THERAPEUTIC CD47 ANTIBODIES

Provided are monoclonal antibodies and antigen-binding fragments thereof that bind to CD47 of multiple mammalian species, block the binding of SIRPalpha and TSP1 to CD47, promote phagocytosis of susceptible cancer cells, and reverse TSP1 inhibition of nitric oxide signaling, as well as monoclonal antibodies and antigen binding fragments thereof that compete with the former for binding to CD47 and that exhibit similar biological activities. Also provided are combinations of any of the foregoing. Such antibody compounds are variously effective in 1) treating tissue ischemia and ischemia-reperfusion injury (IRI) in the setting of organ preservation and transplantation, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, and other instances of surgery and/or trauma in which IRI is a component of pathogenesis; 2) in treating autoimmune and inflammatory diseases; and 3) as anti-cancer agents for treating susceptible cancer cells, promoting their phagocytic uptake and clearance.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This Continuation-in-part application claims the benefit of priority of Continuation-in-part application Ser. No. 14/302,348, filed Jun. 11th 2014, PCT Application Serial No. PCT/US2013/074766, filed Dec. 12, 2013, U.S. Provisional Application Ser. No. 61/833,691, filed Jun. 11th 2013, and U.S. Provisional Application Ser. No. 61/736,301, filed Dec. 12th 2012, the contents of each of which are herein incorporated by reference in their entirety.

INCORPORATION OF SEQUENCE LISTING

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled, “VLX0001-201CIP2 US_SequenceListing”, created on Jun. 11th 2015, which is 168,755 bytes in size. The information in the electronic format of the Sequence Listing is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

The present disclosure relates to antibodies that bind CD47, including that of humans and other mammalian species, and their use in treating conditions and disorders, such as ischemia-reperfusion injury (IRI) and cancers, mediated by this receptor.

CD47 is a cell surface receptor comprised of an extracellular IgV set domain, a 5 membrane spanning transmembrane domain, and a cytoplasmic tail that is alternatively spliced. Two ligands bind CD47: thrombospondin-1 (TSP1), and signal inhibitory receptor protein alpha (SIRPalpha). TSP1 binding to CD47 activates the heterotrimeric G protein Gi, which leads to suppression of intracellular cyclic AMP (cAMP) levels. In addition, the TSP1-CD47 pathway opposes the beneficial effects of the nitric oxide pathway in all vascular cells. The nitric oxide (NO) pathway consists of any of three nitric oxide synthase enzymes (NOS I, NOS II and NOS III) that generate bioactive gas NO using arginine as a substrate. NO can act within the cell in which it is produced, or in neighboring cells, to activate the enzyme soluble guanylyl cyclase that produces the messenger molecule cyclic GMP (cGMP). The proper functioning of the NO-cGMP pathway is essential for protecting the cardiovascular system against stresses including, but not limited to, those resulting from wounding, inflammation, hypertension, metabolic syndrome, ischemia, and ischemia-reperfusion injury (IRI). In the context of these cellular stresses, the inhibition of the NO-cGMP pathway by the TSP1-CD47 system exacerbates the effects of stress. This is a particular problem in the cardiovascular system where both cGMP and cAMP play important protective roles. There are many cases in which ischemia and reperfusion injury cause or contribute to disease, trauma, and poor outcomes of surgical procedures.

SIRPalpha is expressed on hematopoietic cells, including macrophages and dendritic cells. When it engages CD47 on a potential phagocytic target cell, phagocytosis is slowed or prevented. The CD47-SIRPalpha interaction effectively sends a “don't eat me” signal to the phagocyte. Thus, blocking the SIRPalpha-CD47 interaction with a monoclonal antibody in this therapeutic context can provide an effective anti-cancer therapy by promoting, i.e., increasing, the uptake and clearance of cancer cells by the host's immune system by increasing phagocytosis. This mechanism is effective in leukemias, lymphomas, and many types of solid tumors.

U.S. Pat. No. 8,236,313 contemplates antibodies that could be useful in the field of ischemia and blood flow to reverse and/or prevent tissue ischemia and related and associated tissue and cell damage, including antibodies that block CD47.

U.S. Pat. No. 8,101,719 discloses humanized antibodies that bind to CD47 for use in treating hematological disorders. Objects of the invention include humanized anti-CD47 antibodies and small antibody fragments exhibiting reduced antigenicity while retaining their CD47 binding activity and apoptosis-inducing activity. Such antibodies and small fragments are contemplated for use in treating hematological disorders such as various types of leukemias, malignant lymphoma, aplastic anemia, myelodysplastic syndromes, and polycythemia vera.

PCT International Publication WO 2011/143624 discloses chimeric and humanized anti-CD47 monoclonal antibodies for use as reagents for the diagnosis and immunotherapy of diseases associated with CD47 in humans, particularly in cancer therapy, for example to increase phagocytosis of cancer cells expressing CD47. Preferred antibodies are non-activating, i.e., block ligand binding, but do not signal. Disclosed humanized B6H12 and 5F9 antibodies bound soluble human CD47; B6H12 also bound human CD47 on the surface of human CD47-transfected YB2/0 cells. Humanized B6H12 and 5F9 antibodies enabled phagocytosis of CFSE-labeled HL-60 cells by mouse bone marrow- or peripheral blood-derived macrophages in vitro, respectively. Humanized B6H12 utilized human VH-3-7 and VK3-11 frameworks.

U.S. 2013/0142786 discloses non-activating anti-CD47 antibodies that increase the phagocytosis of CD47 expressing cells and these humanized or chimeric anti-CD47 antibodies can be used for therapeutic purposes, particularly in cancer therapy. Amino acid sequences of murine and humanized mAb B6H12, 5F9, and 8B6 heavy and light chain variable regions are disclosed.

Han et al. (2000) J. Biol. Chem. 275(48):37984-37992 discloses the production of mouse anti-CD47 monoclonal antibodies 400 (IgG2b), 410 (IgG1), 420 (IgG2a), 430 (IgG2a), 440 (IgG1), 450 (IgG2a), 460 (IgG1), 470 (IgG2a), and 480 (IgG1) generated by immunizing CD47-deficient mice with the extracellular domain of human CD47. No amino acid sequences of these antibodies, or their CDRs, are disclosed. Of these nine mAbs, three blocked macrophage fusion: 430, 450, and 470. Han et al. discloses no data either demonstrating or suggesting that CD47 plays a role in ischemia-reperfusion injury, autoimmune or inflammatory diseases, or cancer.

PCT International Publication WO 2013/119714 discloses anti-CD47 antibodies that do not cause a significant level of hemagglutination of human red blood cells.

There exists a need for antibodies to human CD47 that selectively block the binding of TSP1 to CD47 to promote the beneficial effects of nitric oxide-cGMP signaling and cAMP signaling in the cardiovascular system in settings in which IRI plays a role in pathogenesis. These situations/diseases include organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of digits/body parts, skin grafting, and trauma. There is also a need for antibodies that block the binding of SIRPalpha to CD47, thus providing novel anti-cancer therapies.

Antibody compounds of the present disclosure meet these needs. They bind to epitopes in the extracellular IgV domain of CD47, variously inhibiting the binding of SIRPalpha and TSP1 to CD47 and receptor activation. Antibodies that block TSP1 and SIRPalpha binding should be therapeutically useful in preventing, treating, or reducing many forms of IRI and treating cancers. Antibodies that block SIRPalpha binding promote phagocytosis of cancer cells. In view of these properties, SIRPalpha blocking antibody compounds should be therapeutically useful in treating a variety of cancers, including hematological cancers and solid tumors.

SUMMARY OF THE INVENTION

Antibody compounds disclosed herein meet these needs by exhibiting the following desirable therapeutic activities:

    • Binding to CD47 of multiple mammalian species;
    • Blocking SIRPalpha and TSP1 binding to CD47;
    • Promoting phagocytosis of cancer cells; and
    • Reversing TSP1 inhibition of nitric oxide signaling.

The present antibodies are useful in reducing, preventing, and/or treating CD47-mediated diseases or conditions (e.g., ischemia reperfusion injury and cancers).

They bind to epitopes in the extracellular IgV domain of CD47, inhibiting TSP1 and SIRPalpha binding to CD47, while inducing little or no agonist activity and promoting tumor cell phagocytic clearance. In view of these properties, antibody compounds of the present disclosure should be therapeutically useful in treating many forms of IRI and cancers.

In addition, the present antibody compounds can possess a number of other desirable properties, including broad reactivity with CD47 of a wide variety of mammalian species, including that of human, mouse, rat, pig, cynomolgus monkey, and dog, making these antibodies useful in both human and veterinary medicine. This feature is further advantageous in that it facilitates preclinical studies including, but not limited to, safety and efficacy studies, in a variety of mammalian species, and therefore the development of such antibodies as human and veterinary therapeutics.

Accordingly, the present disclosure provides:

[1] A monoclonal antibody, or antigen-binding fragment thereof, that:

(i) specifically binds human, rat, mouse, pig, cynomolgus monkey, and dog CD47;
(ii) blocks SIRPalpha and TSP1 binding to CD47;
(iii) promotes phagocytosis of cancer cells; and
(iv) reverses TSP1 inhibition of nitric oxide signaling.

[2] The monoclonal antibody or antigen-binding fragment thereof of [1], which is chimeric or humanized.

[3] The monoclonal antibody, or antigen-binding fragment thereof, of [1] or [2], which comprises three light chain complementarity determining regions (LCDRs 1-3) and three heavy chain complementarity determining regions (HCDRs 1-3), wherein:

LCDR 1 comprises the amino acid sequence RSSQSLVHSNGNTYLH (SEQ ID NO:1);

LCDR 2 comprises the amino acid sequence KVSYRFS (SEQ ID NO:2); and

LCDR 3 comprises the amino acid sequence SQNTHVPRT (SEQ ID NO:3);

HCDR 1 comprises the amino acid sequence GYTFTNYYVF (SEQ ID NO:4);

HCDR 2 comprises the amino acid sequence DINPVNGDTNFNEKFKN (SEQ ID NO:5); and

HCDR 3 comprises the amino acid sequence GGYTMDY (SEQ ID NO:6).

[4] The monoclonal antibody, or antigen-binding fragment thereof, of any one of [1]-[3], which comprises a light chain variable region (LCVR) and a heavy chain variable region (HCVR), wherein said LCVR and said HCVR comprise, respectively, amino acid sequences selected from among the following combinations of LCVRs and HCVRs:

SEQ ID NO:7 and SEQ ID NO:57; SEQ ID NO:8 and SEQ ID NO:58; SEQ ID NO:9 and SEQ ID NO:59; SEQ ID NO:10 and SEQ ID NO:60; SEQ ID NO:11 and SEQ ID NO:61; SEQ ID NO:12 and SEQ ID NO:62; SEQ ID NO:13 and SEQ ID NO:63; SEQ ID NO:14 and SEQ ID NO:64; SEQ ID NO:15 and SEQ ID NO:65; SEQ ID NO:16 and SEQ ID NO:66; SEQ ID NO:17 and SEQ ID NO:67; SEQ ID NO:18 and SEQ ID NO:68; SEQ ID NO:19 and SEQ ID NO:69; SEQ ID NO:20 and SEQ ID NO:70; SEQ ID NO:21 and SEQ ID NO:71; SEQ ID NO:22 and SEQ ID NO:72; SEQ ID NO:23 and SEQ ID NO:73; SEQ ID NO:24 and SEQ ID NO:74; SEQ ID NO:25 and SEQ ID NO:75; SEQ ID NO:26 and SEQ ID NO:76; SEQ ID NO:27 and SEQ ID NO:77; SEQ ID NO:28 and SEQ ID NO:78; SEQ ID NO:29 and SEQ ID NO:79; SEQ ID NO:30 and SEQ ID NO:80; and SEQ ID NO:31 and SEQ ID NO:81,

wherein each one of LCVR SEQ ID NOs:7-31 further comprises a constant domain having the amino acid sequence shown in SEQ ID NO:117, and wherein each one of HCVR SEQ ID NOs:57-81 comprises a constant domain selected from among SEQ ID NO:118, SEQ ID NO:119, SEQ ID NO:120, SEQ ID NO:121, and SEQ ID NO:124.

[5] A monoclonal antibody, or antigen-binding fragment thereof, that competes with said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[4] for binding to CD47, especially human CD47.

[6] A pharmaceutical composition, comprising said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5], and a pharmaceutically or physiologically acceptable carrier, diluent, or excipient.

[7] A monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] for use in human therapy or therapy of companion/pet animals, working animals, sport animals, zoo animals, or therapy of other valuable animals kept in captivity.

[8] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] for use in treating ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[9] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of [8], wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of an appendage or other body part, skin grafting, or trauma.

[10] The monoclonal antibody, or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of [8], wherein said autoimmune or inflammatory disease is selected from the group consisting of arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

[11] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] for use in treating a susceptible cancer.

[12] The monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of [11], which promotes phagocytosis of cells of said susceptible cancer.

[13] The monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of [11] or [12], wherein said susceptible cancer is selected from the group consisting of a leukemia, a lymphoma, ovarian cancer, breast cancer, endometrial cancer, colon cancer (colorectal cancer), rectal cancer, bladder cancer, lung cancer (non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung), bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma (liver cancer, hepatoma), gall bladder cancer, bile duct cancer, esophageal cancer, renal cell carcinoma, thyroid cancer, squamous cell carcinoma of the head and neck (head and neck cancer), testicular cancer, cancer of the endocrine gland, cancer of the adrenal gland, cancer of the pituitary gland, cancer of the skin, cancer of soft tissues, cancer of blood vessels, cancer of brain, cancer of nerves, cancer of eyes, cancer of meninges, cancer of oropharynx, cancer of hypopharynx, cancer of cervix, and cancer of uterus, glioblastoma, meduloblastoma, astrocytoma, glioma, meningioma, gastrinoma, neuroblastoma, melanoma, myelodysplastic syndrome, and sarcomas including, but not limited to, osteosarcoma, Ewing sarcoma, leiomyosarcoma, synovial sarcoma, alveolar soft part sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chrondrosarcoma.

[14] The monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [11]-[13], wherein said leukemia is selected from the group consisting of systemic mastocytosis, acute lymphocytic (lymphoblastic) leukemia (ALL), T cell—ALL, acute myeloid leukemia (AML), myelogenous leukemia, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), chronic myeloid leukemia (CML), myeloproliferative disorder/neoplasm, myelodysplastic syndrome, monocytic cell leukemia, and plasma cell leukemia; lymphomas, including histiocytic lymphoma and T cell lymphoma, B cell lymphomas, including Hodgkin's lymphoma and non-Hodgkin's lymphoma, such as low grade/follicular non-Hodgkin's lymphoma (NHL), cell lymphoma (FCC), mantle cell lymphoma (MCL), diffuse large cell lymphoma (DLCL), small lymphocytic (SL) NHL, intermediate grade/follicular NHL, intermediate grade diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, and Waldenstrom's Macroglobulinemia

[15] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] to treat ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[16] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] to treat a susceptible cancer.

[17] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] for the manufacture of a medicament to treat ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[18] The use of [17], wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of an appendage or other body part, skin grafting, and trauma.

[19] The use of [17] or [18], wherein said autoimmune or inflammatory disease is selected from among arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

[20] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5] for the manufacture of a medicament to treat a susceptible cancer.

[21] A method of treating ischemia or ischemia-reperfusion injury in a patient in need thereof, comprising administering to said patient an effective amount of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5].

[22] The method of [21], wherein said patient is about to be subjected to, or is experiencing, ischemia or ischemia-reperfusion injury.

[23] The method of [21] or [22], wherein said patient is a human.

[24] The method of [21] or [22], wherein said patient is a companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[25] The method of any one of [21]-[24], wherein said ischemia occurs because said patient will undergo, or is undergoing, a surgery selected from the group consisting of integument surgery, soft tissue surgery, composite tissue surgery, cosmetic surgery, surgical resections, reconstructive surgery, skin graft surgery, and limb reattachment surgery.

[26] The method of [25], wherein said skin graft is an autograft.

[27] The method of any one of [21]-[24], wherein said iseheinia occurs because said patient will undergo, or is undergoing, organ transplant surgery,

[28] The method of any one of [21]-[24], wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resection, reconstructive surgery, reattachment of an appendage or other body part, or skin grafting.

[29] The method of any one of [21]-[28], wherein said monoclonal antibody or antigen-binding fragment thereof, or competing monoclonal antibody or antigen binding fragment thereof, is administered before, during, or after said subject undergoes ischemia or surgery, or a combination of any of these time periods.

[30] The method of any one of [21]-[29], further comprising administering to said patient an effective amount of a nitric oxide donor, precursor, or both; a nitric oxide generating topical agent; an agent that activates soluble guanylyl cyclase; an agent that inhibits cyclic nucleotide phosphodiesterases; or any combination of any of the foregoing.

[31] The method of [30], wherein said nitric oxide donor or precursor is selected from the group consisting of NO gas, isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholino-sydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine; and said agent that inhibits cyclic nucleotide phosphodiesterases is selected from the group consisting of sildenafil, tadalafil, vardenafil udenafil, and avanafil.

[32] A method of increasing tissue perfusion in a subject in need thereof, comprising administering to said subject an effective amount of a monoclonal antibody, or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5].

[33] The method of [32], wherein said subject has, or is at risk of developing, at least one disease or condition selected from the group consisting of ischemia-reperfusion injury, myocardial infarction, myocardial ischemia, stroke, cerebral ischemia, sickle cell anemia, and pulmonary hypertension.

[34] The method of [32], wherein said subject has, or is at risk of developing, at least one disease or condition selected from the group consisting of hypertension, atherosclerosis, vasculopathy, ischemia secondary to diabetes, and peripheral vascular disease.

[35] The method of [32], wherein the need for increased tissue perfusion arises because said subject has had, is having, or will have, a surgery selected from the group consisting of integument surgery, soft tissue surgery, composite tissue surgery, skin graft surgery, resection of a solid organ, and reattachment or an appendage or other body part.

[36] The method of [35], wherein said skin graft is an autograft.

[37] The method of [32], wherein the need for increased tissue perfusion arises because said subject has had, is having, or will have, organ transplant surgery.

[38] The method of any one of [32]-[37], further comprising administering to said subject an effective amount of a nitric oxide donor, precursor, or both; a nitric oxide generating topical agent; an agent that activates soluble guanylyl cyclase; an agent that inhibits cyclic nucleotide phosphodiesterases; or any combination of any of the foregoing.

[39] The method of [38], wherein said nitric oxide donor or precursor is selected from the group consisting of NO gas, isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholino-sydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine; and said agent that inhibits cyclic nucleotide phosphodiesterases is selected from the group consisting of sildenafil, tadalafil, vardenafil udenafil, and avanafil.

[40] A method of transplanting a donor organ from an organ donor to an organ recipient, comprising any single step, any combination of steps, or all steps selected from the group consisting of steps i)-iii):

i) administering to said organ donor prior to, during, both prior to and during, after, or any combination thereof, donation of said donor organ an effective amount of said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5], and/or a monoclonal antibody, or antigen-binding fragment thereof, that competes with said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5] for binding to CD47;

ii) contacting said donor organ prior to, during, both prior to and during, after, or any combination thereof, transplantation to said organ recipient, and an effective amount of said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5], and/or a monoclonal antibody, or antigen-binding fragment thereof, that competes with said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5] for binding to CD47; and

iii) administering to said organ recipient prior to, during, both prior to and during, after, or any combination thereof, transplantation of said donor organ to said organ recipient, an effective amount of said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5], and/or a monoclonal antibody, or antigen-binding fragment thereof, that competes with said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5] for binding to CD47.

[41] The method of [40], wherein said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5], or monoclonal antibody, or antigen-binding fragment thereof, that competes with said monoclonal antibody or antigen-binding fragment thereof of any one of [1]-[5] for binding to CD47, reduces ischemia reperfusion injury in said donor organ.

[42] The method of [40] or [41], further comprising administering to said organ donor, said donor organ, said organ recipient, or any combination thereof, an effective amount of a nitric oxide donor, precursor, or both; a nitric oxide generating topical agent; an agent that activates soluble guanylyl cyclase; or an agent that inhibits cyclic nucleotide phosphodiesterases; or any combination of any of the foregoing.

[43] The method of [42], wherein said nitric oxide donor or precursor is selected from the group consisting of NO gas, isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholino-sydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine; and said agent that inhibits cyclic nucleotide phosphodiesterases is selected from the group consisting of sildenafil, tadalafil, vardenafil, udenafil, and avanafil.

[44] A method of treating an autoimmune or inflammatory disease in a patient in need thereof, comprising administering to said patient an effective amount of said monoclonal antibody, or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5].

[45] The method of [44], wherein said autoimmune or inflammatory disease is selected from the group consisting of arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

[46] The method of [44] or [45], wherein said patient is a human.

[47] The method of [44] or [45], wherein said patient is a companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[48] The method of any one of [44]-[47], further comprising administering to said patient an effective amount of a nitric oxide donor, precursor, or both; a nitric oxide generating topical agent; an agent that activates soluble guanylyl cyclase; an agent that inhibits cyclic nucleotide phosphodiesterases; or any combination of any of the foregoing.

[49] The method of [48], wherein said nitric oxide donor or precursor is selected from the group consisting of NO gas, isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholino-sydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine; and said agent that inhibits cyclic nucleotide phosphodiesterases is selected from the group consisting of sildenafil, tadalafil, vardenafil, udenafil, and avanafil.

[50] A method of treating a susceptible cancer in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity in need thereof, comprising administering thereto an effective amount of a monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5].

[51] The method of [50], wherein said susceptible cancer is selected from the group consisting of a leukemia, a lymphoma, ovarian cancer, breast cancer, endometrial cancer, colon cancer (colorectal cancer), rectal cancer, bladder cancer, lung cancer (non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung), bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma (liver cancer, hepatoma), gall bladder cancer, bile duct cancer, esophageal cancer, renal cell carcinoma, thyroid cancer, squamous cell carcinoma of the head and neck (head and neck cancer), testicular cancer, cancer of the endocrine gland, cancer of the adrenal gland, cancer of the pituitary gland, cancer of the skin, cancer of soft tissues, cancer of blood vessels, cancer of brain, cancer of nerves, cancer of eyes, cancer of meninges, cancer of oropharynx, cancer of hypopharynx, cancer of cervix, and cancer of uterus, glioblastoma, meduloblastoma, astrocytoma, glioma, meningioma, gastrinoma, neuroblastoma, melanoma, myelodysplastic syndrome, and sarcomas including, but not limited to, osteosarcoma, Ewing sarcoma, leiomyosarcoma, synovial sarcoma, alveolar soft part sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chrondrosarcoma.

[52] The method of [51], wherein said leukemia is selected from the group consisting of systemic mastocytosis, acute lymphocytic (lymphoblastic) leukemia (ALL), T cell—ALL, acute myeloid leukemia (AML), myelogenous leukemia, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), chronic myeloid leukemia (CML), myeloproliferative disorder/neoplasm, myelodysplastic syndrome, monocytic cell leukemia, and plasma cell leukemia; lymphomas, including histiocytic lymphoma and T cell lymphoma, B cell lymphomas, including Hodgkin's lymphoma and non-Hodgkin's lymphoma, such as low grade/follicular non-Hodgkin's lymphoma (NHL), cell lymphoma (FCC), mantle cell lymphoma (MCL), diffuse large cell lymphoma (DLCL), small lymphocytic (SL) NHL, intermediate grade/follicular NHL, intermediate grade diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, and Waldenstrom's Macroglobulinemia. [53] The method of any one of [50]-[52], wherein said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[45] increases phagocytosis of cells of said susceptible cancer.

[54] The method of [53], wherein said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of any one of [1]-[5], increases phagocytosis of cells of said susceptible cancer and inhibits SIRPalpha binding to CD47.

[55] A humanized monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD47.

[56] The use or method of any one of the previously described embodiments, further comprising administering to said patient an anti-tumor therapeutic treatment selected from the group consisting of surgery, radiation, an anti-tumor or anti-neoplastic agent, and combinations of any of the foregoing.

[57] The use or method of [56], wherein said an anti-tumor or anti-neoplastic agent is a small chemical molecule or a biologic therapeutic.

[58] The use or method of [57], wherein said small chemical molecule or biologic therapeutic is selected from the group consisting of an alkylating agent; an antimetabolite; a natural product; a miscellaneous agent used in cancer therapy; a hormone; an antagonist; a monoclonal antibody or antigen-binding fragment thereof; a cytokine; an antisense oligonucleotide; an siRNA; or a miRNA.

[59] A method of enhancing the therapeutic effect of a soluble guanylyl cyclase activator, comprising administering to a patient in need thereof:

i) an effective amount of a soluble guanylyl cyclase activator, and

ii) a monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of any one of [1]-[5] in an amount effective to enhance said therapeutic effect of said soluble guanylyl cyclase activator.

[60] The method of [59], wherein said therapeutic effect comprises treatment of ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[61] The method of [60], wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of an appendage or other body part, skin grafting, or trauma.

[62] The method of [60], wherein said autoimmune or inflammatory disease is selected from the group consisting of arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

[63] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of any one of [1]-[5] for the manufacture of a medicament to enhance the therapeutic effect of a soluble guanylyl cyclase activator.

[64] The use of [63], wherein said therapeutic effect comprises treatment of ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[65] The use of [64], wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of an appendage or other body part, skin grafting, and trauma.

[66] The use of [64], wherein said autoimmune or inflammatory disease is selected from the group consisting of arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

[67] A method of increasing the level of cGMP in vascular cells, comprising administering to said cells:

i) an effective amount of a soluble guanylyl cyclase activator, and

ii) a monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of any one of [1]-[5] in an amount effective to increase the level of cGMP in said vascular cells.

[68] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of any one of [1]-[5], which is an IgG isotype selected from among IgG1 isotype, IgG2 isotype, IgG3 isotype, and IgG4 isotypes

[69] A pharmaceutical composition, comprising said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68], and a pharmaceutically or physiologically acceptable carrier, diluent, or excipient.

[70] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] for use in human therapy or therapy of companion/pet animals, working animals, sport animals, zoo animals, or therapy of other valuable animals kept in captivity.

[71] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] for use in treating ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[72] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] for use in treating a susceptible cancer.

[73] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] to treat ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[74] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] to treat a susceptible cancer. [75] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] for the manufacture of a medicament to treat ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human or companion/pet animal, working animal, sport animal, zoo animal, or other valuable animal kept in captivity.

[76] Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [68] for the manufacture of a medicament to treat a susceptible cancer.

Expressly encompassed herein is the use of the monoclonal antibodies or antigen-binding fragments thereof of [68]-[76] in any of the methods, uses, compositions, or any other embodiments disclosed herein.

[77] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of any one of [1]-[5], wherein said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG1 isotype, the human IgG1 constant region is modified at amino acid Asn297 to prevent to glycosylation; and/or at amino acid Leu 234 and/or Leu235 to alter Fc receptor interactions; and/or to enhance FcRn binding; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization, optionally further by introduction of a disulfide bond;

i. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG2 isotype, the human IgG2 constant region is modified at amino acid Asn297 to prevent to glycosylation; and/or to enhance FcRn binding; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization, optionally further by introduction of a disulfide bond;

ii. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG3 isotype, the human IgG3 constant region is modified at amino acid Asn297 to prevent to glycosylation; and/or at amino acid 435 to extend half-life; and/or to enhance FcRn binding; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization, optionally further by introduction of a disulfide bond;

iii. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG4 isotype, the human IgG4 constant region is modified within the hinge region to prevent or reduce strand exchange; and/or at amino acid 235 to alter Fc receptor interactions; and/or at amino acid Asn297 to prevent glycosylation; and/or to enhance FcRn binding; and/or to alter antibody-dependent cellular cytotoxicity; and/or complement-dependent cytotoxicity; and/or to induce heterodimerization, optionally further by introduction of a disulfide bond.

[78] The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, of [77], wherein:

i. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG1 isotype, the human IgG1 constant region is modified at amino acid Asn297 to prevent to glycosylation by modification of Asn297→Ala (N297A) or Asn297→Gln(N297Q); and/or at amino acid Leu 234 by modification of Leu234→Ala (L234A) and/or Leu235 by modification of Leu235→Glu (L235E) or Leu235→Ala (L235A) or at both amino acid 234 and 235 by modification of Leu234→Ala and Leu235→Ala to alter Fc receptor interactions; and/or to enhance FcRn binding by modification of Met252→Tyr, Ser254→Thr, Thr256→Glu, Met428→Leu, or Asn434→Ser; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization by modification of Thr366→Trp, and optionally further by introduction of a disulfide bond by modification of Ser354→Cys and Tyr349→Cys on opposite CH3 domains;

ii. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG2 isotype, the human IgG2 constant region is modified at amino acid Asn297 to prevent to glycosylation by modification of Asn297→Ala or Asn297→Gln; and/or to enhance FcRn binding by modification of Met252→Tyr, Ser254→Thr, Thr256→Glu, Met428→Leu, or Asn434→Ser; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization by modification of Thr366→Trp, and optionally further by introduction of a disulfide bond by modification of Ser354→Cys and Tyr349→Cys on opposite CH3 domains;

iii. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG3 isotype, the human IgG3 constant region is modified at amino acid Asn297 to prevent to glycosylation by modification of Asn297→Ala or Asn297→Gln; and/or at amino acid 435 to extend half-life by modification of Arg435→His; and/or to enhance FcRn binding by modification of Met252→Tyr, Ser254→Thr, Thr256→Glu, Met428→Leu, or Asn434→Ser; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization by modification of Thr366→Trp, and optionally further by introduction of a disulfide bond by modification of Ser354→Cys and Tyr349→Cys on opposite CH3 domains;

iv. when said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen-binding fragment thereof, is human IgG4 isotype, the human IgG4 constant region is modified within the hinge region to prevent or reduce strand exchange by modification of Ser228→Pro; and/or at amino acid 235 to alter Fc receptor interactions by modification of Leu235→Glu, or by modification within the hinge and at amino acid 235 by modifying Ser228→Pro and Leu235→Glu; and/or at amino acid Asn297 to prevent glycosylation by modification of Asn297→Ala; and/or to enhance FcRn binding by modification of Met252→Tyr, Ser254→Thr, Thr256→Glu, Met428→Leu, or Asn434→Ser; and/or to alter antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity; and/or to induce heterodimerization by modification of Thr366→Trp, and optionally further by introduction of a disulfide bond by modification of Ser354→Cys and Tyr349→Cys on opposite CH3 domains.

Expressly encompassed herein is the use of the monoclonal antibodies or antigen binding fragments thereof of [77]-[78] in any of the methods, uses, compositions, or any other embodiments disclosed herein.

Further scope of the applicability of the present antibody compounds and methods will become apparent from the detailed description provided below. However, it should be understood that the detailed description and specific examples, while indicating particular embodiments of the disclosure, are given by way of illustration only since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.

BRIEF DESCRIPTION OF THE DRAWINGS

The above and other aspects, features, and advantages of the present disclosure will be better understood from the following detailed descriptions taken in conjunction with the accompanying drawing(s), all of which are given by way of illustration only, and are not limitative of the present disclosure, in which:

FIGS. 1A, 1B, 1C, and 1D show cross species binding curves to human, mouse, rat, and porcine RBCs, respectively, generated using various concentrations of purified antibodies from clones (Cl) Cl 1, Cl 1.1 (hum01 IgG1 N297Q), Cl 13, and Cl 13.1 (hum13 IgG1 N297Q) as described in Example 3. Clones Cl 1 and Cl 13 are as described in Table 3. Clones Cl 1.1 and Cl 13.1 are Fc mutants of clones Cl 1 and Cl 13, respectively, modified to reduce Fc effector function. Each has an Asn297→Gln (N297Q) mutation in the Fc domain (Sazinsky et al. (2008) PNAS 105(51):20167-20172). All of these clones exhibit concentration-dependent binding to all of the species of RBCs tested. RBCs are incubated for 60 minutes on ice with various concentrations of purified antibodies from clones Cl 1, Cl 1.1, Cl 13, and Cl 13.1. Cells are then washed with cold PBS containing EDTA, incubated for an additional hour on ice with FITC labeled donkey anti-human antibody, washed, and antibody binding is analyzed using a BD FACS Aria Cell Sorter (Becton Dickinson) or a C6 Accuri Flow Cytometer (Becton Dickinson).

FIG. 2 shows CD47 mAb-mediated phagocytosis of Jurkat cells. Target (Jurkat) and effector (human macrophages) are combined at a target effector ratio of 4:1, 10 μg/ml of the CD47 mAbs added and incubated for 2 hours at 37 C. Percent phagocytosis is determined by flow cytometry as the percentage of CFSE+/CD14+ cells from the total CD14+ population. All of the clones, Clone 1, Clone 1.1, Clone 1-IgG4 (Clone 1 IgG4 S228P, Clone 1.3), Clone 13, Clone 13.1, and Clone 13-IgG4 (Clone 13 IgG4 S228P, Clone 13.3), except the negative control mAb 2D3, increase phagocytosis of the Jurkat cells by the macrophages irrespective of their affinity to activate Fc receptors.

FIG. 3 shows reversal of TSP1 inhibition of NO-stimulated cGMP production by humanized antibodies of the present disclosure. As described in Example 5, Jurkat JE6.1 cells are incubated overnight in serum-free medium and then incubated with humanized antibodies of the present disclosure or the control chimeric mAb, and with or without TSP1, followed by treatment with or without a NO donor. Cells are lysed 5 minutes later and cGMP is measured. None of the present humanized antibody clones tested, or the control chimeric mAb, has an effect on basal cGMP levels. The control chimeric antibody reverses the TSP1 inhibition, as do humanized Clones 1, 9, 11, and 24 disclosed herein (Cl 1; Cl 9; Cl 11; Cl 24, respectively). PBS: phosphate buffered saline; TSP or TSP: thrombospondin-1; DEA: diethylamine NONOate; chim: chimeric antibody >VxP037-01LC (SEQ ID NO:7))/>VxP037-01HC (SEQ ID NO:57).

FIG. 4 shows reversal of TSP1 inhibition of NO-stimulated cGMP production by humanized antibodies of the present disclosure. As described in Example 5, Jurkat JE6.1 cells are incubated overnight in serum-free medium and then incubated with purified humanized Clone 1 and 13 antibodies, or PBS as the control, and with or without TSP1, followed by treatment with or without a NO donor. Cells are lysed 5 minutes later and cGMP is measured. The humanized antibody clones or PBS have no effect on basal cGMP levels. The humanized Clones 1 and 13 reverse the TSP1 inhibition, while PBS has no effect. PBS: phosphate buffered saline; TSP or TSP1: thrombospondin-1; DEA: diethylamine NONOate.

FIG. 5 shows that treatment of a donor kidney with Clone 1 (Cl 1) at the time of organ harvest is effective in reducing IRI and kidney damage in vivo in a rat kidney transplantation model as assessed by measuring serum creatinine. A syngeneic rat renal transplantation model of IRI with bilaterally nephrectomized recipients is used to evaluate the effect of the anti-CD47 monoclonal antibody Clone 1 on graft function following transplantation. Male Lewis rats weighing 275-300 g are used as both donor and recipient animals. Donor kidneys are flushed with 50 μg of purified Clone 1 or vehicle (phosphate buffered saline, pH 7.2), stored at 4° C. in University of Wisconsin preservation (WU) solution for 6 hours, and then transplanted. Two days following transplantation, kidney function is assessed by measuring serum creatinine. Treatment of donor kidneys with Clone 1 results in improved kidney function compared to controls as measured by a reduction in serum creatinine.

FIG. 6 shows the survival in a DCD (donation after cardiac death) rat kidney transplant model. Male Lewis rats weighing 275-300 g underwent 60 minutes of warm ischemia, prior to flushing the donor kidneys with 50 μg of purified Clone 1.1 or an IgG control mAb. Kidneys are then stored at 4° C. in University of Wisconsin preservation solution (UW) for 6 hours prior to transplantation. In this experiment, survival is monitored over a 7 day time period. All animals that received the IgG control mAb-treated kidney died within 4 days. In contrast, survival is significantly prolonged in the animals that received the Clone1.1 treated kidney, with 30% of the animals surviving for the 7 day duration of the experiment. This shows that with extended periods of warm ischemia, treatment of donor kidneys with Clone 1.1 reduces IRI and increases survival of the recipient.

FIG. 7 shows that purified, humanized antibody Clone 13 (Cl 13) reduces tumor burden in vivo in a syngeneic mouse Acute Promyelocytic Leukemia (APL) model. Murine APL cells (B6APL1) are injected intravenously into C57BL/6 mice randomized into three groups (5-10 mice per group): Group 1: no APL; Group 2: APL with no treatment; Group 3: APL treated with anti-CD47mAb Cl 13. Antibody treatment is initiated on the day of tumor inoculation (day 0), and given in single doses of 10 μg/dose (0.4 mg/kg) by intraperitoneal injection on days 0, 3, and 6. Circulating APL cells (representing the tumor burden) are evaluated at day 25 following tumor inoculation by flow cytometry (CD34+/CD117+ cells). Mice treated with Cl 13 have reduced tumor burden compared to untreated mice at 25 days after tumor inoculation, demonstrating anti-tumor activity of this humanized clone.

FIG. 8 shows that treatment with the CD47 mAb Clone 1.1 significantly reduced tumor growth of the human HepG2 hepatocellular carcinoma model. Male NSG mice are obtained from The Jackson Laboratory (Bar Harbor, Me.) and housed in cages in temperature and light-controlled environments with access to water and food ad libitum. For the heterotopic xenograft model, HepG2-luc2 cells (Perkin Elmer, Waltham, Mass. #134280) are suspended in DMEM containing 25% (v/v), and 1,000,000 cells implanted subcutaneously into the dorsal subcutaneous space of 4- to 8-wk-old NSG mice. After 2 weeks of growth, antibody treatment is begun with twice-weekly intraperitoneal injections of 15 mg/kg of either anti-CD47 antibody Clone 1.1 or an IgG control for 6 weeks. Tumor volumes are calculated twice weekly using (length×width)/0.6. After 6 weeks of treatment, animals are euthanized and tumors were resected, weighed, and fixed in 10% formalin.

FIG. 9 shows binding of Clone 1.1 (IgG1 N297Q) and clone 1.3 (IgG4 S228P) to cynomolgus monkey RBCs by FACS (described previously). Both mAbs exhibited similar binding affinities to cynomolgus RBCs.

FIG. 10 shows that Clones 1.1, 1.2 (IgG4 with mutations of residue S228 to P and of residue L235 to E to reduce Fc effector function, and 1.3 (IgG4 with mutation of residue S228 to P) increase phagocytosis of Jurkat cells by human macrophages while control IgG or the mouse 2D3 antibody that binds to CD47, but does not block the CD47/SIRPalpha interaction, do not.

FIG. 11 shows Clones 1.1, 1.2, 1.3, 13.1, 13.2 and 13.3 all block the interaction of CD47 (expressed on the Jurkat cells) with SIPRalpha while a control antibody that does not bind to CD47 did not block the CD47/SIRPalpha interaction. Absorbance was read at 450 nm.

DETAILED DESCRIPTION OF THE INVENTION

The following detailed description is provided to aid those skilled in the art in practicing the various embodiments of the present disclosure described herein, including all the methods, uses, compositions, etc., described herein. Even so, the following detailed description should not be construed to unduly limit the present disclosure, as modifications and variations in the embodiments herein discussed may be made by those of ordinary skill in the art without departing from the spirit or scope of the present discoveries.

Any feature, or combination of features, described herein is(are) included within the scope of the present disclosure, provided that the features included in any such combination are not mutually inconsistent as will be apparent from the context, this specification, and the knowledge of one of ordinary skill in the art. Additional advantages and aspects of the present disclosure are apparent in the following detailed description and claims.

The contents of all publications, patent applications, patents, and other references mentioned herein are incorporated by reference herein in their entirety. In case of conflict, the present specification, including explanations of terms, will control.

Antibody compounds of the present disclosure can bind to epitopes in the extracellular IgV domain of CD47, inhibiting TSP1 and SIRPalpha binding to CD47 and receptor activation, while inducing little or no agonist activity, and promoting tumor cell phagocytic clearance. In view of these properties, antibody compounds of the present disclosure should be therapeutically useful in treating many forms of IRI and cancers.

The present antibody compounds can also possess a number of other desirable properties, including broad reactivity with CD47 of a wide variety of mammalian species, including that of human, mouse, rat, pig, cynomolgus monkey, and/or dog, i.e., any individual one of these mammalian species, or various combinations thereof, making these antibodies useful in both human and veterinary medicine. This broad reactivity is further advantageous in that it facilitates preclinical studies including, but not limited to, safety and efficacy studies, in a variety of mammalian species, and therefore the development of such antibodies as human and veterinary therapeutics.

Thus, antibody compounds of the present disclosure exhibit the following desirable therapeutic activities:

    • Binding to CD47 of multiple mammalian species;
    • Blocking SIRPalpha and TSP1 binding to CD47;
    • Promoting phagocytosis of cancer cells; and
    • Reversing TSP1 inhibition of nitric oxide signaling
      and are therefore useful in treating ischemia reperfusion injury and cancers.

DEFINITIONS

The following definitions are provided to aid the reader in understanding the various aspects of the present disclosure. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which this disclosure pertains.

A full-length antibody as it exists naturally is an immunoglobulin molecule comprising two heavy (H) chains and two light (L) chains interconnected by disulfide bonds. The amino terminal portion of each chain includes a variable region of about 100-110 or more amino acids primarily responsible for antigen recognition via the complementarity determining regions (CDRs) contained therein. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.

The CDRs are interspersed with regions that are more conserved, termed framework regions (“FR”). Each light chain variable region (LCVR) and heavy chain variable region (HCVR) is composed of 3 CDRs and 4 FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The 3 CDRs of the light chain are referred to as “LCDR1, LCDR2, and LCDR3” and the 3 CDRs of the heavy chain are referred to as “HCDR1, HCDR2, and HCDR3.” The CDRs contain most of the residues which form specific interactions with the antigen. The numbering and positioning of CDR amino acid residues within the LCVR and HCVR regions are in accordance with the well-known Kabat numbering convention. While the light chain CDRs and heavy chain CDRs disclosed herein are numbered 1, 2, and 3, respectively, it is not necessary that they be employed in the corresponding antibody compound light and heavy chain variable regions in that numerical order, i.e., they can be present in any numerical order in a light or heavy chain variable region, respectively.

Light chains are classified as kappa or lambda, and are characterized by a particular constant region as known in the art. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the isotype of an antibody as IgG, IgM, IgA, IgD, or IgE, respectively. IgG antibodies can be further divided into subclasses, e.g., IgG, IgG2, IgG3, IgG4. Each heavy chain type is characterized by a particular constant region with a sequence well known in the art.

The monoclonal antibodies and other antibody compounds useful in the methods and compositions described herein can be any of these isotypes. Furthermore, any of these isotypes can comprise amino acid modifications as follows.

In some embodiments, the antibody constant region is of human IgG1 isotype.

In some embodiments, the human IgG1 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody. For example, this modification can be Asn297→Ala (N297A) or Asn297→Gln (N297Q) (Sazinsky et al. (2008) PNAS 105(51):20167-20172).

In some embodiments, the constant region of the antibody is modified at amino acid Leu234 (Kabat Numbering) to alter Fc receptor interactions. For example, this modification can be Leu234→Ala (L234A).

In some embodiments, the constant region of the antibody is modified at amino acid Leu235 (Kabat Numbering) to alter Fc receptor interactions. For example, this modification can be Leu235→Glu (L235E) or Leu235→Ala (L235A).

In some embodiments, the constant region of the antibody is altered at both amino acid 234 and 235. For example, these modifications can be Leu234→Ala and Leu235→Ala (L234A/L235A) (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest).

In some embodiments, the constant region of the antibody is of human IgG2 isotype.

In some embodiments, the human IgG2 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody. For example, this modification can be Asn297→Ala (N297A) or Asn297→Gln(N297Q).

In some embodiments, the constant region of the antibody is of human IgG3 isotype.

In some embodiments, the human IgG3 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody. For example, this modification can be Asn297→Ala (N297A) or Asn297→Gln(N297Q).

In some embodiments, the human IgG3 constant region is modified at amino acid 435 to extend the half-life. For example, this modification can be Arg435→His (R435H) (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest).

In some embodiments, the constant region of the antibody is of human IgG4 isotype.

In some embodiments, the human IgG4 constant region is modified within the hinge region to prevent or reduce strand exchange. For example, this modification can be Ser228→Pro (S228P) (Angal et al. (1993) Molecular Immunology 30(1):105-108).

In other embodiments, the human IgG4 constant region is modified at amino acid 235 to alter Fc receptor interactions. For example, this can be Leu235→Glu (L235E).

In some embodiments, the human IgG4 constant region is modified within the hinge and at amino acid 228 and in the Fc at amino acid 235. For example, this can be Ser228→Pro and Leu235→Glu (S228P/L235E).

In some embodiments, the human IgG4 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody. For example, this can be Asn297→Ala (N297A). (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest).

In some embodiments, the human IgG constant region is modified to enhance FcRn binding. Examples of Fc mutations that enhance binding to FcRn are Met252→Tyr, Ser254→Thr, Thr256→Glu (M252Y, S254T, and T256E, respectively) (Kabat numbering, Dall'Acqua et al. (2006) J. Biol. Chem. 281(33) 23514-23524), or Met428→Leu and Asn434→Ser (M428L, N434S) (Zalevsky et al. (2010) Nature Biotech. 28(2):157-159). (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest).

In some embodiments, the human IgG constant region is modified to alter antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC), e.g., the amino acid modifications described in Natsume et al. (2008) Cancer Res. 68(10):3863-72; Idusogie et al. (2001) J. Immunol. 166(4):2571-5; Moore et al. (2010) mAbs 2(2):181-189; Lazar et al. (2006) PNAS 103(11):4005-4010; Shields et al. (2001) J. Biol. Chem. 276(9):6591-6604; Stavenhagen et al. (2007) Cancer Res. 67(18):8882-8890; Stavenhagen et al. (2008) Advan. Enzyme Regul. 48:152-164; Alegre et al. (1992) J. Immunol. 148:3461-3468; reviewed in Kaneko and Niwa (2011) Biodrugs 25(1):1-11.

In some embodiments, the human IgG constant region is modified to induce heterodimerization. For example, having an amino acid modification within the CH3 domain at Thr366, which when replaced with a more bulky amino acid, such as Trp (T366W), is able to preferentially pair with a second CH3 domain having amino acid modifications to less bulky amino acids at positions Thr366, Leu368, and Tyr407, e.g., Ser, Ala, and Val, respectively (T366S/L368A/Y407V). Heterodimerization via CH3 modifications can be further stabilized by the introduction of a disulfide bond, for example by changing Ser354 to Cys (S354C) and Tyr349 to Cys (Y349C) on opposite CH3 domains (reviewed in Carter (2001) Journal of Immunological Methods 248:7-15).

As used herein, the term “monoclonal antibody” (mAb) as applied to the present antibody compounds refers to an antibody that is derived from a single copy or clone including, for example, any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. mAbs of the present disclosure preferably exist in a homogeneous or substantially homogeneous population, and can be chimeric or humanized. Complete mAbs contain two heavy chains and two light chains.

“Antigen binding fragments” of such monoclonal antibodies may be desirable for certain applications due to their small size and consequent superior tissue distribution, and include, for example, Fab fragments, Fab′ fragments, F(ab′)2 fragments, Fd fragments, single chain Fv fragments (ScFv), and one-armed antibodies comprising a light chain and a heavy chain. Preferred antigen binding fragments are those that bind to the antigen recognized by the intact antibody. Fc fragments can also be obtained. Monoclonal antibodies and antigen-binding fragments thereof of the present disclosure can be produced, for example, by recombinant technologies, phage display technologies, synthetic technologies, e.g., CDR-grafting, or combinations of such technologies, or other technologies known in the art, including proteolytic digestion of intact antibodies.

“Antibody compounds” refers to mAbs, antigen-binding fragments thereof such as Fabs, etc., and competing antibodies, disclosed herein that specifically bind CD47 of various species, including human, rat, mouse, pig, cynomolgus monkey, and dog CD47, and that exhibit the properties disclosed herein. Thus, the term “mAb” as used herein with respect to antibodies encompassed by the present disclosure includes Fabs and competing antibodies. Additional antibody compounds exhibiting similar functional properties according to the present disclosure can be generated by conventional methods. For example, mice can be immunized with human CD47 or fragments thereof, the resulting antibodies can be recovered and purified, and determination of whether they possess binding and functional properties similar to or the same as the antibody compounds disclosed herein can be assessed by the methods disclosed in Examples 3, 4, and 5, below. Antigen-binding fragments can also be prepared by conventional methods. Methods for producing and purifying antibodies and antigen-binding fragments are well known in the art and can be found, for example, in Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., chapters 5-8 and 15, ISBN 0-87969-314-2.

The phrase “humanized antibodies” refers to monoclonal antibodies and antigen binding fragments thereof, including the antibody compounds disclosed herein, that have binding and functional properties according to the disclosure similar to those disclosed herein, and that have framework and constant regions that are substantially human or fully human surrounding CDRs derived from a non-human antibody. “Framework region” or “framework sequence” refers to any one of framework regions 1 to 4. Humanized antibodies and antigen binding fragments encompassed by the present disclosure include molecules wherein any one or more of framework regions 1 to 4 is substantially or fully human, i.e., wherein any of the possible combinations of individual substantially or fully human framework regions 1 to 4, is present. For example, this includes molecules in which framework region 1 and framework region 2, framework region 1 and framework region 3, framework region 1, 2, and 3, etc., are substantially or fully human. Substantially human frameworks are those that have at least 80% sequence identity to a known human germline framework sequence. Preferably, the substantially human frameworks have at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity, to a framework sequence disclosed herein, or to a known human germline framework sequence.

CDRs encompassed by the present disclosure include not only those specifically disclosed herein, but also CDR sequences having sequence identities of at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to a CDR sequence disclosed herein. Alternatively, CDRs encompassed by the present disclosure include not only those specifically disclosed herein, but also CDR sequences having 1, 2, 3, 4, 5, or 6 amino acid changes at corresponding positions compared to CDR sequences disclosed herein. Such sequence identical, or amino acid modified, CDRs preferably bind to the antigen recognized by the intact antibody.

As used herein, the phrase “sequence identity” means the percentage of identical nucleotide or amino acid residues at corresponding positions in two or more sequences when the sequences are aligned to maximize sequence matching, i.e., taking into account gaps and insertions. Identity can be readily calculated by known methods, including but not limited to those described in: Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988). Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in publicly available computer programs.

Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith & Waterman, by the homology alignment algorithms, by the search for similarity method or, by computerized implementations of these algorithms (GAP, BESTFIT, PASTA, and TFASTA in the GCG Wisconsin Package, available from Accelrys, Inc., San Diego, Calif., United States of America), or by visual inspection. See generally, (Altschul, S. F. et al., J. Mol. Biol. 215: 403-410 (1990) and Altschul et al. Nucl. Acids Res. 25: 3389-3402 (1997)).

One example of an algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in (Altschul, S., et al., NCBI NLM NIH Bethesda, Md. 20894; & Altschul, S., et al., J. Mol. Biol. 215: 403-410 (1990). Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold.

These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always; 0) and N (penalty score for mismatching residues; always; 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value, the cumulative score goes to zero or below due to the accumulation of one or more negative-scoring residue alignments, or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a word length (W) of 11, an expectation (E) of 10, a cutoff of 100, M=5, N=−4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a word length (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix.

In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences. One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P (N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a test nucleic acid sequence is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid sequence to the reference nucleic acid sequence is in one embodiment less than about 0.1, in another embodiment less than about 0.01, and in still another embodiment less than about 0.001.

Fully human frameworks are those that are identical to a known human germline framework sequence. Human framework germline sequences can be obtained from ImMunoGeneTics (IMGT) via their website or from The Immunoglobulin FactsBook by Marie-Paule Lefranc and Gerard Lefranc, Academic Press, 2001, ISBN 012441351. For example, germline light chain frameworks can be selected from the group consisting of: A11, A17, A18, A19, A20, A27, A30, LI, L1I, L12, L2, L5, L15, L6, L8, O12, O2, and 08, and germline heavy chain framework regions can be selected from the group consisting of: VH2-5, VH2-26, VH2-70, VH3-20, VH3-72, VHI-46, VH3-9, VH3-66, VH3-74, VH4-31, VHI-18, VHI-69, VI-13-7, VH3-11, VH3-15, VH3-21, VH3-23, VH3-30, VH3-48, VH4-39, VH4-59, and VH5-5I.

Humanized antibodies in addition to those disclosed herein exhibiting similar functional properties according to the present disclosure can be generated using several different methods. In one approach, the parent antibody compound CDRs are grafted into a human framework that has a high sequence identity with the parent antibody compound framework. The sequence identity of the new framework will generally be at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identical to the sequence of the corresponding framework in the parent antibody compound. In the case of frameworks having fewer than 100 amino acid residues, one, two, three, four, five, six, seven, eight, nine, or ten amino acid residues can be changed. This grafting may result in a reduction in binding affinity compared to that of the parent antibody. If this is the case, the framework can be back-mutated to the parent framework at certain positions based on specific criteria disclosed by Queen et al. (1991) Proc. Natl. Acad. Sci. USA 88:2869. Additional references describing methods useful in humanizing mouse antibodies include U.S. Pat. Nos. 4,816,397; 5,225,539; and 5,693,761; computer programs ABMOD and ENCAD as described in Levitt (1983) J. Mol. Biol. 168:595-620; and the method of Winter and co-workers (Jones et al. (1986) Nature 321:522-525; Riechmann et al. (1988) Nature 332:323-327; and Verhoeyen et al. (1988) Science 239:1534-1536.

The identification of residues to consider for back-mutation can be carried out as follows: When an amino acid falls under the following category, the framework amino acid of the human germ-line sequence that is being used (the “acceptor framework”) is replaced by a framework amino acid from a framework of the parent antibody compound (the “donor framework”): (a) the amino acid in the human framework region of the acceptor framework is unusual for human frameworks at that position, whereas the corresponding amino acid in the donor immunoglobulin is typical for human frameworks at that position; (b) the position of the amino acid is immediately adjacent to one of the CDRs; or (c) any side chain atom of a framework amino acid is within about 5-6 angstroms (center-to-center) of any atom of a CDR amino acid in a three dimensional immunoglobulin model.

When each of the amino acids in the human framework region of the acceptor framework and a corresponding amino acid in the donor framework is generally unusual for human frameworks at that position, such amino acid can be replaced by an amino acid typical for human frameworks at that position. This back-mutation criterion enables one to recover the activity of the parent antibody compound.

Another approach to generating human engineered antibodies exhibiting similar functional properties to the antibody compounds disclosed herein involves randomly mutating amino acids within the grafted CDRs without changing the framework, and screening the resultant molecules for binding affinity and other functional properties that are as good as or better than those of the parent antibody compounds. Single mutations can also be introduced at each amino acid position within each CDR, followed by assessing the effects of such mutations on binding affinity and other functional properties.

Single mutations producing improved properties can be combined to assess their effects in combination with one another.

Further, a combination of both of the foregoing approaches is possible. After CDR grafting, one can back-mutate specific framework regions in addition to introducing amino acid changes in the CDRs. This methodology is described in Wu et al. (1999) J. Mol. Biol. 294:151-162.

The method described in Example 1 below can also be employed.

Applying the teachings of the present disclosure, a person skilled in the art can use common techniques, e.g., site-directed mutagenesis, to substitute amino acids within the presently disclosed CDR and framework sequences and thereby generate further variable region amino acid sequences derived from the present sequences. Up to all naturally occurring amino acids can be introduced at a specific substitution site, including conservative amino acid substitutions as are well known to those of ordinary skill in the art. The methods disclosed herein can then be used to screen these additional variable region amino acid sequences to identify sequences having the indicated in vitro and/or in vivo functions. In this way, further sequences suitable for preparing human engineered antibodies and antigen-binding portions thereof in accordance with the present disclosure can be identified. In some embodiments, amino acid substitution within the frameworks can include one, two, three, four, five, six, seven, eight, nine, or ten positions within any one or more of the 4 light chain and/or heavy chain framework regions disclosed herein. In some embodiments, amino acid substitution within the CDRs is restricted to one, two, three, four, or five positions within any one or more of the 3 light chain and/or heavy chain CDRs. Combinations of the various changes within these framework regions and CDRs described above are also possible.

That the functional properties of the antibody compounds generated by introducing the amino acid modifications discussed above conform to, and are comparable to, those exhibited by the specific molecules disclosed herein can be confirmed by the methods disclosed in the Examples below.

The terms “specifically binds”, “bind specifically”, “specific binding”, and the like as applied to the present antibody compounds refer to the ability of a specific binding agent (such as an antibody) to bind to a target molecular species in preference to binding to other molecular species with which the specific binding agent and target molecular species are admixed. A specific binding agent is said specifically to recognize a target molecular species when it can bind specifically to that target.

“Binding affinity” is a term that refers to the strength of binding of one molecule to another at a site on the molecule. If a particular molecule will bind to or specifically associate with another particular molecule, these two molecules are said to exhibit binding affinity for each other. Binding affinity is related to the association constant and dissociation constant for a pair of molecules, but it is not critical to the methods herein that these constants be measured or determined. Rather, affinities as used herein to describe interactions between molecules of the described methods are generally apparent affinities (unless otherwise specified) observed in empirical studies, which can be used to compare the relative strength with which one molecule (e.g., an antibody or other specific binding partner) will bind two other molecules (e.g., two versions or variants of a peptide). The concepts of binding affinity, association constant, and dissociation constant are well known.

The term “epitope” refers to a specific arrangement of amino acids located on a peptide or protein to which an antibody or antibody fragment binds. Epitopes often consist of a chemically active surface grouping of molecules such as amino acids or sugar side chains, and have specific three dimensional structural characteristics as well as specific charge characteristics. Epitopes can be linear, i.e., involving binding to a single sequence of amino acids, or conformational, i.e., involving binding to two or more sequences of amino acids in various regions of the antigen that may not necessarily be contiguous in the linear sequence.

Monoclonal antibodies or antigen-binding fragments thereof encompassed by the present disclosure that “compete” with the molecules disclosed herein are those that bind human CD47 at site(s) that are identical to, or overlapping with, the site(s) at which the present molecules bind. Competing monoclonal antibodies or antigen-binding fragments thereof can be identified, for example, via an antibody competition assay. For example, a sample of purified or partially purified human CD47 extracellular domain can be bound to a solid support. Then, an antibody compound, or antigen binding fragment thereof, of the present disclosure and a monoclonal antibody or antigen-binding fragment thereof suspected of being able to compete with such disclosure antibody compound are added. One of the two molecules is labeled. If the labeled compound and the unlabeled compound bind to separate and discrete sites on CD47, the labeled compound will bind to the same level whether or not the suspected competing compound is present. However, if the sites of interaction are identical or overlapping, the unlabeled compound will compete, and the amount of labeled compound bound to the antigen will be lowered. If the unlabeled compound is present in excess, very little, if any, labeled compound will bind. For purposes of the present disclosure, competing monoclonal antibodies or antigen-binding fragments thereof are those that decrease the binding of the present antibody compounds to CD47 by about 50%, about 60%, about 70%, about 80%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99%. Details of procedures for carrying out such competition assays are well known in the art and can be found, for example, in Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., pages 567-569, ISBN 0-87969-314-2. Such assays can be made quantitative by using purified antibodies. A standard curve is established by titrating one antibody against itself, i.e., the same antibody is used for both the label and the competitor. The capacity of an unlabeled competing monoclonal antibody or antigen-binding fragment thereof to inhibit the binding of the labeled molecule to the plate is titrated. The results are plotted, and the concentrations necessary to achieve the desired degree of binding inhibition are compared.

Whether monoclonal antibodies or antigen-binding fragments thereof that compete with antibody compounds of the present disclosure in such competition assays possess the same or similar functional properties of the present antibody compounds can be determined via these methods in conjunction with the methods described in Examples 3-5, below. In various embodiments, competing antibodies for use in the therapeutic methods encompassed herein possess biological activities as described herein in the range of from about 50% to about 100% or about 125%, or more, compared to that of the antibody compounds disclosed herein. In some embodiments, competing antibodies possess about 50%, about 60%, about 70%, about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or identical biological activity compared to that of the antibody compounds disclosed herein as determined by the methods disclosed in the Examples presented below.

The term “treating” (or “treat” or “treatment”) means slowing, interrupting, arresting, controlling, stopping, reducing, or reversing the progression or severity of a sign, symptom, disorder, condition, or disease, but does not necessarily involve a total elimination of all disease-related signs, symptoms, conditions, or disorders. The term “treating” and the like refer to a therapeutic intervention that ameliorates a sign, symptom, etc., of a disease or pathological condition after it has begun to develop.

Acute events and chronic conditions can be treated. In an acute event, an antibody or antigen binding fragment thereof is administered at the onset of a symptom, disorder, condition, disease, or procedure, and is discontinued when the acute event ends, or in the case of organ transplantation to the organ, at the time of organ harvest and/or to the transplant recipient at the time of organ transplantation. In contrast, a chronic symptom, disorder, condition, or disease is treated over a more protracted time frame.

The term “effective amount” refers to the amount or dose of an antibody compound of the present disclosure which, upon single or multiple dose administration to a patient or organ, provides the desired treatment or prevention.

The precise effective amount for any particular subject will depend upon their size and health, the nature and extent of their condition, and the therapeutics or combination of therapeutics selected for administration. The effective amount for a given patient is determined by routine experimentation and is within the judgment of a clinician. In some embodiments, an effective dose will generally be from about 0.01 mg/kg to about 50 mg/kg, or about 0.05 mg/kg to about 10 mg/kg of the compositions of the present disclosure in the individual to which it is administered. Therapeutically effective amounts of the present antibody compounds can also comprise an amount in the range of from about 0.1 mg/kg to about 150 mg/kg, from about 0.1 mg/kg to about 100 mg/kg, or from about 0.1 mg/kg to about 50 mg/kg per single dose administered to a harvested organ or to a patient. Known antibody-based pharmaceuticals provide guidance in this respect. For example, Herceptin™ is administered by intravenous infusion of a 21 mg/ml solution, with an initial loading dose of 4 mg/kg body weight and a weekly maintenance dose of 2 mg/kg body weight; Rituxan™ is administered weekly at 375 mg/m2; etc.

A therapeutically effective amount for any individual patient can be determined by the health care provider by monitoring the effect of the antibody compounds on a biomarker, such as serum biomarkers of injury of the treated organ, including but not limited to liver, kidney, lung, intestine, pancreas and heart, changes in pulmonary artery pressures, cell surface CD47 expression in tumor or non-tumor tissues, tumor regression, circulating tumor cells or tumor stem cells, etc. Analysis of the data obtained by these methods permits modification of the treatment regimen during therapy so that optimal amounts of antibody compounds of the present disclosure, whether employed alone or in combination with one another, or in combination with another therapeutic agent, or both, are administered, and so that the duration of treatment can be determined as well. In this way, the dosing/treatment regimen can be modified over the course of therapy so that the lowest amounts of antibody compounds used alone or in combination that exhibit satisfactory efficacy are administered, and so that administration of such compounds is continued only so long as is necessary to successfully treat the patient. Known antibody-based pharmaceuticals provide guidance relating to frequency of administration e.g., whether a pharmaceutical should be delivered daily, weekly, monthly, etc. Frequency and dosage may also depend on the severity of symptoms.

The antibody compounds of the present disclosure can be used as medicaments in human and veterinary medicine, administered by a variety of routes. Veterinary applications include the treatment of companion/pet animals, such as cats and dogs; working animals, such as guide or service dogs, and horses; sport animals, such as horses and dogs; zoo animals, such as primates, cats such as lions and tigers, bears, etc.; and other valuable animals kept in captivity.

Antibody compounds can be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intraperitoneal, intrathecal, intraventricular, transdermal, transcutaneous, topical, subcutaneous, intranasal, enteral, sublingual, intravaginal, intravesiciular or rectal routes. Hypo sprays may also be used to administer the pharmaceutical compositions. Typically, the therapeutic compositions can be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.

Direct delivery of the compositions can generally be accomplished by injection, subcutaneously, intraperitoneally, intravenously, or intramuscularly, or delivered to the interstitial space of a tissue. The compositions can also be administered into a lesion such as a tumor. Dosage treatment may be a single dose schedule or a multiple dose schedule.

In some embodiments, such compositions are formulated for parenteral administration by, for example, intravenous, intramuscular, subcutaneous, etc., administration by infusion, injection, implantation, etc., as is well known in the art. Examples include bolus injection or continuous infusion. Intratumoral administration, for example by injection, is also contemplated.

Such pharmaceutical compositions can be prepared by methods well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy, 21st Edition (2005), Lippincott Williams & Wilkins, Philadelphia, Pa., and comprise one or more antibody compounds disclosed herein, and a pharmaceutically or veterinary acceptable, e.g., physiologically acceptable, carrier, diluent, or excipient.

Combination Therapies

The therapeutic methods encompassed herein include the use of the antibodies disclosed herein alone, and/or in combinations with one another, and/or with antigen-binding fragments thereof, and/or with competing antibodies exhibiting appropriate biological/therapeutic activity, as well, i.e., all possible combinations of these antibody compounds.

In addition, the present therapeutic methods also encompass the use of these antibodies, antigen-binding fragments thereof, competing antibodies, etc., and combinations thereof further in combination with: (1) any one or more of the nitric oxide donor, precursor, or nitric oxide generating topical agents, and/or agents that activate soluble guanylyl cyclase, and/or agents that inhibit cyclic nucleotide phosphodiesterases disclosed herein, or (2) any one or more anti-tumor therapeutic treatments selected from surgery, radiation, anti-tumor or anti-neoplastic agents, and combinations of any of these, or (3) equivalents of any of the foregoing of (1) or (2) as would be apparent to one of ordinary skill in the art, in appropriate combination(s) to achieve the desired therapeutic treatment effect for the particular indication.

Combinations of Antibody Compounds

It should be noted that in all of the therapeutic methods disclosed and claimed herein, the monoclonal antibodies or antigen binding fragments thereof, and monoclonal antibodies or antigen binding fragments thereof that compete with these monoclonal antibodies or antigen binding fragments thereof of the present disclosure that bind to CD47, can be used alone, or in any appropriate combinations with one another, to achieve the greatest treatment efficacy.

Further Therapeutic Combinations to Treat IRI-Related Indications

In addition to administering the combinations of antibody compounds as described immediately above, the methods of the present disclosure, for example those related to treatment of IRI-related indications, can further comprise administering to a patient in need thereof an effective amount of a nitric oxide donor, precursor, or both; a nitric oxide generating topical agent; an agent that activates soluble guanylyl cyclase; an agent that inhibits cyclic nucleotide phosphodiesterases; or any combination of any of the foregoing.

In these methods, the nitric oxide donor or precursor can be selected from NO gas, isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholinosydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine.

The agent that activates soluble guanylyl cyclase can be a non-NO (nitric oxide)-based chemical activator of soluble guanylyl cyclase that increases cGMP levels in vascular cells. Such agents bind soluble guanylyl cyclase in a region other than the NO binding motif, and activate the enzyme regardless of local NO or reactive oxygen stress (ROS). Non-limiting examples of chemical activators of soluble guanylyl cyclase include organic nitrates (Artz et al. (2002) J. Biol. Chem. 277:18253-18256); protoporphyrin IX (Ignarro et al. (1982) Proc. Natl. Acad. Sci. USA 79:2870-2873); YC-1 (Ko et al. (1994) Blood 84:4226-4233); BAY 41-2272 and BAY 41-8543 (Stasch et al. (2001 Nature 410 (6825): 212-5), CMF-1571, and A-350619 (reviewed in Evgenov et al. (2006) Nat. Rev. Drug. Discov. 5:755-768); BAY 58-2667 (Cinaciguat; Frey et al. (2008) Journal of Clinical Pharmacology 48 (12): 1400-10); BAY 63-2521 (Riociguat; Mittendorf et al. (2009) Chemmedchem 4 (5): 853-65). Additional soluble guanylyl cyclase activators are disclosed in Stasch et al. (2011) Circulation 123:2263-2273; Derbyshire and Marletta (2012) Ann. Rev. Biochem. 81:533-559, and Nossaman et al. (2012) Critical Care Research and Practice, Volume 2012, Article ID 290805, pages 1-12.

The agent that inhibits cyclic nucleotide phosphodiesterases can be selected from sildenafil, tadalafil, vardenafil, udenafil, and avanafil.

Further Therapeutic Combinations to Treat Cancer Indications

In addition to the foregoing, the methods of the present disclosure, for example those related to treatment of cancer indications, can further comprise treating the patient via surgery, radiation, and/or administering to a patient in need thereof an effective amount of a chemical small molecule or biologic drug including, but not limited to, a peptide, polypeptide, protein, nucleic acid therapeutic, etc., conventionally used, or currently being developed, to treat cancer. This includes antibodies and antigen-binding fragments other than those disclosed herein, cytokines, antisense oligonucleotides, siRNAs, miRNAs, etc.

As is well known to those of ordinary skill in the art, combination therapies are often employed in cancer treatment as single-agent therapies or procedures may not be sufficient to treat or cure the disease or condition. Conventional cancer treatments often involve surgery, radiation treatment, the administration of a combination of cytotoxic drugs to achieve additive or synergistic effects, and combinations of any or all of these approaches. Especially useful chemotherapeutic and biologic therapy combinations employ drugs that work via different mechanisms of action, increasing cancer cell control or killing, increasing the ability of the immune system to control cancer cell growth, reducing the likelihood of drug resistance during therapy, and minimizing possible overlapping toxicities by permitting the use of reduced doses of individual drugs.

Classes of conventional anti-tumor/anti-neoplastic agents useful in the combination therapies encompassed by the present methods are disclosed, for example, in Goodman & Gilman's The Pharmacological Basis of Therapeutics, Twelfth Edition (2010) L. L. Brunton, B. A. Chabner, and B. C. Knollmann Eds., Section VIII, “Chemotherapy of Neoplastic Diseases”, Chapters 60-63, pp. 1665-1770, McGraw-Hill, NY, and include, for example, alkylating agents; antimetabolites; natural products; a variety of miscellaneous agents; hormones and antagonists; and monoclonal antibodies.

Antibody and small molecule drugs that increase the immune response to cancer by modulating co-stimulatory or inhibitory interactions that influence the T cell response to tumor antigens, including inhibitors of immune checkpoints and modulators of co-stimulatory molecules, are also of particular interest in the context of the combination therapeutic methods encompassed herein and include, but are not limited to, other anti-CD47 antibodies. These agents that are involved in the immune response include IL-10 (Interleukin-10, human cytokin synthesis inhibitory factor, CSIF) and Galectins.

Administration of therapeutic agents that bind to the CD47 protein, for example, antibodies or small molecules that bind to CD47 and prevent interaction between CD47 and SIRPalpha, are administered to a patient, increasing the clearance of cancer cells via phagocytosis. The therapeutic agent that binds to the CD47 protein is combined with a therapeutic agent such as an antibody, a chemical small molecule or biologic drug disclosed herein, directed against one or more additional cellular targets of CD70 (Cluster of Differentiation 70), CD200 (OX-2 membrane glycoprotein, Cluster of Differentiation 200), CD154 (Cluster of Differentiation 154, CD40L, CD40 ligand, Cluster of Differentiation 40 ligand), CD223 (Lymphocyte-activation gene 3, LAG3, Cluster of Differentiation 223), KIR (Killer-cell immunoglobulin-like receptors), GITR (TNFRSF18, glucocorticoid-induced TNFR-related protein, activation-inducible TNFR family receptor, AITR, Tumor necrosis factor receptor superfamily member 18), CD28 (Cluster of Differentiation 28), CD40 (Cluster of Differentiation 40, Bp50, CDW40, TNFRSF5, Tumor necrosis factor receptor superfamily member 5, p50), CD86 (B7-2, Cluster of Differentiation 86), CD160 (Cluster of Differentiation 160, BY55, NK1, NK28), CD258 (LIGHT, Cluster of Differentiation 258, Tumor necrosis factor ligand superfamily member 14, TNFSF14, HVEML, HVEM ligand, herpesvirus entry mediator ligand, LTg), CD270 (HVEM, Tumor necrosis factor receptor superfamily member 14, herpesvirus entry mediator, Cluster of Differentiation 270, LIGHTR, HVEA), CD275 (ICOSL, ICOS ligand, Inducible T-cell Costimulator ligand, Cluster of Differentiation 275), CD276 (B7-H3, B7 homolog 3, Cluster of Differentiation 276), OX40L (OX40 Ligand), B7-H4 (B7 homolog 4, VTCN1, V-set domain-containing T-cell activation inhibitor 1), GITRL (Glucocorticoid-induced tumor necrosis factor receptor-ligand, glucocorticoid-induced TNFR-ligand), 4-1BBL (4-1BB ligand), CD3 (Cluster of Differentiation 3, T3D), CD25 (IL2Rα, Cluster of Differentiation 25, Interleukin-2 Receptor α chain, IL-2 Receptor α chain), CD48 (Cluster of Differentiation 48, B-lymphocyte activation marker, BLAST-1, signaling lymphocytic activation molecule 2, SLAMF2), CD66a (Ceacam-1, Carcinoembryonic antigen-related cell adhesion molecule 1, biliary glycoprotein, BGP, BGP1, BGPI, Cluster of Differentiation 66a), CD80 (B7-1, Cluster of Differentiation 80), CD94 (Cluster of Differentiation 94), NKG2A (Natural killer group 2A, killer cell lectin-like receptor subfamily D member 1, KLRD1), CD96 (Cluster of Differentiation 96, TActILE, T cell activation increased late expression), CD112 (PVRL2, nectin, Poliovirus receptor-related 2, herpesvirus entry mediator B, HVEB, nectin-2, Cluster of Differentiation 112), CD115 (CSF1R, Colony stimulating factor 1 receptor, macrophage colony-stimulating factor receptor, M-CSFR, Cluster of Differentiation 115), CD205 (DEC-205, LY75, Lymphocyte antigen 75, Cluster of Differentiation 205), CD226 (DNAM1, Cluster of Differentiation 226, DNAX Accessory Molecule-1, PTA1, platelet and T cell activation antigen 1), CD244 (Cluster of Differentiation 244, Natural killer cell receptor 2B4), CD262 (DR5, TrailR2, TRAIL-R2, Tumor necrosis factor receptor superfamily member 10b, TNFRSF10B, Cluster of Differentiation 262, KILLER, TRICK2, TRICKB, ZTNFR9, TRICK2A, TRICK2B), CD284 (Toll-like Receptor-4, TLR4, Cluster of Differentiation 284), CD288 (Toll-like Receptor-8, TLR8, Cluster of Differentiation 288), TNFSF15 (Tumor necrosis factor superfamily member 15, Vascular endothelial growth inhibitor, VEGI, TL1A), TDO2 (Tryptophan 2,3-dioxygenase, TPH2, TRPO), IGF-1R (Type 1 Insulin-like Growth Factor), GD2 (Disialoganglioside 2), TMIGD2 (Transmembrane and immunoglobulin domain-containing protein 2), RGMB (RGM domain family, member B), VISTA (V-domain immunoglobulin-containing suppressor of T-cell activation, B7-H5, B7 homolog 5), BTNL2 (Butyrophilin-like protein 2), Btn (Butyrophilin family), TIGIT (T cell immunoreceptor with Ig and ITIM domains, Vstm3, WUCAM), Siglecs (Sialic acid binding Ig-like lectins), Neurophilin, VEGFR (Vascular endothelial growth factor receptor), ILT family (LIRs, immunoglobulin-like transcript family, leukocyte immunoglobulin-like receptors), NKG families (Natural killer group families, C-type lectin transmembrane receptors), MICA (MHC class I polypeptide-related sequence A), TGFβ (Transforming growth factor β), STING pathway (Stimulator of interferon gene pathway), Arginase (Arginine amidinase, canavanase, L-arginase, arginine transamidinase), EGFRvIII (Epidermal growth factor receptor variant III), and HHLA2 (B7-H7, B7y, HERV-H LTR-associating protein 2, B7 homolog 7), inhibitors of PD-1 (Programmed cell death protein 1, PD-1, CD279, Cluster of Differentiation 279), PD-L1 (B7-H1, B7 homolog 1, Programmed death-ligand 1, CD274, cluster of Differentiation 274), PD-L2 (B7-DC, Programmed cell death 1 ligand 2, PDCDiLG2, CD273, Cluster of Differentiation 273), CTLA-4 (Cytotoxic T-lymphocyte-associated protein 4, CD152, Cluster of Differentiation 152), BTLA (B- and T-lymphocyte attenuator, CD272, Cluster of Differentiation 272), Indoleamine 2,3-dioxygenase (IDO, IDO1), TIM3 (HAVCR2, Hepatitis A virus cellular receptor 2, T cell immunoglobulin mucin-3, KIM-3, Kidney injury molecule 3, TIMD-3, T cell immunoglobulin mucin-domain 3), A2A adenosine receptor (ADO receptor), CD39 (ectonucleoside triphosphate diphosphohydrolase-1, Cluster of Differentiation 39, ENTPD 1), and CD73 (Ecto-5′-nucleotidase, 5′-nucleotidase, 5′-NT, Cluster of Differentiation 73), including antibodies and small molecules, and agonists of CD27 (Cluster of Differentiation 27), ICOS (CD278, Cluster of Differentiation 278, Inducible T-cell Co-stimulator), CD137 (4-1BB, Cluster of Differentiation 137, tumor necrosis factor receptor superfamily member 9, TNFRSF9), OX40 (CD134, Cluster of Differentiation 134), and TNFSF25 (Tumor necrosis factor receptor superfamily member 25), including small molecules and antibodies, are also specifically contemplated herein.

YERVOY® (ipilimumab; Bristol-Meyers Squibb) is an example of an approved anti-CTLA-4 antibody.

KEYTRUDA® (pembrolizumab; Merck) and OPDIVO® (nivolumab; Bristol-Meyers Squibb Company) are examples of approved anti-PD-1 antibodies.

Another useful class of compounds for the combination therapies contemplated herein includes modulators of SIRPalpha/CD47 binding such as antibodies to SIRPalpha, as well as soluble protein fragments of this ligand, or CD47 itself, acting as “decoy” molecules inhibiting binding of, or interfering with binding of, SIRPalpha to CD47.

The present disclosure encompasses therapeutic methods comprising not only the administration of any of the individual monoclonal antibodies, antigen binding fragments thereof, or competing antibodies disclosed herein with any one or more of the molecules discussed immediately above, but also combinations of the disclosed monoclonal antibodies, antigen-binding fragments thereof, and competing antibodies in combinations with any one or more of the molecules discussed immediately above, i.e., all possible permutations and combinations of the presently disclosed molecules.

The term “tumor” refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms “cancer”, “cancerous”, and “tumor” are not mutually exclusive as used herein.

The terms “cancer” and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by aberrant cell growth/proliferation. Examples of cancers include, but are not limited to, carcinomas, lymphomas, blastomas, sarcomas, and leukemias.

The term “susceptible cancer” as used herein refers to a cancer, cells of which express CD47 and that are responsive to treatment with an antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of the present disclosure. Exemplary susceptible cancers include, but are not limited to, leukemias, including systemic mastocytosis, acute lymphocytic (lymphoblastic) leukemia (ALL), T cell—ALL, acute myeloid leukemia (AML), myelogenous leukemia, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), chronic myeloid leukemia (CML), myeloproliferative disorder/neoplasm, myelodysplastic syndrome, monocytic cell leukemia, and plasma cell leukemia; lymphomas, including histiocytic lymphoma and T cell lymphoma, B cell lymphomas, including Hodgkin's lymphoma and non-Hodgkin's lymphoma, such as low grade/follicular non-Hodgkin's lymphoma (NHL), cell lymphoma (FCC), mantle cell lymphoma (MCL), diffuse large cell lymphoma (DLCL), small lymphocytic (SL) NHL, intermediate grade/follicular NHL, intermediate grade diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, and Waldenstrom's Macroglobulinemia, ovarian cancer, breast cancer, endometrial cancer, colon cancer (colorectal cancer), rectal cancer, bladder cancer, lung cancer (non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung), bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma (liver cancer, hepatoma), gall bladder cancer, bile duct cancer, esophageal cancer, renal cell carcinoma, thyroid cancer, squamous cell carcinoma of the head and neck (head and neck cancer), testicular cancer, cancer of the endocrine gland, cancer of the adrenal gland, cancer of the pituitary gland, cancer of the skin, cancer of soft tissues, cancer of blood vessels, cancer of brain, cancer of nerves, cancer of eyes, cancer of meninges, cancer of oropharynx, cancer of hypopharynx, cancer of cervix, and cancer of uterus, glioblastoma, meduloblastoma, astrocytoma, glioma, meningioma, gastrinoma, neuroblastoma, melanoma, myelodysplastic syndrome, and sarcomas including, but not limited to, osteosarcoma, Ewing sarcoma, leiomyosarcoma, synovial sarcoma, alveolar soft part sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chrondrosarcoma.

“Phagocytosis” of cancer cells refers to the engulfment and digestion of such cells by macrophages, and the eventual digestion or degradation of these cancer cells and their release extracellularly, or intracellularly, to undergo further processing. Anti-CD47 monoclonal antibodies that block SIRPalpha binding to CD47, the “don't eat me” signal which is highly expressed on cancer cells as compared with normal cells, induce macrophage phagocytosis of cancer cells. SIRPalpha binding to CD47 on cancer cells would otherwise allow these cells to escape macrophage phagocytosis.

The terms “promote”, “promoting”, and the like are used herein synonymously with “increase”, “increasing”, etc.

“Ischemia” refers to a vascular phenomenon in which a decrease in the blood supply to a bodily organ, tissue, or part is caused, for instance, by constriction or obstruction of one or more blood vessels. Ischemia sometimes results from vasoconstriction or thrombosis or embolism. Ischemia can lead to direct ischemic injury, tissue damage due to cell death caused by reduced oxygen supply. Ischemia can occur acutely, as during surgery, or from trauma to tissue incurred in accidents, injuries and war settings, or following harvest of organs intended for subsequent transplantation, for example. It can also occur sub-acutely, as found in atherosclerotic peripheral vascular disease, where progressive narrowing of blood vessels leads to inadequate blood flow to tissues and organs.

When a tissue is subjected to ischemia, a sequence of chemical events is initiated that may ultimately lead to cellular dysfunction and necrosis. If ischemia is ended by the restoration of blood flow, a second series of injurious events ensue, producing additional injury. Thus, whenever there is a transient decrease or interruption of blood flow in a subject, the resultant injury involves two components—the direct injury occurring during the ischemic interval, and the indirect or reperfusion injury that follows.

“Ischemic stroke” can be caused by several different kinds of diseases. The most common problem is narrowing of the arteries in the neck or head. This is most often caused by atherosclerosis, or gradual cholesterol deposition. If the arteries become too narrow, blood cells may collect in them and form blood clots (thrombi). These blood clots can block the artery where they are formed (thrombosis), or can dislodge and become trapped in arteries closer to the brain (embolism). Cerebral stroke can occur when atherosclerotic plaque separates away partially from the vessel wall and occludes the flow of blood through the blood vessel.

“Reperfusion” refers to restoration of blood flow to tissue that is ischemic, due to decrease in blood flow. Reperfusion is a procedure for treating infarction or other ischemia, by enabling viable ischemic tissue to recover, thus limiting further necrosis. However, reperfusion can itself further damage the ischemic tissue, causing reperfusion injury.

In addition to the immediate injury that occurs during deprivation of blood flow, “ischemic/reperfusion injury” involves tissue injury that occurs after blood flow is restored. Current understanding is that much of this injury is caused by chemical products, free radicals, and active biological agents released by the ischemic tissues.

“Nitric oxide donor, precursor, or nitric oxide generating topical agent” refers to a compound or agent that either delivers NO, or that can be converted to NO through enzymatic or non-enzymatic processes. Examples include, but are not limited to, NO gas, isosorbide dinitrite, nitrite, nitroprusside, nitroglycerin, 3-Morpholinosydnonimine (SIN-1), S-nitroso-N-acetyl-penicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine.

“Soluble guanylyl cyclase (sGC)” is the receptor for nitric oxide in vascular smooth muscle. In the cardiovascular system, nitric oxide is endogenously generated by endothelial nitric oxide synthase from L-arginine, and activates soluble guanylyl cyclase in adjacent vascular smooth muscle cells to increase cGMP levels, inducing vascular relaxation. Nitric oxide binds to the normally reduced heme moiety of soluble guanylyl cyclase, and increases the formation of cGMP from GTP, leading to a decrease in intracellular calcium, vasodilation, and anti-inflammatory effects. Oxidation of the heme iron on sGC decreases responsiveness of the enzyme to nitric oxide, and promotes vasoconstriction. The nitric oxide-sGC-cGMP pathway therefore plays an important role in cardiovascular diseases. Nitrogen-containing compounds such as sodium azide, sodium nitrite, hydroxylamine, nitroglycerin, and sodium nitroprusside have been shown to stimulate sGC, causing an increase in cGMP, and vascular relaxation. In contrast to stimulators of sGC, which bind to reduced sGC, activators of sGC activate the oxidized or heme-deficient sGC enzyme that is not responsive to nitric oxide, i.e., they stimulate sGC independent of redox state. While stimulators of sGC can enhance the sensitivity of reduced sGC to nitric oxide, activators of sGC can increase sGC enzyme activity even when the enzyme is oxidized and is therefore less, or unresponsive, to nitric oxide. Thus, sGC activators are non-nitric oxide based. Note the reviews of Nossaman et al. (2012) Critical Care Research and Practice, Volume 2012, article 290805, and Derbyshire and Marletta (2012) Ann. Rev. Biochem. 81:533-559.

“An agent that activates soluble guanylyl cyclase” refers, for example, to organic nitrates (Artz et al. (2002) J. Biol. Chem. 277:18253-18256); protoporphyrin IX (Ignarro et al. (1982) Proc. Natl. Acad. Sci. USA 79:2870-2873); YC-1 (Ko et al. (1994) Blood 84:4226-4233); BAY 41-2272 and BAY 41-8543 (Stasch et al. (2001 Nature 410 (6825): 212-5), CMF-1571, and A-350619 (reviewed in Evgenov et al. (2006) Nat. Rev. Drug. Discov. 5:755-768); BAY 58-2667 (Cinaciguat; Frey et al. (2008) Journal of Clinical Pharmacology 48 (12): 1400-10); BAY 63-2521 (Riociguat; Mittendorf et al. (2009) Chemmedchem 4 (5): 853-65). Additional soluble guanylyl cyclase activators are disclosed in Stasch et al. (2011) Circulation 123:2263-2273; Derbyshire and Marletta (2012) Ann. Rev. Biochem. 81:533-559, and Nossaman et al. (2012) Critical Care Research and Practice, Volume 2012, Article ID 290805, pages 1-12.

Examples of “an agent that inhibits cyclic nucleotide phosphodiesterases” include sildenafil, tadalafil, vardenafil, udenafil, and avanafil.

The singular terms “a”, “an”, and “the” include plural referents unless context clearly indicates otherwise. Similarly, the word “or” is intended to include “and” unless the context clearly indicates otherwise. Hence, comprising A or B means including A, or B, or A and B.

The term “about” as used herein is a flexible word with a meaning similar to “approximately” or “nearly”. The term “about” indicates that exactitude is not claimed, but rather a contemplated variation. Thus, as used herein, the term “about” means within 1 or 2 standard deviations from the specifically recited value, or ±a range of up to 20%, up to 15%, up to 10%, up to 9%, up to 8%, up to 7%, up to 6%, up to 5%, up to 4%, up to 3%, up to 2%, or up to 1% compared to the specifically recited value.

The term “comprising” as used in a claim herein is open-ended, and means that the claim must have all the features specifically recited therein, but that there is no bar on additional features that are not recited being present as well. The term “comprising” leaves the claim open for the inclusion of unspecified ingredients even in major amounts. The term “consisting essentially of” in a claim means that the invention necessarily includes the listed ingredients, and is open to unlisted ingredients that do not materially affect the basic and novel properties of the invention. A “consisting essentially of” claim occupies a middle ground between closed claims that are written in a closed “consisting of” format and fully open claims that are drafted in a “comprising’ format”. These terms can be used interchangeably herein if, and when, this may become necessary.

Furthermore, the use of the term “including”, as well as other related forms, such as “includes” and “included”, is not limiting.

CD47 and Ischemia-Reperfusion Injury (IRI)

Following periods of tissue ischemia, the initiation of blood flow causes damage referred to as “ischemia-reperfusion injury” or IRI. IRI contributes to poor outcomes in many surgical procedures where IRI occurs due to the necessity to stop blood flow for a period of time, in many forms/causes of trauma in which blood flow is interrupted and later restored by therapeutic intervention and in procedures required for organ transplantation, cardio/pulmonary bypass procedures, reattachment of severed body parts, reconstructive and cosmetic surgeries and other situations involving stopping and restarting blood flow. Ischemia itself causes many physiological changes that, by themselves would eventually lead to cell and tissue necrosis and death. Reperfusion poses its own set of damaging events including generation of reactive oxygen species, thrombosis, inflammation and cytokine mediated damage. The pathways that are limited by the TSP1-CD47 system are precisely those that would be of most benefit in combating the damage of IRI. Thus, blocking the TSP1-CD47 pathway, as with the antibody compounds disclosed herein, will provide more robust functioning of these endogenous protective pathways.

The humanized anti-CD47 antibodies, antigen binding fragments thereof, and competing antibodies and antigen binding fragments thereof, of the present disclosure can be used in the methods disclosed in U.S. Pat. No. 8,236,313, the contents of which are herein incorporated by reference in their entirety.

CD47 and Cancer

CD47 has been identified as a novel therapeutic target in hematologic cancers (Majeti et al. (2009) Cell 138(2):286-99), as well as in solid tumors such as colon, prostate, breast, and brain cancers (Willingham et al. (2012) Proc Natl Acad Sci USA 109(17):6662-7). Many human cancers up-regulate cell surface expression of CD47, and those expressing the highest levels of CD47 are the most aggressive and the most lethal for patients. Increased CD47 expression is thought to protect cancer cells from phagocytic clearance by sending a “don't eat me” signal to macrophages via SIRPalpha, an inhibitory receptor that prevents phagocytosis of CD47-bearing cells (Jaiswal et al. (2009) Cell 138(2):271-851; Chao et al. (2010) Science Translational Medicine 2(63):63ra94). Thus, the increase of CD47 expression by many cancers provides them with a cloak of “selfness” that slows their phagocytic clearance by macrophages and dendritic cells. Anti-CD47 mAbs (CD47mAbs) that block the CD47/SIRPalpha interaction enhance phagocytosis of cancer cells in vitro and contribute to control of tumor burden in published human to mouse xenograft tumor models.

Antibodies that block CD47 and prevent its binding to SIRPalpha (“blocking mAbs”) have shown efficacy in human tumor in mouse (xenograft) tumor models. Such blocking CD47mAbs exhibiting this property promote (increase) the phagocytosis of cancer cells by macrophages, which can reduce tumor burden (Majeti et al. (2009) Cell 138(2):286-99) and may ultimately lead to generation of an adaptive immune response to the tumor (Tseng et al. (2013) Proc Natl Acad Sci USA. 110(27):11103-8).

Therapeutic Indications IRI-Related and Autoimmune/Inflammatory Conditions

Administration of a CD47 mAb or antigen binding fragment thereof disclosed herein can be used to treat a number of diseases and conditions in which IRI is a contributing feature, and to treat various autoimmune and inflammatory diseases. These include: organ transplantation in which a mAb or antigen binding fragment thereof of the present disclosure is administered to the donor prior to organ harvest, to the harvested donor organ, to the organ preservation solution, to the recipient patient, or to any combination thereof; skin grafting; surgical resections or tissue reconstruction in which such mAb or fragment is administered either locally by injection to the affected tissue or parenterally to the patient; reattachment of body parts; treatment of traumatic injury; pulmonary hypertension; sickle cell disease (crisis); myocardial infarction; stroke; surgically-induced ischemia; acute kidney disease/kidney failure; any other condition in which IRI occurs and contributes to the pathogenesis of disease; and autoimmune/inflammatory diseases, including arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

CD47 mAbs and antigen binding fragments thereof of the present disclosure can also be used to increase tissue perfusion in a subject in need of such treatment. Such subjects can be identified by diagnostic procedures indicating a need for increased tissue perfusion. In addition, the need for increased tissue perfusion may arise because the subject has had, is having, or will have, a surgery selected from integument surgery, soft tissue surgery, composite tissue surgery, skin graft surgery, resection of a solid organ, organ transplant surgery, or reattachment or an appendage or other body part.

Susceptible Cancers

Presently disclosed mAbs and antigen binding fragments thereof effective as cancer therapeutics can be administered to patients, preferably parenterally, with susceptible hematologic cancers and solid tumors including, but not limited to, leukemias, including systemic mastocytosis, acute lymphocytic (lymphoblastic) leukemia (ALL), T cell—ALL, acute myeloid leukemia (AML), myelogenous leukemia, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), chronic myeloid leukemia (CML), myeloproliferative disorder/neoplasm, myelodysplastic syndrome, monocytic cell leukemia, and plasma cell leukemia; lymphomas, including histiocytic lymphoma and T cell lymphoma, B cell lymphomas, including Hodgkin's lymphoma and non-Hodgkin's lymphoma, such as low grade/follicular non-Hodgkin's lymphoma (NHL), cell lymphoma (FCC), mantle cell lymphoma (MCL), diffuse large cell lymphoma (DLCL), small lymphocytic (SL) NHL, intermediate grade/follicular NHL, intermediate grade diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, and Waldenstrom's Macroglobulinemia, ovarian cancer, breast cancer, endometrial cancer, colon cancer (colorectal cancer), rectal cancer, bladder cancer, lung cancer (non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung), bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma (liver cancer, hepatoma), gall bladder cancer, bile duct cancer, esophageal cancer, renal cell carcinoma, thyroid cancer, squamous cell carcinoma of the head and neck (head and neck cancer), testicular cancer, cancer of the endocrine gland, cancer of the adrenal gland, cancer of the pituitary gland, cancer of the skin, cancer of soft tissues, cancer of blood vessels, cancer of brain, cancer of nerves, cancer of eyes, cancer of meninges, cancer of oropharynx, cancer of hypopharynx, cancer of cervix, and cancer of uterus, glioblastoma, meduloblastoma, astrocytoma, glioma, meningioma, gastrinoma, neuroblastoma, melanoma, myelodysplastic syndrome, and sarcomas including, but not limited to, osteosarcoma, Ewing sarcoma, leiomyosarcoma, synovial sarcoma, alveolar soft part sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chrondrosarcoma.

In certain cases, it may be advantageous to administer the mAb directly to the cancer by injection into the tumor.

Since CD47 expression is up-regulated on many cancers, it may also be desirable to use one or more of the disclosed mAbs as imaging and diagnostic agents when labeled with radioactive or other tracers known to those skilled in the art of in vivo imaging of cancers/tumors.

The following examples describe various aspects of the present disclosure, but should not be considered as limiting the disclosure only to these particularly disclosed embodiments. The materials and methods employed in these examples are for illustrative purposes, and are not intended to limit the practice of the present disclosure thereto. Any materials and methods similar or equivalent to those described herein as would be apparent to one of ordinary skill in the art can be used in the practice or testing of the present compounds and methods.

Example 1 Production of CD47 Antibodies

The humanized antibodies disclosed herein comprise frameworks derived from the human genome. The collection covers the diversity found in the human germ line sequences, yielding functionally expressed antibodies in vivo. The complementarity determining regions (CDRs) in the light and heavy chain variable regions of the target chimeric, non-human antibody VxP037-01LC/VxP037-01HC (SEQ ID NO:7/SEQ ID NO:57) are determined following commonly accepted rules disclosed, for example, in “Protein Sequence and Structure Analysis of Antibody Variable Domains”, In: Antibody Engineering Lab Manual, eds. S. Duebel and R. Kontermann, Springer-Verlag, Heidelberg (2001)). The CDR fragments are synthesized and combined with pools of frameworks to generate full length variable domains. The humanized variable domains are then combined with a secretion signal and human kappa and human IgG1 constant domains, and cloned into a mammalian expression system (e.g., OptiCHO System, Life Technologies, Carlsbad, Calif.) to generate a library of humanized IgG, IgG2, and IgG4 variants. An aliquot of the library is sequenced to ensure high diversity and integrity of the reading frames of the individual clones. Aliquots of the humanized variant library are then re-arrayed as single clones into 96 well plates, mini-prepped (e.g., 96 well Miniprep Kit, Qiagen Hilden, Germany), and transfected into CHO cells (Lipofectamine transfection protocol as recommended by Life Technologies, Carlsbad, Calif.). Transfected CHO cells are grown in DMEM medium with 10% FBS (both from Life Technologies, Carlsbad, Calif.) at 37° C. under 5% CO2. The humanized variants are expressed as full length IgG1 molecules, and secreted into the medium.

The cell culture supernatant containing the humanized IgG variants is then screened for binding to the target antigen, CD47. In parallel, the concentration of each variant is determined in order to calculate specific activity for each clone. The specific activity of each clone is compared to the specific activity of chimeric clone VxP037-01 LC-Pro/VxPO37-01HC-Pro (SEQ ID NO:107/SEQ ID NO:109) expressed on the same plate, and normalized. Top hits from each plate are re-arrayed and re-screened for confirmation. The final candidates are selected by specific activity, functional activity, expression level, and sequence diversity, as well as other criteria, as described below.

A non-glycosylated version (IgG1-N297Q) was created by site directed mutagenesis of heavy chain position 297 to change the asparagine to glutamine (pVxP037-01-HC-IgG1 N297Q-Pro; SEQ ID NO:111). IgG2, IgG4-S228P and IgG4-2SS8P-L235E isotypes were constructed by cloning the heavy chain variable domain in frame with the human IgG2 and IgG4-S228P constant domains (pVxK7b-037-hum01-HC-IgG2-Pro, SEQ ID NO:112 or pVxK7b-037-hum01-HC_IgG4 S228P-Pro, SEQ ID NO:113, pVxK7b-037-hum01-HC-IgG4 S228P L235E-Pro, SEQ ID NO: 122).

Example 2 CD47 Antibody Sequences

The amino acid sequences of the light chain and heavy chain variable regions, the complete light and heavy chains, and the respective encoding nucleotide sequences of the foregoing, of the present human engineered antibodies are listed below in the section entitled “Amino Acid and Nucleic Acid Sequences.”

Also included in this list are complete light chain sequences (SEQ ID NO:107/SEQ ID NO:108), complete heavy chain amino acid and respectively encoding nucleotide sequences of humanized IgG1 (SEQ ID NO:109/SEQ ID NO:110), complete heavy chain amino acid and respectively encoding nucleotide sequences of humanized IgG1 with a N→Q mutation at amino acid position 297 (SEQ ID NO:111/SEQ ID NO:114), IgG2 (SEQ ID NO:112/SEQ ID NO:115), and IgG4 (SEQ ID NO:113/SEQ ID NO: 116) antibodies.

SEQ ID NO:117 shows the amino acid sequence of framework 4+the light chain constant domain amino acid sequence of chimeric complete light chain amino acid sequence SEQ ID NO:107.

SEQ ID NOs: 118, 119, 120, 121, and 124 show the amino acid sequences of framework 4+the heavy chain constant domain amino acid sequences of complete heavy chain amino acid sequences SEQ ID NOs:109, 111, 112, 113, and 122 respectively.

All the light chain variable regions SEQ ID NOs: 7-31 can further comprise SEQ ID NO:117, and all the heavy chain amino acid sequences SEQ ID NOs:57-81 can further comprise any of SEQ ID NOs:118, 119, 120, 121, and 124 thereby describing complete antibody sequences encompassed by this disclosure.

The light chain and heavy chain CDR amino acid sequences are shown in Tables 1 and 2, respectively.

TABLE 1 Light Chain CDRs. CDR1 CDR2 CDR3 RSSQSLVHSNGNTYLH KVSYRFS SQNTHVPRT (SEQ ID NO: 1) (SEQ ID NO: 2) (SEQ ID NO: 3)

TABLE 2 Heavy Chain CDRs. CDR1 CDR2 CDR3 GYTFTNYYVF DINPVNGDTNFNEKFKN GGYTMDY (SEQ ID NO: 4) (SEQ ID NO: 5) (SEQ ID NO: 6)

Example 3 Binding of Antibodies to CD47 of Different Species

Cross species reactivity of humanized antibodies of the present disclosure is determined using freshly isolated red blood cells (RBCs), which display CD47 on their surface, from human, mouse, rat, pig, cynomolgus monkey, and dog according to the methods disclosed in Kamel et al. (2010) Blood. Transfus. 8(4):260-266.

Supernatants containing secreted antibodies are collected from CHO cells transiently transfected with plasmids encoding antibody clones and used as collected, or antibodies are further purified from the supernatants using standard methods. Transfected CHO cells are grown in F-12 medium containing 10% heat inactivated fetal bovine serum (BioWest; S01520). Antibody concentration in the supernatants is determined utilizing a quantitative ELISA. ELISA plates are coated with a donkey anti-human FC antibody (Sigma; Catalog #12136) at 10 μg/ml overnight at 4° C. (Promega; Catalog #W4031). Plates are washed with PBS, and then blocked with casein blocking solution (ThermoScientific; Catalog #37532) for 60 minutes at room temperature. Plates are again washed with PBS, tissue culture supernatants are added, and the plates are incubated for 60 minutes at room temperature. Plates are then washed three times with PBS and incubated with peroxidase-conjugated goat anti-human IgG (Jackson Immunoresearch Labs; Catalog #109-035-003) for 60 minutes at room temperature. Plates are washed three times with PBS, and the peroxidase substrate 3,3′-5,5′-tetramethylbenzidine is added (Sigma; Catalog #T4444). Reactions are terminated by the addition of HCl to 0.7N, and absorbance at 450 nM is determined using a Tecan model Infinite M200 plate reader.

RBCs are incubated for 60 minutes on ice with tissue culture supernatants containing the secreted humanized antibodies at a concentration of 10 ng/ml in a solution of phosphate buffered saline, pH 7.2, 2.5 mM EDTA (PBS+E), or with various concentrations of purified antibodies. Cells are then washed with cold PBS+E, and incubated for an additional hour on ice with FITC labeled donkey anti-human antibody (Jackson Immuno Research Labs, West Grove, Pa.; Catalogue #709-096-149) in PBS+E. Cells are then washed with PBS+E, and antibody binding is analyzed using a BD FACSAria Cell Sorter (Becton Dickinson) or a C6 Accuri Flow Cytometer (Becton Dickinson). Antibody binding is quantitated by comparison of mean fluorescence values relative to that of chimeric antibody >VxP037-01LC (SEQ ID NO:7))/>VxPO37-01HC (SEQ ID NO:57). The mean fluorescence value for each antibody is divided by the mean fluorescence value for the chimeric antibody.

The results obtained from the supernatants are shown in Table 3, where “Chimera” represents chimeric antibody >VxP037-01LC (SEQ ID NO:7))/>VxPO37-01HC (SEQ ID NO:57; complete sequences VxPO37-01LC-Pro/VxP037-01HC-Pro (SEQ ID NO:107/SEQ ID NO:109)), Clone 1 represents >pVxK7b-037-hum01-LC (SEQ ID NO:8)/>pVxK7b-037-hum01-HC (SEQ ID NO:58), Clone 2 represents >pVxK7b-037-hum02-LC (SEQ ID NO:9)/>pVxK7b-037-hum02-HC (SEQ ID NO:59), and so on similarly for remaining clones 3-24. Each antibody also contains a light chain constant domain (SEQ ID:117) and a heavy chain constant domain selected from among SEQ ID NO:118, SEQ ID NO:119, SEQ ID NO:120, and SEQ ID NO:121.

TABLE 3 Binding of Humanized Antibodies to CD47 on the Surface of Red Blood Cells of Different Mammalian Species. Clone No. Human Mouse Rat Pig Dog Chimera 1.0 1.0 1.0 1.0 1.0 1 1.1 1.7 2.7 1.3 1.0 2 1.0 1.2 2.6 1.2 1.0 3 0.7 0.9 1.7 0.9 0.9 4 0.6 0.6 1.0 0.6 0.6 5 1.0 1.0 2.2 1.2 1.1 6 0.9 1.2 2.1 1.1 1.1 7 0.5 0.4 0.8 0.9 0.8 8 0.7 0.7 1.2 0.8 0.8 9 1.2 1.4 3.7 1.6 1.0 10 1.1 1.2 2.9 1.5 1.1 11 0.8 0.7 1.2 1.2 0.8 12 0.8 0.6 1.3 1.4 0.9 13 1.2 1.3 3.1 1.4 1.0 14 1.1 1.5 3.2 1.4 1.3 15 1.0 1.3 2.4 1.2 1.1 16 0.9 1.0 2.1 1.1 1.1 17 0.8 0.9 2.1 1.3 1.3 18 1.0 1.3 2.2 1.2 1.5 19 0.7 1.0 2.6 1.3 1.2 20 1.3 1.5 1.9 1.7 1.1 21 1.2 1.2 2.8 1.4 1.1 22 1.1 1.2 2.8 1.4 1.0 23 1.2 1.4 3.3 1.7 1.1 24 0.8 0.7 1.2 1.1 1.0

FIG. 1 shows cross species binding curves to human, mouse, rat, and porcine RBCs (panels A, B, C, and D, respectively, generated using various concentrations of purified antibodies from clones Cl 1, Cl 1.1, Cl 13, and Cl 13.1. Clones Cl 1 and Cl 13 are as described above in Table 3. Clones Cl 1.1 and Cl 13.1 are Fc mutants of clones Cl 1 and Cl 13, respectively, modified to reduce effector function. Each has an Asn297→Gln (N297Q) mutation in the Fc domain (Sazinsky et al. (2008) PNAS 105(51):20167-20172). All of these clones exhibit concentration-dependent binding to all of the species of RBCs tested. These clones also bind to cynomolgus monkey RBCs as shown in FIG. 9 by the concentration-dependent binding of Clones 1.1 (IgG 1 N297Q mutation) and 1.3 (IgG4 S228P mutation).

Table 4 shows the apparent affinities of these clones to human RBCs determined by non-linear fits (Prism GraphPad software) of the median fluorescence intensities at various antibody concentrations. Clones 1, 1.1, 13, and 13.1 all have apparent Kd values in the low nanomolar range.

TABLE 4 Binding Affinity of Humanized Antibodies to CD47 on Human RBCs. Clone 1 Clone 1.1 Clone 13 Clone 13.1 Kd (ng/ml) 226.2 307.1 86.4 182.9 Kd (apparent) 1.51 2.04 0.58 1.21 nM

Binding activities of humanized clones 1, 3, 5, 8, 13, 14, 17, 20 and 23, either IgG1 N297Q (Clone 1.1, Clone 13.1, etc.) or an IgG4 S228P L235E (Clones 1.2, etc.), to human and mouse CD47 were determined using cell-based ELISA assays with either human OV10 or mouse 4T1 cells expressing cell surface CD47. OV10 cells are grown in IMDM medium containing 10% heat inactivated fetal bovine serum (BioWest; S01520) and 4T1 cells are grown in RMPI medium containing 10% heat inactivated fetal bovine serum (BioWest; S01520). One day before assay, 3×104 cells are plated in 96 well cell bind plates (Corning #3300, VWR #66025-626) so that they are 95-100% confluent at the time of assay. Cells are washed and various concentrations of purified antibodies added in either IMDM or RPMI at 37° C. for 1 hour in 95% O2/5% CO2. Cells are then washed with media and incubated for an additional hour at 37° C. with HRP labeled secondary anti-human antibody (Promega) diluted 1/2500 in media. Cells are washed three times with PBS, and the peroxidase substrate 3,3′,5,5′-tetramethylbenzidine is added (Sigma; Catalog #T4444). Reactions are terminated by the addition of HCl to 0.7N, and absorbance at 450 nM is determined using a Tecan model Infinite M200 plate reader. The apparent binding affinities of these clones to human and mouse cells is determined by non-linear fits (Prism GraphPad software). As shown in Table 5, all of the humanized clones bind to both human and mouse tumor cells with apparent affinities in the picomolar range. Agglutination of human RBCs is assessed following incubation of human RBCs with various concentrations of humanized blocking only clones. Blood is diluted (1:50) and washed 3 times with PBS/EDTA/BSA. RBCs are added to U-bottomed 96 well plates with equal volumes of the antibodies (75 μl) and incubated for 3 hrs at 37° C. and overnight at 4° C. As shown in Table 3, while all clones bind similarly to human RBCs, they exhibit different agglutination activities. Clones 3.1, 3.2, 13.1, 13.2, 20.1, and 20.2 cause agglutination of human RBCs, whereas Clones 1.1 (IgG1 N297Q), 1.2 (IgG4 S228P L235E), 5.1, 5.2, 8.1, 8.2, 14.1, 14.2, 17.1, 17.2, 23.1 and 23.2 do not.

TABLE 5 Humanized Blocking-Only mAb. CD47 Binding Affinity and Red Blood Cell Hemagglutination. Human OV10_ Mouse ELISA 4T1_ Hemag- Kd ELISA glutination [pM] Kd [pM] Human RBC Clone 1.1 147.7 131.6 No hum01 IgG4 S228P L235E Clone 1.2 150.5 175.5 No hum13 IgG1 N297Q Clone 13.1 88.6 86.7 Yes hum 13 IgG4 S228P L235E Clone 13.2 107.8 177.8 Yes hum03 IgG1 N297Q Clone 3.1 115.3 131.7 Yes hum03 IgG4 S228P L235E Clone 3.2 129.3 202 Yes hum05 IgG1 N297Q Clone 5.1 124.7 112.4 No hum05 IgG4 S228P L235E Clone 5.2 107.5 171.8 No hum08 IgG1 N297Q Clone 8.1 114.9 205 No hum08 IgG4 S228P L235E Clone 8.2 121.4 94.9 No hum14 IgG1 N297Q Clone 14.1 96.2 94.9 No hum14 IgG4 S228P L235E Clone 14.2 89.9 101.8 No hum17 IgG1 N297Q Clone 17.1 85.5 197.9 No hum17 IgG4 S228P L235E Clone 17.2 120.9 147 No hum20 IgG1 N297Q Clone 20.1 120.9 236.9 Yes hum20 IgG4 S228P L235E Clone 20.2 113.5 354 Yes hum23 IgG1 N297Q Clone 23.1 99.6 92.2 No hum23 IgG4 S228P L235E Clone 23.2 194.6 192.2 No

These data demonstrate that all of the humanized CD47 mAb clones disclosed herein bind to CD47 of a variety of different mammalian species in addition to human CD47, confirming the useful cross-species reactivity of these antibodies and that certain of these antibodies do not cause agglutination of human RBCs

Example 4 Antibodies to CD47 Enhance Phagocytosis

To assess the effect of humanized CD47 mAbs on phagocytosis of tumor cells by marcrophages in vitro the following method is employed using flow cytometry, essentially as described by Willingham et al. (2012) Proc Natl Acad Sci USA 109(17):6662-7 and Tseng et al. (2013) Proc Natl Acad Sci USA 110(27):11103-8.

Human derived macrophages are derived from leukapheresis of healthy human peripheral blood incubated in human AB serum (Sigma Aldrich) for 24 hours in culture. After 24 hours, all non-adherent cells are removed and the remaining adherent macrophages are incubated in RPMI medium (10% fetal bovine serum (FBS; Hyclone) and antibiotics) for two weeks. In additional experiments, human macrophages are derived from human peripheral blood and incubated in AIM-V media (Life Technologies) for 7-10 days. For the in vitro phagocytosis assay, macrophages are re-plated at a concentration of 5×104 cells per well in 1 ml of RPMI media in a 24 well plate and allowed to adhere for 24 hours. Once the effector macrophages have adhered to the culture dish, the target cancer cells (Jurkat) are labeled with 1 μM 5(6)-Carboxyfluorescein diacetate N-succinimidyl ester (CFSE; Sigma Aldrich) and added to the macrophage cultures at a concentration of 2×105 cells in 1 ml of RPMI media (4:1 target to effector ratio). CD47mAbs (1-10 μg/ml) are added immediately upon mixture of target and effector cells and allowed to incubate at 37° C. for 2-3 hours. After 2-3 hours, all non-phagocytosed cells are removed and the remaining cells are washed three times with phosphate buffered saline (PBS; Sigma Aldrich). Cells are trypsinized, collected into microcentrifuge tubes and incubated in 100 ng of allophycocyanin (APC) labeled CD14 antibodies (BD biosciences) for 30 minutes. Cells are washed once and analyzed by flow cytometry (Accuri C6; BD biosciences) for the percentage of CD14 positive cells that are also CFSE positive indicating complete phagocytosis.

As shown in FIG. 2, both Clone 1 and Clone 13 humanized IgG1 mAbs increase phagocytosis of Jurkat cells by human macrophages grown in serum. Clones 1.1 and Clone 13.1 are identical to Clones 1 and 13 except for a mutation of residue N297 to Q that reduces the affinity of the IgG1 molecule to the Fc receptor. The IgG4 versions of Clone 1 and Clone 13 with a mutation of residue S228 to P to reduce chain strand exchange (also designated as Clones 1.3 and 13.3) also increase phagocytosis of Jurkat cells. The IgG4 isotype also has reduced affinity for activating Fc receptors. As shown in FIG. 10, the IgG4 version of Clone 1 with a mutation of residue S228 to P and a further mutation of residue L235 to E to reduce Fc effector function (designated as Clone 1.2) also increases phagocytosis of Jurkat cells.

Therefore, all isotypes/mutants of Clone 1 and 13 enhance phagocytosis via blocking the CD47/SIRPalpha interaction.

Example 5 Antibodies to CD47 Regulate Nitric Oxide Signaling

The purpose of this experiment is to demonstrate that humanized antibody clones of the present disclosure exhibit the ability to reverse TSP1-mediated inhibition of NO-stimulated cGMP synthesis as, for example, described previously using mouse monoclonal antibodies to CD47 as disclosed by Isenberg et al. (2006) J. Biol. Chem. 281:26069-80.

The method employed to measure cGMP is as described by the manufacturer (CatchPoint Cyclic-GMP Fluorescent Assay Kit, Molecular Devices, Sunnyvale, Calif.). Jurkat JE6.1 cells (ATCC, Manassas, Va.; Catalog #TIB-152) are used as these cells retain the NO-cGMP signaling pathway when grown in culture and exhibit a robust and reproducible inhibitory response to TSP1 ligation of CD47. Cells are grown in Iscove's modified Dulbeccco's medium containing 5% (v/v) heat inactivated fetal bovine serum (BioWest; Catalogue #S01520), 100 units/mL penicillin, 100 μg mL streptomycin (Sigma; Catalogue #P4222) at densities less than 1×106 cells/mL. For the cGMP assay, cells are plated in 96 well tissue culture plates at a density of 1×105 cells/ml in Iscoves modified Dulbecco's medium containing 5% (v/v) heat inactivated fetal bovine serum (BioWest; Catalog #S01520), 100 units/mL penicillin, 100 μg/mL streptomycin (Sigma; #P4222) for 24 hours and then transferred to serum free medium overnight.

The humanized antibodies as disclosed herein, purified from transient transfections in CHO cells as described above in Example 3, as well as the control chimeric antibody, are then added at a final concentration of 20 ng/ml, followed 15 minutes later by 0 or 1 μg/ml human TSP1 (Athens Research and Technology, Athens, Ga., Catalogue #16-20-201319). After an additional 15 minutes, the NO donor, diethylamine (DEA) NONOate (Cayman Chemical, Ann Arbor, Mich., Catalog #82100), is added to half the wells at a final concentration of 1 μM. Five minutes later, the cells are lysed with buffer supplied in the cGMP kit, and aliquots of each well are assayed for cGMP content.

As shown in FIGS. 3 and 4, none of the present humanized antibody clones tested, or the chimeric control mAb, has an effect on basal cGMP levels. As expected, the chimeric antibody VxP037-01LC-Pro/VxP037-01HC-Pro (SEQ ID NO:107/SEQ ID NO: 109)) reverses the TSP1 inhibition.

Humanized Clones 1, 9, 11, 13, and 24 of the present disclosure also significantly reverse TSP1 inhibition, demonstrating that they have the ability to increase NO signaling (FIGS. 3 and 4), suggesting their utility in protecting the cardiovascular system against stresses including, but not limited to, those resulting from wounding, inflammation, hypertension, metabolic syndrome, ischemia, and ischemia-reperfusion injury (IRI).

Example 6 Reduction of Ischemia-Reperfusion Injury In Vivo

The purpose of this experiment is to demonstrate that a humanized antibody clone disclosed herein, i.e., Clone 1, that is shown to regulate nitric oxide signaling in vitro in Example 5, is effective in reducing IRI and kidney damage in vivo in a rat kidney transplant model under standard conditions, i.e., with no warm ischemic time but with cold ischemic time. IRI significantly contributes to delayed graft function and inflammation leading to graft loss, and is exacerbated by the thrombospondin-1/CD47 system through inhibition of nitric oxide signaling.

A syngeneic rat renal transplantation model of IRI with bilaterally nephrectomized recipients is used to evaluate the effect of the anti-CD47 monoclonal antibody Clone 1 on graft function following transplantation as described in Schumacher et al. (2003) Microsurg. 23:389-394 and Karatzas et al. (2007) Microsug. 27:668-672.

Male Lewis rats weighing 275-300 g are obtained from Charles River Laboratories (Wilmington, Mass.). Donor kidneys are flushed with 50 μg of purified Clone 1 or vehicle (phosphate buffered saline, pH 7.2), and stored at 4° C. in University of Wisconsin preservation solution (UW) for 6 hours prior to transplantation. Two days following transplantation, kidney function is assessed by measuring serum creatinine by standard methodology.

As shown in FIG. 5, CD47mAb Clone 1 perfusion of donor kidneys results in improved kidney function compared to controls as measured by a reduction in serum creatinine.

An additional experiment is shown that also demonstrates the ability of CD47mAbs of the present disclosure to improve kidney function of extended criteria organs that have also undergone a 60 minute period of warm ischemic time in addition to a 6 hour cold ischemic time. Male Lewis rats weighing 275-300 g underwent 60 minutes of warm ischemia, prior to flushing the donor kidneys with 50 μg of purified Clone 1.1 or an IgG control mAb. Kidneys are stored at 4° C. in University of Wisconsin preservation solution (UW) for 6 hours prior to transplantation. In this experiment, survival is monitored over a 7 day time period.

As shown in FIG. 6, all animals that received the IgG control mAb-treated kidney die within 4 days. In contrast, survival is significantly increased in the animals that received the Clone1.1 treated kidney, with 30% of the animals surviving for the 7 day duration of the experiment.

Together, these experiments show that with both standard and extended criteria donor kidneys, Clone 1 and Clone 1.1 reduce IRI and increase kidney function and survival outcomes, respectively.

Example 7 Acute Promyelocytic Leukemia (APL) Anti-Tumor Activity In Vivo

The purpose of this experiment is to demonstrate that a humanized antibody clone disclosed herein, i.e., Clone 13, reduces tumor burden in vivo in a mouse leukemia model.

The anti-tumor activity of the anti-CD47mAb Clone13 (Cl 13; clone number as described above in Examples 2 and 3) is determined in a syngeneic murine model of Acute Promyelocytic Leukemia (APL) as described in Ramirez et al. (2009) Blood 113:6206-6214.

Murine APL cells (B6APL1) are injected intravenously into C57BL/6 mice that are randomized into three groups (5-10 mice per group): Group 1: no APL; Group 2: APL with no treatment; Group 3: APL with anti-CD47mAb Cl 13treatment. Antibody treatment is initiated on the day of tumor inoculation (day 0), and given in single doses of 10 μg/dose (0.4 mg/kg) in phosphate buffered saline, pH 7.2, by intraperitoneal injection on days 0, 3, and 6.

Tumor burden is evaluated at day 25 following tumor cell inoculation. Blood samples from each mouse are analyzed for white blood cell count using an automated hemocytometer, and circulating APL cells (representing the tumor burden) are quantified by flow cytometry (CD34+/CD117+ cells).

As shown in FIG. 7, mice treated with Cl 13 have reduced tumor burden compared to untreated mice at 25 days after tumor inoculation, thus demonstrating anti-tumor activity of this humanized clone.

Example 8 HepG2 Anti-Tumor Activity In Vivo

The purpose of this experiment is to demonstrate that a humanized antibody clone disclosed herein, i.e., Clone 1.1, reduces tumor burden in vivo in a mouse xenograft model of human hepatocellular carcinoma (HCC).

Male NSG mice are obtained from The Jackson Laboratory (Bar Harbor, Me.) and housed in cages in temperature and light-controlled environments with access to water and food ad libitum. For the heterotopic xenograft model, HepG2-luc2 cells (Perkin Elmer, Waltham, Mass. #134280) are suspended in DMEM containing 25% (v/v), and 1,000,000 cells implanted subcutaneously into the dorsal subcutaneous space of 4- to 8-wk-old NSG mice. After 2 weeks of growth, antibody treatment is begun with twice-weekly intraperitoneal injections of 15 mg/kg of either anti-CD47 antibody Clone 1.1 or an IgG control for 6 weeks. Tumor volumes are calculated twice weekly using (length×width)/0.6. After 6 weeks of treatment, animals are euthanized and tumors were resected, weighed, and fixed in 10% formalin.

As shown in FIG. 8, treatment with the CD47 mAb Clone 1.1 significantly reduced tumor growth of the HepG2 tumors (p<0.01), demonstrating anti-tumor efficacy on solid tumors.

Embodiments of the disclosure being thus described, it will be obvious that the same may be varied in many ways. Such variations are not to be regarded as a departure from the spirit and scope of the disclosure, and all such modifications as would be obvious to one skilled in the art are intended to be included within the scope of the following claims.

Example 9 Antibodies to CD47 Block CD47/SIRPalpha Binding

To assess the effect of humanized CD47 mAbs on binding of CD47 to SIRPalpha in vitro the following method is employed using binding of CD47 expressing Jurkat cells to SIPRalpha bound to plates.

Polystyrene 96 well tissue culture plates were coated for 60 minutes at 37 degrees with 2 μg/ml SIRP-Fc fusion protein (R and D Systems, cat #4546-SA). Unbound SIRP-Fc fusion was removed and nonspecific protein binding sites were blocked with casein for 60 minutes at 37 degrees (ThermoScientific cat #37528). Blocking solution was removed and plates were washed with PBS. Jurkat cells in RPMI growth medium containing 10% FBS were added (100,000 cells/well), with or without CD47 antibodies at 1 ug/ml. Cells were incubated at 37 degrees for 60 minutes. Cells were aspirated and wells gently washed twice with PBS. Growth medium containing WST-1 reagent was added and plates incubated at 37 degrees for 2 hrs (Cayman Scientific cat #10008883). Absorbance was read at 450 nm. As shown in FIG. 11, Clones 1.1, 1.2, 1.3, 13.1, 13.2 and 13.3 all block the interaction of CD47 (expressed on the Jurkat cells) with SIPRalpha while a control antibody that does not bind to CD47 does not block the CD47/SIRPalpha interaction.

Amino Acid and Nucleic Acid Sequences Light Chain Variable Region Amino Acid Sequences Murine Sequence

>VxP037-01LC: Underlined amino acid sequences represent CDRs (SEQ ID NO: 7) DVVMTQTPLSLSVSLGDQASISCRSSQSLVHSNGNTYLHWYLQKPGQSPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFCSQNTHVP RTFGQG

Humanized Light Chain Sequences

>pVxK7b-037-hum01-LC (SEQ ID NO: 8) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum02-LC (SEQ ID NO: 9) DVVMTQSPLSLPVTLGQPASISCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCSQNTHVP RTFGQG >pVxK7b-037-hum03-LC (SEQ ID NO: 10) DVVMTQSPLSLPVTLGQPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum04-LC (SEQ ID NO: 11) DIQMTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYLQKPGQSPQ LLIYKVSYRFSGIPARFSGSGSGTEFTLTISSLQSEDFAVYYCSQNTHVP RTFGQG >pVxK7b-037-hum05-LC (SEQ ID NO: 12) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYLQKPGQSPQ LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum06-LC (SEQ ID NO: 13) DIQMTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYLQKPGQSPQ LLIYKVSYRFSGIPARFSGSGSGTEFTLTISSLQSEDFAVYYCSQNTHVP RTFGQG >pVxK7b-037-hum07-LC (SEQ ID NO: 14) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYLQKPGQSPQ LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum08-LC (SEQ ID NO: 15) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum09-LC (SEQ ID NO: 16) DVVMTQSPLSLPVTLGQPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum10-LC (SEQ ID NO: 17) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYLQKPGQSPQ LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum11-LC (SEQ ID NO: 18) EIVLTQSPATLSVSPGERATLSCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPSRFSGSGSGTDFTFTISSLEAEDAATYYCSQNTHVP RTFGQG >pVxK7b-037-hum12-LC (SEQ ID NO: 19) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum13-LC (SEQ ID NO: 20) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum14-LC (SEQ ID NO: 21) DVVMTQSPLSLPVTLGQPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum15-LC (SEQ ID NO: 22) AIQLTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCSQNTHVP RTFGQG >pVxK7b-037-hum16-LC (SEQ ID NO: 23) AIQLTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum17-LC (SEQ ID NO: 24) AIQLTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum18-LC (SEQ ID NO: 25) EIVLTQSPATLSVSPGERATLSCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPSRFSGSGSGTDFTFTISSLEAEDAATYYCSQNTHVP RTFGQG >pVxK7b-037-hum19-LC (SEQ ID NO: 26) DVVMTQSPLSLPVTLGQPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum20-LC (SEQ ID NO: 27) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYLQKPGQSPQ LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum21-LC (SEQ ID NO: 28) AIQLTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum22-LC (SEQ ID NO: 29) EIVLTQSPATLSVSPGERATLSCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPSRFSGSGSGTDFTFTISSLEAEDAATYYCSQNTHVP RTFGQG >pVxK7b-037-hum23-LC (SEQ ID NO: 30) DIVMTQTPLSLPVTPGEPASISCRSSQSLVHSNGNTYLHWYQQKPGKAPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQNTHVP RTFGQG >pVxK7b-037-hum24-LC (SEQ ID NO: 31) AIQLTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTYLHWYQQKPGQAPR LLIYKVSYRFSGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCSQNTHVP RTFGQG

Murine Light Chain Variable Region Nucleic Acid Sequence

>VxP037-01LC (SEQ ID NO: 32) GATGTTGTTATGACCCAAACTCCACTCTCCCTGTCTGTCAGTCTTGGAGA TCAAGCCTCCATCTCTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGCCAGTCTCCAAAG CTCCTGATCTACAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGATCAGGGACAGATTTCACACTCAAGATCAGCAGAGTGG AGGCTGAGGATCTGGGAGTTTATTTCTGCTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGAG

Humanized Light Chain Variable Region Nucleic Acid Sequences

>pVxK7b-037-hum01-LC (SEQ ID NO: 33) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum02-LC (SEQ ID NO: 34) GATGTTGTGATGACTCAGTCTCCACTCTCCCTGCCCGTCACCCTTGGACA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCATCAAGGTT CAGCGGCAGTGGATCTGGGACAGAATTCACTCTCACCATCAGCAGCCTGC AGCCTGATGATTTTGCAACTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum03-LC (SEQ ID NO: 35) GATGTTGTGATGACTCAGTCTCCACTCTCCCTGCCCGTCACCCTTGGACA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum04-LC (SEQ ID NO: 36) GACATCCAGATGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGGCAGTCTCCACAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGATCCCAGCCAGGTT CAGTGGCAGTGGGTCTGGGACAGAGTTCACTCTCACCATCAGCAGCCTGC AGTCTGAAGATTTTGCAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum05-LC (SEQ ID NO: 37) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGGCAGTCTCCACAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum06-LC (SEQ ID NO: 38) GACATCCAGATGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGGCAGTCTCCACAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGATCCCAGCCAGGTT CAGTGGCAGTGGGTCTGGGACAGAGTTCACTCTCACCATCAGCAGCCTGC AGTCTGAAGATTTTGCAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum07-LC (SEQ ID NO: 39) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGGCAGTCTCCACAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum08-LC (SEQ ID NO: 40) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum09-LC (SEQ ID NO: 41) GATGTTGTGATGACTCAGTCTCCACTCTCCCTGCCCGTCACCCTTGGACA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum10-LC (SEQ ID NO: 42) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGGCAGTCTCCACAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum11-LC (SEQ ID NO: 43) GAAATTGTGTTGACACAGTCTCCAGCCACCCTGTCTGTGTCTCCAGGGGA AAGAGCCACCCTCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCCTCGAGGTT CAGTGGCAGTGGATCTGGGACAGATTTCACCTTTACCATCAGTAGCCTGG AAGCTGAAGATGCTGCAACATATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum12-LC (SEQ ID NO: 44) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum13-LC (SEQ ID NO: 45) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum14-LC (SEQ ID NO: 46) GATGTTGTGATGACTCAGTCTCCACTCTCCCTGCCCGTCACCCTTGGACA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum15-LC (SEQ ID NO: 47) GCCATCCAGTTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCATCAAGGTT CAGCGGCAGTGGATCTGGGACAGAATTCACTCTCACCATCAGCAGCCTGC AGCCTGATGATTTTGCAACTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum16-LC (SEQ ID NO: 48) GCCATCCAGTTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum17-LC (SEQ ID NO: 49) GCCATCCAGTTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum18-LC (SEQ ID NO: 50) GAAATTGTGTTGACACAGTCTCCAGCCACCCTGTCTGTGTCTCCAGGGGA AAGAGCCACCCTCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCCTCGAGGTT CAGTGGCAGTGGATCTGGGACAGATTTCACCTTTACCATCAGTAGCCTGG AAGCTGAAGATGCTGCAACATATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum19-LC (SEQ ID NO: 51) GATGTTGTGATGACTCAGTCTCCACTCTCCCTGCCCGTCACCCTTGGACA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum20-LC (SEQ ID NO: 52) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGGCAGTCTCCACAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum21-LC (SEQ ID NO: 53) GCCATCCAGTTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTT TCTGGGGTCCCAGACAGGTTCAGTGGCAGTGGGTCAGGCACTGATTTCAC ACTGAAAATCAGCAGGGTGGAGGCTGAGGATGTTGGAGTTTATTACTGTT CTCAAAATACACATGTTCCTCGGACGTTCGGCCAAGGG >pVxK7b-037-hum22-LC (SEQ ID NO: 54) GAAATTGTGTTGACACAGTCTCCAGCCACCCTGTCTGTGTCTCCAGGGGA AAGAGCCACCCTCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCCTCGAGGTT CAGTGGCAGTGGATCTGGGACAGATTTCACCTTTACCATCAGTAGCCTGG AAGCTGAAGATGCTGCAACATATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum23-LC (SEQ ID NO: 55) GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGA GCCGGCCTCCATCTCCTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTATCAGCAGAAACCAGGGAAAGCTCCTAAG CTCCTGATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG >pVxK7b-037-hum24-LC (SEQ ID NO: 56) GCCATCCAGTTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGA CAGAGTCACCATCACTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGG CTCCTCATCTATAAAGTTTCCTACCGATTTTCTGGGGTCCCATCAAGGTT CAGCGGCAGTGGATCTGGGACAGAATTCACTCTCACCATCAGCAGCCTGC AGCCTGATGATTTTGCAACTTATTACTGTTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGG

Heavy Chain Variable Region Amino Acid Sequences Murine Heavy Chain Variable Region Amino Acid Sequence: Underlined Amino Acid Sequences Indicate CDRs

>VxP037-01HC (SEQ ID NO: 57) EVQLQQFGAELVKPGASMKLSCKASGYTFTNYYVFWVKQRPGQGLEWIGD INPVNGDTNFNEKFKNKATLTVDKSSTTTYLQLSSLTSEDSAVYYCTRGG YTMDYWGQG

Humanized Heavy Chain Variable Region Amino Acid Sequences

>pVxK7b-037-hum01-HC (SEQ ID NO: 58) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQG >pVxK7b-037-hum02-HC (SEQ ID NO: 59) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQG >pVxK7b-037-hum03-HC (SEQ ID NO: 60) EVQLVQSGAEVKKPGESLRISCKGSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum04-HC (SEQ ID NO: 61) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQAPGKGLEWVSD INPVNGDTNFNEKFKNRVTISVDTSKNQFSLKLSSVTAADTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum05-HC (SEQ ID NO: 62) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQAPGKGLEWVSD INPVNGDTNFNEKFKNRVTISVDTSKNQFSLKLSSVTAADTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum06-HC (SEQ ID NO: 63) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum07-HC (SEQ ID NO: 64) QVQLQESGPGLVKPGATVKISCKVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum08-HC (SEQ ID NO: 65) QITLKESGPTLVKPTQTLTLTCTFSGYTFTNYYVFWIRQSPSRGLEWLGD INPVNGDTNFNEKFKNRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum09-HC (SEQ ID NO: 66) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum10-HC (SEQ ID NO: 67) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum11-HC (SEQ ID NO: 68) QVQLQESGPGLVKPGATVKISCKVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum12-HC (SEQ ID NO: 69) QVQLQESGPGLVKPGATVKISCKVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum13-HC (SEQ ID NO: 70) EVQLVQSGAEVKKPGESLRISCKGSGYTFTNYYVFWIRQSPSRGLEWLGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum14-HC (SEQ ID NO: 71) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQG >pVxK7b-037-hum15-HC (SEQ ID NO: 72) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum16-HC (SEQ ID NO: 73) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQG >pVxK7b-037-hum17-HC (SEQ ID NO: 74) EVQLVQSGAEVKKPGATVKISCKVSGYTFTNYYVFWIRQPPGKGLEWIGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum18-HC (SEQ ID NO: 75) EVQLVQSGAEVKKPGESLRISCKGSGYTFTNYYVFWIRQSPSRGLEWLGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum19-HC (SEQ ID NO: 76) EVQLVQSGAEVKKPGESLRISCKGSGYTFTNYYVFWIRQSPSRGLEWLGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum20-HC (SEQ ID NO: 77) QITLKESGPTLVKPTQTLTLTCTFSGYTFTNYYVFWVRQAPGQGLEWMGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG >pVxK7b-037-hum21-HC (SEQ ID NO: 78) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum22-HC (SEQ ID NO: 79) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum23-HC (SEQ ID NO: 80) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRLTISKDTSKNQVVLTMTNMDPVDTATYYCARGG YTMDYWGQG >pVxK7b-037-hum24-HC (SEQ ID NO: 81) QVQLQESGPGLVKPGATVKISCKVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTITADKSTSTAYMELSSLRSEDTAVYYCARGG YTMDYWGQG

Murine Heavy Chain Variable Region Nucleic Acid Sequence

>VxP037-01HC (SEQ ID NO: 82) GAGGTCCAGCTGCAGCAGTTTGGGGCTGAACTGGTGAAGCCTGGGGCTTC AATGAAGTTGTCCTGCAAGGCTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGAAACAGAGGCCTGGACAAGGCCTTGAGTGGATTGGAGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAA GGCCACACTGACTGTAGACAAGTCCTCCACCACAACATACTTGCAACTCA GCAGCCTGACATCTGAGGACTCTGCGGTCTATTACTGTACAAGAGGGGGT TATACTATGGACTACTGGGGTCAAGGA

Humanized Heavy Chain Variable Region Nucleic Acid Sequences

>pVxK7b-037-hum01-HC (SEQ ID NO: 83) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum02-HC (SEQ ID NO: 84) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum03-HC (SEQ ID NO: 85) GAAGTGCAGCTGGTGCAGTCTGGAGCAGAGGTGAAAAAGCCCGGGGAGTC TCTGAGGATCTCCTGTAAGGGTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum04-HC (SEQ ID NO: 86) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCAGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATATCAGTAGACACGTCCAAGAACCAGTTCTCCCTGAAGCTGA GCTCTGTGACCGCCGCGGACACGGCTGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum05-HC (SEQ ID NO: 87) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTCCGCCAGGCTCCAGGGAAGGGGCTGGAGTGGGTCAGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATATCAGTAGACACGTCCAAGAACCAGTTCTCCCTGAAGCTGA GCTCTGTGACCGCCGCGGACACGGCTGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum06-HC (SEQ ID NO: 88) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum07-HC (SEQ ID NO: 89) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTGGGGCTAC AGTGAAAATCTCCTGCAAGGTTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum08-HC (SEQ ID NO: 90) CAGATCACCTTGAAGGAGTCTGGTCCTACGCTGGTGAAACCCACACAGAC CCTCACGCTGACCTGCACCTTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGATCAGGCAGTCCCCATCGAGAGGCCTTGAGTGGCTGGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ATTCACCATCTCCAGAGACAACGCCAAGAACTCACTGTATCTGCAAATGA ACAGCCTGAGAGCCGAGGACACGGCTGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum09-HC (SEQ ID NO: 91) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum10-HC (SEQ ID NO: 92) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum11-HC (SEQ ID NO: 93) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTGGGGCTAC AGTGAAAATCTCCTGCAAGGTTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum12-HC (SEQ ID NO: 94) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTGGGGCTAC AGTGAAAATCTCCTGCAAGGTTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum13-HC (SEQ ID NO: 95) GAAGTGCAGCTGGTGCAGTCTGGAGCAGAGGTGAAAAAGCCCGGGGAGTC TCTGAGGATCTCCTGTAAGGGTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGATCAGGCAGTCCCCATCGAGAGGCCTTGAGTGGCTGGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGA >pVxK7b-037-hum14-HC (SEQ ID NO: 96) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum15-HC (SEQ ID NO: 97) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum16-HC (SEQ ID NO: 98) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum17-HC (SEQ ID NO: 99) GAGGTCCAGCTGGTACAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCTAC AGTGAAAATCTCCTGCAAGGTTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGATCCGCCAGCCCCCAGGGAAGGGGCTGGAGTGGATTGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum18-HC (SEQ ID NO: 100) GAAGTGCAGCTGGTGCAGTCTGGAGCAGAGGTGAAAAAGCCCGGGGAGTC TCTGAGGATCTCCTGTAAGGGTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGATCAGGCAGTCCCCATCGAGAGGCCTTGAGTGGCTGGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum19-HC (SEQ ID NO: 101) GAAGTGCAGCTGGTGCAGTCTGGAGCAGAGGTGAAAAAGCCCGGGGAGTC TCTGAGGATCTCCTGTAAGGGTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGATCAGGCAGTCCCCATCGAGAGGCCTTGAGTGGCTGGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum20-HC (SEQ ID NO: 102) CAGATCACCTTGAAGGAGTCTGGTCCTACGCTGGTGAAACCCACACAGAC CCTCACGCTGACCTGCACCTTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCCCCTGGACAAGGGCTTGAGTGGATGGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum21-HC (SEQ ID NO: 103) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum22-HC (SEQ ID NO: 104) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum23-HC (SEQ ID NO: 105) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG ACTCACCATCTCCAAGGACACCTCCAAAAACCAGGTGGTCCTTACAATGA CCAACATGGACCCTGTGGACACAGCCACGTATTACTGTGCAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA >pVxK7b-037-hum24-HC (SEQ ID NO: 106) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTGGGGCTAC AGTGAAAATCTCCTGCAAGGTTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACGATTACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCTGA GCAGCCTGAGATCTGAGGACACGGCCGTGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGA

Chimeric Complete Light Chain Amino Acid Sequence

>VxPO37-01-LC-Pro, below, represents a full length chimeric light chain variable domain (SEQ ID NO:7)+a constant domain amino acid sequence. The underlined amino acid sequence=framework 4+the constant domain. All full length humanized light chain sequences can contain a light chain variable region sequence selected from SEQ ID NOs:7-31 in combination with framework 4+the same constant domain as VxPO37-01-LC-Pro. However, while present, this constant domain is not shown for all the complete humanized light chain amino acid sequences.

>VxP037-01-LC-Pro (SEQ ID NO: 107) DVVMTQTPLSLSVSLGDQASISCRSSQSLVHSNGNTYLHWYLQKPGQSPK LLIYKVSYRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFCSQNTHVP RTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAK VQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACE VTHQGLSSPVTKSFNRGEC

Chimeric Complete Light Chain Nucleic Acid Sequence

The underlined nucleic acid sequence encodes the underlined protein sequence in >VxPO37-01-LC-Pro, above.

>VxP037-01-LC-DNA (SEQ ID NO: 108) GATGTTGTTATGACCCAAACTCCACTCTCCCTGTCTGTCAGTCTTGGAGA TCAAGCCTCCATCTCTTGCAGATCTAGTCAGAGCCTTGTACACAGTAATG GAAACACCTATTTACATTGGTACCTGCAGAAGCCAGGCCAGTCTCCAAAG CTCCTGATCTACAAAGTTTCCTACCGATTTTCTGGGGTCCCAGACAGGTT CAGTGGCAGTGGATCAGGGACAGATTTCACACTCAAGATCAGCAGAGTGG AGGCTGAGGATCTGGGAGTTTATTTCTGCTCTCAAAATACACATGTTCCT CGGACGTTCGGCCAAGGGACCAAGGTGGAAATCAAACGTACGGTGGCTGC ACCATCTGTCTTCATCTTCCCGCCATCTGATGAGCAGTTGAAATCTGGAA CTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTATCCCAGAGAGGCCAAA GTACAGTGGAAGGTGGATAACGCCCTCCAATCGGGTAACTCCCAGGAGAG TGTCACAGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCC TGACGCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGCGAA GTCACCCATCAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGG AGAGTGTTGA

Chimeric and Humanized Complete Heavy Chain Amino Acid Sequences

>VxP037-01-HC-Pro, below, represents a full length chimeric heavy chain variable domain (SEQ ID NO:57)+a constant domain amino acid sequence. The underlined amino acid sequence=framework 4+the constant IgG1 domain. All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as VxP037-01-HC-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.

Chimeric Complete Heavy Chain Amino Acid Sequence

>VxP037-01-HC-Pro (SEQ ID NO: 109) EVQLQQFGAELVKPGASMKLSCKASGYTFTNYYVFWVKQRPGQGLEWIGD INPVNGDTNFNEKFKNKATLTVDKSSTTTYLQLSSLTSEDSAVYYCTRGG YTMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN VNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTL MISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL PPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSD GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

Chimeric Complete Heavy Chain Nucleic Acid Sequence

The underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-01-HC-Pro, above.

>VxP037-01-HC-DNA (SEQ ID NO: 110) GAGGTCCAGCTGCAGCAGTTTGGGGCTGAACTGGTGAAGCCTGGGGCTTC AATGAAGTTGTCCTGCAAGGCTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGAAACAGAGGCCTGGACAAGGCCTTGAGTGGATTGGAGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAA GGCCACACTGACTGTAGACAAGTCCTCCACCACAACATACTTGCAACTCA GCAGCCTGACATCTGAGGACTCTGCGGTCTATTACTGTACAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGAACGCTGGTCACCGTCAGCTCAGC CTCCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCCTCCTCCAAGAGCA CCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCC GAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCA CACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCG TGGTGACCGTGCCCTCCAGCAGCTTGGGCACCCAGACCTACATCTGCAAC GTGAATCACAAGCCCAGCAACACCAAGGTGGACAAGAAAGTTGAGCCCAA ATCTTGTGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAACTCC TGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTC ATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCA CGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGC ATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGT GTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGA GTACAAGTGCAAGGTCAGCAACAAAGCCCTCCCAGCCCCCATCGAGAAAA CCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTG CCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGACCTGCCT GGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATG GGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGAC GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCA GCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACC ACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAATGA

Complete IgG1 N2970, IgG2, IgG4 S228P, and IgG4 S228P L235E Heavy Chain Amino Acid Sequences

>VxP037-01-HC-IgG1 N297Q-Pro, below, represents a full length heavy chain variable domain (SEQ ID NO:57)+a constant domain amino acid sequence. The underlined amino acid sequence=framework 4+the constant IgG1 constant domain containing a N-Q mutation at amino acid position 297. All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >VxP037-01-HC-IgG1 N297Q-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.

>VxP037-01-HC-IgG1-N297Q-Pro (SEQ ID NO: 111) EVQLQQFGAELVKPGASMKLSCKASGYTFTNYYVFWVKQRPGQGLEWIGD INPVNGDTNFNEKFKNKATLTVDKSSTTTYLQLSSLTSEDSAVYYCTRGG YTMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN VNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTL MISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYQSTYR VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL PPSR

>pVxK7b-037-hum01-HC-IgG2-Pro, below, represents a full length heavy chain variable domain (SEQ ID NO:58)+a constant domain amino acid sequence. The underlined amino acid sequence=framework 4+the constant IgG2 domain. All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >pVxK7b-037-hum01-HC-IgG2-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.

>pVxK7b-037-hum01-HC-IgG2-Pro (SEQ ID NO: 112) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCN VDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISR TPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSV LTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYNTTPPMLDSDGSFF LYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

>pVxK7b-037-hum01-HC-IgG4 S228P-Pro, below, represents a full length heavy chain variable domain (SEQ ID NO:58)+a constant domain amino acid sequence. The underlined amino acid sequence=framework 4+the constant IgG4 S228P domain. All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >pVxK7b-037-hum01-HC-IgG4 S228P-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.

>pVxK7b-037-hum01-HC-IgG4 S228P-Pro (SEQ ID NO: 113) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCN VDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMIS RTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS QEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF FLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK

>pVxK7b-037-hum01-HC-IgG4 S228P L235E-Pro, below, represents a full length heavy chain variable domain (SEQ ID NO:58)+a constant domain amino acid sequence. The underlined amino acid sequence=framework 4+the constant IgG4 S228P L235E domain. All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >pVxK7b-037-hum01-HC-IgG4 S228P-L235E-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.

>pVxK7b-037-hum01-HC-IgG4 S228P L235E-Pro (SEQ ID NO: 122) QVQLQESGPGLVKPSQTLSLTCTVSGYTFTNYYVFWVRQARGQRLEWIGD INPVNGDTNFNEKFKNRVTISADKSISTAYLQWSSLKASDTAMYYCARGG YTMDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCN VDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMIS RTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS QEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF FLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK

Complete IgG1 N2970, IgG2, IgG4 S228P, and IgG4 IgG4 S228P L235E Heavy Chain Nucleic Acid Sequences

The underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-01-HC-IgG1 N297Q-Pro, above.

>VxP037-01-HC-IgG1 N297Q-DNA (SEQ ID NO: 114) GAGGTCCAGCTGCAGCAGTTTGGGGCTGAACTGGTGAAGCCTGGGGCTTC AATGAAGTTGTCCTGCAAGGCTTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGAAACAGAGGCCTGGACAAGGCCTTGAGTGGATTGGAGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAA GGCCACACTGACTGTAGACAAGTCCTCCACCACAACATACTTGCAACTCA GCAGCCTGACATCTGAGGACTCTGCGGTCTATTACTGTACAAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGAACGCTGGTCACCGTCAGCTCAGC CTCCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCCTCCTCCAAGAGCA CCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCC GAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCA CACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCG TGGTGACCGTGCCCTCCAGCAGCTTGGGCACCCAGACCTACATCTGCAAC GTGAATCACAAGCCCAGCAACACCAAGGTGGACAAGAAAGTTGAGCCCAA ATCTTGTGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAACTCC TGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTC ATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCA CGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGC ATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACCAGAGCACGTACCGT GTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGA GTACAAGTGCAAGGTCAGCAACAAAGCCCTCCCAGCCCCCATCGAGAAAA CCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTG CCCCCATCCCGGGATGAGCTGACCAAGAACCAGGTCAGCCTGACCTGCCT GGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATG GGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGAC GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCA GCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACC ACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAATGA

The underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-hum 01-HC-IgG2-Pro, above.

>pVxK7b-037-hum01-HC-IgG2-DNA (SEQ ID NO: 115) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGAACGCTGGTCACCGTCAGCTCAGC CTCCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCCTGCTCCAGGAGCA CCTCCGAGAGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCC GAACCGGTGACGGTGTCGTGGAACTCAGGCGCTCTGACCAGCGGCGTGCA CACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCG TGGTGACCGTGCCCTCCAGCAACTTCGGCACCCAGACCTACACCTGCAAC GTAGATCACAAGCCCAGCAACACCAAGGTGGACAAGACAGTTGAGCGCAA ATGTTGTGTCGAGTGCCCACCGTGCCCAGCACCACCTGTGGCAGGACCGT CAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGG ACCCCTGAGGTCACGTGCGTGGTGGTGGACGTGAGCCACGAAGACCCCGA GGTCCAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGA CAAAGCCACGGGAGGAGCAGTTCAACAGCACGTTCCGTGTGGTCAGCGTC CTCACCGTCGTGCACCAGGACTGGCTGAACGGCAAGGAGTACAAGTGCAA GGTGTCCAACAAAGGCCTCCCAGCCCCCATCGAGAAAACCATCTCCAAAA CCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGG GAGGAGATGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTT CTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGA ACAACTACAACACCACACCTCCCATGCTGGACTCCGACGGCTCCTTCTTC CTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGT CTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGA AGAGCCTCTCCCTGTCTCCGGGTAAA

The underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-hum 01-HC-IgG4 S228P-Pro, above.

>pVxK7b-037-hum01-HC-IgG4 S228P-DNA (SEQ ID NO: 116) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGCACCACCGTGACCGTGTCCTCCGC TTCCACCAAGGGCCCATCCGTCTTCCCCCTGGCGCCCTGCTCCAGGAGCA CCTCCGAGAGCACAGCCGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCC GAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCA CACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCG TGGTGACCGTGCCCTCCAGCAGCTTGGGCACGAAGACCTACACCTGCAAC GTAGATCACAAGCCCAGCAACACCAAGGTGGACAAGAGAGTTGAGTCCAA ATATGGTCCCCCATGCCCACCGTGCCCAGCACCTGAGTTCCTGGGGGGAC CATCAGTCTTCCTGTTCCCCCCAAAACCCAAGGACACTCTCATGATCTCC CGGACCCCTGAGGTCACGTGCGTGGTGGTGGACGTGAGCCAGGAAGACCC CGAGGTCCAGTTCAACTGGTACGTGGATGGCGTGGAGGTGCATAATGCCA AGACAAAGCCGCGGGAGGAGCAGTTCAACAGCACGTACCGTGTGGTCAGC GTCCTCACCGTCCTGCACCAGGACTGGCTGAACGGCAAGGAGTACAAGTG CAAGGTGTCCAACAAAGGCCTCCCGTCCTCCATCGAGAAAACCATCTCCA AAGCCAAAGGGCAGCCCCGAGAGCCACAGGTGTACACCCTGCCCCCATCC CAGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGG CTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGG AGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC TTCCTCTACAGCAGGCTAACCGTGGACAAGAGCAGGTGGCAGGAGGGGAA TGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACAC AGAAGAGCCTCTCCCTGTCTCTGGGTAAA

The underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-hum 01-HC-IgG4 S228P L235E-Pro, above.

> pVxK7b-037-hum01-HC-IgG4 S228P L235E-DNA (SEQ ID NO: 123) CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAC CCTGTCCCTCACCTGCACTGTCTCTGGCTACACCTTCACCAACTACTATG TATTCTGGGTGCGACAGGCTCGTGGACAACGCCTTGAGTGGATAGGTGAC ATTAATCCTGTCAATGGTGATACTAACTTCAATGAGAAATTCAAGAACAG AGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAGTGGA GCAGCCTGAAGGCCTCGGACACCGCCATGTATTACTGTGCGAGAGGGGGT TATACTATGGACTACTGGGGCCAGGGCACCACCGTGACCGTGTCCTCCGC TTCCACCAAGGGCCCATCCGTCTTCCCCCTGGCGCCCTGCTCCAGGAGCA CCTCCGAGAGCACAGCCGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCC GAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCA CACCTTCCCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCG TGGTGACCGTGCCCTCCAGCAGCTTGGGCACGAAGACCTACACCTGCAAC GTAGATCACAAGCCCAGCAACACCAAGGTGGACAAGAGAGTTGAGTCCAA ATATGGTCCCCCATGCCCACCGTGCCCAGCACCTGAGTTCGAGGGGGGAC CATCAGTCTTCCTGTTCCCCCCAAAACCCAAGGACACTCTCATGATCTCC CGGACCCCTGAGGTCACGTGCGTGGTGGTGGACGTGAGCCAGGAAGACCC CGAGGTCCAGTTCAACTGGTACGTGGATGGCGTGGAGGTGCATAATGCCA AGACAAAGCCGCGGGAGGAGCAGTTCAACAGCACGTACCGTGTGGTCAGC GTCCTCACCGTCCTGCACCAGGACTGGCTGAACGGCAAGGAGTACAAGTG CAAGGTGTCCAACAAAGGCCTCCCGTCCTCCATCGAGAAAACCATCTCCA AAGCCAAAGGGCAGCCCCGAGAGCCACAGGTGTACACCCTGCCCCCATCC CAGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGG CTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGG AGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTC TTCCTCTACAGCAGGCTAACCGTGGACAAGAGCAGGTGGCAGGAGGGGAA TGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACAC AGAAGAGCCTCTCCCTGTCTCTGGGTAAA

Framework 4+Light Chain Constant Domain Amino Acid Sequence

(SEQ ID NO: 117) TKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVD NALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGL SSPVTKSFNRGEC

Framework 4+Heavy Chain Constant IgG1 Domain

(SEQ ID NO: 118) TLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNS GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTK VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVT CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLH QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKL TVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

Framework 4+Heavy Chain Constant IgG1 N297Q Domain

(SEQ ID NO: 119) TLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNS GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTK VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVT CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYQSTYRVVSVLTVLH QDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSR

Framework 4+Heavy Chain Constant IgG2 Domain

(SEQ ID NO: 120) WGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKP SNTKVDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVT CVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLTVVH QDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYNTTPPMLDSDGSFFLYSKL TVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

Framework 4+Heavy Chain Constant IgG4 S228P Domain

(SEQ ID NO: 121) TTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNS GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTK VDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVV VDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDW LNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQV SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVD KSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK

Framework 4+Heavy Chain Constant IgG4 S228P L235E Domain

(SEQ ID NO: 124) TTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNS GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTK VDKRVESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVV VDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDW LNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQV SLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVD KSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK

Claims

1. A monoclonal antibody, or antigen-binding fragment thereof, that:

(i) specifically binds human, rat, mouse, pig, cynomolgus monkey, and dog CD47;
(ii) blocks SIRPalpha and TSP1 binding to CD47;
(iii) promotes phagocytosis of cancer cells; and
(iv) reverses TSP1 inhibition of nitric oxide signaling.

2. The monoclonal antibody or antigen-binding fragment thereof of claim 1, which is chimeric or humanized.

3. The monoclonal antibody, or antigen-binding fragment thereof, of claim 2, which comprises three light chain complementarity determining regions (LCDRs 1-3) and three heavy chain complementarity determining regions (HCDRs 1-3), wherein: LCDR 1 comprises the amino acid sequence (SEQ ID NO: 1) RSSQSLVHSNGNTYLH LCDR 2 comprises the amino acid sequence (SEQ ID NO: 2) KVSYRFS; and LCDR 3 comprises the amino acid sequence (SEQ ID NO: 3) SQNTHVPRT; HCDR1 comprises the amino acid sequence (SEQ ID NO: 4) GYTFTNYYVF; HCDR 2 comprises the amino acid sequence (SEQ ID NO: 5) DINPVNGDTNFNEKFKN; and HCDR 3 comprises the amino acid sequence (SEQ ID NO: 6) GGYTMDY.

4. The monoclonal antibody, or antigen-binding fragment thereof of claim 3, which comprises a light chain variable region (LCVR) and a heavy chain variable region (HCVR), wherein said LCVR and said HCVR comprise, respectively, amino acid sequences selected from among the following combinations of LCVRs and HCVRs: wherein each one of LCVR SEQ ID NOs:7-31 further comprises a constant domain having the amino acid sequence shown in SEQ ID NO: 117, and wherein each one of HCVR SEQ ID NOs:57-81 comprises a constant domain selected from among SEQ ID NO:118, SEQ ID NO:119, SEQ ID NO:120, SEQ ID NO:121, and SEQ ID NO:124.

SEQ ID NO:7 and SEQ ID NO:57;
SEQ ID NO:8 and SEQ ID NO:58;
SEQ ID NO:9 and SEQ ID NO:59;
SEQ ID NO:10 and SEQ ID NO:60;
SEQ ID NO:11 and SEQ ID NO:61;
SEQ ID NO:12 and SEQ ID NO:62;
SEQ ID NO:13 and SEQ ID NO:63;
SEQ ID NO:14 and SEQ ID NO:64;
SEQ ID NO:15 and SEQ ID NO:65;
SEQ ID NO:16 and SEQ ID NO:66;
SEQ ID NO:17 and SEQ ID NO:67;
SEQ ID NO:18 and SEQ ID NO:68;
SEQ ID NO:19 and SEQ ID NO:69;
SEQ ID NO:20 and SEQ ID NO:70;
SEQ ID NO:21 and SEQ ID NO:71;
SEQ ID NO:22 and SEQ ID NO:72;
SEQ ID NO:23 and SEQ ID NO:73;
SEQ ID NO:24 and SEQ ID NO:74;
SEQ ID NO:25 and SEQ ID NO:75;
SEQ ID NO:26 and SEQ ID NO:76;
SEQ ID NO:27 and SEQ ID NO:77;
SEQ ID NO:28 and SEQ ID NO:78;
SEQ ID NO:29 and SEQ ID NO:79;
SEQ ID NO:30 and SEQ ID NO:80; and
SEQ ID NO:31 and SEQ ID NO:81,

5. A monoclonal antibody, or antigen-binding fragment thereof, that competes with said monoclonal antibody or antigen binding fragment thereof of claim 4 for binding to human CD47.

6. A pharmaceutical composition, comprising said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 1 and a pharmaceutically or physiologically acceptable carrier, diluent, or excipient.

7. A monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 1 for use in human therapy.

8. The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof of claim 1 for use in reducing, preventing, and/or treating ischemia-reperfusion injury, or an autoimmune or inflammatory disease in a human patient.

9. The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 8, wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of an appendage or other body part, skin grafting, or trauma.

10. The monoclonal antibody, or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 8, wherein said autoimmune or inflammatory disease is selected from the group consisting of arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

11. The monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof of claim 1 for use in treating or reducing a susceptible cancer.

12. The monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 11, which promotes phagocytosis of cells of said susceptible cancer.

13. The monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 11 wherein said susceptible cancer is selected from the group consisting of a leukemia, a lymphoma, ovarian cancer, breast cancer, endometrial cancer, colon cancer (colorectal cancer), rectal cancer, bladder cancer, lung cancer (non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung), bronchial cancer, bone cancer, prostate cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, gall bladder cancer, bile duct cancer, esophageal cancer, renal cell carcinoma, thyroid cancer, squamous cell carcinoma of the head and neck (head and neck cancer), testicular cancer, cancer of the endocrine gland, cancer of the adrenal gland, cancer of the pituitary gland, cancer of the skin, cancer of soft tissues, cancer of blood vessels, cancer of brain, cancer of nerves, cancer of eyes, cancer of meninges, cancer of oropharynx, cancer of hypopharynx, cancer of cervix, and cancer of uterus, glioblastoma, meduloblastoma, astrocytoma, glioma, meningioma, gastrinoma, neuroblastoma, melanoma, myelodysplastic syndrome, and a sarcoma.

14. The monoclonal antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 13 wherein:

said leukemia is selected from the group consisting of systemic mastocytosis, acute lymphocytic (lymphoblastic) leukemia (ALL), T cell—ALL, acute myeloid leukemia (AML), myelogenous leukemia, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), chronic myeloid leukemia (CML), myeloproliferative disorder/neoplasm, myelodysplastic syndrome, monocytic cell leukemia, and plasma cell leukemia;
wherein said lymphoma is selected from the group consisting of histiocytic lymphoma and T cell lymphoma, B cell lymphomas, including Hodgkin's lymphoma and non-Hodgkin's lymphoma, such as low grade/follicular non-Hodgkin's lymphoma (NHL), cell lymphoma (FCC), mantle cell lymphoma (MCL), diffuse large cell lymphoma (DLCL), small lymphocytic (SL) NHL, intermediate grade/follicular NHL, intermediate grade diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease NHL, and Waldenstrom's Macroglobulinemia; and
wherein said sarcoma is selected from the group consisting of osteosarcoma, Ewing sarcoma, leiomyosarcoma, synovial sarcoma, alveolar soft part sarcoma, angiosarcoma, liposarcoma, fibrosarcoma, rhabdomyosarcoma, and chrondrosarcoma.

15. Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 1 to reduce, prevent, and/or treat ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human patient.

16. Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 1 to reduce, prevent, and/or treat a susceptible cancer.

17. Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 1 for the manufacture of a medicament to prevent, reduce, and/or treat ischemia-reperfusion injury, or an autoimmune or inflammatory disease, in a human patient.

18. The use of claim 17, wherein said ischemia-reperfusion injury occurs in organ transplantation, acute kidney injury, cardiopulmonary bypass surgery, pulmonary hypertension, sickle cell disease, myocardial infarction, stroke, surgical resections and reconstructive surgery, reattachment of an appendage or other body part, skin grafting, and trauma.

19. The use of claim 17, wherein said autoimmune or inflammatory disease is selected from among arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory bowel disease, lupus, Grave's disease and Hashimoto's thyroiditis, and ankylosing spondylitis.

20. Use of said monoclonal antibody or antigen-binding fragment thereof, or competing antibody or antigen binding fragment thereof, of claim 1 for the manufacture of a medicament to treat or reduce a susceptible cancer.

21. A monoclonal antibody, or antigen-binding fragment thereof, of claim 1 that specifically binds to epitopes in the extracellular IgV domain of CD47.

22. A monoclonal antibody, or antigen-binding fragment thereof, of claim 5 that specifically binds to epitopes in the extracellular IgV domain of CD47.

Patent History
Publication number: 20150274826
Type: Application
Filed: Jun 11, 2015
Publication Date: Oct 1, 2015
Inventors: William A. FRAZIER (St. Louis, MO), Pamela T. MANNING (Chesterfield, MO), Gerhard FREY (St. Louis, MO), Hwai Wen CHANG (St. Louis, MO)
Application Number: 14/737,053
Classifications
International Classification: C07K 16/28 (20060101);