Methods and compositions for reducing immunogenicity

文档序号:1651929 发布日期:2019-12-24 浏览:25次 中文

阅读说明:本技术 用于降低免疫原性的方法和组合物 (Methods and compositions for reducing immunogenicity ) 是由 杰弗里·C·约翰逊 劳伦斯·迪尔思 哈拉兰博斯·哈德杰瓦斯林 孙正勋 坎德萨米·哈里哈兰 于 2018-03-26 设计创作,主要内容包括:本文提供涉及抗CD20抗体例如利妥昔单抗(rituximab)与蛋白质治疗剂例如抗体(例如,特异性结合人CD47的抗体)的组合的方法和用途。(Provided herein are methods and uses involving anti-CD 20 antibodies, such as rituximab (rituximab), in combination with protein therapeutics, such as antibodies (e.g., antibodies that specifically bind human CD 47).)

1. A method of reducing immunogenicity in a subject, the method comprising administering to the subject a combination of rituximab and a protein therapeutic, wherein the immunogenicity is reduced compared to the immunogenicity in the subject when the protein therapeutic is administered alone.

2. The method of claim 1, wherein the protein therapeutic is an antibody therapeutic.

3. The method of claim 1, wherein the protein therapeutic is a cytokine.

4. The method of claim 1, wherein the protein therapeutic is an interleukin.

5. The method of claim 1, wherein the protein therapeutic is not an enzyme.

6. The method of claim 2, wherein the antibody therapeutic is an antibody or antigen-binding fragment thereof that binds CD 47.

7. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a variable heavy chain (VH) Complementarity Determining Region (CDR)1 comprising SEQ ID NO 50, a VH CDR2 comprising SEQ ID NO 72, a VH CDR3 sequence comprising SEQ ID NO 52, a variable light chain (VL) CDR1 comprising SEQ ID NO 53, a VLCDR2 comprising SEQ ID NO 71, and a VL CDR3 comprising SEQ ID NO 55.

8. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30.

9. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47.

10. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30 and a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47.

11. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 51, a VH CDR3 comprising SEQ ID No. 52, a VL CDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 54, and a VL CDR3 comprising SEQ ID No. 55.

12. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from the group consisting of IgG1 isotype, IgG2 isotype, IgG3 isotype, and IgG4 isotype.

13. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from IgG4P and IgG4 PE.

14. The method of claim 6, wherein the antibody or antigen-binding fragment thereof that binds CD47 is a component of a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof that binds CD47 and a pharmaceutically acceptable carrier.

15. The method of any one of claims 6-14, wherein the antibody is chimeric, humanized, or fully human.

16. The method of any one of claims 1-15, wherein the subject is a human.

17. The method of any one of claims 1-15, further comprising administering chemotherapy.

18. The method of claim 17, wherein the chemotherapy is radiation therapy.

19. The method of any one of claims 7-17, wherein the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg.

20. The method of any one of claims 1-19, wherein the rituximab is administered at 300, 325, 350, 375, 400, 425, 450, or 500mg/m2Is administered to the subject.

21. The method of any one of claims 7-20, wherein the rituximab is administered prior to the antibody or antigen-binding fragment thereof that binds CD 47.

22. The method of any one of claims 1-21, wherein the method does not comprise administering a proteasome inhibitor to the subject.

23. The method of claim 22, wherein the method does not comprise administering bortezomib to the subject.

24. The method of any one of claims 1-23, wherein the method does not comprise administering methotrexate to the subject.

25. A method of treating cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof that binds CD47, wherein the method further comprises administering rituximab to the subject.

26. The method of claim 25, wherein the rituximab is administered prior to the antibody or antigen-binding fragment thereof that binds CD 47.

27. The method of claim 25 or 26, further comprising administering radiation or chemotherapy.

28. The method of any one of claims 25-27, further comprising administering another anti-cancer agent.

29. The method of any one of claims 25-28, wherein the cancer is a hematologic cancer.

30. The method of any one of claims 25-28, wherein the cancer is a solid cancer.

31. The method of any one of claims 25-28, wherein the cancer is multiple myeloma, non-hodgkin's lymphoma, Acute Myeloid Leukemia (AML), breast cancer, bladder cancer, non-small cell lung cancer, hepatocellular carcinoma (HCC), sarcoma, or head and neck cancer.

32. The method of claim 31, wherein the cancer is non-hodgkin's lymphoma.

33. The method of claim 32, wherein the non-hodgkin's lymphoma is CD20 positive.

34. The method of claim 31 or 32, wherein the non-hodgkin's lymphoma is relapsed or refractory.

35. The method of any one of claims 25-34, wherein the subject has been previously treated with rituximab.

36. The method of any one of claims 25-35, wherein the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg.

37. The method of any one of claims 25-36, wherein the rituximab is administered at 300, 325, 350, 375, 400, 425, 450, or 500mg/m2Is administered to the subject.

38. The method of any one of claims 25-37, wherein the method does not comprise administering a proteasome inhibitor to the subject.

39. The method of claim 38, wherein the method does not comprise administering bortezomib to the subject.

40. The method of any one of claims 25-39, wherein the method does not comprise administering methotrexate to the subject.

41. The method of any one of claims 25-40, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 72, a VHCDR3 sequence comprising SEQ ID No. 52, a variable light chain (VL) CDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 71, and a VL CDR3 comprising SEQ ID No. 55.

42. The method of any one of claims 25-40, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30.

43. The method of any one of claims 25-40, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47.

44. The method of any one of claims 25-40, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30 and a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47.

45. The method of any one of claims 25-40, wherein the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 51, a VHCDR3 comprising SEQ ID No. 52, a VL CDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 54, and a VL CDR3 comprising SEQ ID No. 55.

46. The method of any one of claims 25-45, wherein the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from the group consisting of IgG1 isotype, IgG2 isotype, IgG3 isotype, and IgG4 isotype.

47. The method of any one of claims 25-45, wherein the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from IgG4P and IgG4 PE.

48. The method of any one of claims 25-47, wherein the antibody or antigen-binding fragment thereof that binds CD47 is a component of a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof that binds CD47 and a pharmaceutically acceptable carrier.

49. The method of any one of claims 25-48, wherein the antibody is chimeric, humanized, or fully human.

50. The method of any one of claims 25-49, wherein the subject is a human.

1. Field of the invention

Provided herein are methods and uses involving anti-CD 20 antibodies, such as rituximab (rituximab), in combination with protein therapeutics, such as antibodies (e.g., antibodies that specifically bind human CD 47).

2. Background of the invention

While protein therapeutics have been used to treat a number of diseases, they may promote an immune response involving the production of anti-drug antibodies when administered to a subject. This may result in reduced efficacy and/or toxicity of the therapeutic agent. Thus, there is a need for methods of reducing such immune responses.

CD47, also known as integrin-associated protein (IAP), ovarian cancer antigen OA3, Rh-associated antigen and MER6, are multi-spanning transmembrane receptors belonging to the immunoglobulin superfamily. Sirpa (signal-regulatory protein a) expressed on macrophages interacts with CD47, and this interaction negatively controls effector functions of innate immune cells, such as host cell phagocytosis. CD47 expression and/or activity is associated with a number of diseases and disorders. Thus, there is a need for therapies targeting CD 47.

3. Summary of the invention

Provided herein are methods and compositions for reducing immunogenicity in a subject, comprising administering to the subject an anti-CD 20 antibody (e.g., rituximab) in combination with a protein therapeutic, wherein the immunogenicity is reduced compared to the immunogenicity in the subject when the protein therapeutic is administered alone. In certain aspects, the protein therapeutic is an antibody therapeutic. In certain aspects, the protein therapeutic is a fusion protein, e.g., an Fc-containing fusion protein, e.g., a soluble receptor fusion protein. In certain aspects, the protein therapeutic is a cytokine. In certain aspects, the protein therapeutic is an interleukin. In certain aspects, the protein therapeutic is not an enzyme. In certain aspects, the subject is a human.

In certain aspects of the methods and compositions provided herein, the protein therapeutic is an antibody, wherein the antibody therapeutic is an antibody or antigen-binding fragment thereof that binds CD 47.

In particular aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 72, a VH CDR3 comprising SEQ ID No. 52, a VLCDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 71, and a VL CDR3 comprising SEQ ID No. 55.

In particular aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 51, a VH CDR3 comprising SEQ ID No. 52, a VLCDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 54, and a VL CDR3 comprising SEQ ID No. 55.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30 and a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from the group consisting of IgG1 isotype, IgG2 isotype, IgG3 isotype, and IgG4 isotype. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from IgG4P and IgG4 PE.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is a component of a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof that binds CD47 and a pharmaceutically acceptable carrier.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is a chimeric, humanized, or fully human antibody.

In certain aspects, the methods provided herein further comprise administering a second therapeutic agent, e.g., a small molecule therapeutic agent, such as a chemotherapeutic therapeutic agent. In a particular aspect, the chemotherapy is radiation therapy.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered at 300, 325, 350, 375, 400, 425, 450, or 500mg/m2Is administered to the subject. In certain aspects, the anti-CD 20 antibody is rituximab. In certain aspects, the CD 47-binding antibodiesThe body or antigen-binding fragment thereof is administered to a subject at a dose of 0.3, 1, 2, 4,8, 15, or 20mg/kg, and the anti-CD 20 antibody, e.g., rituximab, is at 300, 325, 350, 375, 400, 425, 450, or 500mg/m2Is administered to the subject. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered prior to the protein therapeutic. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered 1, 2,3, 4,5, or 6 weeks prior to the protein therapeutic. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered 1, 2,3, 4,5, or 6 days prior to the protein therapeutic. In certain aspects, rituximab is administered prior to an antibody or antigen-binding fragment thereof that binds CD 47. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered 1, 2,3, 4,5, or 6 weeks prior to the antibody or antigen-binding fragment thereof that binds CD 47. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered 1, 2,3, 4,5, or 6 days prior to the antibody or antigen-binding fragment thereof that binds CD 47.

In certain aspects, the methods provided herein do not comprise administering a proteasome inhibitor to the subject. In certain aspects, the methods provided herein do not comprise administering bortezomib to the subject. In certain aspects, the methods provided herein do not include administering methotrexate to the subject.

Provided herein are methods for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof that binds CD47, wherein the method further comprises administering to the subject an anti-CD 20 antibody, e.g., rituximab. In certain aspects, the anti-CD 20 antibody is rituximab. In certain aspects, the subject is a human. In certain aspects, the methods provided herein further comprise administering radiation or chemotherapy. In certain aspects, the methods provided herein further comprise administering another anti-cancer agent. In certain aspects, the cancer is a hematologic cancer. In certain aspects, the cancer is a solid cancer. In certain aspects, the cancer is multiple myeloma, non-hodgkin's lymphoma, Acute Myeloid Leukemia (AML), breast cancer (e.g., triple negative breast cancer), bladder cancer, non-small cell lung cancer (non-small cell lung cancer/carcinosa), hepatocellular carcinoma (HCC), sarcoma, or head and neck cancer. In certain aspects, the cancer is multiple myeloma. In certain aspects, the cancer is non-hodgkin's lymphoma. In particular aspects, the non-hodgkin lymphoma is CD20 positive. In particular aspects, the non-hodgkin's lymphoma is relapsed or refractory. In particular aspects, the subject has previously received a treatment regimen comprising an anti-CD 20 antibody (e.g., rituximab).

Provided herein are methods for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof that binds CD 47. In certain aspects, the subject is a human. In certain aspects, the methods provided herein further comprise administering radiation or chemotherapy. In certain aspects, the methods provided herein further comprise administering another anti-cancer agent. In certain aspects, the cancer is a hematologic cancer. In certain aspects, the cancer is a solid cancer. In certain aspects, the cancer is multiple myeloma, non-hodgkin's lymphoma, Acute Myeloid Leukemia (AML), breast cancer (e.g., triple negative breast cancer), bladder cancer, non-small cell lung cancer (non-small cell lung cancer/carcinosa), hepatocellular carcinoma (HCC), sarcoma, or head and neck cancer. In certain aspects, the cancer is multiple myeloma. In certain aspects, the cancer is non-hodgkin's lymphoma. In particular aspects, the non-hodgkin lymphoma is CD20 positive. In particular aspects, the non-hodgkin's lymphoma is relapsed or refractory. In particular aspects, the subject has previously received a treatment regimen comprising an anti-CD 20 antibody (e.g., rituximab).

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered at 300, 325, 350, 375, 400, 425, 450, or 500mg/m2Is administered to the subject. In certain aspects, the anti-CD 20 antibody, e.g., rituximab, is administered prior to the antibody or antigen-binding fragment thereof that binds CD 47.

In certain aspects, the method does not comprise administering a proteasome inhibitor to the subject. In certain aspects, the method does not comprise administering bortezomib to the subject. In certain aspects, the method does not comprise administering methotrexate to the subject.

In certain aspects of the methods of treating cancer provided herein, the protein therapeutic is an antibody, wherein the antibody therapeutic is an antibody or antigen-binding fragment thereof that binds CD 47.

In particular aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 72, a VH CDR3 comprising SEQ ID No. 52, a VLCDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 71, and a VL CDR3 comprising SEQ ID No. 55.

In particular aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH CDR1 comprising SEQ ID No. 50, a VH CDR2 comprising SEQ ID No. 51, a VH CDR3 comprising SEQ ID No. 52, a VLCDR1 comprising SEQ ID No. 53, a VL CDR2 comprising SEQ ID No. 54, and a VL CDR3 comprising SEQ ID No. 55.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 comprises a VH comprising a sequence selected from the group consisting of SEQ ID NOs 5-30 and a VL comprising a sequence selected from the group consisting of SEQ ID NOs 31-47.

In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from the group consisting of IgG1 isotype, IgG2 isotype, IgG3 isotype, and IgG4 isotype. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is an IgG isotype selected from IgG4P and IgG4 PE. In certain aspects, the antibody or antigen-binding fragment thereof that binds CD47 is chimeric, humanized, or fully human.

4. Detailed description of the preferred embodiments

4.1 method

4.1.1 methods for reducing immunogenicity

Provided herein are methods for reducing immunogenicity in a subject comprising administering to the subject an anti-CD 20 antibody (e.g., rituximab) in combination with a protein therapeutic, wherein the immunogenicity is reduced compared to the immunogenicity in the subject when the protein therapeutic is administered alone. In certain embodiments, the protein therapeutic is an antibody therapeutic. In certain embodiments, the protein therapeutic is a cytokine. In certain embodiments, the cytokine is Bone Morphogenic Protein (BMP), Erythropoietin (EPO), granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony stimulating factor (GM-CSF), interferon alpha (IFN-alpha), interferon beta (IFN-beta), interleukin 2(IL-2), interleukin 11(IL-11), or interferon gamma (IFN-gamma). In certain embodiments, the protein therapeutic is an interleukin. In certain embodiments, the protein therapeutic is not an enzyme.

In certain embodiments of the methods provided herein, the protein therapeutic is an antibody therapeutic, wherein the antibody therapeutic is an antibody or antigen-binding fragment thereof that binds CD 47.

In certain embodiments, an antibody or antigen-binding fragment thereof that binds CD47 is a component of a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof that binds CD47 and a pharmaceutically acceptable carrier.

In certain embodiments, the methods provided herein comprise administering chemotherapy. In a specific embodiment, the chemotherapy is radiation therapy.

In certain embodiments, the anti-CD 20 antibody, e.g., rituximab, is administered prior to and/or concurrently with the protein therapeutic. In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered prior to the protein therapeutic. In certain embodiments, the anti-CD 20 antibody, e.g., rituximab, is administered concurrently with the protein therapeutic.

In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered prior to and/or concurrently with an antibody or antigen-binding fragment thereof that binds CD 47. In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered prior to the antibody or antigen-binding fragment thereof that binds CD 47. In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered concurrently with an antibody or antigen-binding fragment thereof that binds CD 47.

In certain embodiments, the methods provided herein do not comprise administering a proteasome inhibitor to the subject. In certain embodiments, the methods provided herein do not comprise administering bortezomib to the subject. In certain embodiments, the methods provided herein do not include administering methotrexate to the subject.

Immunogenicity may be measured by any method known to those skilled in the art. In certain embodiments, immunogenicity is measured by determining the amount and/or concentration of anti-drug antibodies present in serum. In certain embodiments, immunogenicity is measured by determining the titer of anti-drug antibodies present in serum. In certain embodiments, immunogenicity is measured by determining the amount of protein therapeutic agent neutralized per volume of serum. In certain embodiments, the presence of immunogenicity is indicated by the occurrence of an allergic reaction, cytokine release syndrome, infusion reaction, delayed-type hypersensitivity reaction, and/or cross-reaction with endogenous proteins. In certain embodiments, immunogenicity is measured by a screening assay. In particular embodiments, the screening assay is a direct-binding enzyme-linked immunosorbent assay (ELISA), a bridge ELISA, a radioimmunoprecipitation assay (RIPA), a Surface Plasmon Resonance (SPR) assay, a Bethesda assay, or a bridge electrochemiluminescence assay. In certain embodiments, immunogenicity is measured by a neutralization assay. In particular embodiments, the neutralization assay is a cell-based bioassay or a non-cell based competitive ligand binding assay. In certain embodiments, the anti-drug antibody binds to a protein therapeutic. In certain embodiments, the anti-drug antibody neutralizes the protein therapeutic. In certain embodiments, the anti-drug antibody binds to and neutralizes the protein therapeutic.

In certain aspects, immunogenicity is measured one week after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured two weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured three weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured four weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured five weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured six weeks after the first dose of protein therapeutic.

In certain aspects, immunogenicity is measured weekly after a first dose of a protein therapeutic. In certain aspects, immunogenicity is measured one week after and one week after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured two weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, immunogenicity is measured three weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, immunogenicity is measured four weeks after and weekly after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured five weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, immunogenicity is measured six weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, immunogenicity is measured every other week after the first dose of protein therapeutic.

In certain aspects, the B cell count is measured one week after the first dose of the protein therapeutic. In certain aspects, the B cell count is measured two weeks after the first dose of protein therapeutic. In certain aspects, B cell counts are measured three weeks after the first dose of protein therapeutic. In certain aspects, the B cell count is measured four weeks after the first dose of the protein therapeutic. In certain aspects, B cell counts are measured five weeks after the first dose of protein therapeutic. In certain aspects, B cell counts are measured six weeks after the first dose of protein therapeutic.

In certain aspects, the B cell count is measured one week after the first dose of the protein therapeutic. In certain aspects, the B cell count is measured two weeks after the first dose of protein therapeutic. In certain aspects, B cell counts are measured three weeks after the first dose of protein therapeutic. In certain aspects, the B cell count is measured four weeks after the first dose of the protein therapeutic. In certain aspects, B cell counts are measured five weeks after the first dose of protein therapeutic. In certain aspects, B cell counts are measured six weeks after the first dose of protein therapeutic.

In certain aspects, the B cell count is measured weekly after the first dose of the protein therapeutic. In certain aspects, the B cell count is measured one week after and every week after the first dose of the protein therapeutic. In certain aspects, the B cell count is measured two weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, B cell counts are measured three weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, the B cell count is measured four weeks after and weekly after the first dose of the protein therapeutic. In certain aspects, B cell counts are measured five weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, B cell counts are measured six weeks after the first dose of protein therapeutic and weekly thereafter. In certain aspects, B cell counts are measured every other week after the first dose of protein therapeutic.

In certain aspects, immunogenicity is measured one week after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured two weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured three weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured four weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured five weeks after the first dose of protein therapeutic. In certain aspects, immunogenicity is measured six weeks after the first dose of protein therapeutic.

In certain aspects, immunogenicity is measured weekly after a first dose of an anti-CD 20 antibody, e.g., rituximab. In certain aspects, the anti-CD 20 antibody is rituximab. In certain aspects, immunogenicity is measured one week after the first dose of rituximab and weekly thereafter. In certain aspects, immunogenicity is measured two weeks after the first dose of rituximab and weekly thereafter. In certain aspects, immunogenicity is measured three weeks after the first dose of rituximab and weekly thereafter. In certain aspects, immunogenicity is measured four weeks after the first dose of rituximab and weekly thereafter. In certain aspects, immunogenicity is measured five weeks after the first dose of rituximab and weekly thereafter. In certain aspects, immunogenicity is measured six weeks after the first dose of rituximab and weekly thereafter. In certain aspects, immunogenicity is measured every other week after the first dose of rituximab.

In certain aspects, B cell counts are measured one week after the first dose of an anti-CD 20 antibody, e.g., rituximab. In certain aspects, the anti-CD 20 antibody is rituximab. In certain aspects, B cell counts are measured two weeks after the first dose of rituximab. In certain aspects, B cell counts are measured three weeks after the first dose of rituximab. In certain aspects, B cell counts are measured four weeks after the first dose of rituximab. In certain aspects, B cell counts are measured five weeks after the first dose of rituximab. In certain aspects, B cell counts are measured six weeks after the first dose of rituximab.

In certain aspects, B cell counts are measured one week after the first dose of an anti-CD 20 antibody, e.g., rituximab. In certain aspects, the anti-CD 20 antibody is rituximab. In certain aspects, B cell counts are measured two weeks after the first dose of rituximab. In certain aspects, B cell counts are measured three weeks after the first dose of rituximab. In certain aspects, B cell counts are measured four weeks after the first dose of rituximab. In certain aspects, B cell counts are measured five weeks after the first dose of rituximab. In certain aspects, B cell counts are measured six weeks after the first dose of rituximab.

In certain aspects, B cell counts are measured weekly after a first dose of an anti-CD 20 antibody, e.g., rituximab. In certain aspects, the anti-CD 20 antibody is rituximab. In certain aspects, B cell counts are measured one week after the first dose of rituximab and weekly thereafter. In certain aspects, B cell counts are measured two weeks after the first dose of rituximab and weekly thereafter. In certain aspects, B cell counts are measured three weeks after the first dose of rituximab and weekly thereafter. In certain aspects, B cell counts are measured four weeks after the first dose of rituximab and weekly thereafter. In certain aspects, B cell counts are measured five weeks after the first dose of rituximab and weekly thereafter. In certain aspects, B cell counts are measured six weeks after the first dose of rituximab and weekly thereafter. In certain aspects, B cell counts are measured every other week after the first dose of rituximab.

4.1.2 methods of treating cancer

Provided herein are methods for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof that binds CD47, wherein the method further comprises administering to the subject an anti-CD 20 antibody, e.g., rituximab. In certain embodiments, the anti-CD 20 antibody is rituximab. Also provided herein are methods for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof that binds CD 47.

In certain embodiments, the methods for treating cancer provided herein further comprise administering radiation or chemotherapy. In certain embodiments, the methods provided herein further comprise administering another anti-cancer agent. In certain embodiments, the cancer is a hematologic cancer. In certain embodiments, the cancer is a solid cancer. In certain embodiments, the cancer is multiple myeloma, non-hodgkin's lymphoma, Acute Myeloid Leukemia (AML), breast cancer (e.g., triple negative breast cancer), bladder cancer, non-small cell lung cancer, hepatocellular carcinoma (HCC), sarcoma, or head and neck cancer. In particular embodiments, the cancer is non-hodgkin's lymphoma. In particular embodiments, the non-hodgkin lymphoma is CD20 positive. In particular embodiments, the non-hodgkin's lymphoma is relapsed or refractory. In particular embodiments, the subject has been previously treated with an anti-CD 20 antibody, such as rituximab.

In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered prior to and/or concurrently with an antibody or antigen-binding fragment thereof that binds CD 47. In certain embodiments, the anti-CD 20 antibody is rituximab. In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered prior to the antibody or antigen-binding fragment thereof that binds CD 47. In certain embodiments, an anti-CD 20 antibody, e.g., rituximab, is administered concurrently with an antibody or antigen-binding fragment thereof that binds CD 47.

In certain embodiments, the method does not comprise administering a proteasome inhibitor to the subject. In certain embodiments, the method does not comprise administering bortezomib to the subject. In certain embodiments, the method does not comprise administering methotrexate to the subject. In certain embodiments, the method further comprises administering methotrexate to the subject.

In certain embodiments, an antibody or antigen-binding fragment thereof that binds CD47 is a component of a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof that binds CD47 and a pharmaceutically acceptable carrier.

In certain embodiments, provided herein are methods of using an anti-CD 47 antibody described herein, alone or in combination with an anti-CD 20 antibody (e.g., rituximab), to prevent a condition or disorder, such as cancer.

In particular embodiments, provided herein are methods for managing, treating, preventing, or preventing cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment described herein that specifically binds to CD47 (e.g., human CD47), alone or in combination with an anti-CD 20 antibody (e.g., rituximab). In certain embodiments, provided herein are methods of ameliorating, inhibiting, or reducing the progression or severity of one or more symptoms associated with cancer.

As used herein, "administering" refers to the act of injecting or otherwise physically delivering a substance (e.g., an anti-CD 20 antibody (e.g., rituximab), or an anti-CD 47 antibody or antigen-binding fragment thereof provided herein) to a subject or patient (e.g., a human), for example, by mucosal, topical, intradermal, parenteral, intravenous, subcutaneous, intramuscular delivery, and/or any other physical delivery method described herein or known in the art. In certain embodiments, the anti-CD 20 antibody, e.g., rituximab, is administered intravenously. In certain embodiments, the anti-CD 47 antibodies provided herein are administered intravenously. In certain embodiments, rituximab and the anti-CD 47 antibodies provided herein are administered intravenously.

As used herein, the term effective amount refers to an amount of a composition (e.g., an antibody or pharmaceutical composition provided herein, or an anti-CD 20 antibody (e.g., rituximab), or a pharmaceutical composition provided herein) sufficient to achieve a particular readout, e.g., reduce and/or ameliorate the severity and/or duration of a given condition, disorder, or disease (e.g., cancer, metastasis, or angiogenesis) and/or symptoms associated therewith. This term also encompasses an amount necessary to reduce, slow or ameliorate progression or progression of a given disease, reduce, slow or ameliorate recurrence, development or onset of a given disease, and/or ameliorate or enhance one or more prophylactic or therapeutic effects of another therapy (e.g., a therapy other than an anti-CD 47 antibody provided herein). In some embodiments, an "effective amount" as used herein refers to an amount of an anti-CD 20 antibody (e.g., rituximab) that is associated with reducing the immunogenicity of a protein therapeutic as described herein.

For example, a dose of anti-CD 20 antibody (e.g., rituximab) to be administered in combination with a protein therapeutic agent, such as an anti-CD 47 antibody, may include about 0.1mg/kg body weight to about 100mg/kg body weight. For example, a dose of rituximab to be administered in combination with a protein therapeutic agent (such as an anti-CD 47 antibody) may comprise about 25mg/m2To about 1500mg/m2. In some embodiments, rituximab is administered to a subject at a dose of 0.1mg/kg, 0.5mg/kg, 1mg/kg, 2mg/kg, 5mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 25mg/kg, 30mg/kg, 50mg/kg, 75mg/kg, 100mg/kg, or more. In certain embodiments, rituximab is administered to a subject at a dose of 0.1, 0.3, 0.5, 1, 2, 4,5, 8, 10, 15, 20, 25, 30, 50, 75, or 100 mg/kg. In certain embodiments, rituximab is administered to a subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In some embodiments, rituximab is administered at 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 750, 1000, 1250, 1500mg/m2Or higher doses, are administered to the subject. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject.

The frequency of administration of the anti-CD 20 antibody (e.g., rituximab) can range, for example, from twice a day to once a month. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once a week. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once every two weeks. In certain embodiments, rituximab is administered at 300, 325, 350, 375, 400, 425, 450, 475Or 500mg/m2Is administered to the subject once every four weeks. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once a week. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once every two weeks. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once every four weeks.

For example, a dose of anti-CD 47 antibody or antigen-binding fragment thereof to be administered in combination with an anti-CD 20 antibody (e.g., rituximab) may include about 0.1mg/kg body weight to about 100mg/kg body weight. In some embodiments, the anti-CD 47 antibody is administered to the subject at a dose of 0.1mg/kg, 0.5mg/kg, 1mg/kg, 2mg/kg, 5mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 25mg/kg, 30mg/kg, 50mg/kg, 75mg/kg, 100mg/kg or more. In certain embodiments, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject at a dose of 0.1, 0.3, 0.5, 1, 2, 4,5, 8, 10, 15, 20, 25, 30, 50, 75, or 100 mg/kg. In certain embodiments, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg.

The frequency of administration of the anti-CD 47 antibody or antigen-binding fragment thereof can range, for example, from twice daily to once weekly. In certain embodiments, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject at a dose of 0.3, 1, 2, 4,8, 15, or 20mg/kg once weekly. In certain embodiments, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject twice weekly at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject twice weekly at a dose of 8 mg/kg. In certain embodiments, the antibody or antigen-binding fragment thereof that binds CD47 is administered to a subject once weekly at a dose of 20 mg/kg.

In some embodiments, the anti-CD 20 antibody (e.g., rituximab) is administered at 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 750, 1000, 1250, 1500mg/m2Is administered to a subject and binds to CD47, or an antigen-binding fragment thereof, is administered to a subject at a dose of 0.1, 0.3, 0.5, 1, 2, 4,5, 8, 10, 15, 20, 25, 30, 50, 75, or 100 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2And the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 375mg/m2And the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once a week, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject once a week at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg.

In certain embodiments, the anti-CD 20 antibody (e.g., rituximab) is administered at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 8 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 8 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject weekly at a dose of 20 mg/kg.

In certain embodiments, an anti-CD 20 antibody (e.g., rituximab)At 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 8 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 8 mg/kg. In certain embodiments, rituximab is at 300, 325, 350, 375, 400, 425, 450, 475, or 500mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject once a week at a dose of 20 mg/kg.

In certain embodiments, the anti-CD 20 antibody (e.g., rituximab) is at 375mg/m2Is administered to the subject once a week, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once a week, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 8 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once a week, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 8 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once a week, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject once a week at a dose of 20 mg/kg.

In certain embodiments, the anti-CD 20 antibody (e.g., rituximab) is at 375mg/m2Is administered to a subject biweekly and binds to an antibody to CD47 orThe antigen-binding fragment thereof is administered to a subject twice weekly at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 8 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 8 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject biweekly, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject weekly at a dose of 20 mg/kg.

In certain embodiments, the anti-CD 20 antibody (e.g., rituximab) is at 375mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice weekly at a dose of 0.3, 1, 2, 4,8, 15, or 20 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 8 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject twice a week at a dose of 8 mg/kg. In certain embodiments, rituximab is at 375mg/m2Is administered to the subject once every four weeks, and the antibody or antigen-binding fragment thereof that binds CD47 is administered to the subject once a week at a dose of 20 mg/kg.

As used herein, the term "combination" in the context of administering other therapies refers to the use of more than one therapy. The use of the term "combination" does not limit the order in which the therapies are administered. These therapies may be administered, for example, sequentially, simultaneously or concomitantly.

In certain embodiments of the methods provided herein, the anti-CD 20 antibody (e.g., rituximab) is administered prior to the protein therapeutic agent (e.g., anti-CD 47 antibody). In certain embodiments of the methods provided herein, the anti-CD 20 antibody (e.g., rituximab) is administered concurrently with the protein therapeutic agent (e.g., anti-CD 47 antibody). In certain embodiments of the methods provided herein, the anti-CD 20 antibody (e.g., rituximab) is administered after the protein therapeutic agent (e.g., anti-CD 47 antibody). In certain embodiments of the methods provided herein, the anti-CD 20 antibody (e.g., rituximab) is administered prior to and concurrently with the protein therapeutic agent (e.g., anti-CD 47 antibody).

As used herein, the terms "subject" and "patient" are used interchangeably. As used herein, a subject is a mammal, such as a non-primate (e.g., cows, pigs, horses, cats, dogs, goats, rabbits, rats, mice, etc.) or a primate (e.g., monkey and human), e.g., a human. In one embodiment, the subject is a mammal, e.g., a human, diagnosed with a condition or disorder (e.g., cancer, metastasis, or angiogenesis) provided herein. In another embodiment, the subject is a mammal, e.g., a human, at risk of developing a condition or disorder provided herein (e.g., cancer, metastasis, or angiogenesis). In another embodiment, the subject is a human.

As used herein, "hematologic cancer" refers to cancer of the blood, and includes leukemia, lymphoma, myeloma, and the like. "leukemia" refers to cancer of the blood in which too many white blood cells are produced that are ineffective against infection, thus displacing other parts of the blood that make up the blood, such as platelets and red blood cells. It is well known that cases of leukemia are classified as either acute or chronic. By way of non-limiting example, certain forms of leukemia include Acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), myeloproliferative disease/tumor (MPDS), and myelodysplastic syndrome. "lymphoma" may refer to hodgkin's lymphoma, indolent and aggressive non-hodgkin's lymphoma, burkitt's lymphoma, and follicular lymphoma (both small and large cells), among others. Myeloma may refer to Multiple Myeloma (MM), giant cell myeloma, heavy chain myeloma and light chain, or Bence-Jones myeloma. In certain embodiments, the hematologic cancer is multiple myeloma. In certain embodiments, the hematologic cancer is non-hodgkin's lymphoma.

Non-limiting examples of conditions that may be treated or managed with the anti-CD 47 antibodies described herein, alone or in combination with anti-CD 20 antibodies (e.g., rituximab), include hematologic cancers and/or solid tumors, and diseases or disorders associated with aberrant CD47 expression, activity, and/or signaling include, by way of non-limiting example, hematologic cancers and/or solid tumors. Hematologic cancers include, for example, leukemia, lymphoma, and myeloma. By way of non-limiting example, certain forms of leukemia include Acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), myeloproliferative disease/tumor (MPDS), and myelodysplastic syndrome. By way of non-limiting example, certain forms of lymphoma include hodgkin's lymphoma, indolent and aggressive non-hodgkin's lymphoma, burkitt's lymphoma, and follicular lymphoma (both small and large). By way of non-limiting example, some forms of myeloma include Multiple Myeloma (MM), giant cell myeloma, heavy chain myeloma, and light chain or Bence-Jones myeloma. Solid tumors include, for example, breast, ovarian, lung, pancreatic, prostate, melanoma, colorectal, lung, head and neck, bladder, esophageal, liver, and kidney tumors.

Symptoms associated with cancer and other neoplastic diseases include, for example, inflammation, fever, general malaise, fever, pain (often located in inflamed areas), loss of appetite, weight loss, edema, headache, fatigue, rash, anemia, muscle weakness, muscle fatigue, and abdominal symptoms such as abdominal pain, diarrhea, or constipation.

In particular aspects, provided herein are anti-CD 47 antibodies that can be used alone or in combination with an anti-CD 20 antibody (e.g., rituximab) for treating, delaying progression of, arresting, preventing recurrence of, or alleviating symptoms of a cancer (e.g., MM, NHL, AML, breast cancer (e.g., triple negative breast cancer), bladder cancer, non-small cell lung cancer, hepatocellular carcinoma (HCC), sarcoma, and head and neck cancer). For example, the CD47 antibodies described herein can be used alone or in combination with an anti-CD 20 antibody (e.g., rituximab) for the treatment of hematological malignancies and/or tumors, e.g., hematological malignancies and/or tumors. For example, the CD47 antibodies described herein can be used to treat CD47+ tumors. By way of non-limiting example, the CD47 antibodies described herein can be used to treat non-hodgkin's lymphoma (NHL), Acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Multiple Myeloma (MM), breast cancer (e.g., triple negative breast cancer), ovarian cancer, head and neck cancer, bladder cancer, melanoma, colorectal cancer, pancreatic cancer, lung cancer, leiomyoma, leiomyosarcoma, glioma, glioblastoma, and the like. Solid tumors include, for example, breast tumors, ovarian tumors, lung tumors (e.g., NSCLC), pancreatic tumors, prostate tumors, melanoma, colorectal tumors, lung tumors, head and neck tumors, bladder tumors, esophageal tumors, liver tumors (e.g., hepatocellular carcinoma), sarcomas, and kidney tumors.

In one particular embodiment, provided herein is a method of treating cancer (e.g., a hematological disorder/cancer or a solid cancer) in a subject, comprising administering (e.g., simultaneously or sequentially) to a subject in need thereof (i) an anti-CD 47 antibody or antigen-binding fragment thereof described herein that specifically binds CD47, such as human CD47, alone or in combination with an anti-CD 20 antibody (e.g., rituximab), and (ii) another anti-cancer agent. In certain embodiments, the anti-cancer agent is a chemotherapeutic agent (e.g., a microtubule disassembly blocking agent, an anti-metabolite, a topoisomerase inhibitor, and a DNA cross-linking or damaging agent). In certain embodiments, the anti-cancer agent is a tyrosine kinase inhibitor (e.g., a tyrosine kinase inhibitor)(imatinib mesylate) or(SU11248 or sunitinib)). Other non-limiting examples of tyrosine kinase inhibitors include 706 and AMNI07 (nilotinib). RAD00I, PKC412. Gefitinib (IRESSA)TM) Erlotinib, and methods of use thereofSorafenibPazopanib (VOTRIENT)TM) Asitinib, bosutinib, Cedizibobu (Dasatinib), LapatinibLestaurtinib, neratinib, nilotinibSimaxanil, tositude cloth (PALLADIA)TM) Vandetanib (ZACTIMA)TM) And vartanib.

In a particular aspect, provided herein is a method of treating cancer (e.g., a hematological disorder/cancer or a solid cancer) in a subject, comprising administering (e.g., simultaneously or sequentially) to a subject in need thereof (i) an anti-CD 47 antibody or antigen-binding fragment thereof described herein that specifically binds CD47, such as human CD47, alone or in combination with an anti-CD 20 antibody (e.g., rituximab), and (ii) radiation therapy.

In a particular aspect, provided herein is a method of promoting (e.g., inducing or enhancing) phagocytosis (e.g., macrophage-mediated phagocytic killing of tumor cells), comprising contacting tumor cells with an effective amount of an anti-CD 47 antibody that specifically binds human CD47, described herein, alone or in combination with an anti-CD 20 antibody (e.g., rituximab). Also provided herein are methods of promoting (e.g., inducing or enhancing) phagocytosis (e.g., macrophage-mediated phagocytic killing of tumor cells) in a subject in need thereof (e.g., a subject having tumor cells such as CD 47-expressing tumor cells), comprising administering to the subject an effective amount of an anti-CD 47 antibody described herein that specifically binds human CD47, alone or in combination with an anti-CD 20 antibody (e.g., rituximab).

In one particular aspect, provided herein is a method of reducing tumor volume comprising contacting a tumor with an effective amount of an anti-CD 47 antibody that specifically binds human CD47 described herein, alone or in combination with an anti-CD 20 antibody (e.g., rituximab). Also provided herein are methods of reducing tumor volume in a subject in need thereof (e.g., a subject having a tumor such as a CD 47-expressing tumor) comprising administering to the subject an effective amount of an anti-CD 47 antibody that specifically binds human CD47 described herein, alone or in combination with an anti-CD 20 antibody (e.g., rituximab).

In a particular aspect, provided herein is a method of inhibiting growth or proliferation of a cancer cell, comprising contacting the cancer cell with an effective amount of an anti-CD 47 antibody that specifically binds human CD47 described herein, alone or in combination with an anti-CD 20 antibody (e.g., rituximab). Also provided herein are methods of inhibiting growth or proliferation of cancer cells in a subject in need thereof (e.g., a subject having cancer cells such as CD 47-expressing cancer cells), comprising administering to the subject an effective amount of an anti-CD 47 antibody that specifically binds human CD47 described herein, alone or in combination with an anti-CD 20 antibody (e.g., rituximab).

4.2 composition

4.2.1 protein therapeutics:

the protein therapeutic used in combination with, for example, an anti-CD 20 antibody (e.g., rituximab) in the methods described herein can be any protein therapeutic known to one of skill in the art. In certain embodiments, the protein therapeutic is a cytokine. In certain embodiments, the cytokine is Bone Morphogenic Protein (BMP), Erythropoietin (EPO), granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony stimulating factor (GM-CSF), interferon alpha (IFN-alpha), interferon beta (IFN-beta), interleukin 2(IL-2), interleukin 11(IL-11), or interferon gamma (IFN-gamma). In certain embodiments, the protein therapeutic is an interleukin. In certain embodiments, the protein therapeutic is not an enzyme.

In certain embodiments, the protein therapeutic is a fusion protein, e.g., an Fc-containing fusion protein, e.g., a soluble receptor fusion protein.

4.2.2 antibodies

The protein therapeutic used in combination with, for example, an anti-CD 20 antibody (e.g., rituximab) in the methods described herein can be an antibody therapeutic. In some embodiments, the anti-CD 20 antibody is rituximab. Rituximab: (Baijian (Biogen)/Genetech (Genentech)) is a chimeric murine/human monoclonal IgG directed against the CD20 antigen1Kappa antibodies. Rituximab has a molecular weight of about 145kD and a binding affinity for the CD20 antigen of about 8.0 nM. Rituximab was produced from a suspension culture of mammalian cells (chinese hamster ovary) in nutrient medium containing the antibiotic gentamicin. Gentamicin was not detected in the final product.Is a sterile, clear, colorless, preservative-free liquid concentrate for intravenous administration.Provided at a concentration of 10mg/mL in 100mg/10mL or 500mg/50mL disposable vials. This product was formulated in polysorbate 80(0.7mg/mL), sodium chloride (9mg/mL), sodium citrate dihydrate (7.35mg/mL) and water for injection. The pH was 6.5. In some embodiments, the anti-CD 20 antibody is omeprazole (ocrelizumab). In some embodiments, the anti-CD 20 antibody is ofatumumab. In some embodiments, the anti-CD 20 antibody is obinutuzumab (obinutuzumab). In some embodiments, the anti-CD 20 antibody is tositumomab (tositumomab). In some embodiments, the anti-CD 20 antibody is ibritumomab tiuxetan (ibritumomab). In some embodiments, the anti-CD 20 antibody is ocatuzumab (ocaatuzumab). In some embodiments, the anti-CD 20 antibody is veltuzumab)。

As used herein and unless otherwise specified, the term "about" or "approximately" means within ± 10% of a given value or range. Where integers are required, these terms mean within ± 10% of a given value or range, rounded up or down to the nearest integer.

As used herein, the terms "antibody" and "immunoglobulin" and "Ig" are terms of the art and are used interchangeably herein, and refer to a molecule having an antigen binding site that specifically binds an antigen.

Antibodies provided herein can include, for example, monoclonal antibodies, recombinantly produced antibodies, monospecific antibodies, multispecific antibodies (including bispecific antibodies), human antibodies, humanized antibodies, murine antibodies (e.g., mouse or rat antibodies), chimeric antibodies, synthetic antibodies, and tetrameric antibodies comprising two heavy chain and two light chain molecules. In particular embodiments, antibodies may include, but are not limited to, antibody light chain monomers, antibody heavy chain monomers, antibody light chain dimers, antibody heavy chain dimers, antibody light chain-antibody heavy chain pairs, intrabodies, heteroconjugate antibodies (heteroconjugate antibodies), single domain antibodies, and monovalent antibodies. In one particular embodiment, the antibody may comprise an antigen-binding or epitope-binding fragment, such as, but not limited to, a single chain antibody or a single chain fv (scfv) (e.g., including monospecific, bispecific, etc.), a camelized antibody, an affibody, a Fab fragment, a F (ab')2Fragment and disulfide-linked fv (sdFv). In certain embodiments, the antibodies described herein refer to a polyclonal antibody population.

The antibody can be of any type (e.g., IgG, IgE, IgM, IgD, IgA or IgY), of any class (e.g., IgG)1、IgG2、IgG3、IgG4、IgA1Or IgA2) Or of any subclass (e.g., IgG)2aOr IgG2b) The immunoglobulin molecule of (a). In certain embodiments, the antibodies described herein are IgG antibodies, or classes thereof (e.g., human IgG)1、IgG2、IgG3Or IgG4) Or a subclass. In certain embodiments, described hereinThe antibody is IgG1Antibodies (e.g., human IgG)1) Or a subclass thereof. In certain embodiments, the IgG described herein1Antibodies comprise one or more amino acid substitutions and/or deletions in the constant region. In certain embodiments, the antibody described herein is an IgG4Antibodies (e.g., human IgG)4) Or a subclass thereof. In certain embodiments, the IgG described herein4Antibodies comprise one or more amino acid substitutions and/or deletions in the constant region.

As used herein, an "antigen" is a moiety or molecule that contains an epitope to which an antibody can specifically bind. Thus, the antigen is also specifically bound by the antibody.

As used herein, "epitope" is a term in the art and refers to a localized region of an antigen to which an antibody can specifically bind. The epitope may be a linear epitope or a conformational, non-linear or discontinuous epitope. For example, in the case of a polypeptide antigen, an epitope may be contiguous amino acids of the polypeptide ("linear" epitope), or an epitope may comprise amino acids from two or more non-contiguous regions of the polypeptide ("conformational", "non-linear" or "discontinuous" epitope). One skilled in the art will appreciate that, in general, linear epitopes may or may not be dependent on secondary, tertiary, or quaternary structure.

As used herein, the term "monoclonal antibody" is a term well known in the art and refers to an antibody obtained from a population of homogeneous or substantially homogeneous antibodies. The term "monoclonal" is not limited to any particular method of making an antibody. Generally, a population of monoclonal antibodies can be produced by a cell, a population of cells, or a cell line. In particular embodiments, a "monoclonal antibody" as used herein is an antibody produced by a single cell or cell line, wherein the antibody immunospecifically binds to the CD47 epitope, as determined, for example, by ELISA or other antigen binding or competitive binding assays known in the art or in the examples provided herein. In particular embodiments, the monoclonal antibody can be a chimeric antibody or a humanized antibody. In certain embodiments, the monoclonal antibody is a monovalent antibody or a multivalent (e.g., bivalent) antibody.

As used herein, the term "unnatural amino acid" refers to an amino acid that is not a proteinogenic amino acid, or a post-translationally modified variant thereof. In particular, the term refers to amino acids that are not one of the 20 common amino acids or pyrrolysine or selenocysteine, or post-translationally modified variants thereof.

As used herein, the term "polyclonal antibody" refers to a population of antibodies comprising a plurality of different antibodies that immunospecifically bind to the same and/or different epitopes within one or more antigens.

As used herein, the term "variable region" or "variable domain" refers to a portion of an antibody, usually a portion of an antibody light chain or heavy chain, typically around the amino-terminal 110 to 120 amino acids in the mature heavy chain and around the amino-terminal 90 to 100 amino acids in the mature light chain. The variable region comprises Complementarity Determining Regions (CDRs) flanked by Framework Regions (FRs). In general, the spatial orientation of the CDRs and FRs in the N-terminal to C-terminal direction is as follows: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR 4. Without wishing to be bound by any particular mechanism or theory, it is believed that the CDRs of the light and heavy chains are primarily responsible for the interaction of the antibody with the antigen and the specificity of the antibody for the epitope. In one embodiment, the numbering of amino acid positions of the antibodies described herein is according to the EU Index, e.g., as described in Kabat et al (1991)Sequences of Proteins of Immunological Interest[ protein sequence of immunological significance]Fifth edition, department of Health and Human Services, NIH publication No. 91-3242. In certain embodiments, the variable region is a human variable region. In certain embodiments, the variable region comprises murine (e.g., mouse or rat) CDRs and human Framework Regions (FRs). In particular embodiments, the variable region is a primate (e.g., human or non-human primate) variable region. In certain embodiments, the variable region comprises a murine (e.g., mouse or rat) CDR and a primate (e.g., human or non-human primate) Framework Region (FR). As a non-limiting example, the variable regions described herein are obtained by assembling two or more fragments of a human sequence into a composite human sequence.

In certain aspects, the CDRs of an antibody can be encoded according to (i) KabatSystem No. (Kabat et al (1971) ann.ny acad.sci. [ new york academy of sciences annual press.)]190:382 supplement 391 and Kabat et al (1991)Sequences of Proteins of Immunological Interest[ protein sequence of immunological significance]Fifth edition, department of Health and Human Services (U.S. department of Health and Human Services), NIH publication No. 91-3242); or (ii) the Chothia numbering scheme, which will be referred to herein as "Chothia CDRs" (see, e.g., Chothia and Lesk, 1987, j.mol.biol. [ journal of molecular biology]196: 901-; Al-Lazikani et Al 1997, J.mol.biol. [ journal of molecular biology]273: 927-; chothia et al, 1992, J.mol.biol. [ journal of molecular biology]227: 799-; tramontano A et al 1990, J.mol.biol. [ J.Mol.Biol. [ J.M.]215(1) 175-82; and U.S. patent No. 7,709,226); or (iii) ImmunoGeneTiCs (IMGT) numbering system, e.g., as Lefranc, M. -P., 1999, The Immunologist]7:132-136 and Lefranc, M. -P. et al 1999, Nucleic Acids Res. [ Nucleic acid research ]]27:209-212 ("IMGT CDR"); or (iv) MacCallum et al, 1996, J.mol.biol. [ J.M.]262: 732-. See, e.g., Martin, A., "Protein Sequence and Structure Analysis of Antibody Variable Domains" [ Protein Sequence and Structure Analysis of Antibody Variable Domains]Antibody Engineering]Kontermann and D ü bel eds, Chapter 31, pp 422-439, Berlin Springs-Verlag, Berlin (2001).

With respect to the Kabat numbering system, the CDRs in an antibody heavy chain molecule typically exist at amino acid positions 31 to 35, which optionally may comprise one or two additional amino acids after 35 (referred to as 35A and 35B in the Kabat numbering scheme) (CDR1), amino acid positions 50 to 65(CDR2), and amino acid positions 95 to 102(CDR 3). Using the Kabat numbering system, CDRs in an antibody light chain molecule are typically present at amino acid positions 24 to 34(CDR1), amino acid positions 50 to 56(CDR2), and amino acid positions 89 to 97(CDR 3). As is well known to those skilled in the art, using the Kabat numbering system, the actual linear amino acid sequence of an antibody variable domain may contain fewer or additional amino acids due to shortening or lengthening of the FRs and/or CDRs, and thus, the Kabat numbering of amino acids is not necessarily the same as its linear amino acid numbering.

The antibodies provided herein can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, or IgY), of any class (e.g., IgG1、IgG2、IgG3、IgG4、IgA1Or IgA2) Or of any subclass (e.g., IgG)2aOr IgG2bOr mixtures thereof). In certain embodiments, the antibodies described herein are IgG antibodies (e.g., human IgG) or classes thereof (e.g., human IgG)1、IgG2、IgG3Or IgG4) Or a subclass thereof.

In particular aspects, provided herein are antibodies comprising antibody light and heavy chains (e.g., light and heavy chains, respectively). With respect to light chains, in a particular embodiment, the light chain of an antibody described herein is a kappa (κ) light chain. In another specific embodiment, the light chain of the antibody described herein is a lambda (λ) light chain. In another embodiment, the light chain is a mixed sequence, e.g., the variable portion of the light chain comprises a kappa light chain sequence and the constant region of the light chain comprises a lambda light chain sequence, or vice versa. In certain embodiments, the light chain of an antibody described herein is a human kappa light chain or a human lambda light chain. Non-limiting examples of human constant region sequences have been described in the art, e.g., see U.S. Pat. No. 5,693,780 and Kabat et al (1991)Sequences of Proteins of Immunological Interest[ protein sequence of immunological significance]Fifth edition, department of Health and Human Services, NIH publication No. 91-3242.

In certain embodiments, the anti-CD 47 antibodies or antigen-binding fragments thereof described herein comprise an amino acid sequence having a certain percentage identity relative to a parent antibody.

Determination of percent identity between two sequences (e.g., amino acid sequences or nucleic acid sequences) can also be accomplished using a mathematical algorithm. Non-limiting examples of mathematical algorithms for comparing two sequences are Karlin and Altschul, 1990, proc.natl.acad.sci.u.s.a. [ journal of the american national academy of sciences ], the algorithm of 87:22642268, as modified in Karlin and Altschul, 1993, proc.natl.acad.sci.u.s.a. [ journal of the american national academy of sciences ]90: 58735877. This algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al, 1990, j.mol.biol. [ journal of molecular biology ]215: 403. BLAST nucleotide searches can be performed by setting NBLAST nucleotide program parameters, for example, to a score of 100 and a word length of 12, to obtain nucleotide sequences homologous to the nucleic acid molecules described herein. BLAST protein searches can be performed by setting the XBLAST program parameters, for example, to a score of 50, a word length of 3, to obtain amino acid sequences homologous to the protein molecules described herein. To obtain a Gapped alignment for comparison purposes, Gapped BLAST as described in Altschul et al, (1997) Nucleic Acids Res. [ Nucleic Acids research ]25:33893402 can be used. Alternatively, PSI BLAST can be used to perform an iterative search for detecting distant relationships between molecules (ibid.). When utilizing BLAST, Gapped BLAST, and PSI BLAST programs, default parameters (e.g., of XBLAST and NBLAST) of the respective programs can be used (see, e.g., National Center for Biotechnology Information, NCBI, on the world wide web NCBI. Another preferred, non-limiting example of a mathematical algorithm for sequence comparison is the algorithm of Myers and Miller, 1988, CABIOS 4: 1117. This algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When comparing amino acid sequences using the ALIGN program, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used.

Percent identity between two sequences may be determined with or without allowing gaps using techniques similar to those described above. In calculating percent identity, only exact matches are typically counted.

The anti-CD 47 antibodies described herein also include monoclonal antibodies that specifically bind to CD47, wherein the antibodies do not promote (e.g., induce or enhance) or cause a significant level of agglutination, such as red blood cell hemagglutination ("RBC hemagglutination").

A pharmaceutical composition according to the invention may comprise an antibody of the invention and a pharmaceutically acceptable carrier.

In some embodiments, the antibody or antigen-binding fragment thereof is an IgG isotype. In some embodiments, the constant region of the antibody is a human IgG1 isotype having the following amino acid sequence:

in some embodiments, the human IgG1 constant region is modified at amino acid Asn297(Boxed, Kabat numbering) to prevent glycosylation of the antibody, e.g., Asn297Ala (N297A). In some embodiments, the constant region of the antibody is modified at amino acid Leu235(Kabat numbering) to alter Fc receptor interactions, e.g., Leu235Glu (L235E) or Leu235Ala (L235A). In some embodiments, the constant region of the antibody is modified at amino acid Leu234(Kabat numbering) to alter Fc receptor interactions, e.g., Leu234Ala (L234A). In some embodiments, the constant region of the antibody is altered at both amino acids 234 and 235, such as Leu234Ala and Leu235Ala (L234A/L235A) (EU index in Kabat et al, 1991, Sequences of proteins of Immunological Interest).

In some embodiments, the constant region of the antibody is a human IgG2 isotype having the following amino acid sequence:

in some embodiments, the human IgG2 constant region is modified at amino acid Asn297(Boxed, Kabat numbering) to prevent antibody glycosylation, e.g., Asn297Ala (N297A).

In some embodiments, the constant region of the antibody is a human IgG3 isotype having the following amino acid sequence:

in some embodiments, the human IgG3 constant region is modified at amino acid Asn297(Boxed, Kabat numbering) to prevent antibody glycosylation, e.g., Asn297Ala (N297A). In some embodiments, the human IgG3 constant region is modified at amino acid 435 to increase half-life, e.g., Arg435H is (R435H) (EU index in Kabat et al, 1991, Sequences of proteins of Immunological Interest).

In some embodiments, the constant region of the antibody is a human IgG4 isotype having the following amino acid sequence:

in some embodiments, the human IgG4 constant region is modified within the hinge region to prevent or reduce chain exchange, e.g., Ser228Pro (S228P). In other embodiments, the human IgG4 constant region is modified at amino acid 235 to alter Fc receptor interactions, e.g., Leu235Glu (L235E). In some embodiments, the human IgG4 constant region is modified within the hinge and at amino acid 235, such as Ser228Pro and Leu235Glu (S228P/L235E). In some embodiments, the human IgG4 constant region is modified at amino acid Asn297(Kabat numbering) to prevent glycosylation of the antibody, e.g., Asn297Ala (N297A). In some embodiments of the invention, the human IgG4 constant region is modified at amino acid positions Ser228, Leu235, and Asn297 (e.g., S228P/L235E/N297A). (Kabat et al, 1991, EU index in Sequences of proteins of Immunological Interest). In other embodiments of the invention, the antibody is of the human IgG4 subclass and lacks glycosylation. In these examples, glycosylation may be eliminated by a mutation at position 297(Kabat numbering), e.g., N297A. In other embodiments, glycosylation can be eliminated by producing the antibody in a host cell (e.g., a bacterial or yeast-derived system or a modified mammalian cell expression system) that lacks the ability to post-translationally glycosylate.

In some embodiments, the human IgG constant region is modified to enhance FcRn binding. Examples of Fc mutations which enhance binding to FcRn are Met252Tyr, Ser254Thr, Thr256Glu (M252Y, S254T, T256E, respectively) (Kabat numbering, Dall' Acqua et al, 2006, J.biol Chem [ J. Biochem ], Vol.281 (33) 23514) or Met428Leu and Asn434Ser (M428L, N434S) (Zalevsky et al, 2010, Nature Biotech [ Nature Biotech ], Vol.28 (2) 157-) 159). (EU index in 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), such as amino acid modifications described in: natsume et al, 2008 cancer Res [ cancer research ], 68(10): 3863-72; idusogene et al, 2001J Immunol [ J Immunol ], 166(4): 2571-5; moore et al, 2010 mAbs [ monoclonal antibodies ], 2(2): 181-189; lazar et al, 2006PNAS journal of the national academy of sciences USA, 103(11): 4005-; shields et al, 2001 JBC [ journal of biochemistry ], 276(9): 6591-6604; stavenhagen et al, 2007 Cancer Res [ Cancer research ], 67(18): 8882-; stavenhagen et al, 2008 Advan. enzyme Regul. [ progression of enzyme regulation ], 48: 152-; alegr et al, 1992J Immunol [ J Immunol ], 148: 3461-; 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, a second CH3 domain with amino acid modifications at Thr366 within the CH3 domain, when replaced with a larger volume of amino acids such as Try (T366W), can preferentially pair with amino acid modifications (T366S/L368A/Y407V) with smaller volume amino acids at positions Thr366, Leu368 and Tyr407 (e.g., Ser, Ala and Val, respectively). Heterodimerization via modification of CH3 can be further stabilized by introducing disulfide bonds, for example by changing Ser354 to Cys (S354C) and Y349 to Cys (Y349C) on the opposite CH3 domain (reviewed in Carter, 2001, Journal of immunologicals methods, 248: 7-15).

In other embodiments of the invention, the antibody lacks glycosylation but is unmodified at amino acid Asn297(Kabat numbering). In these embodiments, glycosylation can be eliminated by producing the antibody in a host cell (e.g., a bacterial or yeast-derived system or a modified mammalian cell expression system) that lacks the ability to post-translationally glycosylate.

4.2.2.1 anti-CD 47 antibodies for use in the methods provided herein

In a particular aspect, for use in the methods provided herein are antibodies that specifically bind CD47 (e.g., human CD 47). Such anti-CD 47 antibodies include all antibodies disclosed in U.S. patent application publication No. 2014/0140989, which is incorporated by reference herein in its entirety. Such anti-CD 47 antibodies also include all antibodies disclosed in international patent application publication No. WO 2016/109415, which is incorporated herein by reference in its entirety.

As used herein, the terms "CD 47" or "integrin-associated protein" or "IAP" or "ovarian cancer antigen" or "OA 3" or "Rh-associated antigen" or "MER 6" are used interchangeably and refer to a multi-spanning transmembrane receptor belonging to the immunoglobulin superfamily. The amino acid sequence of exemplary human CD47 (GenBank accession No. Q08722.1 (GI: 1171879), incorporated herein by reference) is provided below. The signal sequence (amino acids 1-18) is underlined.

For clarity, the amino acid sequence of exemplary human CD47, which does not include a signal sequence, is provided below.

The invention also provides pharmaceutical compositions comprising one or more monoclonal antibodies that bind CD47, or an immunologically active fragment thereof, wherein the antibodies do not cause a significant level of hemagglutination of red blood cells after administration.

Hemagglutination is an example of a homotypic interaction in which two CD47 expressing cells aggregate or clump when treated with a bivalent CD47 binding entity. The ability of the antibodies of the present invention to bind CD47 on the cell surface and not cause the cell clumping phenomenon is not limited to red blood cells. It has been observed that the antibodies of the invention bind uniquely to CD47 in a manner that does not promote clumping of CD47 positive cell lines, such as Daudi cells.

In some cases, antibodies for use in the methods provided herein comprise a Variable Heavy (VH) chain region selected from the group consisting of SEQ ID NOS: 5-30. The antibody optionally comprises a Variable Light (VL) chain region selected from the group consisting of SEQ ID NOS: 31-47. In some cases, the antibody comprises a VH chain region selected from the group consisting of SEQ ID NOS: 5-30 and a VL chain region selected from the group consisting of SEQ ID NOS: 31-47. The antibodies of the invention also include antibodies having a variable heavy chain that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to a sequence set forth in at least one of SEQ ID Nos. 5-30 and a variable light chain that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to a sequence set forth in at least one of SEQ ID Nos. 31-47. In other aspects, the antibody comprises a VH region provided by any one of SEQ ID NOs 5,7, 8, 11, 15-17, 20-22, and 27-30 paired with a VL region provided by any one of SEQ ID NOs 31-39, 42, 43, 44, and 47. In another embodiment, the antibody comprises a VH region provided by any one of SEQ ID NOs 5,7, 8, 11, 12, 15-17, 20-22, and 27-30 paired with a VL region provided by any one of SEQ ID NOs 31, 32, 35, 40, 41, 42, 43, 44, and 47. In yet another aspect, the antibody comprises a combination of VH chain regions and VL chain regions selected from the combinations listed in table 1.

TABLE 1 exemplary anti-CD 47 antibody VH/VL combinations.

In some embodiments, the CD47 antibody or antigen-binding fragment thereof for use in the methods provided herein comprises the VH complementarity determining region 1(CDR1) sequence shown in SEQ ID NO 50, SEQ ID NO 57, SEQ ID NO 58, SEQ ID NO 59, SEQ ID NO 60, SEQ ID NO 61, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 64, SEQ ID NO 65, or SEQ ID NO 66, the VH CDR2 sequence shown in SEQ ID NO 51, SEQ ID NO 72, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, or SEQ ID NO 76, the VH CDR3 sequence shown in SEQ ID NO 52 or SEQ ID NO 77, SEQ ID NO 53, SEQ ID NO 67, or the VL CDR1 sequence shown in SEQ ID NO 68, SEQ ID NO 54, SEQ ID NO 69, SEQ ID NO 48, The VL CDR2 sequence shown in SEQ ID NO. 70 or SEQ ID NO. 71 and the VL CDR3 sequence shown in SEQ ID NO. 55. For example, the antibody or immunologically active fragment thereof comprises the VH CDR1 sequence shown in SEQ ID NO. 50, the VH CDR2 sequence shown in SEQ ID NO. 51, the VH CDR3 sequence shown in SEQ ID NO. 52, the VL CDR1 sequence shown in SEQ ID NO. 53, the VL CDR2 sequence shown in SEQ ID NO. 54, and the VL CDR3 sequence shown in SEQ ID NO. 55. In another example, the antibody or immunologically active fragment thereof comprises the VH CDR1 sequence shown in SEQ ID NO. 50, the VH CDR2 sequence shown in SEQ ID NO. 72, the VH CDR3 sequence shown in SEQ ID NO. 52, the VL CDR1 sequence shown in SEQ ID NO. 53, the VL CDR2 sequence shown in SEQ ID NO. 71, and the VL CDR3 sequence shown in SEQ ID NO. 55.

In a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, that specifically bind to human CD47, wherein such anti-CD 47 antibodies are variants of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises one or more amino acid modifications, e.g., 1-15 amino acid modifications, relative to the parent anti-CD 47 antibody. In a particular aspect, the one or more amino acid modifications, e.g., 1-15 amino acid modifications, are within the heavy chain or VH (e.g., SEQ ID NO: 6). In a particular aspect, the one or more amino acid modifications, e.g., 1-15 amino acid modifications, are within the framework region of a VH (e.g., SEQ ID NO: 6). In certain aspects, an anti-CD 47 antibody provided herein as a variant of a parent anti-CD 47 antibody comprises the CDRs (e.g., Kabat CDRs) of the parent anti-CD 47 antibody.

In a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, that specifically bind to human CD47, wherein such anti-CD 47 antibodies are variants of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises one or more amino acid modifications, e.g., 1-15 amino acid modifications, relative to the parent anti-CD 47 antibody. In a particular aspect, the one or more amino acid modifications, e.g., 5 or 14 amino acid modifications, are within the heavy chain or VH (e.g., SEQ ID NO: 6). In a particular aspect, the one or more amino acid modifications, e.g., 5, 10, 13, or 14 amino acid modifications, are within the framework region of a VH (e.g., SEQ ID NO: 6). In a particular aspect, the one or more amino acid modifications, e.g., 5, 13, or 14 amino acid modifications, are within the framework region of a VH (e.g., SEQ ID NO: 6). In certain aspects, an anti-CD 47 antibody provided herein as a variant of a parent anti-CD 47 antibody comprises the CDRs (e.g., Kabat CDRs) of the parent anti-CD 47 antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1, IgG2, IgG3, or IgG4 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1Z allotype isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG4 (such as IgG4P or IgG4PE) isotype antibody.

In a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, that specifically bind to human CD47, wherein such anti-CD 47 antibodies are variants of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in the CF system. In particular embodiments, the parent anti-CD 47 antibody is the antibody AB6.12 (see, e.g., U.S. application publication No. US2014/0140989a1, which is incorporated by reference herein in its entirety). The amino acid sequences of the heavy chain variable region (VH) and light chain variable region (VL) of antibody AB6.12 are provided below, with Kabat CDRs underlined. In a certain aspect, an anti-CD 47 antibody provided herein is a variant of the parent antibody AB6.12 and comprises CDRs (e.g., Kabat CDRs) of the parent antibody AB6.12, e.g., SEQ id nos: 50, 72, 52, 53, 71, and 55. In certain aspects, such an anti-CD 47 antibody is an IgG1, IgG2, IgG3, or IgG4 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1Z allotypic isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG4 (such as IgG4P or IgG4PE) isotype antibody.

anti-CD 47 antibody AB6.12 heavy chain variable region (VH) (Kabat CDR1-3 underlined, SEQ ID NOS: 50, 72 and 52):

QMQLVQSGAEVKKTGSSVKVSCKASGFNIKDYYLHWVRQAPGQALEWMGWIDPDQGDTEYAQKFQDRVTITRDRSMSTAYMELSSLRSEDTAMYYCNAAYGSSSYPMDYWGQGTTVTV(SEQ ID NO:6)

anti-CD 47 antibody AB6.12 light chain variable region (VL) (Kabat CDR1-3 underlined, SEQ ID NOS: 53, 71 and 55):

NIQMTQSPSAMSASVGDRVTITCKASQDIHRYLSWFQQKPGKVPKHLIYRANRLVSGVPSRFSGSGSGTEFTLTISSLQPEDFATYYCLQYDEFPYTFGGGTKVEIK(SEQ ID NO:42)

in a particular embodiment, the anti-CD 47 described herein comprises one or more amino acid modifications (e.g., 1-15 amino acid modifications) of a parent antibody (e.g., a parent antibody selected from the anti-CD 47 antibodies described herein), e.g., in the VH framework regions.

In a particular aspect, provided herein is an antibody, e.g., a monoclonal antibody, that specifically binds human CD47, wherein such anti-CD 47 antibody is a variant of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises a VH comprising the following N-terminal to C-terminal sequence:X1 QX2 QLVQSGAEVKKX3 GX4 SVKVSCKASGFNIKDYYLHWVRQAPGQX5 LEWMGWIDPDQGDTEYAQKX6 QX7 RVTX8 TX9 DX1 0SX11 STAYMELX12 SLRSX1 3 DTAX14 YYCNAAYGSSSYPMDYWGQGTTVTV (SEQ ID NO:89), whereinX1-X14 The underlined amino acid residues of (a), wherein X is arranged orderly from the N-terminus to the C-terminus1Is M or in position X1In the absence of amino acids, X2Is an amino acid having a hydrophobic side chain such as M or V, X3Is T or P, X4Is S or A, X5Is an acid having an aliphatic side chain such as A or G, X6Is F or L, X7Is D or G, X8Is an amino acid having a hydrophobic side chain such as I or M, X9Is R or T, X10Is R or T, X11Is M or T, X12Is S or R, X13Is a negatively charged amino acid such as E or D, and X14Are amino acids such as M or V with hydrophobic side chains.

In a particular aspect, the anti-CD 47 antibodies described herein comprise a VH comprising the sequence of SEQ ID NO:89, wherein at position X1The amino acid at (A) is any amino acid such as M, X2Is not M, X3Is not T, X4Is not S, X5Is not A, X6Is not F, X7Is not D, X8Is not I, X9Is not R, X10Is not R, X11Is not M, X12Is not S, X13Is not E, and/or X14Is not M. In a particular aspect, X1To X14Is not a member of any of 1, 2,3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 of these amino acids. In a particular aspect, the VH amino acid sequence is not that of antibody AB6.12, e.g., the VH amino acid sequence is not SEQ ID No. 6.

In a particular aspect, the anti-CD 47 antibodies described herein comprise a VH comprising the sequence of SEQ ID NO:89, wherein at position X7The amino acid in (A) is not G, X9Is not A and/or X11Is not S. In a particular aspect, X7、X9And X11Is not a member of any of 1, 2 or 3 of these amino acids. In a particular aspect, when in position X7Amino acid of (A)When is G, then X8Is M and/or X10Is T, X9Is not A and/or X11Is not S.

In a particular aspect, the anti-CD 47 antibodies described herein comprise a VH comprising the sequence of SEQ ID NO:89, wherein at position X7The amino acid in (A) is not G, X8Is not M, X9Is not E, X10Is not T, and/or X11Is not T. In a particular aspect, X7To X11Is not a member of any of 1, 2,3 or 4 of these amino acids. In a particular aspect, when in position X7When the amino acid in (A) is G, then X8Is M, X10Is T, X9Is not E, and X11Is T.

In one particular aspect, the anti-CD 47 antibodies described herein comprise a VH comprising the sequence of SEQ ID No. 89, wherein the VH does not comprise the amino acid sequence of SEQ ID No. 5,6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of U.S. application publication No. US2014/0140989a1, which is incorporated herein by reference in its entirety. In one particular aspect, the anti-CD 47 antibodies described herein comprise a VH comprising the consensus sequence of SEQ ID No. 89, wherein the VH does not comprise the framework region of the amino acid sequence of SEQ ID No. 5,6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of U.S. application publication No. US2014/0140989a1, which is incorporated herein by reference in its entirety.

In a particular aspect, X1Is M, X2Is V, X3Is P, X4Is A, X5Is G, X6Is L, X7Is G, X8Is M, X9Is T, X10Is T, X11Is T, X12Is R X13Is D, and/or X14Is V. In a particular embodiment, X1To X14Any 1, 2,3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 of these amino acids.

In a particular aspect, X1Is M, X2Is M, X3Is P, X4Is S, X5Is A, X6Is F, X7Is G, X8Is I, X9Is R, X10Is R, X11Is T, X12Is R, X13Is E, and/or X14Is V. In a particular embodiment, X1To X14Any 1, 2,3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 of these amino acids.

In a particular aspect, the anti-CD 47 antibody provided herein is not antibody AB 6.12. In a particular aspect, the anti-CD 47 antibodies provided herein do not comprise the VH (e.g., SEQ ID NO:6) and/or VL (e.g., SEQ ID NO:42) of antibody AB 6.12.

In a particular aspect, the anti-CD 47 antibodies provided herein comprise one of the following VH amino acid sequences provided in table 2.

Table 2: VH amino acid sequence

In a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, that specifically bind to human CD47, wherein such anti-CD 47 antibodies are variants of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises a VH comprising SEQ ID NO: 90. In certain aspects, such an anti-CD 47 antibody is an IgG1, IgG2, IgG3, or IgG4 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1Z allotype isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG4 (such as IgG4P or IgG4PE) isotype antibody.

In a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, that specifically bind to human CD47, wherein such anti-CD 47 antibodies are variants of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises a VH comprising SEQ ID NO: 91. In certain aspects, such an anti-CD 47 antibody is an IgG1, IgG2, IgG3, or IgG4 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1 isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG1Z allotype isotype antibody. In certain aspects, such an anti-CD 47 antibody is an IgG4 (such as IgG4P or IgG4PE) isotype antibody.

In a particular aspect, the anti-CD 47 antibodies (IgG1-13m) provided herein comprise an IgG1 heavy chain comprising the amino acid sequence shown below:

MQVQLVQSGAEVKKPGASVKVSCKASGFNIKDYYLHWVRQAPGQGLEWMGWIDPDQGDTEYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK(SEQ ID NO:81)

in a particular aspect, the anti-CD 47 antibodies (IgG1-13mZ) provided herein comprise an IgG1-Z allotypic heavy chain comprising the amino acid sequence shown below:

MQVQLVQSGAEVKKPGASVKVSCKASGFNIKDYYLHWVRQAPGQGLEWMGWIDPDQGDTEYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK(SEQ ID NO:82)

in a particular aspect, an anti-CD 47 antibody (IgG1-5m) provided herein comprises an IgG1 heavy chain comprising the amino acid sequence shown below:

MQMQLVQSGAEVKKPGSSVKVSCKASGFNIKDYYLHWVRQAPGQALEWMGWIDPDQGDTEYAQKFQGRVTITRDRSTSTAYMELRSLRSEDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK(SEQ ID NO:83)

in a particular aspect, the anti-CD 47 antibodies (IgG4P-13m) provided herein comprise an IgG4P antibody comprising the amino acid sequence shown below:

MQVQLVQSGAEVKKPGASVKVSCKASGFNIKDYYLHWVRQAPGQGLEWMGWIDPDQGDTEYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK(SEQ ID NO:84)

in a particular aspect, the anti-CD 47 antibodies (IgG4P-5m) provided herein comprise an IgG4P heavy chain comprising the amino acid sequence shown below:

MQMQLVQSGAEVKKPGSSVKVSCKASGFNIKDYYLHWVRQAPGQALEWMGWIDPDQGDTEYAQKFQGRVTITRDRSTSTAYMELRSLRSEDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK(SEQ ID NO:85)

in a particular aspect, the anti-CD 47 antibodies (IgG4PE-13m) provided herein comprise an IgG4PE heavy chain comprising the amino acid sequence shown below:

MQVQLVQSGAEVKKPGASVKVSCKASGFNIKDYYLHWVRQAPGQGLEWMGWIDPDQGDTEYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK(SEQ ID NO:86)

in a particular aspect, the anti-CD 47 antibodies (IgG4PE-5m) provided herein comprise an IgG4PE heavy chain comprising the amino acid sequence shown below:

MQMQLVQSGAEVKKPGSSVKVSCKASGFNIKDYYLHWVRQAPGQALEWMGWIDPDQGDTEYAQKFQGRVTITRDRSTSTAYMELRSLRSEDTAVYYCNAAYGSSSYPMDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK(SEQ ID NO:87)

in a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, that specifically bind to human CD47, wherein such anti-CD 47 antibodies are variants of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises a light chain comprising a kappa or lambda light chain constant region (e.g., a human kappa or lambda light chain constant region), e.g., seq id NO: 88.

In a particular aspect, provided herein are antibodies, e.g., monoclonal antibodies, wherein the anti-CD 47 antibody is a variant of a parent anti-CD 47 antibody, wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent anti-CD 47 antibody when expressed in a CF system, and wherein the anti-CD 47 antibody comprises (i) a VH described herein (e.g., SEQ ID NO:89, 90 or 91) or a heavy chain described herein (e.g., any one of SEQ ID NO: 81-87), and (ii) a light chain comprising a kappa or lambda light chain constant region (e.g., a human kappa or lambda light chain constant region), e.g., SEQ ID NO:88, for example as shown below (anti-CD 47 antibody light chain (Ig κ)), or SEQ ID NO without amino acid M at the N-terminus: 88:

MNIQMTQSPSAMSASVGDRVTITCKASQDIHRYLSWFQQKPGKVPKHLIYRANRLVSGVPSRFSGSGSGTEFTLTISSLQPEDFATYYCLQYDEFPYTFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC(SEQID NO:88)。

in certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a VL domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 42, wherein the antibody specifically binds CD 47. In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a VL domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 42, wherein the antibody specifically binds CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID No. 42 (e.g., SEQ ID NOs 53, 71, and 55).

In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 88, wherein the antibody specifically binds CD 47. In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a light domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 88, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDR1-3) that are identical to the CDRs (e.g., VL CDR1-3) of SEQ ID No. 88 (e.g., SEQ ID NOs 53, 71, and 55).

In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a VH domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 6, wherein the antibody specifically binds CD47, and wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent antibody when produced in the CF system. In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a VH domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 6, wherein the antibody specifically binds CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID No. 6 (e.g., SEQ ID NOs 50, 72, and 52).

In certain embodiments, the antibodies or antigen-binding fragments thereof described herein comprise a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 78, wherein the antibody specifically binds to CD47, and wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent antibody when produced in the CF system. In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a heavy domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 78, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID No. 78 (e.g., SEQ ID NOs 53, 71, and 55).

In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:79, wherein the antibody specifically binds to CD47, and wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher antibody expression titer or yield as compared to the parent antibody when produced in the CF system. In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a heavy domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 79, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID No. 79 (e.g., SEQ ID NOs 53, 71, and 55).

In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a light chain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 80, wherein the antibody specifically binds to CD47, and wherein the anti-CD 47 antibody, when produced using a cell-free (CF) expression system, has a higher titer or yield of antibody expression compared to the parent antibody when produced in the CF system. In certain embodiments, an antibody or antigen-binding fragment thereof described herein comprises a heavy domain having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID No. 80, wherein the antibody specifically binds to CD47, and wherein the antibody comprises CDRs (e.g., VL CDRs 1-3) that are identical to the CDRs (e.g., VL CDRs 1-3) of SEQ ID No. 80 (e.g., SEQ ID NOs 53, 71, and 55).

Table 3 provides a table of the anti-CD 47 antibody amino acid sequences described herein. In certain embodiments, the antibodies described herein comprise any light chain variable region sequence in table 3 and any heavy chain variable region sequence in table 3.

TABLE 3 anti-CD 47 antibody amino acid sequence.

In some embodiments, the CD47 antibodies described herein are used in combination with one or more additional agents or a combination of additional agents. Suitable additional agents include current drugs and/or surgical therapies for the intended application, e.g., cancer. For example, the CD47 antibody can be used in combination with one or more additional chemotherapeutic or antineoplastic agents. Alternatively, the additional chemotherapeutic agent is radiation therapy. In some embodiments, the chemotherapeutic agent is a cell death inducing agent. In some embodiments, the chemotherapeutic agent induces a loss of phospholipid asymmetry across the plasma membrane, e.g., causing cell surface exposure of Phosphatidylserine (PS). In some embodiments, the chemotherapeutic agent induces Endoplasmic Reticulum (ER) stress. In some embodiments, the chemotherapeutic agent is a proteasome inhibitor. In some embodiments, the chemotherapeutic agent induces translocation of the ER protein to the cell surface. In some embodiments, the chemotherapeutic agent induces the translocation of calreticulin and cell surface exposure.

In some embodiments, the CD47 antibody and the additional agent are formulated as a monotherapy composition and the CD47 antibody and the additional agent are administered simultaneously. Alternatively, the CD47 antibody and additional agent are separate from each other, e.g., each formulated in a separate therapeutic composition, and the CD47 antibody and additional agent are administered simultaneously, or the CD47 antibody and additional agent are administered at different times during a treatment regimen. For example, the CD47 antibody is administered prior to administration of an additional agent, the CD47 antibody is administered after administration of an additional agent, or the CD47 antibody and an additional agent are administered in an alternating manner. The CD47 antibody and additional agent are administered in single or multiple doses, as described herein.

In particular aspects, the anti-CD 47 antibodies provided herein comprise one or more unnatural amino acid residue at a site-specific position. See, for example, U.S. application publication No. US2014/0046030a1, which is incorporated by reference herein in its entirety. In particular aspects, unnatural amino acid residues at site-specific positions can have advantages with respect to antibody yield, solubility, binding affinity, and/or activity. Non-limiting examples of unnatural amino acids have been described, see, e.g., U.S. application publication No. US 2014/0066598 a 1.

In a particular aspect, provided herein are anti-CD 47 antibodies conjugated to a conjugate moiety or an agent such as a label or toxin. The conjugate moiety may be any conjugate moiety that those skilled in the art consider useful. For example, the conjugate moiety may be a polymer, such as polyethylene glycol, that can improve antibody stability in vitro or in vivo. The conjugate moiety may be therapeutically active, thereby producing an antibody-drug conjugate. The conjugate moiety may be a molecular payload that is harmful to the target cell. The conjugate moiety may be a label that can be used for detection or diagnosis. In certain aspects, the conjugate moiety is attached to the antibody by a direct covalent bond. In certain aspects, the conjugate moiety is linked to the antibody via a linker. In particular aspects, the conjugate moiety or linker is linked through one of the unnatural amino acids of the anti-CD 47 antibody. Exemplary conjugate moieties and linkers have been described, for example, see U.S. application publication No. US2014/0046030a1, which is incorporated by reference herein in its entirety.

The antibodies or antigen-binding fragments described herein that immunospecifically bind to CD47 (e.g., the ECD of human CD47) can be generated by any method known in the art.

The antibodies described herein may, for example, comprise chimeric antibodies. Chimeric antibodies are molecules in which different portions of the antibody are derived from different immunoglobulin molecules. For example, a chimeric antibody can comprise the variable region of a mouse or rat monoclonal antibody fused to the constant region of a human antibody. Methods for producing chimeric antibodies are known in the art. See, e.g., Morrison, 1985, Science [ Science ]229: 1202; oi et al, 1986, BioTechniques [ Biotechnology ]4: 214; gillies et al, 1989, J.Immunol.methods [ J.Immunol. methods ]125: 191-202; and U.S. Pat. nos. 5,807,715, 4,816,567, 4,816,397 and 6,331,415.

Antibodies or antigen-binding fragments produced using techniques such as those described herein can be isolated using standard well-known techniques. For example, the antibody or antigen-binding fragment can be suitably separated from, for example, culture medium, ascites fluid, serum, cell lysate, synthetic reaction material, and the like, by a conventional immunoglobulin purification procedure (e.g., protein a-agarose, hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography). As used herein, an "isolated" or "purified" antibody is substantially free of cellular material or other proteins from the cell or tissue source from which the antibody is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized, or free of components of the CF expression system used to produce the antibody.

The antibodies described herein include antibody fragments that recognize a particular CD47 antigen, and can be generated by any technique known to those skilled in the art. For example, enzymes such as papain (to produce Fab fragments) or pepsin (to produce F (ab')2Fragments) by immunizationProteolytic cleavage of the globulin molecule to produce Fab and F (ab')2And (3) fragment. A Fab fragment corresponds to one of the two identical arms of an antibody molecule and comprises the complete light chain paired with the VH and CH1 domains of the heavy chain. F (ab')2A fragment comprises two antigen-binding arms of an antibody molecule connected by disulfide bonds in the hinge region. Alternatively, the antibody fragments described herein may be routinely produced by well-known recombinant expression techniques. See, e.g., PCT publication nos. WO 92/22324; mullinax et al, 1992, BioTechniques biotechnologies]12(6) 864-869; sawai et al, 1995, AJRI [ journal of reproductive immunology USA ]]34: 26-34; and Better et al, 1988, Science [ Science]240:1041-1043。

The antibodies described herein can, for example, include humanized antibodies, such as deimmunized or composite human antibodies. The humanized antibody may comprise human constant region sequences. In certain embodiments, the humanized antibody may be selected from any class of immunoglobulin, including IgM, IgG, IgD, IgA, and IgE; and any isotype, including IgG1、IgG2、IgG3And IgG4. In certain embodiments, a humanized antibody may comprise a kappa or lambda light chain constant sequence.

Humanized antibodies may be generated using a variety of techniques known in the art, including, but not limited to, CDR-grafting (European patent No. EP 239,400; International publication No. WO 91/09967; and U.S. Pat. Nos. 5,225,539, 5,530,101 and 5,585,089), veneering (veering) or resurfacing (European patent No. EP 592,106 and EP 519,596; Padlan, 1991, Molecular Immunology [ Molecular Immunology ]28(4/5): 489-498; Studnica et al 1994, Protein Engineering [ Protein Engineering ]7(6): 814; and Roguska et al 1994, PNAS [ national academy of sciences ]91:969-973), chain shuffling (U.S. Pat. No. 5,565,332) and techniques such as those described in: U.S. Pat. No. 6,407,213, U.S. Pat. No. 5,766,886, WO 9317105, Tan et al, J.Immunol. [ J.Immunol ]169: 111925 (2002), Caldas et al, Protein Eng. [ Protein engineering ]13(5):353-60(2000), Morea et al, Methods [ Methods ]20(3): 26779 (2000), Baca et al, J.biol.chem. [ J.Chem. [ J.272 (16):10678-84(1997), Roguska et al, Protein Eng. [ Protein engineering ]9(10): 895904 (1996), Couto et al, Cancer Res. [ Cancer research ]55 (23. supplement: 1994) 5973 s-59s (1995), Couto et al, Cancer Res [ 1718 ] (55-1718), Sandhu. J.95150 (1995), Pelteh.957-957, J.957: 3, J.3: 3, and Peltier.3-73 (73) BioSen et al, J.3 (73) (1995). See also U.S. patent publication US 2005/0042664 a1 (24/2/2005), each of which is incorporated herein by reference in its entirety.

The antibodies described herein can be, for example, multispecific (e.g., bispecific) antibodies. Methods of making multispecific (e.g., bispecific) antibodies have been described, see, e.g., U.S. patent nos. 7951917, 7183076, 8227577, 5837242, 5989830, 5869620, 6132992, and 8586713.

Single domain antibodies, such as antibodies lacking a light chain, can be produced by methods well known in the art. See Riechmann et al, 1999, j.immunol. [ journal of immunology ]231: 25-38; nuttall et al 2000, Curr.pharm.Biotechnol. [ contemporary drug biotechnology ]1(3): 253-phase 263; muyderman, 2001, J.Biotechnol. [ J.Biotech ]74 (4: 277302; U.S. patent nos. 6,005,079; and international publication numbers WO 94/04678, WO 94/25591 and WO 01/44301.

Human antibodies can be produced using any method known in the art. For example, well known transgenic mice that do not express functional endogenous murine immunoglobulins, but can express human immunoglobulin genes, can be used. Alternatively, for example, the phage display technology described above can be used. Furthermore, in some embodiments, human antibodies can be produced, for example, using mouse-human hybridomas. For example, human peripheral blood lymphocytes transformed with epstein-barr virus (EBV) can be fused with mouse myeloma cells to produce mouse-human hybridomas that secrete human monoclonal antibodies, and these mouse-human hybridomas can be screened to determine hybridomas that secrete human monoclonal antibodies that immunospecifically bind to an antigen of interest (e.g., the ECD of human CD 47). Such methods are known and described in the art, see, e.g., Shinmoto et al, Cytotechnology [ cell technology ], 2004, 46: 19-23; naganawa et al, Human Antibodies [ Human Antibodies ], 2005, 14: 27-31.

Antibody variable domains with the desired binding specificity (antibody-antigen binding site) can be fused to immunoglobulin constant domain sequences. The fusion preferably has an immunoglobulin heavy chain constant domain comprising at least a portion of the hinge, CH2, and CH3 regions. Preferably, the first heavy chain constant region (CH1) containing the site necessary for light chain binding is present in at least one of the fusions. The DNA encoding the immunoglobulin heavy chain fusion and, if desired, the immunoglobulin light chain are inserted into separate expression vectors and then co-transfected into a suitable host organism. For further details on the generation of bispecific antibodies see, e.g., Suresh et al, Methods in Enzymology [ Methods in Enzymology ], 121:210 (1986).

According to another approach described in WO 96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers recovered from recombinant cell culture. Preferred interfaces comprise at least a portion of the CH3 region of the antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced by larger side chains (e.g., tyrosine or tryptophan). By replacing the large amino acid side chain with a smaller amino acid side chain (e.g., alanine or threonine), a compensatory "cavity" of the same or similar size to the large side chain is created at the interface of the second antibody molecule. This provides a mechanism for increasing the yield of heterodimers relative to other undesired end products such as dimers.

Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g., F (ab')2Bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical bonding. Brennan et al, Science]229:81(1985) describes a procedure in which intact antibodies are proteolytically cleaved to yield F (ab')2And (3) fragment. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments produced are then converted intoA thionitrobenzoic acid (TNB) derivative. One of the Fab ' -TNB derivatives is then converted to the Fab ' -thiol by reduction with mercaptoethylamine and mixed with an equimolar amount of the other Fab ' -TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as reagents for the selective immobilization of enzymes.

4.2.3 pharmaceutical compositions and kits

Provided herein are compositions, pharmaceutical compositions, and kits comprising one or more protein therapeutics described herein. Also provided herein are compositions, pharmaceutical compositions, and kits comprising an anti-CD 20 antibody (e.g., rituximab), alone or in combination with a protein therapeutic described herein. In particular, provided herein are compositions, pharmaceutical compositions, and kits comprising one or more of the antibodies described herein (e.g., anti-CD 47 antibodies) or antigen-binding fragments thereof, or conjugates thereof. In certain aspects, the compositions (e.g., pharmaceutical compositions) described herein can be used for in vitro, in vivo, or ex vivo use. Non-limiting examples of uses include use for reducing immunogenicity, use for modulating (e.g., inhibiting or inducing/enhancing) CD47 activity, and use for managing or treating disorders (e.g., cancer). In particular embodiments, provided herein are pharmaceutical compositions comprising an antibody (e.g., a humanized antibody) (or antigen-binding fragment thereof) described herein and a pharmaceutically acceptable carrier or excipient.

As used herein, the term "pharmaceutically acceptable" means approved by a regulatory agency of the federal or a state government or listed in the U.S. pharmacopeia, european pharmacopeia, or other generally recognized pharmacopeia for use in mammals, and more particularly in humans.

Formulations containing one or more of the antibodies or antigen-binding fragments thereof provided herein can be prepared for storage by admixing the antibody of the desired purity with an optional physiologically acceptable carrier, excipient, or stabilizer: (Remington’s Pharmaceutical Sciences[ Ramington pharmaceutical science](1990) Easton mark Publishing company, PA (Mack Publishing co., Easton, PA);Remington:The Science and Practice of Pharmacy[ Ramiphene ]Ton: pharmaceutical techniques and practices]21 st edition (2006) of the Barlerley Count Williams publishing Co., Lippincott Williams&Wilkins, Baltimore, MD)). Such formulations may, for example, be in the form of lyophilized formulations or aqueous solutions. Pharmaceutical carriers suitable for administration of the antibodies provided herein include any such carrier known to those of skill in the art to be suitable for a particular mode of administration. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers, such as phosphate, citrate, and other organic acids; and/or nonionic surfactants, such as TWEENTM、PLURONICSTMOr polyethylene glycol (PEG).

Formulations for in vivo administration may be sterile. This can be easily achieved, for example, by filtration through sterile filtration membranes.

In a particular aspect, a pharmaceutical composition for use in the methods provided herein contains a therapeutically effective amount of one or more protein therapeutics provided herein in a pharmaceutically acceptable carrier. In particular aspects, the pharmaceutical compositions for use in the methods provided herein contain a therapeutically effective amount of an anti-CD 20 antibody (e.g., rituximab) alone or in combination with one or more protein therapeutics provided herein in a pharmaceutically acceptable carrier. In particular aspects, a pharmaceutical composition for use in a method provided herein contains a therapeutically effective amount of one or more of the antibodies or antigen-binding fragments provided herein in a pharmaceutically acceptable carrier. Such pharmaceutical compositions can be used to prevent, treat, manage, or ameliorate a condition or disorder described herein, or one or more symptoms thereof.

Compositions for use in the methods provided herein can contain one or more protein therapeutics provided herein. Compositions for use in the methods provided herein may contain rituximab alone or in combination with one or more protein therapeutic agents provided herein. Compositions for use in the methods provided herein can contain one or more of the antibodies or antigen-binding fragments thereof provided herein. In one embodiment, compositions are provided wherein the antibodies or antigen binding fragments described herein are formulated into suitable pharmaceutical formulations, such as solutions, suspensions, powders, sustained release formulations or elixirs in sterile solutions or suspensions for parenteral administration, or transdermal patch preparations and dry powder inhalers.

In one embodiment, the compositions for use in the methods provided herein are formulated for single dose administration. To formulate the composition, a weight fraction of the compound is dissolved, suspended, dispersed, or otherwise mixed in a selected carrier at a concentration effective to alleviate, prevent, or ameliorate one or more symptoms of the condition being treated.

In certain aspects, the anti-CD 20 antibody (e.g., rituximab) is included in a pharmaceutically acceptable carrier in an effective amount sufficient to exert a therapeutically useful effect in the absence or with minimal or negligible adverse side effects in the patient being treated.

In certain aspects, the protein therapeutics for use in the methods provided herein are included in a pharmaceutically acceptable carrier in an effective amount sufficient to exert a therapeutically useful effect in the absence or with minimal or negligible adverse side effects in the patient being treated.

In certain aspects, the antibodies for use in the methods provided herein are contained in a pharmaceutically acceptable carrier in an effective amount sufficient to exert a therapeutically useful effect in the absence or with minimal or negligible adverse side effects in the patient being treated.

The concentration of a protein therapeutic (e.g., an anti-CD 47 antibody) in a pharmaceutical composition for use in the methods provided herein will depend on, for example, the physicochemical characteristics of the antibody, the dosage regimen, and the amount administered, among other factors.

Pharmaceutical compositions for use in the methods described herein are provided for administration to a human or animal (e.g., a mammal) in unit dosage form, such as a sterile parenteral (e.g., intravenous) solution or suspension containing an appropriate amount of the compound or a pharmaceutically acceptable derivative thereof. Also provided are pharmaceutical compositions for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, and oral or nasal solutions or suspensions, and oil-water emulsions containing an appropriate amount of a protein therapeutic or a pharmaceutically acceptable derivative thereof. In one embodiment, the protein therapeutic is formulated and administered in a unit dosage form or in multiple dosage forms. Unit dosage forms, as used herein, refer to physically discrete units suitable for use in association with a human or animal (e.g., mammalian) subject and packaged separately. Each unit dose contains a predetermined amount of the protein therapeutic in combination with a desired pharmaceutical carrier, vehicle or diluent sufficient to produce the desired therapeutic effect. Examples of unit dosage forms include ampoules and syringes and individually packaged tablets or capsules. The unit dosage form may be administered in fractions or multiples thereof. A multi-dose form is a plurality of identical unit dose forms packaged in a single container for administration as separate unit dose forms. Examples of multi-dose forms include vials, tablets or capsules bottles, or bottles. Thus, in a particular aspect, the multi-dose form is a multiple of a unit dose that is not separately packaged.

In certain embodiments, one or more protein therapeutics for use in the methods described herein are liquid pharmaceutical formulations. Liquid compositions that can be pharmaceutically administered can be prepared, for example, by dissolving, dispersing, or otherwise mixing the antibody and optional pharmaceutical adjuvant in a carrier, such as water, saline, aqueous dextrose, glycerol, glycols, and the like, to form a solution or suspension. In certain embodiments, the pharmaceutical compositions provided herein to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting agents, emulsifiers, solubilizing agents, pH buffers, and the like.

Methods of preparing such dosage forms are known or will be apparent to those skilled in the art. See, for exampleRemington’s Pharmaceutical Sciences[ Ramington pharmaceutical science](1990) Easton mark Publishing company, PA (Mack Publishing co., Easton, PA);Remington:The Science and Practice of Pharmacy[ Remington: pharmaceutical techniques and practices]21 st edition (2006) of the Barlerley Count Williams publishing Co., Lippincott Williams&Wilkins,Baltimore,MD)。

In one embodiment, parenteral administration is characterized by subcutaneous, intramuscular, or intravenous injection also contemplated herein. Injectables can be prepared in conventional forms, either as liquid solutions, or as suspensions, solid forms suitable for forming solutions or suspensions in liquids prior to injection, or as emulsions. Injections, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol. Other routes of administration may include enteral, intracerebral, nasal, intraarterial, intracardial, intraosseous infusion, intrathecal, and intraperitoneal administration.

Formulations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products (such as lyophilized powders), including subcutaneous tablets, ready to be combined with a solvent just prior to use, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use, and sterile emulsions. The solution may be an aqueous solution or a non-aqueous solution.

If administered intravenously, suitable carriers include physiological saline or Phosphate Buffered Saline (PBS), and solutions containing thickening and solubilizing agents such as glucose, polyethylene glycol, and polypropylene glycol, and mixtures thereof.

Pharmaceutically acceptable carriers for parenteral formulations include aqueous vehicles, non-aqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, chelating agents (or chelating agents), and other pharmaceutically acceptable materials.

The pharmaceutical carriers also include ethanol, polyethylene glycol and propylene glycol for water-miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.

In certain embodiments, intravenous or intra-arterial infusion of a sterile aqueous solution containing a protein therapeutic for use in the methods described herein is an effective mode of administration. Another example is a sterile aqueous or oily solution or suspension containing the protein therapeutic for use in the methods described herein that is injected as needed to produce the desired pharmacological effect.

In particular embodiments, the protein therapeutics for use in the methods described herein can be suspended in micronized or other suitable form. The form of the resulting mixture depends on a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle.

In other embodiments, the pharmaceutical formulation is a lyophilized powder that can be reconstituted for administration as a solution, emulsion, and other mixture. They may also be reconstituted and formulated as solids or gels.

Lyophilized powders can be prepared, for example, by dissolving the protein therapeutic for use in the methods provided herein in a suitable solvent. In some embodiments, the lyophilized powder is sterile. Suitable solvents may contain excipients that improve the stability or other pharmacological components of the powder or reconstituted solution prepared from the powder. Excipients that may be used include, but are not limited to, dextrose, sorbitol, fructose, corn syrup, xylitol, glycerol, glucose, sucrose, or other suitable agents. Suitable solvents may also contain buffering agents such as citrate, sodium or potassium phosphate, or other such buffering agents known to those skilled in the art, and in one embodiment, have a pH of about neutral. The solution is then sterile filtered and then lyophilized under standard conditions known to those skilled in the art to give an example of a formulation. In one embodiment, the resulting solution will be dispensed into vials for lyophilization. The lyophilized powder may be stored under suitable conditions, such as at about 4 ℃ to room temperature.

Reconstitution of the lyophilized powder with water for injection provides a formulation for parenteral administration. For reconstitution, the lyophilized powder is added to sterile water or other suitable vehicle.

In certain aspects, the protein therapeutics for the methods provided herein can be formulated for topical or external application, e.g., in the form of gels, creams, and lotions for external application to the skin and mucosa, such as in the eye, as well as for the eye or for intracisternal or intraspinal application. Topical use for transdermal delivery as well as for ocular or mucosal administration, or for inhalation therapy is contemplated. Nasal solutions of the active compounds, alone or in combination with other pharmaceutically acceptable excipients, may also be administered.

anti-CD 47 antibodies, anti-CD 20 antibodies (e.g., rituximab), and other protein therapeutics for use in the methods provided herein can also be formulated to target a particular body tissue, organ, or other region of a subject to be treated. Many such targeting methods are well known to those skilled in the art. All such targeting methods are contemplated herein for use in the compositions of the present invention. For non-limiting examples of targeting approaches, see, e.g., U.S. Pat. nos. 6,316,652, 6,274,552, 6,271,359, 6,253,872, 6,139,865, 6,131,570, 6,120,751, 6,071,495, 6,060,082, 6,048,736, 6,039,975, 6,004,534, 5,985,307, 5,972,366, 5,900,252, 5,840,674, 5,759,542, and 5,709,874. In some embodiments, the anti-CD 47 antibodies described herein are targeted to (or otherwise administered to) the visual organ, bone marrow, gastrointestinal tract, lung, brain, or joint. In particular embodiments, the anti-CD 47 antibodies described herein are capable of crossing the blood-brain barrier.

5. Examples of the invention

The examples in this section (i.e., section 5) are provided by way of illustration and not by way of limitation.

5.1 example 1: co-administration of rituximab and anti-CD 47 antibody in cynomolgus monkeys

The objective of this study was to monitor the immunogenicity of a humanized anti-CD 47 antibody comprising the heavy chain variable region CDRs of SEQ ID NOs 50, 72 and 52 and the light chain variable region CDRs of SEQ ID NOs 53, 71 and 55 (hereinafter "anti-CD 47 antibody") when co-administered with rituximab or rituximab and methotrexate and to evaluate its effect on pharmacokinetics after intravenous administration in cynomolgus monkeys.

In the first part of the study, the anti-CD 47 antibody was administered as four intravenous Injections (IV) at a dose of 20mg/kg (doses 1 to 4) to 3 groups of cynomolgus monkeys (5 animals/group, total 15 animals), with the time of administration for group 1 being days 1, 8, 15 and 22 of the study and the time of administration for groups 2 and 3 being days 15, 22, 29 and 36 of the study. Rituximab was administered as four intravenous injections to groups 2 and 3 animals at a dose of 10mg/kg on study days 1, 8, 15 and 22. Methotrexate was administered as three subcutaneous injections to group 3 animals at a dose of 0.4mg/kg on study days 15, 16 and 18.

In the second part of the study, the anti-CD 47 antibody was administered as a single intravenous injection to animals in groups 1 and 3 at a dose of 20mg/kg (dose 5) on study day 78. Methotrexate was administered as multiple subcutaneous injections to group 3 animals on study days 71, 72, 74, 78, 79, 81, 88, 95, 102 and 109 at a dose of 0.4 mg/kg.

Concentration data in cynomolgus monkeys after the first, fourth and fifth dose of anti-CD 47 antibody were used for Pharmacokinetic (PK) assessment to understand the effect of co-administered drugs on PK of anti-CD 47 antibody. Immunogenicity of anti-CD 47 antibodies was assessed by testing anti-drug antibody (ADA) titers in sera weekly throughout the study, where "drug" is an anti-CD 47 antibody. B cell counts were tracked weekly throughout the study.

All animals dosed with anti-CD 47 antibody alone (group 1) became ADA positive before dose 4. anti-CD 47 antibody area under serum concentration-time curve (AUC) 0 to 168 hours after dose 4 for five animals in group 10-168) (measurement of anti-CD 47 antibody serum concentration) is the AUC after dose 10-1683% -37%.

This decrease in AUC after seroconversion was attributed to the ADA-mediated decrease in free anti-CD 47 antibody exposure. The number of CD20+ B cells in these animals was within the normal range. The number of peripheral CD20+ B cells decreased rapidly in all animals receiving rituximab (groups 2 and 3) and within 24 hours after receiving the first dose of rituximab<100 cells/. mu.l. In 8 of 10 animals in groups 2 and 3, B cell numbers began to recover around day 15 after the first rituximab dose. This recovery of B cells can be attributed to anti-rituximab antibodies, as all animals had detectable anti-rituximab antibodies by day 15, with a corresponding decrease in serum rituximab concentration. By day 36, 8 animals showing recovery of B cell numbers became positive for the presence of anti-drug antibodies (ADA), titers varied between 5 and 625, and exposure of all 8 animals was reduced to varying degrees, ranging in AUC after dose 10-168Within 2% -53%.

In animals 11 and 15, which maintained significant rituximab concentrations, peripheral CD20+ B cell counts remained negligible until day 36, at which time a fourth dose of anti-CD 47 antibody was administered to these animals. The two animals that received all 4 doses of anti-CD 47 antibody with effective B cell depletion remained ADA negative for a long period of time until day 71 and the end of the study, respectively. anti-CD 47 antibody concentrations after dose 4 were similar to those observed after dose 1, with week 4 AUC in animals 11 and 150-168115% and 83% of mean AUC, respectively, at week 1.

Monkeys treated with anti-CD 47 antibody alone (group 1) or with a combination of all three agents (group 3) were then challenged again with anti-CD 47 antibody at 12 weeks (day 78). In two animals 11 and 15, in which effective B cell depletion reduced ADA development, the PK profile at week 12 was comparable to that after dose 1 of anti-CD 47 antibody, and AUC at week 120-168Mean AUC after first dose0-168127% and 120%, confirming that there was no ADA-mediated exposure loss in these animals. In addition, serum anti-drug antibody levels were maintained at low levels in both animals, with detectable drug present within three weeks after the last dose.

The concentration of anti-CD 47 antibody in ADA positive animals was below the limit of quantitation at more than 4 days after the last dose. These data indicate that depletion of B cells by rituximab reduces the immune response to the anti-CD 47 antibody and increases exposure to the drug over a long period of time. In addition, no toxicity was observed in monkeys treated with anti-CD 47 antibody plus rituximab or anti-CD 47 antibody plus rituximab plus methotrexate.

5.2 example 2: clinical study of anti-CD 47 antibody in combination with rituximab

Described herein are open label, phase 1 dose escalation and extension studies of anti-CD 47 antibodies administered by Intravenous (IV) infusion in subjects with advanced refractory solid and hematologic cancers. The study consisted of 2 sections. Part a is a dose escalation phase using escalating doses of anti-CD 47 antibody, and part B is a dose escalation and expansion phase in which anti-CD 47 antibody is administered in combination with rituximab to subjects with CD20 positive non-hodgkin's lymphoma (NHL). Based on a review of safety, PK and pharmacodynamic data from part a, expansion can be made at the Maximum Tolerated Dose (MTD) established in the dose escalation phase and/or at lower doses, or alternatively tolerated dosing regimens. The modified 3+3 dose escalation design was used to determine the initial toxicity of the anti-CD 47 antibody. A cohort of 3 to 6 evaluable subjects was treated with the anti-CD 47 antibody in a defined dosing regimen, and in the event of dose-limiting toxicity (DLT) in one subject, the cohort was expanded to a complete cohort of 6 evaluable subjects. In a given dose cohort, subjects 4 to 6 may be enrolled before the first 3 subjects complete cycle 1 to obtain additional safety information and to ensure that there are a sufficient number of evaluable patients for DLT evaluation. No more than one subject per day was enrolled in a given dose escalation cohort.

Doses included weekly (QW)0.3mg/kg IV and every 2 weeks (Q2W)1, 2, 4,8, 15 and 20mg/kg IV. During dose escalation, a decision is made to evaluate the lower dose cohort, the intermediate dose cohort, the alternate dosing interval, or declare the MTD based on a review of clinical and laboratory safety data for the previous dose cohort. All treatments were administered in a 28 day cycle, with anti-CD 47 antibody administered on days 1 and 15. After administering the first dose in any cohort, subjects were observed for at least 28 days (cycle 1, DLT window) before the next higher, regimen-specified dose cohort could be started.

The starting dose for cohort 1 was 0.3mg/kg on day 1 of cycle 1, followed by 1mg/kg QW.

For part B, in relapsed or refractory CD20 positive non-hodgkin lymphoma subjects who had previously received rituximab, two weeks prior to administration of anti-CD 47 antibody, initially once a week at 375mg/m2Rituximab administration was performed. Rituximab is administered at four weekly doses (day-15, day-8, day-1, day 8) before and during cycle 1, once per cycle (day 8) for cycles 2-6, and once every two months thereafter, if there is a response, up to 12 cycles (cycles 8, 10 and 8)12). Serial B cell counts and PK were evaluated. In addition, PK, PD and ADA evaluations were performed as per part a.

Inclusion criteria were:

1. male and female, 18 years old or older, with advanced, recurrent or refractory solid tumors, Multiple Myeloma (MM) or non-hodgkin's lymphoma (NHL) in part a.

In part B, only subjects with relapsed or refractory CD 20-positive NHL were included.

2. At least one of the subjects with solid tumors and NHL has a site of measurable disease.

3. Eastern cooperative tumor group performance status (ECOG PS) is 0 or 1.

4. The subject must have adequate hematopoietic, hepatic, renal, and blood clotting functions as assessed by specific laboratory criteria.

5. Women and men must agree to a contraceptive method and avoid pregnancy up to 8 weeks throughout the study and after the last dose of anti-CD 47 antibody. If part B is included, then the fertile women should continue to use an effective contraceptive method for 12 months after rituximab treatment

Exclusion criteria:

1. high grade lymphoma (Burkitt's lymphoma or lymphoblastic lymphoma), plasma cell leukemia.

2. High grade, rapidly proliferating solid tumors with a wide tumor burden (e.g., small cell lung cancer, germ cell tumors, neuroblastoma).

3. Symptomatic central nervous system involvement.

4. Impaired cardiac function or clinically significant heart disease.

5. Prior Red Blood Cell (RBC) transfusions were received <3 months prior to the start of anti-CD 47 antibodies.

6. Less than or equal to 3 months prior to the initiation of the anti-CD 47 antibody, prior autologous stem cell transplantation was received.

7. Prior allogeneic stem cell transplantation with standard or reduced pretreatment was performed less than or equal to 6 months prior to the initiation of the anti-CD 47 antibody.

8. Less than 5 half-lives or 4 weeks (whichever is shorter) before initiation of the anti-CD 47 antibody, received prior systemic cancer-directed therapy or trial drug.

9. Extensive surgery was performed less than or equal to 2 weeks before the start of the anti-CD 47 antibody.

10. A pregnant or lactating female.

11. Known HIV infection.

12. Known as chronic hepatitis B or hepatitis C (HBV/HCV).

13. Treatment with long-term, therapeutic administration of anticoagulants continues.

14. A history of autoimmune hemolytic anemia or autoimmune thrombocytopenia.

15. A second concurrent cancer history requiring aggressive, sustained systemic treatment.

All references (e.g., publications or patents or patent applications) cited herein are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual reference (e.g., publication or patent application) was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

Other embodiments are within the scope of the following claims.

50页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:抗CLDN-5抗体及含有该抗体的药物

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!