can be performed on the anti-Met antibody. In some embodiments, an inhibition of TGFb induced myofibroblast differentiation assay can be performed on the anti-Met antibody to show that the anti-Met antibody prevents scarring.
  • the anti-Met antibody prevents scarring. In some embodiments, the anti-Met antibody prevents scarring comeal opacity. In some embodiments, the anti-Met antibody has low angiogenesis induction. In some embodiments, the anti-Met antibody has reduced angiogenic response.
  • the anti-Met antibody has reduced angiogenic capacity. In some embodiments, the anti-Met antibody impairs and/or reduces the normal formation of blood vessels in presence of media supportive of angiogenesis. In some embodiments, the anti- Met antibody has reduced angiogenic capacity when the anti-Met antibody is compared to untreated control. In some embodiments, the anti-Met antibody has reduced angiogenic capacity as compared to a sample treated to serum containing media. In some embodiments, the anti-Met antibody attenuates an angiogenic response. In some embodiments, the anti- Met antibody reduces the angiogenic response induce by serum free media. In some embodiments, an angiogenic response is indicated by tube formation in a cell based assay. In some embodiments, an angiogenic response is indicated by tube formation in an endothelial cell tube formation assay.
  • the anti-Met antibody can be evaluated for the ability to prevent differentiation and prevent scarring. In some embodiments, the anti-Met antibody prevents and/or impairs scarring. In some embodiments, the anti-Met antibody prevents scarring. In some embodiments, the anti-Met antibody reduces scarring as compared to other standard treatments. In some embodiments, the anti-Met antibody prevents and/or impairs differentiation. In some embodiments, the anti-Met antibody prevents and/or impairs myofibroblast differentiation. In some embodiments, the anti-Met antibody reduces the loss of comeal transparency. In some embodiments, the anti-Met antibody reduces the loss of comeal transparency by preventing and/or impairing myofibroblast differentiation.
  • the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to modulate factors involved in differentiation.
  • the anti-Met antibody can be evaluated the ability of the anti-Met antibody to modulate factors involved in differentiation, including but not limited to TGFB2, Collagen I, Collagen III (normally upregulated during differentiation), TFGB3, MMP-2, and MMP-9 (normally downregulated during differentiation.
  • the anti-Met antibody modulates factors selected from the group consisting of TGFB2, Collagen I, Collagen III (normally upregulated during differentiation), TFGB3, MMP-2, and MMP-9 (normally downregulated during differentiation.
  • the anti-Met antibody induces a decrease in factors upregulated during normal differentiation.
  • the anti-Met antibody induces an increase in factors downregulated during normal differentiation. In some embodiments, the anti-Met antibody induces a decrease in expression of factors such as SMA. In some embodiments, the anti-Met antibody induces a decrease in expression of factors such as SMA which is indicative of anti-Met antibody potency. Neovascularization.
  • the anti-Met antibody can be evaluated for the ability to prevent neovascularization. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces neovascularization. In some embodiments, the anti-Met antibody inhibits or does not promote neovascularization. In some embodiments, the anti- Met antibody can be evaluated for the ability to prevent angiogenesis. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces angiogenesis. In some embodiments, the anti-Met antibody inhibits angiogenesis.
  • the anti-Met antibody can be further evaluated using depletion assays.
  • the anti-Met antibody can be depleted of specified factors.
  • the anti-Met antibody can be depleted of specified factors, including for example, but not limited to TIMP1 and/or Serpin El.
  • the anti-Met antibody can be depleted of TIMP1 and/or Serpin El.
  • the anti-Met antibody can be depleted of TIMP1.
  • the anti-Met antibody can be depleted of Serpin El.
  • the anti-Met antibody can be evaluated for the ability to prevent, impair, inhibit, and/or reduce inflammation. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation. In some embodiments, the anti-Met antibody inhibits inflammation. In some embodiments, the anti-Met antibody is characterized in vitro and/or in vivo to determine the ability to prevent, impair, inhibit, and/or reduce inflammation. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation in vitro and/or in vivo. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation in vitro.
  • the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation or in vivo.
  • a tissue model can be employed to characterizing preventing, impairing, inhibiting, and/or reducing inflammation in vitro.
  • a 3D tissue model can be employed to characterizing preventing, impairing, inhibiting, and/or reducing inflammation in vitro.
  • a nitrogen mustard (NM) gas bum model can be used to evaluate preventing, impairing, inhibiting, and/or reducing inflammation in vitro.
  • a nitrogen mustard (NM) gas bum model can be used to evaluate preventing, impairing, inhibiting, and/or reducing inflammation in vitro and as a surrogate for in vivo conditions.
  • the cytokine profile in response to treatment with and/or administration of the anti-Met antibody can be determined.
  • the levels of specific cytokines can be determined.
  • the level of IL-8 can be determined.
  • the level of IL-8 expression can be reduced in tissues treated with the anti-Met antibody.
  • the level of IL-8 expression is reduced in tissues treated with the anti-Met antibody and this is indicative of preventing, impairing, inhibiting, and/or reducing inflammation.
  • the present disclosure also provides methods of treatment using the anti-Met antibody of the present disclosure.
  • the anti-Met antibody finds use in the treatment of ocular conditions.
  • the anti-Met antibody finds use in the treatment of ocular conditions, including but not limited to ocular diseases.
  • the ocular disease is associated with the ocular surface.
  • the ocular disease is associated with damaged ocular tissue and/or damaged ocular tissue indications.
  • the anti-Met antibody finds use in the treatment of ocular conditions, including accelerating wound healing.
  • the anti-Met antibody finds use in the treatment of ocular conditions, including reducing scarring.
  • the anti-Met antibody finds use in the treatment of ocular conditions, including reducing inflammation. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing inflammation and thus promoting growth. In some embodiments, the anti-Met antibody finds use in treating ocular conditions such as reducing inflammation at the ocular surface. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing neovascularization. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing neovascularization in the cornea.
  • the anti-Met antibody finds use in the treatment of ocular conditions, including dry eye treatment (including, for example, treatment of severe dry eye, including where the epithelial cells are damaged). In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as restoring the integrity to damaged ocular tissue. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as accelerating the healing of damaged ocular tissue. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as regenerating damaged ocular nerve tissue.
  • the anti-Met antibody finds use in the treatment of ocular conditions, such as regenerating damaged ocular nerve tissue associated with Persistent Comeal Epithelial Defect (PCED).
  • the anti-Met antibody finds use in the treatment of ocular conditions, such as PCED.
  • the anti-Met antibody finds use in the treatment of ocular conditions, such as inflammatory damage to the eye surface.
  • the anti-Met antibody finds use in the treatment of ocular conditions, such as for example GvHD and/or Sjogrens syndrome.
  • the anti-Met antibody finds use in the treatment of ocular conditions, such as surgical debridement.
  • the anti-Met antibody finds use in the treatment of ocular conditions, such as contact lens wear.
  • the anti-Met antibody finds use in accelerating wound healing. In some embodiments, the anti-Met antibody finds use in reducing scarring. In some embodiments, the anti-Met antibody finds use in reducing inflammation. In some embodiments, the anti-Met antibody finds use in reducing inflammation and thus promoting growth. In some embodiments, the anti-Met antibody finds use in reducing inflammation at the ocular surface. In some embodiments, the anti-Met antibody finds use in reducing neovascularization. In some embodiments, the anti-Met antibody finds use in reducing neovascularization in the cornea. In some embodiments, the anti-Met antibody finds use in the protection and repair of retinal epithelial cells and retinal ganglion cells. In some embodiments, the anti-Met antibody finds use in induction of trabecular meshwork regeneration and reduction of intraocular pressure.
  • the composition comprising the anti-Met antibody is administered for the treatment of an ocular disease.
  • treatment comprises administering to a patient in need thereof therapeutically effective amount of an anti-Met antibody composition as described herein to a patient in need thereof.
  • the anti-Met antibody is administered to a patient in need thereof in order to promote or induce ocular wound healing.
  • the anti-Met antibody is administered to a patient in need thereof in order to reduce and/or inhibit neovascularization, reduce and/or inhibit scarring, promote and/or preserve vision, and/or increasing wound closure rate (e.g., decreasing wound closure time).
  • the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit neovascularization. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit reducing scarring. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to promote and/or preserve vision. In some embodiments, the anti-Met antibody is administered to promote and/or induce closing wound faster wound closure (e.g., reduce the amount of time required for wound closure). In some embodiments, the anti-Met antibody prevents, reduces, and/or inhibits or does not promote neovascularization and reducing scarring in order to promote vision preservation.
  • the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit neovascularization and reducing scarring in order to promote vision preservation. In some embodiments, the anti-Met antibody prevents, reduces, and/or inhibits inflammation. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit inflammation.
  • the anti-Met antibody is administered for the treatment of a visual dysfunction following traumatic injury to ocular structures.
  • treatment comprises administering to a patient in need thereof a therapeutically effective amount of an anti-Met antibody composition as described herein
  • the anti-Met antibody composition is administered for the treatment of a traumatic injury of the optic nerve degeneration following concussive injury.
  • the concussive injury to the eye is selected from the group consisting of ocular contusion and blunt injury to the eye.
  • the anti- Met antibody composition is administered for the treatment of a traumatic injury of the optic nerve.
  • treatment comprises administering to a patient in need thereof a therapeutically effective amount of an anti-Met antibody as described herein.
  • the anti-Met antibody composition is administered for ameliorating optic nerve degeneration following concussive injury to the eye.
  • the method for ameliorating optic nerve degeneration comprises administering to the patient a therapeutically effective amount of an anti-Met antibody as described herein.
  • the concussive injury to the eye is selected from the group consisting of ocular contusion and blunt injury to the eye.
  • the concussive injury to the eye an ocular contusion.
  • the concussive injury to the eye a blunt injury to the eye.
  • Efficacy readouts can include a reduced in symptoms and/or decreased disease state, including for example, increased quality of life.
  • reduced in symptoms and/or decreased disease state by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy.
  • reduction in inflammation by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy.
  • a reduction in scarring by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy.
  • the disease or conditions an ocular disease or ocular condition.
  • the disease or condition is a visual dysfunction following traumatic injury to ocular structures.
  • the disease or condition is a concussive (e.g., blunt or non-blunt) injury to the eye.
  • the disease or condition is a bum, including a chemical bum to the eye.
  • the anti-Met antibody composition is administered to a particular targeted area.
  • the particular targeted area is the eye.
  • the anti-Met antibody composition is administered to a particular targeted area and is formulated so as not to spread to other surrounding areas.
  • the anti-Met antibody composition is administered to a particular targeted area and is formulated so as not to spread to other surrounding areas.
  • the anti-Met antibody composition is administered to a particular targeted area and is formulated to stay in the targeted area for at least 1 minute, at least about 2 minutes, 3 at least about minutes, at least about 4 minutes, at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 20 minutes, at least about 30 minutes, at least about 40 minutes, at least about 50 minutes, at least about 60 minutes, at least about 70 minutes, at least about 80 minutes, at least about 90 minutes, or at least about 2 hours.
  • the anti-Met antibody is administered to an affected area immediately after the wound or injury. In some embodiments, the anti-Met antibody is administered to an affected area within 15 seconds, 30 seconds, 1 minutes, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 70 minutes, 80 minutes, 90 minutes, 2 hours, 4 hours, 8 hours, 12 hours, 24 hours, 36 hours, 48 hours, or 96 hours.
  • the anti-Met antibody is administered topically. In some embodiments, the anti-Met antibody is administered by subconjunctival injection. In some embodiments, the anti-Met antibody compositions exhibit ultrapotency when administered to a subject in need thereof. In some embodiments, the anti-Met antibody is administered topically once, two, three, four, five, and/or up to six times daily. In some embodiments, the anti-Met antibody compositions allow for therapeutic efficacy with one drop or one administration per day. In some embodiments, one drop is administered 1, 2, 3, 4, 5, or 6 times per day. In some embodiments, one drop is administered at 1 hour, 2 hour, 3 hour, or 4 hour intervals.
  • one drop is administered at least once per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least twice per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 3 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 4 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks.
  • one drop is administered at least 5 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 6 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks.
  • the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 2 pg - 20 pg of anti-Met antibody per mL; ii. 2 mg - 3 mg monobasic sodium phosphate per mL; iii. 11 mg - 12 mg dibasic sodium phosphate per mL; iv. 11.5 mg - 13 mg mannitol per mL; v. 23 mg - 24 mg trehalose dihydrate; vi.
  • the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 0.004% - 0.08 % w/w of anti-Met antibody ii. 4 % - 5 % w/w monobasic sodium phosphate; iii. 21.5 % - 23 % w/w dibasic sodium phosphate; iv. 23 % - 25 % w/w mannitol; v. 46 % - 48 % w/w trehalose dehydrate; vi. 1 % - 3 % w/w hypromellose; and wherein the pH is about 4.7 to about 7.5.
  • the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 2 pg - 20 pg of anti-Met antibody per mL; ii. 2 mg - 3 mg monobasic sodium phosphate per mL; iii. 11 mg - 12 mg dibasic sodium phosphate per mL; iv. 11.5 mg - 13 mg mannitol per mL; v. 23 mg - 24 mg trehalose dihydrate; vi. 0.5 mg - 2 mg optionally hypromellose per mL; and wherein the pH is about 4.7 to about 7.5.
  • the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 0.004% - 0.08 % w/w of anti-Met antibody ii. 4 % - 5 % w/w monobasic sodium phosphate; iii. 21.5 % - 23 % w/w dibasic sodium phosphate; iv. 23 % - 25 % w/w mannitol; v. 46 % - 48 % w/w trehalose dehydrate; vi.
  • the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 2 pg - 20 pg of anti-Met antibody per mL; ii. 2 mg - 3 mg monobasic sodium phosphate per mL; iii. 11 mg - 12 mg dibasic sodium phosphate per mL; iv. 11.5 mg - 13 mg mannitol per mL; v. 23 mg - 24 mg trehalose dihydrate; and wherein the pH is about 4.7 to about 7.5.
  • the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 0.004% - 0.08 % w/w of anti-Met antibody ii. 4 % - 5 % w/w monobasic sodium phosphate; iii. 21.5 % - 23 % w/w dibasic sodium phosphate; iv. 23 % - 25 % w/w mannitol; v. 46 % - 48 % w/w trehalose dehydrate; and wherein the pH is about 4.7 to about 7.5.
  • a kit can include an anti-Met antibody in a container or the conditioned media for use in preparing an anti-Met antibody, also in a container, as disclosed herein, and instructions for use. Additionally, a kit can include components for mixing to prepare a solution for use in an ocular treatment, and instructions for mixing and use.
  • the container can include at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which an anti-Met antibody in a container or the conditioned media for use in preparing an anti-Met antibody, and in some instances, suitably aliquoted.
  • the kit can contain additional containers into which this component may be placed.
  • Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
  • Containers and/or kits can include labeling with instructions for use and/or warnings.
  • kits comprising a panel of tests and/or assays for characterizing an anti-Met antibody, wherein the panel comprises at least two characterization assays, wherein characterization assays are selected from the group consisting of physical component characterizations, oxidative stress assays, safety analyses, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays.
  • the panel of tests and/or assays identifies an anti-Met antibody as described herein.
  • kits comprising a panel of tests and/or assays for determining consistency between anti-Met antibody lots, wherein the panel comprises one or more characterization assays, wherein characterization assays are selected from the group consisting of physical component characterizations, oxidative stress assays, safety analyses, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays.
  • the panel of tests and/or assays identifies an anti- Met antibody as described herein.
  • HGF/Met axis is known to play a prominent role in comeal wound healing.
  • activation of the pathway through the Met receptor has shown to accelerate wound healing, with potential for anti-scarring effects as well.
  • EXAMPLE 2 Anti-Met monoclonal antibody expression constructs and cloning strategy
  • Expression System & Vector Mammalian, Expression Vector pcDNA3.4 [00175] Host cell line: Expi293F
  • Antibody was obtained from supernatant of cell culture, one-step purification by HiTrap MabSelect SuRe
  • Endotoxin Level 0.1 EU/mg(LAL Endotoxin Assay Kit, GenScript, Cat.No.L00350)
  • Sterility Sterilized via a 0.22 pm filter and packaged aseptically
  • Endotoxin Level 0.1 EU/mg
  • Buffer PBS pH 7.2
  • Figures 3A-3G illustrate the exemplary amino acid sequences and nucleic acid sequences ofHRll.
  • Expi293F cells were grown in serum-free Expi293TM Expression Medium (Thermo Fisher Scientific).
  • DNA and ExpiFectamineTM 293 reagent were mixed at an optimal ratio and then added into the flask with cells ready for transfection.
  • the recombinant plasmids encoding target antibody were transiently co transfected into suspension Expi293F cell cultures.
  • the eluted fractions were pooled and buffer exchanged to the final formulation buffer.
  • the purified protein was analyzed by SDS-PAGE, Western blot analysis to determine the molecular weight and purity.
  • the concentration was determined by A280 method.
  • Met mAb (HR11) peptide promotes comeal wound healing with reduced scarring.
  • a 3.0 mm epithelial defect was created in mouse corneas using a trephine. Wounds were treated with HR11 twice daily at 2.0 mg/mL for seven days. Depicted are representative images of eyes treated with HR11 or vehicle control ( Figure 4).
  • HR11 mAb promotes comeal wound healing. A 3.0 mm epithelial defect was created in mouse corneas. Wounds were treated with HR11 twice daily at 3.0 mg/mL for seven days ( Figure 6).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Ophthalmology & Optometry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present application provides anti-Met antibodies, or antigen-binding proteins, pharmaceutical compositions for wound healing or tissue regeneration that includes the anti- Met antibodies or antigen-binding proteins, as well as methods and processes for making and using such antibodies described herein.

Description

COMPOSITIONS COMPRISING C-MET AGONIST ANTIBODIES AND METHODS
FOR USE IN OCULAR TREATMENT
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to U.S. Application No. 63/165,678 filed March 24, 2021, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.
REFERENCE TO SEQUENCE LISTING FILED ELECTRONICALLY
[0002] The sequence listing contained in the file named “121785-5007- WO_ST25.txt”, created on March 21, 2022, and having a size of 36.0 kilobytes, has been submitted electronically herewith via EFS, and the contents of the txt file are hereby incorporated by reference in their entirety.
BACKGROUND OF THE INVENTION
[0003] Blast and blunt injuries to the eye can cause a series of mechanical disruptions to the ocular contents including commotio retinae, traumatic cataract, disruption of the zonular attachments to the lens, angle recession, iris dialysis, and rupture of the pupillary sphincter. Treatment of these injuries has been limited to mechanical repair (when possible) of the iris, replacement of the crystalline lens with plastic lens implants, and repair of retinal detachments. There has been no treatment to repair the cellular architecture of the retina or the anterior chamber. Furthermore, traumatic optic neuropathy and optic nerve avulsion are among the six leading types of ocular injury that required specialized ophthalmic care during Operation Iraqi Freedom (Cho and Savitsky, “Ocular Trauma Chapter 7”, in Combat Casualty Care: Lessons learned from Oef and Oif, by Brian Eastbridge and Eric Savitsky, pp. 299-342, Ft. Detrick, Md.: Borden Institute (US) Government Printing Office, 2012), incorporated herein by reference in its entirety. Sixty percent of traumatic head injuries result in neuro-ophthalmic abnormalities (Van Stavem, el al. , J Neuro-Ophthamol 21(2): 112-117, 2001) (incorporated herein by reference in its entirety) half of which involve the optic nerves or visual pathways. Traumatic injury to neurons results in axonal damage and irreversible neuronal loss resulting in permanent deficits. While a number of potential neuroprotective therapies have been identified in animals, these single agents have generally failed to translate to therapies in human clinical trials (Turner, et al., J Neurosurg 118(5):1072-1085, 2013, incorporated herein by reference in its entirety). Combination therapies that affect several cellular targets are likely needed to prevent neuronal damage.
[0004] The cornea serves a protective role as the outermost tissue of the eye, however it is highly vulnerable to severe injury and disease. Its lack of blood vessels enables its transparency but also limits its ability to heal. Comeal injury, due to its potential to cause irreversible blindness, requires prompt intervention and aggressive treatment. The critical need for improved ocular surface healing therapies is particularly apparent for chemical bums and in severe comeal diseases, such as ocular manifestations of acute Chronic Graft v. Host Disease (GvHD), Stevens-Johnson Syndrome, Ocular Mucous Membrane Pemphigoid and other conditions giving rise to persistent comeal epithelial defect, which collectively comprise an incidence of over 100,000 cases per year. (See, Dietrich-Ntoukas etal. Cornea. 2012, 31(3):299-310; Stevenson W, et cil, Clin Ophthalmol. 2013, 7:2153- 2158; White KD, et al., J Allergy Clin Immunol Pract. 2018;6(l):38-69; Tauber J. (2002) Autoimmune Diseases Affecting the Ocular Surface. In: Ocular Surface Disease Medical and Surgical Management. Springer, New York, NY.; and Wirostko B, et al, Ocul Surf. 2015 Jul; 13(3): 204-21; and Haring, RS., et al., JAMA Ophthalmol. 2016 Oct 1; 134(10): 1119-1124.)
[0005] Moreover, topical ophthalmic drug development is impeded by many anatomical constraints including tear turnover and dilution, nasolacrimal drainage, and reflex blinking with often less than 5% of the topically administered dose reaching deeper ocular tissues (Gaudana et al, Pharm Res. 26(5): 1197-216 (2009)). In the case of comeal wounds, the initial insult causes rifts in the comeal epithelium thereby enabling the passage of topically applied MSC-S to penetrate the epithelial layers.
[0006] Accordingly, there is a large unmet need in the art for ocular therapies that can target the eye and deliver a therapeutic payload to difficult-to-reach sensory tissue which may have degenerated due to inflammation secondary to trauma (such as for example, bums, acute inflammation, age, and/or oxidative stress). The present invention meets this need by providing anti-Met antibodies, or an antigen-binding proteins thereof, pharmaceutical compositions for wound healing or tissue regeneration that includes the anti-Met antibodies or antigen-binding proteins thereof, as well as methods for making such compositions. BRIEF SUMMARY OF THE INVENTION
[0007] In one aspect, the provide invention provides a composition comprising an anti-Met antibody, wherein the antibody comprises: a) a vhCDRl, vhCDR2, and vhCDR3 from SEQ ID NO:5; and b) a vlCDRl, vlCDR2, and vlCDR3 from SEQ ID NO: 13.
[0008] In some embodiments, the antibody comprises a heavy chain variable domain having at least 80% identity to SEQ ID NO:5 and a light chain variable domain having at least 80% identity to SEQ ID NO: 13. [0009] In some embodiments, the antibody comprises: a) a vhCDRl having 0-2 amino acid substitutions from the vhCDRl from SEQ ID NO:5; b) a vhCDR2 having 0-2 amino acid substitutions from the vhCDR2 from SEQ ID NO:5; c) a vhCDR3 having 0-2 amino acid substitutions from the vhCDR3 from SEQ ID
NO:5; d) a vlCDRl having 0-2 amino acid substitutions from the vlCDRl from SEQ ID NO:13 e) a vlCDR2 having 0-2 amino acid substitutions from the vlCDR2 from SEQ ID NO: 13; and f) a vlCDR3 having 0-2 amino acid substitutions from the vlCDR3 from SEQ ID NO:13.
[0010] In some embodiments, the antibody comprises a heavy chain variable domain comprising SEQ ID NO:5 and a light chain variable domain comprising SEQ ID NO: 13. [0011] In some embodiments, the antibody comprises a CHl-hinge-CH2-CH3 region from human IgGl, IgG2, IgG3, or IgG4.
[0012] In some embodiments, the antibody comprises a CHl-hinge-CH2-CH3 region from human IgGl . [0013] In some embodiments, the antibody comprises a CL region of human kappa constant.
[0014] In some embodiments, the heavy chain comprises SEQ ID NO: 1 and the light chain comprises SEQ ID NO:9. [0015] In some embodiments, the heavy chain further comprising a first signal peptide sequence of SEQ ID NO:3 and the light chain further comprising a second signal peptide sequence of SEQ ID NO: 11.
[0016] In another aspect, the present invention provides a nucleic composition comprising: a) a first nucleic acid encoding the heavy chain variable domain of the anti-Met antibody; and b) a second nucleic acid encoding the light chain variable domain of the same anti-Met antibody.
[0017] In another aspect, the present invention provides an expression vector composition comprising: a) a first expression vector comprising the first nucleic acid; and b) a second expression vector comprising the second nucleic acid.
[0018] In another aspect, the present invention provides an expression vector composition comprising an expression vector comprising the first and second nucleic acids. [0019] In another aspect, the present invention provides a host cell comprising the expression vector composition.
[0020] In another aspect, the present invention provides a method of making an anti- Met antibody comprising: a) culturing the host cell under conditions wherein the antibody is expressed; and b) recovering the anti-Met antibody.
[0021] In another aspect, the present invention provides a method of treating an ocular condition in a subject in need thereof, wherein the method comprises administering to the subject an anti-MET antibody, the antibody comprising: a) a vhCDRl, vhCDR2, and vhCDR3 from SEQ ID NO:5; and b) a vlCDRl, vlCDR2, and vlCDR3 from SEQ ID NO: 13.
[0022] In some embodiments, the antibody comprises a heavy chain variable domain having at least 80% identity to SEQ ID NO:5 and a light chain variable domain having at least 80% identity to SEQ ID NO: 13. [0023] In some embodiments, the antibody comprises: a) a vhCDRl having 0-2 amino acid substitutions from the vhCDRl from SEQ ID NO:5; b) a vhCDR2 having 0-2 amino acid substitutions from the vhCDR2 from SEQ ID NO:5; c) a vhCDR3 having 0-2 amino acid substitutions from the vhCDR3 from SEQ ID
NO:5; d) a vlCDRl having 0-2 amino acid substitutions from the vlCDRl from SEQ ID NO:13 e) a vlCDR2 having 0-2 amino acid substitutions from the vlCDR2 from SEQ ID NO:13; and f) a vlCDR3 having 0-2 amino acid substitutions from the vlCDR3 from SEQ ID NO:13.
[0024] In some embodiments, the antibody used for such a treatment comprises a heavy chain variable domain comprising SEQ ID NO:5 and a light chain variable domain comprising SEQ ID NO: 13.
[0025] In some embodiments, the antibody used for such a treatment comprises a CHl-hinge-CH2-CH3 region from human IgGl, IgG2, IgG3, or IgG4.
[0026] In some embodiments, the antibody used for such a treatment comprises a CHl-hinge-CH2-CH3 region from human IgGl. [0027] In some embodiments, the antibody used for such a treatment comprises a CL region of human kappa constant.
[0028] In some embodiments, the heavy chain of the antibody used for such a treatment comprises SEQ ID NO: 1 and the light chain comprises SEQ ID NO:9. [0029] In some embodiments, the heavy chain of the antibody used for such a treatment further comprising a first signal peptide sequence of SEQ ID NO:3 and the light chain further comprising a second signal peptide sequence of SEQ ID NO: 11.
[0030] In some embodiments, the ocular condition is selected from the group consisting of Chronic Graft v. Host Disease (GvHD), Stevens-Johnson Syndrome, Ocular Mucous Membrane Pemphigoid, Persistent Comeal Epithelial Defect (PCED), dry eye, ocular nerve tissue damage, concussive injury to the eye (such as concussive injury, ocular contusion, or chemical bum), surgical debridement, and contact lens wear.
[0031] In some embodiments, the present disclosures provide for the use of the composition comprising the anti-Met antibody described herein for treating the ocular condition. In some embodiments, the present disclosures provide for the use of the composition for the manufacture of a medicament for treating the ocular condition in a subject in need thereof. BRIEF DESCRIPTION OF THE DRAWINGS
[0001] FIGURE 1. Schematic diagram of an exemplary embodiment of anti-Met antibody composition preparation, processing, and use.
[0002] FIGURE 2: SDS-PAGE and western blot analysis of HR11. Lane Ml : Protein Marker, TaKaRa, Cat. No. 3452. Lane M2: Protein Marker, GenScript, Cat. No. M00521. Lane 1: Reducing condition. Lane 2: Non-reducing condition. Lane P: Human IgGl, Kappa (Sigma, Cat.No.15154) as a positive control. Primary antibody: Goat Anti-Human IgG-HRP (GenScript, Cat. No. A00166). Primary antibody: Goat Anti-Human Kappa-HRP (SouthemBiotech, Cat. No. 2060-05).
[0003] FIGURE 3A-3G: Exemplary amino acid and nucleic acid sequences of HR11.
[0004] FIGURE 4: Mechanical wound efficacy of anti-Met monoclonal antibody (HR11) - depicted are representative images of eyes treated with HR11 or vehicle control.
[0005] FIGURE 5: Mechanical wound efficacy of anti-Met monoclonal antibody (HR11) - percentage of wounds completely closed. [0006] FIGURE 6: Mechanical wound efficacy of anti-Met monoclonal antibody (HR11) - HR11 mAh promotes comeal wound healing. A 3.0 mm epithelial defect was created in mouse corneas. Wounds were treated with HR11 twice daily at 3.0 mg/mL for seven days.
DETAILED DESCRIPTION OF THE INVENTION
I. INTRODUCTION
[0007] c-Met is a typical receptor tyrosine kinase (RTK) present on a cell surface that induces intracellular signal transmission by binding with its ligand hepatocyte growth factor (HGF), thereby facilitating cell growth. Studies conducted so far have revealed that c-Met is found in various human tissues of damaged organs, including the liver, lung, kidney, heart, intestinal mucosa, and skin, and thus is involved in post-damage regeneration of such tissues. c-Met may facilitate liver tissue regeneration after a hepatectomy or damage from liver cancer or cirrhosis; kidney tissue regeneration after simple or partial kidney resection from cancer, infection, renal stones, or renal artery stricture; skin tissue regeneration in patients with skin damage from bums, bedsores, or skin ulcers; and heart tissue regeneration after damage from cardiac infarction.
[0008] c-Met is involved in a variety of mechanisms, for example, cancer occurrence, metastasis, cancer cell migration and invasion and angiogenesis, and in the growth of a variety of cells. Further to the ability to facilitate regeneration and growth of normal tissue cells such as liver, kidney and heart cells, c-Met is known to facilitate growth and proliferation of stem cells when bound to a growth factor HGF.
A. DEFINITIONS
[0009] Terms used in the claims and specification are defined as set forth below unless otherwise specified. In the case of direct conflict with a term used in a parent provisional patent application, the term used in the instant specification shall control.
[0010] The terms “c-Met”, “c-Met protein", “Met”, or “Met protein” as used interchangeably herein refers to a receptor tyrosine kinase (RTK) that binds to a hepatocyte growth factor (HGF). The c-Met protein may include, for example, polypeptides encoded by nucleotide sequences of GenBank Accession Number NM.sub.— 000245, proteins encoded by polypeptide sequences of GenBank Accession Number NM.sub.— 000236, or extracellular domains thereof. The RTK c-Met is involved in a variety of mechanisms, for example, cancer occurrence, metastasis, cancer cell migration and invasion, angiogenesis, cell migration, and cell proliferation.
[0011] By “antigen binding domain” or “ABD” herein is meant a set of six Complementary Determining Regions (CDRs) that, when present as part of a polypeptide sequence, specifically binds a target antigen as discussed herein. Thus, a “Met antigen binding domain” binds Met antigen as outlined herein. As is known in the art, these CDRs are generally present as a first set of variable heavy CDRs (vhCDRs or VHCDRS) and a second set of variable light CDRs (vlCDRs or VLCDRS), each comprising three CDRs: vhCDRl, vhCDR2, vhCDR3 for the heavy chain and vlCDRl, vlCDR2 and vlCDR3 for the light. The CDRs are present in the variable heavy and variable light domains, respectively, and together form an Fv region. Thus, in some cases, the six CDRs of the antigen binding domain are contributed by a variable heavy and variable light chain. In a “Fab” format, the set of 6 CDRs are contributed by two different polypeptide sequences, the variable heavy domain (vh or VH; containing the vhCDRl, vhCDR2 and vhCDR3) and the variable light domain (vl or VL; containing the vlCDRl, vlCDR2 and vlCDR3), with the C-terminus of the vh domain being attached to the N-terminus of the CHI domain of the heavy chain and the C-terminus of the vl domain being attached to the N-terminus of the constant light domain (and thus forming the light chain). “Antigen-binding protein” as used herein refers to a protein that comprises an antigen binding domain.
[0012] The term “human antibody” includes all antibodies that have one or more variable and constant regions derived from human immunoglobulin sequences. In one embodiment, all of the variable and constant domains are derived from human immunoglobulin sequences (a fully human antibody). Human antibodies may be prepared in a variety of ways, including immunization of a mouse that is genetically modified to express human antibodies. One can engineer mouse strains deficient in mouse antibody production with large fragments of the human Ig loci in anticipation that such mice would produce human antibodies in the absence of mouse antibodies. Large human Ig fragments may preserve the large variable gene diversity as well as the proper regulation of antibody production and expression. By exploiting the mouse machinery for antibody diversification and selection and the lack of immunological tolerance to human proteins, the reproduced human antibody repertoire in these mouse strains may yield high affinity fully human antibodies against any antigen of interest, including human antigens. Using the hybridoma technology, antigen-specific human MAbs with the desired specificity may be produced and selected. Human antibodies can also be prepared by panning human antibody libraries expressed on phage, phagemids, ribosomes, or other particles.
[0013] A “humanized antibody” has a sequence that differs from the sequence of an antibody derived from a non-human species by one or more amino acid substitutions, deletions, and/or additions, such that the humanized antibody is less likely to induce an immune response, and/or induces a less severe immune response, as compared to the non human species antibody, when it is administered to a human subject. In one embodiment, certain amino acids in the framework and constant domains of the heavy and/or light chains of the non-human species antibody are mutated to produce the humanized antibody. In another embodiment, the constant domain(s) from a human antibody are fused to the variable domain(s) of anon-human species. In another embodiment, one or more amino acid residues in one or more CDR sequences of a non-human antibody are changed to reduce the likely immunogenicity of the non-human antibody when it is administered to a human subject, wherein the changed amino acid residues either are not critical for immunospecific binding of the antibody to its antigen, or the changes to the amino acid sequence that are made are conservative changes, such that the binding of the humanized antibody to the antigen is not significantly worse than the binding of the non-human antibody to the antigen. Examples of methods for making humanized antibodies may be found in U.S. Pat. Nos. 6,054,297, 5,886,152 and 5,877,293, the disclosures of each of which are incorporated by reference herein.
[0014] The term “chimeric antibody” refers to an antibody that contains one or more regions from one antibody and one or more regions from one or more other antibodies. A “CDR grafted antibody” is an antibody comprising one or more CDRs derived from an antibody of a particular species or isotype and the framework of another antibody of the same or different species or isotype.
[0015] By "modification" herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence or an alteration to a moiety chemically linked to a protein. For example, a modification may be an altered carbohydrate or PEG structure attached to a protein. By "amino acid modification" herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence. For clarity, unless otherwise noted, the amino acid modification is always to an amino acid coded for by DNA, e.g. the 20 amino acids that have codons in DNA and RNA. [0016] By "amino acid substitution" or "substitution" herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with a different amino acid. In particular, in some embodiments, the substitution is to an amino acid that is not naturally occurring at the particular position, either not naturally occurring within the organism or in any organism. For example, the substitution N297A refers to a variant polypeptide, in this case an Fc variant, in which the asparagine at position 297 is replaced with alanine. For clarity, a protein which has been engineered to change the nucleic acid coding sequence but not change the starting amino acid (for example exchanging CGG (encoding arginine) to CGA (still encoding arginine) to increase host organism expression levels) is not an “amino acid substitution”; that is, despite the creation of a new gene encoding the same protein, if the protein has the same amino acid at the particular position that it started with, it is not an amino acid substitution.
[0017] By "amino acid insertion" or "insertion" as used herein is meant the addition of an amino acid sequence at a particular position in a parent polypeptide sequence. For example, -233E or 233E designates an insertion of glutamic acid after position 233 and before position 234. Additionally, -233ADE or A233ADE designates an insertion of AlaAspGlu after position 233 and before position 234.
[0018] By "amino acid deletion" or "deletion" as used herein is meant the removal of an amino acid sequence at a particular position in a parent polypeptide sequence. For example, E233- or E233#, E233() or E233del designates a deletion of glutamic acid at position 233. Additionally, EDA233- or EDA233# designates a deletion of the sequence GluAspAla that begins at position 233.
[0019] By "variant protein" or "protein variant", or "variant" as used herein is meant a protein that differs from that of a parent protein by virtue of at least one amino acid modification. Protein variant may refer to the protein itself, a composition comprising the protein, or the amino sequence that encodes it. In some embodiments, the protein variant has at least one amino acid modification compared to the parent protein, e.g. from about one to about seventy amino acid modifications, in one exemplary embodiment from about one to about five amino acid modifications compared to the parent. As described below, in some embodiments the parent polypeptide, for example an Fc parent polypeptide, is a human wild type sequence, such as the Fc region from IgGl, IgG2, IgG3 or IgG4, although human sequences with variants can also serve as “parent polypeptides”. Exemplary protein variant sequences herein will possess at least about 80% identity with a parent protein sequence, in some embodiments at least about 90% identity, in some embodiments at least about 95-98- 99% identity. Variant protein can refer to the variant protein itself, compositions comprising the protein variant, or the DNA sequence that encodes it. Accordingly, by "antibody variant" or "variant antibody" as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification, "IgG variant" or "variant IgG" as used herein is meant an antibody that differs from a parent IgG (again, in many cases, from a human IgG sequence) by virtue of at least one amino acid modification, and "immunoglobulin variant" or "variant immunoglobulin" as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification. "Fc variant" or "variant Fc" as used herein is meant a protein comprising an amino acid modification in an Fc domain. The Fc variants of the present invention are defined according to the amino acid modifications that compose them. The EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference.) The modification can be an addition, deletion, or substitution. Substitutions can include naturally occurring amino acids and, in some cases, synthetic amino acids. Examples include U.S. Pat. No. 6,586,207; WO 98/48032; WO 03/073238; US2004-0214988A1; WO 05/35727A2; WO 05/74524A2; J. W. Chin et al., (2002), Journal of the American Chemical Society 124:9026-9027; J. W. Chin, & P. G. Schultz, (2002), ChemBioChem 11:1135-1137; J. W. Chin, et al., (2002), PICAS United States of America 99:11020-11024; and, L. Wang, & P. G. Schultz, (2002), Chem. 1-10, all entirely incorporated by reference.
[0020] As used herein, "protein" herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides. The peptidyl group may comprise naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids (see Simon et al., PNAS USA 89(20):9367 (1992), entirely incorporated by reference). The amino acids may either be naturally occurring or synthetic (e.g. not an amino acid that is coded for by DNA); as will be appreciated by those in the art. For example, homo-phenylalanine, citrulline, ornithine and noreleucine are considered synthetic amino acids for the purposes of the invention, and both D- and L-(R or S) configured amino acids may be utilized. The variants of the present invention may comprise modifications that include the use of synthetic amino acids incorporated using, for example, the technologies developed by Schultz and colleagues, including but not limited to methods described by Cropp & Shultz, 2004, Trends Genet. 20(12):625-30, Anderson et al., 2004, Proc Natl Acad Sci USA 101 (2):7566-71, Zhang et al., 2003, 303(5656):371-3, and Chin et al., 2003, Science 301(5635):964-7, all entirely incorporated by reference. In addition, polypeptides may include synthetic derivatization of one or more side chains or termini, glycosylation, PEGylation, circular permutation, cyclization, linkers to other molecules, fusion to proteins or protein domains, and addition of peptide tags or labels.
[0021] By "residue" as used herein is meant a position in a protein and its associated amino acid identity. For example, Asparagine 297 (also referred to as Asn297 or N297) is a residue at position 297 in the human antibody IgGl .
[0022] By "Fab" or "Fab region" as used herein is meant the polypeptide that comprises the VH, CHI, VL, and CL immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody or antibody fragment.
[0023] By "Fv" or "Fv fragment" or "Fv region" as used herein is meant a polypeptide that comprises the VL and VH domains of a single antibody. As will be appreciated by those in the art, these generally are made up of two chains.
[0024] By “single chain Fv” or “scFv” herein is meant a variable heavy domain covalently attached to a variable light domain, generally using a scFv linker as discussed herein, to form a scFv or scFv domain. A scFv domain can be in either orientation from N- to C-terminus (vh-linker-vl or vl-linker-vh). In general, the linker is a scFv linker as is generally known in the art, with the linker peptide predominantly including the following amino acid residues: Gly, Ser, Ala, or Thr. The linker peptide should have a length that is adequate to link two molecules in such a way that they assume the correct conformation relative to one another so that they retain the desired activity. In one embodiment, the linker is from about 1 to 50 amino acids in length, in some embodiments about 1 to 30 amino acids in length. In one embodiment, linkers of 1 to 20 amino acids in length may be used, with from about 5 to about 10 amino acids finding use in some embodiments. Useful linkers include glycine-serine polymers, including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least one (and generally from 3 to 4), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers. Alternatively, a variety of nonproteinaceous polymers, including but not limited to polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol, may find use as linkers, that is may find use as linkers.
[0025] By "IgG subclass modification" or “isotype modification” as used herein is meant an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype. For example, because IgGl comprises a tyrosine and IgG2 a phenylalanine at EU position 296, a F296Y substitution in IgG2 is considered an IgG subclass modification. Similarly, because IgGl has a proline at position 241 and IgG4 has a serine there, an IgG4 molecule with a S241P is considered an IgG subclass modification. Note that subclass modifications are considered amino acid substitutions herein.
[0026] By "amino acid" and "amino acid identity" as used herein is meant one of the 20 naturally occurring amino acids that are coded for by DNA and RNA.
[0027] By "effector function" as used herein is meant a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to ADCC, ADCP, and CDC. In many cases, it is desirable to ablate most or all effector functions using either different IgG isotypes (e.g. IgG4) or amino acid substitutions in the Fc domain; however, preserving binding to the FcRn receptor is desirable, as this contributes to the half-life of the antibodies in human serum.
[0028] By "IgG Fc ligand" as used herein is meant a molecule, for example a polypeptide, from any organism that binds to the Fc region of an IgG antibody to form an Fc/Fc ligand complex. Fc ligands include but are not limited to FcyRIs, FcyRIIs, FcyRIIIs, FcRn, Clq, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcyR. Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcyRs (Davis et ak, 2002, Immunological Reviews 190:123-136, entirely incorporated by reference). Fc ligands may include undiscovered molecules that bind Fc. Particular IgG Fc ligands are FcRn and Fc gamma receptors. By "Fc ligand" as used herein is meant a molecule, for example a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc/Fc ligand complex.
[0029] By "parent polypeptide" as used herein is meant a starting polypeptide that is subsequently modified to generate a variant. The parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide. Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it. Accordingly, by "parent immunoglobulin" as used herein is meant an unmodified immunoglobulin polypeptide that is modified to generate a variant, and by "parent antibody" as used herein is meant an unmodified antibody that is modified to generate a variant antibody. It should be noted that "parent antibody" includes known commercial, recombinantly produced antibodies as outlined below.
[0030] By “Fc” or “Fc region” or “Fc domain” as used herein is meant the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain and in some cases, part of the hinge. Thus Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains. For IgA and IgM, Fc may include the J chain. For IgG, the Fc domain comprises immunoglobulin domains Cy2 and Cy3 (Cy2 and Cy3) and the lower hinge region between Cyl (Cyl) and Cy2 (Cy2). Although the boundaries of the Fc region may vary, the human IgG heavy chain Fc region is usually defined to include residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat. In some embodiments, as is more fully described below, amino acid modifications are made to the Fc region, for example to alter binding to one or more FcyR receptors or to the FcRn receptor.
[0031] By “heavy constant region” herein is meant the CHl-hinge-CH2-CH3 portion of an antibody.
[0032] By "position" as used herein is meant a location in the sequence of a protein. Positions may be numbered sequentially, or according to an established format, for example the EU index for antibody numbering.
[0033] By "target antigen" as used herein is meant the molecule that is bound specifically by the variable region of a given antibody. The target antigen of interest herein is Met, usually human Met and optionally cyno Met, the sequences of which are shown in .
[0034] By "target cell" as used herein is meant a cell that expresses a target antigen.
[0035] By "variable region" as used herein is meant the region of an immunoglobulin that comprises one or more Ig domains substantially encoded by any of the VK (V. kappa), nl (V.lamda), and/or VH genes that make up the kappa, lambda, and heavy chain immunoglobulin genetic loci respectively.
[0036] By "wild type or WT" herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations. A WT protein has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
[0037] The antibodies of the present invention are generally isolated or recombinant. “Isolated,” when used to describe the various polypeptides disclosed herein, means a polypeptide that has been identified and separated and/or recovered from a cell or cell culture from which it was expressed. Ordinarily, an isolated polypeptide will be prepared by at least one purification step. An “isolated antibody,” refers to an antibody which is substantially free of other antibodies having different antigenic specificities. The term “recombinant,” as applied to a polynucleotide means the polynucleotide is the product of various combinations of cloning, restriction or ligation steps, and other procedures resulting in a construct distinct and/or different from a polynucleotide found in nature. The terms respectively include replicates of the original polynucleotide construct and progeny of the original virus construct. The term in vivo ” refers to an event that takes place in a subject's body.
[0038] The term “synthetic antibody” as used herein includes an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage. The term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using synthetic DNA or amino acid sequence technology which is available and well known in the art.
[0039] The term in vitro ” refers to an event that takes places outside of a subject's body. In vitro assays encompass cell-based assays in which cells alive or dead are employed and may also encompass a cell-free assay in which no intact-cells are employed.
[0040] “Specific binding” or “specifically binds to” or is “specific for” a particular antigen or an epitope means binding that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. For example, specific binding can be determined by competition with a control molecule that is similar to the target.
[0041] Specific binding for a particular antigen or an epitope can be exhibited, for example, by an antibody having a KD for an antigen or epitope of at least about 109 M, at least about 1010 M, at least about 1011 M, at least about 10 12 M, at least about 10 13 M, at least about 1014 M, at least about 1015 M, where KD refers to a dissociation rate of a particular antibody-antigen interaction. Typically, an antibody that specifically binds an antigen will have a KD that is 20-, 50-, 100-, 500-, 1000-, 5,000-, 10,000- or more times greater for a control molecule relative to the antigen or epitope.
[0042] Also, specific binding for a particular antigen or an epitope can be exhibited, for example, by an antibody having a KA or Ka for an antigen or epitope of at least 20-, 50-, 100-, 500-, 1000-, 5,000-, 10,000- or more times greater for the epitope relative to a control, where KA or Ka refers to an association rate of a particular antibody-antigen interaction. Binding affinity is generally measured using surface plasmon resonance (e.g. Biacore assay) and flow cytometry with antigen-expressing cells.
[0043]
[0044] The terms “polypeptide”, “protein” and “peptide”, as used herein, refer to any polymer formed from multiple amino acids, regardless of length or posttranslational modification (e.g., phosphorylation or glycosylation), associated, at least in part, by covalent bonding (e.g., “protein” as used herein refers both to linear polymers (chains) of amino acids associated by peptide bonds as well as proteins exhibiting secondary, tertiary, or quaternary structure, which can include other forms of intramolecular and intermolecular association, such as hydrogen and van der Waals bonds, within or between peptide chain(s)). Examples of polypeptides include, but are not limited to, proteins, peptides, oligopeptides, dimers, multimers, variants, and the like. In some embodiments, the polypeptide can be unmodified such that it lacks modifications such as phosphorylation and glycosylation. A polypeptide can contain part or all of a single naturally-occurring polypeptide, or can be a fusion or chimeric polypeptide containing amino acid sequences from two or more naturally-occurring polypeptides.
[0001] By “isolated polypeptide” or “purified polypeptide” is meant a polypeptide that is substantially free from the materials with which the polypeptide is normally associated in nature or in culture. The polypeptides of the invention can be obtained, for example, by extraction from a natural source if available (for example, a mammalian cell), by expression of a recombinant nucleic acid encoding the polypeptide (for example, in a cell or in a cell-free translation system), or by chemically synthesizing the polypeptide. In addition, polypeptide may be obtained by cleaving full length polypeptides. When the polypeptide is a fragment of a larger naturally occurring polypeptide, the isolated polypeptide is shorter than and excludes the full-length, naturally-occurring polypeptide of which it is a fragment.
[0002] The terms “sequence identity,” “percent identity,” and “sequence percent identity” (or synonyms thereof, e.g., “99% identical”) in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences. Suitable programs to determine percent sequence identity include for example the BLAST suite of programs available from the U.S. Government’s National Center for Biotechnology Information BLAST web site. Comparisons between two sequences can be carried using either the BLASTN or BLASTP algorithm. BLASTN is used to compare nucleic acid sequences, while BLASTP is used to compare amino acid sequences. ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or MegAlign, available from DNASTAR, are additional publicly available software programs that can be used to align sequences. One skilled in the art can determine appropriate parameters for maximal alignment by particular alignment software. In certain embodiments, the default parameters of the alignment software are used.
[0003] As used herein “isolated” refers to material removed from its original environment and is thus altered “by the hand of man” from its natural state. [0004] As used herein, “enriched” means to selectively concentrate or to increase the amount of one or more materials by elimination of the unwanted materials or selection and separation of desirable materials from a mixture (e.g., separate cells with specific cell markers from a heterogeneous cell population in which not all cells in the population express the marker). [0005] As used herein, the term “substantially purified” means a population of cells substantially homogeneous for a particular marker or combination of markers. By substantially homogeneous is meant at least 90%, and in some embodiments 95% homogeneous for a particular marker or combination of markers. As used herein, the term “multipotent stem cells” are true stem cells but can only differentiate into a limited number of types. For example, the bone marrow contains multipotent stem cells that give rise to all the cells of the blood but may not be able to differentiate into other cells types.
[0006] By the term “animal-free” when referring to certain compositions, growth conditions, culture media, etc. described herein, is meant that no non-human animal-derived materials, such as bovine serum, proteins, lipids, carbohydrates, nucleic acids, vitamins, etc., are used in the preparation, growth, culturing, expansion, storage or formulation of the certain composition or process. By “no non-human animal-derived materials” is meant that the materials have never been in or in contact with a non-human animal body or substance so they are not xeno-contaminated. Generally, clinical grade materials, such as recombinantly produced human proteins, are used in the preparation, growth, culturing, expansion, storage and/or formulation of such compositions and/or processes.
[0007] By the term “expanded”, in reference to cell compositions, means that the cell population constitutes a significantly higher concentration of cells than is obtained using previous methods. For example, the level of cells per gram of amniotic tissue in expanded compositions of AMP cells is at least 50-fold and up to 150-fold higher than the number of cells in the primary culture after 5 passages, as compared to about a 20-fold increase in such cells using previous methods. In another example, the level of cells per gram of amniotic tissue in expanded compositions of AMP cells is at least 30-fold and up to 100- fold higher than the number of cells in the primary culture after 3 passages. Accordingly, an “expanded” population has at least a 2-fold, and up to a 10-fold, improvement in cell numbers per gram of amniotic tissue over previous methods. The term “expanded” is meant to cover only those situations in which a person has intervened to elevate the number of the cells.
[0008] As used herein, “conditioned medium” is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide support to or affect the behavior of other cells. Such factors include, but are not limited to, hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, chemokines, receptors, inhibitors and granules. The medium containing the cellular factors is the conditioned medium. Examples of methods of preparing conditioned media have been described in U.S. Pat. No. 6,372,494 which is incorporated by reference in its entirety herein. As used herein, conditioned medium also refers to components, such as proteins, that are recovered and/or purified from conditioned medium or from for example, MSC cells.
[0009] As used herein, the term “mesenchymal stem cell composition” or “MSC composition” means conditioned medium that has been derived from MSCs and in some instances has undergone further processing. In some embodiments, “anti-Met antibody composition” can refer to the crude conditioned media derived from the MSC. In some embodiments, “anti-Met antibody composition” can refer to the composition obtained from the crude conditioned media after it has been subjected to further processing as described herein.
[0010] As used herein, the term “suspension” means a liquid containing dispersed components, e.g., cytokines. The dispersed components may be fully solubilized, partially solubilized, suspended or otherwise dispersed in the liquid. Suitable liquids include, but are not limited to, water, osmotic solutions such as salt and/or sugar solutions, cell culture media, and other aqueous or non-aqueous solutions.
[0011] “Amino acid” refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxy glutamate, and O-phosphoserine. Amino acid analogs refer to compounds that have the same basic chemical structure as a naturally occurring amino acid, e.g., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid. Amino acids can be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, can be referred to by their commonly accepted single-letter codes. [0012] An “amino acid substitution” refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence (an amino acid sequence of a starting polypeptide) with a second, different “replacement” amino acid residue. An “amino acid insertion” refers to the incorporation of at least one additional 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 larger “peptide insertions,” can be made, e.g. insertion of about three to about five or even up to about ten, fifteen, or twenty 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.
[0013] “Polypeptide,” “peptide”, and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
[0014] “Nucleic acid” refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double- stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences and as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions can be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al, Nucleic Acid Res. 19:5081, 1991; Ohtsuka e/ a/. , Biol. Chem. 260:2605-2608, 1985; and Cassol et al, 1992; Rossolini et al, Mol. Cell. Probes 8:91-98, 1994). For arginine and leucine, modifications at the second base can also be conservative. The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene. Polynucleotides used herein can be composed of any polyribonucleotide or polydeoxribonucleotide, which can be unmodified RNA or DNA or modified RNA or DNA. For example, polynucleotides can be composed of single- and double- stranded DNA, DNA that is a mixture of single- and double- stranded regions, single- and double- stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that can be single- stranded or, more typically, double- stranded or a mixture of single- and double- stranded regions. In addition, the polynucleotide can be composed of triple- stranded regions comprising RNA or DNA or both RNA and DNA. A polynucleotide can also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons. “Modified” bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications can be made to DNA and RNA; thus, “polynucleotide” embraces chemically, enzymatically, or metabolically modified forms.
[0015] A “vector” is capable of transferring gene sequences to target-cells. Typically, “vector construct,” “expression vector,” and “gene transfer vector,” mean any nucleic acid construct capable of directing the expression of a gene of interest and which can transfer gene sequences to target-cells, which can be accomplished by genomic integration of all or a portion of the vector, or transient or inheritable maintenance of the vector as an extrachromosomal element. Thus, the term includes cloning, and expression vehicles, as well as integrating vectors.
[0016] The term “regulatory element” as used herein includes a nucleotide sequence which controls some aspect of the expression of nucleic acid sequences. Examples of regulatory elements illustratively include an enhancer, an internal ribosome entry site (IRES), an intron, an origin of replication, a polyadenylation signal (pA), a promoter, an enhancer, a transcription termination sequence, and an upstream regulatory domain, which contribute to the replication, transcription, and/or post-transcriptional processing of a nucleic acid sequence. In cases, regulatory elements can also include cv.v-regulatoiy DNA elements as well as transposable elements (TEs). Those of ordinary skill in the art are capable of selecting and using these and other regulatory elements in an expression construct with no more than routine experimentation. Expression constructs can be generated using a genetic recombinant approach or synthetically using well-known methodology.
[0017] A “control element” or “control sequence” is a nucleotide sequence involved in an interaction of molecules contributing to the functional regulation of a polynucleotide, including replication, duplication, transcription, splicing, translation, or degradation of the polynucleotide. The regulation may affect the frequency, speed, or specificity of the process, and may be enhancing or inhibitory in nature. Control elements known in the art include, for example, transcriptional regulatory sequences such as promoters and enhancers. A promoter is a DNA region capable under certain conditions of binding RNA polymerase and initiating transcription of a coding region usually located downstream (in the 3’ direction) from the promoter.
[0018] As used herein, the term “secretome composition” refers to a composition comprising one or more substances which are secreted from a cell. In certain embodiments, a secretome composition may include one or more cytokines, one or more exosomes, and/or one or more microvesicles. A secretome composition may be purified or unpurified. In some embodiments, a secretome composition may further comprise one or more substances that are not secreted from a cell (e.g., culture media, additives, nutrients, etc.). In some a secretome composition does not comprise and or comprises only trace amounts of one or more substances that are not secreted from a cell (e.g., culture media, additives, nutrients, etc.).
[0019] The terms “treatment,” “treat,” or “treating,” and the like, as used herein covers any treatment of a human or nonhuman mammal (e.g., rodent, cat, dog, horse, cattle, sheep, and primates etc.), and includes preventing the disease or condition from occurring in a subject who may be predisposed to the disease or condition but has not yet been diagnosed as having it. It also includes inhibiting (arresting development ol), relieving or ameliorating (causing regression ol), or curing (permanently stopping development or progression) the disease, condition and/or any related symptoms. The terms “treatment,” “treat,” or “treating,” as used herein covers any treatment of a disease or condition of a mammal, particularly a human, and includes: (a) preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it; (b) inhibiting the disease or condition, e.g., arresting its development; (c) relieving and or ameliorating the disease or condition, e.g., causing regression of the disease or condition; or (d) curing the disease or condition, e.g., stopping its development or progression. The population of subjects treated by the methods of the invention includes subjects suffering from the undesirable condition or disease, as well as subjects at risk for development of the condition or disease. In some embodiments, “treatment” (also “treat” or “treating”) refers to any administration of a therapy that partially or completely alleviates, ameliorates, relives, inhibits, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms, features, and/or causes of a particular disease, disorder, and/or condition. In some embodiments, such treatment may be of a subject who does not exhibit signs of the relevant disease, disorder, and/or condition, and/or of a subject who exhibits only early signs of the disease, disorder, and/or condition. Alternatively and/or additionally, such treatment may be of a subject who exhibits one or more established signs of the relevant disease, disorder, and/or condition. In some embodiments, treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, and/or condition. In some embodiments, treatment may be of a subject known to have one or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, and/or condition.
[0020] As used herein, a “wound” is any disruption, from whatever cause, of normal anatomy (internal and/or external anatomy) including but not limited to traumatic injuries such as mechanical ( e.g . contusion, penetrating), thermal, chemical, electrical, radiation, concussive and incisional injuries; elective injuries such as operative surgery and resultant incisional hernias, fistulas, etc. ; acute wounds, chronic wounds, infected wounds, and sterile wounds, as well as wounds associated with disease states (e.g. ocular contusion). A wound is dynamic and the process of healing is a continuum requiring a series of integrated and interrelated cellular processes that begin at the time of wounding and proceed beyond initial wound closure through arrival at a stable wound closure. These cellular processes are mediated or modulated by humoral substances including but not limited to cytokines, lymphokines, growth factors, and hormones. In accordance with the subject invention, “wound healing” refers to improving, by some form of intervention, the natural cellular processes and humoral substances of tissue repair such that healing is faster, and/or the resulting healed area has less scaring and/or the wounded area possesses tissue strength that is closer to that of uninjured tissue and/or the wounded tissue attains some degree of functional recovery.
[0021] As used herein, the terms “a” or “an” means one or more or at least one.
[0022] As used herein, a “therapeutically effective” or “effective” dosage or amount of a composition is an amount sufficient to have a positive effect on a given medical condition. If not immediate, the therapeutically effective or effective dosage or amount may, over period of time, provide a noticeable or measurable effect on a patient's health and well-being.
[0023] As used herein a “composition” or “pharmaceutical composition” refers to an a mixture of at least one compound, such as the compound of the anti-Met antibody provided herein, with at least one and optionally more than one other pharmaceutically acceptable chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
[0024] The term “pharmaceutically acceptable carrier” refers to a carrier or a diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compounds.
[0025] The term “excipient” refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound.
[0026] As used herein, the terms “mix”, “mixing”, and the like describe a mechanical process or a mechanical treatment of the components. For example, mixing can be in the sense of carrying out repeated cycles of pressing and folding or comparable processing steps which lead to an intense compression and mixing of the provided hydrophobic matrices.
[0027] Adult stem cells can be harvested from a variety of adult tissues, including bone marrow, fat, and dental pulp tissue. While all adult stem cells are cable of self-renewal and are considered multipotent, their therapeutic functions vary depending on their origin. As a result, each type of adult stem cell has unique characteristics that make them suitable for certain diseases. Mesenchymal stem cells (MSCs) are typically derived from the mesoderm and are multipotent, nonhematopoietic (non-blood) stem cells isolated from (derived from) capable of differentiating into a variety of tissues, including osteoblasts (e.g., bone cells), chondrocytes (e.g., cartilage cells), myocytes (e.g., muscle cells) and adipocytes (e.g., fat cells which give rise to marrow adipose tissue). As used herein, “isolated” refers to cells removed from their original environment. Stem cells produce factors, such as growth factors, that regulate or are important for regulating multiple biological processes. A growth factor is an agent, such as a naturally occurring substance capable of stimulating cellular growth and/or proliferation and/or cellular differentiation. Typically, growth factors are proteins or steroid hormones. While the terms “growth factor” and “factor” and the like are used interchangeably herein, the term “biological factor” is not limited to growth factors.
[0028] Human mesenchymal stem cells (MSCs), can be characterized by the surface marker profile of CD45-/CD31-/CD73+/CD90+/CD105+/CD44+ (or any suitable subset thereof). (See Bourin etal, Cytotherapy 15(6):641-648 (2013)). Further, appropriate stem cells display the CD34+ positive at the time of isolation, but lose this marker during culturing. Therefore, the full marker profile for one stem cell type that may be used according to the present application includes CD45-/CD31-/CD73+/CD90+/CD105+. In another embodiment utilizing mouse stem cells, the stem cells are characterized by the Sca- 1 marker, instead of CD34, to define what appears to be a homologue to the human cells described above, with the remaining markers remaining the same.
[0029] The phrase “conditioned medium” or “CM” refers to media which includes biological factors secreted by MSCs. This can also be referred to herein as the “secretome”, “MSC-CM”, “anti-Met antibody composition” and/or “MSC derived secretome”. Also provided are processed “conditioned medium” which included biological factors secreted by MSCs and which has been further processed by, for example, filtration, purification, and/or concentration procedures. The “conditioned medium” is obtained by culturing stem cells in media, as described herein in detail, and separating the resulting media, which contains stem cells and their secreted stem cell products (secretome) into conditioned medium that contains biological factors and fewer stem cells than were present prior to separation. The conditioned medium may be used in the methods described herein and is substantially free of stem cells (may contain a small percentage of stem cells) or free of stem cells. Biological factors that may be in the conditioned medium include, but are not limited to, proteins (e.g., cytokines, chemokines, growth factors, enzymes), nucleic acids (e.g, miRNA), lipids (e.g., phospholipids), polysaccharides, and/or combinations thereof. Any combination(s) of these biological factors may be either bound within or on the surface of extracellular vesicles (e.g., exosomes) or separate from extracellular vesicles.
B. ANTIBODIES
[0030] As is discussed below, the term “antibody” is used generally. Traditional antibody structural units typically comprise a tetramer. Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one “light” (typically having a molecular weight of about 25 kDa) and one “heavy” chain (typically having a molecular weight of about 50-70 kDa). Human light chains are classified as kappa and lambda light chains. The present invention is directed to antibodies that generally are based on the IgG class, which has several subclasses, including, but not limited to IgGl, IgG2, IgG3, and IgG4. In general, IgGl, IgG2 and IgG4 are used more frequently than IgG3. It should be noted that IgGl has different allotypes with polymorphisms at 356 (D or E) and 358 (L or M). The sequences depicted herein use the 356D/358M allotype, however the other allotype is included herein. That is, any sequence inclusive of an IgGl Fc domain included herein can have 356E/358L replacing the 356D/358M allotype.
[0031] The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition, generally referred to in the art and herein as the “Fv domain” or “Fv region”. In the variable region, three loops are gathered for each of the V domains of the heavy chain and light chain to form an antigen-binding site. Each of the loops is referred to as a complementarity determining region (hereinafter referred to as a “CDR”), in which the variation in the amino acid sequence is most significant. “Variable” refers to the fact that certain segments of the variable region differ extensively in sequence among antibodies. Variability within the variable region is not evenly distributed. Instead, the V regions consist of relatively invariant stretches called framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called “hypervariable regions” that are each 9-15 amino acids long or longer.
[0032] Each VH and VL is composed of three hypervariable regions (“complementary determining regions,” “CDRs”) and four FRs, arranged from amino- terminus to carboxy -terminus in the following order: FR1-CDR1-FR2-CDR2-FR3-CDR3- FR4.
[0033] The hypervariable region generally encompasses amino acid residues from about amino acid residues 24-34 (LCDR1; “L” denotes light chain), 50-56 (LCDR2) and 89-97 (LCDR3) in the light chain variable region and around about 31-35B (HCDR1; “H” denotes heavy chain), 50-65 (HCDR2), and 95-102 (HCDR3) in the heavy chain variable region; Rabat et al., SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991) and/or those residues forming a hypervariable loop (e.g. residues 26-32 (LCDR1), 50-52 (LCDR2) and 91-96 (LCDR3) in the light chain variable region and 26-32 (HCDR1), 53-55 (HCDR2) and 96-101 (HCDR3) in the heavy chain variable region; Chothia and Lesk (1987) J. Mol. Biol. 196:901-917. Specific CDRs of the invention are described below.
[0034] As will be appreciated by those in the art, the exact numbering and placement of the CDRs can be different among different numbering systems. However, it should be understood that the disclosure of a variable heavy and/or variable light sequence includes the disclosure of the associated (inherent) CDRs. Accordingly, the disclosure of each variable heavy region is a disclosure of the vhCDRs (e.g. vhCDRl, vhCDR2 and vhCDR3) and the disclosure of each variable light region is a disclosure of the vlCDRs (e.g. vlCDRl, vlCDR2 and vlCDR3). A useful comparison of CDR numbering is as below, see Lafranc et al., Dev. Comp. Immunol. 27(l):55-77 (2003):
Figure imgf000028_0001
[0035] Throughout the present specification, the Rabat numbering system is generally used when referring to a residue in the variable domain (approximately, residues 1-107 of the light chain variable region and residues 1-113 of the heavy chain variable region) and the hinge and the EU numbering system for Fc regions (e.g, Rabat et al., supra (1991)).
[0036] The present invention provides a large number of different CDR sets. In this case, a “full CDR set” comprises the three variable light and three variable heavy CDRs, e.g. a vlCDRl, vlCDR2, vlCDR3, vhCDRl, vhCDR2 and vhCDR3. These can be part of a larger variable light or variable heavy domain, respectfully. In addition, as more fully outlined herein, the variable heavy and variable light domains can be on separate polypeptide chains, when a heavy and light chain is used, or on a single polypeptide chain in the case of scFv sequences.
[0037] The CDRs contribute to the formation of the antigen-binding, or more specifically, epitope binding site of antibodies. “Epitope” refers to a determinant that interacts with a specific antigen binding site in the variable region of an antibody molecule known as a paratope. Epitopes are groupings of molecules such as amino acids or sugar side chains and usually have specific structural characteristics, as well as specific charge characteristics. A single antigen may have more than one epitope. [0038] The epitope may comprise amino acid residues directly involved in the binding (also called immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically antigen binding peptide; in other words, the amino acid residue is within the footprint of the specifically antigen binding peptide.
[0039] Epitopes may be either conformational or linear. A conformational epitope is produced by spatially juxtaposed amino acids from different segments of the linear polypeptide chain. A linear epitope is one produced by adjacent amino acid residues in a polypeptide chain. Conformational and nonconformational epitopes may be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
[0040] An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Antibodies that recognize the same epitope can be verified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen, for example “binning.” As outlined below, the invention not only includes the enumerated antigen binding domains and antibodies herein, but those that compete for binding with the epitopes bound by the enumerated antigen binding domains.
[0041] The carboxy -terminal portion of each chain defines a constant region primarily responsible for effector function. Kabat et al. collected numerous primary sequences of the variable regions of heavy chains and light chains. Based on the degree of conservation of the sequences, they classified individual primary sequences into the CDR and the framework and made a list thereof (see SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, E.A. Kabat et al., entirely incorporated by reference).
[0042] In the IgG subclass of immunoglobulins, there are several immunoglobulin domains in the heavy chain. By “immunoglobulin (Ig) domain” herein is meant a region of an immunoglobulin having a distinct tertiary structure. Of interest in the present invention are the heavy chain domains, including, the constant heavy (CH) domains and the hinge domains. In the context of IgG antibodies, the IgG isotypes each have three CH regions. Accordingly, “CH” domains in the context of IgG are as follows: “CHI” refers to positions 118-220 according to the EU index as in Kabat. “CH2” refers to positions 237-340 according to the EU index as in Kabat, and “CH3” refers to positions 341-447 according to the EU index as in Kabat. As shown herein and described below, the pi variants can be in one or more of the CH regions, as well as the hinge region, discussed below.
[0043] Another type of Ig domain of the heavy chain is the hinge region. By “hinge” or “hinge region” or “antibody hinge region” or “immunoglobulin hinge region” herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody. Structurally, the IgG CHI domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237. Thus for IgG the antibody hinge is herein defined to include positions 221 (D221 in IgGl) to 236 (G236 in IgGl), wherein the numbering is according to the EU index as in Kabat. In some embodiments, for example in the context of an Fc region, the lower hinge is included, with the “lower hinge” generally referring to positions 226 or 230.
[0044] The light chain generally comprises two domains, the variable light domain (containing the light chain CDRs and together with the variable heavy domains forming the Fv region), and a constant light chain region (often referred to as CL or CK).
[0045] Another region of interest for additional substitutions, outlined below, is the Fc region.
[0046] In some embodiments, the antibodies herein can be derived from a mixture from different species, e.g. a chimeric antibody and/or a humanized antibody. In general, both “chimeric antibodies” and “humanized antibodies” refer to antibodies that combine regions from more than one species. For example, “chimeric antibodies” traditionally comprise variable region(s) from a mouse (or rat, in some cases) and the constant region(s) from a human. “Humanized antibodies” generally refer to non-human antibodies that have had the variable-domain framework regions swapped for sequences found in human antibodies. Generally, in a humanized antibody, the entire antibody, except the CDRs, is encoded by a polynucleotide of human origin or is identical to such an antibody except within its CDRs. The CDRs, some or all of which are encoded by nucleic acids originating in a non-human organism, are grafted into the beta-sheet framework of a human antibody variable region to create an antibody, the specificity of which is determined by the engrafted CDRs. The creation of such antibodies is described in, e.g., WO 92/11018, Jones, 1986, Nature 321:522-525, Verhoeyen et ak, 1988, Science 239:1534-1536, all entirely incorporated by reference. “Backmutation” of selected acceptor framework residues to the corresponding donor residues is often required to regain affinity that is lost in the initial grafted construct (US 5530101; US 5585089; US 5693761; US 5693762; US 6180370; US 5859205; US 5821337; US 6054297; US 6407213, all entirely incorporated by reference). The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin, and thus will typically comprise a human Fc region. Humanized antibodies can also be generated using mice with a genetically engineered immune system. Roque et al., 2004, Biotechnol. Prog. 20:639-654, entirely incorporated by reference. A variety of techniques and methods for humanizing and reshaping non-human antibodies are well known in the art (See Tsurushita & Vasquez, 2004, Humanization of Monoclonal Antibodies, Molecular Biology of B Cells, 533-545, Elsevier Science (USA), and references cited therein, all entirely incorporated by reference). Humanization methods include but are not limited to methods described in Jones et al., 1986, Nature 321:522-525; Riechmann et al.,1988; Nature 332:323-329; Verhoeyen et al., 1988, Science, 239:1534-1536; Queen et al., 1989, Proc Natl Acad Sci, USA 86:10029- 33; He et al., 1998, J. Immunol. 160: 1029-1035; Carter et al., 1992, Proc Natl Acad Sci USA 89:4285-9, Presta et al., 1997, Cancer Res. 57(20):4593-9; Gorman et al., 1991, Proc. Natl. Acad. Sci. USA 88:4181-4185; O’Connor et al., 1998, Protein Eng 11:321-8, all entirely incorporated by reference. Humanization or other methods of reducing the immunogenicity of nonhuman antibody variable regions may include resurfacing methods, as described for example in Roguska et al., 1994, Proc. Natl. Acad. Sci. USA 91:969-973, entirely incorporated by reference.
[0047] In certain embodiments, the antibodies of the invention comprise a heavy chain variable region from a particular germline heavy chain immunoglobulin gene and/or a light chain variable region from a particular germline light chain immunoglobulin gene (with optional mutations as is generally described herein). For example, such antibodies may comprise or consist of a human antibody comprising heavy or light chain variable regions that are "the product of or "derived from" a particular germline sequence. A human antibody that is "the product of or "derived from" a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequences of human germline immunoglobulins and selecting the human germline immunoglobulin sequence that is closest in sequence (i.e., greatest % identity) to the sequence of the human antibody. A human antibody that is "the product of or "derived from" a particular human germline immunoglobulin sequence may contain amino acid differences as compared to the germline sequence, due to, for example, naturally-occurring somatic mutations or intentional introduction of site-directed mutation. However, a humanized antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the antibody as being derived from human sequences when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences). In certain cases, a humanized antibody may be at least 95, 96, 97, 98 or 99%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene. Typically, a humanized antibody derived from a particular human germline sequence will display no more than 10-20 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene. In certain cases, the humanized antibody may display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene (again, prior to the introduction of any variants herein; that is, the number of variants is generally low, prior to the introduction of the variants of the invention).
[0048] In one embodiment, the parent antibody has been affinity matured, as is known in the art. Structure-based methods may be employed for humanization and affinity maturation, for example as described in USSN 11/004,590. Selection based methods may be employed to humanize and/or affinity mature antibody variable regions, including but not limited to methods described in Wu et al., 1999, J. Mol. Biol. 294:151-162; Baca et al.,
1997, J. Biol. Chem. 272(16): 10678-10684; Rosok et al., 1996, J. Biol. Chem. 271(37): 22611-22618; Rader et al., 1998, Proc. Natl. Acad. Sci. USA 95: 8910-8915; Krauss et al., 2003, Protein Engineering 16(10):753-759, all entirely incorporated by reference. Other humanization methods may involve the grafting of only parts of the CDRs, including but not limited to methods described in USSN 09/810,510; Tan et al., 2002, J. Immunol. 169:1119- 1125; De Pascalis et al., 2002, J. Immunol. 169:3076-3084, all entirely incorporated by reference.
[0049] In one aspect, the present invention provides anti-Met antibodies, or antigen binding proteins, pharmaceutical compositions for wound healing or tissue regeneration that includes the anti-Met antibodies or antigen-binding proteins. C. SPECIFIC ANTI-MET ANTIBODIES
[0050] In one aspect, the present invention provides antigen-binding proteins that bind to Met, including but not limited to full length anti-Met antibodies and ScFvs, pharmaceutical compositions for wound healing or tissue regeneration that includes antigen-binding proteins including but not limited to full length anti-Met antibodies and ScFvs that bind to Met. In some embodiments, the invention provides antigen binding domains, including but not limited to full length antibodies and ScFvs, which contain a number of specific, enumerated sets of 6 CDRs and defined variable heavy (vh, VH or VH) and variable light (vl, VL or VL), that bind to Met.
[0051] In one embodiment, the antigen-binding protein of the present invention is an anti- Met antibody. In some embodiments, the anti-Met antibody is an antibody comprising a set of six CDRs (vhCDRl, vhCDR2, vhCDR3, vlCDRl, vlCDR2 and vlCDR3) fromHRll as depicted in Figure 3. In some embodiments, the anti-Met antibody is an antibody comprising a set of six CDRs (vhCDRl, vhCDR2, vhCDR3, vlCDRl, vlCDR2 and vlCDR3) from HR11, with the exception that at least one of the six CDRs comprises one or two conservative amino acid substitutions. In one embodiment, one CDR comprises one or two conservative amino acid substitutions. In another embodiment, two CDRs comprise one or two conservative amino acid substitutions. In another embodiment, three CDRs comprise one or two conservative amino acid substitutions. In another embodiment, four CDRs comprise one or two conservative amino acid substitutions. In another embodiment, five CDRs comprise one or two conservative amino acid substitutions. In another embodiment, six CDRs comprise one or two conservative amino acid substitutions.
[0052] In some embodiments, variations are made in both the framework regions that retain at least 80%, 85%, 90% or 95% identity to germline gene sequences. The CDRs can have amino acid modifications (e.g., from 1, 2, 3, 4 or 5 amino acid modifications in the set of CDRs (that is, the CDRs can be modified as long as the total number of changes in the set of 6 CDRs is less than 6 amino acid modifications, with any combination of CDRs being changed; e.g., there may be one change in vlCDRl, two in vhCDR2, none in vhCDR3, etc.).
In some embodiments, CDR1 and/or CDR2 can have amino acid modifications (e.g., from 1, 2, 3, 4 or 5 amino acid modifications in either CDR1, CDR2, or both), while CDR3 does not contain modifications. [0053] In one embodiment, the anti-Met antibody is an antibody comprising the variable heavy (vh) domain (SEQ ID NO:5) and variable light (vl) domain (SEQ ID NO: 13) from HR11. In another embodiment, the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 80% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively. In another embodiment, the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 85% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively. In another embodiment, the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 90% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively. In yet another embodiment, the anti-Met antibody is an antibody comprising vh and vl domains having amino acid sequences with at least 95% identity to SEQ ID NO:5 and SEQ ID NO: 13, respectively.
[0054] In some embodiments, the antigen-binding domain of the anti-Met antibody is linked to a human IgG constant domain of IgGl, IgG2, IgG3, IgG4 or IgG4. In some embodiments, the human IgG domain of the anti-Met antibody is IgGl. In some embodiments, the IgGl domain of the anti-Met antibody comprises an amino acid sequence of SEQ ID NO:7.
[0055] In some embodiments, the anti-Met antibody comprises a constant light chain region of human Kappa constant. In some embodiments, the constant light chain region of the anti- Met antibody comprises an amino acid sequence of SEQ ID NO: 15.
[0056] In some embodiments, the anti-Met antibody is an antibody comprising the heavy chain (SEQ ID NO:l) and light chain (SEQ ID NO: 9) fromHRll. In one embodiment, the anti-Met antibody is HR11.
[0057] Descriptions of additional anti-Met antibodies are provided in the US Patent NOs: 10,450,376; 10,407,50; 10,000,569; 9,717,715; 9,808,507; 9,593,164; and 9,487,589, and the US Publication Application NOs: 20200231681; 20200129633; 20180110875; 20180110875; and 20150064191, the disclosures of which have been incorporated by reference in their entireties.
D. OPTIONAL ANTIBODY ENGINEERING
[0058] The antibodies of the invention can be modified, or engineered, to alter the amino acid sequences by amino acid substitutions. As discussed herein, amino acid substitutions can be made to alter the affinity of the CDRs for the antigen (including both increasing and decreasing binding), as well as to alter additional functional properties of the antibodies. For example, the antibodies may be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity. Furthermore, an antibody according to at least some embodiments of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody. Such embodiments are described further below. The numbering of residues in the Fc region is that of the EU index of Kabat.
[0059] In one embodiment, the hinge region of CHI is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Pat. No. 5,677,425 by Bodmer et al. The number of cysteine residues in the hinge region of CHI is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.
[0060] In still another embodiment, the antibody can be modified to abrogate in vivo Fab arm exchange, in particular when IgG4 constant domains are used. Specifically, this process involves the exchange of IgG4 half-molecules (one heavy chain plus one light chain) between other IgG4 antibodies that effectively results in antibodies which are functionally monovalent. Mutations to the hinge region and constant domains of the heavy chain can abrogate this exchange (see Aalberse, RC, Schuurman J., 2002, Immunology 105:9-19). As outlined herein, a mutation that finds particular use in the present invention is the S241P in the context of an IgG4 constant domain. IgG4 finds use in the present invention as it has no significant effector function, and is thus used to block the receptor-ligand binding without cell depletion.
[0061] In some embodiments, amino acid substitutions can be made in the Fc region, in general for altering binding to FcyR receptors. By "Fc gamma receptor", "FcyR" or "FcgammaR" as used herein is meant any member of the family of proteins that bind the IgG antibody Fc region and is encoded by an FcyR gene. In humans this family includes but is not limited to FcyR I (CD64), including isoforms FcyRIa, FcyRIb, and FcyRIc; FcyRII (CD32), including isoforms FcyRIIa (including allotypes H131 and R131), FcyRIIb (including FcyRIIb-l and FcyRIIb-2), and FcyRIIc; and FcyRIII (CD16), including isoforms FcyRIIIa (including allotypes V158 and F158) and FcyRIIIb (including allotypes FcyRIIIb-NAl and FcyRIIIb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference), as well as any undiscovered human FcyRs or FcyR isoforms or allotypes. An FcyR may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys. Mouse FcyRs include but are not limited to FcyR I (CD64), FcyRII (CD32), FcyRIII-l (CD16), and FcyRIII-2 (CD16-2), as well as any undiscovered mouse FcyRs or FcyR isoforms or allotypes.
[0062] There are a number of useful Fc substitutions that can be made to alter binding to one or more of the FcyR receptors. Substitutions that result in increased binding as well as decreased binding can be useful. For example, it is known that increased binding to FcyRIIIa generally results in increased ADCC (antibody dependent cell-mediated cytotoxicity; the cell- mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell. Similarly, decreased binding to FcyRIIb (an inhibitory receptor) can be beneficial as well in some circumstances. Amino acid substitutions that find use in the present invention include those listed in U.S.
Ser. Nos. 11/124,620 (particularly FIG. 41) and U.S. Patent No. 6,737,056, both of which are expressly incorporated herein by reference in their entirety and specifically for the variants disclosed therein.
[0063] In yet other embodiments, the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector functions of the antibody. For example, one or more amino acids selected from amino acid residues 234, 235, 236, 237,
297, 318, 320 and 322 can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody. The effector ligand to which affinity is altered can be, for example, an Fc receptor or the Cl component of complement. This approach is described in further detail in U.S. Pat. Nos. 5,624,821 and 5,648,260, both by Winter et al.
[0064] In another example, one or more amino acids selected from amino acid residues 329, 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered Clq binding and/or reduced or abolished complement dependent cytotoxicity (CDC). This approach is described in further detail in U.S. Pat. Nos. 6,194,551 by Idusogie et al.
[0065] In another example, one or more amino acid residues within amino acid positions 231 and 239 are altered to thereby alter the ability of the antibody to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al. [0066] In yet another example, the Fc region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fey receptor by modifying one or more amino acids at the following positions: 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305,
307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340,
360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439. This approach is described further in PCT Publication WO 00/42072 by Presta. Moreover, the binding sites on human IgGl for FcyRI, FcyRII, FcyRIII and FcRn have been mapped and variants with improved binding have been described (see Shields, R. L. et al. (2001) J. Biol. Chem. 276:6591-6604). Specific mutations at positions 256, 290, 298, 333, 334 and 339 are shown to improve binding to FcyRIII. Additionally, the following combination mutants are shown to improve FcyRIII binding: T256A/S298A, S298A/E333A, S298A/K224A and S298A/E333A/K334A. Furthermore, mutations such as M252Y/S254T/T256E or M428L/N434S improve binding to FcRn and increase antibody circulation half-life (see Chan CA and Carter PJ (2010 ) Nature Rev Immunol 10:301-316).
[0067] In addition, the antibodies of the invention are modified to increase its biological half- life. Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Pat. No. 6,277,375 to Ward. Alternatively, to increase the biological half-life, the antibody can be altered within the Cm or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Pat. Nos. 5,869,046 and 6,121,022 by Presta et al. Additional mutations to increase serum half-life are disclosed in U.S. Patent Nos. 8,883,973, 6,737,056 and 7,371,826 and include 428L, 434A, 434S, and 428L/434S.
[0068] In still another embodiment, the glycosylation of an antibody is modified. For example, an aglycosylated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen or reduce effector function such as ADCC. Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence, for example N297. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site, with an alanine replacement finding use in some embodiments. [0069] Additionally or alternatively, an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures. Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies according to at least some embodiments of the invention to thereby produce an antibody with altered glycosylation. For example, the cell lines Ms704, Ms705, and Ms709 lack the fucosyltransferase gene, FUT8 (a (1,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their carbohydrates. The Ms704, Ms705, and Ms709 FUT8 cell lines are created by the targeted disruption of the FUT8 gene in CHO/DG44 cells using two replacement vectors (see U.S. Patent Publication No. 20040110704 by Yamane et al. and Yamane-Ohnuki et al. (2004) Biotechnol Bioeng 87:614-22). As another example, EP 1,176,195 by Hanai et al. describes a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the a 1,6 bond-related enzyme. Hanai et al. also describe cell lines which have a low enzyme activity for adding fucose to the N-acetylglucosamine that binds to the Fc region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662). PCT Publication WO 03/035835 by Presta describes a variant CHO cell line, Led 3 cells, with reduced ability to attach fucose to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (see also Shields, R. L. et al. (2002) J Biol. Chem. 277:26733-26740). PCT Publication WO 99/54342 by Umana et al. describes cell lines engineered to express glycoprotein-modifying glycosyl transferases (e.g., b( 1.4)-N-acetylglucosaminyl transferase III (GnTIII)) such that antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (see also Umana et al. (1999) Nat. Biotech. 17:176-180). Alternatively, the fucose residues of the antibody may be cleaved off using a fucosidase enzyme. For example, the fucosidase a-L-fucosidase removes fucosyl residues from antibodies (Tarentino, A. L. et al. (1975) Biochem. 14:5516-23).
[0070] Another modification of the antibodies herein that is contemplated by the invention is PEGylation or the addition of other water soluble moieties, typically polymers, e.g., in order to enhance half-life. An antibody can be PEGylated to, for example, increase the biological (e.g., serum) half-life of the antibody. To PEGylate an antibody, the antibody, or fragment thereof, typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment. In some embodiments, the PEGylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer). As used herein, the term "polyethylene glycol" is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Ci-Cio) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide. In certain embodiments, the antibody to be PEGylated is an aglycosylated antibody. Methods for PEGylating proteins are known in the art and can be applied to the antibodies according to at least some embodiments of the invention. See for example, EP 0 154316 by Nishimura et al. and EP 0401 384 by Ishikawa et al.
[0071] In addition to substitutions made to alter binding affinity to FcyRs and/or FcRn and/or increase in vivo serum half-life, additional antibody modifications can be made, as described in further detail below.
[0072] In some cases, affinity maturation is done. Amino acid modifications in the CDRs are sometimes referred to as "affinity maturation". An "affinity matured" antibody is one having one or more alteration(s) in one or more CDRs which results in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s). In some cases, it may be desirable to decrease the affinity of an antibody to its antigen.
[0073] In some embodiments, one or more amino acid modifications are made in one or more of the CDRs of the antibodies of the invention. In general, only 1 or 2 or 3-amino acids are substituted in any single CDR, and generally no more than from 1, 2, 3. 4, 5, 6, 7, 8 9 or 10 changes are made within a set of CDRs. However, it should be appreciated that any combination of no substitutions, 1, 2 or 3 substitutions in any CDR can be independently and optionally combined with any other substitution.
[0074] Affinity maturation can be done to increase the binding affinity of the antibody for the antigen by at least about 10% to 50-100-150% or more, or from 1 to 5 fold as compared to the "parent" antibody. Exemplary affinity matured antibodies will have nanomolar or even picomolar affinities for the antigen. Affinity matured antibodies are produced by known procedures. See, for example, Marks et al., 1992, Biotechnology 10:779-783 that describes affinity maturation by variable heavy chain (VH) and variable light chain (VL) domain shuffling. Random mutagenesis of CDR and/or framework residues is described in: Barbas, et al. 1994, Proc. Nat. Acad. Sci, USA 91:3809-3813; Shier et al., 1995, Gene 169:147-155; Yelton et al., 1995, J. Immunol. 155:1994-2004; Jackson et al., 1995, J. Immunol. 154(7):3310-9; and Hawkins et al, 1992, J. Mol. Biol. 226:889-896, for example.
[0075] Alternatively, amino acid modifications can be made in one or more of the CDRs of the antibodies of the invention that are "silent", e.g. that do not significantly alter the affinity of the antibody for the antigen. These can be made for a number of reasons, including optimizing expression (as can be done for the nucleic acids encoding the antibodies of the invention).
[0076] Thus, included within the definition of the CDRs and antibodies of the invention are variant CDRs and antibodies; that is, the antibodies of the invention can include amino acid modifications in one or more of the CDRs of the enumerated antibodies of the invention. In addition, as outlined below, amino acid modifications can also independently and optionally be made in any region outside the CDRs, including framework and constant regions.
E. NUCLEIC ACID COMPOSITIONS
[0077] Nucleic acid compositions encoding the anti-Met antibodies of the invention are also provided, as well as expression vectors containing the nucleic acids and host cells transformed with the nucleic acid and/or expression vector compositions. As will be appreciated by those in the art, the protein sequences depicted herein can be encoded by any number of possible nucleic acid sequences, due to the degeneracy of the genetic code. An exemplary embodiment of the nucleic acid sequences and expression vectors of the present invention is provided in Figure 3.
[0078] The nucleic acid compositions that encode the antibodies will depend on the format of the antibody. For traditional, tetrameric antibodies containing two heavy chains and two light chains are encoded by two different nucleic acids, one encoding the heavy chain and one encoding the light chain. These can be put into a single expression vector or two expression vectors, as is known in the art, transformed into host cells, where they are expressed to form the antibodies of the invention. In some embodiments, for example when scFv constructs are used, a single nucleic acid encoding the variable heavy chain-linker-variable light chain is generally used, which can be inserted into an expression vector for transformation into host cells. The nucleic acids can be put into expression vectors that contain the appropriate transcriptional and translational control sequences, including, but not limited to, signal and secretion sequences, regulatory sequences, promoters, origins of replication, selection genes, etc.
[0079] Exemplary mammalian host cells for expressing the recombinant antibodies according to at least some embodiments of the invention include Chinese Hamster Ovary (CHO cells), PERC6, HEK293 and others as is known in the art.
[0080] The nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form. A nucleic acid is "isolated" or "rendered substantially pure" when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art.
[0081] To create a scFv gene, the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser)3, such that the VH and VL sequences can be expressed as a contiguous single chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242:423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al., (1990) Nature 348:552-554).
F. COMPOSITIONS AND FORMULATIONS
[0082] According to the present description, compositions comprising antigen binding proteins, including but not limited to anti-Met antibodies such as full length anti- Met antibodies or ScFvs, are provided herein.
[0083] In some embodiments, the anti-Met antibody in the composition of the present invention does not promote angiogenesis. In some embodiments, the anti-Met antibody exhibits anti-angiogenic properties. In some embodiments, the composition comprising the anti-Met antibody provides for reduced angiogenesis. In some embodiments, the composition comprising the anti-Met antibody provides for a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in angiogenesis. In some embodiments, the composition comprising anti-Met antibody provides for a 10%, 20%, 30%, 40%, 50%,
60%, 70%, 80%, 90%, 95%, or 99% reduction in. In some embodiments, the composition comprising the anti-Met antibody provides for a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% reduction in angiogenesis. In some embodiments, the anti-Met antibody has low angiogenesis induction. In some embodiments, the anti-Met antibody has reduced angiogenic response. In some embodiments, the anti-Met antibody has reduced angiogenic capacity. In some embodiments, the anti-Met antibody impairs and/or reduces the normal formation of blood vessels in presence of media supportive of angiogenesis. In some embodiments, the anti-Met antibody has reduced angiogenic capacity when the anti- Met antibody is compared to untreated control. In some embodiments, the anti-Met antibody has reduced angiogenic capacity as compared to a sample treated with serum containing media. In some embodiments, the anti-Met antibody attenuates an angiogenic response. In some embodiments, the anti-Met antibody reduces the angiogenic response induce by serum containing media. In some embodiments, an angiogenic response is indicated by tube formation in a cell based assay. In some embodiments, an angiogenic response is indicated by tube formation in an endothelial cell tube formation assay. In some embodiments, an angiogenic response is indicated by blood vessel formation in a CAM (Chick Chorioallantoic membrane) assay. In some embodiments, an angiogenic response is indicated by blood vessel formation in any blood vessel formation assay known in the art.
[0084] In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 4.5 to about pH 8. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 4.7 to about pH 7.8. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 5.0 to about pH 7.5.
In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 5.5 to about pH 7.5. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 6 to about pH 7.5.
[0085] In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 4.5, about pH 5.0, about pH 5.5, about pH 6.0, about pH 6.5, about pH 7.0, about pH 7.4, about pH 8.0. In some embodiments, the anti-Met antibody composition is formulated at a pH of about pH 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8,
7.9, or 8.0.
[0086] In some embodiments, the anti-Met antibody composition does not comprise certain components. In some embodiments, the anti-Met antibody composition does not comprise certain components found in cellular media. In some embodiments, the anti-Met antibody composition does not comprise one or more components selected from the group consisting of xenobiotic components (for example, animal serum); Phenol red; peptides and biomolecules < 3kDa; antibiotics; protein aggregates (for example, protein aggregates >200nm); cells; cell debris (cell debris do not include exosomes/ Extracellular Vesicles (EVs); for example, non-exosome, non-EV cell debris); hormones (for example, hormones include, but are not limited to insulin and/or hydrocortisone); and/or L-glutamine. In some embodiments, the anti-Met antibody composition does not comprise xenobiotic components. In some embodiments, the anti-Met antibody composition does not comprise Phenol red. In some embodiments, the anti-Met antibody composition does not comprise peptides and biomolecules < 3kDa. In some embodiments, the anti-Met antibody composition does not comprise antibiotics. In some embodiments, the anti-Met antibody composition does not comprise protein aggregates (for example, protein aggregates >200nm). In some embodiments, the anti-Met antibody composition does not comprise cells. In some embodiments, the anti-Met antibody composition does not comprise cell debris (cell debris do not include exosomes/EVs; for example, non-exosome, non-EV cell debris). In some embodiments, the anti-Met antibody composition does not comprise hormones (for example, hormones include, but are not limited to insulin and/or hydrocortisone. In some embodiments, the anti-Met antibody composition does not comprise L-glutamine.
[0087] In some embodiments, the anti-Met antibody further comprises mannitol, lactose, sorbitol, xylitol, sucrose, trehalose, mannose, maltose, lactose, glucose, raffmose, cellobiose, gentiobiose, isomaltose, arabinose, glucosamine, fructose, dextrose, and/or combinations thereof. In some embodiments, the anti-Met antibody further comprises phosphate. In some embodiments, the phosphate source is sodium phosphate or potassium phosphate. In some embodiments, the phosphate source is sodium phosphate. In some embodiments, the phosphate source is potassium phosphate. In some embodiments, the anti-Met antibody further comprises mono/di-sodium phosphate, mannitol, and trehalose, wherein the composition has a pH of about pH 7.4. [0088] In some embodiments, the anti-Met antibody composition can comprise one or more additional agents including but not limited to glycine, glycerol, sodium chloride, potassium chloride, and/or dextrose. In some embodiments, the anti-Met antibody composition can comprise one or more additional agents selected from the group consisting of glycine, glycerol, sodium chloride, potassium chloride, and dextrose. In some embodiments, the anti-Met antibody composition can comprise one or more additional agents selected from the group consisting of glycine and glycerol, and dextrose. In some embodiments, the anti-Met antibody composition can comprise one or more additional agents selected from the group consisting of sodium chloride and potassium chloride.
[0089] In some embodiments, the anti-Met antibody composition is formulated in a buffer system. In some embodiments, the anti-Met antibody composition is formulated in a buffer system including but not limited to di/mono sodium phosphate, sodium citrate/citric acid, boric acid/sodium citrate, boric acid/sodium tetraborate, and/or citric acid/disodium phosphate. In some embodiments, the anti-Met antibody composition is formulated in a buffer system selected from the group consisting of di/mono sodium phosphate, sodium citrate/citric acid, boric acid/sodium citrate, boric acid/sodium tetraborate, and/or citric acid/disodium phosphate. In some embodiments, the anti-Met antibody composition is formulated in a di/mono sodium phosphate buffer system. In some embodiments, the anti- Met antibody composition is formulated in sodium citrate/citric acid buffer system. In some embodiments, the anti-Met antibody composition is formulated in a boric acid/sodium citrate buffer system. In some embodiments, the anti-Met antibody composition is formulated in a boric acid/sodium tetraborate buffer system. In some embodiments, the anti-Met antibody composition is formulated in a citric acid/disodium phosphate buffer system.
[0090] In some embodiments, the phosphate source is sodium phosphate or potassium phosphate. In some embodiments, the phosphate source is sodium phosphate. In some embodiments, the phosphate source is potassium phosphate. In some embodiments, the anti-Met antibody composition comprises di-sodium phosphate/citric acid, mannitol, and trehalose, wherein the composition has a pH of about pH 6.4.
[0091] In some embodiments, the anti-Met antibody composition further comprises a tonicity adjusting or tonicity modifying agent. In some embodiments, tonicity adjusting or tonicity modifying agent includes but is not limited to NaCl, KC1, mannitol, dextrose, sucrose, sorbitol, and/or glycerin. In some embodiments, tonicity adjusting or tonicity modifying agent is selected from the group consisting of NaCl, KC1, mannitol, dextrose, sucrose, sorbitol, and/or glycerin.
[0092] In some embodiments, the anti-Met antibody composition further comprises an adhesive agent. In some embodiments, the anti-Met antibody composition further comprises an adhesive agent including but not limited to hypromellose, Poloxamer 407, Poloxamer 188, Poloxomer 237, Poloxomer 338, Hypromellose, (HPMC), HEC, polycarbophil, polyvinylpyrrolidone (PVP), PVA (polyvinyl alcohol, polyimide, sodium hyaluronate, gellan gum, poly(lactic acid-co-gly colic acid) (PLGA), polysiloxane, polyimide, carboxymethylcellulose (CMC), or hydroxypropyl methylcellulose (HPMC), hydroxy methyl cellulose, hydroxy ethyl cellulose, sodium carboxy methyl cellulose, fibrin glue, polyethyelene glycol, and GelCORE. In some embodiments, the adhesive agent is hypromellose. In some embodiments, the adhesive agent is fibrin glue. In some embodiments, the adhesive agent is a polyethyelene glycol. In some embodiments, the adhesive agent is GelCORE (see, Sani, et cil, Science Advances, Vol. 5, no. 3 (2019)).
[0093] In some embodiments, the anti-Met antibody composition comprises (a) the anti-Met antibody produced by any one of the methods described herein; and (b) a polymer. In some embodiments, the anti-Met antibody compositions provided herein are in the form of a therapeutic bandage (e.g., a polymer impregnated with anti-Met antibody composition). The therapeutic bandage may be configured as needed, depending on the application. In some embodiments, the bandage is in the form or a patch or is configured as mesh.
[0094] In some embodiments, the anti-Met antibody compositions exhibit bio penetrance, for example, ocular penetration, comeal penetration, and/or comeal permeation. In some embodiments, the anti-Met antibody composition exhibits the ability to be absorbed by the eye. In some embodiments, the anti-Met antibody composition exhibits inherent bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits excipient-enabled bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to upregulation of the smaller factors. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of a biopreservative. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of the biopreservative benzalkonium chloride.
[0095] In some embodiments, the anti-Met antibody compositions exhibit long half- life and/or have increased stability as compared to other treatments. In some embodiments, the anti-Met antibody compositions as provided herein allow for an upregulation of proteins that are allow for increased stability of the anti-Met antibody. In some embodiments, the anti-Met antibody compositions as provided herein allow for upregulating chaperone proteins to improve stability of other proteins in the anti-Met antibody.
[0096] In some embodiments, the anti-Met antibody compositions exhibit ultrapotency when administered to a subject in need thereof. In some embodiments, the anti-Met antibody compositions allow for therapeutic efficacy with one drop or one administration per day.
[0097] In some embodiments, the anti-Met antibody composition comprises (a) anti- Met antibody composition produced by any one of the methods described herein; and (b) a polymer. In some embodiments, the anti-Met antibody composition comprises anti-Met antibody composition which is produced as described herein and a polymer. In some embodiments, the anti-Met antibody composition comprises the anti-Met antibody composition which is produced as described herein and a polymer. In some embodiments the polymer can be a biodegradable polymer from which the anti-Met antibody composition and/or anti-Met antibody composition components can be released. In some embodiments, the polymer enables sustained (slow) release of the anti-Met antibody composition components.
[0098] In some embodiments, the anti-Met antibody compositions provided herein are in the form of a therapeutic bandage (e.g., a polymer impregnated with anti-Met antibody composition). The therapeutic bandage may be configured as needed, depending on the application. In some embodiments, the bandage is in the form or a patch or is configured as mesh.
[0099] In some embodiments, the anti-Met antibody compositions exhibit bio penetrance, for example, ocular penetration, comeal penetration, and/or comeal permeation. In some embodiments, the anti-Met antibody composition exhibits the ability to be absorbed by the eye. In some embodiments, the anti-Met antibody composition exhibits inherent bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits excipient-enabled bio-penetrance. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to upregulation of the smaller factors. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of a biopreservative. In some embodiments, the anti-Met antibody composition exhibits bio-penetrance due to the presence of the biopreservative benzalkonium chloride.
[00100] In some embodiments, the anti-Met antibody compositions exhibit long half- life and/or have increased stability as compared to other treatments. In some embodiments, the anti-Met antibody compositions as provided herein allow for an upregulation of proteins that are allow for increased stability of the anti-Met antibody composition. In some embodiments, the anti-Met antibody compositions as provided herein allow for upregulating chaperone proteins to improve stability of other proteins in the anti-Met antibody composition.
[00101] In some embodiments, the anti-Met antibody compositions exhibit ultrapotency when administered to a subject in need thereof. In some embodiments, the anti-Met antibody compositions allow for therapeutic efficacy with one drop or one administration per day.
[00102] In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.1 mg - 10 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.2 mg - 9 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.3 mg - 8 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.4 mg - 7 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.5 mg - 6 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.6 mg - 5 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.7 mg - 4 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.8 mg - 3 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a pharmaceutical formulation comprising about 0.9 mg - 2 mg per 1 mL of anti-Met antibody. [00103] In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.1 mg - 10 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.2 mg - 9 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.3 mg - 8 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.4 mg - 7 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.5 mg - 6 mg per 1 mL of anti-Met antibody.
In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.6 mg - 5 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.7 mg - 4 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.8 mg - 3 mg per 1 mL of anti-Met antibody. In some embodiments, the anti-Met antibody is prepared in a formulation comprising about 0.9 mg - 2 mg per 1 mL of anti-Met antibody.
[00104] In some embodiments, the anti-Met antibody is prepared in a formulation comprising 2 mg - 3 mg per mL of monobasic sodium phosphate. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 4% to 5% per mL of monobasic sodium phosphate. [00105] In some embodiments, the anti-Met antibody is prepared in a formulation comprising 11 mg - 12 mg per mL of dibasic sodium phosphate. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 21.5% to 23% per mL of dibasic sodium phosphate.
[00106] In some embodiments, the anti-Met antibody is prepared in a formulation comprising 11.5 mg - 13 mg per mL of mannitol. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 23% to 25% per mL of mannitol.
[00107] In some embodiments, the anti-Met antibody is prepared in a formulation comprising 23 mg - 25 mg per mL of trehalose dihydrate. In some embodiments, the anti- Met antibody is prepared in a formulation comprising 46% to 48% per mL of trehalose dihydrate.
[00108] In some embodiments, the anti-Met antibody is prepared in a formulation that does not comprise hypromellose. In some embodiments, the anti-Met antibody is prepared in a formulation that optionally comprises hypromellose. In some embodiments, the anti- Met antibody is prepared in a formulation comprising 0.5 mg-2 mg per mL of hypromellose. In some embodiments, the anti-Met antibody is prepared in a formulation comprising 1% to 3% per mL of hypromellose. [00109] In some embodiments, the anti-Met antibody is prepared in a formulation comprising hydrochloric acid and/or sodium hydroxide. In some embodiments, the anti-Met antibody is prepared in a formulation comprising hydrochloric acid. In some embodiments, the anti-Met antibody is prepared in a formulation comprising sodium hydroxide. In some embodiments, the hydrochloric acid and/or sodium hydroxide is employed to obtain the desired pH.
[00110] In some embodiments, the anti-Met antibody is prepared in a formulation comprising the components as provided in Table 1 below
Table 1: anti-Met antibody formulation embodiment.
Figure imgf000049_0001
G. ASSAY METHODS/THERAPEUTIC PROPERTIES i. Anti-Met antibody - Therapeutic Properties
[00111] The anti-Met antibody of the present disclosure exhibits a variety of therapeutic properties, including for example, anti-angiogenic properties (blood vessels and/or lymphatic vessels), anti-fibrotic properties, anti-inflammatory properties, properties promoting cell migration and proliferation, mitogenic promoting properties, anti-oxidative stress/damage properties,
[00112] In some embodiments, the anti-Met antibody exhibits anti-inflammatory properties. In some embodiments, the anti-Met antibody inhibits inflammation. In some embodiments, the anti-Met antibody inhibits inflammation by 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%. 90%, or 100% (e.g., complete reduction in inflammation). In some embodiments, the anti-Met antibody prevents degranulation of mast cells.
[00113] In some embodiments, the anti-Met antibody promotes cell migration and proliferation, including for example, mitogenic and motogenic activities. In some embodiments, the anti-Met antibody promotes mitogenic activities. In some embodiments, the anti-Met antibody promotes motogenic activities. In some embodiments, the anti-Met antibody comprises FGF7, which provides for the cell migration and proliferation activities of the anti-Met antibody.
[00114] In some embodiments, the anti-Met antibody increases cell migration and proliferation by at least 5%, at least 10%, at least 15%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%. at least 90%, or at least 100%, or more. In some embodiments, the anti-Met antibody increases cell migration and proliferation by at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, or more.
[00115] In some embodiments, the anti-Met antibody provides for anti-oxidative stress and or reduction in cellular damage. In some embodiments, the anti-Met antibody comprises anti-oxidative stress and reduction in cellular damage factors. In some embodiments, the anti-oxidative stress and reduction in cellular damage factors include but are not limited to SOD-1, SOD-2, SOD-3, HO-1. In some embodiments, the anti-oxidative stress and reduction in cellular damage factor is selected from the group consisting of SOD- 1, SOD-2, SOD-3, HO-1.
[00116] In some embodiments, the anti-Met antibody accelerates wound healing (shortening wound healing period). In some embodiments, the anti-Met antibody accelerates wound healing by at least 10% to at least 100% or more. In some embodiments, the anti-Met antibody accelerates wound healing by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least or more. [00117] In some embodiments, the anti-Met antibody exhibits anti-scarring properties. In some embodiments, the anti-Met antibody inhibits scar formation. In some embodiments, the anti-Met antibody inhibits scar formation by 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% (e.g., complete prevention of scar formation). ii. Anti-Met antibody - Biophysical/Biochemical Properties
Biochemical and Biophysical Characterization:
[00118] In some embodiments, the present invention provides methods for characterization of the anti-Met antibody. In some embodiments, the characterization will include measuring biophysical parameters. In some embodiments, in order to determine the properties of the anti-Met antibody, various potency assays can be performed on the anti- Met antibody as described herein. In some embodiments, the anti-Met antibody can be subjected to measuring biophysical parameters. In some embodiments, characterization assays include but are not limited to biophysical assays, biochemical assays, and bioassays. In some embodiments, characterization assays can include but are not limited to physical component characterizations, oxidative stress assays, safety analysis, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays. In some embodiments, characterization assays are selected from the group consisting of physical component characterizations, oxidative stress assays, safety analysis, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays.
Oxidative Stress:
[00119] In some embodiments, oxidative stress prevention assays can be performed on the anti-Met antibody. In some embodiments, the anti-Met antibody prevents comeal epithelium damage. In some embodiments, the anti-Met antibody reduces the presence of inflammation. In some embodiments, the anti-Met antibody reduces the presence of inflammation as determined by an increase in the present of anti-inflammation markers. In some embodiments, the anti-Met antibody reduces the presence of inflammation as determined by an increase in the present of anti-inflammation markers, such as, for example, IL-8.
Safety Characterization: [00120] In some embodiments, the anti-Met antibody can be evaluated for blood compatibility and implementing tests for sterility as well as pyrogen and endotoxin levels. In some embodiments, the anti-Met antibody can be evaluated blood compatibility. In some embodiments, evaluating blood compatibility includes assays for hemolysis and hemagglutination. In some embodiments, the anti-Met antibody does not exhibit detrimental effects with systemic exposure. In some embodiments, the anti-Met antibody does not exhibit detrimental effects with systemic exposure, such as with severe ocular bums. In some embodiments, the anti-Met antibody does not exhibit hemagglutination activity. In some embodiments, the anti-Met antibody does not induce hemolysis. In some embodiments, the anti-Met antibody does not induce hemolytic activity.
Stability:
[00121] In some embodiments, the biophysical characteristics of the anti-Met antibody and the composition comprising the peptide can be evaluated and/or determined. In some embodiments, the fluorescence, static light scattering and dynamic light scatting to characterize protein stability metrics. In some embodiments, the following parameters can be measured to further characterize the anti-Met antibody and the composition comprising the peptide: thermal melting, thermal aggregation, Delta G, and/or viscosity. In some embodiments, a thermal melting assay is employed to determine anti-Met antibody stability. In some embodiments, a thermal aggregation assay is employed to determine anti- Met antibody stability. In some embodiments, delta G is employed as a measure for determining anti-Met antibody stability. In some embodiments, viscosity is measured as an anti-Met antibody characteristic. In some embodiments, viscosity is to determine anti-Met antibody stability
[00122] In some embodiments, biophysical metrics can be employed to establish stability parameters for characterizing different anti-Met antibody formulations.
[00123] In some embodiments, the anti-Met antibody compos is stable at -20°C, 4°C, and room temperature (20°C), for at least 7 days. In some embodiments, the anti-Met antibody is stable -20°C, 4°C, and room temperature (20°C), for at least 14 days. In some embodiments, the anti-Met antibody is stable for at least 7 days, at least 1 week, at least 2 weeks, at least 3 weeks, or at least 1 month. In some embodiments, the anti-Met antibody is stable for at least 7 days, at least 14 days, at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month, at least 2 months, or at least 3 months at about -20°C. In some embodiments, the anti-Met antibody is stable for at least 7 days, at least 14 days, at least 1 week, at least 2 weeks, at least 3 weeks, or at least 1 month at about 4°C. In some embodiments, the anti-Met antibody is stable for at least 7 days, at least 14 days, at least 1 week, at least 2 weeks, at least 3 weeks, or at least 1 month at about 20°C (or room temperature).
[00124] In some embodiments, the anti-Met antibody is stable for at least 7 days at about -20°C. In some embodiments, the anti-Met antibody is stable for at least 7 days at about 4°C. In some embodiments, the anti-Met antibody is stable for at least 7 days at about 20°C. In some embodiments, the anti-Met antibody is stable for at least 7 days at about 25°C (room temperature).
[00125] In some embodiments, the anti-Met antibody is stable for at least 14 days at about -20°C. In some embodiments, the anti-Met antibody is stable for at least 14 days at about 4°C. In some embodiments, the anti-Met antibody is stable for at least 14 days at about 20°C (or room temperature). In some embodiments, the anti-Met antibody is stable for at least 14 days at about 25°C (room temperature).
Epithelial barrier integrity assay
[00126] The comeal epithelium, more precisely, the apical surface of the epithelium has a major contribution to the overall barrier properties of the cornea and change to the comeal barrier serves as a sensitive factor for biocompatibility analysis. In some embodiments, the biophysical characteristics of the anti-Met antibody can be evaluated and/or determined such as by an epithelial barrier integrity assay. In some embodiments, the epithelial barrier integrity assay is a transepithelial electrical resistance (TEER). In some embodiments, the transepithelial electrical resistance (TEER) can be assessed to measure overall barrierroperties. In some embodiments, 3D tissues can be transferred into 24-well plates containing 2 mL of TEER buffer and incubated for 10 min. In some embodiments, TEER can be measured using an epithelial volt-ohm meter EVOMO and the EndOhm-12 chamber (World Precision, Sarasota, FL). In some embodiments, at the end of the procedure, tissues can be used for tissue viability assessment using the following formula:
% Barrier integrity = 100 x [TEER (treated tissue)/TEER (placebo control)]
[00127] In some embodiments, TEER can be employed to evaluate the effect on barrier integrity after topical application of the anti-Met antibody. In some embodiments, TEER can be employed to evaluate the effect on barrier integrity after topical application of the anti-Met antibody following comeal epithelial damage caused by topical exposure to nitrogen mustard (NM) utilizing the EpiComeal tissue model (MatTek Corp). In some embodiments, anti-Met antibody can be applied topically, for example at 6 pg/ml (diluted in Placebo solution), as described in Example 6. In some embodiments, EpiComeal tissues were cultured in 5 ml medium at standard culture conditions for 24h.
Bioassays
[00128] In some embodiments, bioassays can be employed to characterize the anti-Met antibody. In some embodiments, bioassays can be related to comeal wound healing: epithelial cell migration and proliferation, stromal cell differentiation (e.g, scarring); neovascularization, and inflammation. In some embodiments, bioassays can be employed to evaluate the ability of the anti-Met antibody to mediate comeal wound healing: epithelial cell migration and proliferation, stromal cell differentiation (scarring); neovascularization; and inflammation.
Migration and Proliferation:
[00129] In some embodiments, the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to promote proliferation and migration. In some embodiments, the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to promote proliferation. In some embodiments, the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to promote migration. In some embodiments, the anti-Met antibody promotes proliferation and/or migration. In some embodiments, the anti-Met antibody promotes proliferation. In some embodiments, the anti-Met antibody promotes migration. In some embodiments, the anti-Met antibody can be evaluated use a transwell migration assay to determined proliferation promoting ability.
[00130] In some embodiments, a migration assay can be employed to evaluate for the ability of the anti-Met antibody to promote migration and proliferation. In some embodiments, a migration assay can be employed to evaluate for the ability of the anti-Met antibody to promote migration and proliferation, wherein the migration assay is an in vitro wound closure assay In some embodiments, the migration assay can include a “scratch assay” (also referred to as a “scratch wound assay”). In some embodiments, the anti-Met antibody promotes migration and this promotion of migration and proliferation is determined and/or examined utilizing a “scratch assay”. Generally, a scratch assay method is based on when artificial gap, also referred to as a “scratch”, occurs on a confluent cell monolayer. In some embodiments, the artificial gap or scratch is a linear gap. In some embodiments, the artificial gap or scratch is a horizontal linear gap. In some embodiments, the artificial gap or scratch is a circular gap. In some embodiments, the artificial gap or scratch is a crosshatched gap. The “scratch” can be monitored for the cells on the edge of the newly created gap migrating and/or proliferating toward the opening to close/cover the “scratch”. See, for example, Liang, C., Park, A. & Guan, J. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc 2, 329-333 (2007).)
[00131] In some embodiments, the wound closure in a scratch assay is characterized as total cells migrated into the wound gap. In some embodiments, the wound closure in a scratch assay is characterized as wound closed, as a percentage. In some embodiments, the wound closure in a scratch assay is characterized as wound remaining, expressed as a percentage. In some embodiments, the wound closure in a scratch assay is characterized as size of gap. In some embodiments, the wound closure in a scratch assay is characterized as surface area of wound. In some embodiments, the wound closure in a scratch assay is characterized as time required for wound closure. In some embodiments, the wound closure in a scratch assay is characterized as rate of wound closure. In some embodiments, the wound closure in a scratch assay is characterized as EC50 from a curve generated from plotting wound closure relative to concentration of the anti-Met antibody at a given time point. In some embodiments, the migration assay can include a transwell migration assay employing comeal epithelial cells (or other cell surrogate once validation) — (e.g., wound closure) can be performed on the anti-Met antibody. In some embodiments, a transwell migration assay employing comeal epithelial as a test for wound closure potency of the anti-Met antibody. In some embodiments, the anti-Met antibody promotes wound closure as determined using a transwell migration assay.
[00132] In some embodiments, in vitro wound closure assays include but are not limited to a “scratch assay” (also referred to as a “scratch wound assay”) or a circular scratch wound method or circular scratch wound assay or circular wound closure assay.
[00133] In some embodiments, human comeal epithelial cell proliferation assays can be performed on the anti-Met antibody. In some embodiments, human comeal epithelial cell proliferation assays are indicative of a test for wound closure properties of the anti-Met antibody. In some embodiments, the anti-Met antibody promotes wound closure as determined using a human comeal epithelial cell proliferation assay.
[00134] In some embodiments, a circular scratch wound method or circular scratch wound assay or circular wound closure assay can be employed. In some embodiments, the Oris™ Cell Migration Assay platform can be employed (see. also, as described herein in Example 6).
[00135] In some embodiments, an endothelial cell tube formation assay can be performed on the anti-Met antibody. In some embodiments, an endothelial cell tube formation assays can be indicative that the anti-Met antibody is not pro-angiogenic. In some embodiments, an endothelial cell tube formation assay provides a measure of the angiogenic potential of the anti-Met antibody. In some embodiments, the anti-Met antibody exhibits anti-angiogenic properties. In some embodiments, the anti-Met antibody is anti- angiogenic properties. In some embodiments, an endothelial cell tube formation assay provides the ratio of anti- angiogenesis signals and pro-angiogenesis signals. In some embodiments, an endothelial cell tube formation assay a negative result will confirm the anthpro ratio is high and will ensure the anti-Met antibody will not promote neovascularization. In some embodiments, an endothelial cell tube formation assay a negative result will confirm the anti: pro ratio is high and will ensure the anti-Met antibody will not promote CNV (choroidal neovascularization) or neovascularization in general. In some embodiments, an inhibition of TGFb induced myofibroblast differentiation assay can be performed on the anti-Met antibody. In some embodiments, an inhibition of TGFb induced myofibroblast differentiation assay can be performed on the anti-Met antibody to show that the anti-Met antibody prevents scarring. In some embodiments, the anti-Met antibody prevents scarring. In some embodiments, the anti-Met antibody prevents scarring comeal opacity. In some embodiments, the anti-Met antibody has low angiogenesis induction. In some embodiments, the anti-Met antibody has reduced angiogenic response.
In some embodiments, the anti-Met antibody has reduced angiogenic capacity. In some embodiments, the anti-Met antibody impairs and/or reduces the normal formation of blood vessels in presence of media supportive of angiogenesis. In some embodiments, the anti- Met antibody has reduced angiogenic capacity when the anti-Met antibody is compared to untreated control. In some embodiments, the anti-Met antibody has reduced angiogenic capacity as compared to a sample treated to serum containing media. In some embodiments, the anti-Met antibody attenuates an angiogenic response. In some embodiments, the anti- Met antibody reduces the angiogenic response induce by serum free media. In some embodiments, an angiogenic response is indicated by tube formation in a cell based assay. In some embodiments, an angiogenic response is indicated by tube formation in an endothelial cell tube formation assay.
Differentiation/Scarring:
[00136] In some embodiments, the anti-Met antibody can be evaluated for the ability to prevent differentiation and prevent scarring. In some embodiments, the anti-Met antibody prevents and/or impairs scarring. In some embodiments, the anti-Met antibody prevents scarring. In some embodiments, the anti-Met antibody reduces scarring as compared to other standard treatments. In some embodiments, the anti-Met antibody prevents and/or impairs differentiation. In some embodiments, the anti-Met antibody prevents and/or impairs myofibroblast differentiation. In some embodiments, the anti-Met antibody reduces the loss of comeal transparency. In some embodiments, the anti-Met antibody reduces the loss of comeal transparency by preventing and/or impairing myofibroblast differentiation.
[00137] In some embodiments, the anti-Met antibody can be evaluated for the ability of the anti-Met antibody to modulate factors involved in differentiation. In some embodiments, the anti-Met antibody can be evaluated the ability of the anti-Met antibody to modulate factors involved in differentiation, including but not limited to TGFB2, Collagen I, Collagen III (normally upregulated during differentiation), TFGB3, MMP-2, and MMP-9 (normally downregulated during differentiation. In some embodiments, the anti-Met antibody modulates factors selected from the group consisting of TGFB2, Collagen I, Collagen III (normally upregulated during differentiation), TFGB3, MMP-2, and MMP-9 (normally downregulated during differentiation. In some embodiments, the anti-Met antibody induces a decrease in factors upregulated during normal differentiation. In some embodiments, the anti-Met antibody induces an increase in factors downregulated during normal differentiation. In some embodiments, the anti-Met antibody induces a decrease in expression of factors such as SMA. In some embodiments, the anti-Met antibody induces a decrease in expression of factors such as SMA which is indicative of anti-Met antibody potency. Neovascularization.
[00138] In some embodiments, the anti-Met antibody can be evaluated for the ability to prevent neovascularization. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces neovascularization. In some embodiments, the anti-Met antibody inhibits or does not promote neovascularization. In some embodiments, the anti- Met antibody can be evaluated for the ability to prevent angiogenesis. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces angiogenesis. In some embodiments, the anti-Met antibody inhibits angiogenesis.
[00139] In some embodiments, the anti-Met antibody can be further evaluated using depletion assays. In some embodiments, the anti-Met antibody can be depleted of specified factors. In some embodiments, the anti-Met antibody can be depleted of specified factors, including for example, but not limited to TIMP1 and/or Serpin El. In some embodiments, the anti-Met antibody can be depleted of TIMP1 and/or Serpin El. In some embodiments, the anti-Met antibody can be depleted of TIMP1. In some embodiments, the anti-Met antibody can be depleted of Serpin El.
Inflammation:
[00140] In some embodiments, the anti-Met antibody can be evaluated for the ability to prevent, impair, inhibit, and/or reduce inflammation. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation. In some embodiments, the anti-Met antibody inhibits inflammation. In some embodiments, the anti-Met antibody is characterized in vitro and/or in vivo to determine the ability to prevent, impair, inhibit, and/or reduce inflammation. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation in vitro and/or in vivo. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation in vitro. In some embodiments, the anti-Met antibody prevents, impairs, inhibits, and/or reduces inflammation or in vivo. In some embodiments, a tissue model can be employed to characterizing preventing, impairing, inhibiting, and/or reducing inflammation in vitro. In some embodiments, a 3D tissue model can be employed to characterizing preventing, impairing, inhibiting, and/or reducing inflammation in vitro. In some embodiments, a nitrogen mustard (NM) gas bum model can be used to evaluate preventing, impairing, inhibiting, and/or reducing inflammation in vitro. In some embodiments, a nitrogen mustard (NM) gas bum model can be used to evaluate preventing, impairing, inhibiting, and/or reducing inflammation in vitro and as a surrogate for in vivo conditions. . In some embodiments, the cytokine profile in response to treatment with and/or administration of the anti-Met antibody can be determined. In some embodiments, the levels of specific cytokines can be determined. In some embodiments, the level of IL-8 can be determined. In some embodiments, the level of IL-8 expression can be reduced in tissues treated with the anti-Met antibody. In some embodiments, the level of IL-8 expression is reduced in tissues treated with the anti-Met antibody and this is indicative of preventing, impairing, inhibiting, and/or reducing inflammation.
H. METHODS OF TREATMENT
[00141] The present disclosure also provides methods of treatment using the anti-Met antibody of the present disclosure. In particular, the anti-Met antibody finds use in the treatment of ocular conditions. In particular, the anti-Met antibody finds use in the treatment of ocular conditions, including but not limited to ocular diseases. In some embodiments, the ocular disease is associated with the ocular surface. In some embodiments, the ocular disease is associated with damaged ocular tissue and/or damaged ocular tissue indications. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including accelerating wound healing. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing scarring. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing inflammation. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing inflammation and thus promoting growth. In some embodiments, the anti-Met antibody finds use in treating ocular conditions such as reducing inflammation at the ocular surface. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing neovascularization. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including reducing neovascularization in the cornea. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, including dry eye treatment (including, for example, treatment of severe dry eye, including where the epithelial cells are damaged). In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as restoring the integrity to damaged ocular tissue. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as accelerating the healing of damaged ocular tissue. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as regenerating damaged ocular nerve tissue. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as regenerating damaged ocular nerve tissue associated with Persistent Comeal Epithelial Defect (PCED). In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as PCED. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as inflammatory damage to the eye surface. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as for example GvHD and/or Sjogrens syndrome. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as surgical debridement. In some embodiments, the anti-Met antibody finds use in the treatment of ocular conditions, such as contact lens wear.
[00142] In some embodiments, the anti-Met antibody finds use in accelerating wound healing. In some embodiments, the anti-Met antibody finds use in reducing scarring. In some embodiments, the anti-Met antibody finds use in reducing inflammation. In some embodiments, the anti-Met antibody finds use in reducing inflammation and thus promoting growth. In some embodiments, the anti-Met antibody finds use in reducing inflammation at the ocular surface. In some embodiments, the anti-Met antibody finds use in reducing neovascularization. In some embodiments, the anti-Met antibody finds use in reducing neovascularization in the cornea. In some embodiments, the anti-Met antibody finds use in the protection and repair of retinal epithelial cells and retinal ganglion cells. In some embodiments, the anti-Met antibody finds use in induction of trabecular meshwork regeneration and reduction of intraocular pressure.
[00143] In some embodiments, the composition comprising the anti-Met antibody is administered for the treatment of an ocular disease. In some embodiments, treatment comprises administering to a patient in need thereof therapeutically effective amount of an anti-Met antibody composition as described herein to a patient in need thereof. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to promote or induce ocular wound healing. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to reduce and/or inhibit neovascularization, reduce and/or inhibit scarring, promote and/or preserve vision, and/or increasing wound closure rate (e.g., decreasing wound closure time). In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit neovascularization. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit reducing scarring. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to promote and/or preserve vision. In some embodiments, the anti-Met antibody is administered to promote and/or induce closing wound faster wound closure (e.g., reduce the amount of time required for wound closure). In some embodiments, the anti-Met antibody prevents, reduces, and/or inhibits or does not promote neovascularization and reducing scarring in order to promote vision preservation. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit neovascularization and reducing scarring in order to promote vision preservation. In some embodiments, the anti-Met antibody prevents, reduces, and/or inhibits inflammation. In some embodiments, the anti-Met antibody is administered to a patient in need thereof in order to prevent, reduce, and/or inhibit inflammation.
[00144] In some embodiments, the anti-Met antibody is administered for the treatment of a visual dysfunction following traumatic injury to ocular structures. In some embodiments, treatment comprises administering to a patient in need thereof a therapeutically effective amount of an anti-Met antibody composition as described herein
[00145] In some embodiments, the anti-Met antibody composition is administered for the treatment of a traumatic injury of the optic nerve degeneration following concussive injury. In some embodiments, the concussive injury to the eye is selected from the group consisting of ocular contusion and blunt injury to the eye.In some embodiments, the anti- Met antibody composition is administered for the treatment of a traumatic injury of the optic nerve. In some embodiments, treatment comprises administering to a patient in need thereof a therapeutically effective amount of an anti-Met antibody as described herein.
[00146] In some embodiments, the anti-Met antibody composition is administered for ameliorating optic nerve degeneration following concussive injury to the eye. In some embodiments the method for ameliorating optic nerve degeneration comprises administering to the patient a therapeutically effective amount of an anti-Met antibody as described herein. In some embodiments, the concussive injury to the eye is selected from the group consisting of ocular contusion and blunt injury to the eye. In some embodiments, the concussive injury to the eye an ocular contusion. In some embodiments, the concussive injury to the eye a blunt injury to the eye. [00147] Efficacy readouts can include a reduced in symptoms and/or decreased disease state, including for example, increased quality of life. In some embodiments, reduced in symptoms and/or decreased disease state by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy. In some embodiments, reduction in inflammation by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy. In some embodiments, a reduction in scarring by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy. In some embodiments, a reduction in neovascularization by 10%, 20%, 30%,
40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% is indicative of therapeutic efficacy.
[00148] In some embodiments, the disease or conditions an ocular disease or ocular condition. In some embodiments, the disease or condition is a visual dysfunction following traumatic injury to ocular structures. In some embodiments, the disease or condition is a concussive (e.g., blunt or non-blunt) injury to the eye. In some embodiments, the disease or condition is a bum, including a chemical bum to the eye.
[00149] In some embodiments, the anti-Met antibody composition is administered to a particular targeted area. In some embodiments, the particular targeted area is the eye. In some embodiments, the anti-Met antibody composition is administered to a particular targeted area and is formulated so as not to spread to other surrounding areas.
[00150] In some embodiments, the anti-Met antibody composition is administered to a particular targeted area and is formulated so as not to spread to other surrounding areas.
[00151] In some embodiments, the anti-Met antibody composition is administered to a particular targeted area and is formulated to stay in the targeted area for at least 1 minute, at least about 2 minutes, 3 at least about minutes, at least about 4 minutes, at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 20 minutes, at least about 30 minutes, at least about 40 minutes, at least about 50 minutes, at least about 60 minutes, at least about 70 minutes, at least about 80 minutes, at least about 90 minutes, or at least about 2 hours.
[00152] In some embodiments, the anti-Met antibody is administered to an affected area immediately after the wound or injury. In some embodiments, the anti-Met antibody is administered to an affected area within 15 seconds, 30 seconds, 1 minutes, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 70 minutes, 80 minutes, 90 minutes, 2 hours, 4 hours, 8 hours, 12 hours, 24 hours, 36 hours, 48 hours, or 96 hours.
[00153] In some embodiments, the anti-Met antibody is administered topically. In some embodiments, the anti-Met antibody is administered by subconjunctival injection. In some embodiments, the anti-Met antibody compositions exhibit ultrapotency when administered to a subject in need thereof. In some embodiments, the anti-Met antibody is administered topically once, two, three, four, five, and/or up to six times daily. In some embodiments, the anti-Met antibody compositions allow for therapeutic efficacy with one drop or one administration per day. In some embodiments, one drop is administered 1, 2, 3, 4, 5, or 6 times per day. In some embodiments, one drop is administered at 1 hour, 2 hour, 3 hour, or 4 hour intervals. In some embodiments, one drop is administered at least once per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least twice per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 3 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 4 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 5 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks. In some embodiments, one drop is administered at least 6 times per day for 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks.
[00154] In some embodiments, the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 2 pg - 20 pg of anti-Met antibody per mL; ii. 2 mg - 3 mg monobasic sodium phosphate per mL; iii. 11 mg - 12 mg dibasic sodium phosphate per mL; iv. 11.5 mg - 13 mg mannitol per mL; v. 23 mg - 24 mg trehalose dihydrate; vi. 0.5 mg - 2 mg hypromellose per mL; and wherein the pH is about 4.7 to about 7.5. [00155] In some embodiments, the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 0.004% - 0.08 % w/w of anti-Met antibody ii. 4 % - 5 % w/w monobasic sodium phosphate; iii. 21.5 % - 23 % w/w dibasic sodium phosphate; iv. 23 % - 25 % w/w mannitol; v. 46 % - 48 % w/w trehalose dehydrate; vi. 1 % - 3 % w/w hypromellose; and wherein the pH is about 4.7 to about 7.5.
[00156] In some embodiments, the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 2 pg - 20 pg of anti-Met antibody per mL; ii. 2 mg - 3 mg monobasic sodium phosphate per mL; iii. 11 mg - 12 mg dibasic sodium phosphate per mL; iv. 11.5 mg - 13 mg mannitol per mL; v. 23 mg - 24 mg trehalose dihydrate; vi. 0.5 mg - 2 mg optionally hypromellose per mL; and wherein the pH is about 4.7 to about 7.5.
[00157] In some embodiments, the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 0.004% - 0.08 % w/w of anti-Met antibody ii. 4 % - 5 % w/w monobasic sodium phosphate; iii. 21.5 % - 23 % w/w dibasic sodium phosphate; iv. 23 % - 25 % w/w mannitol; v. 46 % - 48 % w/w trehalose dehydrate; vi. 1 % - 3 % w/w optionally hypromellose; and wherein the pH is about 4.7 to about 7.5. [00158] In some embodiments, the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 2 pg - 20 pg of anti-Met antibody per mL; ii. 2 mg - 3 mg monobasic sodium phosphate per mL; iii. 11 mg - 12 mg dibasic sodium phosphate per mL; iv. 11.5 mg - 13 mg mannitol per mL; v. 23 mg - 24 mg trehalose dihydrate; and wherein the pH is about 4.7 to about 7.5.
[00159] In some embodiments, the present disclosure provides a method of treatment for an ocular condition in a subject in need thereof comprising administering to the subject an anti-Met antibody composition, wherein the anti-Met antibody composition is a stable anti-Met antibody formulation comprising: i. 0.004% - 0.08 % w/w of anti-Met antibody ii. 4 % - 5 % w/w monobasic sodium phosphate; iii. 21.5 % - 23 % w/w dibasic sodium phosphate; iv. 23 % - 25 % w/w mannitol; v. 46 % - 48 % w/w trehalose dehydrate; and wherein the pH is about 4.7 to about 7.5.
I. KIT
[00160] A kit can include an anti-Met antibody in a container or the conditioned media for use in preparing an anti-Met antibody, also in a container, as disclosed herein, and instructions for use. Additionally, a kit can include components for mixing to prepare a solution for use in an ocular treatment, and instructions for mixing and use.
[00161] The container can include at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which an anti-Met antibody in a container or the conditioned media for use in preparing an anti-Met antibody, and in some instances, suitably aliquoted. Where an additional component is provided, the kit can contain additional containers into which this component may be placed. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained. Containers and/or kits can include labeling with instructions for use and/or warnings.
[00162] The present disclosure is further illustrated by the following examples, which should not be construed as further limiting. The contents of all figures and all references, Genbank sequences, patents and published patent applications cited throughout this application are expressly incorporated herein by reference.
[00163] The present invention can provide kits comprising a panel of tests and/or assays for characterizing an anti-Met antibody, wherein the panel comprises at least two characterization assays, wherein characterization assays are selected from the group consisting of physical component characterizations, oxidative stress assays, safety analyses, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays. In some embodiments, the panel of tests and/or assays identifies an anti-Met antibody as described herein.
[00164] The present invention can provide kits comprising a panel of tests and/or assays for determining consistency between anti-Met antibody lots, wherein the panel comprises one or more characterization assays, wherein characterization assays are selected from the group consisting of physical component characterizations, oxidative stress assays, safety analyses, stability assays, proliferation assays, migration assays, neovascularization assays, differentiation/scarring assays, inflammation assays, and/or an epithelial barrier integrity assays. In some embodiments, the panel of tests and/or assays identifies an anti- Met antibody as described herein.
EXAMPLES
EXAMPLE 1: Agonist Anti-Met monoclonal antibody (HR11)
Summary:
[00165] The HGF/Met axis is known to play a prominent role in comeal wound healing. In particular, activation of the pathway through the Met receptor has shown to accelerate wound healing, with potential for anti-scarring effects as well.
[00166] Reference papers (linked) on HGF and comeal wound healing: • Hepatocyte Growth Factor Suppresses Inflammation and Promotes Epithelium Repair in Comeal Injury
• The role of hepatocyte growth factor in comeal wound healing
• Restoration of Comeal Transparency by Mesenchymal Stem Cells
• Control of Scar Tissue Formation in the Cornea: Strategies in Clinical and Comeal Tissue Engineering
Sequence (single leter amino acid code) :
[00167] Heavy Chain - Human IgGl
• Artificial signal peptide-Heavy chain variable region (human) — humanlgGl constant
MGWSCIILFLVATATGVHSEVQLVESGGGLVQPGGSLRLSCAASGFTFSSYAMSW VRQAPGKGLEWVSAISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCARIVTASWGRWFDSWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAAL GCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYI CNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT PEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVV SVLTVLH QDWLNGKEYKCKV SNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQV SLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK(SEQ ID NO: 17)
[00168] Ligh Chain - Human Ig Kappa Constant
• Artificial signal peptide— light chain variable region (human)-human Ig kappa constant
MGWSCIILFLVATATGVHSDIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQ KPGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPF TF GPGTKVDIKRTV AAP SVFIFPPS DEQLKS GT AS V V CLLNNF YPRE AKV QWKVDN ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC (SEQ ID NO: 19)
EXAMPLE 2: Anti-Met monoclonal antibody expression constructs and cloning strategy
[00169] Order Number: U1125EC070-3 [00170] Protein Name: HR11
[00171] Shipping Condition: Dry Ice
[00172] Lot Number: U1125EC070-3/P7EC001
[00173] Codon Optimization: Yes
[00174] Expression System & Vector: Mammalian, Expression Vector pcDNA3.4 [00175] Host cell line: Expi293F
[00176] Expression Optimization: Yes
[00177] Expression Scale: Expi293F™ Expression Medium culture
[00178] Purification: Antibody was obtained from supernatant of cell culture, one-step purification by HiTrap MabSelect SuRe
[00179] Package: 12.00 mg. 1.00 ml/tube, 4 tubes
[00180] Concentration: 3.00 mg/ml, was determined by A280 (Extinction coefficients: 1.503)
[00181] Purity: About 95%, was estimated by densitometric analysis of the Coomassie Blue-stained SDS-PAGE gel under non-reducing condition
[00182] Endotoxin Level: 0.1 EU/mg(LAL Endotoxin Assay Kit, GenScript, Cat.No.L00350)
[00183] Sterility: Sterilized via a 0.22 pm filter and packaged aseptically
[00184] Storage and Handling: Store at -80°C. Aliquots should be stored at the same temperature after first use to avoid multiple freeze-thaws Storage Buffer: PBS pH 7.2
[00185] SDS-PAGE & Western blot analysis (Figure 2).
Packing list
[00186] Protein (Shipping Condition: -80°C Dry Ice, Store at -80°C)
[00187] Protein name: HR11
[00188] Concentration: 3.00 mg/ml
[00189] Purity: -95%
[00190] Endotoxin Level: 0.1 EU/mg [00191] Buffer: PBS pH 7.2
[00192] Lot: U1125EC070-3/P7EC001
[00193] Total: 12.00 mg. 1.00 ml/tube, 4 tubes
Cloning strategy [00194] Figures 3A-3G illustrate the exemplary amino acid sequences and nucleic acid sequences ofHRll.
General procedure
Plasmid Preparation:
[00195] 1) Transfection grade plasmids (maxi-prep from previous order U9700DL270- 4) were maxi-prepared for Expi293F cell expression.
Cell Culture and Transient Transfection:
[00196] Expi293F cells were grown in serum-free Expi293™ Expression Medium (Thermo Fisher Scientific).
[00197] The cells were maintained in Erlenmeyer Flasks (Coming Inc.) at 37°C with 8% C02 on an orbital shaker (VWR Scientific).
[00198] One day before transfection, the cells were seeded at an appropriate density in Coming Erlenmeyer Flasks.
[00199] On the day of transfection, DNA and ExpiFectamine™ 293 reagent were mixed at an optimal ratio and then added into the flask with cells ready for transfection. [00200] The recombinant plasmids encoding target antibody were transiently co transfected into suspension Expi293F cell cultures.
[00201] The cell culture supernatants collected on day 6 were used for purification. Purification and Analysis:
[00202] Cell culture broth was centrifuged and followed by filtration. [00203] Filtered cell culture supernatant was loaded onto an affinity purification column at an appropriate flowrate.
[00204] After washing and elution with appropriate buffers, the eluted fractions were pooled and buffer exchanged to the final formulation buffer. [00205] The purified protein was analyzed by SDS-PAGE, Western blot analysis to determine the molecular weight and purity.
[00206] The concentration was determined by A280 method.
EXAMPLE 3: Anti-Met (HR11): mechanical wound efficacy
[00207] Met mAb (HR11) peptide promotes comeal wound healing with reduced scarring. A 3.0 mm epithelial defect was created in mouse corneas using a trephine. Wounds were treated with HR11 twice daily at 2.0 mg/mL for seven days. Depicted are representative images of eyes treated with HR11 or vehicle control (Figure 4).
[00208] Percentage of wounds completely closed - HR11 mAb promotes full wound closure whereas vehicle control cannot fully close wounds. A 3.0 mm epithelial defect was created in mouse corneas. Wounds were treated with HR11 twice daily at 3.0 mg/mL for seven days.
Figure imgf000070_0001
Peptide 67 67
[00209] HR11 mAb promotes comeal wound healing. A 3.0 mm epithelial defect was created in mouse corneas. Wounds were treated with HR11 twice daily at 3.0 mg/mL for seven days (Figure 6).
[00210] The examples set forth above are provided to give those of ordinary skill in the art a complete disclosure and description of how to make and use the embodiments of the compositions, systems and methods of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Modifications of the above-described modes for carrying out the invention that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the invention pertains. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually.
[00211] All headings and section designations are used for clarity and reference purposes only and are not to be considered limiting in any way. For example, those of skill in the art will appreciate the usefulness of combining various aspects from different headings and sections as appropriate according to the spirit and scope of the invention described herein.
[00212] All references cited herein are hereby incorporated by reference herein in their entireties and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.
[00213] Many modifications and variations of this application can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments and examples described herein are offered by way of example only, and the application is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which the claims are entitled.

Claims

WHAT IS CLAIMED IS: Product claims
1. A composition comprising an anti-Met antibody, wherein the antibody comprises: a) a vhCDRl, vhCDR2, and vhCDR3 from SEQ ID NO:5; and b) a vlCDRl, vlCDR2, and vlCDR3 from SEQ ID NO: 13.
2. A composition comprising an anti-Met antibody, wherein the antibody comprises a heavy chain variable domain having at least 80% identity to SEQ ID NO:5 and a light chain variable domain having at least 80% identity to SEQ ID NO: 13.
3. A composition comprising an anti-Met antibody, wherein the antibody comprises: a) a vhCDRl having 0-2 amino acid substitutions from the vhCDRl from SEQ ID NO:5; b) a vhCDR2 having 0-2 amino acid substitutions from the vhCDR2 from SEQ ID NO:5; c) a vhCDR3 having 0-2 amino acid substitutions from the vhCDR3 from SEQ ID NO:5; d) a vlCDRl having 0-2 amino acid substitutions from the vlCDRl from SEQ ID NO: 13 e) a vlCDR2 having 0-2 amino acid substitutions from the vlCDR2 from SEQ ID NO: 13; and
1) a vlCDR3 having 0-2 amino acid substitutions from the vlCDR3 from SEQ ID NO: 13.
4. The composition of any of claims 1 to 3, wherein the antibody comprises a heavy chain variable domain comprising SEQ ID NO:5 and a light chain variable domain comprising SEQ ID NO: 13.
5. The composition of any of claims 1 to 4, wherein the antibody comprises a CH1- hinge-CH2-CH3 region from human IgGl, IgG2, IgG3, or IgG4.
6. The composition of any of claims 1 to 5, wherein the antibody comprises a CH1- hinge-CH2-CH3 region from human IgGl.
7. The composition of any of claims 1 to 6, wherein the antibody comprises a CL region of human kappa constant.
8. The composition of any of claims 1 to 7, wherein the heavy chain comprises SEQ ID NO: 1 and the light chain comprises SEQ ID NO:9.
9. The composition of any of claims 1 to 8, wherein the heavy chain further comprising a first signal peptide sequence of SEQ ID NO: 3 and the light chain further comprising a second signal peptide sequence of SEQ ID NO: 11.
Nucleic acid/expression vector/host cell/method of making claims
10. A nucleic composition comprising: a) a first nucleic acid encoding the heavy chain variable domain of the anti- Met antibody according to any of claims 1 to 9; and b) a second nucleic acid encoding the light chain variable domain of the same anti-Met antibody.
11. An expression vector composition comprising: a) a first expression vector comprising the first nucleic acid of claim 10; and b) a second expression vector comprising the second nucleic acid of claim 10.
12. An expression vector composition comprising an expression vector comprising the first and second nucleic acids of claim 10.
13. A host cell comprising the expression vector composition of any of claims 10 to 12.
14. A method of making an anti-Met antibody comprising: a) culturing the host cell of claim 13 under conditions wherein the antibody is expressed; and b) recovering the anti-Met antibody.
Method of treatment claims
15. A method of treating an ocular condition in a subject in need thereof, wherein the method comprises administering to the subject an anti-MET antibody, the antibody comprising: a) a vhCDRl, vhCDR2, and vhCDR3 from SEQ ID NO:5; and b) a vlCDRl, vlCDR2, and vlCDR3 from SEQ ID NO: 13.
16. A method of treating an ocular condition in a subject in need thereof, wherein the method comprises administering to the subject an anti-MET antibody, wherein the antibody comprises a heavy chain variable domain having at least 80% identity to SEQ ID NO: 5 and a light chain variable domain having at least 80% identity to SEQ ID NO: 13.
17. A method of treating an ocular condition in a subject in need thereof, wherein the method comprises administering to the subject an anti-MET antibody, the antibody comprising: a) a vhCDRl having 0-2 amino acid substitutions from the vhCDRl from SEQ ID NO:5; b) a vhCDR2 having 0-2 amino acid substitutions from the vhCDR2 from SEQ ID NO:5; c) a vhCDR3 having 0-2 amino acid substitutions from the vhCDR3 from SEQ ID NO:5; d) a vlCDRl having 0-2 amino acid substitutions from the vlCDRl from SEQ ID NO: 13 e) a vlCDR2 having 0-2 amino acid substitutions from the vlCDR2 from SEQ ID NO: 13; and
I) a vlCDR3 having 0-2 amino acid substitutions from the vlCDR3 from SEQ ID NO: 13.
18. The method of any of claims 15 to 17, wherein the antibody comprises a heavy chain variable domain comprising SEQ ID NO:5 and a light chain variable domain comprising SEQ ID NO: 13.
19. The method of any of claim 15 to 18, wherein the antibody comprises a CHl-hinge- CH2-CH3 region from human IgGl, IgG2, IgG3, or IgG4.
20. The method of any of claims 15 to 19, wherein the antibody comprises a CHl-hinge- CH2-CH3 region from human IgGl .
21. The method of any of claims 15 to 20, wherein the antibody comprises a CL region of human kappa constant.
22. The method of any of claims 15 to 21, wherein the heavy chain comprises SEQ ID NO: 1 and the light chain comprises SEQ ID NO:9.
23. The method of any of claims 15 to 22, wherein the heavy chain further comprising a first signal peptide sequence of SEQ ID NO:3 and the light chain further comprising a second signal peptide sequence of SEQ ID NO: 11.
24. The method of any of claims 15 to 23, wherein the ocular condition is selected from the group consisting of Chronic Graft v. Host Disease (GvHD), Stevens-Johnson Syndrome, Ocular Mucous Membrane Pemphigoid, Persistent Comeal Epithelial Defect (PCED), dry eye, ocular nerve tissue damage, concussive injury to the eye (such as concussive injury, ocular contusion, or chemical bum), surgical debridement, and contact lens wear.
25. Use of the composition of any of the preceding claims for the method as described in any of the preceding claims.
26. Use of the composition of any one of the preceding claims for the manufacture of a medicament for treating an ocular condition in a subject in need thereof.
PCT/US2022/021618 2021-03-24 2022-03-23 Compositions comprising c-met agonist antibodies and methods for use in ocular treatment WO2022204332A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163165678P 2021-03-24 2021-03-24
US63/165,678 2021-03-24

Publications (1)

Publication Number Publication Date
WO2022204332A1 true WO2022204332A1 (en) 2022-09-29

Family

ID=83396072

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/021618 WO2022204332A1 (en) 2021-03-24 2022-03-23 Compositions comprising c-met agonist antibodies and methods for use in ocular treatment

Country Status (1)

Country Link
WO (1) WO2022204332A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003048328A2 (en) * 2001-12-03 2003-06-12 Abgenix, Inc. Antibodies against carboxic anhydrase ix (ca ix) tumor antigen
WO2017194613A2 (en) * 2016-05-10 2017-11-16 Immunovia Ab Method, array and use thereof
US20180327500A1 (en) * 2014-09-16 2018-11-15 Symphogen A/S Anti-met antibodies and compositions
WO2020172475A1 (en) * 2019-02-21 2020-08-27 Regeneron Pharmaceuticals, Inc. Methods of treating ocular cancer using anti-met antibodies and bispecific antigen binding molecules that bind met

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003048328A2 (en) * 2001-12-03 2003-06-12 Abgenix, Inc. Antibodies against carboxic anhydrase ix (ca ix) tumor antigen
US20180327500A1 (en) * 2014-09-16 2018-11-15 Symphogen A/S Anti-met antibodies and compositions
WO2017194613A2 (en) * 2016-05-10 2017-11-16 Immunovia Ab Method, array and use thereof
WO2020172475A1 (en) * 2019-02-21 2020-08-27 Regeneron Pharmaceuticals, Inc. Methods of treating ocular cancer using anti-met antibodies and bispecific antigen binding molecules that bind met

Similar Documents

Publication Publication Date Title
CN111138544B (en) Homodimer type bispecific antibody aiming at Her2 and CD3 and application thereof
JP7331298B2 (en) Antibody-conjugated TSLP and uses thereof
KR102412023B1 (en) Combination of lenalidomide or pomalidomide and cd38 antibody-attenuated interferon-alpha constructs, and the use thereof
KR102282134B1 (en) Human fcrn-binding modified antibodies and methods of use
AU2011282579B2 (en) Fusion proteins of natural human protein fragments to create orderly multimerized immunoglobulin Fc compositions
EP3478717B1 (en) Novel antibody format
BR112020023746A2 (en) antibody, pharmaceutical composition, method for treating cancer, isolated nucleic acid, vector, host cell, process for producing an antibody and diagnostic reagent
TWI653243B (en) Anti-IGF-1R antibody against FcRn binding and use thereof for treating vascular eye diseases
JP7120989B2 (en) Antibodies with low immunogenicity and uses thereof
JP2022522709A (en) Antibodies that bind to CD40 and their use
MX2014011688A (en) Antibodies to bradykinin b1 receptor ligands.
KR20220041881A (en) Anti-PVRIG antibody formulations and uses thereof
JP2023527583A (en) Antibodies that bind to LAG3 and uses thereof
CN113795509A (en) anti-SEMA 3A antibodies and their use for treating ocular or ocular diseases
KR20220103709A (en) Methods of treating cancer using an anti-OX40 antibody in combination with an anti-TIGIT antibody
EP2966085A1 (en) Antibody IgG1 with a modified heavy chain constant region
US20230365680A1 (en) Combination therapy with anti-pvrig antibodies formulations, anti-tigit antibodies, and anti-pd-1 antibodies
US20240279324A1 (en) Multispecific antagonists
WO2022204332A1 (en) Compositions comprising c-met agonist antibodies and methods for use in ocular treatment
AU2015200330A1 (en) Fusion proteins of natural human protein fragments to create orderly multimerized immunoglobulin Fc compositions
US20230212291A1 (en) Methods of cancer treatment using anti-ox40 antibodies in combination with anti-pd1 or anti-pdl1 antibodies
CN114729048A (en) Methods of treating cancer using anti-OX 40 antibodies in combination with TLR agonists
WO2024182443A2 (en) Triple combination therapy with anti-pvrig antibodies, anti-tigit antibodies, and pembrolizumab
TW202408571A (en) Methods of treating lymphoma using anti-tigit antibodies

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22776606

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22776606

Country of ref document: EP

Kind code of ref document: A1