WO2019200006A2 - Novel angiopoietin 2, vegf dual antagonists - Google Patents

Novel angiopoietin 2, vegf dual antagonists Download PDF

Info

Publication number
WO2019200006A2
WO2019200006A2 PCT/US2019/026838 US2019026838W WO2019200006A2 WO 2019200006 A2 WO2019200006 A2 WO 2019200006A2 US 2019026838 W US2019026838 W US 2019026838W WO 2019200006 A2 WO2019200006 A2 WO 2019200006A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
chimeric molecule
acid sequence
ang
Prior art date
Application number
PCT/US2019/026838
Other languages
French (fr)
Inventor
Yuefeng Lu
Jian-Feng Lu
Original Assignee
Askgene Pharma 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 Askgene Pharma Inc. filed Critical Askgene Pharma Inc.
Priority to CN201980024414.9A priority Critical patent/CN112262158A/en
Priority to EP19784270.1A priority patent/EP3775267A4/en
Publication of WO2019200006A2 publication Critical patent/WO2019200006A2/en

Links

Classifications

    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • 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
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • the present application relates to novel molecules comprising binding domains to both VEGF and Ang2.
  • Angiogenesis is implicated in the pathogenesis of a variety of disorders including solid tumors, intraocular neovascular syndromes such as proliferative retinopathies or age- related macular degeneration (AMD), rheumatoid arthritis, and psoriasis (Folkman, J., et al., J. Biol. Chem.267 (1992) 10931-10934; Klagsbrun, M., et al, Annu. Rev. Physiol.53 (1991) 217- 239; and Garner, A., Vascular Diseases, in: Pathobiology of Ocular Disease, A Dynamic Approach, Garner, A., and Klintworth, G. K.
  • VEGF/VEGF-A Human vascular endothelial growth factor
  • VEGF expression contributes to the development of solid tumors by promoting tumor angiogenesis and to the etiology of several additional diseases that are characterized by abnormal angiogenesis (Kim, K. J., et al., 1993. Nature (London) 362, 841– 844; Millauer, B., et al., 1994. Nature (London) 367, 576–579). Consequently, inhibition of VEGF signaling abrogates the development of a wide variety of tumors.
  • retinopathies in which partial or general ischemia of the retina is accompanied by overexpression of VEGF and hyperproliferation of blood vessels, blindness can result (Aiello, L. P et al., 1994. N. Engl. J. Med.331, 1480–1487; Adamis, A. P., et al., , Am. J. Ophthalmol. 118, 445–450). Inhibition of VEGF expression in such disease states can treat or prevent resulting blindness.
  • Ang2 Human angiopoietin-2 (ANG-2 or Ang-2 or Ang2) (alternatively abbreviated with ANGPT2 or ANG2) is described in Maisonpierre, P.C., et al, Science 277 (1997) 55-60 and Cheung,A.H., et al, Genomics 48 (1998) 389-91.
  • Ang2 plays an important role in angiogenesis and its expression levels have been correlated with cancer and eye diseases (Gerald D et al., Cancer Res.2013, 73(6):1649-57; Watanabe et al., Am J Ophthalmol.2005, 139(3):476-81).
  • Dual antagonist RG7716 demonstrated superior efficacy than VEGF antagonist ranibizumab in a recent clinical trial.
  • the reported dosage for RG7716 at 6 mg per dose was rather high considering the volume of administration to eye is typically low, e.g. 50 micoL). This could require a concentration of 120 mg/ml, a significant challenge for formulation development.
  • a dual antagonist with stronger binding affinities to VEGF and/or Ang2 is needed.
  • the present invention includes bi-specific molecules with enhanced binding ability and which result in a reduction in the severity of a disease in a patient treated with a molecule disclosed herein.
  • the present disclosure relates to novel bispecific chimeric molecules comprising binding domains to both VEGF and Ang-2. Further disclosed are methods of using said chimeric molecules to treat a patient of cancer, proliferative retinopathy, neovascular glaucoma, macular edema, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR).
  • proliferative retinopathy neovascular glaucoma
  • macular edema macular edema
  • wAMD wet age-related macular degeneration
  • RVO macular edema following retinal vein occlusion
  • DME diabetic macular edema
  • DR diabetic retinopathy
  • said chimeric molecule comprises one or two VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein:
  • said Ang-2 antagonist peptide comprises an amino acid sequence selected from SEQ ID NO: 8-14;
  • said VEGF-binding moiety is an antibody, an Fab or an scFv; and wherein said antibody, Fab or scFv comprises light chain CDRs as derived from a light chain with an amino acid sequence as shown in SEQ ID NO: 4, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6, and heavy chain CDRs as derived from a heavy chain with an amino acid sequence as shown in SEQ ID NO: 5, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6.
  • said VEGF binding moiety comprises an antibody with a light chain amino acid sequence that is at least 95% identical to that of SEQ ID NO: 4, and heavy chain amino acid sequence that is at least 99% identical to that of SEQ ID NO: 7.
  • said Ang-2 antagonist peptide is fused to one or both of the N- terminals of the heavy chains (HC) of the said antibody optionally through a peptide linker.
  • the peptide-HC fusion polypeptide comprises an amino acid sequence that is at least 99% identical as one selected from SEQ ID NOS:29, 30, and SEQ ID NO:33.
  • said Ang-2 antagonist peptide is fused to one or both of the C- terminal of the heavy chain of the said antibody optionally through a peptide linker.
  • the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence that is at least 99% identical or 100% identical as one selected from SEQ ID NOS: 31, 32, and 34.
  • said Ang-2 antagonist polypeptide is fused to the N-terminals or the C-terminals of the heavy chain of said antibody through a peptide linker; and wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NO: 37, 39, 41, 43, 45, 47, 49, 51, and 53.
  • said VEGF binding moiety is an Fab with a light chain amino acid sequence of at least 95% identity to SEQ ID NO: 4, and a heavy chain amino acid sequence of at least 95% identity to SEQ ID NO: 5.
  • th-e Ang2 antagonist peptide is fused to the N-terminal of the heavy chain of said Fab molecule through a peptide linker.
  • the Ang2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO:19 or SEQ ID NO:20.
  • the Ang-2 antagonist peptide is fused to the C-terminal of the heavy chain of said Fab molecule through a peptide linker.
  • the peptide- heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO: 25 or SEQ ID NO:26.
  • said VEGF binding moiety is an scFv with an amino acid sequence that has at least 95% identity to SEQ ID NO: 6.
  • the Ang-2 antagonist peptide is fused to the N-terminal of the scFv; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NOS:21 and 22.
  • the Ang-2 antagonist peptide is fused to the C-terminal of the scFv; and wherein the peptide-scFv fusion polypeptide has an amino acid sequence selected from SEQ ID NO:27 and SEQ ID NO:28.
  • said chimeric molecule comprises a fusion protein that has one or more VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein said VEGF binding moiety is a VEGF trap with an amino acid sequence having at least 95% identity to SEQ ID NO: 3; wherein said chimeric molecule comprises two identical polypeptide chains, which have an amino acid sequence at least 99% identical as one selected from SEQ ID NOS:15-17, 23 and 24.
  • polynucleotide or polynucleotides encoding any one of the above said chimeric molecules.
  • said polynucleotide comprises a DNA sequence as one selected from SEQ ID NO: 35, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54 and 56.
  • an expression vector or vectors comprising the above said polynucleotide or polynucleotides.
  • composition comprising the chimeric molecule of any one of the above said chimeric molecule and a pharmaceutically acceptable excipient.
  • a method of treating a patient with cancer, proliferative retinopathy, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR) comprising administering to a subject of above said pharmaceutical composition.
  • wAMD wet age-related macular degeneration
  • RVO retinal vein occlusion
  • DME diabetic macular edema
  • DR diabetic retinopathy
  • FIG. 1 Protein A Affinity Chromatography. Approximately 150 ml of the clarified HEK 293 cell culture medium of the transient expression of AMD-B was loaded to a Protein A column (1 x 17 cm (Diameter x Height) of Captiv A Protein A resin) at 3 ml/min. The protein A column was equilibrated with an equilibration buffer (25 mM Tris Buffer, 100 mM NaCl, PH approximately 7.2). The column was washed with the Equilibration buffer and eluted with 2 M ariginine solution, PH 4.
  • an equilibration buffer 25 mM Tris Buffer, 100 mM NaCl, PH approximately 7.2
  • FIG. 4A Blocking of Binding of Ang-1 and Ang-2 to Tie-2 by AMD-A and AMD- B
  • fusion proteins and chimeric molecules which comprise two components: an Ang-2 antagonist peptide operationally linked to a VEGF binding domain, which is selected from an anti-VEGF antibody, an anti-VEGF Fab, an anti-VEGF scFv, or a VEGF receptor extracellular domain-Fc fusion protein (or VEGF Trap).
  • the Ang-2 antagonist peptide and VEGF binding domain are each defined below with reference to percent identity to a reference sequence.
  • chimeric molecules to treat a patient of cancer, proliferative retinopathy, neovascular glaucoma, macular edema, wet age- related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR).
  • proliferative retinopathy neovascular glaucoma
  • macular edema wet age- related macular degeneration
  • wAMD macular edema following retinal vein occlusion
  • DME diabetic macular edema
  • DR diabetic retinopathy
  • Reference to“about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se.
  • description referring to“about X” includes description of“X.”
  • use of“about” preceding any series of numbers includes“about” each of the recited numbers in that series.
  • description referring to “about X, Y, or Z” is intended to describe“about X, about Y, or about Z.”
  • the term“antigen-binding moiety” refers to a polypeptide or a set of interacting polypeptides that specifically bind to an antigen, and includes, but is not limited to, an antibody or antibody fragment, such as a monoclonal antibody, polyclonal, a chimeric antibody, a CDR- grafted antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody (dAb), a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, a functionally active epitope-binding fragment thereof, bifunctional hybrid antibodies, a single chain antibody, and a Fc-containing polypeptide, such as an immunoadhesion.
  • an antibody or antibody fragment such as a monoclonal antibody, polyclonal, a chimeric antibody, a CDR
  • the antibody may be of any heavy chain isotype (e.g., IgG, IgA, IgM, IgE, or IgD). In some embodiments, the antibody may be of any light chain isotype (e.g., kappa or gamma). The antibody may be non-human (e.g.,
  • the antibody is a derivatized antibody.
  • an effective amount refers to an amount of a compound or composition sufficient to treat a specified disorder, condition, or disease, such as ameliorate, palliate, lessen, and/or delay one or more of its symptoms.
  • an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation in the cancer.
  • the effective amount is an amount sufficient to delay development of a cancer.
  • the effective amount is an amount sufficient to prevent or delay recurrence.
  • An effective amount can be administered in one or more administrations.
  • the effective amount of the drug or composition may: (i) reduce the number of epithelioid cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop the cancer cells infiltration into peripheral organs; (iv) inhibit (e.g., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
  • the term“fused” or“fusion” in reference to two or more polypeptide sequences refers to joining of the polypeptide sequences through a backbone peptide bond.
  • pharmaceutically acceptable when used to refer to a compound or composition means that the compound or composition is suitable for administration to a subject, including a human subject, to achieve the treatments described herein, without unduly deleterious side effects in light of the severity of the disease and necessity of the treatment.
  • subject refers to a mammal and includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate.
  • treatment or“treating” is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: alleviating one or
  • treatment is a reduction of a pathological consequence of a disease (such as cancer).
  • the methods of the invention contemplate any one or more of these aspects of treatment.
  • the fusion protein or chimeric molecule comprises an Ang-2 antagonist peptide component, which binds to Angiopoietin 2 (Ang-2) and inhibits the binding of Ang-2 to its receptor.
  • Ang-2 antagonist peptide component which binds to Angiopoietin 2 (Ang-2) and inhibits the binding of Ang-2 to its receptor.
  • Ang-2 Angiopoietin 2
  • One example of the peptide is called 2xCon4(C), as described in WO2004/092215A2 or WO03/05134A2.
  • 2xCon4(C) has an amino acid sequence as shown in SEQ ID NO:1. Additional examples of Ang-2 binding peptides include but are not limited to: L-1-21, L1-7, L1- 10, and L1-15, as described in WO2004/092215A2. Examples of Ang-2 antagonist peptides are shown in SEQ ID NO: 8-14.
  • the chimeric molecule further comprises a VEGF-binding moiety.
  • said VEGF-binding moiety is an anti-VEGF antibody, an anti-VEGF Fab, or an anti-VEGF scFv that inhibits the binding of VEGF to its receptors.
  • VEGF antibody is bevacizumab, which has two heavy chains with amino acid sequence as shown as SEQ ID NO:1, and two light chains with amino acid sequence as shown as SEQ ID NO:2.
  • Another example is ranibizumab, an anti-VEGF Fab.
  • a third example is Brolucizumab (RTH258), which is a humanized single-chain antibody fragment (scFv) against VEGF.
  • said VEGF binding domain is a VEGF receptor-Fc fusion protein which“traps” VEGF (herein, referred to as a“VEGF trap”) and competes with the naturally occurring VEGF cellular receptor to inhibit VEGF.
  • a“VEGF trap” VEGF receptor-Fc fusion protein which“traps” VEGF
  • afilbercept which has an amino acid sequence as shown in SEQ ID NO:3.
  • the VEGF-binding moiety comprises the six complementarity determining regions (CDRs) of Brolucizumab (RTH258), Ranibizumab or Bevacizumab.
  • CDRs complementarity determining regions
  • RTH258 Brolucizumab
  • Ranibizumab Ranibizumab
  • Bevacizumab A number of CDR delineations are known in the art and are encompassed herein. A person of skill in the art can readily determine a CDR for a given delineation based on the sequence of the heavy or light chain variable region.
  • The“Kabat” Complementarity Determining Regions (CDRs) are based on sequence variability and are the most commonly used (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.
  • “Chothia” CDRs refer to the location of the structural loops (Chothia & Lesk, Canonical structures for the hypervariable regions of immunoglobulins, J. Mol. Biol., vol. 196, pp.901-917 (1987)).
  • The“AbM” CDRs represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular’s AbM antibody modeling software.
  • The“Contact” CDRs are based on an analysis of the available complex crystal structures. The residues from each of these CDRs are noted below in Table 1, in reference to common antibody numbering schemes.
  • amino acid number of antibodies refers to the Kabat numbering scheme as described in Kabat et al., supra, including when CDR delineations are made in reference to Kabat, Chothia, AbM, or Contact schemes.
  • the actual linear amino acid sequence may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a framework region (FR) or CDR of the variable domain.
  • a heavy-chain variable domain may include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted residues (e.g. residues 82a, 82b, and 82c, etc. according to Kabat) after heavy-chain FR residue 82.
  • the Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a“standard” Kabat numbered sequence.
  • the CDRs are“extended CDRs,” and encompass a region that begins or terminates according to a different scheme.
  • an extended CDR can be as follows: L24—L36, L26—L34, or L26—L36 (VL-CDR1); L46—L52, L46—L56, or L50—L55 (VL-CDR2); L91—L97 (VL-CDR3); H47—H55, H47—H65, H50—H55, H53—H58, or H53—H65 (VH-CDR2); and/or H93—H102 (VH-CDR3).
  • the Ang-2 peptide can be linked or fused to either the C- or N- terminus of the VEGF antibody (e.g., either the heavy or the light chains) or the VEGF receptor-Fc fusion protein.
  • the Fc portion of the VEGF receptor-Fc fusion protein may be located at either the C- or N-terminus of the VEGF receptor protein. The Fc portion is further defined herein.
  • the present compositions include“Fc fragments” or“Fc regions.”
  • the Fc region excludes the variable regions of the heavy and light chains, the heavy-chain constant region 1 (CH1) and the light-chain constant region 1 (CL1) of the immunoglobulin.
  • the Fc region may further include a hinge region at the heavy- chain constant region.
  • the immunoglobulin Fc region disclosed herein may contain a part
  • the immunoglobulin Fc region may be a fragment having a deletion in a relatively long portion of the amino acid sequence of CH2 and/or CH3.
  • the immunoglobulin Fc region disclosed herein may comprise 1) a CH1 domain, a CH2 domain, a CH3 domain and a CH4 domain, 2) a CH1 domain and a CH2 domain, 3) a CH1 domain and a CH3 domain, 4) a CH2 domain and a CH3 domain, 5) a combination of one or more domains and an immunoglobulin hinge region (or a portion of the hinge region), and 6) a dimer of each domain of the heavy-chain constant regions and the light-chain constant region.
  • the immunoglobulin Fc region disclosed herein includes a native amino acid sequence, or a sequence analogue thereof.
  • An amino acid sequence analogue is a sequence that is different from the native amino acid sequence due to a deletion, an insertion, a non- conservative or conservative substitution or combinations thereof of one or more amino acid residues.
  • analogues are possible, including one in which a region capable of forming a disulfide bond is deleted, or certain amino acid residues are eliminated at the N- terminal end of a native Fc form or a methionine residue is added thereto. Further, to remove effector functions, a deletion may occur in a complement-binding site, such as a C1q-binding site and an ADCC (antibody dependent cell mediated cytotoxicity) site.
  • a complement-binding site such as a C1q-binding site and an ADCC (antibody dependent cell mediated cytotoxicity) site.
  • the aforementioned Fc analogues are analogues that have a biological activity identical to the Fc region disclosed herein or improved structural stability, for example, against heat, pH, or the like.
  • these Fc regions may be obtained from native forms isolated from humans and other animals including cows, goats, pigs, mice, rabbits, hamsters, rats and guinea pigs, or may be recombinants or analogues thereof, obtained from transformed animal cells or microorganisms.
  • they may be obtained from a native immunoglobulin by isolating whole immunoglobulins from human or animal organisms and treating them with a proteolytic enzyme.
  • a human-derived Fc region is a recombinant immunoglobulin Fc region that is obtained from a microorganism.
  • the Fc region may be modified by phosphorylation, sulfation, acrylation, glycosylation, methylation, farnesylation, acetylation, amidation, and the like.
  • the immunoglobulin Fc region disclosed herein may be in the form of having native sugar chains, increased sugar chains compared to a native form or decreased sugar chains compared to the native form, or may be in a deglycosylated form. The increase, decrease or removal of the immunoglobulin Fc sugar chains may be achieved by methods common in the art, such as a chemical method, an enzymatic method and a genetic engineering method using a microorganism.
  • an immunoglobulin Fc region in a deglycosylated or aglycosylated form may be more suitable as a drug carrier.
  • deglycosylation refers to enzymatically removing sugar moieties from an Fc region
  • amino acid sequence means that an Fc region is produced in an unglycosylated form by a prokaryote, preferably E. coli.
  • the immunoglobulin Fc region may be an Fc region that is derived from IgG, IgA, IgD, IgE and IgM, or that is made by combinations thereof or hybrids thereof. In an embodiment, it is derived from IgG or IgM, which are among the most abundant proteins in human blood, and further, wherein an IgG, which is known to enhance the half-lives of ligand-binding proteins is an IgG1, IgG2a, IgG2b and/or IgG3.
  • polypeptide peptide
  • protein protein
  • 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.
  • Methods for obtaining e.g., producing, isolating, purifying,
  • compositions of the chimeric molecules are prepared by mixing the antibody fusion molecules or the antibody fusion molecule drug conjugate having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (see Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • Buffers are used to control the pH in a range which optimizes the therapeutic effectiveness, especially if stability is pH dependent. Buffers are preferably present at concentrations ranging from about 50 mM to about 250 mM.
  • Suitable buffering agents for use with the present invention include both organic and inorganic acids and salts thereof, such as citrate, phosphate, succinate, tartrate, fumarate, gluconate, oxalate, lactate, acetate. Additionally, buffers may comprise histidine and trimethylamine salts such as Tris.
  • Preservatives are added to retard microbial growth, and are typically present in a range from 0.2% - 1.0% (w/v).
  • Suitable preservatives for use with the present invention include octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium halides (e.g., chloride, bromide, iodide), benzethonium chloride; thimerosal, phenol, butyl or benzyl
  • alkyl parabens such as methyl or propyl paraben
  • catechol resorcinol
  • cyclohexanol 3- pentanol
  • m-cresol
  • Tonicity agents sometimes known as“stabilizers” are present to adjust or maintain the tonicity of liquid in a composition. When used with large, charged biomolecules such as proteins and antibodies, they are often termed“stabilizers” because they can interact with the charged groups of the amino acid side chains, thereby lessening the potential for inter- and intra- molecular interactions. Tonicity agents can be present in any amount between 0.1% to 25% by weight, or more preferably between 1% to 5% by weight, taking into account the relative amounts of the other ingredients.
  • Preferred tonicity agents include polyhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol and mannitol.
  • Non-ionic surfactants or detergents are present to help solubilize the therapeutic agent as well as to protect the therapeutic protein against agitation- induced aggregation, which also permits the formulation to be exposed to shear surface stress without causing denaturation of the active therapeutic protein or antibody.
  • Non-ionic surfactants are present in a range of about 0.05 mg/ml to about 1.0 mg/ml, preferably about 0.07 mg/ml to about 0.2 mg/ml.
  • Suitable non-ionic surfactants include polysorbates (20, 40, 60, 65, 80, etc.), polyoxamers (184, 188, etc.), PLURONIC® polyols, TRITON®, polyoxyethylene sorbitan monoethers (TWEEN®-20, TWEEN®-80, etc.), lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose.
  • Anionic detergents that can be used include sodium lauryl sulfate, dioctyle sodium sulfosuccinate and dioctyl sodium sulfonate.
  • Cationic detergents include benzalkonium chloride or benzethonium chloride.
  • compositions may comprise as - or in addition to - the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s) or solubilizing agent(s).
  • compositions useful in the formulation may be different composition/formulation requirements dependent on the different delivery systems.
  • pharmaceutical compositions useful in the formulation may be different composition/formulation requirements dependent on the different delivery systems.
  • pharmaceutical compositions useful in the formulation may be different composition/formulation requirements dependent on the different delivery systems.
  • present invention may be formulated to be administered using a mini-pump or by a mucosal route, for example, as a nasal spray or aerosol for inhalation or ingestible solution, or parenterally in which the composition is formulated by an injectable form, for delivery, by, for example, an intravenous, intramuscular or subcutaneous route.
  • the formulation may be designed to be administered by a number of routes.
  • said formulation is administrated directly in a tumor or tumors.
  • a host cell is a cell that is transfected with an expression vector containing a nucleotide or polynucleotide sequence that encodes one or more protein sequences that can be expressed in a cell.
  • a cell, including a host cell is a mammalian cell, a yeast cell, an insect cell, or a bacteria.
  • a mammalian cell used as a host cell can be a Chinese hamster ovary (“CHO”) cell, a HeLa cell, an HEK cell, including an HEK-293 cell.
  • a yeast cell used as a host cell can be S. cerevisiae or Pichia pastoris.
  • an insect cell used as a host cell can be Sf9, Sf21, Hi-5, Schneider 2 cells, Schneider 3 cells or High Five.
  • a bacterial cell used as a host cell can be E. coli, Corynebacterium or C. glutamicum.
  • an antibody or protein formulation is a lyophilized formulation. In another embodiment, an antibody or protein formulation is an aqueous formulation.
  • a fusion protein or chimeric molecule disclosed herein reduces the severity of a disease by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%.
  • a fusion protein or chimeric molecule disclosed herein reduces the severity of a disease from, e.g., about 5% to about 100%, about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 10% to about 90%, about 20% to about 90%, about 30% to about 90%, about 40% to about 90%, about 50% to about 90%, about 60% to about 90%, about 70% to about 90%, about 10% to about 80%, about 20% to about 80%, about 30% to about 80%, about 40% to about
  • a fusion protein or chimeric molecule disclosed herein may comprise a therapeutic compound in an amount sufficient to allow customary administration to an individual and with other excipients may constitute a pharmaceutical composition.
  • a therapeutic compound disclosed herein may be, e.g., at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, or at least 100 mg of a therapeutic compound.
  • a therapeutic compound disclosed herein may be, e.g., at least 5 mg, at least 10 mg, at least 20 mg, at least 25 mg, at least 50 mg, at least 75 mg, at least 100 mg, at least 200 mg, at least 300 mg, at least 400 mg, at least 500 mg, at least 600 mg, at least 700 mg, at least 800 mg, at least 900 mg, at least 1,000 mg, at least 1,100 mg, at least 1,200 mg, at least 1,300 mg, at least 1,400 mg, or at least 1,500 mg of a therapeutic compound.
  • a therapeutic compound disclosed herein may be in the range of, e.g., about 5 mg to about 100 mg, about 10 mg to about 100 mg, about 50 mg to about 150 mg, about 100 mg to about 250 mg, about 150 mg to about 350 mg, about 250 mg to about 500 mg, about 350 mg to about 600 mg, about 500 mg to about 750 mg, about 600 mg to about 900 mg, about 750 mg to about 1,000 mg, about 850 mg to about 1,200 mg, or about 1,000 mg to about 1,500 mg.
  • a therapeutic compound disclosed herein may be in the range of, e.g., about 10 mg to about 250 mg, about 10 mg to about 500 mg, about 10 mg to about 750 mg, about 10 mg to about 1,000 mg, about 10 mg to about 1,500 mg, about 50 mg to about 250 mg, about 50 mg to about 500 mg, about 50 mg to about 750 mg, about 50 mg to about 1,000 mg, about 50 mg to about 1,500 mg, about 100 mg to about 250 mg, about 100 mg to about 500 mg, about 100 mg to about 750 mg, about 100 mg to about 1,000 mg, about 100 mg to about 1,500 mg, about 200 mg to about 500 mg, about 200 mg to about 750 mg, about 200 mg to about 1,000 mg, about 200 mg to about 1,500 mg, about 5 mg to about 1,500 mg, about 5 mg to about 1,000 mg, or about 5 mg to about 250 mg.
  • a therapeutic compound disclosed herein may comprise a solvent, emulsion or other diluent in an amount sufficient to dissolve a therapeutic compound disclosed herein.
  • a therapeutic compound disclosed herein may comprise a solvent, emulsion or a diluent in an amount of, e.g., less than about 90% (v/v), less than about 80% (v/v),
  • a therapeutic compound disclosed herein may comprise a solvent, emulstion or other diluent in an amount in a range of, e.g., about 1% (v/v) to 90% (v/v), about 1% (v/v) to 70% (v/v), about 1% (v/v) to 60% (v/v), about 1% (v/v) to 50% (v/v), about 1% (v/v) to 40% (v/v), about 1% (v/v) to 30% (v/v), about 1% (v/v) to 20% (v/v), about 1% (v/v) to 10% (v/v), about 2% (v/v) to 50% (v/v), about 2% (v/v) to 40% (v/v), about 2% (v/v) to 30% (v/v), about 2% (v/v) to 20% (v/v), about 2% (v/v) to 10% (v/v), about 4% (v/v) to 50% (v/v/v), about
  • the final concentration of a therapeutic compound disclosed herein in a pharmaceutical composition disclosed herein may be of any concentration desired.
  • the final concentration of a therapeutic compound in a pharmaceutical composition may be a therapeutically effective amount.
  • the final concentration of a therapeutic compound in a pharmaceutical composition may be, e.g., at least 0.00001 mg/mL, at least 0.0001 mg/mL, at least 0.001 mg/mL, at least 0.01 mg/mL, at least 0.1 mg/mL, at least 1 mg/mL, at least 10 mg/mL, at least 25 mg/mL, at least 50 mg/mL, at least 100 mg/mL, at least 200 mg/mL, at least 500 mg/mL, at least 700 mg/mL, at least 1,000 mg/mL, or at least 1,200 mg/mL.
  • the concentration of a therapeutic compound disclosed herein in the solution may be, e.g., at most 1,000 mg/mL, at most 1,100 mg/mL, at most 1,200 mg/mL, at most 1,300 mg/mL, at most 1,400 mg/mL, at most 1,500 mg/mL, at most 2,000 mg/mL, at most 2,000 mg/mL, or at most 3,000 mg/mL.
  • the final concentration of a therapeutic compound in a pharmaceutical composition may be in a range of, e.g., about 0.00001 mg/mL to about 3,000 mg/mL, about 0.0001 mg/mL to about 3,000 mg/mL, about 0.01 mg/mL to about 3,000 mg/mL, about 0.1 mg/mL to about 3,000 mg/mL, about 1 mg/mL to about 3,000 mg/mL, about 250 mg/mL to about 3,000 mg/mL, about 500 mg/mL to about 3,000 mg/mL, about 750 mg/mL to about 3,000 mg/mL, about 1,000 mg/mL to about 3,000 mg/mL, about 100 mg/mL to about 2,000 mg/mL, about 250 mg/mL to about 2,000 mg/mL, about 500 mg/mL to about 2,000
  • aspects of the present specification disclose, in part, treating an individual suffering from a disease, including a cancer.
  • treating refers to reducing or eliminating in an individual a clinical symptom of cancer; or delaying or preventing in an individual the onset of a clinical symptom of a disease, including a cancer.
  • the term "treating" can mean reducing a symptom of a condition characterized by a cancer, including, but not limited to, tumor size, by, e.g., at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% at least 95%, or at least 100%.
  • the actual symptoms associated with cancer are well known and can be determined by a person of ordinary skill in the art by taking into account factors, including, without limitation, the location of the disease, including a cancer, the cause of the disease, including a cancer, the severity of the disease, including a cancer, and/or the tissue or organ affected by the disease, including a cancer.
  • factors including, without limitation, the location of the disease, including a cancer, the cause of the disease, including a cancer, the severity of the disease, including a cancer, and/or the tissue or organ affected by the disease, including a cancer.
  • Those of skill in the art will know the appropriate symptoms or indicators associated with a specific type of disease, including a cancer and will know how to determine if an individual is a candidate for treatment as disclosed herein.
  • a therapeutically effective amount of a therapeutic compound disclosed herein reduces a symptom associated with a disease, including a cancer by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%,
  • a therapeutically effective amount of a therapeutic compound disclosed herein reduces a symptom associated with a disease, including a cancer by, e.g., at most 10%, at most 15%, at most 20%, at most 25%, at most 30%, at most 35%, at most 40%, at most 45%, at most 50%, at most 55%, at most 60%, at most 65%, at most 70%, at most 75%, at most 80%, at most 85%, at most 90%, at most 95% or at most 100%.
  • a therapeutically effective amount of a therapeutic compound disclosed herein reduces a symptom associated with a disease, including a cancer by, e.g., about 10% to about 100%, about 10% to about 90%, about 10% to about 80%, about 10% to about 70%, about 10% to about 60%, about 10% to about 50%, about 10% to about 40%, about 20% to about 100%, about 20% to about 90%, about 20% to about 80%, about 20% to about 20%, about 20% to about 60%, about 20% to about 50%, about 20% to about 40%, about 30% to about 100%, about 30% to about 90%, about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, or about 30% to about 50%.
  • a therapeutically effective amount of a therapeutic compound disclosed herein generally is in the range of about 0.001 mg/kg/day to about 100 mg/kg/day.
  • an effective amount of a therapeutic compound disclosed herein may be, e.g., at least 0.001 mg/kg/day, at least 0.01 mg/kg/day, at least 0.1 mg/kg/day, at least 1.0 mg/kg/day, at least 5.0 mg/kg/day, at least 10 mg/kg/day, at least 15 mg/kg/day, at least 20 mg/kg/day, at least 25 mg/kg/day, at least 30 mg/kg/day, at least 35 mg/kg/day, at least 40 mg/kg/day, at least 45 mg/kg/day, or at least 50 mg/kg/day.
  • an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 0.001 mg/kg/day to about 10 mg/kg/day, about 0.001 mg/kg/day to about 15 mg/kg/day, about 0.001 mg/kg/day to about 20 mg/kg/day, about 0.001 mg/kg/day to about 25 mg/kg/day, about 0.001 mg/kg/day to about 30 mg/kg/day, about 0.001 mg/kg/day to about 35 mg/kg/day, about 0.001 mg/kg/day to about 40 mg/kg/day, about 0.001 mg/kg/day to about 45 mg/kg/day, about 0.001 mg/kg/day to about 50 mg/kg/day, about 0.001 mg/kg/day to about 75 mg/kg/day, or about 0.001 mg/kg/day to about 100 mg/kg/day.
  • an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 0.01 mg/kg/day to about 10 mg/kg/day, about 0.01 mg/kg/day to about 15 mg/kg/day, about 0.01 mg/kg/day to about 20 mg/kg/day, about 0.01 mg/kg/day to about 25 mg/kg/day, about 0.01 mg/kg/day to about 30 mg/kg/day, about 0.01 mg/kg/day to about 35 mg/kg/day, about 0.01 mg/kg/day to about 40 mg/kg/day, about 0.01 mg/kg/day to about 45 mg/kg/day, about 0.01 mg/kg/day to about 50 mg/kg/day, about 0.01
  • an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 0.1 mg/kg/day to about 10 mg/kg/day, about 0.1 mg/kg/day to about 15 mg/kg/day, about 0.1 mg/kg/day to about 20 mg/kg/day, about 0.1 mg/kg/day to about 25 mg/kg/day, about 0.1 mg/kg/day to about 30 mg/kg/day, about 0.1 mg/kg/day to about 35 mg/kg/day, about 0.1 mg/kg/day to about 40 mg/kg/day, about 0.1 mg/kg/day to about 45 mg/kg/day, about 0.1 mg/kg/day to about 50 mg/kg/day, about 0.1 mg/kg/day to about 75 mg/kg/day, or about 0.1 mg/kg/day to about 100 mg/kg/day.
  • an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 1 mg/kg/day to about 10 mg/kg/day, about 1 mg/kg/day to about 15 mg/kg/day, about 1 mg/kg/day to about 20 mg/kg/day, about 1 mg/kg/day to about 25 mg/kg/day, about 1 mg/kg/day to about 30 mg/kg/day, about 1 mg/kg/day to about 35 mg/kg/day, about 1 mg/kg/day to about 40 mg/kg/day, about 1 mg/kg/day to about 45 mg/kg/day, about 1 mg/kg/day to about 50 mg/kg/day, about 1 mg/kg/day to about 75 mg/kg/day, or about 1 mg/kg/day to about 100 mg/kg/day.
  • an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 5 mg/kg/day to about 10 mg/kg/day, about 5 mg/kg/day to about 15 mg/kg/day, about 5 mg/kg/day to about 20 mg/kg/day, about 5 mg/kg/day to about 25 mg/kg/day, about 5 mg/kg/day to about 30 mg/kg/day, about 5 mg/kg/day to about 35 mg/kg/day, about 5 mg/kg/day to about 40 mg/kg/day, about 5 mg/kg/day to about 45 mg/kg/day, about 5 mg/kg/day to about 50 mg/kg/day, about 5 mg/kg/day to about 75 mg/kg/day, or about 5 mg/kg/day to about 100 mg/kg/day.
  • a concentration of a therapeutic compound disclosed herein typically may be between about 50 mg/mL to about 1,000 mg/mL.
  • a therapeutically effective amount of a therapeutic compound disclosed herein may be from, e.g., about 50 mg/mL to about 100 mg/mL, about 50 mg/mL to about 200 mg/mL, about 50 mg/mL to about 300 mg/mL, about 50 mg/mL to about 400 mg/mL, about 50 mg/mL to about 500 mg/mL, about 50 mg/mL to about 600 mg/mL, about 50 mg/mL to about 700 mg/mL, about 50 mg/mL to about 800 mg/mL, about 50 mg/mL to about 900 mg/mL, about 50 mg/mL to about 1,000 mg/mL, about 100 mg/mL to about 200 mg/mL, about 100 mg/mL to about 300 mg/mL, about 100 mg/mL to about 400 mg/mL,
  • Dosing can be single dosage or cumulative (serial dosing), and can be readily determined by one skilled in the art.
  • treatment of a disease, including acancer may comprise a one-time administration of an effective dose of a therapeutic compound or a pharmaceutical composition disclosed herein.
  • treatment of a disease, including a cancer may comprise multiple administrations of an effective dose of a pharmaceutical composition carried out over a range of time periods, such as, e.g., once daily, twice daily, trice daily, once every few days, or once weekly.
  • the timing of administration can vary from individual to individual, depending upon such factors as the severity of an individual's symptoms.
  • an effective dose of a therapeutic compound or pharmaceutical composition disclosed herein can be administered to an individual once daily for an indefinite period of time, or until the individual no longer requires therapy.
  • a person of ordinary skill in the art will recognize that the condition of the individual can be monitored throughout the course of treatment and that the effective amount of a therapeutic compound or pharmaceutical composition disclosed herein that is administered can be adjusted accordingly.
  • a therapeutic compound disclosed herein is capable of reducing the number of cancer cells or tumor size in an individual suffering from a cancer by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% as compared to a patient not receiving the
  • a therapeutic compound is capable of reducing the number of cancer cells or tumor size in an individual suffering from a cancer by, e.g., about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 10% to about 90%, about 20% to about 90%, about 30% to about 90%, about 40% to about 90%, about 50% to about 90%, about 60% to about 90%, about 70% to about 90%, about 10% to about 80%, about 20% to about 80%, about 30% to about 80%, about 40% to about 80%, about 50% to about 80%, or about 60% to about 80%, about 10% to about 70%, about 20% to about 70%, about 30% to about 70%, about 40% to about 70%, or about 50% to about 70% as compared to a patient not receiving the same treatment.
  • a therapeutic compound and its derivatives have half-lives of 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, one month, two months, three months, four months or more.
  • the period of administration of a therapeutic compound is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
  • a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
  • a therapeutically effective amount of a therapeutic compound disclosed herein reduces or maintains a disease, including a cancer cell population and/or tumor cell size in an individual by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or at least 100%.
  • a therapeutically effective amount of a therapeutic compound disclosed herein reduces or maintains a disease or a cancer cell population and/or tumor cell size in an individual by, e.g., at most 10%, at most 15%, at most
  • a therapeutically effective amount of a therapeutic compound disclosed herein reduces or maintains a disease, including a cancer cell population and/or tumor cell size in an individual by, e.g., about 10% to about 100%, about 10% to about 90%, about 10% to about 80%, about 10% to about 70%, about 10% to about 60%, about 10% to about 50%, about 10% to about 40%, about 20% to about 100%, about 20% to about 90%, about 20% to about 80%, about 20% to about 20%, about 20% to about 60%, about 20% to about 50%, about 20% to about 40%, about 30% to about 100%, about 30% to about 90%, about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, or about 30% to about 50%.
  • a pharmaceutical composition or therapeutic compound is administered to an individual.
  • An individual is typically a human being, but can be an animal, including, but not limited to, dogs, cats, birds, cattle, horses, sheep, goats, reptiles and other animals, whether domesticated or not.
  • any individual who is a candidate for treatment is a candidate with some form of disease, including a cancer, whether the cancer is benign or malignant, a tumor, solid or otherwise, a cancer cell not located in a tumor or some other form of cancer.
  • cancer include, but are not limited to, bladder cancer, breast cancer, colon and rectal cancer, endometrial cancer, kidney cancer, renal cancer, leukemia, lung cancer, melanoma, non-Hodgkins lymphoma, pancreatic cancer, prostate cancer, stomach cancer and thyroid cancer.
  • Pre-operative evaluation typically includes routine history and physical examination in addition to thorough informed consent disclosing all relevant risks and benefits of the procedure.
  • Example 1 Production of the Chimeric Molecule Comprising VEGF Antibody and Ang-2 Binding Peptide in HEK293 Cells
  • Chimeric molecules named AMD A, B, C, D and E were expressed through transient expression by HEK-293 cells. Briefly, DNAs (SEQ ID NOs: 58, 59, 60 and 63) for the fusion proteins comprising VEGF antibody light chain with or without Ang2 binding peptides and DNAs (SEQ ID NOs: 57, 61 and 62) for the fusion proteins comprising VEGF antibody heavy chain with Ang2 binding peptides were synthesized and cloned into expression vectors. The complete expression constructs comprising the genes were confirmed by DNA sequencing. DNA constructs were transformed into E. coli DH5alfa competent cells
  • the cell culture media were harvested by clarifying centrifugation at 9000 rpm for 30-60 minutes followed by filtration through 0.22 micrometer filters.
  • the clarified supernants were loaded to a Protein A affinity column and the chimeric molecules (AMD-A, B, C, D and E) were purified.
  • the chimeric molecules were eluted using 2 M arginine solution, pH 4 from the protein A column.
  • Figure 1 shows a representative chromatograph of the Protein A column step.
  • Table 3 summarizes the results from the purification of the chimeric molecules.
  • chimeric molecules containing a total of 2 copies L1-15 peptides (AMD-B and AMD-D), both fused to the N-terminals of the heavy chain, had significantly higher expression levels comparing to the ones with a total of four copies of L1-15 peptides (AMD-A and AMD-C), wherein there is one each of L1-15 peptide fused to the N-terminals of both the light chains and the heavy chains of the antibody.
  • ASD-A and AMD-C L1-15 peptide fused to the N-terminals of both the light chains and the heavy chains of the antibody.
  • AMD-E has one Peptide 2xCon4(C) fused to each of the C-terminus of the heavy chains of Bevacizumab.
  • the purity of the products were analyzed using SDS electrophoresis and/or HPLC methods.
  • DNA for the chimeric molecule comprising the VEGF Receptor-Fc fusion protein (VEGF Trap) and the Ang-2 binding peptide (SEQ ID NO: 64, named as ASKB-E06) is synthesized and cloned into an expression vector.
  • the complete expression construct comprising the DNA gene is confirmed by DNA sequencing.
  • the expression construct is amplified by transforming into DH10B E. coli and culturing the cells overnight.
  • DNA for the expression construct was prepared and purified by endo-free plasmid kit (from QIAGEN ® ).
  • Cell lines stably expressing ASKB-E06 is obtained by transfecting the expression construct into GS -/- Chinese hamster ovarian cells (CHO) by electroporation and screening for transfected CHO cells using a selective culture medium without glutamine (EX-CELL ® CD CHO Fusion Growth Medium). In this manner 32 or more stable minipools are established and the leading mini-pool is selected based on expression level in batch and fed-batch cultures. The expression levels are detected by ELISA titer assay. Single cloning is performed by limited dilution and using clone media, two leading single clones out of more than100 positive clones are selected based on productivity and cell growth in batch and fed-batch culture.
  • the lead clones are expanded and seeded at 0.5x10 6 cells/mL, total 300 mL in 2L shake flasks, and the cells are cultured at 37 o C, 5% CO 2 , 70% HMR conditions and shaking at 120 rpm.
  • the cultures are fed by using 5% Acti CHO ® Feed A + 0.5% Feed B (from GE Health) on Day 3, 6, 7, 8 and 9.
  • the cell viability, viable cell density are monitored every other day, the cultures are harvested on Day 11-13.
  • the cell culture medium is harvested by clarifying approximately 600 mL of the cultured cell medium through centrifugation at 2000 rpm for 10 minutes followed by filtration.
  • the clarified supernant is loaded to a Protein A affinity column and the chimeric molecule is purified.
  • the protein is further purified using ion exchange chromatography, hydrophobic interaction chromatography, hydroxyapatite chromatography, and/or mixed mode chromatography.
  • the product is further concentrated and buffer exchanged using UFDF and further formulated.
  • the purity of the product is analyzed using CE-SDS and HPLC methods.
  • Molecular assays (Octet Binding Affinity, Affinity ELISA, and Blocking ELISA) were developed to assess direct binding of the chimeric molecules to ANG-1, Ang-2 and/or VEGF, and the effect of the chimeric molecules on the Ang1:Tie-2 interaction, Ang-2:Tie-2 interaction and/or VEGF:VEGF receptor interaction. These in vitro assays are described as the following: Octet Affinity
  • VEGF protein Purified recombinant human VEGF protein was ordered from Life-Technologies (Cat.# PHC9391 ). Human Ang1 or Ang2 protein were ordered from R&D System. Analysis was carried out using Octet Red96 from Pall ForteBio. Using anti-human IgG Fc sensors, a sample of chimeric molecule AMD-B, AMD-D, AMD-E or the control antibody Bevacizumab was loaded for 300 seconds at 3ug/mL in the kinetics buffer. Ligands ANG1, ANG2, or VEGF samples were associated for 300 seconds using a dilution series starting at 5 or 10 ug/mL and sequentially diluting 2-fold for 7 wells. Dissociation was run for 600 seconds.
  • Affinity ELISA Purified recombinant human VEGF protein was ordered from Life- Technologies (Cat.# PHC9391 ). VEGF is reconstituted in BSA solution at 0.1 mg/mL as recommended by the manufacturer. Aliquots the samples were made and stored at -20 o C.
  • VEGF vascular endothelial growth factor
  • PBS phosphate buffered saline
  • Tween-20 four times.
  • the wells are then blocked using about 250 microliters per well of about 5 percent BSA in PBS, and the plates were incubated at room temperature for about 2 hours. After incubation, excess blocking solution is discarded, and about 100 microliters of AMD-A, B, C, D or E was added to a well in a dilution series starting at a concentration of about 40 nanomolar and then serially diluting 4- fold in PBS containing about 1 percent BSA.
  • Figure 3 shows the ELISA results of binding of VEGF to AMD-A, B, C, D, and E.
  • the results showed that all the molecules AMD-A, B, C, D and E retained abilities to bind to VEGF.
  • the EC-50 results are summarized in Table 5.
  • the results showed that the AMD-B and AMD-D had VEGF binding affinity close to ASKB1202.
  • AMD- B and AMD-D had stronger VEGF binding affinity than AMD-A and AMD-C.
  • chimeric molecules were assessed in their abilities in blocking the binding of Ang1 and Ang2 to their receptor Tie-2.
  • 96 well microtiter plate (Nunk) was coated with 100 uL final concentration 100 ng/mL of human Tie2-Fc (R&D System, 313-T1) diluted in 0.1 M carbonate (pH9.3) at 4 o C overnight. The plate was then blocked for 2 hours with 5% BSA in PBST (0.05% Tween 20). Purified chimeric molecule, at starting concentration of 1000 ng/mL, was serially diluted with dilution factor of three in PBS with 1% BSA.
  • Human Ang1 or Ang2 protein (R&D System) was added to final concentration of 50 ng/mL and incubated at room temperature for 1 hour. The Chimeric molecule-Ang1 or Chimeric molecule-Ang2 mixture was then added into microtiter plate coated with human Tie2-Fc and incubate for another 1 hour at room temperature. 100 uL anti-Ang1 or anti-Ang2 monoclonal antibody (R&D System) was added into each well at final concentration of 1 ug/mL and incubated for 1 hour at room temperature. Horseradish-peroxidase (HRP) conjugated anti-mouse IgG secondary antibody was added at 1:5000 dilution and incubated for 1 hour at room temperature. Standard colorimetric response was developed by using TMB (Pierce). Absorbance was read at OD450 by spectrophotometer. Between each step, the plate was washed 5 time with 100 uL PBS.
  • HRP horseradish-peroxidase conjugated anti-mouse
  • Figure 5 shows the inhibition of the binding of Ang-2 to Tie-2 by chimeric molecules 712-O and 712-O2.
  • the chimeric molecule 712-O comprises two heavy chain polypeptide chains with an amino acid sequence as shown in SEQ ID NO: 29 and two light chains with an amino acid sequence as shown in SEQ ID NO: 4.
  • the chimeric molecule 712-O2 comprises
  • the Ang-2 antagonist peptide L1-15 is fused to the N-terminals of the heavy chains of a VEGF-binding antibody in the case of 712-O.
  • L1-15 is fused to the C-terminals of the heavy chains.
  • the IC-50’s for the Ang-2 blocking assay were approximately 33 pM for 712-O and approximately 78 pM for 712-O2. Since L1-15, together with other peptides including L1-7, L1-10 and L1-21, was considered an N-terminal fusion peptide and was only tested to be active when it is fused to the N-terminal of the Fc as described in WO2004/092215A2. It was surprised that the chimeric molecule 712-O2 was significantly potent with an IC-50 of approximately 78 pM.
  • Blocking ELISA Results Inhibition of Binding of Ang-1 or Ang-2 to Tie-2.
  • Example 4 Cell-based activity assay: In vitro Human Umbilical Vein Endothelial Cells
  • EBM-2 medium having VEGF-A (50 ng/ml) is added thereto, or EBM-2 medium including VEGF- A (50 ng/ml) and 712-O sample at different concentration is added thereto in each well of a 96- well plate, followed by incubation under 5% CO 2 , at 37° C. for 72 hours. Then, 10 ml of WST-1 solution was added thereto, followed by incubation at 37° C. for 4 hours. Absorbance is measured at 410 nm with a reference of 610 nm. The results are shown in Table 7, which indicated that 712-O had similar or higher potency than Lucentis®. It was more potent than ASKB1202 (a biosimilar of bevacizumab).
  • a lower chamber is filled with 600 ml of EBM- 2 medium (Lonza), EBM-2 with VEGF-A (50 ng/ml), or EBM-2 with VEGF-A (50 ng/ml) and 712- O sample at different concentration.
  • An upper chamber is provided with 100 ml of EBM-2 medium containing 1 ⁇ 10 5 HUVEC.
  • a filter is detached from the Transwell and cells are fixed with methanol for 1 minute and stained with Hematoxylin/Eosin. Cells which do not migrate but are left on an upper surface of the transwell are completely removed with a cotton swab. Five random fields among the cells migrated through the filter are arbitrarily chosen under an optical microscope ( ⁇ 100) and the number thereof is counted.
  • tube formation assay is performed. More specifically, after a 96-well plate is coated with Growth Factor Reduced Matrigel (BD Biosciences, US), 15,000 HUVEC in 100 ml of EBM-2 medium, EBM-2 medium with VEGF-A (50 ng/ml), or EBM-2 medium with VEGF-A (50 ng/ml) and an antibody sample are added to each well, followed by incubation in 37° C. cell incubator for 6 hours. Then, tube formation is observed by using an inverted microscope.
  • Growth Factor Reduced Matrigel BD Biosciences, US
  • Example 5 In vivo anti-tumor activity study: Therapeutic Efficacy Studies With Systemically
  • the chimeric molecule ASKB712-B is administered subcutaneously to A431 tumor- bearing mice at a once-per-day schedule 72 hours after tumor challenge.
  • the doses used are
  • a chimeric molecule which comprises one or two VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein:
  • said Ang-2 antagonist peptide comprises an amino acid sequence selected from SEQ ID NO: 8-14;
  • said VEGF-binding moiety is an antibody, an Fab or an scFv; and wherein said antibody, Fab or scFv comprises light chain CDRs as derived from a light chain with an amino acid sequence as shown in SEQ ID NO: 4, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6, and heavy chain CDRs as derived from a heavy chain with an amino acid sequence as shown in SEQ ID NO: 5, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6.
  • VEGF binding moiety comprises an antibody with a light chain amino acid sequence that is at least 95% identical to that of SEQ ID NO: 4, and heavy chain amino acid sequence that is at least 99% identical to that of SEQ ID NO: 7.
  • Ang-2 antagonist peptide-HC fusion polypeptide comprises an amino acid sequence that has at least 99% identity to one of SEQ ID NOS:29, 30, and SEQ ID NO:33.
  • Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NOS: 31, 32, and 34.
  • Ang-2 antagonist polypeptide is fused to the N-terminals or the C-terminals of the heavy chain of said antibody through a peptide linker; and wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NO: 37, 39, 41, 43, 45, 47, 49, 51, and 53..
  • VEGF binding moiety is an Fab with a light chain amino acid sequence of at least 95% identity to SEQ ID NO: 4, and a heavy chain amino acid sequence of at least 95% identity to SEQ ID NO: 5.
  • Ang-2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO:19 or SEQ ID NO:20.
  • VEGF binding moiety is an scFv with an amino acid sequence having at least 95% identity to SEQ ID NO: 6.
  • Ang2 antagonist peptide is fused to the C-terminal of the scFv optionally; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NO:27 and SEQ ID NO:28.
  • a chimeric molecule comprising a fusion protein that has one or more VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein said VEGF binding moiety is a VEGF trap with an amino acid sequence having at least 95% identity to SEQ ID NO: 3; wherein the chimeric molecule comprises two identical polypeptide chains, each having an amino acid sequence at least 99% identical to one of SEQ ID NOS:15-17, 23 and 24.
  • a method of making the chimeric molecule of any one of claims 1-16 comprising culturing a host cell transfected with one or more expression vectors containing a polynucleotide that encodes a chimeric molecule of one of claims 1 -16 under conditions that allow expression of the chimeric molecule, and isolating the chimeric molecule.
  • a pharmaceutical composition comprising the chimeric molecule of any one of claims 1-16 and a pharmaceutically acceptable excipient.
  • composition of claim 21, wherein the pharmaceutical composition is in the form of a lyophilized formulation or an aqueous solution.
  • a method of treating a patient with cancer, proliferative retinopathy, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR) comprising administering to a subject a pharmaceutical composition of claim 21.
  • wAMD wet age-related macular degeneration
  • RVO retinal vein occlusion
  • DME diabetic macular edema
  • DR diabetic retinopathy
  • the open-ended transitional term“comprising” (and equivalent open-ended transitional phrases thereof like including, containing and having) encompasses all the expressly recited elements, limitations, steps and/or features alone or in combination with unrecited subject matter; the named elements, limitations and/or features are essential, but other unnamed elements, limitations and/or features may be added and still form a construct within the scope of the claim.
  • the meaning of the open-ended transitional phrase“comprising” is being defined as encompassing all the specifically recited elements, limitations, steps and/or features as well as any optional, additional unspecified ones.
  • the meaning of the closed-ended transitional phrase“consisting of” is being defined as only including those elements, limitations, steps and/or features specifically recited in the claim whereas the meaning of the closed-ended transitional phrase“consisting essentially of” is being defined as only including those elements, limitations, steps and/or features specifically recited in the claim and those elements, limitations, steps and/or features that do not materially affect the basic and novel characteristic(s) of the claimed subject matter.
  • the open-ended transitional phrase“comprising” (and equivalent open-ended transitional phrases thereof) includes within its meaning, as a limiting case, claimed subject matter specified by the closed- ended transitional phrases“consisting of” or“consisting essentially of.” As such embodiments described herein or so claimed with the phrase“comprising” are expressly or inherently
  • SEQ ID NO: 1 Bevacizumab Heavy Chain:
  • YTQKSLSLSP G(K) [0144] SEQ ID NO: 4, Protein Sequence for Light Chain, Ranibizumab (VEGF Fab) DIQLTQSPSSLSASVGDRVTITCSASQDISNYLNWYQQKPGKAPKVLIYFTSSLHSGVPSRFS GSGSGTDFTLTISSLQPEDFATYYCQQYSTVPWTFGQGTKVEIKRTVAAPSVFIFPPSDE QLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
  • SEQ ID NO: 5 Protein Sequence for Heavy Chain Ranibizumab (VEGF Fab) EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGE PTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQ GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHL
  • SEQ ID NO:6 Protein Sequence for a VEGF ScFv EIVMTQSPSTLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRF SGSGAEFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGG SGGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGL EWVGFIDPDDDPYYATWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSG WGLDIWGQGTLVTVSS
  • SEQ ID NO:7 Protein Sequence for a heavy chain of a VEGF antibody
  • VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS VMHEALHNHY TQKSLSLSPG K
  • SEQ ID NO 16 Protein Sequence for AMD-I (L1-15 fused to VEGF Trap)
  • SEQ ID NO 19 Protein Sequence for AMD-K Heavy Chain (L1-15 fused to VEGF Fab)
  • SEQ ID NO 20 Protein Sequence for AMD-L Heavy Chain (L1-7 fused to VEGF Fab)
  • SEQ ID NO 21 Protein Sequence for AMD-N (L1-15 fused to VEGF ScFv)
  • SEQ ID NO 23 Protein Sequence for AMD-I-C terminal (L1-15 fused to C- terminal of VEGF Trap)
  • SEQ ID NO 24 Protein Sequence for AMD-J-C terminal (L1-7 fused to C- terminal of VEGF Trap)
  • SEQ ID NO 25 Protein Sequence for AMD-K-C terminal Heavy Chain (L1-15 fused to C-terminal VEGF Fab)
  • SEQ ID NO 26 Protein Sequence for AMD-L-C terminal Heavy Chain (L1-7 fused to VEGF Fab)
  • SEQ ID NO 27 Protein Sequence for AMD-N-C terminal (L1-15 fused to C- terminal VEGF ScFv)
  • SEQ ID NO 28 Protein Sequence for AMD-Q-C terminal (L1-7 fused to VEGF ScFv)
  • SEQ ID NO: 30 Protein Sequence for ASKB712-O3 (L1-15 fused to the N- terminal of an VEGF-binding antibody)
  • Xaa25 is L or deleted
  • Xaa26 is E is deleted
  • n 0, 1, 2, 3, 4, or 5
  • the C- terminal amino acid K may be deleted.
  • SEQ ID NO:31 Protein Sequence for ASKB712-O2 (L1-15 fused to the C- terminal of an VEGF-binding antibody)
  • SEQ ID NO: 32 Protein Sequence for ASKB712-O4 (L1-15 fused to the C- terminal of an VEGF-binding antibody)
  • SEQ ID NO: 33 Protein Sequence for ASKB712-P (L1-7 fused to the N- terminal of an VEGF-binding antibody)
  • SEQ ID NO: 34 Protein Sequence for ASKB712-P2 (L1-7 fused to the C- terminal of an VEGF-binding antibody)
  • Xaa1 is A, G, or deleted;
  • Xaa2 is Q or A or deleted;
  • Xaa26 is L or deleted;
  • Xaa27 is E is deleted;
  • SEQ ID NO 57 DNA sequence (DHAMDH02083016) for 2xCon4(C) fused to the C-terminus of the Heavy Chain of Bevacizumab, with linker peptide
  • SEQ ID NO 58 DNA sequence (DHAMDL083016), for the light chain of
  • SEQ ID NO 59 DNA sequence (LY2.55.1), for peptide L1-15 (no LE) fused to the N-terminus of the light chain of Bevacizumab
  • SEQ ID NO 60 DNA sequence (LY2.55.2), for peptide L1-15 (with LE) fused to the N-terminus of the light chain of Bevacizumab
  • SEQ ID NO 61 DNA sequence (LY2.55.3), for peptide L1-15 (no LE) fused to the N-terminus of the heavy chain of Bevacizumab
  • SEQ ID NO 62 DNA sequence (LY2.55.4), for peptide L1-15 (with LE) fused to the N-terminus of the heavy chain of Bevacizumab
  • SEQ ID NO 63 DNA sequence (LY2.55.5), for the light chain of Bevacizumab ATGGCCTGGATGATGTTGCTTCTCGGACTTCTCGCGTATGGATCAGGGGTGGACTCCG ACATTCAAATGACTCAGTCGCCATCGTCCCTCTCGGCATCCGTGGGAGACAGAGTGA CCATCACTTGTTCCGCCTCGCAAGACATCTCCAACTACCTGAACTGGTACCAGCAGA AGCCCGGGAAGGCCCCCAAAGTGCTCATCTACTTTACTTCCTCACTGCACTCCGGG GTGCCAAGCCGCTTTAGCGGCTCCGGTTCTGGAACCGATTTCACCCTGACCATTAGC TCACTCCAGCCGGAAGATTTCGCTACGTACTACTGCCAGCAGTATTCGACCGTGCCG TGGACTTTCGGACAGGGTACCAAAGTCGAGATCAAGCGGACCGTGGCCCGAG CGTGTTCATTTTCCCGCCTTCCGACGAGCAACTCAAGTCCGTCAGTCGGACAGGGTACCAAAGTCGAAAGCGGACCGTGGCC

Landscapes

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

Description

 
NOVEL ANGIOPOIETIN 2, VEGF DUAL ANTAGONISTS CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to Provisional Patent Application 62/336,552, filed May 13, 2016, Provisional Patent Application 62/459,046, filed February 14, 2017 and Provisional Patent Application 62/448,998, filed January 21, 2017, herein both incorporated by reference in their entirety. The present application also claims priority to US Patent Application US20170327569A1 filed May 11th, 2017, herein incorporated by reference in their entirety. The present application further claims priority to US patent applications 62/655,436 filed on April 10th, 2018, herein incorporated by reference in their entirety.
FIELD OF THE INVENTION
[0002] The present application relates to novel molecules comprising binding domains to both VEGF and Ang2.
INTRODUCTION
[0003] Angiogenesis is implicated in the pathogenesis of a variety of disorders including solid tumors, intraocular neovascular syndromes such as proliferative retinopathies or age- related macular degeneration (AMD), rheumatoid arthritis, and psoriasis (Folkman, J., et al., J. Biol. Chem.267 (1992) 10931-10934; Klagsbrun, M., et al, Annu. Rev. Physiol.53 (1991) 217- 239; and Garner, A., Vascular Diseases, in: Pathobiology of Ocular Disease, A Dynamic Approach, Garner, A., and Klintworth, G. K. (eds.), 2nd edition, Marcel Dekker, New York (1994), pp. 1625- 1710). In the case of solid tumors, the neovascularization allows the tumor cells to acquire a growth advantage and proliferative autonomy compared to the normal cells. Accordingly, a correlation has been observed between density of microvessels in tumor sections and patient survival in a number of cancers (see, e.g., Weidner, N., et al, N Engl J Med. 324 (1991) 1-8; Horak, E.R., et al, Lancet 340 (1992) 1120-1124; and Macchiarini, P., et al, Lancet 340 (1992) 145-146).
[0004] Human vascular endothelial growth factor (VEGF/VEGF-A) is described in, e.g., Leung, D.W., et al, Science 246 (1989) 1306-9; Keck, P.J., et al, Science 246 (1989) 1309-12 and Connolly, D.T., et al, J. Biol. Chem. 264 (1989) 20017-24. The expression of VEGF is potentiated in response to hypoxia, by activated oncogenes, and by a variety of cytokines. VEGF
is involved in the regulation of normal and abnormal angiogenesis and neovascularization associated with tumors and intraocular disorders (Ferrara, N., et al, Endocr. Rev. 18 (1997) 4- 25; Berkman, R.A., et al, J. Clin. Invest.91 (1993) 153-159; Brown, L.F., et al, Human Pathol. 26 (1995) 86- 91; Brown, L.F., et al, Cancer Res. 53 (1993) 4727-4735; Mattern, J., et al, Brit. J. Cancer.73 (1996) 931-934; and Dvorak, H.F., et al, Am. J. Pathol.146 (1995) 1029-1039).
[0005] Deregulated VEGF expression contributes to the development of solid tumors by promoting tumor angiogenesis and to the etiology of several additional diseases that are characterized by abnormal angiogenesis (Kim, K. J., et al., 1993. Nature (London) 362, 841– 844; Millauer, B., et al., 1994. Nature (London) 367, 576–579). Consequently, inhibition of VEGF signaling abrogates the development of a wide variety of tumors.
[0006] In retinopathies, in which partial or general ischemia of the retina is accompanied by overexpression of VEGF and hyperproliferation of blood vessels, blindness can result (Aiello, L. P et al., 1994. N. Engl. J. Med.331, 1480–1487; Adamis, A. P., et al., , Am. J. Ophthalmol. 118, 445–450). Inhibition of VEGF expression in such disease states can treat or prevent resulting blindness.
[0007] Human angiopoietin-2 (ANG-2 or Ang-2 or Ang2) (alternatively abbreviated with ANGPT2 or ANG2) is described in Maisonpierre, P.C., et al, Science 277 (1997) 55-60 and Cheung,A.H., et al, Genomics 48 (1998) 389-91. Ang2 plays an important role in angiogenesis and its expression levels have been correlated with cancer and eye diseases (Gerald D et al., Cancer Res.2013, 73(6):1649-57; Watanabe et al., Am J Ophthalmol.2005, 139(3):476-81).
[0008] Dual antagonist RG7716 demonstrated superior efficacy than VEGF antagonist ranibizumab in a recent clinical trial. However, the reported dosage for RG7716 at 6 mg per dose was rather high considering the volume of administration to eye is typically low, e.g. 50 micoL). This could require a concentration of 120 mg/ml, a significant challenge for formulation development. A dual antagonist with stronger binding affinities to VEGF and/or Ang2 is needed. The present invention includes bi-specific molecules with enhanced binding ability and which result in a reduction in the severity of a disease in a patient treated with a molecule disclosed herein.
SUMMARY
 
[0009] The present disclosure relates to novel bispecific chimeric molecules comprising binding domains to both VEGF and Ang-2. Further disclosed are methods of using said chimeric molecules to treat a patient of cancer, proliferative retinopathy, neovascular glaucoma, macular edema, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR).
[0010] In some aspect, said chimeric molecule comprises one or two VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein:
a) said Ang-2 antagonist peptide comprises an amino acid sequence selected from SEQ ID NO: 8-14; and
b) said VEGF-binding moiety is an antibody, an Fab or an scFv; and wherein said antibody, Fab or scFv comprises light chain CDRs as derived from a light chain with an amino acid sequence as shown in SEQ ID NO: 4, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6, and heavy chain CDRs as derived from a heavy chain with an amino acid sequence as shown in SEQ ID NO: 5, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6.
[0011] In some embodiment, said VEGF binding moiety comprises an antibody with a light chain amino acid sequence that is at least 95% identical to that of SEQ ID NO: 4, and heavy chain amino acid sequence that is at least 99% identical to that of SEQ ID NO: 7.
[0012] In some embodiment, said Ang-2 antagonist peptide is fused to one or both of the N- terminals of the heavy chains (HC) of the said antibody optionally through a peptide linker. In some embodiment, the peptide-HC fusion polypeptide comprises an amino acid sequence that is at least 99% identical as one selected from SEQ ID NOS:29, 30, and SEQ ID NO:33.
[0013] In some embodiment, said Ang-2 antagonist peptide is fused to one or both of the C- terminal of the heavy chain of the said antibody optionally through a peptide linker. In some embodiment, the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence that is at least 99% identical or 100% identical as one selected from SEQ ID NOS: 31, 32, and 34.
[0014] In some embodiment, said Ang-2 antagonist polypeptide is fused to the N-terminals or the C-terminals of the heavy chain of said antibody through a peptide linker; and wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at   least 99% identical or 100% identical as one selected from SEQ ID NO: 37, 39, 41, 43, 45, 47, 49, 51, and 53.
[0015] In some embodiment, said VEGF binding moiety is an Fab with a light chain amino acid sequence of at least 95% identity to SEQ ID NO: 4, and a heavy chain amino acid sequence of at least 95% identity to SEQ ID NO: 5.
[0016] In some embodiment, th-e Ang2 antagonist peptide is fused to the N-terminal of the heavy chain of said Fab molecule through a peptide linker. In some embodiment, the Ang2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO:19 or SEQ ID NO:20.
[0017] In some embodiment, the Ang-2 antagonist peptide is fused to the C-terminal of the heavy chain of said Fab molecule through a peptide linker. In some embodiment, the peptide- heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO: 25 or SEQ ID NO:26.
[0018] In some embodiment, said VEGF binding moiety is an scFv with an amino acid sequence that has at least 95% identity to SEQ ID NO: 6. In some embodiment, the Ang-2 antagonist peptide is fused to the N-terminal of the scFv; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NOS:21 and 22. In some embodiment the Ang-2 antagonist peptide is fused to the C-terminal of the scFv; and wherein the peptide-scFv fusion polypeptide has an amino acid sequence selected from SEQ ID NO:27 and SEQ ID NO:28.
[0019] In some aspect, said chimeric molecule comprises a fusion protein that has one or more VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein said VEGF binding moiety is a VEGF trap with an amino acid sequence having at least 95% identity to SEQ ID NO: 3; wherein said chimeric molecule comprises two identical polypeptide chains, which have an amino acid sequence at least 99% identical as one selected from SEQ ID NOS:15-17, 23 and 24.
[0020] Also disclosed is a polynucleotide or polynucleotides encoding any one of the above said chimeric molecules. In some embodiment, said polynucleotide comprises a DNA sequence as one selected from SEQ ID NO: 35, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54 and 56.
[0021] Also disclosed is an expression vector or vectors comprising the above said polynucleotide or polynucleotides.
[0022] Also disclosed is a host cell comprising the above said vector(s).
[0023] Also disclosed is a method of making any one of the above said chimeric molecules, comprising culturing the above said host cell under conditions that allow expression of the chimeric molecule, and isolating the chimeric molecule.
[0024] Also disclosed is a pharmaceutical composition comprising the chimeric molecule of any one of the above said chimeric molecule and a pharmaceutically acceptable excipient.
[0025] Further provide is a method of treating a patient with cancer, proliferative retinopathy, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR) comprising administering to a subject of above said pharmaceutical composition.
BRIEF DESCRIPTION OF THE DRAWINGS
[0026] Figure 1. Protein A Affinity Chromatography. Approximately 150 ml of the clarified HEK 293 cell culture medium of the transient expression of AMD-B was loaded to a Protein A column (1 x 17 cm (Diameter x Height) of Captiv A Protein A resin) at 3 ml/min. The protein A column was equilibrated with an equilibration buffer (25 mM Tris Buffer, 100 mM NaCl, PH approximately 7.2). The column was washed with the Equilibration buffer and eluted with 2 M ariginine solution, PH 4.
[0027] Figure 2. Kinetics of Ang-1 or Ang-2 Binding to AMD-E As Analyzed by Octet Red96.
[0028] Figure 3. Binding of AMD A-E with VEGF.
[0029] Figure 4A. Blocking of Binding of Ang-1 and Ang-2 to Tie-2 by AMD-A and AMD- B
[0030] Figure 4B. Blocking of Binding of Ang-1 and Ang-2 to Tie-2 by AMD-C and AMD- D
[0031] Figure 5. Blocking of Binding of Ang-2 to Tie-2 by ASKB712-O and ASKB712-O2
DETAILED DESCRIPTION OF THE INVENTION  
[0032] Disclosed herein are fusion proteins and chimeric molecules which comprise two components: an Ang-2 antagonist peptide operationally linked to a VEGF binding domain, which is selected from an anti-VEGF antibody, an anti-VEGF Fab, an anti-VEGF scFv, or a VEGF receptor extracellular domain-Fc fusion protein (or VEGF Trap). The Ang-2 antagonist peptide and VEGF binding domain are each defined below with reference to percent identity to a reference sequence. Further disclosed are methods of using said chimeric molecules to treat a patient of cancer, proliferative retinopathy, neovascular glaucoma, macular edema, wet age- related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR).
[0033] It is understood that aspects and variations of the invention described herein include “consisting” and/or“consisting essentially of” aspects and variations.
Definitions
[0034] As used herein and in the appended claims, the singular forms“a,”“or,” and“the” include plural referents unless the context clearly dictates otherwise.
[0035] Reference to“about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to“about X” includes description of“X.” Additionally, use of“about” preceding any series of numbers includes“about” each of the recited numbers in that series. For example, description referring to “about X, Y, or Z” is intended to describe“about X, about Y, or about Z.”
[0036] The term“antigen-binding moiety” refers to a polypeptide or a set of interacting polypeptides that specifically bind to an antigen, and includes, but is not limited to, an antibody or antibody fragment, such as a monoclonal antibody, polyclonal, a chimeric antibody, a CDR- grafted antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, a Fv, a disulfide linked Fv, a scFv, a single domain antibody (dAb), a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, a functionally active epitope-binding fragment thereof, bifunctional hybrid antibodies, a single chain antibody, and a Fc-containing polypeptide, such as an immunoadhesion. In some embodiments, the antibody may be of any heavy chain isotype (e.g., IgG, IgA, IgM, IgE, or IgD). In some embodiments, the antibody may be of any light chain isotype (e.g., kappa or gamma). The antibody may be non-human (e.g.,
 
from mouse, goat, or any other animal), fully human, humanized, or chimeric. In some embodiments, the antibody is a derivatized antibody.
[0037] The term“effective amount” used herein refers to an amount of a compound or composition sufficient to treat a specified disorder, condition, or disease, such as ameliorate, palliate, lessen, and/or delay one or more of its symptoms. In reference to a disease such as a cancer, an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation in the cancer. In some embodiments, the effective amount is an amount sufficient to delay development of a cancer. In some embodiments, the effective amount is an amount sufficient to prevent or delay recurrence. An effective amount can be administered in one or more administrations. In the case of a cancer, the effective amount of the drug or composition may: (i) reduce the number of epithelioid cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop the cancer cells infiltration into peripheral organs; (iv) inhibit (e.g., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
[0038] The term“fused” or“fusion” in reference to two or more polypeptide sequences (such as an antibody heavy chain, antibody light chain, an antibody heavy chain fragment, an antibody light chain fragment, a drug conjugation moiety, a heterologous peptide, an albumin, or an albumin fragment) refers to joining of the polypeptide sequences through a backbone peptide bond.
[0039] The term“pharmaceutically acceptable” when used to refer to a compound or composition means that the compound or composition is suitable for administration to a subject, including a human subject, to achieve the treatments described herein, without unduly deleterious side effects in light of the severity of the disease and necessity of the treatment.
[0040] The term“subject” refers to a mammal and includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate.
[0041] As used herein,“treatment” or“treating” is an approach for obtaining beneficial or desired results including clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: alleviating one or  
more symptoms resulting from a disease, diminishing the extent of a disease, stabilizing a disease (e.g., preventing or delaying the worsening of the disease), preventing or delaying the spread (e.g., metastasis) of a disease, preventing or delaying the recurrence of a disease, delaying or slowing the progression of a disease, ameliorating a disease state, providing remission (partial or total) of a disease, decreasing the dose of one or more other medications required to treat a disease, delaying the progression of a disease, increasing the quality of life, and/or prolonging survival. Also encompassed by“treatment” is a reduction of a pathological consequence of a disease (such as cancer). The methods of the invention contemplate any one or more of these aspects of treatment.
[0042] It is to be understood that one, some or all of the properties of the various embodiments described herein may be combined to form other embodiments of the present invention.
[0043] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
Ang-2 Antagonist Peptide
[0044] The fusion protein or chimeric molecule comprises an Ang-2 antagonist peptide component, which binds to Angiopoietin 2 (Ang-2) and inhibits the binding of Ang-2 to its receptor. One example of the peptide is called 2xCon4(C), as described in WO2004/092215A2 or WO03/05134A2. 2xCon4(C) has an amino acid sequence as shown in SEQ ID NO:1. Additional examples of Ang-2 binding peptides include but are not limited to: L-1-21, L1-7, L1- 10, and L1-15, as described in WO2004/092215A2. Examples of Ang-2 antagonist peptides are shown in SEQ ID NO: 8-14.
VEGF-Binding Moiety
[0045] The chimeric molecule further comprises a VEGF-binding moiety. In one embodiment, said VEGF-binding moiety is an anti-VEGF antibody, an anti-VEGF Fab, or an anti-VEGF scFv that inhibits the binding of VEGF to its receptors. One example of the VEGF antibody is bevacizumab, which has two heavy chains with amino acid sequence as shown as SEQ ID NO:1, and two light chains with amino acid sequence as shown as SEQ ID NO:2. Another example is ranibizumab, an anti-VEGF Fab. And a third example is Brolucizumab (RTH258), which is a humanized single-chain antibody fragment (scFv) against VEGF.  
[0046] In another embodiment, said VEGF binding domain is a VEGF receptor-Fc fusion protein which“traps” VEGF (herein, referred to as a“VEGF trap”) and competes with the naturally occurring VEGF cellular receptor to inhibit VEGF. One example of the VEGF- receptor Fc fusion protein is afilbercept, which has an amino acid sequence as shown in SEQ ID NO:3.
[0047] In some embodiments, the VEGF-binding moiety comprises the six complementarity determining regions (CDRs) of Brolucizumab (RTH258), Ranibizumab or Bevacizumab. A number of CDR delineations are known in the art and are encompassed herein. A person of skill in the art can readily determine a CDR for a given delineation based on the sequence of the heavy or light chain variable region. The“Kabat” Complementarity Determining Regions (CDRs) are based on sequence variability and are the most commonly used (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).“Chothia” CDRs refer to the location of the structural loops (Chothia & Lesk, Canonical structures for the hypervariable regions of immunoglobulins, J. Mol. Biol., vol. 196, pp.901-917 (1987)). The“AbM” CDRs represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular’s AbM antibody modeling software. The“Contact” CDRs are based on an analysis of the available complex crystal structures. The residues from each of these CDRs are noted below in Table 1, in reference to common antibody numbering schemes. Unless otherwise specified herein, amino acid number of antibodies refers to the Kabat numbering scheme as described in Kabat et al., supra, including when CDR delineations are made in reference to Kabat, Chothia, AbM, or Contact schemes. Using this numbering system, the actual linear amino acid sequence may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a framework region (FR) or CDR of the variable domain. For example, a heavy-chain variable domain may include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted residues (e.g. residues 82a, 82b, and 82c, etc. according to Kabat) after heavy-chain FR residue 82. The Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a“standard” Kabat numbered sequence.
Table 1: CDR Delineations According to Various Schemes
 
Figure imgf000011_0001
[0048] In some embodiments, the CDRs are“extended CDRs,” and encompass a region that begins or terminates according to a different scheme. For example, an extended CDR can be as follows: L24—L36, L26—L34, or L26—L36 (VL-CDR1); L46—L52, L46—L56, or L50—L55 (VL-CDR2); L91—L97 (VL-CDR3); H47—H55, H47—H65, H50—H55, H53—H58, or H53—H65 (VH-CDR2); and/or H93—H102 (VH-CDR3).
Ang-2 antagonist peptide - VEGF-Binding Moiety Fusion Protein
[0049] The Ang-2 peptide can be linked or fused to either the C- or N- terminus of the VEGF antibody (e.g., either the heavy or the light chains) or the VEGF receptor-Fc fusion protein. The Fc portion of the VEGF receptor-Fc fusion protein may be located at either the C- or N-terminus of the VEGF receptor protein. The Fc portion is further defined herein.
[0050] The present compositions include“Fc fragments” or“Fc regions.” The term“Fc fragment” or "immunoglobulin Fc region" as used herein, refers to a protein that contains at least the heavy-chain constant region 2 (CH2) and the heavy-chain constant region 3 (CH3) of an immunoglobulin. In one embodiment, the Fc region excludes the variable regions of the heavy and light chains, the heavy-chain constant region 1 (CH1) and the light-chain constant region 1 (CL1) of the immunoglobulin. The Fc region may further include a hinge region at the heavy- chain constant region. Also, the immunoglobulin Fc region disclosed herein may contain a part  
or all of the Fc region including the heavy-chain constant region 1 (CH1) and/or the light-chain constant region 1 (CL1), except for the variable regions of the heavy and light chains, as long as it has a physiological function substantially similar to or better than the native protein. Also, the immunoglobulin Fc region may be a fragment having a deletion in a relatively long portion of the amino acid sequence of CH2 and/or CH3. That is, the immunoglobulin Fc region disclosed herein may comprise 1) a CH1 domain, a CH2 domain, a CH3 domain and a CH4 domain, 2) a CH1 domain and a CH2 domain, 3) a CH1 domain and a CH3 domain, 4) a CH2 domain and a CH3 domain, 5) a combination of one or more domains and an immunoglobulin hinge region (or a portion of the hinge region), and 6) a dimer of each domain of the heavy-chain constant regions and the light-chain constant region.
[0051] The immunoglobulin Fc region disclosed herein includes a native amino acid sequence, or a sequence analogue thereof. An amino acid sequence analogue is a sequence that is different from the native amino acid sequence due to a deletion, an insertion, a non- conservative or conservative substitution or combinations thereof of one or more amino acid residues.
[0052] Also, other various analogues are possible, including one in which a region capable of forming a disulfide bond is deleted, or certain amino acid residues are eliminated at the N- terminal end of a native Fc form or a methionine residue is added thereto. Further, to remove effector functions, a deletion may occur in a complement-binding site, such as a C1q-binding site and an ADCC (antibody dependent cell mediated cytotoxicity) site. Techniques of preparing such sequence analogues of the immunoglobulin Fc region are disclosed in WO 1997/034631 and WO 1996/032478.
[0053] The aforementioned Fc analogues are analogues that have a biological activity identical to the Fc region disclosed herein or improved structural stability, for example, against heat, pH, or the like.
[0054] In addition, these Fc regions may be obtained from native forms isolated from humans and other animals including cows, goats, pigs, mice, rabbits, hamsters, rats and guinea pigs, or may be recombinants or analogues thereof, obtained from transformed animal cells or microorganisms. Herein, they may be obtained from a native immunoglobulin by isolating whole immunoglobulins from human or animal organisms and treating them with a proteolytic enzyme.  
Papain digests the native immunoglobulin into Fab and Fc regions, and pepsin treatment results in the production of pF'c and F(ab)2 fragments. These fragments may be subjected, for example, to size exclusion chromatography to isolate Fc or pF'c. In another embodiment, a human-derived Fc region is a recombinant immunoglobulin Fc region that is obtained from a microorganism.
[0055] In one embodiment, the Fc region, if desired, may be modified by phosphorylation, sulfation, acrylation, glycosylation, methylation, farnesylation, acetylation, amidation, and the like. In one embodiment, the immunoglobulin Fc region disclosed herein may be in the form of having native sugar chains, increased sugar chains compared to a native form or decreased sugar chains compared to the native form, or may be in a deglycosylated form. The increase, decrease or removal of the immunoglobulin Fc sugar chains may be achieved by methods common in the art, such as a chemical method, an enzymatic method and a genetic engineering method using a microorganism. The removal of sugar chains from an Fc region results in a sharp decrease in binding affinity to the C1q part of the first complement component C1 and a decrease or loss in antibody-dependent cell-mediated cytotoxicity or complement-dependent cytotoxicity, thereby not inducing unnecessary immune responses in-vivo. In this regard, an immunoglobulin Fc region in a deglycosylated or aglycosylated form may be more suitable as a drug carrier.
[0056] As used herein, the term "deglycosylation" refers to enzymatically removing sugar moieties from an Fc region, and the term "aglycosylation" means that an Fc region is produced in an unglycosylated form by a prokaryote, preferably E. coli.
[0057] In one embodiment, the immunoglobulin Fc region may be an Fc region that is derived from IgG, IgA, IgD, IgE and IgM, or that is made by combinations thereof or hybrids thereof. In an embodiment, it is derived from IgG or IgM, which are among the most abundant proteins in human blood, and further, wherein an IgG, which is known to enhance the half-lives of ligand-binding proteins is an IgG1, IgG2a, IgG2b and/or IgG3.
[0058] The terms "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. Methods for obtaining (e.g., producing, isolating, purifying,
 
synthesizing, and recombinantly manufacturing) polypeptides are well known to one of ordinary skill in the art.
Pharmaceutical Compositions
[0059] Pharmaceutical compositions of the chimeric molecules are prepared by mixing the antibody fusion molecules or the antibody fusion molecule drug conjugate having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (see Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG).
[0060] Buffers are used to control the pH in a range which optimizes the therapeutic effectiveness, especially if stability is pH dependent. Buffers are preferably present at concentrations ranging from about 50 mM to about 250 mM. Suitable buffering agents for use with the present invention include both organic and inorganic acids and salts thereof, such as citrate, phosphate, succinate, tartrate, fumarate, gluconate, oxalate, lactate, acetate. Additionally, buffers may comprise histidine and trimethylamine salts such as Tris.
[0061] Preservatives are added to retard microbial growth, and are typically present in a range from 0.2% - 1.0% (w/v). Suitable preservatives for use with the present invention include octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium halides (e.g., chloride, bromide, iodide), benzethonium chloride; thimerosal, phenol, butyl or benzyl  
alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol, 3- pentanol, and m-cresol.
[0062] Tonicity agents, sometimes known as“stabilizers” are present to adjust or maintain the tonicity of liquid in a composition. When used with large, charged biomolecules such as proteins and antibodies, they are often termed“stabilizers” because they can interact with the charged groups of the amino acid side chains, thereby lessening the potential for inter- and intra- molecular interactions. Tonicity agents can be present in any amount between 0.1% to 25% by weight, or more preferably between 1% to 5% by weight, taking into account the relative amounts of the other ingredients. Preferred tonicity agents include polyhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol and mannitol.
[0063] Non-ionic surfactants or detergents (also known as“wetting agents”) are present to help solubilize the therapeutic agent as well as to protect the therapeutic protein against agitation- induced aggregation, which also permits the formulation to be exposed to shear surface stress without causing denaturation of the active therapeutic protein or antibody. Non-ionic surfactants are present in a range of about 0.05 mg/ml to about 1.0 mg/ml, preferably about 0.07 mg/ml to about 0.2 mg/ml.
[0064] Suitable non-ionic surfactants include polysorbates (20, 40, 60, 65, 80, etc.), polyoxamers (184, 188, etc.), PLURONIC® polyols, TRITON®, polyoxyethylene sorbitan monoethers (TWEEN®-20, TWEEN®-80, etc.), lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. Anionic detergents that can be used include sodium lauryl sulfate, dioctyle sodium sulfosuccinate and dioctyl sodium sulfonate. Cationic detergents include benzalkonium chloride or benzethonium chloride.
[0065] The choice of pharmaceutical carrier, excipient or diluent may be selected with regard to the intended route of administration and standard pharmaceutical practice. Pharmaceutical compositions may comprise as - or in addition to - the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s) or solubilizing agent(s).
[0066] There may be different composition/formulation requirements dependent on the different delivery systems. By way of example, pharmaceutical compositions useful in the  
present invention may be formulated to be administered using a mini-pump or by a mucosal route, for example, as a nasal spray or aerosol for inhalation or ingestible solution, or parenterally in which the composition is formulated by an injectable form, for delivery, by, for example, an intravenous, intramuscular or subcutaneous route. Alternatively, the formulation may be designed to be administered by a number of routes. In some embodiment, said formulation is administrated directly in a tumor or tumors.
[0067] In an embodiment, a host cell is a cell that is transfected with an expression vector containing a nucleotide or polynucleotide sequence that encodes one or more protein sequences that can be expressed in a cell. In an embodiment, a cell, including a host cell is a mammalian cell, a yeast cell, an insect cell, or a bacteria. In a further embodiment, a mammalian cell used as a host cell can be a Chinese hamster ovary (“CHO”) cell, a HeLa cell, an HEK cell, including an HEK-293 cell. In another embodiment, a yeast cell used as a host cell can be S. cerevisiae or Pichia pastoris. In an embodiment, an insect cell used as a host cell can be Sf9, Sf21, Hi-5, Schneider 2 cells, Schneider 3 cells or High Five. In a further embodiment, a bacterial cell used as a host cell can be E. coli, Corynebacterium or C. glutamicum.
[0068] In some embodiments, an antibody or protein formulation is a lyophilized formulation. In another embodiment, an antibody or protein formulation is an aqueous formulation.
[0069] In other aspects of this embodiment, a fusion protein or chimeric molecule disclosed herein reduces the severity of a disease by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%. In yet other aspects of this embodiment, a fusion protein or chimeric molecule disclosed herein reduces the severity of a disease from, e.g., about 5% to about 100%, about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 10% to about 90%, about 20% to about 90%, about 30% to about 90%, about 40% to about 90%, about 50% to about 90%, about 60% to about 90%, about 70% to about 90%, about 10% to about 80%, about 20% to about 80%, about 30% to about 80%, about 40% to about
 
80%, about 50% to about 80%, or about 60% to about 80%, about 10% to about 70%, about 20% to about 70%, about 30% to about 70%, about 40% to about 70%, or about 50% to about 70%.
[0070] A fusion protein or chimeric molecule disclosed herein may comprise a therapeutic compound in an amount sufficient to allow customary administration to an individual and with other excipients may constitute a pharmaceutical composition. In aspects of this embodiment, a therapeutic compound disclosed herein may be, e.g., at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, or at least 100 mg of a therapeutic compound. In other aspects of this embodiment, a therapeutic compound disclosed herein may be, e.g., at least 5 mg, at least 10 mg, at least 20 mg, at least 25 mg, at least 50 mg, at least 75 mg, at least 100 mg, at least 200 mg, at least 300 mg, at least 400 mg, at least 500 mg, at least 600 mg, at least 700 mg, at least 800 mg, at least 900 mg, at least 1,000 mg, at least 1,100 mg, at least 1,200 mg, at least 1,300 mg, at least 1,400 mg, or at least 1,500 mg of a therapeutic compound. In yet other aspects of this embodiment, a therapeutic compound disclosed herein may be in the range of, e.g., about 5 mg to about 100 mg, about 10 mg to about 100 mg, about 50 mg to about 150 mg, about 100 mg to about 250 mg, about 150 mg to about 350 mg, about 250 mg to about 500 mg, about 350 mg to about 600 mg, about 500 mg to about 750 mg, about 600 mg to about 900 mg, about 750 mg to about 1,000 mg, about 850 mg to about 1,200 mg, or about 1,000 mg to about 1,500 mg. In still other aspects of this embodiment, a therapeutic compound disclosed herein may be in the range of, e.g., about 10 mg to about 250 mg, about 10 mg to about 500 mg, about 10 mg to about 750 mg, about 10 mg to about 1,000 mg, about 10 mg to about 1,500 mg, about 50 mg to about 250 mg, about 50 mg to about 500 mg, about 50 mg to about 750 mg, about 50 mg to about 1,000 mg, about 50 mg to about 1,500 mg, about 100 mg to about 250 mg, about 100 mg to about 500 mg, about 100 mg to about 750 mg, about 100 mg to about 1,000 mg, about 100 mg to about 1,500 mg, about 200 mg to about 500 mg, about 200 mg to about 750 mg, about 200 mg to about 1,000 mg, about 200 mg to about 1,500 mg, about 5 mg to about 1,500 mg, about 5 mg to about 1,000 mg, or about 5 mg to about 250 mg.
[0071] A therapeutic compound disclosed herein may comprise a solvent, emulsion or other diluent in an amount sufficient to dissolve a therapeutic compound disclosed herein. In other aspects of this embodiment, a therapeutic compound disclosed herein may comprise a solvent, emulsion or a diluent in an amount of, e.g., less than about 90% (v/v), less than about 80% (v/v),
 
less than about 70% (v/v), less than about 65% (v/v), less than about 60% (v/v), less than about 55% (v/v), less than about 50% (v/v), less than about 45% (v/v), less than about 40% (v/v), less than about 35% (v/v), less than about 30% (v/v), less than about 25% (v/v), less than about 20% (v/v), less than about 15% (v/v), less than about 10% (v/v), less than about 5% (v/v), or less than about 1% (v/v). In other aspects of this embodiment, a therapeutic compound disclosed herein may comprise a solvent, emulstion or other diluent in an amount in a range of, e.g., about 1% (v/v) to 90% (v/v), about 1% (v/v) to 70% (v/v), about 1% (v/v) to 60% (v/v), about 1% (v/v) to 50% (v/v), about 1% (v/v) to 40% (v/v), about 1% (v/v) to 30% (v/v), about 1% (v/v) to 20% (v/v), about 1% (v/v) to 10% (v/v), about 2% (v/v) to 50% (v/v), about 2% (v/v) to 40% (v/v), about 2% (v/v) to 30% (v/v), about 2% (v/v) to 20% (v/v), about 2% (v/v) to 10% (v/v), about 4% (v/v) to 50% (v/v), about 4% (v/v) to 40% (v/v), about 4% (v/v) to 30% (v/v), about 4% (v/v) to 20% (v/v), about 4% (v/v) to 10% (v/v), about 6% (v/v) to 50% (v/v), about 6% (v/v) to 40% (v/v), about 6% (v/v) to 30% (v/v), about 6% (v/v) to 20% (v/v), about 6% (v/v) to 10% (v/v), about 8% (v/v) to 50% (v/v), about 8% (v/v) to 40% (v/v), about 8% (v/v) to 30% (v/v), about 8% (v/v) to 20% (v/v), about 8% (v/v) to 15% (v/v), or about 8% (v/v) to 12% (v/v).
[0072] The final concentration of a therapeutic compound disclosed herein in a pharmaceutical composition disclosed herein may be of any concentration desired. In an aspect of this embodiment, the final concentration of a therapeutic compound in a pharmaceutical composition may be a therapeutically effective amount. In other aspects of this embodiment, the final concentration of a therapeutic compound in a pharmaceutical composition may be, e.g., at least 0.00001 mg/mL, at least 0.0001 mg/mL, at least 0.001 mg/mL, at least 0.01 mg/mL, at least 0.1 mg/mL, at least 1 mg/mL, at least 10 mg/mL, at least 25 mg/mL, at least 50 mg/mL, at least 100 mg/mL, at least 200 mg/mL, at least 500 mg/mL, at least 700 mg/mL, at least 1,000 mg/mL, or at least 1,200 mg/mL. In other aspects of this embodiment, the concentration of a therapeutic compound disclosed herein in the solution may be, e.g., at most 1,000 mg/mL, at most 1,100 mg/mL, at most 1,200 mg/mL, at most 1,300 mg/mL, at most 1,400 mg/mL, at most 1,500 mg/mL, at most 2,000 mg/mL, at most 2,000 mg/mL, or at most 3,000 mg/mL. In other aspects of this embodiment, the final concentration of a therapeutic compound in a pharmaceutical composition may be in a range of, e.g., about 0.00001 mg/mL to about 3,000 mg/mL, about 0.0001 mg/mL to about 3,000 mg/mL, about 0.01 mg/mL to about 3,000 mg/mL, about 0.1 mg/mL to about 3,000 mg/mL, about 1 mg/mL to about 3,000 mg/mL, about 250 mg/mL to about 3,000 mg/mL, about 500 mg/mL to about 3,000 mg/mL, about 750 mg/mL to about 3,000 mg/mL, about 1,000 mg/mL to about 3,000 mg/mL, about 100 mg/mL to about 2,000 mg/mL, about 250 mg/mL to about 2,000 mg/mL, about 500 mg/mL to about 2,000  
mg/mL, about 750 mg/mL to about 2,000 mg/mL, about 1,000 mg/mL to about 2,000 mg/mL, about 100 mg/mL to about 1,500 mg/mL, about 250 mg/mL to about 1,500 mg/mL, about 500 mg/mL to about 1,500 mg/mL, about 750 mg/mL to about 1,500 mg/mL, about 1,000 mg/mL to about 1,500 mg/mL, about 100 mg/mL to about 1,200 mg/mL, about 250 mg/mL to about 1,200 mg/mL, about 500 mg/mL to about 1,200 mg/mL, about 750 mg/mL to about 1,200 mg/mL, about 1,000 mg/mL to about 1,200 mg/mL, about 100 mg/mL to about 1,000 mg/mL, about 250 mg/mL to about 1,000 mg/mL, about 500 mg/mL to about 1,000 mg/mL, about 750 mg/mL to about 1,000 mg/mL, about 100 mg/mL to about 750 mg/mL, about 250 mg/mL to about 750 mg/mL, about 500 mg/mL to about 750 mg/mL, about 100 mg/mL to about 500 mg/mL, about 250 mg/mL to about 500 mg/mL, about 0.00001 mg/mL to about 0.0001 mg/mL, about 0.00001 mg/mL to about 0.001 mg/mL, about 0.00001 mg/mL to about 0.01 mg/mL, about 0.00001 mg/mL to about 0.1 mg/mL, about 0.00001 mg/mL to about 1 mg/mL, about 0.001 mg/mL to about 0.01 mg/mL, about 0.001 mg/mL to about 0.1 mg/mL, about 0.001 mg/mL to about 1 mg/mL, about 0.001 mg/mL to about 10 mg/mL, or about 0.001 mg/mL to about 100 mg/mL.
[0073] Aspects of the present specification disclose, in part, treating an individual suffering from a disease, including a cancer. As used herein, the term "treating," refers to reducing or eliminating in an individual a clinical symptom of cancer; or delaying or preventing in an individual the onset of a clinical symptom of a disease, including a cancer. For example, the term "treating" can mean reducing a symptom of a condition characterized by a cancer, including, but not limited to, tumor size, by, e.g., at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% at least 95%, or at least 100%. The actual symptoms associated with cancer are well known and can be determined by a person of ordinary skill in the art by taking into account factors, including, without limitation, the location of the disease, including a cancer, the cause of the disease, including a cancer, the severity of the disease, including a cancer, and/or the tissue or organ affected by the disease, including a cancer. Those of skill in the art will know the appropriate symptoms or indicators associated with a specific type of disease, including a cancer and will know how to determine if an individual is a candidate for treatment as disclosed herein.
[0074] In aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein reduces a symptom associated with a disease, including a cancer by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%,
 
at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or at least 100%. In other aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein reduces a symptom associated with a disease, including a cancer by, e.g., at most 10%, at most 15%, at most 20%, at most 25%, at most 30%, at most 35%, at most 40%, at most 45%, at most 50%, at most 55%, at most 60%, at most 65%, at most 70%, at most 75%, at most 80%, at most 85%, at most 90%, at most 95% or at most 100%. In yet other aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein reduces a symptom associated with a disease, including a cancer by, e.g., about 10% to about 100%, about 10% to about 90%, about 10% to about 80%, about 10% to about 70%, about 10% to about 60%, about 10% to about 50%, about 10% to about 40%, about 20% to about 100%, about 20% to about 90%, about 20% to about 80%, about 20% to about 20%, about 20% to about 60%, about 20% to about 50%, about 20% to about 40%, about 30% to about 100%, about 30% to about 90%, about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, or about 30% to about 50%.
[0075] In yet other aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein generally is in the range of about 0.001 mg/kg/day to about 100 mg/kg/day. In aspects of this embodiment, an effective amount of a therapeutic compound disclosed herein may be, e.g., at least 0.001 mg/kg/day, at least 0.01 mg/kg/day, at least 0.1 mg/kg/day, at least 1.0 mg/kg/day, at least 5.0 mg/kg/day, at least 10 mg/kg/day, at least 15 mg/kg/day, at least 20 mg/kg/day, at least 25 mg/kg/day, at least 30 mg/kg/day, at least 35 mg/kg/day, at least 40 mg/kg/day, at least 45 mg/kg/day, or at least 50 mg/kg/day. In other aspects of this embodiment, an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 0.001 mg/kg/day to about 10 mg/kg/day, about 0.001 mg/kg/day to about 15 mg/kg/day, about 0.001 mg/kg/day to about 20 mg/kg/day, about 0.001 mg/kg/day to about 25 mg/kg/day, about 0.001 mg/kg/day to about 30 mg/kg/day, about 0.001 mg/kg/day to about 35 mg/kg/day, about 0.001 mg/kg/day to about 40 mg/kg/day, about 0.001 mg/kg/day to about 45 mg/kg/day, about 0.001 mg/kg/day to about 50 mg/kg/day, about 0.001 mg/kg/day to about 75 mg/kg/day, or about 0.001 mg/kg/day to about 100 mg/kg/day. In yet other aspects of this embodiment, an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 0.01 mg/kg/day to about 10 mg/kg/day, about 0.01 mg/kg/day to about 15 mg/kg/day, about 0.01 mg/kg/day to about 20 mg/kg/day, about 0.01 mg/kg/day to about 25 mg/kg/day, about 0.01 mg/kg/day to about 30 mg/kg/day, about 0.01 mg/kg/day to about 35 mg/kg/day, about 0.01 mg/kg/day to about 40 mg/kg/day, about 0.01 mg/kg/day to about 45 mg/kg/day, about 0.01 mg/kg/day to about 50 mg/kg/day, about 0.01  
mg/kg/day to about 75 mg/kg/day, or about 0.01 mg/kg/day to about 100 mg/kg/day. In still other aspects of this embodiment, an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 0.1 mg/kg/day to about 10 mg/kg/day, about 0.1 mg/kg/day to about 15 mg/kg/day, about 0.1 mg/kg/day to about 20 mg/kg/day, about 0.1 mg/kg/day to about 25 mg/kg/day, about 0.1 mg/kg/day to about 30 mg/kg/day, about 0.1 mg/kg/day to about 35 mg/kg/day, about 0.1 mg/kg/day to about 40 mg/kg/day, about 0.1 mg/kg/day to about 45 mg/kg/day, about 0.1 mg/kg/day to about 50 mg/kg/day, about 0.1 mg/kg/day to about 75 mg/kg/day, or about 0.1 mg/kg/day to about 100 mg/kg/day.
[0076] In other aspects of this embodiment, an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 1 mg/kg/day to about 10 mg/kg/day, about 1 mg/kg/day to about 15 mg/kg/day, about 1 mg/kg/day to about 20 mg/kg/day, about 1 mg/kg/day to about 25 mg/kg/day, about 1 mg/kg/day to about 30 mg/kg/day, about 1 mg/kg/day to about 35 mg/kg/day, about 1 mg/kg/day to about 40 mg/kg/day, about 1 mg/kg/day to about 45 mg/kg/day, about 1 mg/kg/day to about 50 mg/kg/day, about 1 mg/kg/day to about 75 mg/kg/day, or about 1 mg/kg/day to about 100 mg/kg/day. In yet other aspects of this embodiment, an effective amount of a therapeutic compound disclosed herein may be in the range of, e.g., about 5 mg/kg/day to about 10 mg/kg/day, about 5 mg/kg/day to about 15 mg/kg/day, about 5 mg/kg/day to about 20 mg/kg/day, about 5 mg/kg/day to about 25 mg/kg/day, about 5 mg/kg/day to about 30 mg/kg/day, about 5 mg/kg/day to about 35 mg/kg/day, about 5 mg/kg/day to about 40 mg/kg/day, about 5 mg/kg/day to about 45 mg/kg/day, about 5 mg/kg/day to about 50 mg/kg/day, about 5 mg/kg/day to about 75 mg/kg/day, or about 5 mg/kg/day to about 100 mg/kg/day.
[0077] In liquid and semi-solid formulations, a concentration of a therapeutic compound disclosed herein typically may be between about 50 mg/mL to about 1,000 mg/mL. In aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein may be from, e.g., about 50 mg/mL to about 100 mg/mL, about 50 mg/mL to about 200 mg/mL, about 50 mg/mL to about 300 mg/mL, about 50 mg/mL to about 400 mg/mL, about 50 mg/mL to about 500 mg/mL, about 50 mg/mL to about 600 mg/mL, about 50 mg/mL to about 700 mg/mL, about 50 mg/mL to about 800 mg/mL, about 50 mg/mL to about 900 mg/mL, about 50 mg/mL to about 1,000 mg/mL, about 100 mg/mL to about 200 mg/mL, about 100 mg/mL to about 300 mg/mL, about 100 mg/mL to about 400 mg/mL, about 100 mg/mL to about 500 mg/mL, about 100 mg/mL to about 600 mg/mL, about 100 mg/mL to about 700 mg/mL, about 100 mg/mL to about 800 mg/mL, about 100 mg/mL to about 900 mg/mL, about 100 mg/mL to
 
about 1,000 mg/mL, about 200 mg/mL to about 300 mg/mL, about 200 mg/mL to about 400 mg/mL, about 200 mg/mL to about 500 mg/mL, about 200 mg/mL to about 600 mg/mL, about 200 mg/mL to about 700 mg/mL, about 200 mg/mL to about 800 mg/mL, about 200 mg/mL to about 900 mg/mL, about 200 mg/mL to about 1,000 mg/mL, about 300 mg/mL to about 400 mg/mL, about 300 mg/mL to about 500 mg/mL, about 300 mg/mL to about 600 mg/mL, about 300 mg/mL to about 700 mg/mL, about 300 mg/mL to about 800 mg/mL, about 300 mg/mL to about 900 mg/mL, about 300 mg/mL to about 1,000 mg/mL, about 400 mg/mL to about 500 mg/mL, about 400 mg/mL to about 600 mg/mL, about 400 mg/mL to about 700 mg/mL, about 400 mg/mL to about 800 mg/mL, about 400 mg/mL to about 900 mg/mL, about 400 mg/mL to about 1,000 mg/mL, about 500 mg/mL to about 600 mg/mL, about 500 mg/mL to about 700 mg/mL, about 500 mg/mL to about 800 mg/mL, about 500 mg/mL to about 900 mg/mL, about 500 mg/mL to about 1,000 mg/mL, about 600 mg/mL to about 700 mg/mL, about 600 mg/mL to about 800 mg/mL, about 600 mg/mL to about 900 mg/mL, or about 600 mg/mL to about 1,000 mg/mL.
[0078] Dosing can be single dosage or cumulative (serial dosing), and can be readily determined by one skilled in the art. For instance, treatment of a disease, including acancer may comprise a one-time administration of an effective dose of a therapeutic compound or a pharmaceutical composition disclosed herein. Alternatively, treatment of a disease, including a cancer may comprise multiple administrations of an effective dose of a pharmaceutical composition carried out over a range of time periods, such as, e.g., once daily, twice daily, trice daily, once every few days, or once weekly. The timing of administration can vary from individual to individual, depending upon such factors as the severity of an individual's symptoms. For example, an effective dose of a therapeutic compound or pharmaceutical composition disclosed herein can be administered to an individual once daily for an indefinite period of time, or until the individual no longer requires therapy. A person of ordinary skill in the art will recognize that the condition of the individual can be monitored throughout the course of treatment and that the effective amount of a therapeutic compound or pharmaceutical composition disclosed herein that is administered can be adjusted accordingly.
[0079] In one embodiment, a therapeutic compound disclosed herein is capable of reducing the number of cancer cells or tumor size in an individual suffering from a cancer by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% as compared to a patient not receiving the
 
same treatment. In other aspects of this embodiment, a therapeutic compound is capable of reducing the number of cancer cells or tumor size in an individual suffering from a cancer by, e.g., about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 10% to about 90%, about 20% to about 90%, about 30% to about 90%, about 40% to about 90%, about 50% to about 90%, about 60% to about 90%, about 70% to about 90%, about 10% to about 80%, about 20% to about 80%, about 30% to about 80%, about 40% to about 80%, about 50% to about 80%, or about 60% to about 80%, about 10% to about 70%, about 20% to about 70%, about 30% to about 70%, about 40% to about 70%, or about 50% to about 70% as compared to a patient not receiving the same treatment.
[0080] In a further embodiment, a therapeutic compound and its derivatives have half-lives of 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, one month, two months, three months, four months or more.
[0081] In an embodiment, the period of administration of a therapeutic compound is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
[0082] In aspects, of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein reduces or maintains a disease, including a cancer cell population and/or tumor cell size in an individual by, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or at least 100%. In other aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein reduces or maintains a disease or a cancer cell population and/or tumor cell size in an individual by, e.g., at most 10%, at most 15%, at most  
20%, at most 25%, at most 30%, at most 35%, at most 40%, at most 45%, at most 50%, at most 55%, at most 60%, at most 65%, at most 70%, at most 75%, at most 80%, at most 85%, at most 90%, at most 95% or at most 100%. In yet other aspects of this embodiment, a therapeutically effective amount of a therapeutic compound disclosed herein reduces or maintains a disease, including a cancer cell population and/or tumor cell size in an individual by, e.g., about 10% to about 100%, about 10% to about 90%, about 10% to about 80%, about 10% to about 70%, about 10% to about 60%, about 10% to about 50%, about 10% to about 40%, about 20% to about 100%, about 20% to about 90%, about 20% to about 80%, about 20% to about 20%, about 20% to about 60%, about 20% to about 50%, about 20% to about 40%, about 30% to about 100%, about 30% to about 90%, about 30% to about 80%, about 30% to about 70%, about 30% to about 60%, or about 30% to about 50%.
[0083] A pharmaceutical composition or therapeutic compound is administered to an individual. An individual is typically a human being, but can be an animal, including, but not limited to, dogs, cats, birds, cattle, horses, sheep, goats, reptiles and other animals, whether domesticated or not. Typically, any individual who is a candidate for treatment is a candidate with some form of disease, including a cancer, whether the cancer is benign or malignant, a tumor, solid or otherwise, a cancer cell not located in a tumor or some other form of cancer. Among the most common types of cancer include, but are not limited to, bladder cancer, breast cancer, colon and rectal cancer, endometrial cancer, kidney cancer, renal cancer, leukemia, lung cancer, melanoma, non-Hodgkins lymphoma, pancreatic cancer, prostate cancer, stomach cancer and thyroid cancer. Pre-operative evaluation typically includes routine history and physical examination in addition to thorough informed consent disclosing all relevant risks and benefits of the procedure.
EXAMPLES
[0084] Example 1– Production of the Chimeric Molecule Comprising VEGF Antibody and Ang-2 Binding Peptide in HEK293 Cells
[0085] Chimeric molecules named AMD A, B, C, D and E (see Table 2) were expressed through transient expression by HEK-293 cells. Briefly, DNAs (SEQ ID NOs: 58, 59, 60 and 63) for the fusion proteins comprising VEGF antibody light chain with or without Ang2 binding peptides and DNAs (SEQ ID NOs: 57, 61 and 62) for the fusion proteins comprising VEGF antibody heavy chain with Ang2 binding peptides were synthesized and cloned into expression vectors. The complete expression constructs comprising the genes were confirmed by DNA sequencing. DNA constructs were transformed into E. coli DH5alfa competent cells  
(Invitrogen). Single clone was selected and cultured in LB broth with antibiotics (kanamycin, 25 ug/mL). DNA plasmids were extracted with Qiagen Plasmid Maxi Kit (Qiagen) following manufacture’s protocol. Plasmid concentration was measured by NanoDrop (Thermo Fisher). The expression plasmid constructs containing the DNA sequences encoding the genes of interest, were introduced into HEK-293 cells transiently by using polyethylenimine (PEI). The transfected cells were treated by alproic acid (VPA) 24 hours post transfection to enhance protein expression.
Table 2. AMD Molecules
Figure imgf000025_0001
 
Figure imgf000026_0001
when the peptides were screened and in the subsequent peptibody (Peptide-Fc fusion) molecules.
[0086] After approximately 6 days of culturing, the cell culture media were harvested by clarifying centrifugation at 9000 rpm for 30-60 minutes followed by filtration through 0.22 micrometer filters. The clarified supernants were loaded to a Protein A affinity column and the chimeric molecules (AMD-A, B, C, D and E) were purified. The chimeric molecules were eluted using 2 M arginine solution, pH 4 from the protein A column. Figure 1 shows a representative chromatograph of the Protein A column step. Table 3 summarizes the results from the purification of the chimeric molecules. As shown in Table 3, chimeric molecules containing a total of 2 copies L1-15 peptides (AMD-B and AMD-D), both fused to the N-terminals of the heavy chain, had significantly higher expression levels comparing to the ones with a total of four copies of L1-15 peptides (AMD-A and AMD-C), wherein there is one each of L1-15 peptide fused to the N-terminals of both the light chains and the heavy chains of the antibody. With or without the flanking sequence LE as part of the L1-15 peptide did not appear to affect the expression of the chimeric molecules.
[0087] The expression level of AMD-E was comparable to that of AMD-B and AMD-D (Table 3). AMD-E has one Peptide 2xCon4(C) fused to each of the C-terminus of the heavy chains of Bevacizumab. The purity of the products were analyzed using SDS electrophoresis and/or HPLC methods.
Table 3. Summary of Protein A Affinity Chromatography Purification
Figure imgf000026_0002
  Example 2– Production of the Chimeric Molecule Comprising VEGF Trap and Ang2 Binding
Peptide in CHO Cells
[0088] DNA for the chimeric molecule comprising the VEGF Receptor-Fc fusion protein (VEGF Trap) and the Ang-2 binding peptide (SEQ ID NO: 64, named as ASKB-E06) is synthesized and cloned into an expression vector. The complete expression construct comprising the DNA gene is confirmed by DNA sequencing. The expression construct is amplified by transforming into DH10B E. coli and culturing the cells overnight. DNA for the expression construct was prepared and purified by endo-free plasmid kit (from QIAGEN®).
[0089] Cell lines stably expressing ASKB-E06 is obtained by transfecting the expression construct into GS-/- Chinese hamster ovarian cells (CHO) by electroporation and screening for transfected CHO cells using a selective culture medium without glutamine (EX-CELL® CD CHO Fusion Growth Medium). In this manner 32 or more stable minipools are established and the leading mini-pool is selected based on expression level in batch and fed-batch cultures. The expression levels are detected by ELISA titer assay. Single cloning is performed by limited dilution and using clone media, two leading single clones out of more than100 positive clones are selected based on productivity and cell growth in batch and fed-batch culture. The lead clones are expanded and seeded at 0.5x106 cells/mL, total 300 mL in 2L shake flasks, and the cells are cultured at 37oC, 5% CO2, 70% HMR conditions and shaking at 120 rpm. The cultures are fed by using 5% Acti CHO® Feed A + 0.5% Feed B (from GE Health) on Day 3, 6, 7, 8 and 9. The cell viability, viable cell density are monitored every other day, the cultures are harvested on Day 11-13.
[0090] The cell culture medium is harvested by clarifying approximately 600 mL of the cultured cell medium through centrifugation at 2000 rpm for 10 minutes followed by filtration. The clarified supernant is loaded to a Protein A affinity column and the chimeric molecule is purified. The protein is further purified using ion exchange chromatography, hydrophobic interaction chromatography, hydroxyapatite chromatography, and/or mixed mode chromatography. The product is further concentrated and buffer exchanged using UFDF and further formulated. The purity of the product is analyzed using CE-SDS and HPLC methods.
Example 3– Molecular Assays to Evaluate Dual Antagonist Activities of the Chimeric
Molecules
[0091] Molecular assays (Octet Binding Affinity, Affinity ELISA, and Blocking ELISA) were developed to assess direct binding of the chimeric molecules to ANG-1, Ang-2 and/or VEGF, and the effect of the chimeric molecules on the Ang1:Tie-2 interaction, Ang-2:Tie-2 interaction and/or VEGF:VEGF receptor interaction. These in vitro assays are described as the following: Octet Affinity
[0092] Purified recombinant human VEGF protein was ordered from Life-Technologies (Cat.# PHC9391 ). Human Ang1 or Ang2 protein were ordered from R&D System. Analysis was carried out using Octet Red96 from Pall ForteBio. Using anti-human IgG Fc sensors, a sample of chimeric molecule AMD-B, AMD-D, AMD-E or the control antibody Bevacizumab was loaded for 300 seconds at 3ug/mL in the kinetics buffer. Ligands ANG1, ANG2, or VEGF samples were associated for 300 seconds using a dilution series starting at 5 or 10 ug/mL and sequentially diluting 2-fold for 7 wells. Dissociation was run for 600 seconds. Data was analyzed using a 1:1 model with global fit. A representative binding kinetics graph is shown in Figure 2. The binding affinity results are summarized in Tables 4A, 4B and 4C. The results showed that the chimeric molecules AMD-B, AMD-D, and AMD-E were able to bind to Ang1, Ang2, and VEGF. It was also noticed that the chimeric molecule AMD-B with four L1-15 peptides fused to the N-terminals of the antibody had reduced affinity to VEGF when comparing to the control antibody Bevacizumab. AMD-D and AMD-E showed comparable affinity to VEGF comparing to the control antibody ASKB1202, a biosimilar m An internal control, ASKB 1202, a biosimilar to Bevacizumab developed in-house.
Table 4A. Summary of the Octet Affinity analysis results– Binding of ANG-1.
Figure imgf000028_0001
Table 4B. Summary of the Octet Affinity analysis results– Binding of ANG-2.
Figure imgf000028_0002
Table 4C. Summary of the Octet Affinity analysis results– Binding of VEGF.
 
Figure imgf000029_0002
[0093] Affinity ELISA: Purified recombinant human VEGF protein was ordered from Life- Technologies (Cat.# PHC9391 ). VEGF is reconstituted in BSA solution at 0.1 mg/mL as recommended by the manufacturer. Aliquots the samples were made and stored at -20 oC.
[0094] Using microtiter plates, approximately 100 microliters per well of VEGF is added to each well and the plates were incubated about 2 hours, after which the plates are washed with phosphate buffered saline (PBS) containing about 0.1 percent Tween-20 four times. The wells are then blocked using about 250 microliters per well of about 5 percent BSA in PBS, and the plates were incubated at room temperature for about 2 hours. After incubation, excess blocking solution is discarded, and about 100 microliters of AMD-A, B, C, D or E was added to a well in a dilution series starting at a concentration of about 40 nanomolar and then serially diluting 4- fold in PBS containing about 1 percent BSA. The plates were then incubated overnight at room temperature. After incubation, plates were washed with PBS containing about 0.1 percent Tween-20. Washing was repeated four additional times, after which about 100 microliters per well of goat anti-human IgG(Fc)-HRP (Pierce Chemical Co., catalog # 31416) previously diluted 1:5000 in PBS containing 1 percent BSA was added. Plates were incubated approximately 1 hour at room temperature. Plates were then washed five times in PBS containing about 0.1 percent Tween-20, after which about 100 microliters per well of TMB (3,3',5,5'- Tetramethylbenzidine Liquid Substrate System; Sigma Chemical Company, St. Louis, MO, catalog number T8665) substrate was added and plates are incubated about 5-15 minutes until blue color developed. Absorbance was then read in a spectrophotometer at about 450 nm.
[0095] Figure 3 shows the ELISA results of binding of VEGF to AMD-A, B, C, D, and E. An internal control, ASKB 1202, a biosimilar to Bevacizumab currently in development, was used as a positive control. The results showed that all the molecules AMD-A, B, C, D and E retained abilities to bind to VEGF. The EC-50 results are summarized in Table 5. The results showed that the AMD-B and AMD-D had VEGF binding affinity close to ASKB1202. In addition, AMD- B and AMD-D had stronger VEGF binding affinity than AMD-A and AMD-C.
Table 5. Affinity ELISA Results: Binding of VEGF to AMD-A, B, C, D and E.
Figure imgf000029_0001
 
Figure imgf000030_0001
Blocking ELISA:
[0096] The chimeric molecules were assessed in their abilities in blocking the binding of Ang1 and Ang2 to their receptor Tie-2. 96 well microtiter plate (Nunk) was coated with 100 uL final concentration 100 ng/mL of human Tie2-Fc (R&D System, 313-T1) diluted in 0.1 M carbonate (pH9.3) at 4 oC overnight. The plate was then blocked for 2 hours with 5% BSA in PBST (0.05% Tween 20). Purified chimeric molecule, at starting concentration of 1000 ng/mL, was serially diluted with dilution factor of three in PBS with 1% BSA. Human Ang1 or Ang2 protein (R&D System) was added to final concentration of 50 ng/mL and incubated at room temperature for 1 hour. The Chimeric molecule-Ang1 or Chimeric molecule-Ang2 mixture was then added into microtiter plate coated with human Tie2-Fc and incubate for another 1 hour at room temperature. 100 uL anti-Ang1 or anti-Ang2 monoclonal antibody (R&D System) was added into each well at final concentration of 1 ug/mL and incubated for 1 hour at room temperature. Horseradish-peroxidase (HRP) conjugated anti-mouse IgG secondary antibody was added at 1:5000 dilution and incubated for 1 hour at room temperature. Standard colorimetric response was developed by using TMB (Pierce). Absorbance was read at OD450 by spectrophotometer. Between each step, the plate was washed 5 time with 100 uL PBS.
[0097] The dose dependent inhibition or lack of inhibition of the binding of Ang1 and Ang-2 to receptor Tie-2 are shown in Figure 4. The IC-50 results are summarized in Table 6. The results showed that the chimeric molecules AMD-A, B, C, and D selectively inhibited the binding of Ang2 to Tie-2, with IC-50 in the range of 5-15 ng/ml; while their abilities in inhibiting the binding of Ang-1 to Tie-2 were very weak, if any, despite the fact that they all were able to bind to Ang1. The results also showed that AMD-A and AMD-C, both comprising 4 copies of the peptide L1-15 had lower IC-50 than AMD-B and AMD-D. AMD-E was able to inhibit the association of both Ang-1 and Ang-2 to their receptor Tie-2.
[0098] Figure 5 shows the inhibition of the binding of Ang-2 to Tie-2 by chimeric molecules 712-O and 712-O2. The chimeric molecule 712-O comprises two heavy chain polypeptide chains with an amino acid sequence as shown in SEQ ID NO: 29 and two light chains with an amino acid sequence as shown in SEQ ID NO: 4. The chimeric molecule 712-O2 comprises
 
two heavy chain polypeptide chains with an amino acid sequence as shown in SEQ ID NO: 31 and two light chains with an amino acid sequence as shown in SEQ ID NO: 4.
[0099] The Ang-2 antagonist peptide L1-15 is fused to the N-terminals of the heavy chains of a VEGF-binding antibody in the case of 712-O. In the case of 712-O2, L1-15 is fused to the C-terminals of the heavy chains. The IC-50’s for the Ang-2 blocking assay were approximately 33 pM for 712-O and approximately 78 pM for 712-O2. Since L1-15, together with other peptides including L1-7, L1-10 and L1-21, was considered an N-terminal fusion peptide and was only tested to be active when it is fused to the N-terminal of the Fc as described in WO2004/092215A2. It was surprised that the chimeric molecule 712-O2 was significantly potent with an IC-50 of approximately 78 pM.
Table 6. Blocking ELISA Results: Inhibition of Binding of Ang-1 or Ang-2 to Tie-2.
Figure imgf000031_0001
Example 4 - Cell-based activity assay: In vitro Human Umbilical Vein Endothelial Cells
(HUVEC) Tube-Formation Assay
[0100] In order to confirm whether or not ASKB-E06 inhibits angiogenesis, proliferation, migration, and differentiation assays of human umbilical vein endothelial cells (HUVEC) are performed.
a) Proliferation Inhibition of HUVEC by 712-O
[0101] After 10,000 HUVEC were added to 100 ml of EBM-2 medium (Lonza, Switzerland), EBM-2 medium having VEGF-A (50 ng/ml) is added thereto, or EBM-2 medium including VEGF- A (50 ng/ml) and 712-O sample at different concentration is added thereto in each well of a 96- well plate, followed by incubation under 5% CO2, at 37° C. for 72 hours. Then, 10 ml of WST-1 solution was added thereto, followed by incubation at 37° C. for 4 hours. Absorbance is measured at 410 nm with a reference of 610 nm. The results are shown in Table 7, which indicated that 712-O had similar or higher potency than Lucentis®. It was more potent than ASKB1202 (a biosimilar of bevacizumab).
 
Table 7. HUVEC Assay Results
Figure imgf000032_0001
(2) Migration Inhibition of HUVEC by 712-O
[0102] After a bottom of Transwells, (Corning Inc., US) having a pore size of 8-mm is coated with 0.1% gelatin and mounted in a 24-well plate, a lower chamber is filled with 600 ml of EBM- 2 medium (Lonza), EBM-2 with VEGF-A (50 ng/ml), or EBM-2 with VEGF-A (50 ng/ml) and 712- O sample at different concentration. An upper chamber is provided with 100 ml of EBM-2 medium containing 1×105 HUVEC. After incubation in 37° C. cell incubator for 4 hours, a filter is detached from the Transwell and cells are fixed with methanol for 1 minute and stained with Hematoxylin/Eosin. Cells which do not migrate but are left on an upper surface of the transwell are completely removed with a cotton swab. Five random fields among the cells migrated through the filter are arbitrarily chosen under an optical microscope (×100) and the number thereof is counted.
(3) Inhibition of Tube Formation by 712-O
[0103] In order to confirm that ASKB-E06 can inhibit differentiation of HUVEC, tube formation assay is performed. More specifically, after a 96-well plate is coated with Growth Factor Reduced Matrigel (BD Biosciences, US), 15,000 HUVEC in 100 ml of EBM-2 medium, EBM-2 medium with VEGF-A (50 ng/ml), or EBM-2 medium with VEGF-A (50 ng/ml) and an antibody sample are added to each well, followed by incubation in 37° C. cell incubator for 6 hours. Then, tube formation is observed by using an inverted microscope.
Example 5 - In vivo anti-tumor activity study: Therapeutic Efficacy Studies With Systemically
Administered Dual Antagonist Chimeric Molecules
[0104] The chimeric molecule ASKB712-B is administered subcutaneously to A431 tumor- bearing mice at a once-per-day schedule 72 hours after tumor challenge. The doses used are  
1000, 200, 40 and 8ug/mouse/day. A total of 20 doses is given to all animals. Tumor volumes and body weights are recorded three times/week. At the end of the study, animals are sacrificed, and their sera are collected for measuring ASKB712-B levels by ELISA. Tumors and a panel of normal tissues are collected from all groups.
[0105] The non-limiting examples provided herein are for illustrative purposes only in order to facilitate a more complete understanding of the disclosed subject matter. These examples should not be construed to limit any of the embodiments described in the present specification, including those pertaining to the fusion peptides, pharmaceutical compositions, or methods and uses for treating cancer, proliferative retinopathies, AMD or RA.
[0106] A chimeric molecule, which comprises one or two VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein:
a) said Ang-2 antagonist peptide comprises an amino acid sequence selected from SEQ ID NO: 8-14; and
b) said VEGF-binding moiety is an antibody, an Fab or an scFv; and wherein said antibody, Fab or scFv comprises light chain CDRs as derived from a light chain with an amino acid sequence as shown in SEQ ID NO: 4, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6, and heavy chain CDRs as derived from a heavy chain with an amino acid sequence as shown in SEQ ID NO: 5, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6.
[0107] The chimeric molecule of claim 1, wherein said VEGF binding moiety comprises an antibody with a light chain amino acid sequence that is at least 95% identical to that of SEQ ID NO: 4, and heavy chain amino acid sequence that is at least 99% identical to that of SEQ ID NO: 7.
[0108] The chimeric molecule of claims 2, wherein said Ang-2 antagonist peptide is fused to the N-terminal of the heavy chain (HC) of the said antibody optionally through a peptide linker.
[0109] The chimeric molecule of claim 3, wherein the Ang-2 antagonist peptide-HC fusion polypeptide comprises an amino acid sequence that has at least 99% identity to one of SEQ ID NOS:29, 30, and SEQ ID NO:33.
[0110] The chimeric molecule of claim 2, wherein said Ang-2 antagonist peptide is fused to the C-terminal of the heavy chain of the said antibody optionally through a peptide linker.  
[0111] The chimeric molecule of claim 5, wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NOS: 31, 32, and 34.
[0112] The chimeric molecule of claim 2, wherein said Ang-2 antagonist polypeptide is fused to the N-terminals or the C-terminals of the heavy chain of said antibody through a peptide linker; and wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NO: 37, 39, 41, 43, 45, 47, 49, 51, and 53..
[0113] The chimeric molecule of claim 1, wherein said VEGF binding moiety is an Fab with a light chain amino acid sequence of at least 95% identity to SEQ ID NO: 4, and a heavy chain amino acid sequence of at least 95% identity to SEQ ID NO: 5.
[0114] The chimeric molecule of claim 8, wherein the Ang-2 antagonist peptide is fused to the N-terminal of the heavy chain of said Fab molecule through a peptide linker.
[0115] The chimeric molecule of claim 9, wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO:19 or SEQ ID NO:20.
[0116] The chimeric molecule of claim 8, wherein the Ang-2 antagonist peptide is fused to the C-terminal of the heavy chain of said Fab molecule through a peptide linker.
[0117] Chimeric molecule of claim 11, wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical to SEQ ID NO: 25 or SEQ ID NO:26.
[0118] The chimeric molecule of claim 1, wherein said VEGF binding moiety is an scFv with an amino acid sequence having at least 95% identity to SEQ ID NO: 6.
[0119] The chimeric molecule of claim 13, wherein the Ang-2 antagonist peptide is fused to the N-terminal of the scFv; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NOS:21 and 22.
[0120] The chimeric molecule of claim 13, wherein the Ang2 antagonist peptide is fused to the C-terminal of the scFv optionally; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NO:27 and SEQ ID NO:28.  
[0121] A chimeric molecule comprising a fusion protein that has one or more VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein said VEGF binding moiety is a VEGF trap with an amino acid sequence having at least 95% identity to SEQ ID NO: 3; wherein the chimeric molecule comprises two identical polypeptide chains, each having an amino acid sequence at least 99% identical to one of SEQ ID NOS:15-17, 23 and 24.
[0122] A polynucleotide or polynucleotides encoding the chimeric molecule of any one of claims 1-16.
[0123] An expression vector or vectors containing a polynucleotide or polynucleotides of claim 17.
[0124] A host cell transfected with one or more of the expression vectors of claim 18.
[0125] A method of making the chimeric molecule of any one of claims 1-16, comprising culturing a host cell transfected with one or more expression vectors containing a polynucleotide that encodes a chimeric molecule of one of claims 1 -16 under conditions that allow expression of the chimeric molecule, and isolating the chimeric molecule.
[0126] A pharmaceutical composition comprising the chimeric molecule of any one of claims 1-16 and a pharmaceutically acceptable excipient.
[0127] The pharmaceutical composition of claim 21, wherein the pharmaceutical composition contains one or more acceptable carriers.
[0128] The pharmaceutical composition of claim 21, wherein the pharmaceutical composition is in the form of a lyophilized formulation or an aqueous solution.
[0129] The pharmaceutical compositions of claim 21, wherein the pharmaceutical composition includes one or more of carriers, an excipient, a diluent, a suitable binder, a lubricant, a suspension agent, a coating agent or a solubilizing agent.
[0130] A method of treating a patient with cancer, proliferative retinopathy, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR) comprising administering to a subject a pharmaceutical composition of claim 21.
[0131] In closing, it is to be understood that although aspects of the present specification are highlighted by referring to specific embodiments, one skilled in the art will readily appreciate  
that these disclosed embodiments are only illustrative of the principles of the subject matter disclosed herein. Therefore, it should be understood that the disclosed subject matter is in no way limited to a particular compound, composition, article, apparatus, methodology, protocol, and/or reagent, etc., described herein, unless expressly stated as such. In addition, those of ordinary skill in the art will recognize that certain changes, modifications, permutations, alterations, additions, subtractions and sub-combinations thereof can be made in accordance with the teachings herein without departing from the spirit of the present specification. It is therefore intended that the following appended claims and claims hereafter introduced are interpreted to include all such changes, modifications, permutations, alterations, additions, subtractions and sub-combinations as are within their true spirit and scope.
[0132] Certain embodiments of the present invention are described herein, including the best mode known to the inventors for carrying out the invention. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the present invention to be practiced otherwise than specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described embodiments in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
[0133] Groupings of alternative embodiments, elements, or steps of the present invention are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other group members disclosed herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.
[0134] Unless otherwise indicated, all numbers expressing a characteristic, item, quantity, parameter, property, term, and so forth used in the present specification and claims are to be understood as being modified in all instances by the term“about.” As used herein, the term “about” means that the characteristic, item, quantity, parameter, property, or term so qualified encompasses a range of plus or minus ten percent above and below the value of the stated  
characteristic, item, quantity, parameter, property, or term. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specification and attached claims are approximations that may vary. For instance, as mass spectrometry instruments can vary slightly in determining the mass of a given analyte, the term "about" in the context of the mass of an ion or the mass/charge ratio of an ion refers to +/-0.50 atomic mass unit. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical indication should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
[0135] Use of the terms“may” or“can” in reference to an embodiment or aspect of an embodiment also carries with it the alternative meaning of“may not“ or“cannot.” As such, if the present specification discloses that an embodiment or an aspect of an embodiment may be or can be included as part of the inventive subject matter, then the negative limitation or exclusionary proviso is also explicitly meant, meaning that an embodiment or an aspect of an embodiment may not be or cannot be included as part of the inventive subject matter. In a similar manner, use of the term“optionally” in reference to an embodiment or aspect of an embodiment means that such embodiment or aspect of the embodiment may be included as part of the inventive subject matter or may not be included as part of the inventive subject matter. Whether such a negative limitation or exclusionary proviso applies will be based on whether the negative limitation or exclusionary proviso is recited in the claimed subject matter.
[0136] Notwithstanding that the numerical ranges and values setting forth the broad scope of the invention are approximations, the numerical ranges and values set forth in the specific examples are reported as precisely as possible. Any numerical range or value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Recitation of numerical ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate numerical value falling within the range. Unless otherwise indicated herein, each individual value of a numerical range is incorporated into the present specification as if it were individually recited herein.
[0137] The terms“a,”“an,”“the” and similar references used in the context of describing the present invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Further, ordinal indicators– such as“first,”“second,”“third,” etc.– for identified elements are used to distinguish between the elements, and do not indicate or imply a required  
or limited number of such elements, and do not indicate a particular position or order of such elements unless otherwise specifically stated. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g.,“such as”) provided herein is intended merely to better illuminate the present invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the present specification should be construed as indicating any non-claimed element essential to the practice of the invention.
[0138] When used in the claims, whether as filed or added per amendment, the open-ended transitional term“comprising” (and equivalent open-ended transitional phrases thereof like including, containing and having) encompasses all the expressly recited elements, limitations, steps and/or features alone or in combination with unrecited subject matter; the named elements, limitations and/or features are essential, but other unnamed elements, limitations and/or features may be added and still form a construct within the scope of the claim. Specific embodiments disclosed herein may be further limited in the claims using the closed-ended transitional phrases“consisting of” or“consisting essentially of” in lieu of or as an amended for “comprising.” When used in the claims, whether as filed or added per amendment, the closed- ended transitional phrase“consisting of” excludes any element, limitation, step, or feature not expressly recited in the claims. The closed-ended transitional phrase“consisting essentially of” limits the scope of a claim to the expressly recited elements, limitations, steps and/or features and any other elements, limitations, steps and/or features that do not materially affect the basic and novel characteristic(s) of the claimed subject matter. Thus, the meaning of the open-ended transitional phrase“comprising” is being defined as encompassing all the specifically recited elements, limitations, steps and/or features as well as any optional, additional unspecified ones. The meaning of the closed-ended transitional phrase“consisting of” is being defined as only including those elements, limitations, steps and/or features specifically recited in the claim whereas the meaning of the closed-ended transitional phrase“consisting essentially of” is being defined as only including those elements, limitations, steps and/or features specifically recited in the claim and those elements, limitations, steps and/or features that do not materially affect the basic and novel characteristic(s) of the claimed subject matter. Therefore, the open-ended transitional phrase“comprising” (and equivalent open-ended transitional phrases thereof) includes within its meaning, as a limiting case, claimed subject matter specified by the closed- ended transitional phrases“consisting of” or“consisting essentially of.” As such embodiments described herein or so claimed with the phrase“comprising” are expressly or inherently  
unambiguously described, enabled and supported herein for the phrases“consisting essentially of” and“consisting of.”
[0139] All patents, patent publications, and other publications referenced and identified in the present specification are individually and expressly incorporated herein by reference in their entirety for the purpose of describing and disclosing, for example, the compositions and methodologies described in such publications that might be used in connection with the present invention. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.
[0140] Lastly, the terminology used herein is for the purpose of describing particular embodiments only and is not intended to limit the scope of the present invention, which is defined solely by the claims. Accordingly, the present invention is not limited to that precisely as shown and described.
SEQUENCES
[0141] SEQ ID NO: 1, Bevacizumab Heavy Chain:
10 20 30 40 50 60
EVQLVESGGG LVQPGGSLRL SCAASGYTFT NYGMNWVRQA PGKGLEWVGW INTYTGEPTY
70 80 90 100 110 120
AADFKRRFTF SLDTSKSTAY LQMNSLRAED TAVYYCAKYP HYYGSSHWYF DVWGQGTLVT
130 140 150 160 170 180
VSSASTKGPS VFPLAPSSKS TSGGTAALGC LVKDYFPEPV TVSWNSGALT SGVHTFPAVL
190 200 210 220 230 240
QSSGLYSLSS VVTVPSSSLG TQTYICNVNH KPSNTKVDKK VEPKSCDKTH TCPPCPAPEL
250 260 270 280 290 300
LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE
310 320 330 340 350 360
 
QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS
370 380 390 400 410 420
REEMTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK
430 440 450
SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK [0142] SEQ ID NO: 2, Bevacizumab Light Chain:
10 20 30 40 50 60
DIQMTQSPSS LSASVGDRVT ITCSASQDIS NYLNWYQQKP GKAPKVLIYF TSSLHSGVPS
70 80 90 100 110 120
RFSGSGSGTD FTLTISSLQP EDFATYYCQQ YSTVPWTFGQ GTKVEIKRTV AAPSVFIFPP
130 140 150 160 170 180
SDEQLKSGTA SVVCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT
190 200 210
LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC [0143] SEQ ID NO: 3, VEGF Trap Aflibercept
10 20 30 40 50 60
SDTGRPFVEM YSEIPEIIHM TEGRELVIPC RVTSPNITVT LKKFPLDTLI PDGKRIIWDS
70 80 90 100 110 120
RKGFIISNAT YKEIGLLTCE ATVNGHLYKT NYLTHRQTNT IIDVVLSPSH GIELSVGEKL
130 140 150 160 170 180
VLNCTARTEL NVGIDFNWEY PSSKHQHKKL VNRDLKTQSG SEMKKFLSTL TIDGVTRSDQ
190 200 210 220 230 240
GLYTCAASSG LMTKKNSTFV RVHEKDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR
250 260 270 280 290 300
TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN
310 320 330 340 350 360
GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS  
370 380 390 400 410 420
DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH
430
YTQKSLSLSP G(K) [0144] SEQ ID NO: 4, Protein Sequence for Light Chain, Ranibizumab (VEGF Fab) DIQLTQSPSSLSASVGDRVTITCSASQDISNYLNWYQQKPGKAPKVLIYFTSSLHSGVPSRFS GSGSGTDFTLTISSLQPEDFATYYCQQYSTVPWTFGQGTKVEIKRTVAAPSVFIFPPSDE QLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLS KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
[0145] SEQ ID NO: 5, Protein Sequence for Heavy Chain Ranibizumab (VEGF Fab) EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGE PTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQ GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHL
[0146] SEQ ID NO:6, Protein Sequence for a VEGF ScFv EIVMTQSPSTLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRF SGSGSGAEFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGG SGGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGL EWVGFIDPDDDPYYATWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSG WGLDIWGQGTLVTVSS
[0147] SEQ ID NO:7, Protein Sequence for a heavy chain of a VEGF antibody
EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGE PTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQ GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPP CPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0148] SEQ ID NO: 8, L1-7
 
AQQTNFMPM DDLEQRLYEQ FILQQG
[0149] SEQ ID NO: 9, L1-10
AQQKFQPLD ELEQTLYEQF MLQQA
[0150] SEQ ID: NO: 10, L1-15
AQQKYQPLD ELDKTLYDQF MLQQG
[0151] SEQ ID NO: 11, L1-7B
QTNFMPM DDLEQRLYEQ FILQQG
[0152] SEQ ID NO: 12, L1-10B
QKFQPLD ELEQTLYEQF MLQQA
[0153] SEQ ID NO: 13, L1-15B
QKYQPLD ELDKTLYDQF MLQQG
[0154] SEQ ID NO: 14, CVX-060:
QKYQPLDEKDKTLYDQFMLQQG
[0155] SEQ ID NO 15, L1-15 fused to the N-terminus of the VEGF Trap, with linker peptide GGGGSGGGGSGGGGS
10 20 30 40 50 60
AQQKYQPLDE LDKTLYDQFM LQQGGGGGSG GGGSGGGGSS DTGRPFVEMY SEIPEIIHMT
70 80 90 100 110 120
EGRELVIPCR VTSPNITVTL KKFPLDTLIP DGKRIIWDSR KGFIISNATY KEIGLLTCEA
130 140 150 160 170 180
TVNGHLYKTN YLTHRQTNTI IDVVLSPSHG IELSVGEKLV LNCTARTELN VGIDFNWEYP
190 200 210 220 230 240
SSKHQHKKLV NRDLKTQSGS EMKKFLSTLT IDGVTRSDQG LYTCAASSGL MTKKNSTFVR
250 260 270 280 290 300
VHEKDKTHTC PPCPAPELLG GPSVFLFPPK PKDTLMISRT PEVTCVVVDV SHEDPEVKFN
310 320 330 340 350 360
WYVDGVEVHN AKTKPREEQY NSTYRVVSVL TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI
 
370 380 390 400 410 420
SKAKGQPREP QVYTLPPSRD ELTKNQVSLT CLVKGFYPSD IAVEWESNGQ PENNYKTTPP
430 440 450 460 470
VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS VMHEALHNHY TQKSLSLSPG K [0156] SEQ ID NO 16, Protein Sequence for AMD-I (L1-15 fused to VEGF Trap)
Xaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFMLQQGXaa25Xaa26(GGGGS)nSDTGRPF VEMYSEIPEIIHMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRIIWDSRKGFIISNATY KEIGLLTCEATVNGHLYKTNYLTHRQTNTIIDVVLSPSHGIELSVGEKLVLNCTARTELNVG IDFNWEYPSSKHQHKKLVNRDLKTQSGSEMKKFLSTLTIDGVTRSDQGLYTCAASSGLM TKKNSTFVRVHEKDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVS HEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSN KALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNG QPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL SPG(K)
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa25 is L or deleted; Xaa5 is Y or F; Xaa12 is D or E; Xaa13 is Q or K; Xaa 17 is D or E; Xaa26 is E is deleted; n = 0, 1, 2, 3, 4, or 5; and the C-terminal amino acid K may be deleted.
[0157] SEQ ID NO 17, Protein Sequence for AMD-J (L1-7 fused to VEGF Trap)
Xaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27(GGGGS)nSDTGRPFVEMYSEIPEII HMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRIIWDSRKGFIISNATYKEIGLLTCEAT VNGHLYKTNYLTHRQTNTIIDVVLSPSHGIELSVGEKLVLNCTARTELNVGIDFNWEYPSS KHQHKKLVNRDLKTQSGSEMKKFLSTLTIDGVTRSDQGLYTCAASSGLMTKKNSTFVRV HEKDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN WYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKT ISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG(K) wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; n = 0, 1, 2, 3, 4, or 5; and the C-terminal amino acid K may be deleted.
[0158] SEQ ID NO 19, Protein Sequence for AMD-K Heavy Chain (L1-15 fused to VEGF Fab)
 
Xaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFMLQQGXaa25Xaa26(GGGGS)nEVQLVES GGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADF KRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVS SASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHL
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa5 is Y or F; Xaa12 is D or E;
Xaa13 is Q or K; Xaa 17 is D or E; Xaa25 is L or deleted; Xaa26 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0159] SEQ ID NO 20, Protein Sequence for AMD-L Heavy Chain (L1-7 fused to VEGF Fab)
Xaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27(GGGGS)nEVQLVESGGGLVQPGG SLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADFKRRFTFSLDT SKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVSSASTKGPSVF PLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV VTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHL
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0160] SEQ ID NO 21, Protein Sequence for AMD-N (L1-15 fused to VEGF ScFv)
Xaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFMLQQGXaa25Xaa26(GGGGS)nEIVMTQSP STLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRFSGSGSGA EFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGGSGGGGSG GGGSEVQLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGLEWVGFID PDDDPYYATWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSGWGLDIWG QGTLVTVSS
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa5 is Y or F; Xaa12 is D or E;
Xaa13 is Q or K; Xaa 17 is D or E; Xaa25 is L or deleted; Xaa26 is E is deleted; and n = 0, 1, 2, 3, 4, or 5
SEQ ID NO 22, Protein Sequence for AMD-Q (L1-7 fused to VEGF ScFv)
Xaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27(GGGGS)nEIVMTQSPSTLSASV GDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRFSGSGSGAEFTLTISS LQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGGSGGGGSGGGGSEV  
QLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGLEWVGFIDPDDDPYY ATWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSGWGLDIWGQGTLVTV SS
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0161] SEQ ID NO 23, Protein Sequence for AMD-I-C terminal (L1-15 fused to C- terminal of VEGF Trap)
SDTGRPFVEMYSEIPEIIHMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRIIWDSRKG FIISNATYKEIGLLTCEATVNGHLYKTNYLTHRQTNTIIDVVLSPSHGIELSVGEKLVLNCTA RTELNVGIDFNWEYPSSKHQHKKLVNRDLKTQSGSEMKKFLSTLTIDGVTRSDQGLYTC AASSGLMTKKNSTFVRVHEKDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVT CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKE YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHY TQKSLSLSPGA(GGGGS)nXaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFMLQQGXa a
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa5 is Y or F; Xaa12 is D or E;
Xaa13 is Q or K; Xaa 17 is D or E; Xaa25 is L or deleted; Xaa26 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0162] SEQ ID NO 24, Protein Sequence for AMD-J-C terminal (L1-7 fused to C- terminal of VEGF Trap)
SDTGRPFVEMYSEIPEIIHMTEGRELVIPCRVTSPNITVTLKKFPLDTLIPDGKRIIWDSRKGFII SNATYKEIGLLTCEATVNGHLYKTNYLTHRQTNTIIDVVLSPSHGIELSVGEKLVLNCTART ELNVGIDFNWEYPSSKHQHKKLVNRDLKTQSGSEMKKFLSTLTIDGVTRSDQGLYTCAA SSGLMTKKNSTFVRVHEKDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYK CKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVE WESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYT QKSLSLSPGA(GGGGS)nXaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27 wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
 
[0163] SEQ ID NO 25, Protein Sequence for AMD-K-C terminal Heavy Chain (L1-15 fused to C-terminal VEGF Fab)
EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPT YAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGT LVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHL(GGGG
Figure imgf000046_0001
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa5 is Y or F; Xaa12 is D or E;
Xaa13 is Q or K; Xaa 17 is D or E; Xaa25 is L or deleted; Xaa26 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0164] SEQ ID NO 26, Protein Sequence for AMD-L-C terminal Heavy Chain (L1-7 fused to VEGF Fab)
EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPT YAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGT LVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHL(GGGG S)nXaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0165] SEQ ID NO 27, Protein Sequence for AMD-N-C terminal (L1-15 fused to C- terminal VEGF ScFv)
EIVMTQSPSTLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRF SGSGSGAEFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGG SGGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGL EWVGFIDPDDDPYYATWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSG WGLDIWGQGTLVTVSS(GGGGS)nXaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFML QQGXaa25Xaa26; wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa5 is Y or F; Xaa12 is D or E; Xaa13 is Q or K; Xaa 17 is D or E; Xaa25 is L or deleted; Xaa26 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0166] SEQ ID NO 28, Protein Sequence for AMD-Q-C terminal (L1-7 fused to VEGF ScFv)
 
EIVMTQSPSTLSASVGDRVIITCQASEIIHSWLAWYQQKPGKAPKLLIYLASTLASGVPSRFS GSGSGAEFTLTISSLQPDDFATYYCQNVYLASTNGANFGQGTKLTVLGGGGGSGGGGS GGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCTASGFSLTDYYYMTWVRQAPGKGLE WVGFIDPDDDPYYATWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAGGDHNSGW GLDIWGQGTLVTVSS(GGGGS)n
Xaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0167] SEQ ID NO: 29, Protein Sequence for ASKB712-O (L1-15 fused to the N- terminal of an VEGF-binding antibody)
AQQKYQPLDELDKTLYDQFMLQQGLEGGGGSGGGGSGGGGSEVQLVESGGGLVQPGGS LRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADFKRRFTFSLDTS KSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVSSASTKGPSVFP LAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVV TVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPP KPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQV SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNV FSCSVMHEALHNHYTQKSLSLSPGK
[0168] SEQ ID NO: 30, Protein Sequence for ASKB712-O3 (L1-15 fused to the N- terminal of an VEGF-binding antibody)
Xaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFMLQQGXaa25Xaa26(GGGGS)nEVQLVES GGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADF KRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVS SASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAA GGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREE QYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPS REEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVD KSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK; wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa5 is Y or F; Xaa12 is D or E; Xaa13 is Q or K; Xaa 17 is D
 
or E; Xaa25 is L or deleted; Xaa26 is E is deleted; n = 0, 1, 2, 3, 4, or 5; and the C- terminal amino acid K may be deleted.
[0169] SEQ ID NO:31, Protein Sequence for ASKB712-O2 (L1-15 fused to the C- terminal of an VEGF-binding antibody)
EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPT YAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGT LVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCP APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKT KPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGAGGGGSGGGGSGGGGSA QQKYQPLDELDKTLYDQFMLQQGLE
[0170] SEQ ID NO: 32, Protein Sequence for ASKB712-O4 (L1-15 fused to the C- terminal of an VEGF-binding antibody)
EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGE PTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQ GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPP CPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGA
(GGGGS)nXaa1Xaa2QKXaa5QPLDELXaa12Xaa13TLYXaa17QFMLQQGXaa25Xaa26 wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa25 is L or deleted; Xaa5 is Y or F; Xaa12 is D or E; Xaa13 is Q or K; Xaa 17 is D or E; Xaa26 is E is deleted; n = 0, 1, 2, 3, 4, or 5.
[0171] SEQ ID NO: 33, Protein Sequence for ASKB712-P (L1-7 fused to the N- terminal of an VEGF-binding antibody)
Xaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27(GGGGS)nEVQLVESGGGLVQP GGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADFKRRFTFSL DTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVSSASTKGPS  
VFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLS SVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFL FPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKN QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQ GNVFSCSVMHEALHNHYTQKSLSLSPGK
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted; and n = 0, 1, 2, 3, 4, or 5.
[0172] SEQ ID NO: 34, Protein Sequence for ASKB712-P2 (L1-7 fused to the C- terminal of an VEGF-binding antibody)
EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPT YAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGT LVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCP APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKT KPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGA(GGGGS)n
Xaa1Xaa2QTNFMPMDDLEQRLYEQFILQQGXaa26Xaa27
wherein Xaa1 is A, G, or deleted; Xaa2 is Q or A or deleted; Xaa26 is L or deleted; Xaa27 is E is deleted;
[0173] SEQ ID NO: 35 - 712O_L7_3xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCCG CTCAGACCAACTTCATGCCTATGGACGACCTGGAACAGCGGCTGTACGAGCAGTTCA TCCTGCAGCAAGGACTGGAAGGCGGCGGAGGATCTGGCGGAGGCGGTAGCGGAG GCGGTGGATCTGAAGTGCAGCTGGTTGAAAGTGGCGGCGGATTGGTTCAGCCTGGC GGATCTCTGAGACTGTCTTGTGCCGCCTCTGGCTACGACTTCACCCACTACGGCATG AATTGGGTCCGACAGGCTCCCGGCAAAGGCCTGGAATGGGTCGGATGGATCAACAC CTATACCGGCGAGCCTACCTACGCCGCCGATTTCAAGCGGAGATTCACCTTCTCCCT GGACACCTCCAAGTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCCGAGGACA CCGCCGTGTACTACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACT TCGACGTGTGGGGACAGGGCACACTGGTCACAGTGTCCTCCGCCTCTACCAAGGGA  
CCCTCTGTGTTTCCTCTGGCTCCCTCCAGCAAGTCCACCTCTGGTGGAACAGCTGCT CTGGGCTGCCTGGTCAAGGACTACTTTCCTGAGCCTGTGACCGTGTCCTGGGCTTCT GGTGCTCTGACATCTGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCT GTACTCTCTGTCCTCTGTCGTGACCGTGCCTTCCAGCTCTCTGGGAACCCAGACCTA CATCTGCAATGTGAACCACAAGCCTTCCAACACCAAGGTCGACAAGAAGGTGGAACC CAAGTCCTGCGATAAGACCCACACCTGTCCTCCATGTCCTGCTCCAGAAGCTGCTGG CGGCCCATCCGTGTTTCTGTTCCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCG GACCCCTGAAGTGACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGA AGTTCAATTGGTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGA GAGGAACAGTACAACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCA GGATTGGCTGAACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTG CTCCTATCGAAAAGACCATCTCCAAGGCCAAGGGCCAGCCTAGGGAACCCCAGGTT TACACCTTGCCTCCAAGCCGGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTG CCTCGTGAAGGGCTTCTACCCTTCCGATATCGCCGTGGAATGGGAGAGCAATGGCC AGCCTGAGAACAACTACAAGACCACTCCTCCTGTGCTGGACTCCGACGGCTCATTCT TCCTGTACTCCAAGCTGACAGTGGACAAGTCCAGATGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAATCACTACACACAGAAGTCCCTGTCT CTGTCCCCTGGCAAGTAA
[0174] SEQ ID NO: 36 - 712O_L7_2xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCCG CTCAGACCAACTTCATGCCTATGGACGACCTGGAACAGCGGCTGTACGAGCAGTTCA TCCTGCAGCAAGGACTGGAAGGCGGCGGAGGATCTGGAGGCGGTGGATCTGAAGT GCAGCTGGTTGAAAGTGGCGGCGGATTGGTTCAGCCTGGCGGATCTCTGAGACTGT CTTGTGCCGCCTCTGGCTACGACTTCACCCACTACGGCATGAATTGGGTCCGACAG GCTCCCGGCAAAGGCCTGGAATGGGTCGGATGGATCAACACCTATACCGGCGAGCC TACCTACGCCGCCGATTTCAAGCGGAGATTCACCTTCTCCCTGGACACCTCCAAGTC TACCGCCTACCTGCAGATGAACTCCCTGAGAGCCGAGGACACCGCCGTGTACTACT GCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTCGACGTGTGGGGA CAGGGCACACTGGTCACAGTGTCCTCCGCCTCTACCAAGGGACCCTCTGTGTTTCCT CTGGCTCCCTCCAGCAAGTCCACCTCTGGTGGAACAGCTGCTCTGGGCTGCCTGGT CAAGGACTACTTTCCTGAGCCTGTGACCGTGTCCTGGGCTTCTGGTGCTCTGACATC TGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTGTACTCTCTGTCCTC TGTCGTGACCGTGCCTTCCAGCTCTCTGGGAACCCAGACCTACATCTGCAATGTGAA
 
CCACAAGCCTTCCAACACCAAGGTCGACAAGAAGGTGGAACCCAAGTCCTGCGATA AGACCCACACCTGTCCTCCATGTCCTGCTCCAGAAGCTGCTGGCGGCCCATCCGTG TTTCTGTTCCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGTG ACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGTA CGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTACA ACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAAC GGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAAA GACCATCTCCAAGGCCAAGGGCCAGCCTAGGGAACCCCAGGTTTACACCTTGCCTC CAAGCCGGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTGCCTCGTGAAGGGC TTCTACCCTTCCGATATCGCCGTGGAATGGGAGAGCAATGGCCAGCCTGAGAACAA CTACAAGACCACTCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAA GCTGACAGTGGACAAGTCCAGATGGCAGCAGGGCAACGTGTTCTCCTGCTCCGTGA TGCACGAGGCCCTGCACAATCACTACACACAGAAGTCCCTGTCTCTGTCCCCTGGCA AGTAA
[0175] SEQ ID NO: 37 - 712O_L7_2xGS_Protein sequence
MEFGLSWLFLVAILKGALAAQTNFMPMDDLEQRLYEQFILQQGLEGGGGSGGGGSEVQLV ESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAA DFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVT VSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAV LQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPE AAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPR EEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLP PSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0176] SEQ ID NO: 38 - 712O_L7_1xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCCG CTCAGACCAACTTCATGCCTATGGACGACCTGGAACAGCGGCTGTACGAGCAGTTCA TCCTGCAGCAAGGACTGGAAGGAGGCGGTGGATCTGAAGTGCAGCTGGTTGAAAGT GGCGGCGGATTGGTTCAGCCTGGCGGATCTCTGAGACTGTCTTGTGCCGCCTCTGG CTACGACTTCACCCACTACGGCATGAATTGGGTCCGACAGGCTCCCGGCAAAGGCC TGGAATGGGTCGGATGGATCAACACCTATACCGGCGAGCCTACCTACGCCGCCGAT TTCAAGCGGAGATTCACCTTCTCCCTGGACACCTCCAAGTCTACCGCCTACCTGCAG ATGAACTCCCTGAGAGCCGAGGACACCGCCGTGTACTACTGCGCTAAGTACCCCTA  
CTACTACGGCACCAGCCACTGGTACTTCGACGTGTGGGGACAGGGCACACTGGTCA CAGTGTCCTCCGCCTCTACCAAGGGACCCTCTGTGTTTCCTCTGGCTCCCTCCAGCA AGTCCACCTCTGGTGGAACAGCTGCTCTGGGCTGCCTGGTCAAGGACTACTTTCCTG AGCCTGTGACCGTGTCCTGGGCTTCTGGTGCTCTGACATCTGGCGTGCACACCTTTC CAGCTGTGCTGCAGTCCTCCGGCCTGTACTCTCTGTCCTCTGTCGTGACCGTGCCTT CCAGCTCTCTGGGAACCCAGACCTACATCTGCAATGTGAACCACAAGCCTTCCAACA CCAAGGTCGACAAGAAGGTGGAACCCAAGTCCTGCGATAAGACCCACACCTGTCCT CCATGTCCTGCTCCAGAAGCTGCTGGCGGCCCATCCGTGTTTCTGTTCCCTCCAAAG CCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGTGACCTGCGTGGTGGTGGA TGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGTACGTGGACGGCGTGGAAG TGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTACAACTCCACCTACAGAGTG GTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAACGGCAAAGAGTACAAGTG CAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAAAGACCATCTCCAAGGCCAA GGGCCAGCCTAGGGAACCCCAGGTTTACACCTTGCCTCCAAGCCGGGAAGAGATGA CCAAGAACCAGGTGTCCCTGACCTGCCTCGTGAAGGGCTTCTACCCTTCCGATATCG CCGTGGAATGGGAGAGCAATGGCCAGCCTGAGAACAACTACAAGACCACTCCTCCT GTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAAGCTGACAGTGGACAAGTCC AGATGGCAGCAGGGCAACGTGTTCTCCTGCTCCGTGATGCACGAGGCCCTGCACAA TCACTACACACAGAAGTCCCTGTCTCTGTCCCCTGGCAAGTAA
[0177] SEQ ID NO: 39 - 712O_L7_1xGS_DNA sequence
MEFGLSWLFLVAILKGALAAQTNFMPMDDLEQRLYEQFILQQGLEGGGGSEVQLVESGGGL VQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADFKRRF TFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVSSAST KGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL YSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGP SVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREE MTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0178] SEQ ID NO: 40712O_L10_3xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTTTGGCCG CTCAGCAGAAGTTTCAGCCTCTGGACGAGCTGGAACAGACCCTGTACGAGCAGTTC ATGCTCCAGCAGGCTTTGGAAGGCGGCGGAGGATCTGGCGGAGGCGGTAGCGGAG  
GCGGTGGATCTGAAGTGCAGCTGGTTGAAAGTGGCGGCGGATTGGTTCAGCCTGGC GGATCTCTGAGACTGTCTTGTGCCGCCTCTGGCTACGACTTCACCCACTACGGCATG AATTGGGTCCGACAGGCTCCCGGCAAAGGCCTGGAATGGGTCGGATGGATCAACAC CTATACCGGCGAGCCTACCTACGCCGCCGATTTCAAGCGGAGATTCACCTTCTCCCT GGACACCTCCAAGTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCCGAGGACA CCGCCGTGTACTACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACT TCGACGTGTGGGGACAGGGCACACTGGTCACAGTGTCCTCCGCCTCTACCAAGGGA CCCTCTGTGTTTCCTCTGGCTCCCTCCAGCAAGTCCACCTCTGGTGGAACAGCTGCT CTGGGCTGCCTGGTCAAGGACTACTTTCCTGAGCCTGTGACCGTGTCCTGGGCTTCT GGTGCTCTGACATCTGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCT GTACTCTCTGTCCTCTGTCGTGACCGTGCCTTCCAGCTCTCTGGGAACCCAGACCTA CATCTGCAATGTGAACCACAAGCCTTCCAACACCAAGGTCGACAAGAAGGTGGAACC CAAGTCCTGCGATAAGACCCACACCTGTCCTCCATGTCCTGCTCCAGAAGCTGCTGG CGGCCCATCCGTGTTTCTGTTCCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCG GACCCCTGAAGTGACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGA AGTTCAATTGGTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGA GAGGAACAGTACAACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCA GGATTGGCTGAACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTG CTCCTATCGAAAAGACCATCTCCAAGGCCAAGGGCCAGCCTAGGGAACCCCAGGTT TACACCTTGCCTCCAAGCCGGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTG CCTCGTGAAGGGCTTCTACCCTTCCGATATCGCCGTGGAATGGGAGAGCAATGGCC AGCCTGAGAACAACTACAAGACCACTCCTCCTGTGCTGGACTCCGACGGCTCATTCT TCCTGTACTCCAAGCTGACAGTGGACAAGTCCAGATGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAATCACTACACACAGAAGTCCCTGTCT CTGTCCCCTGGCAAGTAA
[0179] SEQ ID NO: 41712O_L10_3xGS_Protein sequence
MEFGLSWLFLVAILKGALAAQQKFQPLDELEQTLYEQFMLQQALEGGGGSGGGGSGGGGS EVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGE PTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQ GTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVH TFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPP CPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA KTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP
 
QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0180] SEQ ID NO: 42712O_L10_2xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTTTGGCCG CTCAGCAGAAGTTTCAGCCTCTGGACGAGCTGGAACAGACCCTGTACGAGCAGTTC ATGCTCCAGCAGGCTTTGGAAGGCGGCGGAGGATCTGGAGGCGGTGGATCTGAAG TGCAGCTGGTTGAAAGTGGCGGCGGATTGGTTCAGCCTGGCGGATCTCTGAGACTG TCTTGTGCCGCCTCTGGCTACGACTTCACCCACTACGGCATGAATTGGGTCCGACAG GCTCCCGGCAAAGGCCTGGAATGGGTCGGATGGATCAACACCTATACCGGCGAGCC TACCTACGCCGCCGATTTCAAGCGGAGATTCACCTTCTCCCTGGACACCTCCAAGTC TACCGCCTACCTGCAGATGAACTCCCTGAGAGCCGAGGACACCGCCGTGTACTACT GCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTCGACGTGTGGGGA CAGGGCACACTGGTCACAGTGTCCTCCGCCTCTACCAAGGGACCCTCTGTGTTTCCT CTGGCTCCCTCCAGCAAGTCCACCTCTGGTGGAACAGCTGCTCTGGGCTGCCTGGT CAAGGACTACTTTCCTGAGCCTGTGACCGTGTCCTGGGCTTCTGGTGCTCTGACATC TGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTGTACTCTCTGTCCTC TGTCGTGACCGTGCCTTCCAGCTCTCTGGGAACCCAGACCTACATCTGCAATGTGAA CCACAAGCCTTCCAACACCAAGGTCGACAAGAAGGTGGAACCCAAGTCCTGCGATA AGACCCACACCTGTCCTCCATGTCCTGCTCCAGAAGCTGCTGGCGGCCCATCCGTG TTTCTGTTCCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGTG ACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGTA CGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTACA ACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAAC GGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAAA GACCATCTCCAAGGCCAAGGGCCAGCCTAGGGAACCCCAGGTTTACACCTTGCCTC CAAGCCGGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTGCCTCGTGAAGGGC TTCTACCCTTCCGATATCGCCGTGGAATGGGAGAGCAATGGCCAGCCTGAGAACAA CTACAAGACCACTCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAA GCTGACAGTGGACAAGTCCAGATGGCAGCAGGGCAACGTGTTCTCCTGCTCCGTGA TGCACGAGGCCCTGCACAATCACTACACACAGAAGTCCCTGTCTCTGTCCCCTGGCA AGTAA
[0181] SEQ ID NO: 43712O_L10_2xGS_Protein sequence
 
MEFGLSWLFLVAILKGALAAQQKFQPLDELEQTLYEQFMLQQALEGGGGSGGGGSEVQLV ESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAA DFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVT VSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAV LQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPE AAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPR EEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLP PSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0182] SEQ ID NO: 44712O_L10_3xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTTTGGCCG CTCAGCAGAAGTTTCAGCCTCTGGACGAGCTGGAACAGACCCTGTACGAGCAGTTC ATGCTCCAGCAGGCTTTGGAAGGAGGCGGTGGATCTGAAGTGCAGCTGGTTGAAAG TGGCGGCGGATTGGTTCAGCCTGGCGGATCTCTGAGACTGTCTTGTGCCGCCTCTG GCTACGACTTCACCCACTACGGCATGAATTGGGTCCGACAGGCTCCCGGCAAAGGC CTGGAATGGGTCGGATGGATCAACACCTATACCGGCGAGCCTACCTACGCCGCCGA TTTCAAGCGGAGATTCACCTTCTCCCTGGACACCTCCAAGTCTACCGCCTACCTGCA GATGAACTCCCTGAGAGCCGAGGACACCGCCGTGTACTACTGCGCTAAGTACCCCT ACTACTACGGCACCAGCCACTGGTACTTCGACGTGTGGGGACAGGGCACACTGGTC ACAGTGTCCTCCGCCTCTACCAAGGGACCCTCTGTGTTTCCTCTGGCTCCCTCCAGC AAGTCCACCTCTGGTGGAACAGCTGCTCTGGGCTGCCTGGTCAAGGACTACTTTCCT GAGCCTGTGACCGTGTCCTGGGCTTCTGGTGCTCTGACATCTGGCGTGCACACCTTT CCAGCTGTGCTGCAGTCCTCCGGCCTGTACTCTCTGTCCTCTGTCGTGACCGTGCCT TCCAGCTCTCTGGGAACCCAGACCTACATCTGCAATGTGAACCACAAGCCTTCCAAC ACCAAGGTCGACAAGAAGGTGGAACCCAAGTCCTGCGATAAGACCCACACCTGTCC TCCATGTCCTGCTCCAGAAGCTGCTGGCGGCCCATCCGTGTTTCTGTTCCCTCCAAA GCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGTGACCTGCGTGGTGGTGG ATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGTACGTGGACGGCGTGGAA GTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTACAACTCCACCTACAGAGT GGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAACGGCAAAGAGTACAAGT GCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAAAGACCATCTCCAAGGCC AAGGGCCAGCCTAGGGAACCCCAGGTTTACACCTTGCCTCCAAGCCGGGAAGAGAT GACCAAGAACCAGGTGTCCCTGACCTGCCTCGTGAAGGGCTTCTACCCTTCCGATAT
 
CGCCGTGGAATGGGAGAGCAATGGCCAGCCTGAGAACAACTACAAGACCACTCCTC CTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAAGCTGACAGTGGACAAGT CCAGATGGCAGCAGGGCAACGTGTTCTCCTGCTCCGTGATGCACGAGGCCCTGCAC AATCACTACACACAGAAGTCCCTGTCTCTGTCCCCTGGCAAGTAA
[0183] SEQ ID NO: 45712O_L10_1xGS_Protein sequence
MEFGLSWLFLVAILKGALAAQQKFQPLDELEQTLYEQFMLQQALEGGGGSEVQLVESGGGL VQPGGSLRLSCAASGYDFTHYGMNWVRQAPGKGLEWVGWINTYTGEPTYAADFKRRF TFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPYYYGTSHWYFDVWGQGTLVTVSSAST KGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL YSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPEAAGGP SVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREE MTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
[0184] SEQ ID NO: 46712O_C terminal L7_1xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCCG AAGTGCAGTTGGTTGAATCTGGTGGCGGATTGGTGCAGCCTGGCGGATCTCTGAGA CTGTCTTGTGCCGCCTCTGGCTACGATTTCACCCACTACGGCATGAATTGGGTCCGA CAGGCTCCTGGCAAAGGCCTGGAATGGGTCGGATGGATCAATACCTATACCGGCGA GCCTACCTACGCCGCCGACTTCAAGAGAAGATTCACCTTCTCCCTGGACACCTCCAA GTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCTGAGGACACCGCCGTGTACT ACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTTGATGTGTGGG GACAGGGCACCCTGGTCACCGTTTCTTCCGCTTCTACAAAGGGACCCAGCGTGTTC CCTCTGGCTCCTAGCTCTAAGTCTACCTCTGGCGGAACCGCTGCTCTGGGCTGTCTG GTCAAGGATTACTTCCCTGAGCCTGTGACCGTGTCCTGGAATAGTGGTGCTCTGACA TCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCTGGCCTGTACTCTCTGTCC TCTGTCGTGACCGTCCCTTCTAGCTCTCTGGGCACCCAGACCTACATCTGCAACGTG AACCACAAGCCTTCCAACACTAAGGTGGACAAGAAGGTGGAACCCAAGTCCTGCGAT AAGACCCACACCTGTCCTCCATGTCCTGCACCTGAAGCTGCTGGCGGACCCTCTGT GTTCCTGTTTCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGT GACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGT ACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTAC AACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAA  
CGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAA AGACCATCTCTAAGGCTAAGGGCCAGCCTCGGGAACCTCAGGTTTACACACTGCCTC CAAGCCGGGAAGAGATGACCAAGAATCAGGTGTCCCTGACCTGCCTCGTGAAGGGC TTCTACCCTTCCGATATCGCCGTCGAATGGGAGTCCAATGGCCAGCCTGAGAACAAC TACAAGACAACCCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAAG CTGACAGTGGACAAGTCTCGGTGGCAGCAGGGCAACGTGTTCTCCTGTTCTGTGAT GCACGAGGCCCTGCACAACCACTACACACAGAAGTCACTCTCCCTTTCCCCGGGCgc tGGCGGCGGAGGATCTGCTCAGACCAACTTCATGCCTATGGACGACCTGGAACAGC GGCTGTACGAGCAGTTCATCCTGCAGCAAGGACTGGAAtga
[0185] SEQ ID NO: 47712O_C terminal L7_1xGS_Protein sequence
MEFGLSWLFLVAILKGALAEVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAP GKGLEWVGWINTYTGEPTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYP YYYGTSHWYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKV DKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE NNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG AGGGGSAQTNFMPMDDLEQRLYEQFILQQGLE*
[0186] SEQ ID NO: 48712O_L7_C_2xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCCG AAGTGCAGTTGGTTGAATCTGGTGGCGGATTGGTGCAGCCTGGCGGATCTCTGAGA CTGTCTTGTGCCGCCTCTGGCTACGATTTCACCCACTACGGCATGAATTGGGTCCGA CAGGCTCCTGGCAAAGGCCTGGAATGGGTCGGATGGATCAATACCTATACCGGCGA GCCTACCTACGCCGCCGACTTCAAGAGAAGATTCACCTTCTCCCTGGACACCTCCAA GTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCTGAGGACACCGCCGTGTACT ACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTTGATGTGTGGG GACAGGGCACCCTGGTCACCGTTTCTTCCGCTTCTACAAAGGGACCCAGCGTGTTC CCTCTGGCTCCTAGCTCTAAGTCTACCTCTGGCGGAACCGCTGCTCTGGGCTGTCTG GTCAAGGATTACTTCCCTGAGCCTGTGACCGTGTCCTGGAATAGTGGTGCTCTGACA TCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCTGGCCTGTACTCTCTGTCC TCTGTCGTGACCGTCCCTTCTAGCTCTCTGGGCACCCAGACCTACATCTGCAACGTG AACCACAAGCCTTCCAACACTAAGGTGGACAAGAAGGTGGAACCCAAGTCCTGCGAT  
AAGACCCACACCTGTCCTCCATGTCCTGCACCTGAAGCTGCTGGCGGACCCTCTGT GTTCCTGTTTCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGT GACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGT ACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTAC AACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAA CGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAA AGACCATCTCTAAGGCTAAGGGCCAGCCTCGGGAACCTCAGGTTTACACACTGCCTC CAAGCCGGGAAGAGATGACCAAGAATCAGGTGTCCCTGACCTGCCTCGTGAAGGGC TTCTACCCTTCCGATATCGCCGTCGAATGGGAGTCCAATGGCCAGCCTGAGAACAAC TACAAGACAACCCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAAG CTGACAGTGGACAAGTCTCGGTGGCAGCAGGGCAACGTGTTCTCCTGTTCTGTGAT GCACGAGGCCCTGCACAACCACTACACACAGAAGTCACTCTCCCTTTCCCCGGGCgc tGGCGGCGGAGGATCTGGCGGAGGCGGTAGCGCTCAGACCAACTTCATGCCTATGG ACGACCTGGAACAGCGGCTGTACGAGCAGTTCATCCTGCAGCAAGGACTGGAAtga
[0187] SEQ ID NO: 49712O_L7_C_2xGS_protein sequence
MEFGLSWLFLVAILKGALAEVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAP GKGLEWVGWINTYTGEPTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYP YYYGTSHWYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKV DKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE NNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG AGGGGSGGGGSAQTNFMPMDDLEQRLYEQFILQQGLE
[0188] SEQ ID NO: 50712O_L7_C_3xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCCG AAGTGCAGTTGGTTGAATCTGGTGGCGGATTGGTGCAGCCTGGCGGATCTCTGAGA CTGTCTTGTGCCGCCTCTGGCTACGATTTCACCCACTACGGCATGAATTGGGTCCGA CAGGCTCCTGGCAAAGGCCTGGAATGGGTCGGATGGATCAATACCTATACCGGCGA GCCTACCTACGCCGCCGACTTCAAGAGAAGATTCACCTTCTCCCTGGACACCTCCAA GTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCTGAGGACACCGCCGTGTACT ACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTTGATGTGTGGG GACAGGGCACCCTGGTCACCGTTTCTTCCGCTTCTACAAAGGGACCCAGCGTGTTC  
CCTCTGGCTCCTAGCTCTAAGTCTACCTCTGGCGGAACCGCTGCTCTGGGCTGTCTG GTCAAGGATTACTTCCCTGAGCCTGTGACCGTGTCCTGGAATAGTGGTGCTCTGACA TCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCTGGCCTGTACTCTCTGTCC TCTGTCGTGACCGTCCCTTCTAGCTCTCTGGGCACCCAGACCTACATCTGCAACGTG AACCACAAGCCTTCCAACACTAAGGTGGACAAGAAGGTGGAACCCAAGTCCTGCGAT AAGACCCACACCTGTCCTCCATGTCCTGCACCTGAAGCTGCTGGCGGACCCTCTGT GTTCCTGTTTCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAGT GACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGGT ACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTAC AACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAA CGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAAA AGACCATCTCTAAGGCTAAGGGCCAGCCTCGGGAACCTCAGGTTTACACACTGCCTC CAAGCCGGGAAGAGATGACCAAGAATCAGGTGTCCCTGACCTGCCTCGTGAAGGGC TTCTACCCTTCCGATATCGCCGTCGAATGGGAGTCCAATGGCCAGCCTGAGAACAAC TACAAGACAACCCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAAG CTGACAGTGGACAAGTCTCGGTGGCAGCAGGGCAACGTGTTCTCCTGTTCTGTGAT GCACGAGGCCCTGCACAACCACTACACACAGAAGTCACTCTCCCTTTCCCCGGGCgc tGGCGGCGGAGGATCTGGCGGAGGCGGTAGCGGTGGTGGTGGATCTGCTCAGACC AACTTCATGCCTATGGACGACCTGGAACAGCGGCTGTACGAGCAGTTCATCCTGCAG CAAGGACTGGAAtga
[0189] > SEQ ID NO: 51712O_L7_C_3xGS_Protein sequence
MEFGLSWLFLVAILKGALAEVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAP GKGLEWVGWINTYTGEPTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYP YYYGTSHWYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKV DKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE NNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG AGGGGSGGGGSGGGGSAQTNFMPMDDLEQRLYEQFILQQGLE
[0190] SEQ ID NO: 52712O_L15_C_1xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGC CGAAGTGCAGTTGGTTGAATCTGGTGGCGGATTGGTGCAGCCTGGCGGATCTCTGA  
GACTGTCTTGTGCCGCCTCTGGCTACGATTTCACCCACTACGGCATGAATTGGGTCC GACAGGCTCCTGGCAAAGGCCTGGAATGGGTCGGATGGATCAATACCTATACCGGC GAGCCTACCTACGCCGCCGACTTCAAGAGAAGATTCACCTTCTCCCTGGACACCTCC AAGTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCTGAGGACACCGCCGTGTA CTACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTTGATGTGTG GGGACAGGGCACCCTGGTCACCGTTTCTTCCGCTTCTACAAAGGGACCCAGCGTGT TCCCTCTGGCTCCTAGCTCTAAGTCTACCTCTGGCGGAACCGCTGCTCTGGGCTGTC TGGTCAAGGATTACTTCCCTGAGCCTGTGACCGTGTCCTGGAATAGTGGTGCTCTGA CATCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCTGGCCTGTACTCTCTGT CCTCTGTCGTGACCGTCCCTTCTAGCTCTCTGGGCACCCAGACCTACATCTGCAACG TGAACCACAAGCCTTCCAACACTAAGGTGGACAAGAAGGTGGAACCCAAGTCCTGC GATAAGACCCACACCTGTCCTCCATGTCCTGCACCTGAAGCTGCTGGCGGACCCTCT GTGTTCCTGTTTCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAA GTGACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTG GTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGT ACAACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTG AACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGA AAAGACCATCTCTAAGGCTAAGGGCCAGCCTCGGGAACCTCAGGTTTACACACTGCC TCCAAGCCGGGAAGAGATGACCAAGAATCAGGTGTCCCTGACCTGCCTCGTGAAGG GCTTCTACCCTTCCGATATCGCCGTCGAATGGGAGTCCAATGGCCAGCCTGAGAACA ACTACAAGACAACCCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCA AGCTGACAGTGGACAAGTCTCGGTGGCAGCAGGGCAACGTGTTCTCCTGTTCTGTG ATGCACGAGGCCCTGCACAACCACTACACACAGAAGTCACTCTCCCTTTCCCCGGG
CgctGGCGGCGGAGGATCTGCCCAGCAGAAGTATCAGCCTCTGGACGAGCTGGACAA GACCCTGTACGACCAGTTCATGCTCCAGCAGGGACTGGAAtga
[0191] SEQ ID NO: 53712O_L15_C_1xGS_Protein sequence
MEFGLSWLFLVAILKGALAEVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQAP GKGLEWVGWINTYTGEPTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYP YYYGTSHWYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKV DKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE
 
NNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG AGGGGSAQQKYQPLDELDKTLYDQFMLQQGLE
[0192] SEQ ID NO: 54712O_L15_C_2xGS_DNA sequence
TGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGCC GAAGTGCAGTTGGTTGAATCTGGTGGCGGATTGGTGCAGCCTGGCGGATCTCTGAG ACTGTCTTGTGCCGCCTCTGGCTACGATTTCACCCACTACGGCATGAATTGGGTCCG ACAGGCTCCTGGCAAAGGCCTGGAATGGGTCGGATGGATCAATACCTATACCGGCG AGCCTACCTACGCCGCCGACTTCAAGAGAAGATTCACCTTCTCCCTGGACACCTCCA AGTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCTGAGGACACCGCCGTGTAC TACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTTGATGTGTGG GGACAGGGCACCCTGGTCACCGTTTCTTCCGCTTCTACAAAGGGACCCAGCGTGTT CCCTCTGGCTCCTAGCTCTAAGTCTACCTCTGGCGGAACCGCTGCTCTGGGCTGTCT GGTCAAGGATTACTTCCCTGAGCCTGTGACCGTGTCCTGGAATAGTGGTGCTCTGAC ATCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCTGGCCTGTACTCTCTGTC CTCTGTCGTGACCGTCCCTTCTAGCTCTCTGGGCACCCAGACCTACATCTGCAACGT GAACCACAAGCCTTCCAACACTAAGGTGGACAAGAAGGTGGAACCCAAGTCCTGCG ATAAGACCCACACCTGTCCTCCATGTCCTGCACCTGAAGCTGCTGGCGGACCCTCTG TGTTCCTGTTTCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAAG TGACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTGG TACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGTA CAACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGA ACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGAA AAGACCATCTCTAAGGCTAAGGGCCAGCCTCGGGAACCTCAGGTTTACACACTGCCT CCAAGCCGGGAAGAGATGACCAAGAATCAGGTGTCCCTGACCTGCCTCGTGAAGGG CTTCTACCCTTCCGATATCGCCGTCGAATGGGAGTCCAATGGCCAGCCTGAGAACAA CTACAAGACAACCCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCAA GCTGACAGTGGACAAGTCTCGGTGGCAGCAGGGCAACGTGTTCTCCTGTTCTGTGA TGCACGAGGCCCTGCACAACCACTACACACAGAAGTCACTCTCCCTTTCCCCGGGCg ctGGCGGCGGAGGATCTGGCGGAGGCGGTAGCGCCCAGCAGAAGTATCAGCCTCTG GACGAGCTGGACAAGACCCTGTACGACCAGTTCATGCTCCAGCAGGGACTGGAAtga
[0193] SEQ ID NO: 55712O_L15_C_2xGS_Protein sequence
MEFGLSWLFLVAILKGALAEVQLVESGGGLVQPGGSLRLSCAASGYDFTHYGMNWVRQ APGKGLEWVGWINTYTGEPTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAK
 
YPYYYGTSHWYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT KVDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH EDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNK ALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQ PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS PGAGGGGSGGGGSAQQKYQPLDELDKTLYDQFMLQQGLE
[0194] SEQ ID NO: 56712O_L15_C_3xGS_DNA sequence
ATGGAATTCGGCCTGTCTTGGCTGTTCCTGGTGGCCATTCTGAAGGGCGCTCTGGC CGAAGTGCAGTTGGTTGAATCTGGTGGCGGATTGGTGCAGCCTGGCGGATCTCTGA GACTGTCTTGTGCCGCCTCTGGCTACGATTTCACCCACTACGGCATGAATTGGGTCC GACAGGCTCCTGGCAAAGGCCTGGAATGGGTCGGATGGATCAATACCTATACCGGC GAGCCTACCTACGCCGCCGACTTCAAGAGAAGATTCACCTTCTCCCTGGACACCTCC AAGTCTACCGCCTACCTGCAGATGAACTCCCTGAGAGCTGAGGACACCGCCGTGTA CTACTGCGCTAAGTACCCCTACTACTACGGCACCAGCCACTGGTACTTTGATGTGTG GGGACAGGGCACCCTGGTCACCGTTTCTTCCGCTTCTACAAAGGGACCCAGCGTGT TCCCTCTGGCTCCTAGCTCTAAGTCTACCTCTGGCGGAACCGCTGCTCTGGGCTGTC TGGTCAAGGATTACTTCCCTGAGCCTGTGACCGTGTCCTGGAATAGTGGTGCTCTGA CATCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCTGGCCTGTACTCTCTGT CCTCTGTCGTGACCGTCCCTTCTAGCTCTCTGGGCACCCAGACCTACATCTGCAACG TGAACCACAAGCCTTCCAACACTAAGGTGGACAAGAAGGTGGAACCCAAGTCCTGC GATAAGACCCACACCTGTCCTCCATGTCCTGCACCTGAAGCTGCTGGCGGACCCTCT GTGTTCCTGTTTCCTCCAAAGCCTAAGGACACCCTGATGATCTCTCGGACCCCTGAA GTGACCTGCGTGGTGGTGGATGTGTCTCACGAGGACCCAGAAGTGAAGTTCAATTG GTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCTAGAGAGGAACAGT ACAACTCCACCTACAGAGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTG AACGGCAAAGAGTACAAGTGCAAGGTGTCCAACAAGGCCCTGCCTGCTCCTATCGA AAAGACCATCTCTAAGGCTAAGGGCCAGCCTCGGGAACCTCAGGTTTACACACTGCC TCCAAGCCGGGAAGAGATGACCAAGAATCAGGTGTCCCTGACCTGCCTCGTGAAGG GCTTCTACCCTTCCGATATCGCCGTCGAATGGGAGTCCAATGGCCAGCCTGAGAACA ACTACAAGACAACCCCTCCTGTGCTGGACTCCGACGGCTCATTCTTCCTGTACTCCA AGCTGACAGTGGACAAGTCTCGGTGGCAGCAGGGCAACGTGTTCTCCTGTTCTGTG ATGCACGAGGCCCTGCACAACCACTACACACAGAAGTCACTCTCCCTTTCCCCGGG
CgctGGCGGCGGAGGATCTGGCGGAGGCGGTAGCGGTGGTGGTGGATCTGCCCAGC  
AGAAGTATCAGCCTCTGGACGAGCTGGACAAGACCCTGTACGACCAGTTCATGCTCC AGCAGGGACTGGAAtga
[0195] SEQ ID NO 57, DNA sequence (DHAMDH02083016) for 2xCon4(C) fused to the C-terminus of the Heavy Chain of Bevacizumab, with linker peptide
GGGGSGGGGSGGGGS ATGGGTTGGTCCTGTATCATTCTTTTCCTCGTCGCCACTGCCACCGGAGTCCACTCAGA AGTCCAGTTGGTGGAGTCGGGAGGAGGACTGGTGCAGCCAGGCGGCTCCCTGCGC CTGTCCTGCGCGGCGTCCGGGTACACCTTCACCAACTACGGCATGAACTGGGTCCG CCAGGCCCCCGGAAAGGGGCTGGAATGGGTCGGCTGGATCAACACTTACACCGGA GAACCTACCTACGCTGCCGATTTCAAGCGGCGCTTTACTTTCTCGCTGGACACCTCC AAGAGCACCGCCTATCTCCAAATGAACTCCCTGCGGGCCGAGGATACCGCCGTGTA CTATTGCGCGAAGTACCCCCACTATTACGGTTCGTCCCATTGGTACTTCGACGTCTG GGGCCAGGGAACTCTTGTCACTGTGTCCTCCGCATCCACCAAGGGACCGTCAGTGT TCCCCCTGGCCCCGTCCTCCAAAAGCACTAGCGGAGGAACCGCAGCCTTGGGATGC CTCGTCAAGGACTACTTTCCCGAGCCTGTCACCGTGTCGTGGAACTCCGGTGCCCT CACTTCGGGCGTGCACACGTTCCCAGCGGTGCTGCAGTCCAGCGGACTGTACTCGC TGTCCTCCGTCGTGACCGTGCCTTCATCGAGCCTGGGGACCCAGACCTACATTTGCA ACGTGAACCACAAGCCCTCCAACACCAAAGTGGACAAGAAGGTCGAACCAAAGAGC TGCGACAAGACCCACACTTGCCCGCCGTGCCCGGCCCCTGAGTTGCTGGGTGGTCC ATCGGTGTTCCTGTTCCCGCCTAAGCCGAAGGACACACTCATGATCAGCAGGACCC CCGAAGTGACCTGTGTGGTGGTCGACGTGTCACATGAAGATCCCGAGGTCAAGTTC AATTGGTACGTGGACGGAGTGGAAGTGCATAATGCCAAGACTAAGCCGAGAGAGGA ACAGTACAACTCCACCTACCGGGTGGTGTCAGTGCTGACCGTGCTCCATCAGGACT GGCTCAACGGGAAGGAGTACAAGTGCAAAGTGTCGAACAAGGCTCTCCCCGCCCCT ATCGAGAAAACCATTAGCAAGGCTAAGGGACAGCCGCGGGAGCCGCAAGTGTACAC CCTGCCCCCGAGCCGCGAAGAAATGACTAAGAACCAAGTGTCCCTGACCTGTCTCG TGAAAGGGTTCTACCCGTCGGACATCGCTGTGGAGTGGGAGTCTAATGGTCAACCT GAGAACAACTACAAGACTACTCCCCCTGTGCTGGACTCCGATGGTTCCTTTTTCCTG TACTCAAAGCTGACCGTGGACAAGTCCAGATGGCAGCAGGGCAACGTGTTCAGCTG CTCCGTGATGCATGAAGCACTTCACAACCACTACACCCAGAAGTCCCTCAGCCTGTC TCCGGGGAAGGGCGGCGGAGGAGGGGCCCAGCAGGAAGAGTGTGAATGGGACCC CTGGACTTGTGAACACATGGGCGGCGGCGGCTCCGGTGGAGGAGGATCCGGCGGA
 
GGGGGCAGCGCGACGCACCAGGAGGAGTGCGAATGGGATCCATGGACTTGCGAAC ACATGCTGGAGTGA
[0196] SEQ ID NO 58, DNA sequence (DHAMDL083016), for the light chain of
Bevacizumab
ATGGGTTGGTCCTGTATTATCCTCTTTCTCGTCGCCACTGCCACCGGAGTGCACTCAGA TATTCAGATGACCCAGAGCCCCTCCTCACTGTCCGCTTCCGTGGGGGACCGCGTGA CTATCACTTGCTCGGCTTCCCAAGATATCTCCAACTACCTGAACTGGTACCAGCAGA AGCCCGGAAAGGCCCCGAAAGTGCTCATCTACTTCACCTCATCGCTGCACTCGGGA GTGCCCTCAAGATTTTCCGGCTCCGGAAGCGGGACCGACTTCACTCTTACCATCTCA TCGTTGCAACCAGAGGATTTCGCGACCTACTACTGTCAGCAGTACTCCACGGTGCCG TGGACCTTCGGACAAGGCACCAAAGTGGAGATCAAGAGGACTGTGGCGGCCCCGA GCGTGTTCATTTTCCCTCCTTCCGACGAGCAGCTGAAAAGCGGCACCGCCTCGGTC GTGTGCCTCCTGAACAACTTCTACCCGCGGGAAGCCAAGGTCCAGTGGAAGGTCGA CAACGCGCTGCAGAGCGGAAATTCCCAGGAGAGCGTGACCGAACAGGACTCCAAG GACAGCACCTATTCCCTGTCGTCTACACTGACCCTGAGCAAGGCCGACTACGAGAA GCATAAGGTCTACGCATGCGAAGTGACCCACCAAGGTCTTTCCTCCCCTGTGACCAA GTCCTTCAACCGGGGCGAATGCTGA
[0197] SEQ ID NO 59, DNA sequence (LY2.55.1), for peptide L1-15 (no LE) fused to the N-terminus of the light chain of Bevacizumab
ATGGCCTGGATGATGTTGCTTCTCGGACTTCTCGCGTATGGATCAGGGGTGGATAGCG CGCAACAGAAGTACCAGCCTTTGGACGAACTGGACAAGACCCTGTACGACCAGTTCA TGCTGCAACAGGGAGGGGGCGGTGGATCCGGGGGCGGCGGCTCCGGCGGTGGCG GATCCGACATTCAAATGACTCAGTCGCCATCGTCCCTCTCGGCATCCGTGGGAGACA GAGTGACCATCACTTGTTCCGCCTCGCAAGACATCTCCAACTACCTGAACTGGTACC AGCAGAAGCCCGGGAAGGCCCCCAAAGTGCTCATCTACTTTACTTCCTCACTGCACT CCGGGGTGCCAAGCCGCTTTAGCGGCTCCGGTTCTGGAACCGATTTCACCCTGACC ATTAGCTCACTCCAGCCGGAAGATTTCGCTACGTACTACTGCCAGCAGTATTCGACC GTGCCGTGGACTTTCGGACAGGGTACCAAAGTCGAGATCAAGCGGACCGTGGCCGC CCCGAGCGTGTTCATTTTCCCGCCTTCCGACGAGCAACTCAAGTCCGGCACTGCCTC CGTGGTCTGCCTGCTGAACAATTTCTACCCCCGCGAGGCTAAGGTCCAGTGGAAGG TCGATAACGCACTGCAGTCCGGAAACAGCCAAGAGAGCGTGACCGAACAGGACTCC AAGGACTCAACTTACTCGCTGAGCTCCACCCTGACCCTGTCGAAGGCCGACTACGAA
 
AAGCACAAAGTGTACGCCTGCGAAGTGACACATCAGGGCCTGTCATCCCCTGTCAC CAAGTCCTTCAACCGGGGAGAGTGCTGATAA
[0198] SEQ ID NO 60, DNA sequence (LY2.55.2), for peptide L1-15 (with LE) fused to the N-terminus of the light chain of Bevacizumab
ATGGCCTGGATGATGTTGCTTCTCGGACTTCTCGCGTATGGATCAGGGGTGGATAGCG CGCAACAGAAGTACCAGCCTTTGGACGAACTGGACAAGACCCTGTACGACCAGTTCA TGCTGCAACAGGGACTGGAAGGGGGCGGTGGATCCGGGGGCGGCGGCTCCGGCG GTGGCGGATCCGACATTCAAATGACTCAGTCGCCATCGTCCCTCTCGGCATCCGTG GGAGACAGAGTGACCATCACTTGTTCCGCCTCGCAAGACATCTCCAACTACCTGAAC TGGTACCAGCAGAAGCCCGGGAAGGCCCCCAAAGTGCTCATCTACTTTACTTCCTCA CTGCACTCCGGGGTGCCAAGCCGCTTTAGCGGCTCCGGTTCTGGAACCGATTTCAC CCTGACCATTAGCTCACTCCAGCCGGAAGATTTCGCTACGTACTACTGCCAGCAGTA TTCGACCGTGCCGTGGACTTTCGGACAGGGTACCAAAGTCGAGATCAAGCGGACCG TGGCCGCCCCGAGCGTGTTCATTTTCCCGCCTTCCGACGAGCAACTCAAGTCCGGC ACTGCCTCCGTGGTCTGCCTGCTGAACAATTTCTACCCCCGCGAGGCTAAGGTCCA GTGGAAGGTCGATAACGCACTGCAGTCCGGAAACAGCCAAGAGAGCGTGACCGAAC AGGACTCCAAGGACTCAACTTACTCGCTGAGCTCCACCCTGACCCTGTCGAAGGCC GACTACGAAAAGCACAAAGTGTACGCCTGCGAAGTGACACATCAGGGCCTGTCATC CCCTGTCACCAAGTCCTTCAACCGGGGAGAGTGCTGATAA
[0199] SEQ ID NO 61, DNA sequence (LY2.55.3), for peptide L1-15 (no LE) fused to the N-terminus of the heavy chain of Bevacizumab
ATGGCTTGGATGATGCTGCTGCTTGGCCTTCTCGCATACGGTTCCGGAGTCGATAGCG CCCAACAGAAGTACCAGCCTCTGGACGAACTGGATAAGACCCTGTACGATCAGTTCA TGCTGCAACAGGGGGGCGGCGGAGGATCGGGCGGTGGTGGATCCGGCGGCGGCG GATCCGAAGTGCAGCTCGTGGAGAGCGGGGGCGGACTCGTGCAGCCGGGAGGTTC GCTGAGATTGTCCTGTGCCGCCTCCGGTTACACCTTTACCAATTACGGGATGAACTG GGTCCGCCAGGCCCCCGGAAAGGGACTGGAATGGGTCGGCTGGATCAACACATATA CCGGAGAGCCCACCTACGCCGCGGACTTCAAGCGGAGATTCACCTTTTCACTGGAT ACGTCAAAGTCAACTGCATACCTCCAGATGAACTCCCTTAGGGCGGAAGATACCGCC GTGTACTACTGCGCCAAGTACCCGCACTATTACGGGTCCAGCCATTGGTACTTCGAC GTCTGGGGACAGGGGACCCTCGTGACCGTCAGCAGCGCCTCCACCAAGGGCCCGT CCGTGTTCCCTCTTGCGCCGTCGTCCAAAAGCACTTCCGGCGGCACTGCCGCCCTG GGCTGCCTCGTGAAGGATTACTTCCCGGAACCGGTCACCGTGTCGTGGAACTCCGG   AGCCCTGACTTCGGGTGTCCACACCTTCCCTGCGGTGCTGCAGAGCTCCGGTCTGT ACTCCCTCTCTTCCGTGGTCACGGTGCCCTCCTCATCACTGGGAACCCAGACCTACA TCTGCAACGTGAACCACAAGCCCTCAAACACTAAGGTCGACAAGAAAGTCGAACCGA AGTCGTGCGACAAGACCCACACTTGCCCTCCGTGCCCGGCTCCCGAGCTGCTGGG GGGCCCTTCCGTGTTTTTGTTCCCGCCGAAACCAAAGGACACTCTGATGATCAGCCG CACTCCGGAAGTGACCTGTGTGGTGGTGGACGTGTCCCACGAGGACCCAGAAGTGA AATTCAATTGGTACGTGGATGGCGTGGAAGTGCACAACGCTAAGACTAAGCCCCGC GAGGAACAGTACAACAGCACTTACCGGGTGGTGTCGGTGCTCACCGTGCTGCACCA AGATTGGCTCAACGGGAAGGAGTACAAGTGCAAAGTCTCCAACAAGGCCCTGCCCG CACCTATTGAAAAGACCATCAGCAAGGCCAAGGGACAGCCCCGGGAGCCCCAGGTC TACACCCTGCCTCCCTCGCGCGAAGAGATGACTAAGAACCAAGTGTCCCTGACCTGT CTGGTCAAGGGATTCTATCCTTCCGACATTGCCGTGGAATGGGAGTCCAACGGGCA GCCAGAGAACAACTACAAGACCACTCCACCTGTGCTGGACTCCGACGGGTCCTTCTT CTTGTACTCGAAGCTGACCGTGGACAAGTCCCGGTGGCAGCAGGGAAACGTGTTCA GCTGCTCCGTGATGCACGAGGCCTTGCATAATCATTACACCCAAAAGTCGCTGAGCT TGAGCCCGGGAAAGTGATAA
[0200] SEQ ID NO 62, DNA sequence (LY2.55.4), for peptide L1-15 (with LE) fused to the N-terminus of the heavy chain of Bevacizumab
ATGGCTTGGATGATGCTGCTGCTTGGCCTTCTCGCATACGGTTCCGGAGTCGATAGCG CCCAACAGAAGTACCAGCCTCTGGACGAACTGGATAAGACCCTGTACGATCAGTTCA TGCTGCAACAGGGGCTTGAGGGCGGCGGAGGATCGGGCGGTGGTGGATCCGGCG GCGGCGGATCCGAAGTGCAGCTCGTGGAGAGCGGGGGCGGACTCGTGCAGCCGG GAGGTTCGCTGAGATTGTCCTGTGCCGCCTCCGGTTACACCTTTACCAATTACGGGA TGAACTGGGTCCGCCAGGCCCCCGGAAAGGGACTGGAATGGGTCGGCTGGATCAA CACATATACCGGAGAGCCCACCTACGCCGCGGACTTCAAGCGGAGATTCACCTTTTC ACTGGATACGTCAAAGTCAACTGCATACCTCCAGATGAACTCCCTTAGGGCGGAAGA TACCGCCGTGTACTACTGCGCCAAGTACCCGCACTATTACGGGTCCAGCCATTGGTA CTTCGACGTCTGGGGACAGGGGACCCTCGTGACCGTCAGCAGCGCCTCCACCAAG GGCCCGTCCGTGTTCCCTCTTGCGCCGTCGTCCAAAAGCACTTCCGGCGGCACTGC CGCCCTGGGCTGCCTCGTGAAGGATTACTTCCCGGAACCGGTCACCGTGTCGTGGA ACTCCGGAGCCCTGACTTCGGGTGTCCACACCTTCCCTGCGGTGCTGCAGAGCTCC GGTCTGTACTCCCTCTCTTCCGTGGTCACGGTGCCCTCCTCATCACTGGGAACCCAG ACCTACATCTGCAACGTGAACCACAAGCCCTCAAACACTAAGGTCGACAAGAAAGTC
 
GAACCGAAGTCGTGCGACAAGACCCACACTTGCCCTCCGTGCCCGGCTCCCGAGCT GCTGGGGGGCCCTTCCGTGTTTTTGTTCCCGCCGAAACCAAAGGACACTCTGATGAT CAGCCGCACTCCGGAAGTGACCTGTGTGGTGGTGGACGTGTCCCACGAGGACCCA GAAGTGAAATTCAATTGGTACGTGGATGGCGTGGAAGTGCACAACGCTAAGACTAAG CCCCGCGAGGAACAGTACAACAGCACTTACCGGGTGGTGTCGGTGCTCACCGTGCT GCACCAAGATTGGCTCAACGGGAAGGAGTACAAGTGCAAAGTCTCCAACAAGGCCC TGCCCGCACCTATTGAAAAGACCATCAGCAAGGCCAAGGGACAGCCCCGGGAGCCC CAGGTCTACACCCTGCCTCCCTCGCGCGAAGAGATGACTAAGAACCAAGTGTCCCT GACCTGTCTGGTCAAGGGATTCTATCCTTCCGACATTGCCGTGGAATGGGAGTCCAA CGGGCAGCCAGAGAACAACTACAAGACCACTCCACCTGTGCTGGACTCCGACGGGT CCTTCTTCTTGTACTCGAAGCTGACCGTGGACAAGTCCCGGTGGCAGCAGGGAAAC GTGTTCAGCTGCTCCGTGATGCACGAGGCCTTGCATAATCATTACACCCAAAAGTCG CTGAGCTTGAGCCCGGGAAAGTGATAA
[0201] SEQ ID NO 63, DNA sequence (LY2.55.5), for the light chain of Bevacizumab ATGGCCTGGATGATGTTGCTTCTCGGACTTCTCGCGTATGGATCAGGGGTGGACTCCG ACATTCAAATGACTCAGTCGCCATCGTCCCTCTCGGCATCCGTGGGAGACAGAGTGA CCATCACTTGTTCCGCCTCGCAAGACATCTCCAACTACCTGAACTGGTACCAGCAGA AGCCCGGGAAGGCCCCCAAAGTGCTCATCTACTTTACTTCCTCACTGCACTCCGGG GTGCCAAGCCGCTTTAGCGGCTCCGGTTCTGGAACCGATTTCACCCTGACCATTAGC TCACTCCAGCCGGAAGATTTCGCTACGTACTACTGCCAGCAGTATTCGACCGTGCCG TGGACTTTCGGACAGGGTACCAAAGTCGAGATCAAGCGGACCGTGGCCGCCCCGAG CGTGTTCATTTTCCCGCCTTCCGACGAGCAACTCAAGTCCGGCACTGCCTCCGTGGT CTGCCTGCTGAACAATTTCTACCCCCGCGAGGCTAAGGTCCAGTGGAAGGTCGATAA CGCACTGCAGTCCGGAAACAGCCAAGAGAGCGTGACCGAACAGGACTCCAAGGACT CAACTTACTCGCTGAGCTCCACCCTGACCCTGTCGAAGGCCGACTACGAAAAGCACA AAGTGTACGCCTGCGAAGTGACACATCAGGGCCTGTCATCCCCTGTCACCAAGTCCT TCAACCGGGGAGAGTGCTGATAA
[0202] SEQ ID NO 64, 2xCon4(C) fused to the C-terminus of the VEGF Trap
10 20 30 40 50 60
SDTGRPFVEM YSEIPEIIHM TEGRELVIPC RVTSPNITVT LKKFPLDTLI PDGKRIIWDS
70 80 90 100 110 120
RKGFIISNAT YKEIGLLTCE ATVNGHLYKT NYLTHRQTNT IIDVVLSPSH GIELSVGEKL
130 140 150 160 170 180  
VLNCTARTEL NVGIDFNWEY PSSKHQHKKL VNRDLKTQSG SEMKKFLSTL TIDGVTRSDQ
190 200 210 220 230 240
GLYTCAASSG LMTKKNSTFV RVHEKDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR
250 260 270 280 290 300
TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN
310 320 330 340 350 360
GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS
370 380 390 400 410 420
DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH
430 440 450 460 470 480
YTQKSLSLSP GKGGGGGAQQ EECEWDPWTC EHMGSGSATG GSGSTASSGS GSATHQEECE
490
WDPWTCEHML E
 

Claims

1. A chimeric molecule, which comprises one or two VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein:
a) said Ang-2 antagonist peptide comprises an amino acid sequence selected from SEQ ID NO: 8-14; and
b) said VEGF-binding moiety is an antibody, an Fab or an scFv; and wherein said antibody, Fab or scFv comprises light chain CDRs as derived from a light chain with an amino acid sequence as shown in SEQ ID NO: 4, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6, and heavy chain CDRs as derived from a heavy chain with an amino acid sequence as shown in SEQ ID NO: 5, or derived from a scFv with an amino acid sequence as shown in SEQ ID NO: 6.
2. The chimeric molecule of claim 1, wherein said VEGF binding moiety comprises an antibody with a light chain amino acid sequence that is at least 95% identical to that of SEQ ID NO: 4, and heavy chain amino acid sequence that is at least 99% identical to that of SEQ ID NO: 7.
3. The chimeric molecule of claims 2, wherein said Ang-2 antagonist peptide is fused to the N-terminal of the heavy chain (HC) of the said antibody optionally through a peptide linker.
4. The chimeric molecule of claim 3, wherein the Ang-2 antagonist peptide-HC fusion polypeptide comprises an amino acid sequence that has at least 99% identity to one of SEQ ID NOS:29, 30, and SEQ ID NO:33.
5. The chimeric molecule of claim 2, wherein said Ang-2 antagonist peptide is fused to the C- terminal of the heavy chain of the said antibody optionally through a peptide linker.
6. The chimeric molecule of claim 5, wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NOS: 31, 32, and 34.
7. The chimeric molecule of claim 2, wherein said Ang-2 antagonist polypeptide is fused to the N-terminals or the C-terminals of the heavy chain of said antibody through a peptide linker; and wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide comprises an amino acid sequence at least 99% identical or 100% identical as one selected from SEQ ID NO: 37, 39, 41, 43, 45, 47, 49, 51, and 53..
8. The chimeric molecule of claim 1, wherein said VEGF binding moiety is an Fab with a light chain amino acid sequence of at least 95% identity to SEQ ID NO: 4, and a heavy chain amino acid sequence of at least 95% identity to SEQ ID NO: 5.
 
9. The chimeric molecule of claim 8, wherein the Ang-2 antagonist peptide is fused to the N- terminal of the heavy chain of said Fab molecule through a peptide linker.
10. The chimeric molecule of claim 9, wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical as that of SEQ ID NO:19 or SEQ ID NO:20.
11. The chimeric molecule of claim 8, wherein the Ang-2 antagonist peptide is fused to the C- terminal of the heavy chain of said Fab molecule through a peptide linker.
12. Chimeric molecule of claim 11, wherein the Ang-2 antagonist peptide-heavy chain fusion polypeptide has an amino acid sequence at least 99% identical to SEQ ID NO: 25 or SEQ ID NO:26.
13. The chimeric molecule of claim 1, wherein said VEGF binding moiety is an scFv with an amino acid sequence having at least 95% identity to SEQ ID NO: 6.
14. The chimeric molecule of claim 13, wherein the Ang-2 antagonist peptide is fused to the N-terminal of the scFv; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NOS:21 and 22.
15. The chimeric molecule of claim 13, wherein the Ang2 antagonist peptide is fused to the C-terminal of the scFv optionally; and wherein the peptide-scFv fusion has an amino acid sequence selected from SEQ ID NO:27 and SEQ ID NO:28.
16. A chimeric molecule comprising a fusion protein that has one or more VEGF-binding moieties and one or two Ang-2 antagonist peptides, wherein said VEGF binding moiety is a VEGF trap with an amino acid sequence having at least 95% identity to SEQ ID NO: 3; wherein the chimeric molecule comprises two identical polypeptide chains, each having an amino acid sequence at least 99% identical to one of SEQ ID NOS:15-17, 23 and 24.
17. A polynucleotide or polynucleotides encoding the chimeric molecule of any one of claim 1.
18. An expression vector or vectors containing a polynucleotide or polynucleotides of claim 17.
19. A host cell transfected with one or more of the expression vectors of claim 18.
20. A method of making the chimeric molecule of any one of claims 1, comprising culturing a host cell transfected with one or more expression vectors containing a polynucleotide that encodes a chimeric molecule of one of claims 1 under conditions that allow expression of the chimeric molecule, and isolating the chimeric molecule.
 
21. A pharmaceutical composition comprising the chimeric molecule of any one of claims 1- 16 and a pharmaceutically acceptable excipient.
22. The pharmaceutical composition of claim 21, wherein the pharmaceutical composition contains one or more acceptable carriers.
23. The pharmaceutical composition of claim 21, wherein the pharmaceutical composition is in the form of a lyophilized formulation or an aqueous solution.
24. The pharmaceutical compositions of claim 21, wherein the pharmaceutical composition includes one or more of carriers, an excipient, a diluent, a suitable binder, a lubricant, a suspension agent, a coating agent or a solubilizing agent.
25. A method of treating a patient with cancer, proliferative retinopathy, wet age-related macular degeneration (wAMD), macular edema following retinal vein occlusion (RVO), diabetic macular edema (DME), or diabetic retinopathy (DR) comprising administering to a subject a pharmaceutical composition of claim 21.
 
PCT/US2019/026838 2018-04-10 2019-04-10 Novel angiopoietin 2, vegf dual antagonists WO2019200006A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201980024414.9A CN112262158A (en) 2018-04-10 2019-04-10 Novel angiopoietin 2, VEGF bispecific antagonists
EP19784270.1A EP3775267A4 (en) 2018-04-10 2019-04-10 Novel angiopoietin 2, vegf dual antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862655436P 2018-04-10 2018-04-10
US62/655,436 2018-04-10

Publications (1)

Publication Number Publication Date
WO2019200006A2 true WO2019200006A2 (en) 2019-10-17

Family

ID=68164868

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/026838 WO2019200006A2 (en) 2018-04-10 2019-04-10 Novel angiopoietin 2, vegf dual antagonists

Country Status (3)

Country Link
EP (1) EP3775267A4 (en)
CN (1) CN112262158A (en)
WO (1) WO2019200006A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109310756A (en) * 2016-05-13 2019-02-05 奥美药业有限公司 Novel vascular generates element 2, VEGF bispecific antagonist
WO2022049165A1 (en) 2020-09-04 2022-03-10 F. Hoffmann-La Roche Ag Antibody that binds to vegf-a and ang2 and methods of use
WO2022256820A1 (en) * 2021-06-03 2022-12-08 Gensun Biopharma Inc. Multispecific antagonists

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL305344A (en) * 2021-03-31 2023-10-01 Hangzhou Jiayin Biotech Ltd Fusion molecules targeting vegf and angiopoietin and uses thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI761959B (en) * 2014-11-07 2022-04-21 瑞士商諾華公司 Methods for treating ocular diseases
AR103477A1 (en) * 2015-01-28 2017-05-10 Lilly Co Eli VEGFA / ANG2 COMPOUNDS
US10654922B2 (en) * 2016-05-13 2020-05-19 Askgene Pharma Inc. Angiopoietin 2, VEGF dual antagonists

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109310756A (en) * 2016-05-13 2019-02-05 奥美药业有限公司 Novel vascular generates element 2, VEGF bispecific antagonist
WO2022049165A1 (en) 2020-09-04 2022-03-10 F. Hoffmann-La Roche Ag Antibody that binds to vegf-a and ang2 and methods of use
US11732036B2 (en) 2020-09-04 2023-08-22 Hoffmann-La Roche Inc. Antibody that binds to VEGF-A and ANG2 and methods of use
WO2022256820A1 (en) * 2021-06-03 2022-12-08 Gensun Biopharma Inc. Multispecific antagonists

Also Published As

Publication number Publication date
EP3775267A2 (en) 2021-02-17
EP3775267A4 (en) 2022-05-18
CN112262158A (en) 2021-01-22

Similar Documents

Publication Publication Date Title
US11746147B2 (en) Angiopoietin 2, VEGF dual antagonists
CN107847595B (en) Antibodies specific for hyperphosphorylated tau protein and methods of use thereof
JP6779876B2 (en) Anti-transferrin receptor antibody and how to use it
US11059885B2 (en) Angiopoietin 2, VEGF dual antagonists
TWI747922B (en) Antibodies specific for hyperphosphorylated tau and methods of use thereof
KR102598180B1 (en) Anti-transthyretin antibody
KR102619359B1 (en) Anti-transthyretin antibody
EP3775267A2 (en) Novel angiopoietin 2, vegf dual antagonists
KR102581734B1 (en) Method for detecting transthyretin
KR102649788B1 (en) Anti-transthyretin antibody
KR20180020169A (en) Anti-transferrin receptor antibody with adjusted affinity
MXPA06011199A (en) Humanized anti-tgf-beta antibodies.
JP2020536534A (en) Anti-tau antibody and its use
EA034747B1 (en) Method of preventing or inhibiting photophobia or light aversion using anti-cgrp antibodies
JP2018504893A (en) Compositions and methods for antibodies targeting BMP6
KR20160037173A (en) Therapeutic fusion protein
KR20170085595A (en) Blood brain barrier receptor antibodies and methods of use
US20210032350A1 (en) Antibodies to galectin-3 and methods of use thereof
JP2022506719A (en) Tau recognition antibody
JP2024518545A (en) TREM2 agonists
KR20220027825A (en) anti-EphA4 antibody
CN114364695A (en) Antibody specifically recognizing C5A and application thereof
CN114026119A (en) Antigen binding molecules that bind PDGF-B and PDGF-D and uses thereof
JP6853392B2 (en) Fusion with anti-CD38 antibody and attenuated interferon alfa-2B
CN113166264B (en) Isolated antigen binding proteins and uses thereof

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: 19784270

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019784270

Country of ref document: EP

Effective date: 20201110