Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AGONIST ANTIBODIES AGAINST HUMAN CD137 IN CANCER THAT EXPRESS MHC I
Document Type and Number:
WIPO Patent Application WO/2019/199896
Kind Code:
A9
Abstract:
The present disclosure relates to methods of treating, or ameliorating one or more symptoms of cancer, using agonistic antibodies, and antigen-binding fragments thereof, that bind CD137, wherein the cancer cells express major histocompatibility complex I (MHC I).

Inventors:
ESKIOCAK UGUR (US)
Application Number:
PCT/US2019/026675
Publication Date:
March 05, 2020
Filing Date:
April 09, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
COMPASS THERAPEUTICS LLC (US)
International Classes:
C07K16/28; A61P35/00; C07K16/30; A61K39/00
Attorney, Agent or Firm:
MANDRAGOURAS, Amy, E. et al. (US)
Download PDF:
Claims:
WE CLAIM:

1. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

2. The method of claim 1, wherein the MHC I expression is detected by the presence of any one or more markers selected from the group consisting of: MHC I protein, MHC I mRNA, beta- 2-microglobulin chain (b2M) protein, b2M chain mRNA, MHC class I alpha chain protein, and MHC class I alpha chain mRNA.

3. The method of claim 2, wherein the MHC I expression is detected with an agent directed to any one or more of the markers.

4. The method of claim 3, wherein the agent is an antibody or a nucleic acid probe.

5. The method of claim 4, wherein the antibody or probe comprises a detectable label.

6. The method of claim 2, wherein the marker is detected using a method selected from any one or more of the following: reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR, real-time RT-PCR, RNase protection assay (RPA), northern blotting, nucleic acid microarray using DNA, western blotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), tissue immuno staining, immunoprecipitation assay, complement fixation assay, fluorescence-activated cell sorting (FACS), mass spectrometry, magnetic bead-antibody immunoprecipitation, or protein chip.

7. The method of any one of claims 1-6, wherein the agonist monoclonal antibody or antigen binding portion thereof binds human CD137 with an affinity (KD) of about 30-100 nM.

8. The method of any one of claims 1-7, wherein the agonist monoclonal antibody or antigen binding portion comprises a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid.

9. The method of any one of claims 1-8, wherein the agonist monoclonal antibody or antigen binding portion comprises a heavy chain CDR3 comprising the amino acid sequence DX1X2X3X4LX5X6X7X8YX9YYX10, wherein Xi is any amino acid, wherein X2 is a non-polar amino acid, wherein X3 is a non-polar amino acid, wherein X4 is any amino acid, wherein X5 is a polar amino acid, wherein X6 is any amino acid, wherein X7 is any amino acid, wherein X8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid.

10. The method of any one of claims 1-9, wherein the agonist monoclonal antibody or antigen binding portion thereof specifically binds to an epitope on human CD 137 comprising one or more of residues El 11, T113, K114, N126, 1132 and P135 of SEQ ID NO: 3.

11. The method of any one of claims 1-10, wherein the agonist monoclonal antibody or antigen binding portion thereof specifically binds to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

12. The method of any one of claims 1-11, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises an epitope comprising 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

13. The method of claim 12, wherein the epitope comprises 6 amino acid residues.

14. The method of any one of claims 1-13, wherein the agonist monoclonal antibody or antigen binding portion thereof specifically binds to an epitope comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3).

15. The method of any one of claims 1-14, wherein the agonist monoclonal antibody or antigen binding portion thereof specifically binds to an epitope comprising the residues EFTK of SEQ ID NO: 3 (corresponding to amino acid residues 111-114 of SEQ ID NO: 3).

16. The method of any one of claims 1-15, wherein the agonist monoclonal antibody or antigen binding portion thereof specifically binds to an epitope, wherein said epitope is a non-linear epitope.

17. The method of any one of claims 1-16, wherein the agonist monoclonal antibody or antigen binding portion thereof has abrogated binding when a mutation of residue K114 of SEQ ID NO: 3 is present.

18. The method of any one of claims 1-17, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises a heavy chain CDR3 comprising the amino acid sequence DXPFXLDXXYYYYYX, wherein X is any amino acid.

19. The method of claim 18, wherein mutation of residues D95, L100, Y 100E, Y 100G, Y 100H, or combinations thereof, of the heavy chain CDR3, results in loss of binding to human CD 137.

20. The method of any one of claims 18 or 19, wherein the agonist monoclonal antibody or antigen binding portion thereof has a loss of binding to human CD 137 as a result of the mutation of residues P97, F98, D100A, Y100D, Y100F, or combinations thereof to alanine.

21. The method of any one of claims 18 or 19, wherein the monoclonal antibody or antigen binding portion thereof has increased binding to human CD 137 as a result of the mutation of residues P97, F98, D100A, Y 100D, Y 100F, or combinations thereof to any residue except alanine.

22. The method of any one of claims 8, 9, or 19, wherein X is any amino acid except for alanine.

23. The method of claim 9, wherein X2 is proline, wherein X3 is phenylalanine or tryptophan, wherein X5 is aspartic acid or glutamic acid wherein X8 is tyrosine, and wherein X9 is tyrosine.

24. The method of any one of claims 1-23, wherein the agonist monoclonal antibody or antigen binding portion thereof binds human CD137 with an affinity (KD) of about 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55-75 nM, 40-70 nM, 50-80 nM, or 60-90 nM.

25. The method of any one of claims 1-24, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain CDRs, wherein heavy chain CDR3 comprises the amino acid sequence set forth in SEQ ID NO: 68.

26. The method of any one of claims 1-25, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; and

(b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively.

27. The method of any one of claims 1-26, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence of SEQ ID NO: 6.

28. The method of any one of claims 1-27, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively; and (b) SEQ ID NO: 101 and 6, respectively.

29. The method of any one of claims 1-28, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 6.

30. The method of any one of claims 1-29, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively; and

(b) SEQ ID NO: 101 and 6, respectively.

31. The method of any one of claims 1-30, wherein the agonist monoclonal antibody or antigen binding portion thereof specifically binds to and agonizes human CD 137.

32. A method of inducing or enhancing one or more of the following in a cancer cell:

(a) dimerization of CD 137 trimers;

(b) multimerization of CD 137 trimers;

(c) human CDl37-mediated T cell activation;

(d) human CDl37-mediated cytotoxic T cell response;

(e) human CDl37-mediated T cell proliferation; and

(f) human CDl37-meduated cytokine production,

comprising contacting the cancer cell with an effective amount of an agonist monoclonal antibody that specifically binds human CD137, or antigen binding portion thereof, and wherein the cancer cell expresses MHC I.

33. The method of claim 32, wherein the agonist monoclonal antibody or antigen binding portion thereof induces or enhances dimerization of CD 137 trimers in a manner that is not Fc receptor binding dependent.

34. The method of claim 32, wherein the agonist monoclonal antibody or antigen binding portion thereof induces or enhances dimerization of CD 137 trimers in a manner which is enhanced by Fc receptor binding.

35. The method of any one of claims 1-34, wherein the agonist monoclonal antibody or antigen binding portion thereof cross-reacts with cynomolgus CD 137 and/or mouse CD 137.

36. The method of any one of claims 1-35, wherein the agonist monoclonal antibody is selected from the group consisting of an IgGl, an IgG2, and IgG3, an IgG4, and IgM, and IgAl, and IgA2, and IgD, and an IgE antibody.

37. The method of any one of claims 1-36, wherein the agonist monoclonal antibody is an IgGl antibody or IgG4 antibody.

38. The method of any one of claims 1-37, wherein treatment with the agonist monoclonal antibody or antigen binding portion thereof reduces or inhibits tumor growth.

39. The method of any one of claims 1-38, wherein treatment with the agonist monoclonal antibody or antigen binding portion thereof treats a disorder mediated by human CD 137 in a subject.

40. The method of any one of claims 1-39, wherein treatment with the agonist monoclonal antibody or antigen binding portion thereof treats cancer in a subject.

41. The method of claim 40, wherein the cancer is selected from the group consisting of melanoma, glioma, renal, colon, lung, prostate, breast, and head and neck cancer.

42. The method of any one of claims 1-41, wherein the agonist monoclonal antibody or antigen binding portion thereof binds Fc gamma receptor.

43. The method of any one of claims 38-41, wherein depletion of CD4+ T cells, CD8+ T cells, Natural Killer cells, or combinations thereof, reduces the efficacy of the antibody or antigen binding portion thereof.

44. The method of any one of claims 1-43, wherein the agonist monoclonal antibody does not significantly induce or enhance intrahepatic and/or intrasplenic T cell activation and/or T cell proliferation.

45. The method of any one of claims 1-44, wherein the agonist monoclonal antibody binds to human CD137 with an equilibrium dissociation constant KD of 1 x 106 or less.

46. The method of any one of claims 1-45, wherein the agonist monoclonal antibody is selected from the group consisting of: human antibody, humanized antibody, chimeric antibody, and deimmunized antibody.

47. A method of detecting MHC I in a subject having cancer, comprising:

(i) contacting a biological sample comprising cancer cells from the subject with an agent directed to MHC I; and

(ii) detecting the agent bound to MHC I; and

(iii) administering to the subject an agonist anti-CD 137 antibody, or antigen-binding fragment thereof.

48. A method of determining whether a subject having cancer would be amenable to agonist CD137 antibody therapy, comprising:

(i) contacting a biological sample comprising cancer cells from the subject with an agent directed to MHC I; and

(ii) detecting the agent bound to MHC I, wherein the presence of MHC I indicates that the cancer is amenable to treatment with an agonist CD 137 antibody, or antigen-binding fragment thereof.

49. The method of claim 48, wherein the method further comprises determining that the cancer is amenable to treatment with an agonist CD 137 antibody, or antigen-binding fragment thereof, and administering to the subject a therapeutically effective amount of an agonist anti-CD 137 antibody, or antigen-binding fragment thereof.

50. The method of any one of claims 47-49, wherein detecting MHC I is performed by detecting any one or more markers selected from the group consisting of: MHC I protein, MHC I mRNA, beta-2-microglobulin chain (b2M) protein, b2M chain mRNA, MHC class I alpha chain protein, and MHC class I alpha chain mRNA.

51. The method of claim 47 or 48, wherein the agent is an antibody or a nucleic acid probe.

52. The method of claim 51, wherein the antibody or probe comprises a detectable label.

53. The method of claim 47 or 48, wherein the detecting is performed by a reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR, real-time RT-PCR, RNase protection assay (RPA), northern blotting, nucleic acid microarray using DNA, western blotting, enzyme- linked immunosorbent assay (ELISA), radioimmunoassay (RIA), tissue immunostaining, immunoprecipitation assay, complement fixation assay, fluorescence-activated cell sorting (FACS), mass spectrometry, magnetic bead-antibody immunoprecipitation, protein chip, or any combination thereof.

54. The method of any one of claims 1-7, wherein the agonist monoclonal antibody or antigen binding portion thereof binds to an epitope on human CD137 comprising Kl 14 of SEQ ID NO: 3.

55. The method of claim 54, wherein the epitope comprises residues El 11, Tl 13, and Kl 14 of SEQ ID NO: 3.

56. The method of any one of claims 54-55, wherein the epitope comprises residues El 11, T113, K114, N126 and 1132 of SEQ ID NO: 3.

57. The method of any one of claims 54-56, wherein the epitope comprises residues El 11, Tl 13, Kl 14, N126, 1132 and P135 of SEQ ID NO: 3.

58. The method of claim 54, wherein the epitope comprises one or more residues El l l, Tl l3, K114, N126, 1132 and P135 of SEQ ID NO: 3.

59. The method of claim 54, wherein the agonist monoclonal antibody or antigen binding portion thereof binds to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

60. The method of claim 59, wherein the epitope comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,

14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

61. The method of claim 54, wherein the agonist monoclonal antibody or antigen binding portion thereof binds to an epitope comprising ELTK (corresponding to amino acid residues 111- 114 of SEQ ID NO: 3).

62. The method of claim 61 , wherein the epitope further comprises one or more residues N 126, 1132 and P135 of SEQ ID NO: 3.

63. The method of any one of claims 54-62, wherein the epitope is a non-linear epitope.

64. The method of any one of claims 54-63, wherein mutation of residue Kl 14 of SEQ ID NO:

3 abrogates binding of the agonistic isolated monoclonal antibody or antigen binding portion thereof.

65. The method of any one of claims 54-64, wherein the agonistic isolated monoclonal antibody or antigen binding portion binds human CD137 with an affinity (KD) of about 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55-75 nM, 40-70 nM, 50-80 nM, or 60-90 nM.

66. The method of any one of claims 54-65, wherein the agonist monoclonal antibody or antigen binding portion thereof binds to a non-ligand binding region of the extracellular domain of human CD 137.

67. The method of any one of claims 1-6, wherein the agonistic isolated monoclonal antibody or antigen binding portion thereof:

(i) binds human CD137 with an affinity (KD) of about 30-100 nM;

(ii) binds to a non-ligand binding region of the extracellular domain of human CD 137; and

(iii) binds to an epitope on human CD137 comprising Kl 14, of SEQ ID NO: 3.

68. The method of any one of claims 1-6, wherein the agonistic isolated monoclonal antibody or antigen binding portion thereof:

(i) binds human CD137 with an affinity (KD) of about 30-100 nM;

(ii) does not inhibit the interaction between human CD 137 and human CD 137 ligand (CD137L); and

(iii) binds to an epitope on human CD137 comprising Kl 14 of SEQ ID NO: 3.

69. The method of any one of claims 54-67, wherein the agonist monoclonal antibody or antigen binding portion thereof does not inhibit the interaction between CD 137 and CD137L.

70. The method of any one of claims 66-67, wherein the non-ligand binding region spans cysteine rich domain (CRD) III and CRD IV.

71. The method of any one of claims 54-70, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid.

72. The method of any one of claims 54, 55, and 63-71, wherein the epitope comprises El 11, T113, K114 and P135 of SEQ ID NO: 3.

73. The method of any one claims 54-71, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises a heavy chain CDR3 comprising the amino acid sequence DXPFXLDXXYYYYYX (SEQ ID NO: 127), wherein X is any amino acid.

74. The method of any one of claims 71-73, wherein mutation of residues D95, L100, Y 100E, Y100G, Y100H, or combinations thereof, of the heavy chain CDR3, to alanine results in loss of binding to human CD 137.

75. The method of any one of claims 71-74, wherein mutation of residues P97, F98, D100A, Y100D, Y100F, or combinations thereof, to alanine results in reduction of binding to human CD137.

76. The method of any one of claims 54-75, wherein the agonistic isolated monoclonal antibody or antigen binding portion does not inhibit the formation of a trimer of CDl37:CDl37L monomers.

77. The method of any one of claims 54, 65, 66, 69-71 and 73-76, wherein the agonist monoclonal antibody or antigen binding portion thereof binds to an epitope located within amino acid residues 111-135 of SEQ ID NO: 3.

78. The method of claim 77, wherein the epitope is a non-linear epitope.

79. The method of any one of claims 54-78, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain CDRs, wherein heavy chain CDR3 comprises the amino acid sequence set forth in SEQ ID NO: 68.

80. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; and

(b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively.

81. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence of SEQ ID NO: 6.

82. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 4 and 6, respectively; and

(b) SEQ ID NOs: 101 and 6, respectively.

83. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 6.

84. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 4 and 6, respectively; and

(b) SEQ ID NOs: 101 and 6, respectively.

85. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chains comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 129 and 133, respectively; and

(b) SEQ ID NOs: 131 and 133, respectively.

86. The method according to any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 70, 79 and 90, respectively;

(c) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 71, 80 and 91, respectively;

(d) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 72, 81 and 92, respectively;

(e) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 73, 82 and 91, respectively; (f) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 74, 83 and 93, respectively;

(g) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 75, 84 and 91, respectively;

(h) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 74, 85 and 94, respectively;

(i) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 76, 86 and 95, respectively;

(j) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 77, 87 and 93, respectively;

(k) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 88 and 90, respectively;

(l) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 57 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(m) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 58 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(n) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 59 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(o) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 60 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; (p) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 61 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(q) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 58 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(r) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 62 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(s) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 52, 63 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(t) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 64 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(u) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 65 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(v) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(w) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 107, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; and

(x) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 109, 110 and 92, respectively.

87. The method according to any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 101 and 103; and wherein the light chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46 and 105.

88. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively; and

(b) SEQ ID NOs: 102 and 7, respectively.

89. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences having at least 90% identity to the nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively; and

(b) SEQ ID NOs: 102 and 7, respectively.

90. The method of claim 89, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences having at least 90% identity to SEQ ID NOs: 5 and 7, respectively.

91. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NO: 5 and 7, respectively;

(b) SEQ ID NO: 5 and 29, respectively;

(c) SEQ ID NO: 5 and 31, respectively;

(d) SEQ ID NO: 5 and 33, respectively;

(e) SEQ ID NO: 5 and 35, respectively;

(f) SEQ ID NO: 5 and 37, respectively;

(g) SEQ ID NO: 5 and 39, respectively; (h) SEQ ID NO: 5 and 41, respectively;

(i) SEQ ID NO: 5 and 43, respectively;

(j) SEQ ID NO: 5 and 45, respectively;

(k) SEQ ID NO: 5 and 47, respectively;

(l) SEQ ID NO: 9 and 7, respectively;

(m) SEQ ID NO: 11 and 7, respectively;

(n) SEQ ID NO: 13 and 7, respectively;

(o) SEQ ID NO: 15 and 7, respectively;

(p) SEQ ID NO: 17 and 7, respectively;

(q) SEQ ID NO: 19 and 7, respectively;

(r) SEQ ID NO: 21 and 7, respectively;

(s) SEQ ID NO: 23 and 7, respectively;

(t) SEQ ID NO: 25 and 7, respectively;

(u) SEQ ID NO: 27 and 7, respectively;

(v) SEQ ID NO: 102 and 7, respectively;

(w) SEQ ID NO: 104 and 7, respectively; and

(x) SEQ ID NO: 5 and 106, respectively.

92. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively;

(b) SEQ ID NO: 4 and 28, respectively;

(c) SEQ ID NO: 4 and 30, respectively;

(d) SEQ ID NO: 4 and 32, respectively;

(e) SEQ ID NO: 4 and 34, respectively;

(f) SEQ ID NO: 4 and 36, respectively;

(g) SEQ ID NO: 4 and 38, respectively;

(h) SEQ ID NO: 4 and 40, respectively;

(i) SEQ ID NO: 4 and 42, respectively;

(j) SEQ ID NO: 4 and 44, respectively; (k) SEQ ID NO: 4 and 46, respectively;

(D SEQ ID NO: 8 and 6, respectively;

(m) SEQ ID NO: 10 and 6, respectively;

(n) SEQ ID NO: 12 and 6, respectively;

(o) SEQ ID NO: 14 and 6, respectively;

(P) SEQ ID NO: 16 and 6, respectively;

(q) SEQ ID NO: 18 and 6, respectively;

(r) SEQ ID NO: 20 and 6, respectively;

(s) SEQ ID NO: 22 and 6, respectively;

(t) SEQ ID NO: 24 and 6, respectively;

(u) SEQ ID NO: 26 and 6, respectively;

(V) SEQ ID NO: 101 and 6, respectively;

(w) SEQ ID NO: 103 and 6, respectively; and

(x) SEQ ID NO: 4 and 105, respectively.

93. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 101 and 103; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46 and 105.

94. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively;

(b) SEQ ID NO: 4 and 28, respectively;

(c) SEQ ID NO: 4 and 30, respectively;

(d) SEQ ID NO: 4 and 32, respectively; (e) SEQ ID NO: 4 and 34, respectively;

(f) SEQ ID NO: 4 and 36, respectively;

(g) SEQ ID NO: 4 and 38, respectively;

(h) SEQ ID NO: 4 and 40, respectively;

(i) SEQ ID NO: 4 and 42, respectively;

CD SEQ ID NO: 4 and 44, respectively;

(k) SEQ ID NO: 4 and 46, respectively;

(l) SEQ ID NO: 8 and 6, respectively;

(m) SEQ ID NO: 10 and 6, respectively;

(n) SEQ ID NO: 12 and 6, respectively;

(o) SEQ ID NO: 14 and 6, respectively;

(P) SEQ ID NO: 16 and 6, respectively;

(q) SEQ ID NO: 18 and 6, respectively;

(r) SEQ ID NO: 20 and 6, respectively;

(s) SEQ ID NO: 22 and 6, respectively;

(t) SEQ ID NO: 24 and 6, respectively;

(u) SEQ ID NO: 26 and 6, respectively;

(V) SEQ ID NO: 101 and 6, respectively;

(w) SEQ ID NO: 103 and 6, respectively; and

(x) SEQ ID NO: 4 and 105, respectively.

95. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain sequences comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 129 and 133, respectively; and

(b) SEQ ID NOs: 131 and 133, respectively.

96. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain sequences having amino acid sequences set forth in SEQ ID NOs: 129 and 133, respectively.

97. The method of any one of claims 54-79, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain sequences having amino acid sequences set forth in SEQ ID NOs: 131 and 133, respectively.

98. The method of any one of claims 54-97, wherein the agonist monoclonal antibody or antigen binding portion thereof exhibits at least one or more of the following properties selected from the group consisting of:

(a) induces or enhances dimerization of CD 137 trimers;

(b) induces or enhances multimerization of CD 137 trimers;

(c) induces or enhances T cell activation;

(d) induces or enhances a cytotoxic T cell response;

(e) induces or enhances T cell proliferation;

(f) induces or enhances immune cell cytokine production; and

(g) any combination of properties (a)-(f).

99. The method of any one of claims 54-98, wherein the agonist monoclonal antibody or antigen binding portion thereof exhibits at least one or more of the following properties relative to a reference antibody that binds human CD137, selected from the group consisting of:

(a) does not induce or enhance intrahepatic T cell activation;

(b) does not induce or enhance intrahepatic T cell proliferation;

(c) does not induce or enhance intrasplenic T cell activation;

(d) does not induce or enhance intrasplenic T cell proliferation;

(e) does not induce or enhance macrophage activation;

(f) does not induce or enhance macrophage differentiation;

(g) does not induce or enhance alanine aminotransferase (ALT) activity; and

(h) any combination of properties (a) - (g).

100. The method of claim 99, wherein the reference antibody is urelumab.

101. The method of any one of claims 98-100, wherein the properties of the agonist monoclonal antibody or antigen binding portion thereof are not dependent on Fc receptor binding.

102. The method of any one of claims 98-100, wherein the properties of the agonist monoclonal antibody or antigen binding portion thereof are enhanced by Fc receptor binding.

103. The method of any one of claims 54-102, wherein the agonist monoclonal antibody or antigen binding portion thereof cross-reacts with cynomolgus CD137, mouse CD137, or both.

104. The method of any one of claims 54-94 and 97-103, wherein the agonistic isolated monoclonal antibody is selected from the group consisting of an IgGl, an IgG2, an IgG3, an IgG4, an IgM, an IgAl, an IgA2, an IgD, and an IgE antibody.

105. The method of claim 104, wherein the agonistic isolated monoclonal antibody is an IgGl antibody or IgG4 antibody.

106. The method of any one of claims 54-94 and 97-103, wherein the agonistic isolated monoclonal antibody comprises a wild-type human IgGl or wild type human IgG4 heavy chain constant region.

107. The method of any one of claims 54-94 and 97-103, wherein the agonistic isolated monoclonal antibody comprises a mutant IgGl heavy chain constant region.

108. The method of any one of claims 54-94 and 97-103, wherein the agonistic isolated monoclonal antibody comprises a mutant IgG4 heavy chain constant region.

109. The method of claim 108, wherein the mutant IgG4 heavy chain constant region comprises a substitution at Ser228.

110. The method of claim 109, wherein the mutant IgG4 heavy chain constant region comprises an S228P substitution.

111. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof induces or enhances T cell activation in the subject.

112. The method of claim 111, wherein the T cell activation occurs in a tumor microenvironment.

113. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof induces or enhances a cytotoxic T cell response in the subject.

114. The method of claim 113, wherein the cytotoxic T cell response occurs in a tumor microenvironment.

115. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof induces or enhances cytokine production of an immune cell in the subject.

116. The method of claim 115, wherein the cytokine produced is IL-2, TNFa, IL-13, IFNy, or combinations thereof.

117. The method of claim 115 or claim 116, wherein the cytokine production occurs in a tumor microenvironment.

118. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof induces or enhances T cell proliferation in the subject.

119. The method of claim 118, wherein the T cell proliferation occurs in a tumor microenvironment.

120. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof reduces or inhibits tumor growth in the subject.

121. The method of any one of the preceding claims, wherein the agonistic isolated monoclonal antibody or antigen binding fragment thereof increases infiltration of immune cells into a tumor microenvironment in the subject.

122. The method of claim 121, wherein the immune cells express CD45.

123. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof reduces the quantity of T regulatory (Treg) cells in a tumor microenvironment in the subject.

124. The method of claim 123, wherein the Treg cells express CD4, FOXP-3 and CD25.

125. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof reduces the quantity of macrophages is reduced in a tumor microenvironment in the subject.

126. The method of claim 125, wherein the macrophages express CD45 and CD1 lb.

127. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof reduces T cell exhaustion in a tumor microenvironment in the subject.

128. The method of claim 127, wherein reduction of T cell exhaustion comprises a decrease in expression of TIGIT, PD-l, LAG-3, or combinations thereof.

129. The method of any one of the preceding claims, wherein the cancer is selected from the group consisting of melanoma, glioma, renal, breast, hematological, and head and neck cancer.

130. The method of claim 129, wherein the hematological cancer is a B cell lymphoma.

131. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof induces an anti-tumor memory immune response in the subject.

132. The method of any one of the preceding claims, wherein the agonist monoclonal antibody or antigen binding portion thereof binds Fc gamma receptor.

133. The method of any one of the preceding claims, wherein depletion of CD4+ T cells, CD8+ T cells, Natural Killer cells, or combinations thereof, reduces the efficacy of the agonistic isolated monoclonal antibody or antigen binding portion thereof.

Description:
AGONIST ANTIBODIES AGAINST HUMAN CD137 IN CANCER THAT EXPRESS MHC I

CROSS-REFERNCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Patent Application Serial No. 62/655,066, filed on April 9, 2018 and U.S. Provisional Patent Application Serial No.

62/755,644, filed on November 5, 2018. The content of these applications is hereby incorporated by reference in their entirety.

BACKGROUND

In recent years, an increasing body of evidence suggests the immune system operates as a significant barrier to tumor formation and progression. The principle that naturally-occurring T cells with anti-tumor potential or activity exist in a patient with cancer has rationalized the development of immunotherapeutic approaches in oncology. Immune cells, such as T cells, macrophages, and natural killer cells, can exhibit anti-tumor activity and effectively control the occurrence and growth of malignant tumors. Tumor- specific or -associated antigens can induce immune cells to recognize and eliminate malignancies (Chen & Mellman, (2013) Immunity 39(1): 1-10). In spite of the existence of tumor- specific immune responses, malignant tumors often evade or avoid immune attack through a variety of immunomodulatory mechanisms resulting in the failure to control tumor occurrence and progression (Motz & Coukos, (2013) Immunity 39(l):6l-730). Indeed, an emerging hallmark of cancer is the exploitation of these immunomodulatory mechanisms and the disablement of anti-tumor immune responses, resulting in tumor evasion and escape from immunological killing (Hanahan and Weinberg (2011) Cell 144(5):646-674).

Novel approaches in the immunotherapy of cancer involve counteracting these immune evasion and escape mechanisms and inducing the endogenous immune system to reject tumors. CD137 (alternatively known as “tumor necrosis factor receptor superfamily member 9” (TNFRSF9), 4-1BB, and“induced by lymphocyte activation” (ILA)) is a transmembrane co stimulatory receptor protein belonging to the tumor necrosis factor superfamily. CD 137 is a T cell co-stimulatory receptor induced upon TCR activation (Nam et ah, (2005) Curr Cancer Drug Targets 5:357-363; Watts et ah, (2005) Annu Rev Immunol 23:23-68). In addition to its expression on activated CD4+ and CD8+ T cells, CD 137 is also expressed on CD4+CD25+ regulatory T cells, activated natural killer (NK) and NK-T cells, monocytes, neutrophils, and dendritic cells.

Under physiological conditions, CD137 is ligated by CD137 ligand (CD137L), an agonist membrane molecule present on antigen-presenting cells including B cells, monocytes, macrophages, and dendritic cells (Watts et ah, (2005) Annu Rev Immunol 23:23-68). Upon interaction with its ligand, CD 137 leads to increased TCR-induced T-cell proliferation, cytokine production, functional maturation, and prolonged CD8+ T-cell survival. The potential of CD 137 co-stimulation using various agonists (e.g. agonistic antibodies, recombinant CD 137L protein, and CD 137- specific aptamers) in enabling the immune system to attack tumors has been documented in numerous models (Dharmadhikari et ah, (2016) Oncoimmunology 5(4):el 113367 and references therein). A recent report on the clinical evaluation of an agonistic CD 137 antibody (Urelumab, BMS-663513; Bristol-Myers Squibb) documented the observation of treatment- related adverse events in human subjects, including indications of severe hep ato toxicity (transaminitis) correlating with antibody dose (Segal et ah, (2016) Clin Cancer Res 23(8): 1929- 1936). In contrast, a different agonistic CD137 antibody (Utomilumab, PF-05082566; Pfizer) tested in combination with an anti-PD-l antibody (pembrolizumab), though not resulting in any dose-limiting toxicities, showed comparable results to anti-PD-l antibody therapy alone (Tolcher, A. et ah, (2017) Clin Cancer Res 23(18): 5349-5357). These results highlight that for patients with various diseases and conditions, including cancer, that are amenable to treatment with a CD137 agonist, there continues to be an unmet need for novel agonistic antibodies that bind to human CD 137 and exhibit characteristics sufficient for the development of a safe and efficacious therapeutic.

SUMMARY OF THE DISCLOSURE

The present disclosure is based, in part, on the discovery that cancer cells expressing MHC I are more responsive to anti-cancer therapy using anti-CDl37 agonist antibodies, and antigen-binding fragments thereof, disclosed herein. Thus, the present discovery can be applied to identify a cancer patient population that would be particularly amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein.

Unexpectedly, the anti-CD 137 monoclonal antibodies of the disclosure were found to agonize CD 137 and induce protective anti-tumor immunity in vivo with a concomitant reduction in the potential for toxicity-related events. Notably, the anti-CDl37 antibodies described herein are efficacious against diverse tumor types, and over a wide dose range. The anti-CD 137 agonist antibodies of the disclosure were also found to induce and/or enhance cytokine production, expansion of CD8+ T cells, and protective anti-tumor immunity and thus are particularly effective in treating cancer. Moreover, the anti-CD 137 agonist antibodies of the disclosure were found to bind a unique epitope on human CD 137. In addition, the disclosure also features agonist anti- CD 137 antibodies having an affinity (KD) that is optimal for maximizing anti-tumor immunity while avoiding toxicity-related events associated with CD137 agonism. Moreover, as exemplified in the working examples, the antibodies described herein are therapeutically effective against very large tumors. For example, treatment of tumor-bearing mice with agonist anti-CD 137 antibodies described herein resulted in complete regression of tumors as large as 1,800 mm 3 . As set forth in FIG. 15, treatment of such mice also resulted in protective immunity. And coincident with the observed efficacy were positive immunophenotypic changes in the tumor microenvironment, such as increased immune cell infiltration with concomitant reductions in regulatory T cell and exhausted T cell populations (see, e.g., FIGs. 22A-22D).

As described above, agonism of CD 137 has been associated with certain adverse events, including hepatotoxicity-related deaths in humans (see, e.g., Segal et al. (2017) Clin Cancer Res 23(8): 1929-1935). Similar toxicities resulting from treatment with agonist anti-CDl37 antibodies (such as the 3H3 antibody) have also been observed in animal models (see, e.g., Bartkowiak et al. (2018) Clin Cancer Res 24(5): 1138- 1151). Yet, the agonist anti-CDl37 antibodies described herein have minimal effects on the liver, as determined by, e.g., plasma levels of liver enzymes (e.g., alanine aminotransferase (ALT)) and immune cell infiltration. For example, there was no evidence of increased intrahepatic or intrasplenic immune cell infiltration in mice treated with the antibodies. Thus, the CD137 binding antibodies described herein are not only highly efficacious, but also sparing of certain toxicities associated with CD 137 agonism.

While the disclosure is not bound by any particular theory or mechanism of action, the superior therapeutic and toxicity-sparing properties of the CD 137 binding antibodies described herein are believed to derive in part from one or both of their affinity and the novel epitope to which they bind. That is, the antibodies described herein share a common, novel epitope that is distinct from that of other agonist anti-CD 137 antibodies. And, as exemplified in the working examples, engagement of this epitope by the antibodies described herein gives rise to differentiated in vitro activity, such as effects on regulatory T cell proliferation, cytokine production by CD8 + T cells and macrophages, and intracellular signaling, as compared to agonist antibodies that bind to different epitopes of CD 137. Furthermore, it has been demonstrated that an affinity range (a“sweet spot”) for CD 137 binding antibodies is particularly optimal for anti tumor activity. For example, antibodies of intermediate affinity were shown to be more efficacious against large tumors as compared to antibodies with higher or lower affinity.

These characteristics provide antibodies with superior in vivo anti-tumor efficacy, which efficacy is enhanced against cancer cells that express MHC I.

Accordingly, in some aspects, the disclosure provides a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

In some aspects, the MHC I expression is detected by the presence of any one or more markers selected from the group consisting of: MHC I protein, MHC I mRNA, beta-2- microglobulin chain (b2M) protein, b2M chain mRNA, MHC class I alpha chain domain protein (e.g., ocl, oc2, or oc3), and MHC class I alpha chain domain mRNA. As commonly known in the art, the alpha chains are encoded by any one of human leukocyte antigen (HLA) -A, -B, and -C genes, which are three of the major types of human MHC class I cell surface receptors. Thus, those skilled in the art would recognize that any one or more of the class I alpha chains and/or their specific domains (e.g., ocl, oc2, or oc3), can be suitable for detecting MHC I expression. In some embodiments, the class I alpha chain is an HLA-A alpha chain. In some aspects, the class I alpha chain is a HLA-B alpha chain. In some embodiments, the class I alpha chain is an HLA-C alpha chain.

In some aspects, MHC I expression is detected with an agent directed to any one or more of the markers. For example, the agent is an antibody or a nucleic acid probe. In some aspects, the antibody or probe comprises a detectable label. Those skilled in the art can readily determine the appropriate agent, e.g., antibody or probe, depending on the particular MHC I (e.g., HLA-A, HLA- B, or HLA-C) being detected. While the HLA-A, -B, and -C genes encoding the alpha chains of the MHC I complex are known to be polymorphic, those skilled in the art can readily determine their sequences, and select and/or design a suitable detecting agent directed to the alpha chain and/or the b2M chain. An exemplary amino acid sequence of MHC I (HLA-A) is shown in SEQ ID NO: 129 (its nucleotide sequence is shown in SEQ ID NO: 130); the amino acid sequence of b2M is shown in SEQ ID NO: 131 (its nucleotide sequence is shown in SEQ ID NO: 132).

In some aspects, the marker is detected using a method selected from any one or more of the following: reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR, real-time RT-PCR, RNase protection assay (RPA), northern blotting, nucleic acid microarray using DNA, western blotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), radioimmunodiffusion, Ouchterlony immunodiffusion, rocket Immunoelectrophoresis, tissue immunostaining, immunoprecipitation assay, complement fixation assay, fluorescence- activated cell sorting (FACS), mass spectrometry, magnetic bead-antibody immunoprecipitation, or protein chip. The protein or mRNA marker can be detected using any other suitable methods known in the art.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion (fragment) thereof is any one or more of the anti-CD 137 antibody disclosed herein. In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof binds human CD137 with an affinity (KD) of about 40-100 nM.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion comprises a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid. In some aspects, X is any amino acid except for alanine.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion comprises a heavy chain CDR3 comprising the amino acid sequence DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid. In some aspects, X 2 is proline, X 3 is phenylalanine or tryptophan, X5 is aspartic acid or glutamic acid, X 8 is tyrosine, and X9 is tyrosine.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion, binds to an epitope on human CD137 comprising Kl 14 of SEQ ID NO: 3. In some aspects, the epitope comprises residues El 11, Tl 13, and Kl 14 of SEQ ID NO: 3. In some aspects, the epitope comprises residues El 11, T113, K114, N126, 1132 and P135 of SEQ ID NO: 3. In some aspects, the epitope comprises one or more residues El 11, T113, K114, N126, 1132 and P135 of SEQ ID NO: 3.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion binds to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3. In some aspects, the epitope comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion binds to an epitope comprising ELTK (corresponding to amino acid residues 111- 114 of SEQ ID NO: 3). In some aspects, the epitope further comprises one or more residues N 126, 1132 and P135 of SEQ ID NO: 3.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion specifically binds to an epitope, wherein said epitope is a non-linear epitope.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion has abrogated binding when a mutation of residue K114 of SEQ ID NO: 3 is present.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion comprises a heavy chain CDR3 comprising the amino acid sequence DXPFXLDXXYYYYYX (SEQ ID NO: 127), wherein X is any amino acid. In some aspects, X is any amino acid except for alanine.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion has a loss of binding to human CD137 as a result of the mutation of residues P97, F98, D100A, Y100D, Y100F, or combinations thereof to alanine.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion has increased binding to human CD137 as a result of the mutation of residues P97, F98, D100A, Y 100D, Y 100F, or combinations thereof to any residue except alanine.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion binds to a non-ligand binding region of the extracellular domain of human CD137. In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion binds human CD137 with an affinity (K D ) of about 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55-75 nM, 40-70 nM, 50-80 nM, or 60-90 nM.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain CDRs, wherein heavy chain CDR3 comprises the amino acid sequence set forth in SEQ ID NO: 68.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion:

(i) binds human CD137 with an affinity (K D ) of about 30-100 nM;

(ii) binds to a non-ligand binding region of the extracellular domain of human CD 137; and

(iii) binds to an epitope on human CD137 comprising Kl 14, of SEQ ID NO: 3. In some aspects, the non-ligand binding region spans cysteine rich domain (CRD) III and CRD IV.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion:

(i) binds human CD137 with an affinity (K D ) of about 30-100 nM;

(ii) does not inhibit the interaction between human CD 137 and human CD 137 ligand (CD137L); and

(iii) binds to an epitope on human CD137 comprising Kl 14 of SEQ ID NO: 3. In some aspects, the non-ligand binding region spans cysteine rich domain (CRD) III and CRD IV.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion does not inhibit the interaction between CD 137 and CD137L.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion does not inhibit the formation of a trimer of CDl37:CDl37L monomers.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion comprises heavy and light chain CDRs selected from the group consisting of: (a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; and (b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively. In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence of SEQ ID NO: 6.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of: (a) SEQ ID NO: 4 and 6, respectively; and (b) SEQ ID NO: 101 and 6, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 6.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of: (a) SEQ ID NO: 4 and 6, respectively; and (b) SEQ ID NO: 101 and 6, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chains comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 129 and 133, respectively; and

(b) SEQ ID NOs: 131 and 133, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereofcomprises heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; (b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

70, 79 and 90, respectively;

(c) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

71, 80 and 91, respectively;

(d) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

72, 81 and 92, respectively;

(e) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

73, 82 and 91, respectively;

(f) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

74, 83 and 93, respectively;

(g) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

75, 84 and 91, respectively;

(h) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 74, 85 and 94, respectively;

(i) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

76, 86 and 95, respectively;

(j) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs:

77, 87 and 93, respectively;

(k) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 88 and 90, respectively; (l) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 57 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(m) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 58 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(n) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 59 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(o) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 60 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(p) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 61 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(q) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 58 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(r) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 62 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(s) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 52, 63 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(t) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 64 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(u) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 65 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; (v) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(w) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 107, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively;

(x) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 109, 110 and 92, respectively;

(y) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(z) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(aa) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(bb) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(cc) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(dd) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(ee) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively; (ff) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(gg) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(hh) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(ii) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(jj) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(kk) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(11) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively; and

(mm) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 101 and 103; and wherein the light chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46 and 105. In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively; and

(b) SEQ ID NOs: 102 and 7, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences having at least 90% identity to the nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively; and

(b) SEQ ID NOs: 102 and 7, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences having at least 90% identity to SEQ ID NOs: 5 and 7, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NO: 5 and 7, respectively;

(b) SEQ ID NO: 5 and 29, respectively;

(c) SEQ ID NO: 5 and 31, respectively;

(d) SEQ ID NO: 5 and 33, respectively;

(e) SEQ ID NO: 5 and 35, respectively;

(f) SEQ ID NO: 5 and 37, respectively;

(g) SEQ ID NO: 5 and 39, respectively;

(h) SEQ ID NO: 5 and 41, respectively;

(i) SEQ ID NO: 5 and 43, respectively;

(j) SEQ ID NO: 5 and 45, respectively;

(k) SEQ ID NO: 5 and 47, respectively;

(l) SEQ ID NO: 9 and 7, respectively;

(m) SEQ ID NO: 11 and 7, respectively;

(n) SEQ ID NO: 13 and 7, respectively; (o) SEQ ID NO: 15 and 7, respectively;

(p) SEQ ID NO: 17 and 7, respectively;

(q) SEQ ID NO: 19 and 7, respectively;

(r) SEQ ID NO: 21 and 7, respectively;

(s) SEQ ID NO: 23 and 7, respectively;

(t) SEQ ID NO: 25 and 7, respectively;

(u) SEQ ID NO: 27 and 7, respectively;

(v) SEQ ID NO: 102 and 7, respectively;

(w) SEQ ID NO: 104 and 7, respectively; and

(x) SEQ ID NO: 5 and 106, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively;

(b) SEQ ID NO: 4 and 28, respectively;

(c) SEQ ID NO: 4 and 30, respectively;

(d) SEQ ID NO: 4 and 32, respectively;

(e) SEQ ID NO: 4 and 34, respectively;

(f) SEQ ID NO: 4 and 36, respectively;

(g) SEQ ID NO: 4 and 38, respectively;

(h) SEQ ID NO: 4 and 40, respectively;

(i) SEQ ID NO: 4 and 42, respectively;

(j) SEQ ID NO: 4 and 44, respectively;

(k) SEQ ID NO: 4 and 46, respectively;

(l) SEQ ID NO: 8 and 6, respectively;

(m) SEQ ID NO: 10 and 6, respectively;

(n) SEQ ID NO: 12 and 6, respectively;

(o) SEQ ID NO: 14 and 6, respectively;

(p) SEQ ID NO: 16 and 6, respectively;

(q) SEQ ID NO: 18 and 6, respectively;

(r) SEQ ID NO: 20 and 6, respectively; (s) SEQ ID NO: 22 and 6, respectively;

(t) SEQ ID NO: 24 and 6, respectively;

(u) SEQ ID NO: 26 and 6, respectively;

(v) SEQ ID NO: 101 and 6, respectively;

(w) SEQ ID NO: 103 and 6, respectively; and

(x) SEQ ID NO: 4 and 105, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 101 and 103; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46 and 105.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively;

(b) SEQ ID NO: 4 and 28, respectively;

(c) SEQ ID NO: 4 and 30, respectively;

(d) SEQ ID NO: 4 and 32, respectively;

(e) SEQ ID NO: 4 and 34, respectively;

(f) SEQ ID NO: 4 and 36, respectively;

(g) SEQ ID NO: 4 and 38, respectively;

(h) SEQ ID NO: 4 and 40, respectively;

(i) SEQ ID NO: 4 and 42, respectively;

(j) SEQ ID NO: 4 and 44, respectively;

(k) SEQ ID NO: 4 and 46, respectively;

(l) SEQ ID NO: 8 and 6, respectively;

(m) SEQ ID NO: 10 and 6, respectively;

(n) SEQ ID NO: 12 and 6, respectively; (o) SEQ ID NO: 14 and 6, respectively;

(p) SEQ ID NO: 16 and 6, respectively;

(q) SEQ ID NO: 18 and 6, respectively;

(r) SEQ ID NO: 20 and 6, respectively;

(s) SEQ ID NO: 22 and 6, respectively;

(t) SEQ ID NO: 24 and 6, respectively;

(u) SEQ ID NO: 26 and 6, respectively;

(v) SEQ ID NO: 101 and 6, respectively;

(w) SEQ ID NO: 103 and 6, respectively; and

(x) SEQ ID NO: 4 and 105, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain sequences comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 129 and 133, respectively; and

(b) SEQ ID NOs: 131 and 133, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain sequences having amino acid sequences set forth in SEQ ID NOs: 129 and 133, respectively.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof comprises heavy and light chain sequences having amino acid sequences set forth in SEQ ID NOs: 131 and 133, respectively.

In some embodiments, the present disclosure also provides a method of inducing or enhancing one or more of the following in a cancer cell: (a) dimerization of CD 137 trimers; (b) multimerization of CD137 trimers; (c) human CDl37-mediated T cell activation; (d) human CDl37-mediated cytotoxic T cell response; (e) human CDl37-mediated T cell proliferation; and (f) human CDl37-meduated cytokine production, comprising contacting the cancer cell with an effective amount of an agonist monoclonal antibody that specifically binds human CD137, or antigen binding portion thereof, as disclosed herein, and wherein the cancer cell expresses MHC I. In some embodiments, the agonist monoclonal antibody or antigen binding portion thereof induces or enhances dimerization of CD 137 trimers in a manner that is not Fc receptor binding dependent.

In some embodiments, the agonist monoclonal antibody or antigen binding portion thereof induces or enhances dimerization of CD 137 trimers in a manner which is enhanced by Fc receptor binding.

In some embodiments, the agonist monoclonal antibody or antigen binding portion thereof cross-reacts with cynomolgus CD137 and/or mouse CD137.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof exhibits at least one or more of the following properties selected from the group consisting of:

(a) induces or enhances dimerization of CD 137 trimers;

(b) induces or enhances multimerization of CD 137 trimers;

(c) induces or enhances T cell activation;

(d) induces or enhances a cytotoxic T cell response;

(e) induces or enhances T cell proliferation;

(f) induces or enhances immune cell cytokine production; and

(g) any combination of properties (a)-(f).

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof exhibits at least one or more of the following properties relative to a reference antibody that binds human CD137, selected from the group consisting of:

(a) does not induce or enhance intrahepatic T cell activation;

(b) does not induce or enhance intrahepatic T cell proliferation;

(c) does not induce or enhance intrasplenic T cell activation;

(d) does not induce or enhance intrasplenic T cell proliferation;

(e) does not induce or enhance macrophage activation;

(f) does not induce or enhance macrophage differentiation;

(g) does not induce or enhance alanine aminotransferase (ALT) activity; and

(h) any combination of properties (a) - (g). In some aspects, the reference antibody is urelumab. In some aspects the agonistic antibody or antigen-binding fragment induces or enhances human CDl37-mediated T cell activation in the tumor microenvironment, but does not significantly induce or enhance human CDl37-mediated T cell activation in the spleen and/or liver. In some aspects, the agonistic antibody or antigen-binding fragment induces or enhances human CDl37-mediated cytotoxic T cell response in the tumor microenvironment, but does not significantly induce or enhance human CDl37-mediated cytotoxic T cell response in the spleen and/or liver. In some aspects, the agonistic antibody or antigen-binding fragment induces human CDl37-mediated T cell proliferation in the tumor microenvironment, but does not significantly induce human CDl37-mediated T cell proliferation in the spleen and/or liver. In some aspects, the agonistic antibody or antigen binding fragment induces or enhances human CDl37-mediated cytokine production in the tumor microenvironment, but does not significantly induce or enhance human CDl37-mediated cytokine production in the spleen and/or liver. In some aspects, the properties of the agonistic antibody or antigen-binding fragment are not Fc gamma receptor binding dependent. In some aspects, the properties of the agonistic antibody or antigen-binding fragment are enhanced by Fc gamma receptor binding.

In any of the foregoing or related aspects, the agonistic antibody or antigen-binding fragment cross-reacts with cynomolgus CD137 and/or mouse CD137. In any of the foregoing or related aspects, the agonistic antibody is selected from the group consisting of an IgGl, an IgG2, and IgG3, an IgG4, and IgM, and IgAl, and IgA2, and IgD, and an IgE antibody. In some aspects, the agonistic antibody is an IgGl antibody or IgG4 antibody.

In any of the foregoing or related aspects, the isolated agonistic monoclonal antibody comprises an IgGl heavy chain constant region or an IgG4 heavy chain constant region. In some aspects, the isolated agonistic monoclonal antibody comprises an IgGl heavy chain constant region. In some aspects, the IgGl heavy chain constant region is a wild-type human IgGl heavy chain constant region. In some aspects, the IgGl heavy chain constant region comprises an amino acid substitution relative to a wild-type human IgGl heavy chain constant region. In some aspects, the isolated agonistic monoclonal antibody comprises an IgG4 heavy chain constant region. In some aspects, the IgG4 heavy chain constant region is a wild-type human IgG4 heavy chain constant region. In some aspects, the IgG4 heavy chain constant region comprises an amino acid substitution relative to a wild-type human IgG4 heavy chain constant region. In some aspects, the amino acid substitution is an amino acid substitution at position Ser228 according to EU numbering. In some aspects, the amino acid substitution at position Ser228 is S228P. In some aspects, the IgG4 heavy chain constant region comprises an amino acid substitution relative to a wild-type human IgG4 heavy chain constant region, wherein the amino acid substitution is an amino acid substitution at position Ser228 according to EU numbering, and wherein amino acid substitution at position Ser228 is S228P.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof induces or enhances T cell activation in the subject. In some aspects, the T cell activation occurs in a tumor microenvironment.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof induces or enhances a cytotoxic T cell response in the subject. In some aspects, the cytotoxic T cell response occurs in a tumor microenvironment.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof induces or enhances cytokine production in the subject. In some aspects, the cytokine produced is IL-2, TNFoc, IL-13, IFNy, or combinations thereof. In some aspects, the cytokine produced is IF-2. In some aspects, the cytokine produced is TNFoc. In some aspects, the cytokine produced is IF-13. In some aspects, the cytokine produced is IFNy. In some aspects, the cytokine produced is IF-2 and TNFoc. In some aspects, the cytokine produced is IF-2 and IF-13. In some aspects, the cytokine produced is IF-2 and IFNy. In some embodiments, the cytokine produced is TNFoc and IF-13. In some aspects, the cytokine produced is TNFoc and IFNy. In some aspects, the cytokine produced is IF-13 and IFNy. In some aspects, the cytokine produced is IF-2, TNFoc and IF-13. In some aspects, the cytokine produced is IF-2, TNFoc and IFNy. In some aspects, the cytokine produced is IFNy TNFoc and IF-13. In other aspects, cytokine production occurs in a tumor microenvironment. In yet other aspects, cytokine production does not significantly occur in the spleen and/or liver of the subject.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof induces or enhances T cell proliferation in the subject. In some aspects, the T cell proliferation occurs in a tumor microenvironment. In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof reduces or inhibits tumor growth in the subject. In any of the foregoing or related aspects, the agonistic isolated monoclonal antibody or antigen binding fragment thereof increases infiltration of immune cells into a tumor microenvironment in the subject. In some aspects, the immune cells express CD45 In any of the foregoing or related aspects, wherein the agonist monoclonal antibody or antigen binding portion thereof reduces the quantity of T regulatory (Treg) cells in a tumor microenvironment in the subject. In some aspects, the Treg cells express CD4, FOXP-3 and CD25. In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof reduces the quantity of macrophages is reduced in a tumor microenvironment in the subject. In some aspects, the macrophages express CD45 and CDl lb.

In any of the foregoing or related embodiments, the isolated agonistic monoclonal antibody or antigen-binding portion thereof reduces T cell exhaustion in a tumor microenvironment in the subject. In some embodiments, reduction of T cell exhaustion comprises a decrease in expression of TIGIT, PD-l, LAG-3, or combinations thereof. In some embodiments, reduction of T cell exhaustion comprises a decrease in expression of a combination of TIGIT and PD-l.

In any of the foregoing or related aspects, the isolated agonistic monoclonal antibody or antigen-binding portion thereof induces an anti-tumor memory immune response in the subject.

In any of the foregoing or related aspects, the isolated agonistic monoclonal antibody or antigen-binding portion thereof binds Fc gamma receptor.

In any of the foregoing or related aspects, depletion of CD4+ T cells, CD8+ T cells, Natural Killer cells, or combinations thereof, reduces the efficacy of the isolated agonistic monoclonal antibody or antigen-binding portion thereof.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof reduces or inhibits tumor (cancer) growth.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof treats a disorder mediated by human CD 137 in a subject. In some embodiments, the agonist monoclonal antibody or antigen binding portion thereof treats cancer in a subject. In any of the foregoing or related aspects, the cancer is selected from the group consisting of melanoma, glioma, renal, colon, lung, prostate, breast, and head and neck cancer, among others as disclosed herein.

In any of the foregoing or related aspects, the agonist monoclonal antibody or antigen binding portion thereof binds Fc gamma receptor.

In any of the foregoing or related aspects, the agonist monoclonal antibody does not significantly induce or enhance intrahepatic and/or intrasplenic T cell activation and/or T cell proliferation. In any of the foregoing or related aspects, the agonist monoclonal antibody binds to human CD137 with an equilibrium dissociation constant K D of 1 X 10 6 or less.

In any of the foregoing or related aspects, the agonist monoclonal antibody is selected from the group consisting of: human antibody, humanized antibody, chimeric antibody, and deimmunized antibody. In any of the foregoing or related aspects, the agonist monoclonal antibody is selected from the group consisting of: a single chain antibody, a single chain Fv fragment (scFv), an Fd fragment, an Fab fragment, an Fab’ fragment, an F(ab’) 2 fragment, and a bispecific antibody.

In some aspects, the present disclosure also provides a method of detecting MHC I in a subject having cancer, comprising: (i) contacting a biological sample comprising cancer cells from the subject with an agent directed to MHC I; and (ii) detecting the agent bound to MHC I; and (iii) administering to the subject an agonist anti-CD 137 antibody, or antigen -binding fragment thereof, as disclosed herein. In some aspect, the agent is an antibody or a nucleic acid probe. In some embodiments, the antibody or probe comprises a detectable label. Such detectable labels are commonly known in the art.

In some aspects, the present disclosure also provides a method of determining whether a subject having cancer would be amenable to agonist CD 137 antibody, or antigen-binding fragment thereof, therapy. The method comprises contacting a biological sample comprising cancer cells from the subject with an agent directed to MHC I, and detecting the agent bound to MHC I according to known methods, and as described herein, wherein the presence of MHC I indicates that the cancer is amenable to treatment with an agonist CD 137 antibody, or an antigen binding fragment thereof.

In some aspects, detecting MHC I expression is performed by detecting the presence of any one or more markers selected from the group consisting of: MHC I protein, MHC I mRNA, beta-2-microglobulin chain (b2M) protein, b2M chain mRNA, MHC class I alpha chain domain protein (e.g., ocl, oc2, or oc3), and MHC class I alpha chain domain mRNA. As commonly known in the art, the alpha chains are encoded by any one of human leukocyte antigen (HLA) -A, -B, and -C genes, which are three of the major types of human MHC class I cell surface receptors. Thus, those skilled in the art would recognize that any one or more of the class I alpha chains and/or their specific domains (e.g., ocl, oc2, or oc3), can be suitable for detecting MHC I expression. In some aspects, MHC I expression is detected with an agent directed to any one or more of the markers. For example, the agent is an antibody or a nucleic acid probe. In some embodiments, the antibody or probe comprises a detectable label. Those skilled in the art can readily determine the appropriate agent, e.g., antibody or probe, depending on the particular MHC I (e.g., HLA-A, HLA-B, or HLA-C) being detected. While the HLA-A, -B, and -C genes encoding the alpha chains of the MHC I complex are known to be polymorphic, those skilled in the art can readily determine their sequences, and select and/or design a suitable detecting agent directed to the alpha chain and/or the b2M chain. An exemplary amino acid sequence of MHC I (HLA-A) is shown in SEQ ID NO: 129 (its nucleotide sequence is shown in SEQ ID NO: 130); the amino acid sequence of b2M is shown in SEQ ID NO: 131 (its nucleotide sequence is shown in SEQ ID NO: 132).

In some aspects, the detecting is performed by any method known in the art and include, e.g., a reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR, real-time RT-PCR, RNase protection assay (RPA), northern blotting, nucleic acid microarray using DNA, western blotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), radioimmunodiffusion, Ouchterlony immunodiffusion, rocket Immunoelectrophoresis, tissue immuno staining, immunoprecipitation assay, complement fixation assay, fluorescence-activated cell sorting (FACS), mass spectrometry, magnetic bead-antibody immunoprecipitation, protein chip, or any combination thereof.

In some aspects, the method further comprises determining that the cancer is amenable to treatment with an agonist CD 137 antibody, or antigen -binding fragment thereof (by the presence of MHC I), and administering to the subject a therapeutically effective amount of an agonist anti- CD 137 antibody, or antigen-binding fragment thereof.

In some aspects, the present disclosure provides a method of identifying a cancer patient likely to respond to treatment with an antibody that specifically binds to CD137, the method comprising contacting a tumor sample from the patient with an antibody that specifically binds to MHC I, or an antigen -binding fragment thereof; and detecting the presence of the MHC I, wherein the presence of MHC I indicates the patient is likely to respond to treatment. In some aspects, the method comprises detecting the presence of MHC I by determining an amount of MHC I in the sample, wherein the amount of MHC I relative to a control indicates the subject is likely to respond to treatment with the agent. In some aspects, if the amount of MHC I in the sample is increased relative to the control indicates the patient is likely to respond to treatment with the agent.

In some aspects, the present disclosure provides a method of identifying a cancer patient likely to respond to treatment with an antibody that specifically binds to CD137, the method comprising contacting a tumor sample from the patient with an antibody that specifically binds to beta-2-microglobulin chain (b2M), or an antigen-binding fragment thereof; and detecting the presence of the b2M, wherein the presence of b2M indicates the patient is likely to respond to treatment. In some aspects, the method comprises detecting the presence of b2M by determining an amount of b2M in the sample, wherein the amount of b2M I relative to a control indicates the subject is likely to respond to treatment with the agent. In some aspects, if the amount of b2M in the sample is increased relative to the control indicates the patient is likely to respond to treatment with the agent.

In some aspects, the present disclosure provides method of identifying a cancer patient likely to respond to treatment with an antibody that specifically binds to CD137, the method comprising contacting a tumor sample from the patient with an antibody that specifically binds to MHC class I alpha chain protein, or an antigen-binding fragment thereof; and detecting the presence of the MHC class I alpha chain, wherein the presence of MHC class I alpha chain indicates the patient is likely to respond to treatment. In some aspects, the method comprises detecting the presence of MHC class I alpha chain protein by determining an amount of MHC class I alpha chain in the sample, wherein the amount of MHC class I alpha chain relative to a control indicates the subject is likely to respond to treatment with the agent. In some aspects, if the amount of MHC class I alpha chain protein in the sample is increased relative to the control indicates the patient is likely to respond to treatment with the agent.

In some aspects, the disclosure provides a method of detecting a tumor susceptible to treatment with an antibody that specifically binds to CD137, the method comprising determining an expression level of a panel of polypeptides comprising MHC I protein, beta-2-micro globulin chain (b2M) protein, and MHC class I alpha chain protein, in a tumor sample, wherein the expression level of the panel of polypeptides in the tumor sample indicates the tumor is susceptible to treatment with the agent. In some aspects, determining an expression level of the panel of polypeptides comprises comparing the expression levels of the panel of polypeptides in the tumor sample to an expression level of the panel of polypeptides in a reference sample. In some aspects, if the expression level of MHC I in the tumor sample is increased relative to the reference sample indicates the tumor is susceptible to treatment with the agent.

In some aspects, the disclosure provides a kit comprising a container comprising one or more reagents for detecting the presence of MHC I, optionally beta-2-microglobulin chain (b2M) and/or MHC class I alpha chain in a tumor sample, and a package insert comprising instructions for determining an amount or expression level of MHC I, optionally b2M and/or MHC class I alpha chain in the tumor sample.

In some aspects, the kit comprises a reagent for detecting the presence of b2M in the sample and instructions for determining an amount or expression level of b2M in the tumor sample

In some aspects, the kit comprises a reagent for detecting the presence of MHC class I alpha chain in the sample and instructions for determining an amount or expression level of MHC class I alpha chain in the tumor sample.

In some aspects of the disclosure, determining an amount or expression level of any one or more of MHC I, b2M, and MHC class I alpha chain, or a combination thereof comprises comparing the amount or expression level of MHC I, b2M, or MHC class I alpha chain, or a combination thereof in a tumor sample relative to the amount or expression level in a reference sample.

In some embodiments, the kit comprises an antibody specifically reactive with MHC I. In some embodiments, the kit comprises an antibody specifically reactive with b2M. In some embodiments, the kit comprises an antibody specifically reactive with MHC class I alpha chain. In some embodiments, the kit comprises an antibody specifically reactive with one or more of MHC I, b2M, and MHC class I alpha chain.

In some embodiments, the one or more antibodies is covalently attached to a label.

In some embodiments, the label is a fluorescent molecule, a chromogenic molecule, or a radioactive molecule.

BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee. FIG. 1 provides graphs depicting the distribution of binding affinities of affinity matured clones of the parental anti-CD 137 antibody mAbl.

FIG. 2 provides a schematic showing the results of mAbl CDRH3 alanine scanning, as measured by binding affinity (K D ) to human or mouse CD 137.

FIG. 3A shows the amino acid sequence of human CD137 (residues 24-159 of SEQ ID NO: 3) wherein residues comprising an epitope bound by mAbl, mAb4 or mAb5 are indicated in bold.

FIG. 3B is a graph depicting kinetic binding data of mAbl to the extracellular domain of mouse and rat CD 137 as determined by surface plasmon resonance.

FIG. 3C provides x-ray crystallography images of human CD 137 bound to CDl37L( shown in grey) and residues El 11, Tl 13, Kl 14 and P135 shown as spheres.

FIG. 3D provides x-ray crystallography images of human CD 137 bound to CD137L (shown in grey) in trimeric formation, and residues E111, T113, K114 and P 135 shown as spheres.

FIG. 4A provides a scatterplot of flow cytometric data depicting an increase in TIGIT (top) or PD-l (bottom) expression on CD44+ T cells in response to anti-CDl37 antibodies.

FIG. 4B provides graphs depicting the quantification of CD8+ CD44+ T cells expressing TIGIT (top) or PD-l (bottom) in the spleen of mice after treatment with anti-CDl37 antibodies.

FIG. 4C provides graphs depicting the quantification of CD8+ T cells in the spleen of mice after treatment with anti-CD 137 antibodies, as percentage of CD45+ cells (left) or cell number per spleen (right).

FIG. 5A provides graphs showing individual CT26 tumor volumes in mice after treatment with anti-CDl37 antibodies at indicated dosages.

FIG. 5B is a graph showing the mean tumor volumes provided in FIG. 5A.

FIG. 5C is a Kaplan-Meier graph showing overall survival of mice with tumors after treatment with anti-CD 137 antibodies.

FIG. 5D is a graph showing tumor volume in mice re-challenged with tumorigenic CT26 cells.

FIG. 6A provides graphs showing individual CT26 tumor volumes in mice after treatment with parental and affinity-matured anti-CD 137 antibodies.

FIG. 6B is a graph providing the mean tumor volumes provided in FIG. 6A. FIG. 7 provides graphs depicting the percentage of CD8+ or CD4+ T cells, from splenic T cells (top) and tumor infiltrating leukocytes (bottom) after treatment with anti-CD 137 antibodies at indicated dosages.

FIG. 8 provides graphs showing individual tumor volumes when mice were treated with mAbl, with or without lymphocyte depleting antibodies. CD4+ T cells were depleted with GK1.5 (middle graph), CD8+ T cells were depleted with YTS 169.4 (second graph from the right), and NK cells were depleted with an anti-asialo-GMl antibody (last graph on the right).

FIG. 9 provides graphs showing individual tumor volumes in mice having either CT26 tumors (colon carcinoma), EMT-6 tumors (breast carcinoma), A20 tumors (B cell lymphoma), or MC38 tumors (colon carcinoma) and treated with mAb8 or isotype control antibody.

FIGs. 10A-10C show the in vivo anti-tumor efficacy of anti-CD 137 antibodies administered at 150 mg/mouse. Individual tumor volumes are shown in 10A, mean tumor volumes are shown in 10B and percent survival is shown in 10C.

FIGs. 11A-11C show the in vivo anti-tumor efficacy of anti-CDl37 antibodies administered at 20 mg/mouse. Individual tumor volumes are shown in 11A, mean tumor volumes are shown in 11B and percent survival is shown in 11C.

FIG. 12 provides graphs showing individual tumor volumes in mice having CT26 tumors and treated with varying doses of mAbl ( i.e 12.5, 25, 50, 100 or 200mg) or isotype control.

FIGs. 13A and 13B show the contribution of Fc binding in the anti-tumor efficacy of mAbl. FIG. 13A shows mAbl as an IgG4 isotype or an IgG4 aglycosylated isotype. Mean tumor volumes are shown on the top and individual tumor volumes are shown on the bottom. FIG. 13B shows mAbl as an IgG4 isotype or an IgGl aglycosylated isotype. Mean tumor volumes are shown on the top and individual tumor volumes are shown on the bottom.

FIGs. 14A-14D show the in vivo anti-tumor efficacy of anti-CD 137 antibodies in mice with large established tumors (i.e., 500mm 3 ) prior to receiving treatment. Individual tumor volumes are shown in 14A and 14D, mean tumor volumes are shown in 14B and percent survival is shown in 14C.

FIG. 15 provides a Kaplan-Meier survival graph showing protective anti-tumor immunity in mice previously treated with mAbl, mAb8 or isotype control from FIGs. 14A-14C and considered cured, re-challenged with CT26 cells in an opposing flank. FIG. 16A provides scatterplots of flow cytometric data showing the expansion of CD45+ intrahepatic T cells following treatment with anti-CDl37 antibodies at indicated dosages.

FIG. 16B provides graphs depicting the quantification of intrahepatic CD8+ T cells (left) and CD4+ T cells (right) following treatment with anti-CDl37 antibodies at indicated dosages.

FIG. 17A provides graphs depicting the percentage of CD3+, CD4+, or CD8+ T cells, from splenic T cells after treatment of mice with affinity-matured anti-CDl37 antibodies.

FIG. 17B provides graphs depicting the percentage of CD3+, CD4+, or CD8+ T cells from liver T cells after treatment of mice with affinity-matured anti-CDl37 antibodies.

FIG. 18A provides graphs depicting the percentage of splenic CD8+CD44+ T cells expressing TIGIT, PD-l, or LAG3 after treatment of mice with affinity-matured anti-CD 137 antibodies.

FIG. 18B provides graphs depicting the percentage of liver CD8+CD44+ T cells expressing TIGIT, PD-l, or LAG3 after treatment of mice with affinity-matured anti-CD 137 antibodies.

FIG. 19A provides graphs depicting the percentage of splenic CD4+CD44+ T cells expressing TIGIT, PD-l, or LAG3 after treatment of mice with affinity-matured anti-CD 137 antibodies.

FIG. 19B provides graphs depicting the percentage of liver CD4+CD44+ T cells expressing TIGIT, PD-l, or LAG3 after treatment of mice with affinity-matured anti-CD 137 antibodies.

FIGs. 20A-20C provide graphs of in vivo indicators of toxicity resulting from multiple administrations of anti-CD 137 antibodies mAbl, mAb8 or 3H3 at varying doses. FIG. 20A is a graph showing percentage of CD8+ T cells in the liver after administration of the anti-CD 137 antibodies. FIG. 20B is a graph showing alanine aminotransferase (ALT) activity in the plasma of mice administered anti-CDl37 antibodies. FIG. 20C is a graph showing the levels of TNFoc in the plasma of mice administered anti-CDl37 antibodies.

FIG. 21 provides representative images of sectioned livers stained with hematoxylin and eosin (H&E) from mice treated with mAbl, mAb8, 3H3 or isotype control as described in FIGs. 20A-20C. Arrows indicate infiltration of immune cells.

FIGs. 22A-22D provide representative FACS plots showing immune cell reprogramming in the tumor microenvironment. Mice having CT26 tumors were administered multiple doses of mAb8 or isotype control (days 0, 3, 6 and 9). FIG. 22A shows overall immune cell infiltration based on CD45 expression. FIG. 22B shows reduction in Treg cells as measured by FOXP-3 and CD25 expression. FIG. 22C shows reduction of T-cell exhaustion as measured by PD-l and TIGIT expression. FIG. 22D shows reduction of tumor-associated macrophages as measured by F4/80 and CDl lb expression.

FIG. 23 shows immunophenotyping analysis of spleens from mice having CT26 tumors and treated with either anti-CD 137 antibodies mAbl and 3H3, or isotype control.

FIG. 24 is a graph showing the concentration of IL-2 (pg/ml) produced by murine T cells in an OVA stimulation assay, when stimulated with the anti-CDl37 antibodies indicated. Along with Atezolizumab (anti-PD-Ll antibody), a murine anti-PD-l (RMP1-14) was used as a comparator.

FIGs. 25A and 25B are graphs showing the percentage of murine CD8+ T cells expressing either CD25 (25A) or TIGIT (25B) when stimulated with the anti-CD 137 antibodies indicated, in an OVA stimulation assay. Along with Atezolizumab (anti-PD-Ll antibody), a murine anti-PD-l (RMP1-14) and murine anti-CDl37 (3H3) were used as comparators.

FIG. 26 provides bar graphs depicting the quantification of cytokines (IL-2, TNFa, IL-13, and IFNy) produced by CD3+ T cells following incubation with plate-bound anti-CD 137 antibodies. Cytokine levels are shown as fold increase over baseline activation by an anti-CD3 antibody.

FIGs. 27A-27C provide graphs depicting the dose-response of IFNy production in a mixed lymphocyte reaction following treatment with anti-CDl37 antibodies. An anti-PDl antibody (Keytruda; Merck) was used as a control.

FIG. 28 is a graph showing IFNy production from human T cells co-cultured with CHO cells engineered to express CD32 (CHO-CD32 cells) in the presence of anti-CD 137 antibodies mAbl, mAb8, mAb4 or mAb5, or isotype control.

FIG. 29 is a graph showing proliferation of Treg cells when co-cultured with CHO cells engineered to express CD32 (CHO-CD32 cells) in the presence or absence of anti-CD 137 antibodies mAbl, mAb8, mAb4 or mAb5, isotype control.

FIG. 30 provides graphs showing NFK and SRF signaling in CCL-119 cells transduced with luciferase reporters for NFK or SRF in the presence of mAbl, mAB8, mAb4 or mAb5 at varying concentrations. FIG. 31 provides graphs showing induction of IL-6, TNFoc, or IL-27 by bone marrow- derived mouse macrophages stimulated with TLR9 agonist CpG in the presence of anti-CD 137 antibodies mAbl, 3H3 or LOB 12.3, or isotype control.

FIG. 32 provides a graph showing induction of TNFoc by human monocyte derived macrophages stimulated with LPS in the presence of anti-CD 137 antibodies mAbl, mAb4 or mAb5, or isotype control.

FIG. 33 provides a graph showing effect of anti-CD 137 antibodies on macrophage differentiation as determined by CD64 expression of THP1 monocytes cultured with PM A in the presence of anti-CDl37 antibodies mAbl, mAb4 or mAb5, or isotype control.

FIGs. 34A-34C provides graphs showing percentage of hCD45+, hCD8+ or hCD4+ from immunocompetent mice that received human PBMCs and anti-CDl37 antibodies mAbl, mAb4 or mAb5, or isotype control.

FIG. 35A shows CT26 cells, a colon cancer cell line, stained with a fluorescent antibody against beta-2 microglobulin (B2M) or isotype control, and analyzed using flow cytometry.

FIG. 35B shows CT26 cells stained with a fluorescent antibody against major histocompatibility complex I (MHC I) or isotype control, analyzed using flow cytometry.

FIG. 36A shows B 16-F10 cells, a skin melanoma cell line, stained with a fluorescent antibody against beta-2 microglobulin (B2M) or isotype control, and analyzed using flow cytometry.

FIG. 36B shows B 16-F10 cells stained with a fluorescent antibody against major histocompatibility complex I (MHC I) or isotype control, analyzed using flow cytometry.

FIG. 37 shows CT26 cells, EMT6 cells, A20 cells, MC38 cells, RENCA cells, MB49 cells, 4T1 cells, ID-8 cells, B 16-F10 cells, PANC02 cells, HEPA1-6 cells, and Rl.l cells stained with a fluorescent antibody against major histocompatibility complex I (MHC I) or isotype control and analyzed using flow cytometry.

FIG. 38A shows CT26 cells stained with a fluorescent antibody against major histocompatibility complex I (MHC I) as compared to isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 25 pg weekly of mAb8 or vehicle. FIG. 38B shows EMT6 cells stained with a fluorescent antibody against MHC I or isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 5pg weekly of mAb8 or vehicle.

FIG. 38C shows A20 cells stained with a fluorescent antibody against MHC I as compared to isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 200 pg weekly of mAb8 or control antibodies.

FIG. 38D shows MC38 cells stained with a fluorescent antibody against MHC I or isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 12.5 pg weekly of mAb8 or control antibodies. Histogram curves for MHC I and isotype control is indicated in blue and red, respectively.

FIG. 39A shows B 16-F10 cells stained with a fluorescent antibody against major histocompatibility complex I (MHC I) or isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 150 pg of mAb8 or isotype control antibodies on days 0, 3, 6, and 9.

FIG. 39B shows PANC02 cells stained with a fluorescent antibody against MHC I or isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 150 pg of mAb8 or isotype control antibodies.

FIG. 39C shows 4T1 cells stained with a fluorescent antibody against MHC I or isotype control which were analyzed using flow cytometry; and graphs of tumor volume in mice following treatment with either 50pg of mAbl or isotype control antibodies on days 0, 3, 6, and 9. Units for middle panel of FIG. 39C is the same as the lower panel of FIG. 39C.

FIG. 40 depicts expression levels of MHC I, B2M, and PDL1, in CT26 parental cells (parental CTN) and CT26- 2M KO cells (CT26 B2M KO 873 CNT and CT26 B2M KO 882 CNT).

FIG. 41A graphically depicts tumor volume in mice injected with CT26 parental cells (CT26 Par CNT) or CT26- 2M KO cells (CT26 B2M KO 882 or CT26 B2M KO 883 ) following treatment with mAb8 or control (x-axis: days post-commencement of treatment, y-axis: tumor volume in mm 3 ).

FIG. 41B graphically depicts tumor volume in mice injected with CT26 parental cells (CT26 Par CNT) or OT26-b2M KO cells (CT26 B2M KO 882 or CT26 B2M KO 883 ) following treatment with mAb8 or control. FIG. 41C graphically depicts percent survival in mice injected with CT26 parental cells (CT26 Par CNT) or CT26-p2M KO cells (CT26 B2M KO 882 or CT26 B2M KO 883 ) following treatment with mAh 8 or control.

DETAILED DESCRIPTION

Cancer therapy with agonist anti-CD 137 antibodies has been shown to induce immune- mediated tumor rejections in mice, and analogous agents of this kind are currently being tested in cancer patients. Previous reports have indicated that administration of anti-CDl37 antibodies can induce significant accumulations of polyclonal infiltrates of T lymphocytes in the liver (Dubrot et al., (2010) Cancer Immunology, Immunotherapy 59(8): 1223-1233), suggestive of hepatic inflammation and the potential for drug-induced liver toxicity. A recent report on the clinical evaluation of an agonistic anti-CDl37 antibody (Urelumab, BMS-663513; Bristol-Myers Squibb) documented the observation of treatment-related adverse events in human subjects, including indications of severe hepatotoxicity (transaminitis) correlating with antibody dose (Segal et al., (2016) Clin Cancer Res 23(8): 1929-1936).

The present disclosure relates to methods and composition for treating cancer in a subject. The disclosure is based, at least in part, on the surprising discovery that treatment of cancer cells expressing major histocompatibility complex I (MHC I) is enhanced when treatment includes the anti-CD 137 agonist antibodies, and antigen-binding fragments thereof, disclosed herein.

Thus, the present discovery can be utilized to identify a cancer patient population that would be particularly amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein. For example, a patient population, wherein the patient or subject has cancer cells that express MHC I.

In some embodiments, the disclosure provides methods for the treatment of cancer in a subject with the isolated monoclonal antibodies of the disclosure, or antigen binding portions thereof, that specifically bind to an epitope of human CD 137 and agonize human CD 137. In some embodiments, the subject has cancer cells that express MHC I. In some embodiments, the antibody or antigen binding portion thereof competes with mAbl for binding to the epitope of human CD137. In some aspects, the anti-CDl37 agonist antibodies of the disclosure induce cytokine production and expansion of CD8+ T cells in the tumor microenvironment, and protective anti-tumor immunity in vivo with a concomitant reduction in the potential for toxicity- related events, as compared to the anti-mouse CD137 3H3 antibody (Melero et al. (1997) Nature Medicine 3(6):682-685; Uno et al. (2006) Nature Medicine l2(6):693-696) and to at least two anti-human CD137 antibodies in clinical development (BMS-6635l3/Urelumab, Bristol-Meyers Squibb, and PF-05082566/Utomilumab, Pfizer).

Definitions

Terms used in the claims and specification are defined as set forth below unless otherwise specified.

It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.

As used herein, "about" will be understood by persons of ordinary skill and will vary to some extent depending on the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill given the context in which it is used, "about" will mean up to plus or minus 10% of the particular value.

As used herein, the term "agonist" refers to any molecule that partially or fully promotes, induces, increases, and/or activates a biological activity of a native polypeptide disclosed herein (e.g., CD137). Suitable agonist molecules specifically include agonist antibodies or antibody fragments, fragments or amino acid sequence variants of native polypeptides, peptides, antisense oligonucleotides, small organic molecules, etc. In some embodiments, activation in the presence of the agonist is observed in a dose-dependent manner. In some embodiments, the measured signal (e.g., biological activity) is at least about 5%, at least about 10%, at least about 15%, at least about

20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about

45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about

70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about

95%, or at least about 100% higher than the signal measured with a negative control under comparable conditions. Also disclosed herein, are methods of identifying agonists suitable for use in the methods of the disclosure. For example, these methods include, but are not limited to, binding assays such as enzyme-linked immuno-absorbent assay (ELISA), Forte Bio© systems, and radioimmunoassay (RIA). These assays determine the ability of an agonist to bind the polypeptide of interest (e.g., a receptor or ligand, e.g., CD137) and therefore indicate the ability of the agonist to promote, increase or activate the activity of the polypeptide. Efficacy of an agonist can also be determined using functional assays, such as the ability of an agonist to activate or promote the function of the polypeptide. For example, a functional assay may comprise contacting a polypeptide with a candidate agonist molecule and measuring a detectable change in one or more biological activities normally associated with the polypeptide. The potency of an agonist is usually defined by its EC50 value (concentration required to activate 50% of the agonist response). The lower the EC50 value the greater the potency of the agonist and the lower the concentration that is required to activate the maximum biological response.

As used herein, the term’’alanine scanning” refers to a technique used to determine the contribution of a specific wild-type residue to the stability or function(s) (e.g., binding affinity) of a given protein or polypeptide. The technique involves the substitution of an alanine residue for a wild-type residue in a polypeptide, followed by an assessment of the stability or function(s) (e.g., binding affinity) of the alanine-substituted derivative or mutant polypeptide and comparison to the wild-type polypeptide. Techniques to substitute alanine for a wild-type residue in a polypeptide are known in the art.

The term "ameliorating" refers to any therapeutically beneficial result in the treatment of a disease state, e.g., cancer, including prophylaxis, lessening in the severity or progression, remission, or cure thereof.

As used herein, the term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxyglutamate, and O-phospho serine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid.

Amino acids can be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, can be referred to by their commonly accepted single-letter codes. As used here, a“polar amino acid” refers to an amino acid comprising a side chain that prefers to reside in an aqueous environment. In some embodiments, a polar amino acid is selected from the group consisting of: arginine, asparagine, aspartic acid, glutamic acid, glutamine, histidine, lysine, serine, theronine and tyrosine. Polar amino acids can be positive, negatively or neutrally charged. As used herein, a“non-polar amino acid” refers to an amino acid selected from the group consisting of: alanine, cysteine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan and valine.

As used herein, an "amino acid substitution" refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence (an amino acid sequence of a starting polypeptide) with a second, different "replacement" amino acid residue. An "amino acid insertion" refers to the incorporation of at least one additional amino acid into a predetermined amino acid sequence. While the insertion will usually consist of the insertion of one or two amino acid residues, larger "peptide insertions," can also be made, e.g. insertion of about three to about five or even up to about ten, fifteen, or twenty amino acid residues. The inserted residue(s) may be naturally occurring or non- naturally occurring as disclosed above. An "amino acid deletion" refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.

As used herein, the term "antagonist" refers to any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity of a native polypeptide disclosed herein. Suitable antagonist molecules specifically include antagonist antibodies or antibody fragments, fragments or amino acid sequence variants of native polypeptides, peptides, antisense oligonucleotides, small organic molecules, etc. In some embodiments, inhibition in the presence of the antagonist is observed in a dose-dependent manner. In some embodiments, the measured signal (e.g., biological activity) is at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100% lower than the signal measured with a negative control under comparable conditions. Also disclosed herein, are methods of identifying antagonists suitable for use in the methods of the disclosure. For example, these methods include, but are not limited to, binding assays such as enzyme-linked immuno-absorbent assay (ELISA), Forte Bio© systems, and radioimmunoassay (RIA). These assays determine the ability of an antagonist to bind the polypeptide of interest (e.g., a receptor or ligand) and therefore indicate the ability of the antagonist to inhibit, neutralize or block the activity of the polypeptide. Efficacy of an antagonist can also be determined using functional assays, such as the ability of an antagonist to inhibit the function of the polypeptide or an agonist. For example, a functional assay may comprise contacting a polypeptide with a candidate antagonist molecule and measuring a detectable change in one or more biological activities normally associated with the polypeptide. The potency of an antagonist is usually defined by its IC50 value (concentration required to inhibit 50% of the agonist response). The lower the IC50 value the greater the potency of the antagonist and the lower the concentration that is required to inhibit the maximum biological response.

As used herein, the term“amount” or“level” refers to a detectable quantity, level or abundance of a substance (e.g., a protein or a biomarker). When referring to a polypeptide or a biomarker, such as those described herein, the terms“level of expression” or“expression level” in general are used interchangeably and generally refer to a detectable amount of a polypeptide or biomarker in a biological sample (e.g., on the surface of a cell). In some aspects, a detectable amount or detectable level of a biomarker is associated with a likelihood of a response to an agent, such as those described herein.

As used herein, the term "anti-CDl37 agonist antibody" (used interchangeably with the term "anti-CDl37 antibody") refers to an antibody that specifically binds to CD137 and partially or fully promotes, induces, increases, and/or activates CD137 biological activity, response, and/or downstream pathway(s) mediated by CD137 signaling or other CDl37-mediated function. In some embodiments, an anti-CD 137 agonist antibody binds to CD 137 and allows binding of CD137L. In some embodiments, an anti-CD 137 agonist antibody binds to CD 137 and induces multimerization of CD137. In some embodiments, an anti-CDl37 agonist antibody binds to CD137 and induces the dimerization of CD137 trimers. In some embodiments, an anti-CDl37 agonist antibody binds to CD 137 and induces the multimerization of CD 137 trimers. Examples of anti-CD 137 agonist antibodies are provided herein. Methods for detecting formation of a trimentrimer complex are known to those of skill in the art. For example, electron microscopy has been shown to detect such complexes, see, e.g., Won, E. The Journal of Biological Chemistry, Vol. 285 (12): 9202-9210 (2010)

As used herein, the term“anti-CDl37 mAbl” (used interchangeably with“mAbl”) refers to an exemplary anti-CD 137 agonist antibody that comprises the variable heavy chain (V H ) amino acid sequence:

EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGSGGSTY

Y ADS VKGRFTIS RDN S KNTLYLQMN S LR AEDT A V Y Y C AKDS PFLLDD Y Y Y Y Y YMD VW GKGTTVTVSS (SEQ ID NO: 4),

and the variable light chain (V L ) amino acid sequence:

DIQMTQS PSSVSASV GDR VTITCRAS QGIS S WLA W Y QQKPGKAPKLLIY A AS S LQS G VPS RFS GS GS GTDFTLTIS S LQPEDF AT Y YCQQGHLFPITF GGGTK VEIK (SEQ ID NO: 6).

As used herein, the term“anti-CD 137 mAb8” (used interchangeably with“mAb8”) refers to an exemplary anti-CD 137 agonist antibody that comprises the variable heavy chain ((V H ) amino acid sequence:

E V QLLES GGGLV QPGGS LRLS C A AS GFTFRN Y AMS W VRQ APGKGLE W V S AIS GS GDTT

Y Y ADS VKGRFTIS RDN S KNTLYLQMN S LR AEDT A V Y Y C AKDS PFLLDD YYYYY YMD V WGKGTTVTVSS (SEQ ID NO: 101);

and the variable light chain (V L ) amino acid sequence:

DIQMTQS PSSVSASV GDR VTITCRAS QGIS S WLA WY QQKPGKAPKLLIY A AS S LQS G VPS RFS GS GS GTDFTLTIS S LQPEDF AT Y YCQQGHLFPITF GGGTK VEIK (SEQ ID NO: 6).

As used herein, the term“anti-CDl37 mAblO” (used interchangeably with“mAblO”) refers to an exemplary anti-CDl37 agonist antibody that comprises the variable heavy chain ((V H ) amino acid sequence:

E V QLLES GGGLV QPGGS LRLS C A AS GFTFY GY AMS W VRQ APGKGLE WV A AIS GS GDS T

Y Y ADS VKGRFTIS RDN S KNTLYLQMN S LR AEDT A V Y Y C AKDS PFLLDD YYYYY YMD V WGKGTTVTVSS (SEQ ID NO: 26);

and the variable light chain (V L ) amino acid sequence:

DIQMTQS PSSVSASV GDR VTITCRAS QGIS S WLA WY QQKPGKAPKLLIY A AS S LQS G VPS RFS GS GS GTDFTLTIS S LQPEDF AT Y YCQQGHLFPITF GGGTK VEIK (SEQ ID NO: 6). As used herein, the term“antibody” refers to a whole antibody comprising two light chain polypeptides and two heavy chain polypeptides. Whole antibodies include different antibody isotypes including IgM, IgG, IgA, IgD, and IgE antibodies. The term“antibody” includes a polyclonal antibody, a monoclonal antibody, a chimerized or chimeric antibody, a humanized antibody, a primatized antibody, a deimmunized antibody, and a fully human antibody. The antibody can be made in or derived from any of a variety of species, e.g., mammals such as humans, non-human primates (e.g., orangutan, baboons, or chimpanzees), horses, cattle, pigs, sheep, goats, dogs, cats, rabbits, guinea pigs, gerbils, hamsters, rats, and mice. The antibody can be a purified or a recombinant antibody.

As used herein, the terms“antibody fragment,”“antibody portion”, “antigen -binding fragment,”“antigen binding portion” or similar terms refer to a fragment of an antibody that retains the ability to bind to a target antigen (e.g., CD137) and inhibit the activity of the target antigen. Such fragments include, e.g., a single chain antibody, a single chain Fv fragment (scFv), an Fd fragment, a Fab fragment, a Fab’ fragment, or an F(ab’) 2 fragment. An scFv fragment is a single polypeptide chain that includes both the heavy and light chain variable regions of the antibody from which the scFv is derived. In addition, intrabodies, minibodies, triabodies, and diabodies are also included in the definition of antibody and are compatible for use in the methods described herein. See, e.g., Todorovska et al., (2001) J. Immunol. Methods 248(l):47-66; Hudson and Kortt, (1999) J. Immunol. Methods 231(1): 177-189; Poljak, (1994) Structure 2(12): 1121-1123; Rondon and Marasco, (1997) Annu. Rev. Microbiol. 51:257-283, the disclosures of each of which are incorporated herein by reference in their entirety.

As used herein, the term“antibody fragment” also includes, e.g., single domain antibodies such as camelized single domain antibodies. See, e.g., Muyldermans et al., (2001) Trends Biochem. Sci. 26:230-235; Nuttall et al., (2000) Curr. Pharm. Biotech. 1:253-263; Reichmann et al., (1999) J. Immunol. Meth. 231:25-38; PCT application publication nos. WO 94/04678 and WO 94/25591; and U.S. patent no. 6,005,079, all of which are incorporated herein by reference in their entireties. In some embodiments, the disclosure provides single domain antibodies comprising two VH domains with modifications such that single domain antibodies are formed.

In some embodiment, an antigen-binding fragment includes the variable region of a heavy chain polypeptide and the variable region of a light chain polypeptide. In some embodiments, an antigen-binding fragment described herein comprises the CDRs of the light chain and heavy chain polypeptide of an antibody.

As used herein, the term“antibody- antigen complex” or“immune complex” or“complex” refers to a molecular structure formed upon the specific binding of an antibody to an antigen (e.g., a biomarker). The bound antibody and antigen act as a unitary object. In some embodiments, a first antibody is bound to an antigen, thereby forming a complex. In some embodiments, the complex is detected by the binding of a second antibody to the first antibody, thereby indirectly detecting the antigen.

The term“antigen presenting cell” or“APC” is a cell that displays foreign antigen complexed with MHC on its surface. T cells recognize this complex using T cell receptor (TCR). Examples of APCs include, but are not limited to, dendritic cells (DCs), peripheral blood mononuclear cells (PBMC), monocytes (such as THP-l), B lymphoblastoid cells (such as C1R.A2, 1518 B-LCL) and monocyte-derived dendritic cells (DCs). Some APCs internalize antigens either by phagocytosis or by receptor-mediated endocytosis.

The term“antigen presentation” refers to the process by which APCs capture antigens and enables their recognition by T cells, e.g., as a component of an MHC-I and/or MHC-II conjugate.

As used herein, the term "apoptosis" refers to the process of programmed cell death that occurs in multicellular organisms (e.g. humans). The highly-regulated biochemical and molecular events that result in apoptosis can lead to observable and characteristic morphological changes to a cell, including membrane blebbing, cell volume shrinkage, chromosomal DNA condensation and fragmentation, and mRNA decay. A common method to identify cells, including T cells, undergoing apoptosis is to expose cells to a fluorophore-conjugated protein (Annexin V). Annexin V is commonly used to detect apoptotic cells by its ability to bind to phosphatidylserine on the outer leaflet of the plasma membrane, which is an early indicator that the cell is undergoing the process of apoptosis.

As used herein,“MHC molecules” refers to two types of molecules, MHC class I and MHC class II. MHC class I molecules present antigen to specific CD8+ T cells and MHC class II molecules present antigen to specific CD4+ T cells. Antigens delivered exogenously to APCs are processed primarily for association with MHC class II. In contrast, antigens delivered endogenously to APCs are processed primarily for association with MHC class I. The term“MHC I” as used herein includes the MHC I complex as well as one or more of the subunits that make up MHC I (e.g., the class I alpha chain having 3 alpha subdomains (ocl, oc2, oc3) and beta2-microglobulin). As described herein, the alpha chains of the MHC I molecule are encoded by highly polymorphic HLA-A, HLA-B, and HLA-C genes. Thus, as those skilled in the art would recognize,“MHC I” includes the MHC class I molecules encoded by the HLA-A, HLA- B, and HLA-C genes, their fragments, as well as their polymorphs. In some embodiments, “detecting MHC I” can be performed by detecting the presence of beta2-microglobulin. Thus, a method that“detects MHC I” includes the detection of any one or more of the subunits of MHC I, or MHC I in its entirety (e.g., by use of an antibody that recognizes the entire complex).

As used herein, “marker” refers to MHC I itself, or any of the subunit(s) and/or component(s) that indicate the presence of MHC I (e.g., any one of more of the 3 alpha subdomains of the class I alpha chains, and polymorphs thereof, and beta2-micro globulin).

As used herein, the term“binds to immobilized CD137,” refers to the ability of a human antibody of the disclosure to bind to CD137, for example, expressed on the surface of a cell or which is attached to a solid support.

As used herein, the term“biomarker” refers to a characteristic (e.g., biological marker) that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention, including therapeutic interventions. In some embodiments, a biomarker is a molecular biomarker. In some embodiment, a molecular biomarker is a protein biomarker. In some embodiments, a molecular biomarker is a nucleic acid biomarker. In some embodiments, a biomarker is a histologic biomarker. In some embodiments, a biomarker is a radiographic biomarker. In some embodiments, a biomarker is a physiologic characteristic.

As used herein, the term“bispecific” or“bifunctional antibody” refers to an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, (1990) Clin. Exp. Immunol. 79:315- 321; Kostelny et al., (1992) J. Immunol. 148: 1547-1553.

Traditionally, the recombinant production of bispecific antibodies is based on the co expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chain/light-chain pairs have different specificities (Milstein and Cuello, (1983) Nature 305:537- 539). Antibody variable domains with the desired binding specificities (antibody- antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion of the heavy chain variable region is preferably with an immunoglobulin heavy-chain constant domain, including at least part of the hinge, CH2, and CH3 regions. For further details of illustrative currently known methods for generating bispecific antibodies see, e.g., Suresh et al., (1986) Methods Enzymol. 121:210; PCT Publication No. WO 96/27011; Brennan et al., (1985) Science 229:81; Shalaby et al., J. Exp. Med. (1992) 175:217-225; Kostelny et al., (1992) J. Immunol. 148(5): 1547-1553; Hollinger et al., (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Gruber et al., (1994) J. Immunol. 152:5368; and Tutt et al., (1991) J. Immunol. 147:60. Bispecific antibodies also include cross-linked or heteroconjugate antibodies. Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.

Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. See, e.g., Kostelny et al. (1992) J Immunol 148(5): 1547-1553. The leucine zipper peptides from the Fos and Jun proteins may be linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers may be reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The“diabody” technology described by Hollinger et al. (1993) Proc Natl Acad Sci USA 90:6444-6448 has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (scFv) dimers has also been reported. See, e.g., Gruber et al. (1994) J Immunol 152:5368. Alternatively, the antibodies can be“linear antibodies” as described in, e.g., Zapata et al. (1995) Protein Eng. 8(10): 1057-1062. Briefly, these antibodies comprise a pair of tandem Fd segments (VH-CH1-VH-CH1) which form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific.

Antibodies with more than two valencies (e.g., trispecific antibodies) are contemplated and described in, e.g., Tutt et al. (1991) J Immunol 147:60. The disclosure also embraces variant forms of multi- specific antibodies such as the dual variable domain immunoglobulin (DVD-Ig) molecules described in Wu et al. (2007) Nat Biotechnol 25(11): 1290-1297. The DVD-Ig molecules are designed such that two different light chain variable domains (VL) from two different parent antibodies are linked in tandem directly or via a short linker by recombinant DNA techniques, followed by the light chain constant domain. Similarly, the heavy chain comprises two different heavy chain variable domains (VH) linked in tandem, followed by the constant domain CH1 and Fc region. Methods for making DVD-Ig molecules from two parent antibodies are further described in, e.g., PCT Publication Nos. WO 08/024188 and WO 07/024715. In some embodiments, the bispecific antibody is a Fabs-in- Tandem immunoglobulin, in which the light chain variable region with a second specificity is fused to the heavy chain variable region of a whole antibody. Such antibodies are described in, e.g., International Patent Application Publication No. WO 2015/103072.

As used herein, "cancer antigen" refers to (i) tumor- specific antigens, (ii) tumor- associated antigens, (iii) cells that express tumor- specific antigens, (iv) cells that express tumor- associated antigens, (v) embryonic antigens on tumors, (vi) autologous tumor cells, (vii) tumor- specific membrane antigens, (viii) tumor- associated membrane antigens, (ix) growth factor receptors, (x) growth factor ligands, and (xi) any other type of antigen or antigen-presenting cell or material that is associated with a cancer.

The term "carcinoma" is art recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. The anti-CDl37 antibodies described herein can be used to treat patients who have, who are suspected of having, or who may be at high risk for developing any type of cancer, including renal carcinoma or melanoma. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary. The term also includes carcinosarcomas, which include malignant tumors composed of carcinomatous and sarcomatous tissues. An "adenocarcinoma" refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.

As used herein the term "compete", when used in the context of antigen-binding proteins (e.g., immunoglobulins, antibodies, or antigen-binding fragments thereof) that compete for binding to the same epitope, refers to a interaction between antigen-binding proteins as determined by an assay (e.g., a competitive binding assay; a cross-blocking assay), wherein a test antigen binding protein (e.g., a test antibody) inhibits (e.g., reduces or blocks) specific binding of a reference antigen-binding protein (e.g., a reference antibody, such as mAbl) to a common antigen (e.g., CD137 or a fragment thereof). In some embodiments, the antibodies described herein cross compete with mAbl (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 4 and 6, respectively), mab8 (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 101 and 6, respectively) or mAblO (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 26 and 6, respectively).

A polypeptide or amino acid sequence "derived from" a designated polypeptide or protein refers to the origin of the polypeptide. Preferably, the polypeptide or amino acid sequence which is derived from a particular sequence has an amino acid sequence that is essentially identical to that sequence or a portion thereof, wherein the portion consists of at least 10-20 amino acids, preferably at least 20-30 amino acids, more preferably at least 30-50 amino acids, or which is otherwise identifiable to one of ordinary skill in the art as having its origin in the sequence. Polypeptides derived from another peptide may have one or more mutations relative to the starting polypeptide, e.g., one or more amino acid residues which have been substituted with another amino acid residue or which has one or more amino acid residue insertions or deletions.

A polypeptide can comprise an amino acid sequence which is not naturally occurring. Such variants necessarily have less than 100% sequence identity or similarity with the starting molecule. In certain embodiments, the variant will have an amino acid sequence from about 75% to less than 100% amino acid sequence identity or similarity with the amino acid sequence of the starting polypeptide, more preferably from about 80% to less than 100%, more preferably from about 85% to less than 100%, more preferably from about 90% to less than 100% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%) and most preferably from about 95% to less than 100%, e.g., over the length of the variant molecule.

In certain embodiments, there is one amino acid difference between a starting polypeptide sequence and the sequence derived there from. Identity or similarity with respect to this sequence is defined herein as the percentage of amino acid residues in the candidate sequence that are identical (i.e., same residue) with the starting amino acid residues, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. In certain embodiments, a polypeptide consists of, consists essentially of, or comprises an amino acid sequence selected from a sequence set forth in any one of Tables 5-8. In certain embodiments, a polypeptide includes an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to an amino acid sequence selected from a sequence set forth in any one of Tables 5-8. In certain embodiments, a polypeptide includes a contiguous amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a contiguous amino acid sequence selected from a sequence set forth in any one of Tables 5-8. In certain embodiments, a polypeptide includes an amino acid sequence having at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 200, 300, 400, or 500 (or any integer within these numbers) contiguous amino acids of an amino acid sequence selected from a sequence set forth in any one of Tables 5-8.

In certain embodiments, the antibodies of the disclosure are encoded by a nucleotide sequence. Nucleotide sequences of the invention can be useful for a number of applications, including: cloning, gene therapy, protein expression and purification, mutation introduction, DNA vaccination of a host in need thereof, antibody generation for, e.g., passive immunization, PCR, primer and probe generation, and the like. In certain embodiments, the nucleotide sequence of the invention comprises, consists of, or consists essentially of, a nucleotide sequence selected from a sequence set forth in any one of Tables 5-8. In certain embodiments, a nucleotide sequence includes a nucleotide sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a nucleotide sequence selected from a sequence set forth in any one of Tables 5-8. In certain embodiments, a nucleotide sequence includes a contiguous nucleotide sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a contiguous nucleotide sequence selected from a sequence set forth in any one of Tables 5-8. In certain embodiments, a nucleotide sequence includes a nucleotide sequence having at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 200, 300, 400, or 500 (or any integer within these numbers) contiguous nucleotides of a nucleotide sequence selected from a sequence set forth in any one of Tables 5-8.

It will also be understood by one of ordinary skill in the art that the antibodies suitable for use in the methods disclosed herein may be altered such that they vary in sequence from the naturally occurring or native sequences from which they were derived, while retaining the desirable activity of the native sequences. For example, nucleotide or amino acid substitutions leading to conservative substitutions or changes at "non-essential" amino acid residues may be made. Mutations may be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis.

The antibodies suitable for use in the methods disclosed herein may comprise conservative amino acid substitutions at one or more amino acid residues, e.g., at essential or non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a nonessential amino acid residue in a binding polypeptide is preferably replaced with another amino acid residue from the same side chain family. In certain embodiments, a string of amino acids can be replaced with a structurally similar string that differs in order and/or composition of side chain family members. Alternatively, in certain embodiments, mutations may be introduced randomly along all or part of a coding sequence, such as by saturation mutagenesis, and the resultant mutants can be incorporated into binding polypeptides of the invention and screened for their ability to bind to the desired target.

As used herein, the term antigen“cross-presentation” refers to presentation of exogenous protein antigens to T cells via MHC class I and class II molecules on APCs.

As used herein, the term“cross -reacts” refers to the ability of an antibody of the disclosure to bind to CD137 from a different species. For example, an antibody of the present disclosure which binds human CD137 may also bind another species of CD137. As used herein, cross reactivity is measured by detecting a specific reactivity with purified antigen in binding assays (e.g., SPR, ELISA) or binding to, or otherwise functionally interacting with, cells physiologically expressing CD137. Methods for determining cross-reactivity include standard binding assays as described herein, for example, by Biacore™ surface plasmon resonance (SPR) analysis using a Biacore™ 2000 SPR instrument (Biacore AB, Uppsala, Sweden), or flow cytometric techniques. As used herein, the term“cytotoxic T lymphocyte (CTL) response” refers to an immune response induced by cytotoxic T cells. CTL responses are mediated primarily by CD8 + T cells.

As used herein the terms "diagnose" or "diagnosis" or "diagnosing" refer to distinguishing or identifying a disease, syndrome or condition or distinguishing or identifying a person having a particular disease, syndrome, or condition.

As used herein, the term“diagnostic antibody” denotes an antibody used for the detection and visualization of its target antigen (e.g., a biomarker). In some embodiments, diagnostic antibody is used e.g. in assay systems (e.g. ELISA), or for imaging techniques (e.g., IHC). A diagnostic antibody may e.g., be a labeled therapeutic antibody.

As used herein, the term“diagnostic biomarker” refers to a biomarker used to detect or confirm presence of a disease or condition of interest or to identify individuals with a subtype of the disease.

As used herein, the term "dimerization" refers to the formation of a macromolecular complex by two, usually non-covalently bound, macromolecules, such as proteins or multimers of proteins. Homodimerization refers to the process of dimerization when the macromolecules (e.g., proteins) are identical in nature. Heterodimerization refers to the process of dimerization when the macromolecules (e.g., proteins) are non-identical in nature. Methods for determining dimerization are known to those of skill in the art. For example, such methods include, but are not limited to, yeast two-hybrid assay, fluorescence resonance energy transfer (FRET), bioluminescence resonance energy transfer (BRET), protein mass spectrometry, evanescent wave methods, size exclusion chromatography, analytical ultracentrifugation, scattering techniques, NMR spectroscopy, isothermal titration calorimetry, fluorescence anisotropy, fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), proximity imaging (PRIM) and bimolecular fluorescence complementation (BiFC) (see e.g., Gell D.A., Grant R.P., Mackay J.P. (2012) The Detection and Quantitation of Protein Oligomerization. In: Matthews J.M. (eds) Protein Dimerization and Oligomerization in Biology. Advances in Experimental Medicine and Biology, vol 747. Springer, New York, NY; and Xie, Q. et al. Methods Mol Biol, 2011; 680: 3-28).

As used herein, the terms "dimerization of CD 137" refers to the dimerization of two CD 137 trimers. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance dimerization of CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance dimerization of CD 137 relative to the amount of dimerization in the absence of an anti-CD 137 agonist antibody. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance dimerization of CD 137 relative to the amount of dimerization in the presence of a reference anti-CDl37 agonist antibody. In some embodiments, dimerization is increased by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.

As used herein, the term“EC50” refers to the concentration of an antibody or an antigen binding portion thereof, which induces a response, either in an in vitro or an in vivo assay, which is 50% of the maximal response, i.e., halfway between the maximal response and the baseline.

As used herein, the term“effective dose” or“effective dosage” is defined as an amount sufficient to achieve or at least partially achieve the desired effect. The term“therapeutically effective dose” is defined as an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient’s own immune system.

As used herein, the term“epitope” or“antigenic determinant” refers to a determinant or site on an antigen (e.g., CD137) to which an antigen-binding protein (e.g., an immunoglobulin, antibody, or antigen-binding fragment) specifically binds. The epitopes of protein antigens can be demarcated into“linear epitopes” and“conformational epitopes”. As used herein, the term“linear epitope” refers to an epitope formed from a contiguous, linear sequence of linked amino acids. Linear epitopes of protein antigens are typically retained upon exposure to chemical denaturants (e.g., acids, bases, solvents, cross-linking reagents, chaotropic agents, disulfide bond reducing agents) or physical denaturants (e.g. thermal heat, radioactivity, or mechanical shear or stress). In some embodiments, an epitope is non-linear, also referred to as an interrupted epitope. As used herein, the term“conformational epitope” or“non-linear epitope” refers to an epitope formed from noncontiguous amino acids juxtaposed by tertiary folding of a polypeptide. Conformational epitopes are typically lost upon treatment with denaturants. An epitope typically includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acids in a unique spatial conformation. In some embodiments, an epitope includes fewer than 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6 or 5 amino acids in a unique spatial conformation. Generally, an antibody, or antigen-binding fragment thereof, specific for a particular target molecule will preferentially recognize and bind to a specific epitope on the target molecule within a complex mixture of proteins and/or macromolecules. In some embodiments, an epitope does not include all amino acids of the extracellular domain of human CD 137.

Also encompassed by the present disclosure are antibodies that bind to an epitope on CD 137 which comprises all or a portion of an epitope recognized by the particular antibodies described herein ( e.g ., the same or an overlapping region or a region between or spanning the region).

As used herein, the term“epitope mapping” refers to a process or method of identifying the binding site, or epitope, of an antibody, or antigen binding fragment thereof, on its target protein antigen. Epitope mapping methods and techniques are provided herein.

As used herein, the term "CD 137" refers to a specific member of the tumor necrosis factor receptor (TNFR) family of transmembrane proteins. Alternative names and acronyms for CD 137 in the art include“tumor necrosis factor receptor superfamily member 9” (TNFRSF9), 4-1BB and “induced by lymphocyte activation” (IFA) (Alderson et al., (1994) Eur J Immunol 24(9):22l9- 2227; Schwarz et al., (1993) Gene 134(2):295-298). An exemplary amino acid sequence of full- length human CD137, including leader, transmembrane, and cytoplasmic domains is set forth in Table 7 (SEQ ID NO: 3) and here:

MGNSCYNIVATFFFVFNFERTRSFQDPCSNCPAGTFCDNNRNQICSPCPPNSFSSAGGQR TCDICRQCKGVFRTRKECSSTSNAECDCTPGFHCFGAGCSMCEQDCKQGQEFTKKGCK DCCFGTFNDQKRGICRPWTNCSFDGKSVFVNGTKERDVVCGPSPADFSPGASSVTPPAP AREPGHS PQIIS FFFAFT S T AFFFFFFFFTFRFS V VKRGRKKFFYIFKQPFMR PVQTTQEEDGCSCRFPEEEEGGCEF.

As used herein, the term“CD137F” or“CD 137 ligand” refers to a member of the tumor necrosis factor (TNF) family of transmembrane proteins. Alternative names and acronyms for CD137F in the art include“tumor necrosis factor superfamily member 9” (TNFSF9) and 4-1BB ligand (4-1BBF) (Alderson et al., (1994) Eur J Immunol 24(9) :2219-2227). An exemplary amino acid sequence of full-length CD137L is set forth in Table 7 (SEQ ID NO: 97).

As used herein, the term“expression” generally refers to the process by which information contained within a gene is converted into the structures (e.g., a protein biomarker) present and operating in the cell. Therefore, as used herein,“expression” may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide). Fragments of the transcribed polynucleotide, the translated polypeptide, or polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide) shall also be regarded as expressed whether they originate from a transcript generated by alternative splicing or a degraded transcript, or from a post-translational processing of the polypeptide, e.g., by proteolysis.“Expressed genes” include those that are transcribed into a polynucleotide as mRNA and then translated into a polypeptide, and also those that are transcribed into RNA but not translated into a polypeptide (for example, transfer and ribosomal RNAs).“Elevated expression,”“elevated expression levels,” or“elevated levels” refers to an increased expression or increased levels of a substance within a sample relative to a control sample, such as an individual or individuals who are not suffering from the disease or disorder (e.g., cancer) or an internal control. In some embodiments, the elevated expression of a substance (e.g., a protein or a biomarker) in a sample refers to an increase in the amount of the substance of about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% relative to the amount of the substance in a control sample, as determined by techniques known in the art (e.g., IHC).“Reduced expression,”“reduced expression levels,” or“reduced levels” refers to a decrease expression or decreased levels of a biomarker in an individual relative to a control, such as an individual or individuals who are not suffering from the disease or disorder (e.g., cancer) or an internal control. In some embodiments, reduced expression is little or no expression. In some embodiments, the reduced expression of a substance (e.g., a protein or a biomarker) in a sample refers to a decrease in the amount of the substance of about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% relative to the amount of the substance in a control sample, as determined by techniques known in the art (e.g., IHC).

As used herein, the term“extent of binding” refers to the level of binding of an antibody (e.g., a diagnostic antibody) to an antigen (e.g., a biomarker) present in a sample. The extent of antibody binding to an antigen can be determined by any of the methods known in the art for determining binding levels of antibodies such as ELISA, Western Blotting, or FACS. The extent of binding may be determined using any detection system such as secondary immunoglobulins or fragments thereof linked to a detectable marker. Exemplary detectable markers include, without limitation, radioactive groups, fluorescent or chromogenic molecules, an enzyme capable of catalyzing a reaction yielding a detectable product (such as a color reaction), and a biotin group capable of being detected by avidin.

As used herein, the terms“Fc-mediated effector functions” or“Fc effector functions” refer to the biological activities of an antibody other than the antibody’s primary function and purpose. For example, the effector functions of a therapeutic agnostic antibody are the biological activities other than the activation of the target protein or pathway. Examples of antibody effect functions include Clq binding and complement dependent cytotoxicity; Fc receptor binding; antibody- dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor); lack of activation of platelets that express Fc receptor; and B cell activation. Many effector functions begin with Fc binding to an Fey receptor.

As used herein, the term“Fc receptor” refers to a polypeptide found on the surface of immune effector cells, which is bound by the Fc region of an antibody. In some embodiments, the Fc receptor is an Fey receptor. There are three subclasses of Fey receptors, FcyRI (CD64), FcyRII (CD32) and FycRIII (CD16). All four IgG isotypes (IgGl, IgG2, IgG3 and IgG4) bind and activate Fc receptors FcyRI, FcyRIIA and FcyRIIIA. FcyRIIB is an inhibitory receptor, and therefore antibody binding to this receptor does not activate complement and cellular responses. FcyRI is a high affinity receptor that binds to IgG in monomeric form, whereas FcyRIIA and FcyRIIA are low affinity receptors that bind IgG only in multimeric form and have slightly lower affinity. The binding of an antibody to an Fc receptor and/or Clq is governed by specific residues or domains within the Fc regions. Binding also depends on residues located within the hinge region and within the CH2 portion of the antibody. In some embodiments, the agonistic and/or therapeutic activity of the antibodies described herein is dependent on binding of the Fc region to the Fc receptor (e.g., FcyR). In some embodiments, the agonistic and/or therapeutic activity of the antibodies described herein is enhanced by binding of the Fc region to the Fc receptor (e.g., FcyR).

As used herein, the term“glycosylation pattern” is defined as the pattern of carbohydrate units that are covalently attached to a protein, more specifically to an immunoglobulin protein. A glycosylation pattern of a heterologous antibody can be characterized as being substantially similar to glycosylation patterns which occur naturally on antibodies produced by the species of the nonhuman transgenic animal, when one of ordinary skill in the art would recognize the glycosylation pattern of the heterologous antibody as being more similar to said pattern of glycosylation in the species of the nonhuman transgenic animal than to the species from which the CH genes of the transgene were derived.

As used herein, the term“hematological cancer” includes a lymphoma, leukemia, myeloma or a lymphoid malignancy, as well as a cancer of the spleen and lymph nodes. Exemplary lymphomas include both B cell lymphomas (a B-cell hematological cancer) and T cell lymphomas. B-cell lymphomas include both Hodgkin's lymphomas and most non-Hodgkin's lymphomas. Non limiting examples of B cell lymphomas include diffuse large B-cell lymphoma, follicular lymphoma, mucosa-associated lymphatic tissue lymphoma, small cell lymphocytic lymphoma (overlaps with chronic lymphocytic leukemia), mantle cell lymphoma (MCL), Burkitt's lymphoma, mediastinal large B cell lymphoma, Waldenstrom macroglobulinemia, nodal marginal zone B cell lymphoma, splenic marginal zone lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, lymphomatoid granulomatosis. Non-limiting examples of T cell lymphomas include extranodal T cell lymphoma, cutaneous T cell lymphomas, anaplastic large cell lymphoma, and angioimmunoblastic T cell lymphoma. Hematological malignancies also include leukemia, such as, but not limited to, secondary leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, and acute lymphoblastic leukemia. Hematological malignancies further include myelomas, such as, but not limited to, multiple myeloma and smoldering multiple myeloma. Other hematological and/or B cell- or T-cell- associated cancers are encompassed by the term hematological malignancy.

As used herein, the term“human antibody” includes antibodies having variable and constant regions (if present) of human germline immunoglobulin sequences. Human antibodies of the disclosure can include amino acid residues not encoded by human germline immunoglobulin sequences ( e.g ., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo ) (See, e.g., Lonberg et ah, (1994) Nature 368(6474): 856-859); Lonberg, (1994) Handbook of Experimental Pharmacology 113:49-101; Lonberg & Huszar, (1995) Intern. Rev. Immunol. 13:65-93, and Harding & Lonberg, (1995) Ann. N.Y. Acad. Sci. 764:536-546). However, the term“human antibody” does not include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences ( i.e . humanized antibodies).

As used herein, the term“heterologous antibody” is defined in relation to the transgenic non-human organism producing such an antibody. This term refers to an antibody having an amino acid sequence or an encoding nucleic acid sequence corresponding to that found in an organism not consisting of the transgenic non-human animal, and generally from a species other than that of the transgenic non-human animal.

As used herein, the term“indicates” or“indicating” refers to a relationship between the presence of a substance or substances (e.g., biomarkers) and one or more events (e.g., response to treatment). In some embodiments, indicates refers to a positive relationship or positive correlation in which as one increases, the other increases as well. In some embodiments, indicates refers to a negative relationship or negative correlation in which as one increases, the other decreases. The present disclosure provides biomarkers, the levels of which indicate or predict a likely outcome, such as presence, amount or level of a biomarker (e.g., MHC I) in a patient sample, and the likelihood a patient will respond to treatment with an agent. For example, the presence, amount or level of a biomarker can indicate a likelihood of a positive clinical outcome for the patient, such as an increased likelihood of long-term survival without recurrence and/or a positive response to treatment. Such a positive indication may be demonstrated statistically in various ways, e.g., by a low hazard ratio. In another example, the presence, amount or level of a biomarker may negatively indicate a likelihood of good clinical outcome for the patient. In this case, for example, the patient may have a decreased likelihood of a positive response to treatment. Such a negative indication may be demonstrated statistically in various ways, e.g., by a high hazard ratio.

The terms“inducing an immune response” and“enhancing an immune response” are used interchangeably and refer to the stimulation of an immune response (i.e., either passive or adaptive) to a particular antigen. The term“induce” as used with respect to inducing CDC or ADCC refer to the stimulation of particular direct cell killing mechanisms.

As used herein, a subject“in need of prevention,”“in need of treatment,” or“in need thereof,” refers to one, who by the judgment of an appropriate medical practitioner (e.g., a doctor, a nurse, or a nurse practitioner in the case of humans; a veterinarian in the case of non-human mammals), would reasonably benefit from a given treatment (such as treatment with a composition comprising an anti-CD 137 antibody).

The term "in vivo" refers to processes that occur in a living organism.

As used herein, the term“isolated antibody” is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to human CD 137 is substantially free of antibodies that specifically bind antigens other than CD137). An isolated antibody that specifically binds to an epitope may, however, have cross-reactivity to other CD137 proteins from different species. However, the antibody continues to display specific binding to human CD137 in a specific binding assay as described herein. In addition, an isolated antibody is typically substantially free of other cellular material and/or chemicals. In some embodiments, a combination of“isolated” antibodies having different CD 137 specificities is combined in a well-defined composition.

As used herein, the term“isolated nucleic acid molecule” refers to nucleic acids encoding antibodies or antibody portions ( e.g ., VH, VL, CDR3) that bind to CD137, is intended to refer to a nucleic acid molecule in which the nucleotide sequences encoding the antibody or antibody portion are free of other nucleotide sequences encoding antibodies or antibody portions that bind antigens other than CD137, which other sequences may naturally flank the nucleic acid in human genomic DNA. For example, a sequence selected from a sequence set forth in any one of Tables 5- 8corresponds to the nucleotide sequences comprising the heavy chain (VH) and light chain (VL) variable regions of anti-CD 137 antibody monoclonal antibodies described herein.

As used herein,“isotype” refers to the antibody class (e.g., IgM or IgGl) that is encoded by heavy chain constant region genes. In some embodiments, a human monoclonal antibody of the disclosure is of the IgGl isotype. In some embodiments, a human monoclonal antibody of the disclosure is of the IgGl isotype and comprises a mutation. In some embodiments, a human monoclonal antibody of the disclosure is of the IgG2 isotype. In some embodiments, a human monoclonal antibody of the disclosure is of the IgG3 isotype. In some embodiments, a human monoclonal antibody of the disclosure is of the IgG4 isotype. In some embodiments, a human monoclonal antibody of the disclosure is of the IgG4 isotype and comprises a mutation. In some embodiments, the mutation is a substitution at Ser228. In some embodiments, the substitution at Ser228 is S228P.

As used herein, the term“isotype switching” refers to the phenomenon by which the class, or isotype, of an antibody changes from one Ig class to one of the other Ig classes.

As used herein the term“KD” or“K D ” refers to the equilibrium dissociation constant of a binding reaction between an antibody and an antigen. The value of K D is a numeric representation of the ratio of the antibody off-rate constant (kd) to the antibody on-rate constant (ka). The value of K D is inversely related to the binding affinity of an antibody to an antigen. The smaller the K D value the greater the affinity of the antibody for its antigen. Affinity is the strength of binding of a single molecule to its ligand and is typically measured and reported by the equilibrium dissociation constant (K D ), which is used to evaluate and rank order strengths of bimolecular interactions.

As used herein, the term“kd” or“k d ” (alternatively“koff’ or“k 0ff ”) is intended to refer to the off-rate constant for the dissociation of an antibody from an antibody/antigen complex. The value of kd is a numeric representation of the fraction of complexes that decay or dissociate per second, and is expressed in units sec 1 .

As used herein, the term“ka” or“k a ” (alternatively“kon” or“k on ”) is intended to refer to the on-rate constant for the association of an antibody with an antigen. The value of ka is a numeric representation of the number of antibody/antigen complexes formed per second in a 1 molar (1M) solution of antibody and antigen, and is expressed in units M^sec 1 .

As used herein, the term“likelihood” generally refers to an increase in the probability of an event (e.g., a patient will respond to treatment) and may be expressed as a probability value, fraction, or percentage. The term“likelihood” may include the concept of“probability” as used in mathematics and by persons of ordinary skill in the art of statistics. The term“likelihood” when used in reference to a patient’s response to treatment, the term contemplates an increase in the probability that the patient responds positively to treatment. The term“likelihood” when used in reference to a tumor’ s response to treatment contemplates a change in the amount of one or more biomarkers present in the tumor or tumor sample that may evidence a progression in treating the tumor.

As used herein, the terms "linked," "fused", or "fusion", are used interchangeably. These terms refer to the joining together of two more elements or components or domains, by whatever means including chemical conjugation or recombinant means. Methods of chemical conjugation (e.g., using heterobifunctional crosslinking agents) are known in the art.

As used herein,“local administration” or“local delivery,” refers to delivery that does not rely upon transport of the composition or agent to its intended target tissue or site via the vascular system. For example, the composition may be delivered by injection or implantation of the composition or agent or by injection or implantation of a device containing the composition or agent. Following local administration in the vicinity of a target tissue or site, the composition or agent, or one or more components thereof, may diffuse to the intended target tissue or site.

As used herein, the term“monitor,” carries its common usage and can refer to the surveillance or continual observation of a process or event (e.g., a response to treatment). For example, the term“monitoring a patient’s response to treatment” contemplates, for example, observation or surveillance of disease progression. The monitoring can be performed, for example, by following or determine the expression of biomarkers over time.

As used herein, the term“monitoring biomarker” refers to a biomarker measured serially for assessing status of a disease or medical conditions or for evidence of exposure to (or effect of) a medical product or agent.

As used herein, the term“monoclonal antibody” refers to an antibody which displays a single binding specificity and affinity for a particular epitope. Accordingly, the term“human monoclonal antibody” refers to an antibody which displays a single binding specificity, and which has variable and optional constant regions derived from human germline immunoglobulin sequences. In some embodiments, human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.

As used herein, the term“multimerization” refers to the formation of a macromolecular complex comprising more than two macromolecules such as proteins, typically bound by non- covalent interactions. Methods for determining multimerization are known to those of skill in the art and are described supra for dimerization. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance multimerization of CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance multimerization of CD 137 relative to the amount of multimerization in the absence of an anti-CD 137 agonist antibody. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance multimerization of CD 137 relative to the amount of multimerization in the presence of a reference anti-CDl37 agonist antibody. In some embodiments, multimerization is increased by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.

As used herein, the term“naturally-occurring” as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring. As used herein, the term“nonswitched isotype” refers to the isotypic class of heavy chain that is produced when no isotype switching has taken place; the CH gene encoding the nonswitched isotype is typically the first CH gene immediately downstream from the functionally rearranged VDJ gene. Isotype switching has been classified as classical or non-classical isotype switching. Classical isotype switching occurs by recombination events which involve at least one switch sequence region in the transgene. Non-classical isotype switching may occur by, for example, homologous recombination between human qm and human å m (d-associated deletion). Alternative non-classical switching mechanisms, such as intertransgene and/or interchromosomal recombination, among others, may occur and effectuate isotype switching.

As used herein, the term "nucleic acid" refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double- stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences and as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions can be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081, 1991; Ohtsuka et al., Biol. Chem. 260:2605-2608, 1985; and Cassol et al, 1992; Rossolini et al, Mol. Cell. Probes 8:91-98, 1994). For arginine and leucine, modifications at the second base can also be conservative. The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene.

As used herein, the term“nucleic acid probe” refers to a fragment of DNA or RNA that is labeled with a tag. The nucleic acid probe is added to a DNA or RNA sample to detect and/or recover nucleotide sequences that are complementary to the sequence in the probe. The probe hybridizes to a single-stranded nucleic acid (DNA or RNA) whose base sequence allows probe- target base pairing due to complementarity between the nucleic acid probe and target. Unless otherwise indicated, the term encompasses nucleic acid probes labeled with tags including radioactive labels, fluorophores, enzymes, or nucleotides modified with digoxygenin or biotin.

Polynucleotides used herein can be composed of any polyribonucleotide or polydeoxribonucleotide, which can be unmodified RNA or DNA or modified RNA or DNA. For example, polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double- stranded regions, single- and double- stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that can be single- stranded or, more typically, double-stranded or a mixture of single- and double- stranded regions. In addition, the polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA. A polynucleotide can also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons. "Modified" bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications can be made to DNA and RNA; thus, "polynucleotide" embraces chemically, enzymatically, or metabolically modified forms.

A nucleic acid is“operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence. With respect to transcription regulatory sequences, operably linked means that the DNA sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame. For switch sequences, operably linked indicates that the sequences are capable of effecting switch recombination.

As used herein, the term“panel of biomarkers” includes a group of markers, the quantity or activity of each member of which indicates responsiveness to treatment with an agent as described herein. The determination of a panel of biomarkers can provide more sensitive and specific information needed to evaluate a tumor or a patient. In some embodiments, the panel of biomarkers include one, two, three, four, five, six, seven, eight, or nine or more biomarkers. In some embodiments, the panel comprises MHC I, beta-2-microglobulin chain (b2M), and MHC class I alpha chain. In some embodiments, the panel comprises MHC I and b2M. In some embodiments, the panel comprises MHC I and MHC class I alpha chain. In some embodiments, the panel comprises b2M and MHC class I alpha chain.

As used herein, the term“paratope”, also“antigen-binding site” refers to a portion of an antibody, or antigen-binding fragment thereof, which recognizes and binds to an epitope on an antigen, comprising the set of complementarity determining regions (CDRs) located within variable heavy and light chains. As used herein, “parenteral administration,” “administered parenterally,” and other grammatically equivalent phrases, refer to modes of administration other than enteral and topical administration, usually by injection, and include, without limitation, intravenous, intranasal, intraocular, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, intracerebral, intracranial, intracarotid and intrasternal injection and infusion.

As used herein, the term“patient” includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment. “Patient” is sometimes used interchangeably with“subject”.

The term "percent identity," in the context of two or more nucleic acid or polypeptide sequences, refer to two or more sequences or subsequences that have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned for maximum correspondence, as measured using one of the sequence comparison algorithms described below (e.g., BLASTP and BLASTN or other algorithms available to persons of skill) or by visual inspection. Depending on the application, the "percent identity" can exist over a region of the sequence being compared, e.g., over a functional domain, or, alternatively, exist over the full length of the two sequences to be compared. For sequence comparison, typically one sequence acts as a reference sequence to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.

Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat'l. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by visual inspection (see generally Ausubel et ah, infra). One example of an algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in Altschul et ah, J. Mol. Biol. 215:403-410 (1990). Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information website.

As generally used herein, “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.

As used herein, a“pharmaceutically acceptable carrier” refers to, and includes, any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. The compositions can include a pharmaceutically acceptable salt, e.g., an acid addition salt or a base addition salt (see, e.g., Berge et al. (1977) J Pharm Sci 66: 1-19).

As used herein, the terms "polypeptide," "peptide", and "protein" are used interchangeably to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.

As used herein, the term“preventing” when used in relation to a condition, refers to administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition.

As used herein, the term“purified” or“isolated” as applied to any of the proteins (antibodies or fragments) described herein refers to a polypeptide that has been separated or purified from components (e.g., proteins or other naturally-occurring biological or organic molecules) which naturally accompany it, e.g., other proteins, lipids, and nucleic acid in a prokaryote expressing the proteins. Typically, a polypeptide is purified when it constitutes at least 60 (e.g., at least 65, 70, 75, 80, 85, 90, 92, 95, 97, or 99) %, by weight, of the total protein in a sample.

As used herein, the term“rearranged” refers to a configuration of a heavy chain or light chain immunoglobulin locus wherein a V segment is positioned immediately adjacent to a D-J or J segment in a conformation encoding essentially a complete V H or V L domain, respectively. A rearranged immunoglobulin gene locus can be identified by comparison to germline DNA; a rearranged locus will have at least one recombined heptamer/nonamer homology element.

As used herein, the term“receptor clustering” refers to a cellular process that results in grouping or local accumulation of a set of receptors at a particular cellular location, often to induce or amplify a signaling response. Many protein receptors bind cognate ligands and cluster, i.e., form dimers, trimers, oligomers or multimers, upon binding their cognate ligands. For example, the PDGF receptor and TNF receptor superfamily members form dimers and trimers upon ligand binding, respectively. Cognate ligand-induced clustering (e.g., dimerization, multimerization) induces signal transduction through the receptor. Accordingly, the antibodies, or antigen-binding fragments thereof, of the present disclosure can activate a receptor by binding to more than one receptor and induce or stabilize dimerization, trimerization, and/or multimerization with or without cognate ligand binding.

Receptor clustering and multimerization is needed for TNFR signaling (Wajant (2015) Cell Death Differ 22(11): 1727-1741), and in particular for TNFRSF activation. 4-1BB (CD137), CD40, GITR, CD27, DR3, DR5, and Fas are some of the TNFSF receptors known to require clustering in order to trigger downstream signaling. Experimental evidence that the 4-1BB receptor must be cross-linked to signal comes from Rabu et al. These authors reported that a l-trimer form of human 4-1BBL had no activating effects on human T cells whereas cross-linking the protein into 2- or more trimers led to a strongly activating protein (Rabu et ah, (2005) J Biol Chem 280:41472- 41481). Accordingly, in some embodiments, an anti-CDl37 agonist antibody induces the multimerization of 2 or more trimers of CD 137.

As used herein, the term“recombinant host cell” (or simply“host cell”) is intended to refer to a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term“host cell” as used herein.

As used herein, the term“recombinant human antibody” includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies comprise variable and constant regions that utilize particular human germline immunoglobulin sequences are encoded by the germline genes, but include subsequent rearrangements and mutations which occur, for example, during antibody maturation. As known in the art (see, e.g. , Lonberg (2005) Nature Biotech. 23(9): 1117-1125), the variable region contains the antigen binding domain, which is encoded by various genes that rearrange to form an antibody specific for a foreign antigen. In addition to rearrangement, the variable region can be further modified by multiple single amino acid changes (referred to as somatic mutation or hypermutation) to increase the affinity of the antibody to the foreign antigen. The constant region will change in further response to an antigen ( i.e ., isotype switch). Therefore, the rearranged and somatically mutated nucleic acid molecules that encode the light chain and heavy chain immunoglobulin polypeptides in response to an antigen may not have sequence identity with the original nucleic acid molecules, but instead will be substantially identical or similar (i.e., have at least 80% identity).

As used herein, the term“reference antibody” (used interchangeably with“reference mAb”) or“reference antigen-binding protein” refers to an antibody, or an antigen-binding fragment thereof, that binds to a specific epitope on human CD 137 and is used to establish a relationship between itself and one or more distinct antibodies. In some embodiments, the relationship is the binding of the reference antibody and the one or more distinct antibodies to the same epitope on CD 137. As used herein, the term connotes an anti-CD 137 antibody that is useful in a test or assay, such as those described herein, (e.g., a competitive binding assay), as a competitor, wherein the assay is useful for the discovery, identification or development, of one or more distinct antibodies that bind to the same epitope. The variable heavy (V H ) and light chain (V L ) amino acid sequences of an exemplary reference antibody (mAbl) are provided in Table 7 (V H l, SEQ ID NO. 4; V H 2, SEQ ID NO. 6). In some embodiments, the term connotes an anti- CD 137 antibody that is useful in a test or assay, as a comparator, wherein the assay is useful for distinguishing characteristics of the antibodies (e.g., hepatotoxicity, anti-tumor efficacy). In some embodiments, the reference antibody is urelumab. In some embodiments, the reference antibody is utomilumab.

As used herein, the term“sample” refers to a composition that is obtained or derived from a patient, subject or individual of interest that contains or may contain a cellular or molecular entity, signature or substance (e.g., a biomarker) that is to be detected, quantified, identified, or otherwise characterized, for example based on physical, biochemical, chemical, or physiological characteristics, or a combination thereof. A“tumor sample” refers to a sample taken from a subject having a tumor (e.g., a cancer patient), wherein the sample comprising tumor cells. In some embodiments, a tumor sample comprises malignant (tumor) and non-malignant cells, optionally, extracellular components. A“reference sample” (alternatively “control sample”) refers to a sample, cell, tissue, standard, or level that is used for comparison purposes. For example, in some embodiments a reference sample is a sample known to contain one or more biomarkers of interest. The amount or level of a biomarker in a reference sample is referred to as a“reference amount.” In certain embodiments of the disclosure, a reference sample is a sample obtained from a healthy and/or non-diseased part of the body of the same patient, subject or individual. For example, healthy and/or non-diseased cells or tissues adjacent to the diseased cells or tissues (e.g., cells or tissues adjacent to a tumor). In another embodiment, a reference sample is obtained from an untreated tissue and/or cell of the body of the same patient, subject or individual. In another embodiment, a reference sample is obtained from a healthy and/or non-diseased part of the body (e.g., tissues or cells) of an individual who is not the patient or subject. In another embodiment, a reference sample is obtained from an untreated tissue and/or cell of the body of an individual who is not the patient or subject. A sample may be obtained by any technique (e.g., biopsy) known to a person skilled in the art. In some embodiments, samples may comprise tissue (referred to as a “tissue sample”) or cells (referred to as a“cell sample”).

As used herein, the terms“specific binding,”“selective binding,”“selectively binds,” and “specifically binds,” refer to antibody binding to an epitope on a predetermined antigen. Typically, the antibody binds with an equilibrium dissociation constant (K D ) of approximately less than 10 6 M, such as approximately less than 10 7 , 10 8 M, 10 9 M or 10 10 M or even lower when determined by surface plasmon resonance (SPR) technology in a BIACORE 2000 instrument using recombinant human CD 137 as the analyte and the antibody as the ligand and binds to the predetermined antigen with an affinity that is at least two-fold greater than its affinity for binding to a non-specific antigen ( e.g ., BSA, casein) other than the predetermined antigen or a closely- related antigen. The phrases“an antibody recognizing an antigen” and“an antibody specific for an antigen” are used interchangeably herein with the term“an antibody which binds specifically to an antigen.”

As used herein, the term“switch sequence” refers to those DNA sequences responsible for switch recombination. A“switch donor” sequence, typically a m switch region, will be 5' (i.e., upstream) of the construct region to be deleted during the switch recombination. The“switch acceptor” region will be between the construct region to be deleted and the replacement constant region (e.g., g, e, etc.). As there is no specific site where recombination always occurs, the final gene sequence will typically not be predictable from the construct.

As used herein, the term“subject” includes any human or non-human animal. For example, the methods and compositions of the present invention can be used to treat a subject with an immune disorder. The term“non-human animal” includes all vertebrates, e.g., mammals and non mammals, such as non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc. “Subject” is sometimes used interchangeably with“patient”.

For nucleic acids, the term“substantial homology” indicates that two nucleic acids, or designated sequences thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides. Alternatively, substantial homology exists when the segments will hybridize under selective hybridization conditions, to the complement of the strand.

The percent identity between two sequences is a function of the number of identical positions shared by the sequences ( i.e ., % homology = # of identical positions/total # of positions x 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.

The percent identity between two nucleotide sequences can be determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. The percent identity between two nucleotide or amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (CABIOS, 4: 11-17 (1989)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol. (48):444- 453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.

The nucleic acid and protein sequences of the present disclosure can further be used as a “query sequence” to perform a search against public databases to, for example, identify related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to the nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to the protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al, (1997) Nucleic Acids Res. 25(l7):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs ( e.g ., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov.

The nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form. A nucleic acid is“isolated” or“rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, L. Ausubel, et al, ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987).

The nucleic acid compositions of the present disclosure, while often in a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures thereof may be mutated, in accordance with standard techniques to provide gene sequences. Lor coding sequences, these mutations, may affect amino acid sequence as desired. In particular, DNA sequences substantially homologous to or derived from native V, D, J, constant, switches and other such sequences described herein are contemplated (where "derived" indicates that a sequence is identical or modified from another sequence).

As used herein, the term ’’tumor microenvironment” (alternatively ’’cancer microenvironment”; abbreviated TME) refers to the cellular environment or milieu in which the tumor or neoplasm exists, including surrounding blood vessels as well as non-cancerous cells including, but not limited to, immune cells, fibroblasts, bone marrow-derived inflammatory cells, and lymphocytes. Signaling molecules and the extracellular matrix also comprise the TME. The tumor and the surrounding microenvironment are closely related and interact constantly. Tumors can influence the microenvironment by releasing extracellular signals, promoting tumor angiogenesis and inducing peripheral immune tolerance, while the immune cells in the microenvironment can affect the growth and evolution of tumor cells.

The term“T cell” refers to a type of white blood cell that can be distinguished from other white blood cells by the presence of a T cell receptor on the cell surface. There are several subsets of T cells, including, but not limited to, T helper cells (a.k.a. T H cells or CD4 + T cells) and subtypes, including T H I , T H 2, T H 3, T H 17, T H 9, and T FH cells, cytotoxic T cells ( i.e Tc cells, CD8 + T cells, cytotoxic T lymphocytes, T-killer cells, killer T cells), memory T cells and subtypes, including central memory T cells (TCM cells), effector memory T cells (TEM and TEMRA cells), and resident memory T cells (TRM cells), regulatory T cells (a.k.a. T reg cells or suppressor T cells) and subtypes, including CD4 + FOXP3 + T reg cells, CD4 + FOXP3 T reg cells, Trl cells, Th3 cells, and T reg l7 cells, natural killer T cells (a.k.a. NKT cells), mucosal associated invariant T cells (MAITs), and gamma delta T cells (gd T cells), including Vy9/ V 52 T cells. Any one or more of the aforementioned or unmentioned T cells may be the target cell type for a method of use of the invention.

As used herein, the term”T cell activation” or’’activation of T cells” refers to a cellular process in which mature T cells, which express antigen- specific T cell receptors on their surfaces, recognize their cognate antigens and respond by entering the cell cycle, secreting cytokines or lytic enzymes, and initiating or becoming competent to perform cell-based effector functions. T cell activation requires at least two signals to become fully activated. The first occurs after engagement of the T cell antigen- specific receptor (TCR) by the antigen-major histocompatibility complex (MHC), and the second by subsequent engagement of co- stimulatory molecules (e.g., CD28). These signals are transmitted to the nucleus and result in clonal expansion of T cells, upregulation of activation markers on the cell surface, differentiation into effector cells, induction of cytotoxicity or cytokine secretion, induction of apoptosis, or a combination thereof.

As used herein, the term“T cell-mediated response” refers to any response mediated by T cells, including, but not limited to, effector T cells (e.g., CD8 + cells) and helper T cells (e.g., CD4 + cells). T cell mediated responses include, for example, T cell cytotoxicity and proliferation.

As used herein, the terms“therapeutically effective amount” or“therapeutically effective dose,” or similar terms used herein are intended to mean an amount of an agent (e.g., an anti- CD 137 antibody or an antigen-binding fragment thereof) that will elicit the desired biological or medical response (e.g., an improvement in one or more symptoms of a cancer).

The terms“treat,”“treating,” and“treatment,” as used herein, refer to therapeutic or preventative measures described herein. The methods of“treatment” employ administration to a subject, in need of such treatment, a human antibody of the present disclosure, for example, a subject in need of an enhanced immune response against a particular antigen or a subject who ultimately may acquire such a disorder, in order to prevent, cure, delay, reduce the severity of, or ameliorate one or more symptoms of the disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.

As used herein, the term “unrearranged” or “germline configuration” refers to the configuration wherein the V segment is not recombined so as to be immediately adjacent to a D or J segment.

As used herein, the term“vector” is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a“plasmid,” which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification,“plasmid” and“vector” may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors ( e.g ., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains. Preferred methods and materials are described below, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the presently disclosed methods and compositions. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.

Methods of Detecting/Identifying MHC I-expressing Cancer Cells

As described herein, the anti-CD 137 agonist antibody, or antigen-binding fragment thereof, described herein demonstrate anti-tumor efficacy in cancer cells that express MHC I. Thus, the present discovery can be applied to identify a cancer patient population that would be amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein, thereby enhancing therapeutic efficacy of the anti-CDl37 agonist antibodies.

Thus, in certain embodiments, the present disclosure provides methods for detecting the presence (or absence) of MHC I in a biological sample, e.g., a biopsy obtained from a subject having or suspected of having any one or more types of cancer disclosed herein, thereby allowing the identification of a subject amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein.

As used herein, cancer that is“amenable” to agonist anti-CD 137 antibody therapy includes those cancers that, upon being administered a therapeutically effective amount of an agonist anti- CD 137 antibody, can benefit from the therapy so as to be“treated”, as used herein.

In some embodiments, the present disclosure also provides methods for determining whether a subject having cancer would be amenable to agonist CD 137 antibody, or antigen binding fragment thereof, therapy. The method comprises contacting a biological sample, e.g., a biopsy obtained from a subject having or suspected of having any one or more types of cancer disclosed herein, from the subject with an agent directed to MHC I, and detecting the agent bound to MHC I according to known methods, and as described herein, wherein the presence of MHC I indicates that the cancer is amendable to treatment with an agonist CD 137 antibody, or an antigen binding fragment thereof.

In some embodiments, the method further comprises determining that the cancer is amenable to treatment with an agonist CD 137 antibody, or antigen-binding fragment thereof (by the presence of MHC I), and administering to the subject a therapeutically effective amount of an agonist anti-CD 137 antibody, or antigen-binding fragment thereof.

In some embodiments, detecting MHC I expression is performed by detecting the presence of any one or more markers selected from the group consisting of: MHC I protein, MHC I mRNA, beta-2-microglobulin chain (b2M) protein, b2M chain mRNA, MHC class I alpha chain domain protein (e.g., ocl, oc2, or oc3), and MHC class I alpha chain domain mRNA. As commonly known in the art, the alpha chains are encoded by any one of human leukocyte antigen (HLA) -A, -B, and -C genes, which are three of the major types of human MHC class I cell surface receptors. Thus, those skilled in the art would recognize that any one or more of the class I alpha chains and/or their specific domains (e.g., ocl, oc2, or oc3), can be suitable for detecting MHC I expression.

In some embodiments, MHC I expression is detected with an agent directed to any one or more of the markers. For example, the agent is an antibody or a nucleic acid probe. In some embodiments, the antibody or probe comprises a detectable label. Those skilled in the art can readily determine the appropriate agent, e.g., antibody or probe, depending on the particular MHC I (e.g., HLA-A, HLA-B, or HLA-C) being detected. While the HLA-A, -B, and -C genes encoding the alpha chains of the MHC I complex are known to be polymorphic, those skilled in the art can readily determine their sequences, and select and/or design a suitable detecting agent directed to the alpha chain and/or the b2M chain.

An exemplary amino acid sequence of MHC I (HLA-A) is shown in SEQ ID NO: 134 (its nucleotide sequence is shown in SEQ ID NO: 135); the amino acid sequence of b2M is shown in SEQ ID NO: 136 (its nucleotide sequence is shown in SEQ ID NO: 137). As those of skill in the art would recognize, the present methods contemplate the use of a sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 134. As those of skill in the art would recognize, the present methods contemplate the use of a sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 135. As those of skill in the art would recognize, the present methods contemplate the use of a sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 136. As those of skill in the art would recognize, the present methods contemplate the use of a sequence that is at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 137.

In some embodiments, MHC I is detected in a sample or tumor using an agent that binds MHC I protein or MHC I mRNA. In some embodiments, the agent that binds MHC I protein is an antibody. In some embodiments, the agent that binds MHC I mRNA is a nucleic acid probe.

In some embodiments, MHC I is detected in a sample or a tumor using an agent that binds beta-2-microglobulin (b2M protein) or b2M mRNA. In some embodiments, the agent that binds b2M protein is an antibody. In some embodiments, the agent that binds b2M mRNA is a nucleic acid probe.

In some embodiments, MHC I is detected in a sample or a tumor using an agent that binds MHC class I alpha chain protein or MHC class I alpha chain mRNA. In some embodiments, the agent that binds MHC class I alpha chain protein is an antibody. In some embodiments, the agent that binds MHC class I alpha chain mRNA is a nucleic acid probe.

The presence of MHC I can be determined by any of the methods known in the art, and include, for example, detecting MHC I in its entirety, or detecting any one or more of its subunits and variants, as described herein. Further, various agents can be used to detect the presence of the markers (e.g., MHC I itself, or any of the subunit(s) and/or component(s) that indicate the presence of MHC I (e.g., any one of more of the 3 alpha subdomains of the class I alpha chains, and polymorphs thereof, and beta2-microglobulin), depending on the assay and/or detection method used. Non-limiting examples of an agent that binds to the marker are nucleic acid probes, or protein binding agents such as antibodies, antibody fragments, or aptamers, or combinations thereof.

For example, an antibody against any one or more of the protein markers can be used if the detection method to be used is, e.g., ELISA. In some embodiments, the antibody is an anti- MHC I antibody, such as MHC Class I (H-2Kd/H-2Dd) antibody from ebioscience (#12-5998- 82) or ebioscience (#(14-5998-82). In some embodiments, the antibody is an anti^2M antibody, such as anti-mouse b2M antibody from Biolegend (#154506) or anti-human b2M antibody from Biolegend (#316302). In some embodiments, the antibody is an anti-MHC class I alpha chain antibody, such as Human HLA Class I antibody from R& D Systems (#MAB7098). Other agents, such as nucleic acid probes against any one or more of the mRNA markers can be used if the detection method to be used is, e.g., RT-PCR. Depending on the application, the agent can include a detectable label commonly known in the art.

As those of skill in the art would recognize, the presence of a marker can be determined by detecting the level of marker mRNA, marker protein, or combinations thereof (e.g., MHC I protein, MHC I mRNA, beta-2-microglobulin chain (b2M) protein, b2M chain mRNA, MHC class I alpha chain protein, and MHC class I alpha chain mRNA).

Suitable biological assays for detecting the marker include, but are not limited to, PCR based methods, hybridization-based methods, sequencing methods, protein detection methods, or combinations thereof. Examples of suitable PCR based methods comprise any form of reverse transcriptase polymerase chain reaction (RT-PCR), any form of quantitative reverse transcriptase polymerase chain reaction (QRT-PCR), competitive RT-PCR, and real-time RT-PCR. Examples of suitable hybridization methods comprise nuclease protection assay, Northern blot analysis, in situ hybridization, and microarray-based analysis based on any suitable arrays such as Affymetrix GENECHIP™, ILLUMINA™ DASL™ arrays, printed cDNA arrays, and the likes. Suitable sequencing methods comprise next generation sequencing (NGS) technologies. Suitable protein detecting methods comprises a Western blot analysis, an enzyme-linked immunosorbent assay (ELISA), immunohistochemistry analysis, an immunoprecipitation followed by an SDS-PAGE analysis, a proteomics analysis, such as a quantitative proteomics analysis. Other assays include e.g., RNase protection assay (RPA), radioimmunoassay (RIA), radioimmunodiffusion, Ouchterlony immunodiffusion, rocket Immunoelectrophoresis, tissue immunostaining, complement fixation assay, fluorescence-activated cell sorting (FACS), mass spectrometry, magnetic bead-antibody immunoprecipitation, protein chip, or any combination thereof.

In some embodiments, the marker (e.g., protein or nucleic acid) level need not be quantified; instead, the presence of the marker at any detectable level indicates that the cancer cell is amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein. In some embodiments, the level of the marker is quantified. For example, if the marker is 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 12%, 14%, 16%, 18%, 20%, 22%, 24%, 26%, 28%, 30%, 32%, 34%, 36%, 38%, 40%, 42%, 44%, 46%, 48%, 50%, 60%, 70%, 80%, 90%, 100%, 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, or more above the level of a negative control, the cancer cell is amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein. A negative control is any suitable control appropriate for use in a given assay. For example, as exemplified herein, a negative control is an isotype control antibody in an assay that uses an antibody to a marker (e.g., in flow cytometry).

If the presence of the marker in a cancer cell is detected, and/or if the marker is quantified to be at a level above that of a negative control such that the cancer cell is amenable to anti-cancer therapy using the anti-CD 137 agonist antibodies disclosed herein, the patient is administered a therapeutically effective amount of an agonist anti-CD 137 antibody, or antigen -binding fragment thereof, as disclosed herein, according to the methods disclosed herein.

Accordingly, in some aspects, the disclosure provides a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the subject has cancer cells that express major histocompatibility complex I (MHC I). The presence of MHC I cancer cells can be determined as disclosed above.

Anti-CD137 Antibodies and Antigen-binding Fragments Thereof

The present disclosure provides antibodies that specifically bind to and agonize CD 137 in a population of cancer cells that express MHC I. In some aspects, the disclosure provides anti- CD137 agonist antibodies that are useful for the treatment of cancer in subjects with cancer cells that express MHC I. In some embodiments, the anti-CDl37 agonist antibodies induce cytokine production. In some embodiments, the anti-CD 137 agonist antibodies increase the number of CD8+ T cells in the tumor microenvironment. In some embodiments, the anti-CDl37 agonist antibodies induce protective anti-tumor immunity. The disclosure also provides anti-CD 137 agonist antibodies that, upon administration in vivo , do not substantially increase intrasplenic or intrahepatic CD4+ and/or CD8+ T cell populations.

Human CD137 is a 255 amino acid transmembrane polypeptide (SEQ ID NO: 3; Accession No. NM_001561; NP_00l552) and a member of the phylogenetically-conserved tumor necrosis factor receptor (TNFR) superfamily. CD137 (alternatively 4-1BB, TNFR superfamily 9) and its ligand (CD137L) are involved in the regulation of a wide range of immune activities. CD137 ligand cross-links its receptor, CD137, which is expressed on activated T cells, and co-stimulates T cell activities. CD137 is an activation-induced co- stimulatory molecule. Recent studies have revealed that CDl37-mediated anti-cancer effects are largely based on its ability to activate T cells, in particular, to induce a cytotoxic T lymphocyte (CTL) response, and induce cytokine production, in particular, high amounts of IFNy (Ye et ah, (2014) Clin Cancer Res 20(l):44-55). CD 137 ligand is a transmembrane protein on the cell surface and transmit signals into the cells on which it is expressed, a phenomenon referred to as“reverse signaling” or“back signaling”). CD 137 ligand expression is found on most types of leukocytes and on some nonimmune cells. In monocytic cells (monocytes, macrophages, and DCs), CD137 ligand signaling induces activation, migration, survival, and differentiation.

Accordingly, in some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to and agonizes CD 137 and allows or promotes CD137L binding. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to and agonizes CD137. In some embodiments, the anti-CDl37 antibodies provided by the disclosure bind to and agonize CD137 and co-stimulate activation of T cells.

In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen-binding fragment thereof, described herein, has one or more of the following properties or characteristics: a) specifically binds to human CD 137;

b) binds to human and cynomolgus CD 137; and

c) binds to human and mouse CD 137.

In some embodiments, an anti-CD 137 agonist antibody, or antigen-binding fragment thereof, described herein, binds to CD137 and co-stimulates T cell activities. In some embodiments, an anti-CD 137 agonist antibody, or antigen-binding fragment thereof, described herein, binds to CD 137 and induces or enhances T cell activation, a cytotoxic T lymphocyte (CTL) response, T cell proliferation, cytokine production, or a combination thereof. In some embodiments, an anti-CD 137 agonist antibody, or antigen-binding fragment thereof, described herein, binds to CD 137 and induces or enhances T cell activation, a cytotoxic T lymphocyte (CTL) response, T cell proliferation, cytokine production, or a combination thereof, in a tumor microenvironment. In some embodiments, an anti-CDl37 antibody, or antigen-binding fragment thereof, described herein, does not significantly induce or enhance intrahepatic and/or intrasplenic T cell activation and/or T cell proliferation. In some embodiments, an anti-CD 137 antibody, described herein, binds to CD137 and induces the production of IFN Y . In some embodiments, the antibodies provided by the disclosure bind to CD 137 and induce the production of IL-2, TNF- Oi , IL-13, or a combination thereof.

In some embodiments, the anti-CD 137 antibodies described herein specifically bind to and agonize CD137. In some embodiments, agonism of CD137 is measured by determining the concentration of cytokines produced by immune cells. Methods for analyzing cytokine production are known in the art and utilized in the Examples. In some embodiments, an increase in cytokine production by immune cells indicates CD 137 agonism. In some embodiments, agonism of CD 137 is measured by analyzing T cell proliferation. In some embodiments, an increase in T cell proliferation indicates CD137 agonism. In some embodiments, agonism of CD137 is measured by measuring the level of cell signaling either through quantitation of phosphorylation of relevant molecules or expression of a gene reporter after a relevant promoter. In some embodiments, an increase in cell signaling indicates CD 137 agonism. In some embodiments, agonism of CD 137 is measured by measuring the volume of a tumor. In some embodiments, a decrease in the volume of a tumor indicates CD 137 agonism.

In some embodiments, the anti-CD 137 antibodies described herein induce, increase or stabilize oligomerization, multimerization, or other higher order clustering of CD 137. In some embodiments, the clustering of CD 137 on the cell surface is observed via fluorescence microscopy.

Provided herein are isolated monoclonal antibodies or antigen binding fragments thereof, that bind to and agonize CD 137. In some embodiments, the antibodies or antigen binding fragments thereof, (i) bind human CD137 with an affinity (KD) of about 40-l00nM (e.g., between about 40 nM and about 100 nM); (ii) bind an epitope on human CD137 described herein; and/or (iii) comprise a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126). In some embodiments, the antibodies or antigen binding fragments thereof, (i) bind human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM); (ii) bind an epitope on human CD137 described herein; and/or (iii) comprise a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126).

Affinity for CD137

In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM or between about 40 nM and about 100 nM). In some embodiments, the affinity of the anti-CDl37 antibody to human CD137 is at least two (e.g., at least three, four, five, six, seven, eight, nine, or 10) fold higher than the affinity of mAblO for mouse CD 137. In some embodiments, the affinity of the anti-CD 137 antibody is no greater than 500, 450, 400, 350, 300, 250, 200, 250, 200, 175, 150, 125, 110, or 100 nM. In some embodiments, the affinity of the anti-CD 137 antibody to human CD 137 is at least two (e.g., at least three, four, five, six, seven, eight, nine, or 10) fold higher than the affinity of mAblO for mouse CD137, but no greater than 500, 450, 400, 350, 300, 250, 200, 250, 200, 175, 150, 125, 110, or 100 nM. The affinity of the antibody is the strength of binding to a single CD 137 polypeptide. In some embodiments, affinity is indicated by the equilibrium dissociation constant (KD). The value of KD is inversely related to the binding affinity of an antibody to an antigen. Accordingly, the smaller the KD value, the greater the affinity of the antibody for its antigen.

Methods for determining the affinity of an antibody for its antigen are known in the art. An exemplary method for determining binding affinity employs surface plasmon resonance. Surface plasmon resonance is an optical phenomenon that allows for the analysis of realtime biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin. 51: 19- 26; Jonsson, U., i (1991) Biotechniques 11 :620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8: 125-131; and Johnsson, B., et al. (1991) Anal. Biochem. 198:268-277.

In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM). In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 40-100 nM (e.g., between about 40 nM and about 100 nM). In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 30- 40 nM, 40-50 nM, 50-60 nM, 60-70 nM, 70-80 nM, 80-90 nM, 90-100 nM, 45-55 nM, 55-65 nM, 75-85 nM, 85-95 nM, 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55-75 nM, 40-70 nM, 50-80 nM, or 60-90 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 60-80 nM. In some embodiments, the anti- CD137 antibodies described herein bind human CD137 with an affinity (KD) of about 60-75 nM. Iln some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 60-90 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD137 with an affinity (KD) of about 50-80 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 40-70 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 55-75 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 65-75 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 60-80 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 55-85 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 50-90 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 45-95 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 85-95 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 75-85 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 75-85 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 55-65 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 45-55 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 80-90 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 70-80 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 60-70 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 50-60 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 40-50 nM. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 30-40 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 30 nM, about 31 nM, about 32 nM, about 33 nM, about 34 nM, about 35 nM, about 36 nM, about 37 nM, about 38 nM, about 39 nM, about 40 nM, about 41 nM, about 42 nM, about 43 nM, about 44 nM, about 45 nM, about 46 nM, about 47 nM, about 48 nM, about 49 nM, about 50 nM, about 51 nM, about 52 nM, about 53 nM, about 54 nM, about 55 nM, about 56 nM, about

57 nM, about 58 nM, about 59 nM, about 60 nM, about 61 nM, about 62 nM, about 63 nM, about

64 nM, about 65 nM, about 66 nM, about 67 nM, about 68 nM, about 69 nM, about 70 nM, about

71 nM, about 72 nM, about 73 nM, about 74 nM, about 75 nM, about 76 nM, about 77 nM, about

78 nM, about 79 nM, about 80 nM, about 81 nM, about 82 nM, about 83 nM, about 84 nM, about

85 nM, about 86 nM, about 87 nM, about 88 nM, about 89 nM, about 90 nM, about 91 nM, about

92 nM, about 93 nM, about 94 nM, about 95 nM, about 96 nM, about 97 nM, about 98 nM, about

99 nM, about 100 nM, about 101 nM, about 102 nM, about 103 nM, about 104 nM, about 105 nM, about 106 nM, about 107 nM, about 108 nM, about 109 nM or about 110 nM.

In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of at least 30 nM but less than about 110 nM, at least 31 nM but less than about 109 nM, at least 32 nM but less than about 108 nM, at least 33 nM but less than about 107 nM, at least 34 nM but less than about 106 nM, at least 35 nM but less than about 105 nM, at least 36 nM but less than about 104 nM, at least 37 nM but less than about 103 nM at least 38 nM but less than about 102 nM, at least 39 nM but less than about 101 nM, at least 40 nM but less than about 100 nM; at least 41 nM but less than about 99 nM; least 42 nM but less than about 98 nM; least 43 nM but less than about 97 nM; at least 44 nM but less than about 96 nM; at least 45 nM but less than about 95 nM; at least 46 nM but less than about 94 nM; at least 47 nM but less than about 93 nM; at least 48 nM but less than about 92 nM; at least 49 nM but less than about 91 nM; at least 50 nM but less than about 90 nM; at least 51 nM but less than about 89 nM; at least 52 nM but less than about 88 nM; at least 53 nM but less than about 87 nM; at least 54 nM but less than about 86 nM; at least 55 nM but less than about 85 nM; at least 56 nM but less than about 84 nM; at least 57 nM but less than about 83 nM; at least 58 nM but less than about 82 nM; at least 59 nM but less than about 81 nM; at least 60 nM but less than about 80 nM; at least 61 nM but less than about 79 nM; at least 62 nM but less than about 78 nM; at least 63 nM but less than about 77 nM; at least 64 nM but less than about 76 nM; or at least 65 nM but less than about 75 nM. In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of at least 40 nM but less than about 100 nM.

In some embodiments, the anti-CDl37 antibodies described herein cross-react with CD137 polypeptides from more than one species. In some embodiments, the anti-CDl37 antibodies described herein bind cynomolgus CD137 and human CD137. In some embodiments, the anti- CD 137 antibodies described herein bind mouse CD 137 and human CD 137. In some embodiments, the anti-CDl37 antibodies described herein bind human CD137, mouse CD137 and cynomolgus CD137.

CD137 Epitope Binding

In some embodiments, the isolated monoclonal antibody, or antigen binding portion thereof, that specifically binds to human CD137, binds to an epitope on human CD137 comprising one or more (e.g., one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or all 25) of amino acids 111-132 of SEQ ID NO:3. In some embodiments, the isolated monoclonal antibody, or antigen binding portion thereof, that specifically binds to human CD137, binds to an epitope within amino acids 111-132 of SEQ ID NO:3. In some aspects, the disclosure provides an isolated monoclonal antibody, or antigen binding portion thereof, that specifically binds to human CD137, binds to all or a portion of amino acids 111-132 of SEQ ID NO:3. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 comprising residue Kl 14 of SEQ ID NO: 3. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD137 comprising residues El 11, T113 and K114 of SEQ ID NO: 3. In some embodiments, an isolated anti-CD 137 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD137 comprising residues El l l, T113, K114, N126 and 1132 of SEQ ID NO: 3. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 comprising El l l, Tl l3, K114, N126, 1132 and P135 of SEQ ID NO: 3. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD137 comprising one or more residues El l l, T113, K114, N126, 1132 and P135 of SEQ ID NO: 3.

In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 comprising a sequence of one or more amino acid residues corresponding to amino acid positions 100 to 135, 101 to 135, 102 to 135, 103 to 135, 104 to 135, 105 to 135, 106 to 135, 107 to 135, 108 to 135, 109 to 135, 110 to 135, or 111 to 135 of SEQ ID NO: 3. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3. In some embodiments, the epitope comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 within amino acid positions 100 to 135, 101 to 135, 102 to 135, 103 to 135, 104 to 135, 105 to 135, 106 to 135, 107 to 135, 108 to 135, 109 to 135, 110 to 135, or 111 to 135 of SEQ ID NO: 3. In some embodiments, an isolated anti-CD 137 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD137 within amino acid positions 111 to 135 of SEQ ID NO: 3. In some embodiments, the epitope comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3). In some embodiments, amino acid residue Ll 12 can be another amino acid residue.

In some embodiments, the epitope is a non-linear epitope. In some embodiments, mutation of amino acid residue K114 abrogates bindings of an isolated anti-CDl37 agonist antibody, or antigen binding fragment thereof, described herein, to human CD 137.

In some embodiments, isolated anti-CD 137 agonist antibody, or antigen binding fragment thereof, described herein, binds to an epitope of human CD 137 comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3, wherein the epitope comprises at least amino acid K114, and wherein the antibody or antigen binding portion thereof binds mouse CD 137 and does not bind rat CD 137. In some embodiments, the epitope is a non-linear epitope. In some embodiments, the antibody or antigen binding portion thereof binds mouse CD 137 and cynomolgus CD 137 and does not bind rat CD 137. In some embodiments, binding of an isolated anti-CD 137 agonist antibody, or antigen binding fragment thereof, described herein, to human, mouse, rat and cynomolgus CD 137 is determined by surface plasmon resonance (SPR). In some embodiments, the antibody or antigen binding portion thereof binds to mouse, cynomolgus or human CD137 with an affinity that is at least 10, 20, 30, 40, 50, 100, 200, 500 or 1000 times greater than the antibody’s affinity for rat CD137. In some embodiments, the antibody or antigen binding portion thereof binds to mouse, cynomolgus or human CD 137 with an affinity that is at least 10, 20, 30, 40, 50, 100, 200, 500 or 1000 times greater than the antibody’s affinity for a CD137 polypeptide that does not comprise a lysine at position 114 relative to human CD137 of SEQ ID NO: 3.

In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen-binding fragment thereof, described herein, binds to an epitope of human CD 137 and competes with mAbl for binding to the epitope of human CD137. In some embodiments, an isolated anti-CDl37 agonist antibody, or antigen-binding fragment thereof, described herein, binds to and agonizes CD 137. In some embodiments, the anti-CD 137 antibodies provided by the disclosure bind to and agonize CD137 and co-stimulate activation of T cells.

The present disclosure provides antibodies that compete for binding to an epitope on CD 137 which comprises all or a portion of an epitope recognized by one or more particular reference antibodies described herein (e.g., mAbl). In some embodiments, the anti-CDl37 antibodies bind to an epitope of human CD 137 and compete with a reference antibody (e.g., mAbl) for binding to the epitope of human CD 137 and wherein the antibody, or antigen binding fragment thereof, binds human CD137 with an equilibrium dissociation constant KD of 1 X 10 6 or less. In some embodiments, the anti-CDl37 antibodies bind to an epitope on CD137, wherein one or more mutations to the epitope inhibit, reduce, or block binding to both the antibodies and a reference antibody (e.g., mAbl). In some embodiments, the reference antibody is the mAbl antibody, described herein. In some embodiments, the reference antibody is any one antibody provided in any one of Tables 5-8.

Accordingly, the anti-CDl37 antibodies provided by the disclosure may be assessed through x-ray crystallographic analysis of a crystal structure comprising an antibody bound to CD137, or a fragment or portion thereof. In some aspects, the epitopes that bound by the antibodies provided by the disclosure are identified by determining the residues on the human CD 137 antigen that reside or are located within 4 angstroms (A) of an antibody paratope residue, e.g., mAbl.

In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is at least 3 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is at least 4 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is at least 5 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is at least 6 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is at least 7 amino acid residues. In some embodiments, the epitope bound by the anti- CD137 antibodies described herein is at least 8 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is at least 9 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is at least 10 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is at least 12 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is at least 3 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is at least 13 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is at least 14 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is at least 15 amino acid residues.

In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 25 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 24 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 23 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 22 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 21 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 20 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 19 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 18 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 17 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 16 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 15 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 14 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 13 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 12 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 11 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 10 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 9 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 8 amino acid residues. In some embodiments, the epitope bound by the anti-CD 137 antibodies described herein is fewer than 7 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 6 amino acid residues. In some embodiments, the epitope bound by the anti-CDl37 antibodies described herein is fewer than 5 amino acid residues.

In some embodiments, the anti-CD 137 antibodies described herein bind to an epitope of fewer than 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6 or 5 amino acids and comprises amino acid residue Kl 14 of SEQ ID NO: 3.

Variable Resigns

In some embodiments, provided herein are isolated monoclonal antibodies or antigen binding fragments thereof, comprising heavy and light chain variable sequences as set forth in any one of Tables 5-8.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69,

78 and 89, respectively;

(b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 70,

79 and 90, respectively;

(c) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 71,

80 and 91, respectively; (d) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 72, and 92, respectively;

(e) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 73, and 91, respectively;

(f) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 74, and 93, respectively;

(g) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 75, and 91, respectively;

(h) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 74, and 94, respectively;

(i) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 76, and 95, respectively;

(j) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 77, and 93, respectively;

(k) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 90, respectively;

(l) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 57 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(m) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 58 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively; (n) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 59 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(o) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 49, 60 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(p) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 61 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(q) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 58 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(r) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 62 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(s) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 52, 63 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(t) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 64 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(u) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 50, 65 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(v) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively;

(w) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 107, 56 and, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, and 89, respectively; (x) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 109, 110 and 92, respectively;

(y) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(z) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(aa) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(bb) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(cc) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(dd) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(ee) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(ff) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(gg) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively; (hh) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(ii) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively;

(jj) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively;

(kk) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively;

(11) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively; and

(mm) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 101 and 103; and wherein the light chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46 and 105.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain CDRs, wherein heavy chain CDR3 comprises the amino acid sequence set forth in SEQ ID NO: 68.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively; (b) SEQ ID NO: 4 and 28, respectively;

(c) SEQ ID NO: 4 and 30, respectively;

(d) SEQ ID NO: 4 and 32, respectively;

(e) SEQ ID NO: 4 and 34, respectively;

(f) SEQ ID NO: 4 and 36, respectively;

(g) SEQ ID NO: 4 and 38, respectively;

(h) SEQ ID NO: 4 and 40, respectively;

(i) SEQ ID NO: 4 and 42, respectively;

(j) SEQ ID NO: 4 and 44, respectively;

(k) SEQ ID NO: 4 and 46, respectively;

(l) SEQ ID NO: 8 and 6, respectively;

(m) SEQ ID NO: 10 and 6, respectively;

(n) SEQ ID NO: 12 and 6, respectively;

(o) SEQ ID NO: 14 and 6, respectively;

(p) SEQ ID NO: 16 and 6, respectively;

(q) SEQ ID NO: 18 and 6, respectively;

(r) SEQ ID NO: 20 and 6, respectively;

(s) SEQ ID NO: 22 and 6, respectively;

(t) SEQ ID NO: 24 and 6, respectively;

(u) SEQ ID NO: 26 and 6, respectively;

(v) SEQ ID NO: 101 and 6, respectively;

(w) SEQ ID NO: 103 and 6, respectively; and

(x) SEQ ID NO: 4 and 105, respectively.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 101 and 103; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46 and 105. In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NO: 4 and 6, respectively;

(b) SEQ ID NO: 4 and 28, respectively;

(c) SEQ ID NO: 4 and 30, respectively;

(d) SEQ ID NO: 4 and 32, respectively;

(e) SEQ ID NO: 4 and 34, respectively;

(f) SEQ ID NO: 4 and 36, respectively;

(g) SEQ ID NO: 4 and 38, respectively;

(h) SEQ ID NO: 4 and 40, respectively;

(i) SEQ ID NO: 4 and 42, respectively;

(j) SEQ ID NO: 4 and 44, respectively;

(k) SEQ ID NO: 4 and 46, respectively;

(l) SEQ ID NO: 8 and 6, respectively;

(m) SEQ ID NO: 10 and 6, respectively;

(n) SEQ ID NO: 12 and 6, respectively;

(o) SEQ ID NO: 14 and 6, respectively;

(p) SEQ ID NO: 16 and 6, respectively;

(q) SEQ ID NO: 18 and 6, respectively;

(r) SEQ ID NO: 20 and 6, respectively;

(s) SEQ ID NO: 22 and 6, respectively;

(t) SEQ ID NO: 24 and 6, respectively;

(u) SEQ ID NO: 26 and 6, respectively;

(v) SEQ ID NO: 101 and 6, respectively;

(w) SEQ ID NO: 103 and 6, respectively; and

(x) SEQ ID NO: 4 and 105, respectively.

In some embodiments, provided herein are antibodies that specifically bind human CD 137 comprising heavy chain and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NO: 5 and 7, respectively; (b) SEQ ID NO: 5 and 29, respectively;

(c) SEQ ID NO: 5 and 31, respectively;

(d) SEQ ID NO: 5 and 33, respectively;

(e) SEQ ID NO: 5 and 35, respectively;

(f) SEQ ID NO: 5 and 37, respectively;

(g) SEQ ID NO: 5 and 39, respectively;

(h) SEQ ID NO: 5 and 41, respectively;

(i) SEQ ID NO: 5 and 43, respectively;

(j) SEQ ID NO: 5 and 45, respectively;

(k) SEQ ID NO: 5 and 47, respectively;

(l) SEQ ID NO: 9 and 7, respectively;

(m) SEQ ID NO: 11 and 7, respectively;

(n) SEQ ID NO: 13 and 7, respectively;

(o) SEQ ID NO: 15 and 7, respectively;

(p) SEQ ID NO: 17 and 7, respectively;

(q) SEQ ID NO: 19 and 7, respectively;

(r) SEQ ID NO: 21 and 7, respectively;

(s) SEQ ID NO: 23 and 7, respectively;

(t) SEQ ID NO: 25 and 7, respectively;

(u) SEQ ID NO: 27 and 7, respectively;

(v) SEQ ID NO: 102 and 7, respectively;

(w) SEQ ID NO: 104 and 7, respectively; and

(x) SEQ ID NO: 5 and 106, respectively.

In some embodiments, provided herein are antibodies that specifically bind human CD 137 comprising heavy chain and light chain variable regions encoded by nucleotide sequences having at least 90% identity to the nucleotide sequences selected from the group consisting of:

(a) SEQ ID NO: 5 and 7, respectively;

(b) SEQ ID NO: 5 and 29, respectively;

(c) SEQ ID NO: 5 and 31, respectively;

(d) SEQ ID NO: 5 and 33, respectively;

(e) SEQ ID NO: 5 and 35, respectively; (f) SEQ ID NO: 5 and 37, respectively;

(g) SEQ ID NO: 5 and 39, respectively;

(h) SEQ ID NO: 5 and 41, respectively;

(i) SEQ ID NO: 5 and 43, respectively;

(j) SEQ ID NO: 5 and 45, respectively;

(k) SEQ ID NO: 5 and 47, respectively;

(l) SEQ ID NO: 9 and 7, respectively;

(m) SEQ ID NO: 11 and 7, respectively;

(n) SEQ ID NO: 13 and 7, respectively;

(o) SEQ ID NO: 15 and 7, respectively;

(p) SEQ ID NO: 17 and 7, respectively;

(q) SEQ ID NO: 19 and 7, respectively;

(r) SEQ ID NO: 21 and 7, respectively;

(s) SEQ ID NO: 23 and 7, respectively;

(t) SEQ ID NO: 25 and 7, respectively;

(u) SEQ ID NO: 27 and 7, respectively;

(v) SEQ ID NO: 102 and 7, respectively;

(w) SEQ ID NO: 104 and 7, respectively; and

(x) SEQ ID NO: 5 and 106, respectively.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain variable regions, wherein the heavy chain variable region is encoded by a nucleotide sequence selected from the group consisting of SEQ ID NOs: 5, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 102 and 104; and wherein the light chain variable region is encoded by a nucleotide sequence selected from the group consisting of SEQ ID NOs: 7, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47 and 106.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chain variable regions, wherein the heavy chain variable region is encoded by a nucleotide sequence having at least 90% identity to a nucleotide sequence selected from the group consisting of SEQ ID NOs: 5, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 102 and 104; and wherein the light chain variable region is encoded by a nucleotide sequence having at least 90% identity to a nucleotide sequence selected from the group consisting of SEQ ID NOs: 7, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47 and 106.

In some embodiments, provided herein are anti-CD 137 antibodies that specifically bind to human CD 137 and comprise a heavy chain CDR3 having the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid. In some embodiments, X is any amino acid except for alanine. In some embodiments, mutation of residues D95, L100, Y 100E, Y 100G, and/or Y 100H of SEQ ID NO: 126, results in loss of binding to human CD137.

In some embodiments, provided herein are anti-CD 137 antibodies that specifically bind to human CD 137 and comprise a heavy chain CDR3 having the amino acid sequence DXPFXLDXXYYYYYX (SEQ ID NO: 127), wherein X is any amino acid. In some embodiments, mutation of residues F98, D100A, Y100D, and/or Y100F, and/or Y100H of SEQ ID NO: 126, to alanine results in loss of binding to human CD137. In some embodiments, mutation of residues F98, D100A, Y 100D, and/or Y 100F, and/or Y 100H of SEQ ID NO: 126, to any residue except for alanine, results in an increase in binding to human CD 137.

In some embodiments, provided herein are anti-CD 137 antibodies that specifically bind to human CD 137 and comprise a heavy chain CDR3 having the amino acid sequence DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid. In some embodiments, X 2 is proline, wherein X 3 is phenylalanine or tryptophan, wherein X5 is aspartic acid or glutamic acid, wherein X 8 is tyrosine, and wherein X9 is tyrosine.

The role of an amino acid residue within the heavy chain CDR3 of an antibody or antigen binding portion thereof, in binding to a specified target (e.g., CD137) can be determined by methods known to one of skill in the art. In some embodiments, an initial analysis using alanine scanning is completed to determine the critical residues for antigen binding. As described herein, alanine scanning is a technique used to determine the contribution of a specific wild-type residue to the stability or function(s) (e.g., binding affinity) of given protein or polypeptide. The technique involves the substitution of an alanine residue for a wild-type residue in a polypeptide, followed by an assessment of the stability or function(s) (e.g., binding affinity) of the alanine-substituted derivative or mutant polypeptide and comparison to the wild-type polypeptide. In some embodiments, the residues identified as not critical are further evaluated to modulate the binding of the antibody to the antigen (e.g., increase or decrease binding). A non-limiting example of such analysis is deep mutational scanning. This method allows for the evaluation of large numbers of mutations. In some embodiments, each amino acid residue within the heavy chain CDR3 is mutated to every amino acid residue (except for alanine), and binding is assessed. Other methods for analyzing the effect of amino acid residue mutations are known in the art. In some embodiments, these methods are utilized to assess the role of residues in all of the heavy chain and light chain CDRs in binding to human CD 137.

Exemplary CD137 Binding Antibodies

In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM). In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of about 40-100 nM (e.g., between about 40 nM and about 100 nM). In some embodiments, the anti-CD 137 antibodies described herein bind an epitope on human CD 137 described supra (e.g., comprising K114). In some embodiments, the anti-CDl37 antibodies described herein comprise a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126). In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an affinity (KD) of 30-100 nM (e.g., between about 30 nM and about 100 nM) and bind an epitope on human CD137 described supra (e.g., comprising K114). In some embodiments, the anti-CD 137 antibodies described herein bind human CD 137 with an affinity (KD) of 30-100 nM (e.g., between about 30 nM and about 100 nM) and comprise a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126). In some embodiments, the anti-CDl37 antibodies described herein bind an epitope on human CD137 described supra (e.g., comprising Kl 14) and comprise a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126). In some embodiments, the anti- CD137 antibodies described herein bind human CD137 with an affinity (KD) of 30-l00nM (e.g., between about 30 nM and about 100 nM), bind an epitope on human CD137 described supra (e.g., comprising K114), and comprise a heavy chain CDR3 comprising the amino acid sequence DXXXXLXXXXYXYYX (SEQ ID NO: 126). In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM); and

(ii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM); and

(ii) bind to an epitope on human CD 137 comprising one or more residues El l l, Tl l3, K114, N126, 1132 and P135 of SEQ ID NO: 3.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope on human CD 137 comprising one or more residues El l l, Tl l3, K114, N126, 1132 and P135 of SEQ ID NO: 3;

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid; or

(iv) combinations thereof.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope on human CD 137 comprising one or more residues El l l, Tl l3, K114, N126, 1132 and P135 of SEQ ID NO: 3;

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X2 is a non-polar amino acid, wherein X3 is a non-polar amino acid, wherein X4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid.; or

(iv) combinations thereof.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) specifically bind to an epitope on human CD 137 comprising one or more residues El 11, T113, K114, N126, 1132 and P135 of SEQ ID NO: 3; and

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope on human CD 137 comprising one or more residues El l l, Tl l3, K114, N126, 1132 and P135 of SEQ ID NO: 3; and

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM); and

(ii) bind to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3; (iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid; or

(iv) combinations thereof.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3;

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid; or

(iv) combinations thereof.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3; and

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising a sequence of one or more amino acid residues corresponding to amino acid positions 111 to 135 of SEQ ID NO: 3; and

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X2 is a non-polar amino acid, wherein X3 is a non-polar amino acid, wherein X4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity of about 30-100 nM (e.g., between about 30 nM and about 100 nM); and

(ii) bind to an epitope comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3).

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3);

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid; or

(iv) combinations thereof.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3);

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid; or

(iv) combinations thereof.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM); (ii) bind to an epitope comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3); and

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DXXXXLXXXXYXYYX (SEQ ID NO: 126), wherein X is any amino acid.

In some embodiments, the anti-CD 137 antibodies

(i) bind human CD137 with an affinity (K D ) of about 30-100 nM (e.g., between about 30 nM and about 100 nM);

(ii) bind to an epitope comprising ELTK (corresponding to amino acid residues 111-114 of SEQ ID NO: 3); and

(iii) comprise a heavy chain CDR3 comprising the amino acid sequence

DX1X2X3X4LX5X6X7X8YX9YYX10 (SEQ ID NO: 128), wherein Xi is any amino acid, wherein X 2 is a non-polar amino acid, wherein X 3 is a non-polar amino acid, wherein X 4 is any amino acid, wherein X5 is a polar amino acid, wherein X 6 is any amino acid, wherein X 7 is any amino acid, wherein X 8 is a polar amino acid, wherein X9 is a polar amino acid, and wherein X10 is any amino acid.

In some embodiments, the anti-CDl37 antibodies described supra comprise heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 48, 56 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively; and

(b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 51, 108 and 68, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 69, 78 and 89, respectively.

In some embodiments, the anti-CDl37 antibodies described supra comprise heavy and light chain CDRs selected from the group consisting of:

(a) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143, and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151, and 154, respectively; and

(b) heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145, and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151, and 154, respectively. In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence of SEQ ID NO: 6.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 4 and 6, respectively; and

(b) SEQ ID NOs: 101 and 6, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions comprising amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 4 and 6, respectively;

(b) SEQ ID NOs: 101 and 6, respectively; and

(c) SEQ ID NOs: 26 and 6, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively; and

(b) SEQ ID NOs: 102 and 7, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions encoded by nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively;

(b) SEQ ID NOs: 102 and 7, respectively; and

(c) SEQ ID NOs: 27 and 7, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4 and 101; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 6.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 26 and 101 ; and wherein the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 6.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions, wherein the heavy chain variable region is encoded by a nucleotide sequence which is least 90% identical to the nucleotide sequence selected from the group consisting of SEQ ID NOs: 5 and 102; and wherein the light chain variable region is encoded by a nucleotide sequence which is at least 90% identical to the nucleotide sequence of SEQ ID NO: 7.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions, wherein the heavy chain variable region is encoded by a nucleotide sequence which is least 90% identical to the nucleotide sequence selected from the group consisting of SEQ ID NOs: 5, 27 and 102; and wherein the light chain variable region is encoded by a nucleotide sequence which is at least 90% identical to the nucleotide sequence of SEQ ID NO: 7.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 4 and 6, respectively; and

(a) SEQ ID NOs: 101 and 6, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions comprising amino acid sequences at least 90% identical to the amino acid sequences selected from the group consisting of:

(a) SEQ ID NOs: 4 and 6, respectively;

(b) SEQ ID NOs: 101 and 6, respectively; and

(c) SEQ ID NOs: 26 and 6, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions encoded by nucleotide sequences at least 90% identical to the nucleotide sequences selected from the group consisting of:

(a) SEQ ID NOs: 5 and 7, respectively; and

(b) SEQ ID NOs: 102 and 7, respectively.

In some embodiments, the anti-CD 137 antibodies comprise heavy and light chain variable regions encoded by nucleotide sequences at least 90% identical to the nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 5 and 7, respectively;

(b) SEQ ID NOs: 102 and 7, respectively; and

(c) SEQ ID NOs: 27 and 7, respectively.

In some embodiments, the anti-CDl37 antibodies described herein have at least the functional properties of mAbl (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 4 and 6, respectively), mab8 (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 101 and 6, respectively) or mAblO (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 26 and 6, respectively). In some embodiments, the functional properties of an antibody described herein include but are not limited to: induction or enhancement of dimerization of CD137; induction or enhancement of multimerization of CD137; induction or enhancement of CDl37-mediated T cell activation; induction or enhancement of CDl37-mediated cytotoxic T cell response; induction or enhancement of CDl37-mediated T cell proliferation; induction or enhancement of CD137- mediated cytokine production; lack of induction or enhancement of intrahepatic and/or intrasplenic T cell activation and/or T cell proliferation; and reduction or inhibition of tumor growth.

In some embodiments, the anti-CDl37 antibodies described herein bind human CD137 with an equilibrium dissociation constant KD of 1 X 10 6 or less. In some embodiments, the anti- CD137 antibodies described herein bind human CD137 with an equilibrium dissociation constant KD at least equivalent to that of mAbl (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 4 and 6, respectively), mab8 (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 101 and 6, respectively) or mAblO (i.e., an antibody comprising the heavy and light chain variable sequences of SEQ ID NOs: 26 and 6, respectively).

In some embodiments, the anti-CDl37 antibodies described herein comprise a human IgGl heavy chain constant region or a human IgG4 heavy chain constant region. In some embodiments, the anti-CD 137 antibodies described herein comprise a human wild-type IgGl heavy chain constant region or a human wild-type IgG4 heavy chain constant region. In some embodiments, the anti-CD 137 antibodies described herein comprise a human wild-type IgGl heavy chain constant region as set forth in SEQ ID NO: 1. In some embodiments, the anti-CDl37 antibodies described herein comprise a human wild-type IgG4 heavy chain constant region as set forth in SEQ ID NO: 138. In some embodiments, the anti-CDl37 antibodies described herein comprise a mutant IgGl heavy chain constant region an IgGl heavy chain constant region comprising an amino acid mutation relative to the human wild-type IgGl) or a mutant IgG4 heavy chain constant region ( i.e an IgG4 heavy chain constant region comprising an amino acid mutation relative to the human wild-type IgG4). In some embodiments, the anti-CD 137 antibodies described herein comprise a mutant IgG4 heavy chain constant region, wherein the mutant IgG4 heavy chain constant region comprises an amino acid substitution at residue Ser228 according to EU numbering. In some embodiments, the amino acid substitution at residue Ser228 is S228P. In some embodiments, the anti-CD 137 antibodies described herein comprise an IgG4 heavy chain constant region, wherein the c-terminal lysine residue is removed. In some embodiments, the anti- CD 137 antibodies described herein comprise an IgG4 heavy chain constant region wherein the c- terminal lysine residue is removed and comprises the S228P amino acid substitution. In some embodiments, the anti-CD 137 antibodies described herein comprise an IgG4 heavy chain constant region as set forth in SEQ ID NO: 2.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chains comprising the amino acid sequences set forth in SEQ ID NOs: 129 and 133, respectively. In some embodiments, the anti-CD 137 antibodies described herein comprise heavy and light chains comprising the amino acid sequences set forth in SEQ ID NOs: 130 and 133, respectively. In some embodiments, the anti-CD 137 antibodies described herein comprise heavy and light chains comprising the amino acid sequences set forth in SEQ ID NOs: 131 and 133, respectively. In some embodiments, the anti-CD 137 antibodies described herein comprise heavy and light chains comprising the amino acid sequences set forth in SEQ ID NOs: 132 and 133, respectively.

In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chains comprising amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least, 96%, at least 97%, at least 98%, or at least 99% identity to SEQ ID NOs: 129 and 133, respectively. In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chains comprising amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least, 96%, at least 97%, at least 98%, or at least 99% identity to SEQ ID NOs: 130 and 133, respectively. In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chains comprising amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least, 96%, at least 97%, at least 98%, or at least 99% identity to SEQ ID NOs: 131 and 133, respectively. In some embodiments, the anti-CDl37 antibodies described herein comprise heavy and light chains comprising amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least, 96%, at least 97%, at least 98%, or at least 99% identity to SEQ ID NOs: 132 and 133, respectively.

CDR Numbering Systems

The system described by Kabat, also referred to as“numbered according to Kabat,” "Kabat numbering", "Kabat definitions", and "Kabat labeling," provides an unambiguous residue numbering system applicable to any variable domain of an antibody, and provides precise residue boundaries defining the three CDRs of each chain. (Kabat et al., Sequences of Proteins of Immunological Interest, National Institutes of Health, Bethesda, Md. (1987) and (1991), the contents of which are incorporated by reference in their entirety. These CDRs are referred to as Kabat CDRs and comprise about residues 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3) in the light chain variable domain, and 31-35 (CDR1), 50-65 (CDR2) and 95-102 (CDR3) in the heavy chain variable domain. When the CDRs are defined according to Kabat, the light chain FR residues are positioned at about residues 1-23 (LCFR1), 35-49 (LCFR2), 57-88 (LCFR3), and 98-107 (LCFR4) and the heavy chain FR residues are positioned about at residues 1-30

(HCFR1), 36-49 (HCFR2), 66-94 (HCFR3), and 103-113 (HCFR4) in the heavy chain residues. The "EU index as in Kabat" refers to the residue numbering of the human IgGl EU antibody.

Other CDR numbering systems are also used in the art. Chothia and coworkers found that certain sub-portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. (Chothia et al. (1987) J. Mol. Biol. 196: 901-917; and Chothia et al. (1989) Nature 342: 877-883). These sub-portions were designated as Ll, L2, and L3 or Hl, H2, and H3 where the "L" and the "H" designates the light chain and the heavy chains regions, respectively. These CDRs can be referred to as“Chothia CDRs,”“Chothia numbering,” or“numbered according to Chothia,” and comprise about residues 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3) in the light chain variable domain, and 26-32 (CDR1), 52-56 (CDR2) and 95-102 (CDR3) in the heavy chain variable domain. Mol. Biol. 196:901-917 (1987).

The system described by MacCallum, also referred to as“numbered according to

MacCallum,” or“MacCallum numbering” comprises about residues 30-36 (CDR1), 46-55 (CDR2) and 89-96 (CDR3) in the light chain variable domain, and 30-35 (CDR1), 47-58 (CDR2) and 93-101 (CDR3) in the heavy chain variable domain. MacCallum et al. ((1996) J. Mol. Biol. 262(5):732-745).

The system described by AbM, also referred to as“numbering according to AbM,” or “AbM numbering" comprises about residues 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3) in the light chain variable domain, and 26-35 (CDR1), 50-58 (CDR2) and 95-102 (CDR3) in the heavy chain variable domain.

The IMGT (INTERNATIONAL IMMUNOGENETICS INFORMATION SYSTEM) numbering of variable regions can also be used, which is the numbering of the residues in an immunoglobulin variable heavy or light chain according to the methods of the IMGT, as described in Lefranc, M.-P., "The IMGT unique numbering for immunoglobulins, T cell

Receptors and Ig-like domains", The Immunologist, 7, 132-136 (1999), and is expressly incorporated herein in its entirety by reference. As used herein, "IMGT sequence numbering" or “numbered according to IMTG,” refers to numbering of the sequence encoding a variable region according to the IMGT. For the heavy chain variable domain, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable domain, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.

In some embodiments of the anti-CD 137 antibody described herein, the CDRs recited herein comprise about residues 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3) in the light chain variable domain, and 27-35 (CDR1), 49-60 (CDR2) and 93-102 (CDR3) in the heavy chain variable domain, when numbered according to Chothia numbering. In some embodiments, CDR2 in the light chain variable domain can comprise amino acids 49-56, when numbered according to Chothia numbering.

In some aspects and embodiments of the anti-CD 137 antibody described herein, the antibody or antigen-binding portion thereof comprises a heavy chain variable region comprising an amino acid sequence that is at least 90% identical ( e.g ., at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95, at least 96%, at least 97%, at least 98%, or at least 99% identical) to SEQ ID NO: 4 and/or a light chain variable region comprising an amino acid sequence that is at least 90% identical (e.g., at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical) to SEQ ID NO: 6. In some such embodiments, the heavy chain variable region comprises an amino acid sequence that differs by 15 amino acids or less, 14 amino acids or less, 13 amino acids or less, 12 amino acids or less, 11 amino acids or less, 10 amino acids or less, 9 amino acids or less, 8 amino acids or less, 7 amino acids or less, 6 amino acids or less, 5 amino acids or less, 4 amino acids or less, 3 amino acids or less, 2 amino acids or less, or 1 amino acid from SEQ ID NO: 4.

In some such embodiments, the light chain variable region comprises an amino acid sequence that differs by 15 amino acids or less, 14 amino acids or less, 13 amino acids or less, 12 amino acids or less, 11 amino acids or less, 10 amino acids or less, 9 amino acids or less, 8 amino acids or less, 7 amino acids or less, 6 amino acids or less, 5 amino acids or less, 4 amino acids or less,

3 amino acids or less, 2 amino acids or less, or 1 amino acid from SEQ ID NO: 6.

In some embodiments, the CDRs of the antibody or antigen-binding portion thereof comprise about residues 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3) in the light chain variable domain of SEQ ID NO: 6, and 27-35 (CDR1), 49-60 (CDR2) and 93-102 (CDR3) in the heavy chain variable domain of SEQ ID NO: 4, when numbered according to Chothia numbering. In some embodiments, CDR2 in the light chain variable domain SEQ ID NO: 6 can comprise amino acids 49-56, when numbered according to Chothia numbering.

The disclosure also provides, in some embodiments, an antibody or antigen-binding portion thereof that comprises heavy chain CDRs of the heavy chain variable region of SEQ ID NO: 4, and light chain CDRs of the light chain variable region of SEQ ID NO: 6, wherein the heavy and light chain CDR residues are numbered according to Rabat.

The disclosure also provides, in some embodiments, an antibody or antigen-binding portion thereof that comprises heavy chain CDRs of the heavy chain variable region of SEQ ID NO: 4, and light chain CDRs of the light chain variable region of SEQ ID NO: 6, wherein the heavy and light chain CDR residues are numbered according to Chothia.

The disclosure also provides, in some embodiments, an antibody or antigen-binding portion thereof that comprises heavy chain CDRs of the heavy chain variable regions of SEQ ID NO: 4, and light chain CDRs of the light chain variable region of SEQ ID NO: 6, wherein the heavy and light chain CDR residues are numbered according to MacCallum.

The disclosure also provides, in some embodiments, an antibody or antigen-binding portion thereof that comprises heavy chain CDRs of the heavy chain variable regions of SEQ ID NO: 4, and light chain CDRs of the light chain variable region of SEQ ID NO: 6, wherein the heavy and light chain CDR residues are numbered according to AbM. The disclosure also provides, in some embodiments, an antibody or antigen-binding portion thereof that comprises heavy chain CDRs of the heavy chain variable regions of SEQ ID NO: 4, and light chain CDRs of the light chain variable region of SEQ ID NO: 6, wherein the heavy and light chain CDR residues are numbered according to IMGT.

Characterization and Functions of CD137 Binding Antibodies

T _ Affinity

In some embodiments, an anti-CDl37 antibody described herein binds human CD137 with an affinity (KD) of about 40-100 nM (e.g., between about 40 nM and about 100 nM) as determined by an antigen-binding assay. In some embodiments, an anti-CDl37 antibody described herein binds human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) as determined by an antigen-binding assay. In some embodiments, an anti-CDl37 antibody described herein binds human CD137 with an affinity (KD) of about 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55-75 nM, 40-70 nM, 50-80 nM, or 60-90 nM as determined by an antigen-binding assay.

In some embodiments, the antigen-binding assay determines a binding affinity of the anti- CD 137 antibody for a CD 137 polypeptide. In some embodiments, the antigen binding assay is surface plasmon resonance. Accordingly, in some embodiments an anti-CD 137 antibody described herein binds human CD137 with an affinity (KD) of about 40-100 nM (e.g., between about 40 nM and about 100 nM) as determined using surface plasmon resonance. In some embodiments, an anti-CDl37 antibody described herein binds human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) as determined using surface plasmon resonance. In some embodiments, an anti-CDl37 antibody described herein binds human CD137 with an affinity (KD) of about 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55- 75 nM, 40-70 nM, 50-80 nM, or 60-90 nM as determined using surface plasmon resonance.

The phrase "surface plasmon resonance" includes an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin. 51: 19-26; Jonsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8: 125-131; and Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277. In some embodiments, the antigen binding assay is biolayer interferometry (BLI). Accordingly, in some embodiments an anti-CD 137 antibody described herein binds human CD137 with an affinity (KD) of about 40-100 nM (e.g., between about 40 nM and about 100 nM) as determined using biolayer interferometry. In some embodiments, an anti-CD 137 antibody described herein binds human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) as determined using biolayer interferometry. In some embodiments, an anti-CDl37 antibody described herein binds human CD137 with an affinity (KD) of about 45-95 nM, 50-90 nM, 55-85 nM, 60-80 nM, 65-75 nM, 55-75 nM, 40-70 nM, 50-80 nM, or 60-90 nM as determined using biolayer interferometry.

The phrase“biolayer interferometry” or“BLI” includes an optical phenomenon that allows for the measurement of sub-nanometer changes in the thickness of its optical layer detection surface. In some embodiments, biomolecules bind at a sensor surface and change the optical layer thickness. The magnitude of the optical layer thickness change is proportional to the mass or molecular weight of the binding molecule. In some embodiments, CD137 is immobilized to the sensor surface to measure binding by an antibody, wherein binding creates a change in the molecular weight to produce a corresponding change in the optical layer thickness. In some embodiments, BLI is performed with an OCTET system (ForteBio).

II. Immune Cell Effects

In some embodiments, an anti-CDl37 antibody described herein induces or enhances cytokine production by an immune cell as determined by a cytokine assay. In some embodiments, the cytokine assay determines an amount of at least one cytokine secreted from an immune cell contacted with the anti-CD 137 antibody, wherein an increase in the amount of the at least one cytokine indicates induction or enhancement of cytokine production by the anti-CD 137 antibody. In some embodiments, an increase in cytokine production is at least 1 fold, 2 fold, 3 fold, 4 fold or 5 fold more compared to a control antibody (e.g., an antibody that does not bind to CD 137 and does not induce cytokine production).

In some embodiments, an anti-CDl37 antibody described herein induces or enhances cytokine production by an immune cell as determined by a cytokine assay, wherein the cytokine assay comprises the following steps:

(i) contacting the immune cell with the anti-CD 137 antibody; and (ii) determining an amount of at least one cytokine produced by the immune cell, wherein an increase in the amount of the at least one cytokine indicates the anti-CD 137 antibody induces or enhances cytokine production by the immune cell.

In some embodiments, an anti-CDl37 antibody described herein induces or enhances cytokine production by an immune cell as determined by a cytokine assay, wherein the cytokine assay comprises the following steps:

(i) contacting the immune cell with an anti-CD 137 antibody; and

(ii) determining an amount of at least one cytokine produced by the immune cell, and

(iii) comparing the amount of the at least one cytokine produced by the immune cell to an amount secreted from a reference immune cell,

wherein the reference immune cell is contacted with a control antibody, and wherein an increase in the amount of the at least one cytokine produced from the immune cell relative to the reference immune cell indicates induction or enhancement of human CDl37-mediated cytokine production.

In some embodiments, an anti-CDl37 antibody described herein induces or enhances cytokine production by an immune cell as determined by a cytokine assay, wherein the cytokine assay comprises the following steps:

(i) contacting an immune cell with an anti-CD 137 antibody;

(ii) determining an amount of at least one cytokine produced by the immune cell, and

(iii) comparing the amount of the at least one cytokine produced by the immune cell to an amount or level secreted from a reference immune cell,

wherein the reference immune cell is not contacted with the anti-CD 137 antibody, and wherein an increase in the amount of the at least one cytokine produced from the immune cell relative to the reference immune cell indicates induction or enhancement of human CD137- mediated cytokine production by the immune cell.

In some embodiments, the at least one cytokine is selected from a group consisting of IL- 2, IFNy, TNFa, IL-13, and combinations thereof. In some embodiments, the cytokine is IL-2. In some embodiments, the cytokine is IFNy. In some embodiments, the cytokine is TNFa. In some embodiments, the cytokine is IL-13. In some embodiments, an anti-CDl37 antibody induces or enhances IL-2 production. In some embodiments, an anti-CDl37 antibody induces or enhances TNFa production. In some embodiments, an anti-CD 137 antibody induces or enhances IL-13 production. In some embodiments, the cytokine produced is IL-2. In some embodimentss, the cytokine produced is TNFa. In some embodiments, the cytokine produced is IL-13. In some embodiments, the cytokine produced is IFNy. In some embodiments, the cytokine produced is IL- 2 and TNFa. In some embodiments, the cytokine produced is IL-2 and IL-13. In some embodiments, the cytokine produced is IL-2 and IFNy. In some embodiments, the cytokine produced is TNFa and IL-13. In some embodiments, the cytokine produced is TNFa and IFNy. In some embodiments, the cytokine produced is IL-13 and IFNy. In some embodiments, the cytokine produced is IL-2, TNFa and IL-13. In some embodiments, the cytokine produced is IL- 2, TNFa and IFNy. In some embodiments, the cytokine produced is IFNy, TNFa and IL-13.

In some embodiments, the immune cell is a T cell. In some embodiments, the reference immune cell is a T cell. In some embodiments the T cell is a CD8+ T cell.

In some embodiments, the cytokine assay is a cytokine bead array assay. A cytokine bead array assay is a bead-based immunoassay that allows for multianalyte flow cytometric determination of multiple cytokines in a sample. The use of microspheres of different size or color is the basis of a cytokine bead array assay, wherein each microsphere (or“bead”) is coated with an antibody that specifically binds to an antigen (e.g., a cytokine). Antibody-coated beads are then introduced to a sample in combination with detector antibodies. The bead: antigen: detector antibody complexes are then analyzed by flow cytometry. Commercially available cytokine bead array assays include, but are not limited to, BD™ Cytometric Bead Array Systems (BD Biosciences) and LUMINEX® Assays (R&D Systems). In some embodiments, induction or enhancement of human CDl37-mediated cytokine production is determined by a cytokine bead array assay. In some embodiments, induction or enhancement of human CDl37-mediated cytokine production is determined by a LUMINEX® Assay.

In some embodiments, the cytokine assay is a Meso Scale Discovery (MSD) assay (Meso Scale Diagnostics; Rockville, MD). An MSD assay is a commercially available assay based on detection of electrochemiluminescent-labeled antibodies that specifically bind to an antigen (e.g., a cytokine) of interest. An MSD assay comprises high binding carbon electrodes in the bottom of microplate wells that allow for attachment of biological reagents (e.g., capture antibodies specific for a cytokine). MSD assays use electrochemiluminescent labels that are conjugated to detection antibodies. A sample is added to the microplate wells and electricity is applied to the plate electrodes by an MSD instrument leading to light emission by the electrochemiluminescent labels. Light intensity is measured to quantify analytes (e.g., cytokines) in the sample. In some embodiments, induction or enhancement of human CDl37-mediated cytokine production is determined by a Meso Scale Discovery (MSD) assay.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay. In some embodiments, the T cell activation assay determines an amount of at least one cytokine secreted from T cells contacted with an anti-CD 137 antibody described herein, wherein an increase in the amount of the at least one cytokine indicates induction or enhancement of T cell activation. In some embodiments, an increase in cytokine production is at least 1 fold, 2 fold, 3 fold, 4 fold or 5 fold more compared to a control antibody (e.g., an antibody that does not bind to CD137 and does not induce cytokine production).

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) isolating T cells from a subject;

(ii) contacting the T cells with an anti-CD 137 antibody; and

(iii) determining an amount of at least one cytokine secreted by the T cells after (ii), wherein an increase in the level of the at least one cytokine indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) isolating T cells from a subject;

(ii) contacting the T cells with an anti-CD 137 antibody;

(iii) determining an amount of at least one cytokine secreted by the T cells; and

(iv) comparing the amount of the at least one cytokines produced by the T cells to an amount or level secreted from reference T cells,

wherein the reference T cells are not contacted with the anti-CD 137 antibody, and wherein an increase in the amount of the at least one cytokine produced from the T cells relative to the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation. In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) isolating T cells from a subject;

(ii) contacting the T cells with an anti-CD 137 antibody;

(iii) determining an amount of at least one cytokine secreted by the T cells; and

(iv) comparing the amount of the at least one cytokine produced by the T cells to an amount secreted from reference T cells,

wherein the reference T cells are contacted with a control antibody, and wherein an increase in the amount of the at least one cytokine produced from the T cells relative to the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, the T cell activation assay comprises determining the level of at least one cytokine secreted by the T cells after contact with an anti-CD 137 antibody described herein, wherein the at least one cytokine is selected from the group consisting of IL-2, IFNy, TNFa and IL-13. In some embodiments, the cytokine is IL-2. In some embodiments, the cytokine is IFNy. In some embodiments, the cytokine is TNFa. In some embodiments, the cytokine is IL-13. In some embodiments, the T cell activation assay comprises a cytokine assay, such as those described herein, to determine the amount of the at least one cytokine. In some embodiments, the cytokine produced is IL-2. In some embodiments, the cytokine produced is TNFa. In some embodiments, the cytokine produced is IL-13. In some embodiments, the cytokine produced is IFNy. In some embodiments, the cytokine produced is IL-2 and TNFy. In some embodiments, the cytokine produced is IL-2 and IL-13. In some embodiments, the cytokine produced is IL-2 and IFNa. In some embodiments, the cytokine produced is TNFa and IL-13. In some embodiments, the cytokine produced is TNFa and IFNy. In some embodiments, the cytokine produced is IL-13 and IFNy. In some embodiments, the cytokine produced is IL-2, TNFa and IL-13. In some embodiments, the cytokine produced is IL-2, TNFa and IFNy. In some embodiments, the cytokine produced is IFNy, TNFa and IL-13.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises detecting surface expression of at least one activation marker on T cells, and wherein an increase in the expression level of the at least one activation marker indicates induction or enhancement of T cell activation. In some embodiments,“increase in surface expression” refers to at least a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% increase in surface expression relative to surface expression in the presence of a control antibody or in the absence of an antibody.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay in vitro , wherein the T cell activation assay comprises the following steps:

(i) isolating T cells from a subject;

(ii) contacting the T cells with an anti-CD 137 antibody; and

(iii) detecting surface expression of at least one activation marker on the T cells, wherein an increase in surface expression of at least one activation marker indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) isolating T cells from a subject;

(ii) contacting the T cells with an anti-CD 137 antibody;

(iii) determining surface expression of at least one activation marker on the T cells; and

(iv) comparing surface expression of at least one activation marker on the T cells to surface expression of the at least one activation marker on reference T cells,

wherein the reference T cells are not contacted with the anti-CD 137 antibody, and wherein an increase in surface expression of at least one activation marker on the T cells relative to the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) isolating T cells from a subject;

(ii) contacting the T cells with an anti-CD 137 antibody;

(iii) determining surface expression of at least one activation marker on the T cells,

(iv) comparing the surface expression of the at least one activation marker on the T cells to surface expression of the at least one activation marker on reference T cells, wherein the reference T cells are contacted with a control antibody, and wherein an increase in surface expression of the at least one activation marker on the T cells relative to surface expression of the at least one activation marker on the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay in vivo , wherein the T cell activation assay comprises the following steps:

(i) administering the anti-CD 137 antibody to a subject;

(ii) isolating T cells from the subject; and

(iii) detecting surface expression of at least one activation marker on the T cells, wherein an increase in surface expression of at least one activation marker indicates the anti-CDl37 antibody induces or enhances CDl37-mediated T cell activation.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) administering the anti-CD 137 antibody to a subject;

(ii) isolating T cells from the subject;

(iii) determining surface expression of at least one activation marker on the T cells after; and

(iv) comparing surface expression of the at least one activation marker on the T cells to surface expression of the at least one activation marker on reference T cells,

wherein the reference T cells are isolated from a subject not administered the anti-CD 137 antibody, and wherein an increase in surface expression of the at least one activation marker on the T cells relative to the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein induces or enhances T cell activation as determined by a T cell activation assay, wherein the T cell activation assay comprises the following steps:

(i) administering the anti-CD 137 antibody to a subject;

(ii) isolating T cells from the subject;

(iii) determining surface expression of at least one activation marker on the T cells; and (iv) comparing surface expression of the at least one activation marker on the T cells to surface expression of the at least one activation marker on reference T cells,

wherein the reference T cells are isolated from a subject contacted with a control antibody, and wherein an increase in surface expression of the at least one activation marker on the T cells relative to surface expression of the at least one activation marker on the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein does not induce or enhance intrahepatic T cell activation as determined by a T cell activation assay in vivo , wherein the T cell activation assay comprises the following steps:

(i) administering the anti-CD 137 to a subject;

(ii) isolating T cells from the liver of the subject;

(iii) detecting surface expression of at least one activation marker on the T cells; and

(iv) comparing the surface expression of the at least one activation marker on the T cells to surface expression of the at least one activation marker on reference T cells,

wherein the reference T cells are isolated from a subject not administered the anti-CD 137 antibody, optionally, wherein the reference T cells are isolated from a subject administered a control antibody, and wherein an absence of an increase in surface expression of the at least one activation marker on the T cells relative to surface expression of the at least one activation marker on the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments, an anti-CD 137 antibody described herein does not induce or enhance intrasplenic T cell activation as determined by a T cell activation assay in vivo, wherein the T cell activation assay comprises the following steps:

(i) administering the anti-CD 137 to a subject;

(ii) isolating T cells from the spleen of the subject;

(iii) detecting surface expression of at least one activation marker on the T cells; and

(iv) comparing surface expression of the at least one activation marker on the T cells to surface expression of the at least one activation marker on reference T cells,

wherein the reference T cells are isolated from a subject not administered the anti-CD 137 antibody, optionally, wherein the reference T cells are isolated from a subject administered a control antibody, and wherein an absence in an increase in surface expression of the at least one activation marker on the T cells relative to surface expression of the at least one activation marker on the reference T cells indicates the anti-CD 137 antibody induces or enhances T cell activation.

In some embodiments“does not induce or enhance” is intended to refer to the absence of an activity (e.g., T cell activation) or a lack of increase of an activity relative to an increase by a reference antibody.

In some embodiments, a surface expression of a T cell activation marker is equivalent to the surface expression in the absence of an antibody. In some embodiments a surface expression of a T cell activation marker is less than the surface expression in the presence of a reference antibody that induces or enhance surface expression at least 1 fold, 5 fold, 10 fold, 50 fold, or 100 fold higher compared to surface expression in the absence of an antibody.

In some embodiments, the at least one activation marker is selected from the group consisting of CD25, CD69 and CD40L. In some embodiments, the one or more activation markers is CD25.

In some embodiments, T cells are isolated from a subject having a tumor. In some embodiments, the T cells are isolated from the tumor. In some embodiments, the control antibody is an isotype control antibody.

In some embodiments, an anti-CDl37 antibody described herein induces or enhances infiltration of one or more immune cells into a tumor microenvironment as determined by an immune cell infiltration assay. In some embodiments, an anti-CD 137 antibody described herein decreases infiltration of one or more immune cells into a tumor microenvironment as determined by an immune cell infiltration assay.

In some embodiments, the immune cell infiltration assay determines a quantity of immune cells expressing one or more immune cell markers in a tumor. In some embodiments, the one or more immune cell markers is labeled with an antibody. In some embodiments, the one or more immune cell markers is selected from the group consisting of CD45, CD25, FOXP3, CD4, CD8, F4/80, CDl lb, TIGIT and PD-l. In some embodiments, the quantity of immune cells expressing the one or more immune cell markers in a tumor is determined by flow cytometry. Methods of quantifying immune cells expressing one or more immune cell markers by flow cytometry are known in the art.

In some embodiments, the anti-CD 137 antibody induces or enhances infiltration of one or more immune cells into a tumor microenvironment relative to a reference antibody, as determined by an immune cell infiltration assay. In some embodiments, the reference antibody is an antibody comprising the same isotype as the anti-CD 137 antibody and does not specifically bind to CD 137. In some embodiments, the reference antibody is an antibody comprising the same isotype as the anti-CD 137 antibody and specifically binds to CD 137. In some embodiments, the reference antibody is an antibody comprising the different isotype as the anti-CD 137 antibody and does not specifically bind to CD137. In some embodiments, the reference antibody is an antibody comprising a different isotype as the anti-CD 137 antibody and specifically binds to CD 137.

In some embodiments, an anti-CDl37 antibody described herein increases infiltration of immune cells expressing CD45 into a tumor microenvironment in a subject as determined by an immune cell infiltration assay, wherein the assay comprises the following steps:

(i) administering the anti-CD 137 antibody to a subject having a tumor;

(ii) obtaining a sample of the tumor;

(iii) contacting the sample with an fluorescently-labeled detection antibody that specifically binds to CD45, wherein the detection antibody fluorescently-labels the immune cells expressing CD45; and

(iv) determining a quantity of the fluorescently-labeled immune cells expressing CD45 by flow cytometry,

wherein an increase in the quantity of fluorescently-immune cells expressing CD45 in the tumor indicates the anti-CD 137 antibody induces or enhances infiltration of immune cells into the tumor microenvironment. In some embodiments, an increase in the quantity of immune cells expressing CD45 is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or 80% of total cells in the tumor microenvironment.

In some embodiments, an anti-CDl37 antibody described herein reduces or inhibits infiltration of one or more immune cells into a tumor microenvironment as determined by an immune cell infiltration assay. In some embodiments, the anti-CDl37 antibody decreases infiltration of one or more immune cells into a tumor microenvironment relative to a reference antibody, as determined by an immune cell infiltration assay. In some embodiments, the reference antibody is an antibody comprising the same isotype as the anti-CD 137 antibody and does not specifically bind to CD137. In some embodiments, the reference antibody is an antibody comprising the same isotype as the anti-CD 137 antibody and specifically binds to CD 137. In some embodiments, the reference antibody is an antibody comprising the different isotype as the anti- CD 137 antibody and does not specifically bind to CD 137. In some embodiments, the reference antibody is an antibody comprising a different isotype as the anti-CD 137 antibody and specifically binds to CD137. In some embodiments, a decrease in immune cells is less than 40%, 35%, 30%, 25%, 20%, 15%, 10%, or 5% of total cells in a tumor microenvironment.

In some embodiments, an anti-CDl37 antibody described herein decreases infiltration of tumor associated macrophages into a tumor microenvironment in a subject as determined by an immune cell infiltration assay, wherein the assay comprises the following steps:

(i) obtaining a sample of the tumor;

(ii) contacting the sample with one or more antibodies that label the tumor associated macrophage, wherein the one or more antibodies specifically bind to an immune cell marker selected from the group consisting of F4/80, CD1 lb, CD45 and a combination thereof; and

(iii) determining a quantity of the labeled tumor associated macrophages by flow cytometry. In some embodiments, tumor-associated macrophages are less than 40%, 35%, 30%, 25%, 20%, 15%, 10%, or 5% of immune cells in the tumor microenvironment. In some embodiments, tumor-associated macrophages express F4/80, CDl lb and CD45.

In some embodiments, an anti-CDl37 antibody described herein decreases infiltration of T regulatory cells (Tregs) into a tumor microenvironment in a subject as determined by an immune cell infiltration assay, wherein the assay comprises the following steps:

(i) obtaining a sample of the tumor;

(ii) contacting the sample with one or more antibodies that label the tumor associated macrophage, wherein the one or more antibodies specifically bind to an immune cell marker selected from the group consisting of CD25, FOXP-3, CD4 and a combination thereof; and

(iii) determining a quantity of the labeled Treg cells by flow cytometry. In some embodiments, Treg cells are less than 35%, 30%, 25%, 20%, 15%, 10%, or 5% of CD4+ T cells in the tumor microenvironment. In some embodiments, Treg cells express CD4, FOXP-3 and CD25.

In some embodiments, an anti-CD 137 antibody described herein protects T cells from T cell exhaustion and/or reverses T cell exhaustion as determined by a T cell exhaustion assay. Exhausted T cells can be distinguished from other T cell dysfunctions such as anergy and senescence based on their underlying molecular mechanisms (Crespo et ah, (2013) Curr Opin Immunol 25(2):24l-22l). Whereas anergy occurs during priming due to the absence of costimulatory signals and senescence is growth arrest after extensive proliferation, exhausted T cells arise from T cells which initially gained and provided T cell effector function, but that exhibit a gradual deterioration of T cell effector function due to continuous T cell receptor (TCR) stimulation from persistent antigen and inflammatory mediators, both of which commonly occur in tumors (Wherry & Kurachi (2015) Nat Rev Immunol l5(8):486-99). Hallmarks of T cell exhaustion include, but are not limited to, continuous deterioration of in vivo and/or ex vivo T cell function, an increased expression of multiple inhibitory receptors (IRs) (e.g., PD-l, CTLA-4, LAG-3, TIM-3, CD244, CD160, TIGIT), progressive loss or decrease of effector cytokine secretion (e.g., IL-2, interferon gamma (IFNy), tumor necrosis factor alpha (TNFa)), loss or decrease of CC chemokine (b-chemokine) production, poor responsiveness to IL-7 and IL-15, loss or decrease of proliferative capacity, loss or decrease of in vivo and/or ex vivo cytolytic activity, altered cell metabolism and a different transcriptional profile relative to non-exhausted T cells. Severely exhausted T cells can succumb to deletion (Yi et al., (2010) Immunology l29(4):474- 481).

In some embodiments, an anti-CD 137 antibody described herein protects T cells from T cell exhaustion and/or reverses T cell exhaustion as determined by a T cell exhaustion assay wherein the T cell exhaustion assay determines an amount or level of one or more effector cytokines secreted from T cells treated with an anti-CD 137 antibody described herein, wherein the amount or level of the one or more effector cytokines indicates protection from or reversion of T cell exhaustion. In some embodiments, the T cell exhaustion assay comprises the following steps:

(i) isolating of T cells from a subject (e.g., a human subject);

(ii) contacting the T cells with an antigen that induces T cell exhaustion;

(iii) contacting the T cells with the anti-CD 137 antibody;

(iv) determining an amount of one or more effector cytokines secreted from the T cells; and;

(v) comparing the amount or level of the one or more effector cytokines secreted from the T cells to an amount or level secreted from reference T cells,

wherein the reference T cells are not contacted with the antigen that induces T cell exhaustion, and wherein the difference in the amount or level of the one or more effector cytokines secreted from the T cells and reference T cells indicates protection from or reversion of T cell exhaustion. In some embodiments, the one or more effector cytokines is selected from IL-2, IFNy, and TNFa. In some embodiments, the amount or level of the one or more effector cytokines is determined by ELISA. ELIS As suitable for the determination of the amount or level of the one or more effector cytokines are known in the art. In some embodiments, the amount or level of the one or more effector cytokines is determined by Meso Scale Discovery. In some embodiments, the amount or level of the one or more effector cytokines is determined by any one of the cytokine production assays described herein.

The gradual dysfunction of exhausted T cells is accompanied by the expression of IRs, which transmit inhibitory signals to the nucleus upon interaction with ligands on target cells. Accordingly, in some embodiments, an anti-CD 137 antibody described herein protects T cells from T cell exhaustion and/or reverses T cell exhaustion as determined by a T cell exhaustion assay wherein the T cell exhaustion assay determines an expression level of one or more inhibitory receptors on T cells treated with an anti-CD 137 antibody described herein, wherein the expression level of the one or more inhibitory receptors indicates protection from or reversion of T cell exhaustion. In some embodiments, the T cell exhaustion assay comprises the following steps:

(i) isolating of T cells from a subject (e.g., a human subject);

(ii) contacting the T cells with an antigen that induces T cell exhaustion;

(iii) contacting the T cells with the anti-CD 137 antibody;

(iv) determining an expression level of one or more inhibitory receptors on T cells; and

(v) comparing the expression level of one or more inhibitory receptors on T cells to an amount or level secreted from reference T cells, wherein the reference T cells are not contacted with the antigen that induces T cell exhaustion, and wherein the difference in the expression level of one or more inhibitory receptors on T cells and reference T cells indicates protection from or reversion of T cell exhaustion.

In some embodiments, the one or more inhibitory receptors is selected from TIGIT and PD-l In some embodiments, the expression level of the one or more inhibitory receptors is determined by flow cytometry. Methods to determine expression levels of inhibitory receptors on immune cells (e.g. T cells) by flow cytometry are known in the art.

In some embodiments, the amount of exhausted T cells is less than 20%, 15%, 10% or 5% of total CD8+ or CD4+ T cells in a tumor microenvironment. Where the assays described herein refer to‘isolating T cells from a subject’; it is to be understood that the assay may suitably be performed on T cells previously isolated from a subject.

Where the assays described herein refer to (i) administering the anti-CD 137 antibody to a subject and (ii) isolating T cells from the subject; it is to be understood that the assay may suitably be performed on T cells previously isolated from a subject to whom the anti-CD 137 antibody has been administered.

Where the assays described herein refer to‘obtaining a sample of the tumor’; it is to be understood that the assay may suitably be performed on a sample of a tumor previously isolated from a subject.

Where the assays described herein refer to (i) administering the anti-CD 137 antibody to a subject having a tumor and (ii) obtaining a sample of the tumor; it is to be understood that the assay may suitably be performed a sample of a tumor previously isolated from a subject to whom the anti-CD 137 antibody has been administered.

III. Non-Ligand Binding

In some embodiments, an anti-CD 137 antibody described herein binds to a non-ligand binding region of CD137, as determined by a ligand binding assay. A ligand binding assay (LBA) is an assay, or an analytic procedure, that provides a measure of the interactions that occur between two reactant molecules (e.g., a receptor and ligand polypeptides). Suitably, the LBA provides a measure of the degree of affinity between the two reactant molecules (e.g., a receptor and ligand polypeptides). For example, in some embodiments a ligand binding assay is used to determine the presence, rate, extent of binding, or combinations thereof, of a ligand molecule (e.g., CD137L) to a receptor (e.g., CD 137). In some embodiments, to determine the presence, rate and/or extent of ligand binding to a receptor, a ligand binding assay comprises detecting the formation of a ligand:receptor complex. In some embodiments, to determine the presence, rate and/or extent of ligand binding to a receptor, a ligand binding assay comprises determining the dissociation of a ligand:receptor complex.

In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection of a fluorescently-labeled ligand in complex with a receptor. In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of fluorescently-labeled receptor in complex with a ligand. In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of a fluorescently-labeled antibody that specifically binds to the ligand:receptor complex. Methods of detecting and quantifying fluorescence are known in the art and include, but are not limited to, fluorescence polarization (FP) and fluorescence anisotropy (FA).

In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of a radioactively-labeled ligand in complex with a receptor. In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of radioactively-labeled receptor in complex with a ligand. In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of a radioactively-labeled antibody that specifically binds to the ligand:receptor complex. Methods of detecting and quantifying radioactivity are known in the art and include, but are not limited to, quantitative autoradiography and scintillation counting.

In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of a bioluminescently-labeled ligand in complex with a receptor. In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of bioluminescently-labeled receptor in complex with a ligand. In some embodiments, the formation and/or dissociation of a ligand:receptor complex is determined by detection and/or quantification of an amount of a bioluminescently-labeled antibody that specifically binds to the ligand:receptor complex. Methods of detecting and quantifying bioluminescence are known in the art and include, but are not limited to, luminometry.

In some embodiment, formation and/or dissociation of the ligand:receptor complex is determined by surface plasmon resonance (SPR) as described supra.

In some embodiments, a ligand binding assay determines if an antibody that specifically binds to a receptor (e.g., an anti-CDl37 antibody) affects the formation of a ligand:receptor complex by determining the presence, rate and/or extent of ligand binding to the receptor in the presence of the antibody. In some embodiments, an antibody (e.g., an anti-CD 137 antibody) that specifically binds to a receptor (e.g., CD137) and decreases, disrupts or blocks the formation of a ligand:receptor complex (e.g., a CDl37:CDl37L complex) is known as a“ligand blocking antibody”. In some embodiments, a“ligand blocking antibody” may decrease the formation of a ligand:receptor complex (e.g., a CDl37:CDl37L complex) by at least 10%, at least 20%, at least 30%, at least 40% or at least 50% compared to the formation of the ligand:receptor complex (e.g., the CDl37:CDl37L complex) which occurs in the absence of the ligand blocking antibody. In some embodiments, an antibody (e.g., an anti-CD 137 antibody) that specifically binds to a receptor (e.g., CD137) and does not decrease, disrupt or block the formation of a ligand:receptor complex (e.g., a CDl37:CDl37L complex) is referred to as a“non-ligand blocking antibody”. In some embodiments, a“non-ligand blocking antibody” may decrease the formation of a ligand:receptor complex (e.g., a CDl37:CDl37L complex) by less than 10%, less than 5%, less than 2% or less than 1% compared to the formation of the ligand:receptor complex (e.g., the CDl37:CDl37L complex) which occurs in the absence of the non-ligand blocking antibody. Accordingly, in some embodiments a ligand binding assay characterizes an antibody that binds to a receptor as a“ligand blocking antibody” or a“non-ligand blocking antibody”.

In some embodiments, a ligand binding assay characterizes an antibody that specifically binds to a receptor and promotes the formation of a ligand:receptor complex. In some embodiments, a ligand binding assay characterizes an antibody that specifically binds to a receptor and stabilizes the formation of a ligand:receptor complex. In some embodiments, the induction and/or stabilization of the formation of a ligand:receptor complex by an antibody contributes to the antibody’s agonistic effect. In some embodiments, an anti-CDl37 antibody described herein agonizes CD137, as determined by a ligand binding assay.

In some embodiments, an isolated anti-CDl37 antibody, or antigen-binding fragment thereof, described herein, binds to CD 137 and induces CD137L binding as determined by a ligand binding assay (LB A).

In some embodiments, an isolated anti-CDl37 antibody, or antigen-binding fragment thereof, described herein, binds to CD 137 and induces CD137L binding as determined by a ligand binding assay, wherein the ligand binding assay comprises the following steps:

(i) combining an anti-CD 137 antibody with CD 137 and CD137L at various concentrations, wherein CD 137 and CD137L form a CDl37:CDl37L complex, and

(ii) detecting the CDl37:CDl37L complex in the presence of the anti-CD 137 antibody over time, wherein an increase in CDl37:CDl37L complex in the presence of the anti-CD 137 antibody indicates the anti-CD 137 antibody induces CD137L binding to CD 137. The increase in CDl37:CDl37L complex in the presence of the anti-CDl37 antibody may be at least 1.5-fold, at least 2-fold, at least 5-fold, at least 10-fold, or at least 20-fold greater that the amount CDl37:CDl37L complex in the absence of the anti-CD 137 antibody.

In some embodiments, an isolated anti-CDl37 antibody, or antigen-binding fragment thereof, described herein, binds to a non-ligand binding region of CD 137 as determined by a ligand binding assay, wherein the ligand binding assay comprises the following steps:

(i) combining an anti-CD 137 antibody with CD 137 and CD137L at various concentrations, wherein CD 137 and CD137L form a CDl37:CDl37L complex, and

(ii) detecting the CDl37:CDl37L complex in the presence of the anti-CD 137 antibody over time,

wherein no change in the CDl37:CDl37L complex in the presence of the anti-CD 137 antibody indicates the anti-CD 137 antibody binds to a non-ligand binding region of CD 137. In some embodiments, less than a 2% change in CDl37:CDl37L complex indicates the anti-CDl37 antibody binds to a non-ligand binding region of CD137. In some embodiments, less than a 5% change in CDl37:CDl37L complex indicates the anti-CDl37 antibody binds to a non-ligand binding region of CD137. In some embodiments, less than a 10% change in CDl37:CDl37L complex indicates the anti-CD 137 antibody binds to a non-ligand binding region of CD 137.

In some embodiments, an anti-CD 137 antibody described herein binds to a non-ligand binding region of CD137, as determined by biolayer interferometry. In some embodiments, an anti-CDl37 antibody described herein binds to a non-ligand binding region of CD137, as determined by surface plasmon resonance imaging (SPRi). In some embodiments, CD137 and CD137L is sequentially applied to a sensor pre-loaded with an anti-CDl37 antibody (i.e., the antibody is captured on a sensor). In some embodiments, the binding of an anti-CDl37 antibody to a non-ligand binding region is indicated by an increase in response upon exposure to CD137L.

IV. Functions of CD 137 Binding Antibodies

In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and inhibit or reduce T cell exhaustion. In some embodiments, the anti-CDl37 agonist antibodies described herein bind to human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and induce or enhance T cell activation. In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD 137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and induce or enhance cytokine production by immune cells. In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and induce or enhance T cell proliferation. In some embodiments, the anti- CD 137 agonist antibodies described herein bind to human CD 137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and exhibit anti-tumor efficacy. In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD 137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and inhibit or reduce macrophage differentiation and/or activation. In some embodiments, the anti- CD 137 agonist antibodies described herein bind to human CD 137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and induce or enhance NFD D signaling. In some embodiments, the anti-CDl37 agonist antibodies described herein bind to human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD 137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and do not induce hepatotoxicity. In some embodiments, the anti-CDl37 agonist antibodies described herein bind to human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and bind to a non-ligand binding domain on extracellular CD137. In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD 137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and do not inhibit CD137 and CD137L interaction. In some embodiments, the anti-CD 137 agonist antibodies described herein bind to human CD137 with an affinity (KD) of about 30-100 nM (e.g., between about 30 nM and about 100 nM) and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce T cell exhaustion and induce or enhance T cell activation. In some embodiments, the anti- CD 137 agonist antibodies described herein inhibit or reduce T cell exhaustion and induce or enhance cytokine production by immune cells. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce T cell exhaustion and induce or enhance T cell proliferation. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce T cell exhaustion and exhibit anti-tumor efficacy. In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce T cell exhaustion and inhibit or reduce macrophage differentiation and/or activation. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce T cell exhaustion and induce or enhance NFD D signaling. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce T cell exhaustion and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce T cell exhaustion and do not induce hepatotoxicity. In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce T cell exhaustion and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce T cell exhaustion and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce T cell exhaustion and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell activation and induce or enhance cytokine production by immune cells. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell activation and induce or enhance T cell proliferation. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell activation and exhibit anti-tumor efficacy. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell activation and inhibit or reduce macrophage differentiation and/or activation. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell activation and induce or enhance NFK signaling. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell activation and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell activation and do not induce hepatotoxicity. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell activation and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell activation and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti- CD 137 agonist antibodies described herein induce or enhance T cell activation and bind to an epitope comprising K114 of SEQ ID NO: 3.

In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance cytokine production by immune cells and induce or enhance T cell proliferation. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance cytokine production by immune cells and exhibit anti-tumor efficacy. In some embodiments, the anti- CD 137 agonist antibodies described herein induce or enhance cytokine production by immune cells and inhibit or reduce macrophage differentiation and/or activation. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance cytokine production by immune cells and induce or enhance NFK signaling. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance cytokine production by immune cells and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance cytokine production by immune cells and do not induce hepatotoxicity. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance cytokine production by immune cells and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance cytokine production by immune cells and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance cytokine production by immune cells and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell proliferation and exhibit anti-tumor efficacy. In some embodiments, the anti- CD 137 agonist antibodies described herein induce or enhance T cell proliferation and inhibit or reduce macrophage differentiation and/or activation. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell proliferation and induce or enhance NFD D signaling. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell proliferation and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell proliferation and do not induce hepatotoxicity. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell proliferation and bind to a non-ligand binding domain on extracellular CD137. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance T cell proliferation and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance T cell proliferation and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CDl37 agonist antibodies described herein exhibit anti tumor efficacy and inhibit or reduce macrophage differentiation and/or activation. In some embodiments, the anti-CD 137 agonist antibodies described herein exhibit anti-tumor efficacy and induce or enhance NFK signaling. In some embodiments, the anti-CDl37 agonist antibodies described herein exhibit anti-tumor efficacy and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CD 137 agonist antibodies described herein exhibit anti-tumor efficacy and do not induce hepatotoxicity. In some embodiments, the anti-CD 137 agonist antibodies described herein exhibit anti-tumor efficacy and bind to a non ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein exhibit anti-tumor efficacy and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein exhibit anti-tumor efficacy and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce macrophage differentiation and/or activation and induce or enhance NFK signaling. In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce macrophage differentiation and/or activation and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce macrophage differentiation and/or activation and do not induce hepatotoxicity. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce macrophage differentiation and/or activation and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein inhibit or reduce macrophage differentiation and/or activation and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein inhibit or reduce macrophage differentiation and/or activation and bind to an epitope comprising K114 of SEQ ID NO: 3. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance NFK signaling and induce or enhance immune cell infiltration into a tumor microenvironment. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance NFK signaling and do not induce hepatotoxicity. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance NFK signaling and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance NFK signaling and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance NFK signaling and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance immune cell infiltration into a tumor microenvironment and do not induce hepatotoxicity. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance immune cell infiltration into a tumor microenvironment and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein induce or enhance immune cell infiltration into a tumor microenvironment and do not inhibit CD137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein induce or enhance immune cell infiltration into a tumor microenvironment and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CD 137 agonist antibodies described herein do not induce hepatotoxicity and bind to a non-ligand binding domain on extracellular CD 137. In some embodiments, the anti-CD 137 agonist antibodies described herein do not induce hepatotoxicity and do not inhibit CD137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein do not induce hepatotoxicity and bind to an epitope comprising Kl 14 of SEQ ID NO: 3.

In some embodiments, the anti-CDl37 agonist antibodies described herein bind to a non ligand binding domain on extracellular CD 137 and do not inhibit CD 137 and CD137L interaction. In some embodiments, the anti-CDl37 agonist antibodies described herein bind to a non-ligand binding domain on extracellular CD137 and bind to an epitope comprising K114 of SEQ ID NO: 3. In some embodiments, the anti-CDl37 agonist antibodies described herein do not inhibit CD137 and CD137L interaction and bind to an epitope comprising Kl 14 of SEQ ID NO: 3. Epitope Mapping

The disclosure provides anti-CD 137 antibodies, or antigen binding fragments thereof, that specifically bind to an epitope of human CD137 and compete with a reference mAb (e.g., mAbl) for binding to the epitope of human CD 137. Methods to characterize, map, or otherwise elucidate the epitope of an anti-CD 137 antibody can be grouped into structural, functional, or computational methods. A particularly suitable structural method to determine the precise molecular architecture of the interaction between an antibody and the corresponding antigen to which it binds is x-ray crystallography (alternatively“x-ray co-crystallography). A crystal structure of a bonded antibody- antigen pair enables very accurate determination of key interactions between individual amino acids from both side chains and main chain atoms in both the epitope of the antigen and the paratope of the antibody. Amino acids that are within 4 angstroms (A) of each other are generally considered to be contacting residues. The methodology typically involves purification of antibody and antigen, formation and purification of the complex, followed by successive rounds of crystallization screens and optimization to obtain diffraction-quality crystals. Structural solution is obtained following x-ray crystallography frequently at a synchrotron source. Other structural methods for epitope mapping include, but are not limited to, hydrogen-deuterium exchange coupled to mass spectrometry, crosslinking-coupled mass spectrometry, and nuclear magnetic resonance (NMR) (see, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996); Abbott et ah, (2014) Immunology l42(4):526-535).

Functional methods for epitope mapping are well known in the art and typically involve an assessment or quantification of antibody binding to whole proteins, protein fragments or peptides. Functional methods for epitope mapping can be used, for example, to identify linear or conformational epitopes and/or can be used to infer when two or more distinct antibodies bind to the same or similar epitopes. Functional methods for epitope mapping include, for example, immunoblotting and immunoprecipitation assays, wherein overlapping or contiguous peptides from CD137 are tested for reactivity with an anti-CDl37 antibody, e.g., mAbl. Other functional methods for epitope mapping include array-based oligopeptide scanning (alternatively known as “overlapping peptide scanning” or“pepscan analysis”), site-directed mutagenesis (e.g., alanine scanning mutagenesis), and high-throughput mutagenesis mapping (e.g., shotgun mutagenesis mapping). Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli el al., Methods in Enzymology 9:242 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et al., J. Immunol. 137:3614 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Press (1988)); solid phase direct label RIA using 1-125 label (see Morel et al., Mol. Immunol. 25(l):7 (1988)); solid phase direct biotin-avidin EIA (Cheung et al., Virology 176:546 (1990)); and direct labeled RIA. (Moldenhauer et al., Scand. J. Immunol. 32:77 (1990)). Typically, such assays involve the use of purified antigen bound to a solid surface or cells and either 1) an unlabeled test antigen-binding protein and a labeled reference antigen-binding protein, or 2) a labeled test antigen-binding protein and an unlabeled reference antigen-binding protein. Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test antigen-binding protein. Usually the test antigen-binding protein is present in excess. Antigen-binding proteins identified by competition assay (competing antigen-binding proteins) include antigen-binding proteins binding to the same epitope as the reference antigen-binding proteins (e.g., mAbl) and antigen-binding proteins binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antigen-binding protein (e.g., mAbl) for steric hindrance to occur. Additional details regarding methods for determining competitive binding are provided in the examples herein. Usually, when a competing antigen- binding protein is present in excess (e.g., about 1-, about 5-, about 10-, about 20- about 50-, or about lOO-fold excess), it will inhibit (e.g., reduce or block) specific binding of a reference antigen-binding protein to a common antigen by at least about 40-45%, about 45-50%, about 50-55%, about 55-60%, about 60-65%, about 65-70%, about 70-75% or about 75% or more. In some instances, binding is inhibited by at least about 80-85%, about 85-90%, about 90-95%, about 95-97%, or about 97% or more.

The site-directed mutagenesis method involves targeted site-directed mutagenesis where critical amino acids are identified by systematically introducing substitutions along the protein sequence and then determining the effects of each substitution on antibody binding. This may be done by "alanine scanning mutagenesis" (Cunningham and Wells (1989) Science 244: 1081-085), or some other form of point mutagenesis of amino acid residues in CD137. Without being bound by theory, two or more antibodies (e.g., a test antibody and a reference antibody, e.g., mAbl) have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of the first antibody reduce or eliminate binding of the second or more antibodies.

Shotgun mutagenesis mapping utilizes a comprehensive plasmid-mutation library for the target gene, with each clone in the library bearing a unique amino acid mutation and the entire library covering every amino acid in the target protein. The clones that constitute the mutation library are individually arranged in microplates, expressed within living mammalian cells, and tested for immunoreactivity with antibodies of interest. Amino acids critical for antibody epitopes are identified by a loss of reactivity and are then mapped onto a protein structure to visualize epitopes. Expression of the target protein antigen within mammalian cells often provides the native structure of the target protein antigen, which allows both linear and conformational epitope structures to be mapped on complex proteins. (Paes et al., J. Am. Chem. Soc. 131 (20): 6952-6954 (2009); Banik and Doranz, Genetic Engineering and Biotechnology News 3(2): 25-28 (2010)).

The epitope bound by an anti-CD 137 antibody may also be determined using peptide scanning methods. In peptide scanning, libraries of short peptide sequences from overlapping segments of the target protein, CD 137 are tested for their ability to bind antibodies of interest. The peptides are synthesized and screened for binding, e.g. using ELISA or BIACORE, or on a chip, by any of the multiple methods for solid-phase screening (Reineke et al, Curr. Opin. Biotechnol. 12: 59-64, 2001) as in the "pepscan" methodology (WO 84/03564; WO 93/09872).

A recently developed technology termed CLIPS (chemical linkage of peptides onto scaffolds) may be used to map conformational epitopes. The loose ends of the peptides are affixed onto synthetic scaffolds, so that the scaffolded peptide may be able to adopt the same spatial structure as the corresponding sequence in the intact protein. CLIPS technology is used to fix linear peptides into cyclic structures ('single-loop' format), and to bring together different parts of a protein binding site ('double- loop', 'triple-loop', etc. format), so as to create conformational epitopes that may be assayed for antibody binding. (US Pat. No. 7,972,993).

The epitopes bound by antibodies provided by the disclosure may also be mapped using computational methods. In these methods, for example, libraries of peptide fragments are displayed on the surface of the phage or cell. Epitopes are then mapped by screening antibodies against these fragments using selective binding assays. A number of computational tools have been developed which allow the prediction of conformational epitopes based upon linear affinity- selected peptides obtained using phage display (Mayrose et al., (2007) Bioinformatics 23:3244- 3246). Methods are also available for the detection of conformational epitopes by phage display. Microbial display systems may also be used to express properly folded antigenic fragments on the cell surface for identification of conformational epitopes (Cochran et al., J. Immunol. Meth. 287: 147-158, 2004; Rockberg et al., Nature Methods 5: 1039-1045, 2008).

Methods involving proteolysis and mass spectroscopy may also be used to determine antibody epitopes (Baerga-Ortiz et al., Protein Sci. 2002 June; 1 1 (6): 1300-1308). In limited proteolysis, the antigen is cleaved by different proteases, in the presence and in the absence of the antibody, and the fragments are identified by mass spectrometry. The epitope is the region of the antigen that becomes protected from proteolysis upon binding of the antibody (Suckau et al., Proc. Natl. Acad. Sci. USA 87: 9848-9852, 1990). Additional proteolysis-based methods include, for example, selective chemical modification (Fiedler et al., Bioconjugate Chemistry 1998, 9(2): 236- 234, 1998), epitope excision (Van de Water et al., Clin. Immunol. Immunopathol. 1997, 85(3): 229-235, 1997), and the recently developed method of hydrogen-deuterium (H/D) exchange (Flanagan, N., Genetic Engineering and Biotechnology News 3(2): 25-28, 2010).

In some embodiments, the anti-CDl37 antibodies described herein bind to an epitope located within amino acid residues 111-135 of SEQ ID NO: 3 as determined by mutagenesis and mammalian display. In some embodiments, the anti-CDl37 antibodies described herein bind to an epitope comprising Kl 14 of SEQ ID NO: 3 as determined by mutagenesis and mammalian display. In some embodiments, the anti-CDl37 antibodies described herein bind to an epitope comprising El 11, T113 and K114 of SEQ ID NO: 3 as determined by mutagenesis and mammalian display. In some embodiments, the anti-CDl37 antibodies described herein bind to an epitope comprising El 11, T113, K114 and P135 of SEQ ID NO: 3 as determined by mutagenesis and mammalian display. In some embodiments, the anti-CD 137 antibodies described herein bind to an epitope comprising El 11, T113, K114, N126, 1132 and P135 of SEQ ID NO: 3 as determined by mutagenesis and mammalian display.

Methods for Producing the Anti-CD137 Antibodies and Antigen-binding Fragments Thereof

The disclosure also features methods for producing any of the anti-CDl37 antibodies or antigen-binding fragments thereof described herein. In some embodiments, methods for preparing an antibody described herein can include immunizing a subject (e.g., a non-human mammal) with an appropriate immunogen. Suitable immunogens for generating any of the antibodies described herein are set forth herein. For example, to generate an antibody that binds to CD137, a skilled artisan can immunize a suitable subject (e.g., a non-human mammal such as a rat, a mouse, a gerbil, a hamster, a dog, a cat, a pig, a goat, a horse, or a non-human primate) with a full-length CD 137 polypeptide such as a full-length human CD 137 polypeptide comprising the amino acid sequence depicted in SEQ ID NO. 3.

A suitable subject (e.g., a non-human mammal) can be immunized with the appropriate antigen along with subsequent booster immunizations a number of times sufficient to elicit the production of an antibody by the mammal. The immunogen can be administered to a subject (e.g., a non-human mammal) with an adjuvant. Adjuvants useful in producing an antibody in a subject include, but are not limited to, protein adjuvants; bacterial adjuvants, e.g., whole bacteria (BCG, Corynebacterium parvum or Salmonella Minnesota ) and bacterial components including cell wall skeleton, trehalose dimycolate, monophosphoryl lipid A, methanol extractable residue (MER) of tubercle bacillus, complete or incomplete Freund’s adjuvant; viral adjuvants; chemical adjuvants, e.g., aluminum hydroxide, and iodoacetate and cholesteryl hemisuccinate. Other adjuvants that can be used in the methods for inducing an immune response include, e.g., cholera toxin and parapoxvirus proteins. See also Bieg et al. (1999) Autoimmunity 3 l(l): l5-24. See also, e.g., Lodmell et al. (2000) Vaccine l_8: 1059-1066; Johnson et al. (1999) J Med Chem 42:4640-4649; Baldridge et al. (1999) Methods 19: 103-107; and Gupta et al. (1995) Vaccine 13(14): 1263-1276.

In some embodiments, the methods include preparing a hybridoma cell line that secretes a monoclonal antibody that binds to the immunogen. For example, a suitable mammal such as a laboratory mouse is immunized with a CD 137 polypeptide as described above. Antibody- producing cells (e.g., B cells of the spleen) of the immunized mammal can be isolated two to four days after at least one booster immunization of the immunogen and then grown briefly in culture before fusion with cells of a suitable myeloma cell line. The cells can be fused in the presence of a fusion promoter such as, e.g., vaccinia virus or polyethylene glycol. The hybrid cells obtained in the fusion are cloned, and cell clones secreting the desired antibodies are selected. For example, spleen cells of B alb/c mice immunized with a suitable immunogen can be fused with cells of the myeloma cell line PAI or the myeloma cell line Sp2/0-Ag 14. After the fusion, the cells are expanded in suitable culture medium, which is supplemented with a selection medium, for example HAT medium, at regular intervals in order to prevent normal myeloma cells from overgrowing the desired hybridoma cells. The obtained hybridoma cells are then screened for secretion of the desired antibodies, e.g., an antibody that binds to CD137.

In some embodiments, a skilled artisan can identify an anti-CD 137 antibody from a non- immune biased library as described in, e.g., U.S. patent no. 6,300,064 (to Knappik et ah; Morphosys AG) and Schoonbroodt et al. (2005) Nucleic Acids Res 33(9):e8l.

In some embodiments, the methods described herein can involve, or be used in conjunction with, e.g., phage display technologies, bacterial display, yeast surface display, eukaryotic viral display, mammalian cell display, and cell-free (e.g., ribosomal display) antibody screening techniques (see, e.g., Etz et al. (2001) J Bacteriol 183:6924-6935; Cornells (2000) Curr Opin Biotechnol 11:450-454; Klemm et al. (2000) Microbiology 146:3025-3032; Kieke et al. (1997) Protein Eng 10: 1303- 1310; Yeung et al. (2002) Biotechnol Prog 18:212-220; Boder et al. (2000) Methods Enzymology 328:430-444; Grabherr et al. (2001) Comb Chem High Throughput Screen 4: 185-192; Michael et al. (1995) Gene Ther 2:660-668; Pereboev et al. (2001) J Virol 75:7107- 7113; Schaffitzel et al. (1999) J Immunol Methods 231:119-135; and Hanes et al. (2000) Nat Biotechnol 18:1287-1292).

Methods for identifying antibodies using various phage display methods are known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. Such phage can be utilized to display antigen-binding domains of antibodies, such as Fab, Fv, or disulfide-bond stabilized Fv antibody fragments, expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage used in these methods are typically filamentous phage such as fd and M13. The antigen binding domains are expressed as a recombinantly-fused protein to any of the phage coat proteins pill, pVIII, or pIX. See, e.g., Shi et al. (2010) JMB 397:385-396. Examples of phage display methods that can be used to make the immunoglobulins, or fragments thereof, described herein include those disclosed in Brinkman et al. (1995) J Immunol Methods 182:41-50; Ames et al. (1995) J Immunol Methods 184:177-186; Kettleborough et al. (1994) Eur J Immunol 24:952- 958; Persic et al. (1997) Gene 187:9-18; Burton et al. (1994) Advances in Immunology 57:191- 280; and PCT publication nos. WO 90/02809, WO 91/10737, WO 92/01047, WO 92/18619, WO 93/11236, WO 95/15982, and WO 95/20401. Suitable methods are also described in, e.g., U.S. patent nos. 5,698,426; 5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698; 5,427,908; 5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969,108. In some embodiments, the phage display antibody libraries can be generated using mRNA collected from B cells from the immunized mammals. For example, a splenic cell sample comprising B cells can be isolated from mice immunized with CD 137 polypeptide as described above. mRNA can be isolated from the cells and converted to cDNA using standard molecular biology techniques. See, e.g., Sambrook et al. (1989)“Molecular Cloning: A Laboratory Manual, 2 nd Edition,” Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Harlow and Lane (1988), supra; Benny K. C. Lo (2004), supra; and Borrebaek (1995), supra. The cDNA coding for the variable regions of the heavy chain and light chain polypeptides of immunoglobulins are used to construct the phage display library. Methods for generating such a library are described in, e.g., Merz et al. (1995) J Neurosci Methods 62(l-2):2l3-9; Di Niro et al. (2005) Biochem J 388(Pt 3):889-894; and Engberg et al. (1995) Methods Mol Biol 51:355-376.

In some embodiments, a combination of selection and screening can be employed to identify an antibody of interest from, e.g., a population of hybridoma-derived antibodies or a phage display antibody library. Suitable methods are known in the art and are described in, e.g., Hoogenboom (1997) Trends in Biotechnology 15:62-70; Brinkman et al. (1995), supra; Ames et al. (1995), supra; Kettleborough et al. (1994), supra; Persic et al. (1997), supra; and Burton et al. (1994), supra. For example, a plurality of phagemid vectors, each encoding a fusion protein of a bacteriophage coat protein (e.g., pill, pVIII, or pIX of M13 phage) and a different antigen combining region are produced using standard molecular biology techniques and then introduced into a population of bacteria (e.g., E. coli). Expression of the bacteriophage in bacteria can, in some embodiments, require use of a helper phage. In some embodiments, no helper phage is required (see, e.g., Chasteen et al., (2006) Nucleic Acids Res 34(2l):el45). Phage produced from the bacteria are recovered and then contacted to, e.g., a target antigen bound to a solid support (immobilized). Phage may also be contacted to antigen in solution, and the complex is subsequently bound to a solid support.

A subpopulation of antibodies screened using the above methods can be characterized for their specificity and binding affinity for a particular antigen (e.g., human CD137) using any immunological or biochemical based method known in the art. For example, specific binding of an antibody to CD137, may be determined for example using immunological or biochemical based methods such as, but not limited to, an ELISA assay, SPR assays, immunoprecipitation assay, affinity chromatography, and equilibrium dialysis as described above. Immunoassays which can be used to analyze immunospecific binding and cross -reactivity of the antibodies include, but are not limited to, competitive and non-competitive assay systems using techniques such as Western blots, RIA, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays. Such assays are routine and well known in the art.

It is understood that the above methods can also be used to determine if, e.g., an anti- CD137 antibody does not bind to full-length, human CD137 and/or CD137 proteins.

In embodiments where the selected CDR amino acid sequences are short sequences (e.g., fewer than 10-15 amino acids in length), nucleic acids encoding the CDRs can be chemically synthesized as described in, e.g., Shiraishi et al. (2007) Nucleic Acids Symposium Series 51(1): 129- 130 and U.S. Patent No. 6,995,259. For a given nucleic acid sequence encoding an acceptor antibody, the region of the nucleic acid sequence encoding the CDRs can be replaced with the chemically synthesized nucleic acids using standard molecular biology techniques. The 5’ and 3’ ends of the chemically synthesized nucleic acids can be synthesized to comprise sticky end restriction enzyme sites for use in cloning the nucleic acids into the nucleic acid encoding the variable region of the donor antibody. Alternatively, fragments of chemically synthesized nucleic acids, together capable of encoding an antibody, can be joined together using DNA assembly techniques known in the art (e.g. Gibson Assembly).

In some embodiments, the anti-CD 137 antibodies described herein comprise an altered heavy chain constant region that has reduced (or no) effector function relative to its corresponding unaltered constant region. Effector functions involving the constant region of the anti-CD 137 antibody may be modulated by altering properties of the constant or Fc region. Altered effector functions include, for example, a modulation in one or more of the following activities: antibody- dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), apoptosis, binding to one or more Fc-receptors, and pro-inflammatory responses. Modulation refers to an increase, decrease, or elimination of an effector function activity exhibited by a subject antibody containing an altered constant region as compared to the activity of the unaltered form of the constant region. In particular embodiments, modulation includes situations in which an activity is abolished or completely absent. An altered constant region with altered FcR binding affinity and/or ADCC activity and/or altered CDC activity is a polypeptide which has either an enhanced or diminished FcR binding activity and/or ADCC activity and/or CDC activity compared to the unaltered form of the constant region. An altered constant region which displays increased binding to an FcR binds at least one FcR with greater affinity than the unaltered polypeptide. An altered constant region which displays decreased binding to an FcR binds at least one FcR with lower affinity than the unaltered form of the constant region. Such variants which display decreased binding to an FcR may possess little or no appreciable binding to an FcR, e.g., 0 to 50% (e.g., less than 50, 49, 48, 47, 46, 45, 44, 43,

42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17,

16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1%) of the binding to the FcR as compared to the level of binding of a native sequence immunoglobulin constant or Fc region to the FcR. Similarly, an altered constant region that displays modulated ADCC and/or CDC activity may exhibit either increased or reduced ADCC and/or CDC activity compared to the unaltered constant region. For example, in some embodiments, the anti-CD 137 antibody comprising an altered constant region can exhibit approximately 0 to 50% (e.g., less than 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13,

12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1%) of the ADCC and/or CDC activity of the unaltered form of the constant region. An anti-CD 137 antibody described herein comprising an altered constant region displaying reduced ADCC and/or CDC may exhibit reduced or no ADCC and/or CDC activity.

In some embodiments, an anti-CD 137 antibody described herein exhibits reduced or no effector function. In some embodiments, an anti-CDl37 antibody comprises a hybrid constant region, or a portion thereof, such as a G2/G4 hybrid constant region (see e.g., Burton et al. (1992) Adv Immun 5J_: l-l8; Canfield et al. (1991) J Exp Med 173: 1483-1491; and Mueller et al. (1997) Mol Immunol 34(6):44l-452). See above.

In some embodiments, an anti-CDl37 antibody may contain an altered constant region exhibiting enhanced or reduced complement dependent cytotoxicity (CDC). Modulated CDC activity may be achieved by introducing one or more amino acid substitutions, insertions, or deletions in an Fc region of the antibody. See, e.g., U.S. patent no. 6,194,551. Alternatively, or additionally, cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated may have improved or reduced internalization capability and/or increased or decreased complement- mediated cell killing. See, e.g., Caron et al. (1992) J Exp Med 176:1191-1195 and Shopes (1992) Immunol 148:2918-2922: PCT publication nos. WO 99/51642 and WO 94/29351; Duncan and Winter (1988) Nature 322:738-40; and U.S. Patent Nos. 5,648,260 and 5,624,821.

Recombinant Antibody Expression and Purification

The antibodies or antigen-binding fragments thereof described herein can be produced using a variety of techniques known in the art of molecular biology and protein chemistry. For example, a nucleic acid encoding one or both of the heavy and light chain polypeptides of an antibody can be inserted into an expression vector that contains transcriptional and translational regulatory sequences, which include, e.g., promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, transcription terminator signals, polyadenylation signals, and enhancer or activator sequences. The regulatory sequences include a promoter and transcriptional start and stop sequences. In addition, the expression vector can include more than one replication system such that it can be maintained in two different organisms, for example in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification.

Several possible vector systems are available for the expression of cloned heavy chain and light chain polypeptides from nucleic acids in mammalian cells. One class of vectors relies upon the integration of the desired gene sequences into the host cell genome. Cells which have stably integrated DNA can be selected by simultaneously introducing drug resistance genes such as E. coli gpt (Mulligan and Berg (1981) Proc Natl Acad Sci USA 78:2072) or Tn5 neo (Southern and Berg (1982) Mol Appl Genet l_:327). The selectable marker gene can be either linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection (Wigler et al. (1979) Cell 16:77). A second class of vectors utilizes DNA elements which confer autonomously replicating capabilities to an extrachromosomal plasmid. These vectors can be derived from animal viruses, such as bovine papillomavirus (Sarver et al. (1982) Proc Natl Acad Sci USA, 79:7147), cytomegalovirus, polyoma virus (Deans et al. (1984) Proc Natl Acad Sci USA 81:1292), or SV40 virus (Lusky and Botchan (1981) Nature 293:79).

The expression vectors can be introduced into cells in a manner suitable for subsequent expression of the nucleic acid. The method of introduction is largely dictated by the targeted cell type, discussed below. Exemplary methods include CaP0 4 precipitation, liposome fusion, cationic liposomes, electroporation, viral infection, dextran-mediated transfection, polybrene-mediated transfection, protoplast fusion, and direct microinjection.

Appropriate host cells for the expression of antibodies or antigen-binding fragments thereof include yeast, bacteria, insect, plant, and mammalian cells. Of particular interest are bacteria such as E. coli, fungi such as Saccharomyces cerevisiae and Pichia pastoris, insect cells such as SF9, mammalian cell lines (e.g., human cell lines), as well as primary cell lines.

In some embodiments, an antibody or fragment thereof can be expressed in, and purified from, transgenic animals (e.g., transgenic mammals). For example, an antibody can be produced in transgenic non-human mammals (e.g., rodents) and isolated from milk as described in, e.g., Houdebine (2002) Curr Opin Biotechnol 13(6}: 625-629; van Kuik-Romeijn et al. (2000) Transgenic Res 9(2): 155-159; and Pollock et al. (1999) J Immunol Methods 23 l(l-2): l47-l57.

The antibodies and fragments thereof can be produced from the cells by culturing a host cell transformed with the expression vector containing nucleic acid encoding the antibodies or fragments, under conditions, and for an amount of time, sufficient to allow expression of the proteins. Such conditions for protein expression will vary with the choice of the expression vector and the host cell, and will be easily ascertained by one skilled in the art through routine experimentation. For example, antibodies expressed in E. coli can be refolded from inclusion bodies (see, e.g., Hou et al. (1998) Cytokine 10:319-30). Bacterial expression systems and methods for their use are well known in the art (see Current Protocols in Molecular Biology, Wiley & Sons, and Molecular Cloning— A Faboratory Manual—3rd Ed., Cold Spring Harbor Faboratory Press, New York (2001)). The choice of codons, suitable expression vectors and suitable host cells will vary depending on a number of factors, and may be easily optimized as needed. An antibody (or fragment thereof) described herein can be expressed in mammalian cells or in other expression systems including but not limited to yeast, baculovirus, and in vitro expression systems (see, e.g., Kaszubska et al. (2000) Protein Expression and Purification 18:213-220).

Following expression, the antibodies and fragments thereof can be isolated. An antibody or fragment thereof can be isolated or purified in a variety of ways known to those skilled in the art depending on what other components are present in the sample. Standard purification methods include electrophoretic, molecular, immunological, and chromatographic techniques, including ion exchange, hydrophobic, affinity, and reverse-phase HPFC chromatography. For example, an antibody can be purified using a standard anti-antibody column (e.g., a protein-A or protein-G column). Ultrafiltration and diafiltration techniques, in conjunction with protein concentration, are also useful. See, e.g., Scopes (1994)“Protein Purification, 3 rd edition,” Springer- Verlag, New York City, New York. The degree of purification necessary will vary depending on the desired use. In some instances, no purification of the expressed antibody or fragments thereof will be necessary.

Methods for determining the yield or purity of a purified antibody or fragment thereof are known in the art and include, e.g., Bradford assay, UV spectroscopy, Biuret protein assay, Lowry protein assay, amido black protein assay, high pressure liquid chromatography (HPLC), mass spectrometry (MS), and gel electrophoretic methods (e.g., using a protein stain such as Coomassie Blue or colloidal silver stain).

Modification of the Antibodies or Antigen-Binding Fragments Thereof

The antibodies or antigen-binding fragments thereof can be modified following their expression and purification. The modifications can be covalent or non-covalent modifications. Such modifications can be introduced into the antibodies or fragments by, e.g., reacting targeted amino acid residues of the polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues. Suitable sites for modification can be chosen using any of a variety of criteria including, e.g., structural analysis or amino acid sequence analysis of the antibodies or fragments.

In some embodiments, the antibodies or antigen -binding fragments thereof can be conjugated to a heterologous moiety. The heterologous moiety can be, e.g., a heterologous polypeptide, a therapeutic agent (e.g., a toxin or a drug), or a detectable label such as, but not limited to, a radioactive label, an enzymatic label, a fluorescent label, a heavy metal label, a luminescent label, or an affinity tag such as biotin or streptavidin. Suitable heterologous polypeptides include, e.g., an antigenic tag (e.g., FLAG (DYKDDDDK; SEQ ID NO: 98), polyhistidine (6-His; HHHHHH; SEQ ID NO: 99), hemagglutinin (HA; YPYDVPDYA; SEQ ID NO: 100), glutathione-S-transferase (GST), or maltose-binding protein (MBP)) for use in purifying the antibodies or fragments. Heterologous polypeptides also include polypeptides (e.g., enzymes) that are useful as diagnostic or detectable markers, for example, luciferase, a fluorescent protein (e.g., green fluorescent protein (GFP)), or chloramphenicol acetyl transferase (CAT). Suitable radioactive labels include, e.g., 32 P, 33 P, 14 C, 125 I, 131 I, 35 S, and 3 H. Suitable fluorescent labels include, without limitation, fluorescein, fluorescein isothiocyanate (FITC), green fluorescent protein (GFP), DYLIGHT™ 488, phycoerythrin (PE), propidium iodide (PI), PerCP, PE-ALEXA FLUOR® 700, Cy5, allophycocyanin, and Cy7. Luminescent labels include, e.g., any of a variety of luminescent lanthanide (e.g., europium or terbium) chelates. For example, suitable europium chelates include the europium chelate of diethylene triamine pentaacetic acid (DTPA) or tetraazacyclododecane-l,4,7,l0-tetraacetic acid (DOTA). Enzymatic labels include, e.g., alkaline phosphatase, CAT, luciferase, and horseradish peroxidase.

Two proteins (e.g., an antibody and a heterologous moiety) can be cross-linked using any of a number of known chemical cross linkers. Examples of such cross linkers are those which link two amino acid residues via a linkage that includes a“hindered” disulfide bond. In these linkages, a disulfide bond within the cross-linking unit is protected (by hindering groups on either side of the disulfide bond) from reduction by the action, for example, of reduced glutathione or the enzyme disulfide reductase. One suitable reagent, 4-succinimidyloxycarbonyl-0C-methyl-a(2- pyridyldithio) toluene (SMPT), forms such a linkage between two proteins utilizing a terminal lysine on one of the proteins and a terminal cysteine on the other. Heterobifunctional reagents that cross-link by a different coupling moiety on each protein can also be used. Other useful cross linkers include, without limitation, reagents which link two amino groups (e.g., N-5-azido-2- nitrobenzoyloxysuccinimide), two sulfhydryl groups (e.g., l,4-bis-maleimidobutane), an amino group and a sulfhydryl group (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester), an amino group and a carboxyl group (e.g., 4-[p-azidosalicylamido]butylamine), and an amino group and a guanidinium group that is present in the side chain of arginine (e.g., p-azidophenyl glyoxal monohydrate).

In some embodiments, a radioactive label can be directly conjugated to the amino acid backbone of the antibody. Alternatively, the radioactive label can be included as part of a larger molecule (e.g., 125 I in meta-[ 125 I]iodophenyl-N-hydroxysuccinimide ([ 125 I]mIPNHS) which binds to free amino groups to form meta-iodophenyl (mIP) derivatives of relevant proteins (see, e.g., Rogers et al. (1997) J Nucl Med 38: 1221-1229) or chelate (e.g., to DOTA or DTPA) which is in turn bound to the protein backbone. Methods of conjugating the radioactive labels or larger molecules/chelates containing them to the antibodies or antigen-binding fragments described herein are known in the art. Such methods involve incubating the proteins with the radioactive label under conditions (e.g., pH, salt concentration, and/or temperature) that facilitate binding of the radioactive label or chelate to the protein (see, e.g., U.S. Patent No. 6,001,329).

Methods for conjugating a fluorescent label (sometimes referred to as a“fluorophore”) to a protein (e.g., an antibody) are known in the art of protein chemistry. For example, fluorophores can be conjugated to free amino groups (e.g., of lysines) or sulfhydryl groups (e.g., cysteines) of proteins using succinimidyl (NHS) ester or tetrafluorophenyl (TFP) ester moieties attached to the fluorophores. In some embodiments, the fluorophores can be conjugated to a heterobifunctional cross-linker moiety such as sulfo-SMCC. Suitable conjugation methods involve incubating an antibody protein, or fragment thereof, with the fluorophore under conditions that facilitate binding of the fluorophore to the protein. See, e.g., Welch and Redvanly (2003) “Handbook of Radiopharmaceuticals: Radiochemistry and Applications,” John Wiley and Sons (ISBN 0471495603).

In some embodiments, the antibodies or fragments can be modified, e.g., with a moiety that improves the stabilization and/or retention of the antibodies in circulation, e.g., in blood, serum, or other tissues. For example, the antibody or fragment can be PEGylated as described in, e.g., Lee et al. (1999) Bioconjug Chem 10(6): 973-8; Kinstler et al. (2002) Advanced Drug Deliveries Reviews 54:477-485; and Roberts et al. (2002) Advanced Drug Delivery Reviews 54:459-476 or HESylated (Fresenius Kabi, Germany; see, e.g., Pavisic et al. (2010) Int J P harm 387(1-2): 110-119). The stabilization moiety can improve the stability, or retention of, the antibody (or fragment) by at least 1.5 (e.g., at least 2, 5, 10, 15, 20, 25, 30, 40, or 50 or more) fold.

In some embodiments, the antibodies or antigen-binding fragments thereof described herein can be glycosylated. In some embodiments, an antibody or antigen-binding fragment thereof described herein can be subjected to enzymatic or chemical treatment, or produced from a cell, such that the antibody or fragment has reduced or absent glycosylation. Methods for producing antibodies with reduced glycosylation are known in the art and described in, e.g., U.S. patent no. 6,933,368; Wright et al. (1991) EMBO J 10(10):2717-2723; and Co et al. (1993) Mol Immunol 30: 1361.

Pharmaceutical Compositions and Formulations

The present disclosure also provides for a pharmaceutical composition comprising an anti- CD 137 antibody with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and/or adjuvant to be used with the methods disclosed herein. Such pharmaceutical compositions can be used in a subject having cancer, whose cancer cells express MHC I, as disclosed herein.

In certain embodiments, acceptable formulation materials preferably are nontoxic to recipients at the dosages and concentrations employed. In certain embodiments, the formulation material(s) are for s.c. and/or I.V. administration. In certain embodiments, the pharmaceutical composition can contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolality, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition. In certain embodiments, suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen- sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta- cyclodextrin); fillers; monosaccharides; disaccharides; and other carbohydrates (such as glucose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emulsifying agents; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; salt- forming counterions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (such as sucrose or sorbitol); tonicity enhancing agents (such as alkali metal halides, preferably sodium or potassium chloride, mannitol sorbitol); delivery vehicles; diluents; excipients and/or pharmaceutical adjuvants. (Remington's Pharmaceutical Sciences, 18th Edition, A. R. Gennaro, ed., Mack Publishing Company (1995). In certain embodiments, the formulation comprises PBS; 20 mM NaOAC, pH 5.2, 50 mM NaCl; and/or 10 mM NAOAC, pH 5.2, 9% Sucrose. In certain embodiments, the optimal pharmaceutical composition will be determined by one skilled in the art depending upon, for example, the intended route of administration, delivery format and desired dosage. See, for example, Remington's Pharmaceutical Sciences, supra. In certain embodiments, such compositions may influence the physical state, stability, rate of in vivo release and/or rate of in vivo clearance of the anti-CD 137 antibody.

In certain embodiments, the primary vehicle or carrier in a pharmaceutical composition can be either aqueous or non-aqueous in nature. For example, in certain embodiments, a suitable vehicle or carrier can be water for injection, physiological saline solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. In certain embodiments, the saline comprises isotonic phosphate-buffered saline. In certain embodiments, neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. In certain embodiments, pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which can further include sorbitol or a suitable substitute therefore. In certain embodiments, a composition comprising an anti-CD 137 antibody can be prepared for storage by mixing the selected composition having the desired degree of purity with optional formulation agents (Remington's Pharmaceutical Sciences, supra) in the form of a lyophilized cake or an aqueous solution. Further, in certain embodiments, a composition comprising an anti-CD 137 antibody can be formulated as a lyophilizate using appropriate excipients such as sucrose.

In certain embodiments, the pharmaceutical composition can be selected for parenteral delivery. In certain embodiments, the compositions can be selected for inhalation or for delivery through the digestive tract, such as orally. The preparation of such pharmaceutically acceptable compositions is within the ability of one skilled in the art.

In certain embodiments, the formulation components are present in concentrations that are acceptable to the site of administration. In certain embodiments, buffers are used to maintain the composition at physiological pH or at a slightly lower pH, typically within a pH range of from about 5 to about 8.

In certain embodiments, when parenteral administration is contemplated, a therapeutic composition can be in the form of a pyrogen-free, parenterally acceptable aqueous solution comprising an anti-CD 137 antibody, in a pharmaceutically acceptable vehicle. In certain embodiments, a vehicle for parenteral injection is sterile distilled water in which an anti-CD 137 antibody is formulated as a sterile, isotonic solution, and properly preserved. In certain embodiments, the preparation can involve the formulation of the desired molecule with an agent, such as injectable microspheres, bio-erodible particles, polymeric compounds (such as polylactic acid or polyglycolic acid), beads or liposomes, that can provide for the controlled or sustained release of the product which can then be delivered via a depot injection. In certain embodiments, hyaluronic acid can also be used, and can have the effect of promoting sustained duration in the circulation. In certain embodiments, implantable drug delivery devices can be used to introduce the desired molecule.

In certain embodiments, a pharmaceutical composition can be formulated for inhalation. In certain embodiments, an anti-CD 137 antibody can be formulated as a dry powder for inhalation. In certain embodiments, an inhalation solution comprising an anti-CDl37 antibody can be formulated with a propellant for aerosol delivery. In certain embodiments, solutions can be nebulized. Pulmonary administration is further described in PCT application No. PCT/US94/001875, which describes pulmonary delivery of chemically modified proteins.

In certain embodiments, it is contemplated that formulations can be administered orally. In certain embodiments, an anti-CDl37 antibody that is administered in this fashion can be formulated with or without those carriers customarily used in the compounding of solid dosage forms such as tablets and capsules. In certain embodiments, a capsule can be designed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systemic degradation is minimized. In certain embodiments, at least one additional agent can be included to facilitate absorption of an anti-CD 137 antibody. In certain embodiments, diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binders can also be employed.

In certain embodiments, a pharmaceutical composition can involve an effective quantity of an anti-CD 137 antibody in a mixture with non-toxic excipients which are suitable for the manufacture of tablets. In certain embodiments, by dissolving the tablets in sterile water, or another appropriate vehicle, solutions can be prepared in unit-dose form. In certain embodiments, suitable excipients include, but are not limited to, inert diluents, such as calcium carbonate, sodium carbonate or bicarbonate, lactose, or calcium phosphate; or binding agents, such as starch, gelatin, or acacia; or lubricating agents such as magnesium stearate, stearic acid, or talc.

Additional pharmaceutical compositions will be evident to those skilled in the art, including formulations involving an anti-CD 137 antibody in sustained- or controlled-delivery formulations. In certain embodiments, techniques for formulating a variety of other sustained- or controlled-delivery means, such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, are also known to those skilled in the art. See for example, PCT Application No. PCT/US 93/00829 which describes the controlled release of porous polymeric microparticles for the delivery of pharmaceutical compositions. In certain embodiments, sustained-release preparations can include semipermeable polymer matrices in the form of shaped articles, e.g. films, or microcapsules. Sustained release matrices can include polyesters, hydrogels, polylactides (U.S. Pat. No. 3,773,919 and EP 058,481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et ah, Biopolymers, 22:547-556 (1983)), poly (2-hydroxyethyl-methacrylate) (Langer et ah, J. Biomed. Mater. Res., 15: 167-277 (1981) and Langer, Chem. Tech., 12:98- 105 (1982)), ethylene vinyl acetate (Langer et ah, supra) or poly-D(-)-3-hydroxybutyric acid (EP 133,988). In certain embodiments, sustained release compositions can also include liposomes, which can be prepared by any of several methods known in the art. See, e.g., Eppstein et al, Proc. Natl. Acad. Sci. USA, 82:3688-3692 (1985); EP 036,676; EP 088,046 and EP 143,949.

The pharmaceutical composition to be used for in vivo administration typically is sterile. In certain embodiments, this can be accomplished by filtration through sterile filtration membranes. In certain embodiments, where the composition is lyophilized, sterilization using this method can be conducted either prior to or following lyophilization and reconstitution. In certain embodiments, the composition for parenteral administration can be stored in lyophilized form or in a solution. In certain embodiments, parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.

In certain embodiments, once the pharmaceutical composition has been formulated, it can be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. In certain embodiments, such formulations can be stored either in a ready-to- use form or in a form (e.g., lyophilized) that is reconstituted prior to administration.

In certain embodiments, kits are provided for producing a single-dose administration unit. In certain embodiments, the kit can contain both a first container having a dried protein and a second container having an aqueous formulation. In certain embodiments, kits containing single and multi-chambered pre-filled syringes (e.g., liquid syringes and lyosyringes) are included.

In certain embodiments, the effective amount of a pharmaceutical composition comprising an anti-CD 137 antibody to be employed therapeutically will depend, for example, upon the therapeutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment, according to certain embodiments, will thus vary depending, in part, upon the molecule delivered, the indication for which an anti-CD 137 antibody is being used, the route of administration, and the size (body weight, body surface or organ size) and/or condition (the age and general health) of the patient. In certain embodiments, the clinician can titer the dosage and modify the route of administration to obtain the optimal therapeutic effect.

In certain embodiments, the frequency of dosing will take into account the pharmacokinetic parameters of an anti-CD 137 antibody in the formulation used. In certain embodiments, a clinician will administer the composition until a dosage is reached that achieves the desired effect. In certain embodiments, the composition can therefore be administered as a single dose or as two or more doses (which may or may not contain the same amount of the desired molecule) over time, or as a continuous infusion via an implantation device or catheter. Further refinement of the appropriate dosage is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them. In certain embodiments, appropriate dosages can be ascertained through use of appropriate dose-response data.

In certain embodiments, the route of administration of the pharmaceutical composition is in accord with known methods, e.g. orally, through injection by intravenous, intraperitoneal, intracerebral (intra-parenchymal), intracerebroventricular, intramuscular, subcutaneously, intra ocular, intraarterial, intraportal, or intralesional routes; by sustained release systems or by implantation devices. In certain embodiments, the compositions can be administered by bolus injection or continuously by infusion, or by implantation device. In certain embodiments, individual elements of the combination therapy may be administered by different routes.

In certain embodiments, the composition can be administered locally via implantation of a membrane, sponge or another appropriate material onto which the desired molecule has been absorbed or encapsulated. In certain embodiments, where an implantation device is used, the device can be implanted into any suitable tissue or organ, and delivery of the desired molecule can be via diffusion, timed-release bolus, or continuous administration. In certain embodiments, it can be desirable to use a pharmaceutical composition comprising an anti-CD 137 antibody in an ex vivo manner. In such instances, cells, tissues and/or organs that have been removed from the patient are exposed to a pharmaceutical composition comprising an anti-CD 137 antibody after which the cells, tissues and/or organs are subsequently implanted back into the patient. In certain embodiments, an anti-CDl37 antibody can be delivered by implanting certain cells that have been genetically engineered, using methods such as those described herein, to express and secrete the polypeptides. In certain embodiments, such cells can be animal or human cells, and can be autologous, heterologous, or xenogeneic. In certain embodiments, the cells can be immortalized. In certain embodiments, in order to decrease the chance of an immunological response, the cells can be encapsulated to avoid infiltration of surrounding tissues. In certain embodiments, the encapsulation materials are typically biocompatible, semi-permeable polymeric enclosures or membranes that allow the release of the protein product(s) but prevent the destruction of the cells by the patient's immune system or by other detrimental factors from the surrounding tissues.

Applications

The compositions described herein can be used in diagnostic and therapeutic applications. For example, detectably-labeled antigen-binding molecules can be used in assays to detect the presence or amount of the target antigens in a sample (e.g., a biological sample). The compositions can be used in in vitro assays for studying inhibition of target antigen function (e.g. CD137- mediated cellular signaling or response). In some embodiments, e.g., in which the compositions bind to and activate a target antigen (e.g. a protein or polypeptide), the compositions can be used as positive controls in assays designed to identify additional novel compounds that also induce activity of the target protein or polypeptide and/or are otherwise are useful for treating a disorder associated with the target protein or polypeptide. For example, a CDl37-activating composition can be used as a positive control in an assay to identify additional compounds (e.g., small molecules, aptamers, or antibodies) that induce, increase, or stimulate CD 137 function. The compositions can also be used in therapeutic methods as elaborated on below.

Determination of Biomarkers

The presence of, an amount, or expression level of, one or more biomarkers described herein in a sample can be detected or determined by a number of methodologies and techniques, which are known in the art and understood by the skilled artisan, including, but not limited to, immunohistochemistry (IHC), immunofluorescence (IF), Western blot analysis, immunoprecipitation, molecular binding assays, enzyme-linked immunosorbent assay (ELISA), enzyme-linked immunofiltration assay (ELIFA), flow cytometry, MassARRAY, proteomics, quantitative blood based assays (e.g., Serum ELISA), biochemical enzymatic activity assays, in situ hybridization, fluorescence in situ hybridization (FISH), Southern analysis, Northern analysis, whole genome sequencing, polymerase chain reaction (PCR) including quantitative real time PCR (qRT-PCR) and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like, RNA-Seq, microarray analysis, gene expression profiling, and/or serial analysis of gene expression (SAGE), as well as any one of the wide variety of assays that can be performed by protein, gene, and/or tissue array analysis. Typical protocols for evaluating the status of genes and gene products are found, for example in Ausubel et ah, eds., 1 995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting) and 18 (PCR Analysis). Multiplexed immunoassays such as those available from Rules Based Medicine or Meso Scale Discovery ("MSD") may also be used.

Protein Biomarker Techniques

In some embodiments, the amount of a biomarker (e.g., major histocompatibility complex I (MHC I)) is measured by determining the protein expression level of the biomarker. There are a number of techniques that measure or determine protein expression levels known in the art and described herein that can be used in the methods provided by the disclosure. For example, in some embodiments, a protein expression level of a biomarker (e.g., MHC I) is determined using a method selected from the group consisting of flow cytometry (e.g., fluorescence-activated cell sorting (FACS™)), Western blot, enzyme-linked immunosorbent assay (ELISA), immunoprecipitation, immunohistochemistry (IHC), immunofluorescence, radioimmunoassay, dot blotting, immunodetection methods, HPLC, surface plasmon resonance, optical spectroscopy, mass spectrometry, and HPLC.

In some embodiments, a sample is contacted with an antibody that specifically binds to a biomarker (e.g., an anti-MHC I antibody) described herein under conditions permissive for binding of the biomarker, and the presence of a complex formed by the antibody and the biomarker is detected. In some embodiments, a sample is contacted with a combination of antibodies that specifically bind to a combination of biomarkers described herein. In some embodiments, the protein expression level of the biomarker (e.g., MHC I) is determined in tumor-infiltrating immune cells (e.g., tumor-infiltrating macrophages). In some embodiments, the protein expression level of a biomarker (e.g., MHC I) is determined in tumor cells. In some embodiments, the protein expression level of the biomarker is determined in tumor-infiltrating immune cells and in tumor cells.

In some embodiments, the amount of a biomarker protein (e.g., MHC I) in a sample is determined using IHC and staining protocols. IHC staining of tissue sections has been shown to be a reliable method of determining or detecting the presence of proteins in a sample. In some embodiments, the biomarker is selected from MHC I, beta-2-microglobulin chain (b2M), and MHC class I alpha chain. In some embodiments, expression level of a biomarker is determined by performing IHC analysis of a sample (e.g., a tumor sample obtained from a patient) with an antibody. In some embodiments, IHC staining intensity of the biomarker in a sample is determined relative to a reference sample. In some embodiments, the reference sample is obtained from a control cell line or non-malignant cell line, a tissue sample from non-cancerous patient, or a tumor sample that does not express or contain a biomarker described herein.

Two general methods of IHC are available; direct and indirect assays. The direct assay uses a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction. In contrast, an indirect assay includes an unconjugated primary antibody that binds to the antigen and a labeled secondary antibody that binds to the primary antibody. The secondary antibody is conjugated to an enzymatic label, a chromogenic or fluorogenic substrate to provide visualization of the antigen.

The primary and/or secondary antibody used for IHC typically will be labeled with a detectable moiety. Numerous labels are available which can be generally grouped into the following categories: (a) radioisotopes, such as 35S, 14C, 1251 , 3H, and 1311; (b) colloidal gold particles; (c) fluorescent labels including, but not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially- available fluorophores such as SPECTRUM ORANGE7 and SPECTRUM GREEN7 and/or derivatives of any one or more of the above; and various enzyme- substrate labels are available (see e.g. U.S. Patent No. 4,275,149 which is included herein by reference in its entirety). Examples of enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; see, e.g., U.S. Patent No. 4,737,456), luciferin, 2,3- dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, b-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), I acto peroxidase, microperoxidase, and the like.

Examples of enzyme- substrate combinations include, for example, horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate; alkaline phosphatase (AP) with para- Nitrophenyl phosphate as chromogenic substrate; and b-D-galactosidase (b-D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl-p-D-galactosidase) or fluorogenic substrate (e.g., 4- methylumbelliferyl-p- D-galactosidase). For a general review of these, see, for example, U.S. Patent Nos. 4,275,149 and 4,318,980.

In some embodiments, samples can be prepared for IHC analysis, for example, manually, or using an automated staining instrument (e.g., a Ventana BenchMark XT or Benchmark ULTRA instrument). Samples thus prepared can be mounted on slides and coverslipped. Slide evaluation is then determined, for example, using a microscope, and staining intensity criteria, routinely used in the art, can be employed. It is understood by one skilled in the art that when a sample from a tumor is analyzed using IHC, staining is generally determined or assessed in tumor cell(s) and/or tissue (as opposed to stromal or surrounding tissue that may be present in the sample). In some embodiments, when cells and/or tissue from a tumor are examined using IHC, staining includes determining or assessing in tumor-infiltrating immune cells, including intratumoral or peritumoral immune cells. In some embodiments, the presence of a biomarker (e.g., MHC I) is detected by IHC in >0% of the sample, for example, in at least 1 % of the sample, in at least 5% of the sample, in at least 10% of the sample, in at least 15% of the sample, in at least 15% of the sample, in at least 20% of the sample, in at least 25% of the sample, in at least 30% of the sample, in at least 35% of the sample, in at least 40% of the sample, in at least 45% of the sample, in at least 50% of the sample, in at least 55% of the sample, in at least 60% of the sample, in at least 65% of the sample, in at least 70% of the sample, in at least 75% of the sample, in at least 80% of the sample, in at least 85% of the sample, in at least 90% of the sample, in at least 95% of the sample, or more. Samples can be scored using any of the criteria described herein, for example, by a pathologist or automated image analysis.

In some embodiments, the presence of major histocompatibility complex I (MHC) in a sample is detected by the presence of any one or more markers selected from the group consisting of: MHC I, beta-2-microglobulin chain (b2M), and MHC class I alpha chain. In some embodiments, the presence of major histocompatibility complex I (MHC) in a sample is detected using an antibody that binds any one or more markers selected from the group consisting of: MHC I protein, beta-2-microglobulin chain (b2M) protein, and MHC class I alpha chain protein.

In some embodiments, the amount of MHC I protein in a sample is determined using an anti-MHC I diagnostic antibody. In some embodiments, the anti-MHC I diagnostic antibody specifically binds human MHC I. In some embodiments, the anti-MHC I diagnostic antibody is a non-human antibody. In some embodiments, the anti-MHC I diagnostic antibody is a rat, mouse, or rabbit antibody. In some embodiments, the anti-MHC I diagnostic antibody is a monoclonal antibody. In some embodiments, the anti-MHC I diagnostic antibody is directly labeled. In other embodiments, the anti-MHC I diagnostic antibody is indirectly labeled. Anti-MHC I diagnostic antibodies are available from a variety of commercial sources, such as BD Biosciences, ebiosciences, BioLegend, Abeam, and the like. In addition, one skilled in the art can make monoclonal antibodies suitable for detection based on the known sequences of MHC I molecules and techniques for generating monoclonal antibodies.

In some embodiments, the amount of beta-2-micro globulin chain (b2M) protein in a sample is determined using an anti^2M diagnostic antibody. In some embodiments, the anti^2M diagnostic antibody specifically binds human b2M. In some embodiments, the anti^2M diagnostic antibody is a non-human antibody. In some embodiments, the anti^2M diagnostic antibody is a rat, mouse, or rabbit antibody. In some embodiments, the anti^2M diagnostic antibody is a monoclonal antibody. In some embodiments, the anti^2M diagnostic antibody is directly labeled. In other instances, the anti^2M diagnostic antibody is indirectly labeled. Anti- b2M diagnostic antibodies are available from a variety of commercial sources, such as BD Biosciences, ebiosciences, BioLegend, Abeam, and the like. One skilled in the art can make monoclonal antibodies suitable for detection based on the known sequence of b2M and techniques for generating monoclonal antibodies. In some embodiments, the amino acid sequence of b2M that can be used for generating monoclonal antibodies is set forth in SEQ ID NO: 136.

In some embodiments, the amount of MHC class I alpha chain protein in a sample is determined using an anti-MHC class I alpha chain diagnostic antibody. In some embodiments, the anti-MHC class I alpha chain diagnostic antibody specifically binds human MHC class I alpha chain. In some embodiments, the anti-MHC class I alpha chain diagnostic antibody is a non-human antibody. In some embodiments, the anti-MHC class I alpha chain diagnostic antibody is a rat, mouse, or rabbit antibody. In some embodiments, the anti-MHC class I alpha chain diagnostic antibody is a monoclonal antibody. In some embodiments, the anti-MHC class I alpha chain diagnostic antibody is directly labeled. In other embodiments, the anti-MHC class I alpha chain diagnostic antibody is indirectly labeled. In some embodiments, the anti-MHC class I alpha chain antibody is Human HLA Class I antibody from R& D Systems (#MAB7098). Other anti-Class I alpha chain protein diagnostic antibodies are available from a variety of commercial sources, such as BD Biosciences, ebiosciences, BioLegend, Abeam, and the like. One skilled in the art can make monoclonal antibodies suitable for detection based on the known sequence of MHC class I alpha chain and techniques for generating monoclonal antibodies. In some embodiments, the amino acid sequence of human HLA-A that can be used for generating monoclonal antibodies is set forth in SEQ ID NO: 134.

In some embodiments, the expression level of a biomarker described herein is detected in tumor-infiltrating immune cells, tumor cells, or combinations thereof using IHC. Tumor- infiltrating immune cells include, but are not limited to, intratumoral immune cells, peritumoral immune cells or any combinations thereof, and other tumor stroma cells (e.g., fibroblasts). Such tumor infiltrating immune cells may be T lymphocytes (such as CD8+ T lymphocytes and/or CD4+ T lymphocytes), B lymphocytes, or other bone marrow-lineage cells including granulocytes (neutrophils, eosinophils, basophils), monocytes, macrophages, dendritic cells (e.g., interdigitating dendritic cells), histiocytes, and natural killer cells. In some embodiments, the staining for a biomarker, described herein, is detected as membrane staining, cytoplasmic staining and combinations thereof. In some embodiments, the absence of a biomarker, described herein, is detected as absent or no staining in the sample.

Nucleic Acid Biomarker Techniques

In some embodiments, the expression level of a biomarker, described herein, can be a nucleic acid expression level. In some embodiments, the nucleic acid expression level is determined using qPCR, rtPCR, RNA-seq, multiplex qPCR or RT-qPCR, microarray analysis, SAGE, MassARRAY technique, or in situ hybridization (e.g., FISH). In some embodiments, the expression level of a biomarker (e.g., b2M) is determined in tumor cells, tumor infiltrating immune cells, stromal cells, or combinations thereof. In some embodiments, the expression level of a biomarker described herein is determined in tumor-infiltrating immune cells. In some embodiments, the expression level of a biomarker described herein is determined in tumor cells.

Methods for the evaluation of mRNAs in cells are known in the art and include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled riboprobes specific for the one or more genes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for one or more of the genes, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like). In addition, such methods can include one or more steps that allow one to determine the levels of target mRNA in a biological sample (e.g., by simultaneously examining the levels a comparative control mRNA sequence of a "housekeeping" gene such as an actin family member).

In some embodiments, a nucleic acid probe is selected using techniques known in the art. In some embodiments, a nucleic acid probe is selected using specific criteria. In some

embodiments, the criteria include the length of a probe, Tm of a probe (e.g., the temperature at which 50% of double stranded DNA molecules are dissociated into two single stranded DNAs), and a threshold value of the sequence homology with other sequences. After a candidate nucleic acid probe meeting the criteria has been selected, the candidate nucleic acid probe is examined with regard to Tm and sequence homology to determine whether or not the candidate nucleic acid probe is unique to a target sequence and whether or not the candidate nucleic acid probe is a sequence that can easily induce cross hybridization. A preferable nucleic acid probe among the candidate nucleic acid probes meeting the criteria is selected as a sequence that specifically hybridizes to a target sequence.

In some embodiments, the amount of MHC I mRNA in a sample is determined using a nucleic acid probe that hybridizes with MHC I mRNA. In some embodiments, the nucleic acid probe that hybridizes with MHC I mRNA is directly labeled. In other embodiments, the nucleic acid probe that hybridizes with MHC I mRNA is indirectly labeled. In some embodiments, the nucleic acid probe that hybridizes with MHC I mRNA is radioactively labeled. In some embodiments, the nucleic acid probe that hybridizes with MHC I mRNA is fluorescently labeled. One skilled in the art can make nucleic acid probes that hybridize with MHC I mRNA suitable for detection based on the known sequence of MHC I mRNA and techniques for nucleic acid probes. In some embodiments, the amount of b2M chain mRNA in a sample is determined using a nucleic acid probe that hybridizes with b2M chain mRNA. In some embodiments, the nucleic acid probe that hybridizes with b2M chain mRNA is directly labeled. In other embodiments, the nucleic acid probe that hybridizes with b2M chain mRNA is indirectly labeled. In some embodiments, the nucleic acid probe that hybridizes with b2M chain mRNA is radioactively labeled. In some embodiments, the nucleic acid probe that hybridizes with b2M chain mRNA is fluorescently labeled. One skilled in the art can make nucleic acid probe that hybridizes with b2M chain mRNA suitable for detection based on the known sequence of b2M chain mRNA and techniques for nucleic acid probes. In some embodiments, the nucleotide sequence of b2M that can be used for generating nucleic acid probes is set forth in SEQ ID NO: 137.

In some embodiments, the amount of MHC class I alpha chain mRNA in a sample is determined using a nucleic acid probe that hybridizes with MHC class I alpha chain mRNA. In some embodiments, the nucleic acid probe that hybridizes with MHC class I alpha chain mRNA is directly labeled. In other embodiments, the nucleic acid probe that hybridizes with MHC class I alpha chain mRNA is indirectly labeled. In some embodiments, the nucleic acid probe that hybridizes with MHC class I alpha chain mRNA is radioactively labeled. In some embodiments, the nucleic acid probe that hybridizes with MHC class I alpha chain mRNA is fluorescently labeled. One skilled in the art can make nucleic acid probe that hybridizes with MHC class I alpha chain mRNA suitable for detection based on the known sequence of MHC class I alpha chain mRNA and techniques for nucleic acid probes. In some embodiments, the cDNA sequence of human HLA-A that can be used for generating nucleic acid probes is set forth in SEQ ID NO: 135.

In some embodiments, the sequence of the amplified target cDNA can be determined. Methods include protocols which examine or detect mRNAs, such as target mRNAs, in a tissue or cell sample by microarray technologies. Using nucleic acid microarrays, test and control mRNA samples from test and control tissue samples are reverse transcribed and labeled to generate cDNA probes. The probes are then hybridized to an array of nucleic acids immobilized on a solid support. The array is configured such that the sequence and position of each member of the array is known. For example, a selection of genes whose expression correlates with increased or reduced clinical benefit of treatment comprising an agent that specifically binds to b2M, wherein the agent comprises an Fc domain, can be arrayed on a solid support. Hybridization of a labeled probe with a particular array member indicates that the sample from which the probe was derived expresses that gene.

Inclusion of any of the diagnostic methods described herein as part of any method directed to methods for identifying patients likely to benefit from treatment as described herein (e.g., selection of a therapeutic treatment or intervention) or to the development of treatments (e.g., enrollment patients in clinical trials) provides an advantage over those methods that do not include the diagnostic methods, in that a patient population whose members are predicted to need and/or not need, benefit, or respond to treatment can be identified.

Biomarkers can be a substance or a biological event whose detection indicates a particular physiological state (e.g., a diseased state). For example, the presence of an antibody in the serum of a patient may indicate an infection. Biomarkers measured in patients before treatment can be used to identify suitable patients for inclusion in a clinical trial. Biomarker changes after treatment may predict or identify safety problems related to a candidate drug, or reveal pharmacologic activity expected to predict an eventual benefit of treatment. Biomarkers can reduce uncertainty in drug development and evaluation by providing quantifiable predictions about drug performance, and they can contribute to dose selection. Composite biomarkers include several individual biomarkers in a stated algorithm which reaches a single interpretive readout when a single biomarker fails to provide all the relevant information required for assessment.

A surrogate endpoint is a biomarker that is intended to substitute for a clinical end point and is expected, based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence, to predict clinical benefit.

Samples

In some embodiments of the methods provided by the disclosure, a sample includes any relevant biological sample (e.g., a tumor sample) suitable for use in the methods provided by the disclosure (e.g., sections of tissues such as biopsy or tissue removed during surgical or other procedures, autopsy samples, and frozen sections taken for histological purposes). Samples can be derived or obtained from blood and blood fractions or products (e.g., serum, buffy coat, plasma, platelets, red blood cells, peripheral blood mononuclear cells (PBMC)), sputum, cheek cells, cultured cells (e.g., primary cultures, explants, and transformed cells), stool, urine, other biological or bodily fluids (e.g., prostatic fluid, gastric fluid, intestinal fluid, renal fluid, lung fluid, cerebrospinal fluid, pleural and abdominal effusions, and the like), etc. A sample can be processed according to techniques understood by those in the art. A sample can be without limitation fresh, frozen or fixed. In some embodiments, a sample is obtained from a tumor or malignant tissue. In some embodiments, a sample comprises malignant and non-malignant cells. In some embodiments, a sample comprises formalin-fixed paraffin-embedded (FFPE) tissue or fresh frozen (FF) tissue. In some embodiments, a sample comprises cultured cells, including primary or immortalized cell lines derived from a subject. In some embodiments, a sample refer to an extract from a sample from a subject. For example, in some embodiments, a sample comprises DNA, RNA or protein extracted from a tissue or a bodily fluid. Many techniques and commercial kits are available for such purposes. In some embodiments, a fresh sample from the individual is treated with an agent to preserve RNA prior to further processing, e.g., cell lysis and extraction. Samples can include frozen samples collected for other purposes. In some embodiments, samples are associated with information such as age, gender, and clinical symptoms present in the subject from which the sample is obtained; source or location of the sample; and information regarding methods of collection, treatment, and/or storage of the sample.

In some embodiments, a sample is obtained from a biopsy. A biopsy comprises the process of removing a tissue sample for diagnostic or prognostic evaluation. Any biopsy technique known in the art can be applied to the methods provided by the disclosure. The biopsy technique applied can depend on the tissue type to be evaluated (e.g., colon, prostate, kidney, bladder, lymph node, liver, bone marrow, blood cell, lung, breast, etc.), the size and type of the tumor (e.g., solid or suspended, blood or ascites), among other factors. Representative biopsy techniques include, but are not limited to, excisional biopsy, incisional biopsy, needle biopsy, surgical biopsy, liquid biopsy and bone marrow biopsy. In some embodiments, a sample is obtained from an“excisional biopsy”, which refers to the removal of an entire tumor mass with a small margin of normal tissue surrounding it. An“incisional biopsy” refers to the removal of a wedge of tissue that includes a cross-sectional diameter of the tumor. In some embodiments, a sample is obtained from a“core needle biopsy” of the tumor mass, or a“fine-needle aspiration biopsy” which generally obtains a suspension of cells from within the tumor mass. Biopsy techniques are discussed, for example, in Harrison's Principles of Internal Medicine, Kasper, et ah, eds., l6th ed., 2005, Chapter 70, and throughout Part V. In some embodiments, the amount of a biomarker, described herein, in a first sample is increased or elevated as compared to amount in a second sample. In some embodiments, the amount of a biomarker, described herein, in a first sample is decreased or reduced as compared to amount in a second sample. In some embodiments, the second sample is a reference sample. Additional disclosure for determining the amount of a biomarker are described herein.

In some embodiments, a reference sample is a single sample or a combination of multiple samples from the same subject that are obtained at one or more different time points than when the test sample is obtained. For example, a reference sample may be obtained at an earlier time point from the same subject than when the test sample is obtained. Such reference sample can be useful if the reference sample is obtained during initial diagnosis of cancer and the test sample is later obtained when the cancer has progressed (e.g., becomes metastatic).

In some embodiments, a reference sample is a combination of multiple samples from one or more healthy subjects who are not the subject being tested. In some embodiments, a reference sample is a combination of multiple samples from one or more subjects with a disease or disorder (e.g., cancer) who are not the subject being tested. In some embodiments, a reference sample is pooled RNA samples from normal tissues or pooled plasma or serum samples from one or more subjects who are not the subject being tested. In some embodiments, a reference sample is pooled RNA samples from tumor tissues or pooled plasma or serum samples from one or more subjects with a disease or disorder (e.g., cancer) who are not the subject being tested. In some embodiments, elevated or increased expression refers to an overall increase of about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or greater, in the level of a biomarker (e.g., protein or nucleic acid (e.g., gene or mRNA)), detected by standard art-known methods such as those described herein, as compared to a reference sample. In some embodiments, the elevated expression refers to the increase in expression level/amount of a biomarker in the sample wherein the increase is at least about any of l.5x, l.75x, 2x, 3x, 4x, 5x, 6x, 7x, 8x, 9x, lOx, 25x, 50x, 75x, or lOOx the expression level/amount of the respective biomarker in a reference sample. In some embodiments, elevated expression refers to an overall increase of greater than about 1.5-fold, about 1.75-fold, about 2- fold, about 2.25-fold, about 2.5-fold, about 2.75-fold, about 3.0-fold, or about 3.25-fold as compared to a reference sample.

In some embodiments, reduced expression refers to an overall reduction of about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99% or greater, in the level of biomarker (e.g., protein or nucleic acid (e.g., gene or mRNA)) in a sample, as compared to a reference sample. In certain embodiments, reduced expression refers to the decrease in amount of a biomarker in the sample, wherein the decrease is about 0.9x, about 0.8x, about 0.7x, about 0.6x, about 0.5x, about 0.4x, about 0.3x, about 0.2x, about O.lx, about 0.05x, or about O.Olx the amount of a biomarker in the reference sample.

Modification of Detection Agents

Agents that specifically bind markers for the detection of MHC I expression (e.g., markers including MHC I, beta-2-microglobulin chain (b2M), MHC class I alpha chain), can be modified following their expression and purification.

In some embodiments, the modified agent is an antibody. In some embodiments, the modified agent is a polypeptide comprising an Fc domain. The modifications can be covalent or noncovalent modifications. Such modifications can be introduced into the antibodies or fragments by, e.g., reacting targeted amino acid residues of the polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues. Suitable sites for modification can be chosen using any of a variety of criteria including, e.g., structural analysis or amino acid sequence analysis of the antibodies or fragments.

In some embodiments, the modified agent is a nucleic acid probe. In some embodiments, nucleic acids are labeled at their 5’ end. In some embodiments, nucleic acids are labeled at their 3” end. In some embodiments, nucleic acids are labeled throughout the molecule. In some embodiments, the labeling of nucleic acids throughout the molecule is accomplished by nick translation, random priming, PCT, or in vitro transcription using labeled dNTPs or NTPs. In some embodiments, the nucleic acids are end labeled by end-labeling protocols or PCT using primers bearing the required modification. In some embodiments, nucleic acid probes are enzymatically labeled. In some embodiments, nucleic acid probes are chemically labeled. In some embodiments, nucleic acid probes are radioactively labeled. In some embodiments, nucleic acid probes are fluorescently labeled. In some embodiments, nucleic acid probes are modified with biotin. In some embodiments, nucleic acid probes are modified with digoxygenin.

In some embodiments, the agents that specifically bind markers for the detection of MHC I expression (e.g., MHC I, beta-2-microglobulin chain (b2M), MHC class I alpha chain), can be conjugated to a heterologous moiety. In some embodiments, the agent conjugated to a heterologous moiety is an antibody. In some embodiments, the agent conjugated to a heterologous moiety is a polypeptide comprising an Fc domain. In some embodiments, the agent conjugated to a heterologous moiety is a nucleic acid probe. The heterologous moiety can be, e.g., a heterologous polypeptide, a therapeutic agent (e.g., a toxin or a drug), or a detectable label such as, but not limited to, a radioactive label, an enzymatic label, a fluorescent label, a heavy metal label, a luminescent label, or an affinity tag such as biotin or streptavidin. Suitable heterologous polypeptides include, e.g., an antigenic tag (e.g., FLAG (DYKDDDDK (SEQ ID NO: 98)), polyhistidine (6-His; HHHHHH (SEQ ID NO: 99), hemagglutinin (HA; YPYDVPDYA (SEQ ID NO: 100)), glutathione-S-transferase (GST), or maltose-binding protein (MBP)) for use in purifying the antibodies or fragments. Heterologous polypeptides also include polypeptides (e.g., enzymes) that are useful as diagnostic or detectable markers, for example, luciferase, a fluorescent protein (e.g., green fluorescent protein (GFP)), or chloramphenicol acetyl transferase (CAT). Suitable radioactive labels include, e.g., 32P, 33P, 14C, 1251, 1311, 35S, and 3H. Suitable fluorescent labels include, without limitation, fluorescein, fluorescein isothiocyanate (FITC), green fluorescent protein (GFP), DYLIGHT™ 488, phycoerythrin (PE), propidium iodide (PI), PerCP, PE-ALEXA FLUOR® 700, Cy5, allophycocyanin, and Cy7. Luminescent labels include, e.g., any of a variety of luminescent lanthanide (e.g., europium or terbium) chelates. For example, suitable europium chelates include the europium chelate of diethylene triamine pentaacetic acid (DTPA) or tetraazacyclododecane-l,4,7,l0-tetraacetic acid (DOTA). Enzymatic labels include, e.g., alkaline phosphatase, CAT, luciferase, and horseradish peroxidase.

Two proteins (e.g., an antibody and a heterologous moiety) can be crosslinked using any of a number of known chemical cross linkers. Examples of such cross linkers are those which link two amino acid residues via a linkage that includes a“hindered” disulfide bond. In these linkages, a disulfide bond within the cross-linking unit is protected (by hindering groups on either side of the disulfide bond) from reduction by the action, for example, of reduced glutathione or the enzyme disulfide reductase. One suitable reagent, 4-succinimidyloxycarbonyl-a-methyl-a(2-pyridyldithio) toluene (SMPT), forms such a linkage between two proteins utilizing a terminal lysine on one of the proteins and a terminal cysteine on the other. Heterobifunctional reagents that cross-link by a different coupling moiety on each protein can also be used. Other useful cross-linkers include, without limitation, reagents which link two amino groups (e.g., N-5-azido-2- nitrobenzoyloxysuccinimide), two sulfhydryl groups (e.g., l,4-bis-maleimidobutane), an amino group and a sulfhydryl group (e.g., mmaleimidobenzoyl-N-hydroxysuccinimide ester), an amino group and a carboxyl group (e.g., 4-[p-azidosalicylamido]butylamine), and an amino group and a guanidinium group that is present in the side chain of arginine (e.g., p-azidophenyl glyoxal monohydrate).

In some embodiments, a radioactive label can be directly conjugated to the amino acid backbone of the agent. In some embodiments, a radioactive label can be directly conjugated to the nucleic acid sequence of the agent. Alternatively, the radioactive label can be included as part of a larger molecule (e.g., 1251 in meta-[l25I]iodophenyl-N-hydroxysuccinimide ([l25I]mIPNHS) which binds to free amino groups to form meta-iodophenyl (mIP) derivatives of relevant proteins (see, e.g., Rogers et al. (1997) J Nucl Med 38: 1221-1229) or chelate (e.g., to DOTA or DTPA) which is in turn bound to the protein backbone or the nucleic acid sequence. Methods of conjugating the radioactive labels or larger molecules/chelates containing them to the antibodies, polypeptides comprising an Fc domain, or nucleic acid probes described herein are known in the art. Such methods involve incubating the proteins with the radioactive label under conditions (e.g., pH, salt concentration, and/or temperature) that facilitate binding of the radioactive label or chelate to the protein (see, e.g., U.S. Patent No. 6,001,329).

Methods for conjugating a fluorescent label (sometimes referred to as a“fluorophore”) to a protein (e.g., an antibody) are known in the art. For example, fluorophores can be conjugated to free amino groups (e.g., of lysines) or sulfhydryl groups (e.g., cysteines) of proteins using succinimidyl (NHS) ester or tetrafluorophenyl (TFP) ester moieties attached to the fluorophores. In some embodiments, the fluorophores can be conjugated to a heterobifunctional cross-linker moiety such as sulfo-SMCC. Suitable conjugation methods involve incubating an antibody protein, or fragment thereof, with the fluorophore under conditions that facilitate binding of the fluorophore to the protein. See, e.g., Welch and Redvanly (2003) “Handbook of Radiopharmaceuticals: Radiochemistry and Applications,” John Wiley and Sons (ISBN 0471495603). Methods for conjugating a fluorescent label (sometimes referred to as a “fluorophore”) to a nucleic acid (e.g., a nucleic acid probe) are known in the art. Kits

In some embodiments, the disclosure provides a kit comprising an anti-CD 137 antibody described herein. In some embodiments, a kit includes an anti-CD 137 antibody as disclosed herein, and instructions for use. The kits may comprise, in a suitable container, an anti-CD 137 antibody, one or more controls, and various buffers, reagents, enzymes and other standard ingredients well known in the art.

The container can include at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which an anti-CD 137 antibody may be placed, and in some instances, suitably aliquoted. Where an additional component is provided, the kit can contain additional containers into which this component may be placed. The kits can also include a means for containing an anti-CDl37 antibody and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained. Containers and/or kits can include labeling with instructions for use and/or warnings.

In some embodiments, a kit comprises a container comprising an anti-CDl37 antibody and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the anti- CD 137 antibody, and instructions for treating or delaying progression of cancer or reducing or inhibiting tumor growth in a subject in need thereof. In some embodiments, a kit comprises a container comprising an anti-CD 137 antibody and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the anti-CD 137 antibody, and instructions for administering the anti-CD 137 antibody to a subject in need thereof, alone or in combination with another agent, for treating or delaying progression of cancer or reducing or inhibiting tumor growth in the subject.

The disclosure also provides kits comprising a container with one or more reagents for detecting the presence of major histocompatibility complex I (MHC I) in a tumor sample, and a package insert comprising instructions for determining an amount or expression level of MHC I in the tumor sample. In some embodiments, the kit comprises a reagent for detecting the presence of MHC I protein in the sample and instructions for determining an amount or expression level of MHC I protein in the tumor sample. In some embodiments, the kit comprises a reagent for detecting the presence of beta-2-microglobulin chain (b2M) protein in the sample and instructions for determining an amount or expression level of beta-2-microglobulin chain (b2M) protein in the tumor sample. In some embodiments, the kit comprises a reagent for detecting the presence of MHC class I alpha chain protein in the sample and instructions for determining an amount or expression level of MHC class I alpha chain protein in the tumor sample. In other embodiments, the kit comprises a reagent for detecting the presence of one or more polypeptides in the sample selected from the group consisting of MHC I, b2M, MHC class I alpha chain, or a combination thereof, and instructions for determining an amount or expression level of the one or more polypeptides in the tumor sample.

In some embodiments, the kits can comprise, in a suitable container, an agent that specifically binds one or more of MHC I protein, b2M protein, and MHC class I alpha chain protein, or a combination thereof, wherein the agent comprises an antibody, one or more controls, and various buffers, reagents, enzymes and other standard ingredients well known in the art. In some embodiments, a kit with an agent that specifically binds one or more of MHC I protein, b2M protein, and MHC class I alpha chain protein, wherein the agent comprises an antibody, includes instructions for determining an amount or expression level of the one or more polypeptides in the tumor sample.

In other embodiments, the kit comprises an anti-tumor agent (e.g., an anti-CD 137 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis. In some embodiments, a kit comprises an anti-tumor agent (e.g., an anti-CD 137 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis, and further comprises an agent that specifically binds MHC I protein, wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and includes instructions for detecting expression of MHC I protein.

In some embodiments, a kit comprises an anti-tumor agent (e.g., an anti-CDl37 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis, and further comprises an agent that specifically binds b2M protein, wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and includes instructions for detecting expression of b2M protein.

In some embodiments, a kit comprises an anti-tumor agent (e.g., an anti-CDl37 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis, and further comprises an agent that specifically binds MHC class I alpha chain protein, wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and includes instructions for detecting expression of MHC class I alpha chain mRNA.

In some aspects, a kit includes a composition comprising an agent that specifically binds CD 137 (e.g., an anti-CD 137 antibody of the disclosure), wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and an optional pharmaceutically acceptable carrier, for use in treating or delaying progression of cancer in a subject comprising MHC I expressing malignant cells, wherein the treatment comprises administration of the agent. In some embodiments, a kit described herein includes instructions for detecting expression of MHC I, diagnosing a patient and selecting a treatment.

In some embodiments, the disclosure provides a kit comprising a container which includes at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which an agent that specifically binds MHC I protein, b2M protein, or MHC class I alpha chain protein, or a combination thereof, described herein can be placed, and in some instances, suitably aliquoted. Where an additional component is provided, the kit can contain additional containers into which this component may be placed. The kits can also include a means for containing an anti-tumor agent as described herein and any other reagent containers in close confinement for commercial sale. Such containers can include injection or blowmolded plastic containers into which the desired vials are retained. Containers and/or kits can include labeling with instructions for use and/or warnings.

In some aspects, the disclosure provides a kit comprising a medicament comprising a composition comprising an agent that specifically binds binds CD137 (e.g., an anti-CDl37 antibody of the disclosure), wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for detection of MHC I protein expressing malignant cells, and, optionally, b2M protein, MHC class I alpha chain protein, or a combination thereof, and administration of the medicament, for treating or delaying progression of cancer in a subject.

In some aspects, the disclosure provides a kit comprising a container comprising a composition comprising an agent that specifically binds CD 137 (e.g., an anti-CD 137 antibody of the disclosure), wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for detection of MHC I protein expressing malignant cells and, optionally, b2M protein, MHC class I alpha chain protein, or a combination thereof, and administration of the composition, for treating or delaying progression of cancer in a subject.

In some embodiments, the kits described herein comprise an assay for detecting MHC I protein expression on malignant cells.

In some embodiments, the kit comprises a reagent for detecting the presence of MHC I mRNA in the sample and instructions for determining an mRNA levels of MHC I mRNA in the tumor sample. In some embodiments, the kit comprises a reagent for detecting the presence of b2M mRNA in the sample and instructions for determining mRNA levels of b2M mRNA in the tumor sample. In some embodiments, the kit comprises a reagent for detecting the presence of MHC class I alpha chain mRNA in the sample and instructions for determining mRNA levels of MHC class I alpha chain mRNA in the tumor sample. In other aspects, the kit comprises a reagent for detecting the presence of one or more mRNAs in the sample selected from the group consisting of MHC I mRNA, b2M mRNA, MHC class I alpha chain mRNA, or a combination thereof, and instructions for detecting mRNA levels of the one or more mRNAs in the tumor sample.

In some aspects, the kit can comprise, in a suitable container, an agent that specifically binds one or more of MHC I mRNA, b2M mRNA, and MHC class I alpha chain mRNA, or a combination thereof, wherein the agent comprises a nucleic acid probe, one or more controls, and various buffers, reagents, enzymes and other standard ingredients well known in the art. In some embodiments, a kit comprising an agent that specifically binds one or more of MHC I mRNA, b2M mRNA, and MHC class I alpha chain mRNA, wherein the agent comprises a nucleic acid probe, includes instructions for detecting mRNA levels of the one or more mRNAs in the tumor sample.

In other aspects, the kit comprises an anti-tumor agent (e.g., an anti-CDl37 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis. In some embodiments, a kit comprises an anti-tumor agent (e.g., an anti-CD 137 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis, and further comprises an agent that specifically binds MHC I mRNA, wherein the agent comprises a nucleic acid probe, described herein, and includes instructions for detecting expression of MHC I mRNA. In some embodiments, a kit comprises an anti-tumor agent (e.g., an anti-CDl37 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis, and further comprises an agent that specifically binds b2M mRNA, wherein the agent comprises a nucleic acid probe, described herein, and includes instructions for detecting expression of b2M mRNA.

In some embodiments, a kit comprises an anti-tumor agent (e.g., an anti-CDl37 antibody of the disclosure) as described herein and instructions for use to treat or delay progression of cancer in a subject following diagnosis, and further comprises an agent that specifically binds MHC class I alpha chain mRNA, wherein the agent comprises a nucleic acid probe, described herein, and includes instructions for detecting expression of MHC class I alpha chain mRNA.

In some aspects, a kit includes a composition comprising an agent that specifically binds CD 137 (e.g., an anti-CD 137 antibody of the disclosure), wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and an optional pharmaceutically acceptable carrier, for use in treating or delaying progression of cancer in a subject comprising MHC I expressing malignant cells, wherein the treatment comprises administration of the agent. In some embodiments, a kit described herein includes instructions for detecting expression of MHC I, diagnosing a patient and selecting a treatment.

In some aspects, the disclosure provides a kit comprising a container which includes at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which an agent that specifically binds MHC I mRNA, b2M mRNA, or MHC class I alpha chain mRNA, or a combination thereof, described herein can be placed, and in some instances, suitably aliquoted. Where an additional component is provided, the kit can contain additional containers into which this component may be placed. The kits can also include a means for containing an anti-tumor agent as described herein and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blowmolded plastic containers into which the desired vials are retained. Containers and/or kits can include labeling with instructions for use and/or warnings.

In some aspects, the disclosure provides a kit comprising a medicament comprising a composition comprising an agent that specifically binds CD 137 (e.g., an anti-CD 137 antibody of the disclosure), wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for detection of MHC I mRNA expressing malignant cells, and, optionally, b2M mRNA, MHC class I alpha chain mRNA, or a combination thereof, and administration of the medicament, for treating or delaying progression of cancer in a subject.

In some aspects, the disclosure provides a kit comprising a container comprising a composition comprising an agent that specifically binds CD 137 (e.g., an anti-CD 137 antibody of the disclosure), wherein the agent comprises an antibody or polypeptide comprising an Fc domain, described herein, and an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for detection of MHC I mRNA expressing malignant cells and, optionally, b2M mRNA, MHC class I alpha chain mRNA, or a combination thereof, and administration of the composition, for treating or delaying progression of cancer in a subject.

In some embodiments, the kits described herein comprise an assay for detecting MHC I mRNA expression on malignant cells.

Methods of Use

The compositions of the present invention have numerous in vitro and in vivo utilities involving the detection and/or quantification of CD 137 and/or the agonism of CD 137 function.

The above-described compositions are useful in, inter alia, methods for treating or preventing a variety of cancers in a subject, particularly cancers that express MHC I, such as colon cancer. The compositions can be administered to a subject, e.g., a human subject, using a variety of methods that depend, in part, on the route of administration. The route can be, e.g., intravenous injection or infusion (IV), subcutaneous injection (SC), intraperitoneal (IP) injection, intramuscular injection (IM), or intrathecal injection (IT). The injection can be in a bolus or a continuous infusion.

Administration can be achieved by, e.g., local infusion, injection, or by means of an implant. The implant can be of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. The implant can be configured for sustained or periodic release of the composition to the subject. See, e.g., U.S. Patent Application Publication No. 20080241223; U.S. Patent Nos. 5,501,856; 4,863,457; and 3,710,795; EP488401; and EP 430539, the disclosures of each of which are incorporated herein by reference in their entirety. The composition can be delivered to the subject by way of an implantable device based on, e.g., diffusive, erodible, or convective systems, e.g., osmotic pumps, biodegradable implants, electrodiffusion systems, electroosmosis systems, vapor pressure pumps, electrolytic pumps, effervescent pumps, piezoelectric pumps, erosion-based systems, or electromechanical systems.

In some embodiments, an anti-CDl37 antibody or antigen-binding fragment thereof is therapeutically delivered to a subject by way of local administration.

A suitable dose of an antibody or fragment thereof described herein, which dose is capable of treating or preventing cancer in a subject, can depend on a variety of factors including, e.g., the age, sex, and weight of a subject to be treated and the particular inhibitor compound used. For example, a different dose of a whole anti-CD 137 antibody may be required to treat a subject with cancer as compared to the dose of a CDl37-binding Fab’ antibody fragment required to treat the same subject. Other factors affecting the dose administered to the subject include, e.g., the type or severity of the cancer. For example, a subject having metastatic melanoma may require administration of a different dosage of an anti-CD 137 antibody than a subject with glioblastoma. Other factors can include, e.g., other medical disorders concurrently or previously affecting the subject, the general health of the subject, the genetic disposition of the subject, diet, time of administration, rate of excretion, drug combination, and any other additional therapeutics that are administered to the subject. It should also be understood that a specific dosage and treatment regimen for any particular subject will also depend upon the judgment of the treating medical practitioner (e.g., doctor or nurse). Suitable dosages are described herein. In some embodiments, the anti-CDl37 antibodies described herein are effective at both high and low doses.

A pharmaceutical composition can include a therapeutically effective amount of an anti- CD 137 antibody or antigen-binding fragment thereof described herein. Such effective amounts can be readily determined by one of ordinary skill in the art based, in part, on the effect of the administered antibody, or the combinatorial effect of the antibody and one or more additional active agents, if more than one agent is used. A therapeutically effective amount of an antibody or fragment thereof described herein can also vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody (and one or more additional active agents) to elicit a desired response in the individual, e.g., reduction in tumor growth. For example, a therapeutically effective amount of an anti-CDl37 antibody can inhibit (lessen the severity of or eliminate the occurrence of) and/or prevent a particular disorder, and/or any one of the symptoms of the particular disorder known in the art or described herein. A therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.

Suitable human doses of any of the antibodies or fragments thereof described herein can further be evaluated in, e.g., Phase I dose escalation studies. See, e.g., van Gurp et al. (2008) Am J Transplantation 8(8): 1711-1718; Hanouska et al. (2007) Clin Cancer Res 13(2, part l):523-53 l; and Hetherington et al. (2006) Antimicrobial Agents and Chemotherapy 50(10): 3499-3500.

In some embodiments, the composition contains any of the antibodies or antigen-binding fragments thereof described herein and one or more (e.g., two, three, four, five, six, seven, eight, nine, 10, or 11 or more) additional therapeutic agents such that the composition as a whole is therapeutically effective. For example, a composition can contain an anti-CD 137 antibody described herein and an alkylating agent, wherein the antibody and agent are each at a concentration that when combined are therapeutically effective for treating or preventing a cancer (e.g., melanoma) in a subject.

Toxicity and therapeutic efficacy of such compositions can be determined by known pharmaceutical procedures in cell cultures or experimental animals (e.g., animal models of any of the cancers described herein). These procedures can be used, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. An antibody or antigen-binding fragment thereof that exhibits a high therapeutic index is preferred. While compositions that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue and to minimize potential damage to normal cells and, thereby, reduce side effects.

The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such antibodies or antigen-binding fragments thereof lies generally within a range of circulating concentrations of the antibodies or fragments that include the ED 50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For an anti- CD 137 antibody described herein, the therapeutically effective dose can be estimated initially from cell culture assays. A dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the EC 50 (i.e., the concentration of the antibody which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. In some embodiments, e.g., where local administration (e.g., to the eye or a joint) is desired, cell culture or animal modeling can be used to determine a dose required to achieve a therapeutically effective concentration within the local site.

In some embodiments, the methods can be performed in conjunction with other therapies for cancer. For example, the composition can be administered to a subject at the same time, prior to, or after, radiation, surgery, targeted or cytotoxic chemotherapy, chemoradiotherapy, hormone therapy, immunotherapy, gene therapy, cell transplant therapy, precision medicine, genome editing therapy, or other pharmacotherapy.

As described above, the compositions described herein (e.g., anti-CD 137 compositions) can be used to treat a variety of cancers such as but not limited to: Kaposi's sarcoma, leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, myeloblasts promyelocyte myelomonocytic monocytic erythroleukemia, chronic leukemia, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, mantle cell lymphoma, primary central nervous system lymphoma, Burkitt’s lymphoma and marginal zone B cell lymphoma, Polycythemia vera Lymphoma, Hodgkin's disease, non-Hodgkin' s disease, multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, solid tumors, sarcomas, and carcinomas, fibrosarcoma, myxosarcoma, liposarcoma, chrondrosarcoma, osteogenic sarcoma, osteosarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangio sarcoma, lymphangioendothelio sarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon sarcoma, colorectal carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, non- small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, retinoblastoma, nasopharyngeal carcinoma, esophageal carcinoma, basal cell carcinoma, biliary tract cancer, bladder cancer, bone cancer, brain and central nervous system (CNS) cancer, cervical cancer, choriocarcinoma, colorectal cancers, connective tissue cancer, cancer of the digestive system, endometrial cancer, esophageal cancer, eye cancer, head and neck cancer, gastric cancer, intraepithelial neoplasm, kidney cancer, larynx cancer, liver cancer, lung cancer (small cell, large cell), melanoma, neuroblastoma; oral cavity cancer (for example lip, tongue, mouth and pharynx), ovarian cancer, pancreatic cancer, retinoblastoma, rhabdomyosarcoma, rectal cancer; cancer of the respiratory system, sarcoma, skin cancer, stomach cancer, testicular cancer, thyroid cancer, uterine cancer, and cancer of the urinary system.

In some embodiments, an anti-CD 137 antibody or an antigen-binding fragment thereof described herein can be administered to a subject as a monotherapy. Alternatively, as described above, the antibody or fragment thereof can be administered to a subject as a combination therapy with another treatment, e.g., another treatment for a cancer. For example, the combination therapy can include administering to the subject (e.g., a human patient) one or more additional agents that provide a therapeutic benefit to a subject who has, or is at risk of developing, cancer. Chemotherapeutic agents suitable for co-administration with compositions of the present invention include, for example: taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxyanthrancindione, mitoxantrone, mithramycin, actinomycin D, 1 -dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Further agents include, for example, antimetabolites (e.g., methotrexate, 6- mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g. mechlorethamine, thioTEPA, chlorambucil, melphalan, carmustine (BSNU), lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, cis-dichlordiamine platinum (II)(DDP), procarbazine, altretamine, cisplatin, carboplatin, oxaliplatin, nedaplatin, satraplatin, or triplatin tetranitrate), anthracycline (e.g. daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g. dactinomcin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g. vincristine and vinblastine) and temozolomide. In some embodiments, an anti-CD 137 antibody and the one or more additional active agents are administered at the same time. In other embodiments, the anti-CDl37 antibody is administered first in time and the one or more additional active agents are administered second in time. In some embodiments, the one or more additional active agents are administered first in time and the anti- CD 137 antibody is administered second in time.

An anti-CD 137 antibody or an antigen-binding fragment thereof described herein can replace or augment a previously or currently administered therapy. For example, upon treating with an anti-CD 137 antibody or antigen-binding fragment thereof, administration of the one or more additional active agents can cease or diminish, e.g., be administered at lower levels or dosages. In some embodiments, administration of the previous therapy can be maintained. In some embodiments, a previous therapy will be maintained until the level of the anti-CD 137 antibody reaches a level sufficient to provide a therapeutic effect. The two therapies can be administered in combination.

Monitoring a subject (e.g., a human patient) for an improvement in a cancer, as defined herein, means evaluating the subject for a change in a disease parameter, e.g., a reduction in tumor growth. In some embodiments, the evaluation is performed at least one (1) hour, e.g., at least 2, 4, 6, 8, 12, 24, or 48 hours, or at least 1 day, 2 days, 4 days, 10 days, 13 days, 20 days or more, or at least 1 week, 2 weeks, 4 weeks, 10 weeks, 13 weeks, 20 weeks or more, after an administration. The subject can be evaluated in one or more of the following periods: prior to beginning of treatment; during the treatment; or after one or more elements of the treatment have been administered. Evaluation can include evaluating the need for further treatment, e.g., evaluating whether a dosage, frequency of administration, or duration of treatment should be altered. It can also include evaluating the need to add or drop a selected therapeutic modality, e.g., adding or dropping any of the treatments for a cancer described herein.

In some embodiments, an anti-CD 137 antibody or an antigen-binding fragment thereof described herein is administered to modulate a T-cell response in a patient, for example, by increasing T-cell activation and/or proliferation. Crosslinking of CD137 strongly enhances T cell proliferation, IFN Y production and secretion, and the cytolytic activity of T cells. Accordingly, in some embodiments, an anti-CDl37 agonist antibody, or an antigen-binding fragment thereof, of the present disclosure is administered to a patent in need thereof to induce or increase T-cell activation, enhance T cell proliferation, induce the production and/or secretion of IFN Y , and/or induce a cytolytic T cell response.

In some embodiments, an anti-CD 137 antibody or an antigen-binding fragment thereof described herein is useful to modulate or shift the T-cell population in a patient from a T H 2/T reg T cell population to a T H 1/T H 17 T cell population to thereby improve or enhance an anti-tumor response in the patient. Studies have shown that while CD137 is expressed in both T-cell subsets, Thl and Th2 T cells, CD137 is expressed at higher levels on CD8+ T cells than on CD4+ T cells. Accordingly, CD137 mainly co-stimulates CD8+ T cells. Accordingly, an anti-CDl37 antibody, or an antigen-binding fragment thereof, as described herein, is administered to a patient to enhance an anti-tumor response, for example, by modulating or shifting the T-cell response and/or T cell population in the patient from a T fZ/T reg T cell response and or T cell population to a T H 1/T H 17 T cell response and/or T cell population in the patient.

In some cancers (e.g. melanoma and ovarian cancer), natural tumor-infiltrating lymphocytes (TILs) can be enriched through optimized cell culture methods and provide a source of tumor-reactive lymphocytes useful for adoptive immunotherapy. Adoptive TIL therapy can result in durable tumor regression for some types of cancer, which warrants the development and optimization of TIL-based approaches for cancer. Currently, the identification and expansion of natural tumor-reactive TILs remains challenging due to low level and/or rarity of antigen- specific CD8+ T cells. CD137 expression by T cells is activation dependent, which provides an opportunity to capture CDl37-expressing activated T cells from circulation or from tumor samples. Accordingly, an anti-CD 137 antibody, or an antigen-binding fragment thereof, as described herein, can be employed for the selective enrichment of activated, antigen- specific T cells.

In some embodiments, the efficacy of the anti-CD 137 antibodies described herein is dependent on a competent immune system. Specifically, in some embodiments, depletion of CD4+ T cells, CD8+ T cells and/or Natural Killer cells reduces the efficacy of the anti-CD 137 antibodies. In some embodiments, depletion of CD4+ T cells, CD8+ T cells and/or Natural Killer cells reduces the inhibition or reduction of tumor growth by the anti-CD 137 antibodies described herein. In some embodiments, depletion of CD4+ T cells, CD8+ T cells and/or Natural Killer cells reduces the inhibition or reduction of tumor growth by the anti-CD 137 antibodies described herein. In some embodiments, the efficacy of the anti-CD 137 antibodies described herein is dependent on an infiltration of immune cells into a tumor microenvironment. In some embodiments, the infiltration of immune cells into a tumor microenvironment is coupled with a lack of infiltration into the spleen and/or liver.

In some embodiments, the anti-CD 137 antibodies described herein induce a protective anti tumor memory immune response. Memory T cells are a subset of antigen- specific T cells that persist long-term after having encountered and responded to their cognate antigen. Such cells quickly expand to large numbers of effector cells upon re-exposure to their cognate antigen. Accordingly, in some embodiments the anti-CD 137 antibodies described herein stimulate the production of memory T cells to a cancer antigen. In some embodiments, a subject that has received an anti-CD 137 antibody described herein to treat or cure a cancer, develops memory T cells specific to the cancer. In some embodiments, a subject that has received an anti-CD 137 antibody described herein to treat or cure a cancer, develops an anti-tumor memory immune response upon re-exposure to the cancer. In some embodiments, the anti-tumor memory immune response comprises stimulating memory T cells to become effector cells. In some embodiments, a subject that has received an anti-CD 137 antibody described herein to treat or cure a cancer, develops an anti-tumor memory immune response to a cancer antigen.

In some embodiments, the anti-CDl37 antibodies described herein induce immune re programming with a tumor microenvironment. Specifically, in some embodiments, the anti- CD 137 antibodies induce immune infiltration; reduce, inhibit or prevent Treg proliferation; reduce, inhibit or prevent tumor-associated macrophage proliferation; and protect or reverse T cell exhaustion.

In some embodiments, the anti-CDl37 antibodies induce infiltration of immune cells into a tumor microenvironment relative. In some embodiments, the anti-CD 137 antibodies increase immune cell infiltration by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 105%, at least 110%, at least 115%, at least 120%, at least 125%, at least 130%, at least 135%, at least 140%, at least 145%, or at least 150%. In some embodiments, immune cell infiltration is determined by measuring the level of CD45 expression on cells isolated from a tumor microenvironment. Methods for measuring protein expression are known to those of skill in the art and described herein.

In some embodiments, the anti-CDl37 antibodies prevent or inhibit an increase in Treg cells in a tumor microenvironment. In some embodiments, prevention or inhibition is relative to the amount of Treg cells in a tumor microenvironment in the absence of an anti-CD 137 antibody. In some embodiments, prevention or inhibition of an increase in Treg cells is relative to a reference antibody. In some embodiments, Treg cells are detected by expression of CD25 and FOX-3P on CD4+ T cells isolated from a tumor microenvironment. Methods for measuring protein expression are known to those of skill in the art and described herein.

In some embodiments, the anti-CDl37 antibodies prevent or inhibit an increase in tumor- associated macrophages in a tumor microenvironment. In some embodiments, prevention or inhibition is relative to the amount of tumor-associated macrophages in a tumor microenvironment in the absence of an anti-CDl37 antibody. In some embodiments, prevention or inhibition of an increase in tumor-associated macrophages is relative to a reference antibody. In some embodiments, tumor-associated macrophages are detected by expression of CDl lb and F4/80 on CD45+ immune cells isolated from a tumor microenvironment. Methods for measuring protein expression are known to those of skill in the art and described herein.

In some embodiments, the anti-CDl37 antibodies protect T cells from T cell exhaustion in a tumor microenvironment. In some embodiments, the anti-CDl37 antibodies reverse T cell exhaustion in a tumor microenvironment. In some embodiments, T cell exhaustion in a tumor microenvironment is reduced in the presence of an anti-CD 137 antibody described herein, relative to a tumor microenvironment in the absence of the anti-CD 137 antibody. In some embodiments, T cell exhaustion is determined by analyzing CD8+ T cells or CD4+ T cells for expression of co- inhibitory receptors (e.g., PD-l, TIGIT or LAG-3). In some embodiments, T cell exhaustion is detected by expression of PD-l and TIGIT on CD4+ or CD8+ T cells isolated from a tumor microenvironment.

In some embodiments, an anti-CD 137 antibody, or an antigen-binding fragment thereof, described herein, can be employed in methods of detection and/or quantification of human CD 137 in a biological sample. Accordingly, an anti-CD 137 antibodies, or an antigen-binding fragment thereof, as described herein, is used to diagnose, prognose, and/or determine progression of disease (e.g., cancer) in a patient.

Other Embodiments

1. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

2. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

3. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

4. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

5. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

6. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 140, 144 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

7. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

8. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

9. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 141, 145 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

10. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

11. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

12. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 142, 146 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

13. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 148, 151 and 154, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

14. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 149, 152 and 155, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

15. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an agonist monoclonal antibody, or antigen-binding fragment thereof, that specifically binds human CD137, wherein the agonist monoclonal antibody or antigen binding portion thereof comprises heavy chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 139, 143 and 147, respectively, and light chain CDR1, CDR2 and CDR3 sequences set forth in SEQ ID NOs: 150, 153 and 156, respectively, and wherein the subject has cancer cells that express major histocompatibility complex I (MHC I).

EXAMPLES

While the present disclosure has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the disclosure. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present disclosure. All such modifications are intended to be within the scope of the disclosure. Example 1: Synthetic Hu an Monoclonal Antibodies Produced in Yeast Exhibit Binding To Recombinant Human CD137

Purified CD137 protein antigen was biotinylated using the EZ-Link Sulfo-NHS- Biotinylation Kit (Thermo Scientific). CD137 antigens were concentrated to ~lmg/mL and buffer exchanged into PBS before addition of 1:7.5 molar ratio biotinylation reagent (EZ-Link Sulfo- NHS -Biotinylation Kit, Thermo Scientific, Cat #21425.). The mixture was held at 4°C overnight prior to another buffer exchange to remove free biotin in the solution. Biotinylation was confirmed through Streptavidin sensor binding of the labeled proteins on a ForteBio. Successful biotinylation of the CD 137 protein antigen was confirmed via detectable binding to a streptavidin-linked biosensor installed on ForteBio Octet™ Red384 Interferometer (Pall ForteBio, Menlo Park, CA) according to the manufacturer's guidelines (data not shown).

Eight naive human synthetic yeast-based antibody libraries each of -10 9 diversity were designed, generated, and propagated as described previously (see, e.g., W02009036379; WO2010105256; W02012009568; Xu et ah, Protein Eng Des Sel. 2013 Oct;26(l0):663-70). Eight parallel selections were performed, using the eight naive libraries against biotinylated human CDl37-Fc fusion.

For the first two rounds of selection, a magnetic bead sorting technique utilizing the Miltenyi MACS system was performed, essentially as described (Siegel etal., J Immunol Methods. 2004 Mar;286(l-2): 141-53). Briefly, yeast cells (-10 10 cells/library) were incubated with 10 mL of 10 nM biotinylated human CDl37-Fc fusion antigen for 15 minutes at room temperature in FACS wash buffer PBS with 0.1% BSA. After washing once with 50 mL ice-cold wash buffer, the cell pellet was resuspended in 40 mL wash buffer, and 500 pl Streptavidin MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany. Cat # 130-048-101) were added to the yeast and incubated for 15 minutes at 4°C. Next, the yeast were pelleted, resuspended in 5 mL wash buffer, and loaded onto a MACS LS column (Miltenyi Biotec, Bergisch Gladbach, Germany. Cat.# 130- 042-401). After the 5 mL was loaded, the column was washed three times with 3 mL FACS wash buffer. The column was then removed from the magnetic field, and the yeast were eluted with 5 mL of growth media and then grown overnight.

Subsequent to the two rounds of MACS, three rounds of sorting were performed using flow cytometry (FACS), which are described in the following three paragraphs. Selection strategy employing 8 parallel selections with Fc antigen

The eight libraries from the MACS selections were taken through three rounds of FACS selections. Approximately lxlO 8 yeast per library were pelleted, washed three times with wash buffer, and incubated with 10 nM of biotinylated human CDl37-Fc fusion and 10 nM of biotinylated murine CDl37-Fc fusion antigen separately for 10 minutes at room temperature. Yeast were then washed twice and stained with goat anti-human F(ab’) 2 kappa-FITC diluted 1: 100 (Southern Biotech, Birmingham, Alabama, Cat# 2062-02) and either streptavidin-Alexa Fluor 633 (Life Technologies, Grand Island, NY, Cat # S21375) diluted 1:500, or Extravidin-phycoerthyrin (Sigma- Aldrich, St Louis, Cat # E4011) diluted 1:50, secondary reagents for 15 minutes at 4°C. After washing twice with ice-cold wash buffer, the cell pellets were resuspended in 0.4 mL wash buffer and transferred to strainer-capped sort tubes. Sorting was performed using a FACS ARIA sorter (BD Biosciences) and sort gates were determined to select only CD137 binding. The murine- and human- selected populations from the first round of FACS were brought forward into the next round.

The second and third round of FACS for the above selected populations involved positive sorts for binders to human and/or murine CD 137 reagents; or negative sorts to decrease polyspecific reagent binders (Xu et al, PEDS. 2013 Oct;26(l0):663-70). Depending on the amount of polyspecific binding or target binding of a specific selection output, a positive sort followed a negative sort or vice versa, to enrich for a full binding population with limited amount of polyspecific binding. Competition selections were also performed with control mAbs from the literature. For competition selections, mAb4 (urelumab; Bristol-Myers Squibb; CAS Number: 934823-49-1) and mAb5 (utomilumab; Pfizer; CAS Number: 1417318-27-4) were pre-complexed to biotinylated human CDl37-Fc fusion. Antibodies that bind and do not bind in the presence of the control mAbs were selected for on FACS. The outputs of these rounds were plated and isolates were picked for sequencing and characterization.

Affinity maturation of clones identified in naive selections

Heavy chains from the first FACS sorting round against biotinylated human CD 137 Fc fusion outputs were used to prepare light chain diversification libraries used for four additional selection rounds. The first of these selection rounds utilized Miltenyi MACs beads conjugated with 10 nM biotinylated human CDl37-Fc fusion as antigen. Subsequent to the MACs bead selections, three rounds of FACS sorting were performed. The first of these rounds used biotinylated human CDl37-Fc fusion at lOnM. The second FACS round for the above involved positive sorts for binders to mouse CD137 reagents, competition sorts with previously mentioned control mAbs or negative sorts to decrease polyspecific reagent binders as described above. The third and final round of FACS selection was done using either biotinylated murine CD137 Fc fusion at 10 nM or biotinylated human monomeric CD137 at 50 nM. Individual colonies from each FACS selection round described above were picked for sequencing characterization.

IgG and Fab production and purification

Yeast clones were grown to saturation and then induced for 48 hours at 30°C with shaking. After induction, yeast cells were pelleted and the supernatants were harvested for purification. IgGs were purified using a Protein A column and eluted with acetic acid, pH 2.0. Fab fragments were generated by papain digestion and purified over CaptureSelect IgG-CHl affinity matrix (LifeTechnologies, Cat # 1943200250).

Example 2: Epitope Binning and Determination of Human Anti-CD137 Antibody Affinity to Recombinant CD137

Epitope binning of the antibodies isolated in Example 1 was performed on a Forte Bio Octet Red384 system (Pall Forte Bio Corporation, Menlo Park, CA) using a standard sandwich format binning assay. CD 137 control antibody IgGs were loaded onto AHQ sensors and unoccupied Fc-binding sites on the sensor were blocked with a non-relevant human IgGl antibody. The sensors were then exposed to 100 nM target antigen followed by exposure to the isolated antibodies identified as described in Example 1. Data were processed using ForteBio’s Data Analysis Software 7.0. Additional binding by the second antibody after antigen association indicates an unoccupied epitope (non-competitor), while no binding indicates epitope blocking (competitor) (data not shown).

Affinity of the CD 137 antibodies was determined by measuring their kinetic constants (k a , k d , KD) on ForteBio Octet. ForteBio affinity measurements were performed generally as previously described (Estep et al, MAbs. 2013 5(2):270-8). Briefly, ForteBio affinity measurements were performed by loading antibodies (IgGs) on-line onto AHQ sensors. Sensors were equilibrated off-line in assay buffer for 30 minutes and then monitored on-line for 60 seconds for baseline establishment. For avid binding measurement, sensors with loaded IgGs were exposed to 100 nM antigen (human, cyno, or murine CD137) for 3 minutes, afterwards they were transferred to assay buffer for 3 minutes for off-rate measurement. Monovalent binding measurements were obtained by loading human CDl37-Fc fusion on AHQ sensors followed by exposure to 200 nM antibody Fab in solution. Kinetics data were fit using a 1: 1 binding model in the data analysis software provided by ForteBio (data not shown).

Determination of whether antibodies were ligand blocking was also assessed. Specifically, ligand blocking experiments were performed both on Octet HTX (ForteBio) and on label-free MX96 SPRi (Caterra). mAbl was captured on Octet sensor or MX96 chip sensor. CD137 and CD137L were sequentially applied to the sensors pre-loaded with mAbl. An increase in response upon exposure to CD137L indicated non-competition between mAbl and CD137L for binding to CD 137. On the other hand, a lack of change in the signal indicated competition, which was the case for control antibody mAb5. mAbl did not inhibit binding of CD137L to CD 137 (data not show), and therefore was considered a non-ligand blocking antibody.

Example 3: Distribution of Binding Affinities of Affinity-Matured Anti-CD137 Antibodies

Affinity matured anti-CDl37 antibodies were generated using 2 mutant libraries. The first library contained mutations in the heavy chain and the second library contained mutations in the light chain, wherein donor diversity in light chain CDR1, CDR2 and CDR3 was created. The mutant libraries went through 3 rounds of phage panning aimed at increasing affinity and maintaining cross-reactivity with mouse CD137. In each round, an off-rate competition step was employed after initial binding to biotinylated antigens (i.e., 1 hour incubation with excess unlabeled antigen or parental IgG at 37°C).

The resulting anti-CD 137 antibodies from different selection rounds were plotted on k d /k a double log plots. Apparent association and dissociation kinetic rate constants (k a and k d values) were determined on an SPRi reader (MX96, Carterra) in a running buffer of PBS-T 0.01%. Anti human CD 137 antibodies were covalently printed on a Carboxymethyldextran hydrogel 50L chip (Xantec bioanalytics) on a CFM (Carterra). Freshly mixed activating reagents (150 ml 0.4 M EDC and 150 ml 0.1 M sulfo-NHS in H20) were used to activate the surface of the SPR substrate for 7 minutes. Antibodies at 10 mg/ml in acetic acid buffer pH 4.5 were used for printing for 15 minutes. The printed chip was then quenched on SPRi reader (MX96, Carterra) with 1 M ethanolamine for 15 minutes. For kinetics analysis, purified recombinant his tagged human CD137 (0, 2.05, 5.12, 12.8, 32, 80, 200, 500 nM) was injected sequentially. For each concentration, there was 5 minutes of association followed by 10 minutes of dissociation. Data were processed and analyzed in SPR Inspection Tool and Scrubber softwares. The kinetic data were referenced with the interstitial reference spots and double-referenced to a buffer cycle, and then fit globally to a 1 : 1 binding model to determine their apparent association and dissociation kinetic rate constants (k a and k d values). The ratio k d /k a was used to derive the K D value of each antigen/mAb interaction, i.e. Ko=k d /k a .

Antibodies with K D (k d /k a ) between 10-20 nM are shown as upright triangles, while the ones with K D lower than 10 nM are shown as upside down triangles (FIG. 1). Affinity maturation of only the heavy chains (top panels) or only the light chains (bottom panels) both resulted in the isolation of anti-CD 137 antibodies with higher binding affinities than the parental antibody (mAbl) (FIG. 1). The heavy chain and light chain variable regions of mAbl are set forth in SEQ ID NOs: 4 and 6, respectively.

Example 4: Identification of Critical Binding Residues Comprising Heavy Chain CDR3 (CDRH3) of Anti-CD137 Antibodies

To determine which amino acid residues within CDRH3 are critical for the binding of mAbl to mouse and human CD 137 polypeptides, alanine scanning was performed. A set of polynucleotides encoding derivatives of the mAbl open reading frame was generated, wherein each derivative contained a single alanine residue substitution at a wild-type amino acid residue position comprising CDRH3. Positions D95 through M100I of SEQ ID NO: 4 were each mutated to alanine by replacing the wild-type codon with the alanine codon GCC. The amino acid sequences of each CDRH3 of each mAbl alanine-substituted derivative are set forth in SEQ ID NOs: 111-125. The polynucleotides encoding each of the 15 mAbl alanine-substituted derivatives were individually cloned into an expression vector (aglyco-IgGl, DID-2600) via Gibson Assembly. Each mAbl alanine-substituted derivative was expressed and purified using standard techniques known in the art. Binding affinities of each mAbl alanine-substituted derivative for human and mouse CD137 were determined via Wasatch SPR kinetics measurements for human CD137 (huCDl37) or equilibrium cell-binding assays for mouse CD137 (mCDl37). Table 1 provides the calculated dissociation constants (KD) for each mutant. When“Weak” is noted in the table there was measurable binding above background but not enough confidence in the curve fitting to assign an accurate KD value. In Table 1,“NB” signifies that no binding was observed during the determination of binding affinities and indicates which alanine substitutions in CDRH3 resulted in an antibody that did not bind to CD 137.

Table 1: Binding affinity (KD) of alanine scanning clones for human and mouse CD137

The retention, weakening, or loss of binding affinity resulting from mutations to alanine informed the determination of which residues were required for CD 137 binding and which residues tolerated mutations. FIG. 2 summarizes the binding data for alanine scanning of CDRH3 with wild-type amino acid identity indicated at each position. CDRH3 positions are color-coded based on the effects of mutating the position to alanine, as shown. This analysis resulted in the following consensus sequence: OXPFXLDXX YYYYYX. When bolded residues in the consensus sequence were mutated to alanine there was a complete loss of binding and these residues were therefore necessary for mAbl binding to CD 137. When italicized residues in the consensus sequence were mutated to alanine the antibody was still able to bind CD 137 but with a weaker affinity indicating these residues played a partial role in binding but were not absolutely required. When residue positions denoted with an X in the consensus sequence were mutated to alanine there was little to no change in binding affinity. Thus, these residues tolerated mutations and were not critical to the binding interaction. Example 5: Epitope Mapping by Scanning Saturation Mutagenesis and Homolog Comparison

Functional mapping of the CD 137 epitope by scanning saturation mutagenesis library and homology comparison were performed to identify residues important for antibody binding to CD 137. Combinatorial libraries of CD 137 mutants with single point mutations at all residue positions to every possible amino acid substitution except cysteine were generated and tested for their ability to bind to mAbl, mAb4, and mAb5. A library consisting of genes encoding each point mutant of CD 137 were synthesized from a commercial supplier and cloned into a mammalian display expression vector. Mammalian display was used to present a library of variant human CD 137 extracellular domains, with each variant having at least one point mutation relative to wild type human CD 137.

The library of cells displaying CD 137 variants was stained with non-overlapping antibodies (i) mAb4 and mAbl or (ii) mAb4 and mAb5. Populations of cells with reduced binding to one antibody but not the other were enriched by FACS. Each population was sequenced by Illumina sequencing to identify mutations in positions that specifically disrupted binding to each antibody but did not affect correct folding of CD 137 or binding to the non-overlapping antibody.

For mAbl, K114 was identified as the most important residue important for binding to CD137, with 34% of all mutations observed occurring in that position, and all amino acid substitutions observed. El 11, T113, and P135 are also important for binding, with 10% of mutations observed in each of those positions. Additionally, N126 and 1132 was observed in the population that had partial decrease in binding for mAbl. FIG. 3A shows the residues comprising the epitope for mAbl, mAb4 and mAb5. mAb4 and mAb5 had binding epitopes that were distinct from mAbl. For mAb4, N42 was the most important residue with 50% of all mutations observed in that position, followed by R41 and D38. For mAb5, 1132 was the most important with 32% of all mutations occurring in that position, followed by N126, G96, K114, and L95.

Point mutants isolated from the library screen were expressed as soluble proteins and tested for binding to mAbl. All 4 mutations tested at K114 (R, E, N, T) abolished binding to mAbl. Mutations at T113 and P135 also disrupted binding. 1/2 point mutants at El 11, 1/3 mutants at N126, and 1/4 mutants at 1132 showed no binding. Likewise, 3/3 mutants at N42 did not bind to mAb4, and 3/4 mutants at 1132 did not bind to mAb5. Additionally, CD 137 homologs were tested for their binding to mAbl. mAbl was able to bind to mouse CD137, but not to rat CD137, as shown in FIG. 3B. To determine if there was a difference in the residues comprising the epitope for mAbl between mouse CD137 and rat CD137, the amino acid sequences of CD 137 homologs from human, cynomolgus monkey, rat, and mouse were aligned for comparison. All of the amino acid residues comprising the mAbl epitope are present in human, cynomolgus monkey, and mouse, but not in rat. Lysine 114 (Kl 14) of the human CD 137 sequence, as well as the corresponding lysine in the cynomolgus monkey and mouse CD137 sequences, is glutamic acid (E) in the rat CD137 sequence, further indicating that Kl 14 of the human CD 137 sequence is at least one of the critical binding residues for mAbl.

FIGs. 3C and 3D show the crystal structure of human CD137 bound to CD137L (Bitra A et ah, J Biol Chem 2018, 293(26):9958-9969), wherein residues El 11, T113, K114 and P135 are shown as spheres. As can be seen, these residues are located away from the CD 137 ligand (CD137L) binding domain, shown in grey.

Example 6: Effect of Anti-CD137 Antibodies on Immune Regulators and CD8+ T Cells in Mice

Three anti-CD 137 antibodies generated in Example 1, mAbl, mAb2 and mAb3, were further analyzed for their efficacy. These antibodies were mouse cross-reactive and comprised the constant regions of the human IgG4 isotype containing the S228P mutation to prevent Fab shuffling. The 3H3 monoclonal antibody, known to stimulate mouse CD137 signaling in vivo and elicit anti-tumor immunity (Melero et al. (1997) Nature Medicine 3(6):682-685; Uno et al. (2006) Nature Medicine l2(6):693-696), was used as a comparator (BioXcell cat# BE0239; lot number 5926/1115). Notably, antibody 3H3 has similar properties to that of urelumab (Bristol-Myers Squibb; CAS Number: 934823-49-1), a fully human IgG4-S228P agonistic antibody that targets the extracellular domain of CD137, but does not block ligand binding. In addition, anti-Rat IgG4 was used as an isotype control (BioXcell cat# BE0089; lot number 5533/5679-316J1). Dilutions were made in PBS to achieve desired dose per mice, as indicated, in 100 pL injection volume.

The antibodies (100 pg) were administered intraperitoneally on days 0, 3, 6 to non-tumor bearing female B alb/c mice and spleens were harvested on day 9. Levels of PD-l and TIGIT expression on CD8+CD44+ T cells were measured by flow cytometry. Specifically, single cell suspensions from the spleens were obtained by mechanical disruption and passing through a 40 mih cell strainer. Red blood cells were lysed using ACK buffer. The cell suspensions were stained with the following antibodies: CD45 (clone 30-F11, eBioscience), CD8 (clone 53-6.7, BD Biosciences), CD4 (clone RM-45, BD Biosciences), CD44 (clone IM7, eBioscience), PD-l (RMP1-30, eBioscience) and TIGIT (GIGD7, eBioscience). Data acquisition was carried out on the MACSQuant Analyzer flow cytometer (Milenyi) and data were analyzed using the FlowJo software, version 10.

Antibody 3H3 caused a significant increase in expression of both PD-l and TIGIT, whereas only antibody mAbl increased expression compared to mAb2 and mAb3 (FIGs. 4A and 4B). In addition, expansion of CD8+ T cells was assessed by analyzing the percentage of splenic CD45+ cells or number of CD8+ T cells per spleen. Similarly, antibody 3H3 caused the highest expansion of CD8+ T cells, with mAbl resulting in the highest levels of CD8+ T cell expansion relative to mAb2 and mAb3 (FIG. 4C). Accordingly, mAbl was selected for further testing.

Example 7: Efficacy of Anti-CD137 Antibodies in Tumor-Bearing Mice

Given the ability of mAbl to enhance CD8+ T cell expansion, as shown in Example 6, mAbl was further analyzed for anti-tumor activity using a subcutaneous model of syngeneic colon cancer. Specifically, CT26 tumor cells (passage 3) were maintained under aseptic conditions in DMEM Medium (Gibco cat# 11965-092), containing 10% 56°C -heat inactivated FBS (Gibco 10438-034), 1 mM sodium pyruvate (Gibco cat. # 11360-070), IX NEAA (Gibco cat# 11140-050) and IX MEM Vitamin solution (Gibco cat#l 1120-052). Cells were maintained at 37°C and 5% C0 2 . Upon reaching 50-70 % confluence, cells were passaged at a ratio of 1: 10, for a total of two passages, prior to in vivo implantation. Cells were harvested and counted using a Hemacytometer (Hausser Scientific Bright-Line #1492).

Balb/c female mice were purchased from Charles River Laboratories and were nine weeks old at the start of study. CT26 tumor cells (1 xlO 5 cells per mouse in 0.1 mL PBS) were injected subcutaneously into the right flank of each mouse, and tumor volume was calculated twice weekly (Length* (Width A 2)/2) using dial calipers. On day 7 post-tumor inoculation, animals were sorted into groups of eight, and treatments were initiated. Body weights were recorded three times per week for the duration of the study. mAbl was administered at three different dosages (100, 50 or 25 mg/mouse), 3H3 at two different dosages (50 or 10 mg/mouse) and the isotype control antibody at a dosage of 50 mg/mouse. All mice were dosed intraperitoneally at days 0, 3, 6 and 9.

Expansion of CD8+ T cells in the tumors was confirmed in vivo for both mAbl and 3H3 antibodies (data not shown). Individual tumor volumes are shown in FIG. 5A and mean tumor volumes are shown in FIG. 5B. mAbl treatment resulted in inhibition of tumor growth compared to the control group at all three dosages. Moreover, treatment with mAbl resulted in the complete regressions in 6 out of 8 mice at the 25 pg dose level, 5 out of 8 mice at the 50 pg dose level and 3 out of 8 mice at the 100 pg dose level.

Overall survival in each treatment group is shown in FIG. 5C. Strong anti-tumor activity of mAbl against CT26 tumors was reflected as extended overall survival. Long term survival (>60 days) were observed in 80% of the mice at the 25 pg dose level, 62% of the mice at the 50 pg dose level and 38% of the mice at the 100 pg dose level.

Mice with no palpable tumor at day 70 were considered cured and re-challenged with subcutaneous injection of CT26 cells in the opposite flank. Specifically, mice with eradicated tumors were injected again with lxlO 5 CT26 cells in the left flank and tumor volume was calculated twice weekly (Length*(Width A 2)/2) using dial calipers. Five non-immunized (naive) mice were injected in the same manner as a control, respectively. Results of the re-challenge experiment are shown in FIG. 5D. Twenty-two days after the subcutaneous injection of CT26 cells, none of the re-challenged mice formed tumors. In contrast, all of the naive mice that were injected with the same cells formed tumors. Therefore, all mice that were considered cured rejected CT26 tumors suggesting that mAbl can induce long-term protective memory.

Example 8: Efficacy of Affinity-Matured Anti-CD137 Antibodies in Tumor-Bearing Mice

The affinity-matured monoclonal antibodies generated in Example 4 were analyzed for anti-tumor activity using the same subcutaneous model of syngeneic colon cancer (CT26) essentially as described in Example 7. Specifically, 6 affinity- matured clones (mAb7-mAbl2) were generated with IgG4 constant regions and tested accordingly. The sequences of the heavy chain and light chain variable regions are provided in the chart below, along with their KD values to mouse CD 137 (determined by ForteBio Octet, described in Example 2) and human CD 137 (determined by Carterra, described in Example 4).

Parental mAbl, the 3H3 antibody (data not shown), and an IgG4 isotype antibody were used as controls. All mice were dosed with 50pg of mAb/mouse intraperitoneally at days 0, 3, 7 and 10. Spleens and livers were harvested on day 13 after therapy initiation.

Individual tumor volumes are shown in FIG. 6A and mean tumor volumes are shown in FIG. 6B. Consistent with the results from Example 7, treatment with parental mAbl resulted in a reduction in tumor volume. Further, administration of all affinity-matured clones derived from mAbl (mAb7-mAbl2) to tumor-bearing mice resulted in an inhibition of tumor growth compared to mice treated with the isotype control antibody.

Example 9: Effect of Anti-CD137 Antibodies on T Cells in Tumor-Bearing Mice

To determine the effect of anti-CD 137 antibodies (i.e., 3H3 and mAbl) on the level of T cells in tumor-bearing mice, B alb/c mice with CT26 tumors, as described in Example 7, were intraperitoneally injected with antibodies on days 0 and 3, and tissues were harvested on day 7. mAbl was administered at three different dosages (100, 50 or 25 pg/mouse), 3H3 at two different dosages (50 or 10 pg/mouse) and the isotype control antibody at a dosage of 50 pg/mouse.

Single cell suspensions from the spleen were obtained as described in Example 6 and tumor cell suspensions were obtained by enzymatic and mechanical digestion using tumor dissociation kit (Miltenyi cat# 130-096-730). Cell suspensions were treated with complete medium to inactivate the enzymes and then passed through a 40 pm cell strainer. Red blood cells were lysed using ACK buffer. Cells were stained with antibodies against CD45, CD8 and CD4, and analyzed as described in Example 6. FIG. 7 shows the number of CD4+ and CD8+ T cells, as a percentage of CD45+ cells, found in the spleen and tumor. These results indicated that mAbl selectively expands tumor- infiltrating CD8+ T cells as compared to splenic CD8+ T cells.

Example 10: Effect of CD4+, CD8+, or NK lymphocytes Depletion on Anti-Tumor Efficacy of Anti-CD137 Antibodies In Vivo

To assess the mechanism of action of anti-CDl37 antibodies, Balb/c mice with CT26 tumors, as described in Example 7, were intraperitoneally injected with mAbl alone or in combination with anti-CD4 (GK1.5), anti-CD8 (YTS 169.4), or anti-asialo-GMl (targets NK cells) antibodies to deplete these specific lymphocyte subsets from the animals. Mice treated only with the mAbl antibody were administered l50pg of antibody on days 6, 9, 12, 19, and 26. The mice treated with l50pg mAbl in combination with 500pg anti-CD4, anti-CD8, or 50uL of anti-asialo- GMl antibodies administered on days -1, 0, 5, 10, 15, and 20. Effective depletion was confirmed by FACS analysis (data not shown).

Individual tumor volumes are shown in FIG. 8. Consistent with the results from Example 7, treatment with parental mAbl resulted in a reduction in tumor volume. Further, administration of mAbl in combination with lymphocyte-depleting anti-CD4, anti-CD8, or anti-asialo-GMl antibodies reduced the anti-tumor activity of the mAbl antibody. These results indicated cooperation between innate and adaptive immunity for anti-tumor efficacy of the anti-CD 137 antibodies described herein.

Example 11: Anti-Tumor Efficacy of Anti-CD137 Antibodies in Various Tumor Models

To determine whether an anti-CD 137 antibody had anti-tumor efficacy in different tumor models, mAb8 was administered to mice having either CT26 tumors (colon carcinoma; as described above), EMT-6 tumors (breast carcinoma), A20 tumors (B cell lymphoma) or MC38 tumors (colon carcinoma).

For all tumor models, female mice were purchased from Charles River Laboratories and were 7-9 weeks old at the start of study. For each tumor type appropriate syngeneic mouse strain was used (Balb/c for CT26, EMT-6 and A20; C57BL/6 for MC38). EMT6 tumor cells (5 xlO 4 ) cells per mouse in 0.05 mL PBS) were injected into the right mammary fat pad of each mouse. CT26 tumor cells (1 xlO 5 cells per mouse), A20 tumor cells (5 xlO 6 cells per mouse) and MC38 tumor cells (5 xlO 5 cells per mouse) were injected subcutaneously into the right flank of each mouse, and tumor volume was calculated twice weekly (Length*(Width A 2)/2) using dial calipers. Upon reaching 50-100 mm 3 sized tumors, the mice were randomized to receive mAb8 or isotype control (day 0). Mice with orthoptic EMT6 tumors received 12.5 pg on days 0, 3, 6 and 9. Mice with A20 (200 pg/mouse) and MC38 (12.5 pg/mouse) tumors received 5 doses once a week. All mice were dosed intraperitoneally.

As shown in FIG. 9, mAb8 was effective in all four tumor models tested, indicating a wide range of efficacy for varying cancer types. Treatment with mAb8 resulted in tumor regressions in mice carrying 8/8 CT26, 3/8 EMT6, 5/8 A20 tumors and delayed growth in majority of the remaining mice carrying EMT6, A20 and MC38.

Example 12: Effect of Dosage of Anti-CD137 Antibodies

To further characterize the anti-tumor efficacy of the anti-CDl37 antibodies, a dosage study was performed using the same subcutaneous model of syngeneic colon cancer (CT26) essentially as described in Example 7. Specifically, parental mAbl and affinity matured antibodies mAb8 and mAblO were administered intraperitoneally at doses of either l50pg (high dose) or 20pg (low dose) per mouse on days 0, 3, 6 and 9, with 8 mice per treatment group. One group of mice (n=8) was administered an IgG4 isotype control at a dose of l50pg.

Individual tumor volumes, mean tumor volume and percent survival of mice treated at the l50pg are shown in FIG. 10A, FIG. 10B, and FIG. 10C, respectively. Individual tumor volumes, mean tumor volume and percent survival of mice treated at the 20pg are shown in FIG. 11A, FIG. 11B, and FIG. 11C, respectively. These results indicated that treatment with the parental mAbl and the affinity-matured mAb8 and mAblO antibodies resulted in a reduction in tumor volume and an increase in mouse survival at both high and low doses.

In a separate dosage study utilizing the CT26 tumor model, additional doses of parental mAbl were tested. Specifically, mAbl was administered intraperitoneally at the following doses: l2.5pg, 25pg, 50pg, lOOpg and 200pg. FIG. 12 shows the results of the dosage study, indicating efficacy over a wide dose range. Treatment with mAbl resulted in tumor regressions in at least 3/8 mice in each dose level with optimum dose range (50-100 pg/mouse) leading to 7/8 mice with eradicated tumors. Example 13: Effect of Fc-Receptor Binding on Anti-Tumor Efficacy of Anti-CD137 Antibodies

To determine the contribution of Fc-receptor binding on the anti-tumor activity of anti- CD137 antibodies, aglycosylated IgGl and IgG4 versions of mAbl were generated. CT26 tumors were established in mice as described in Example 7. Mice received l50ug of either (a) isotype control; (b) mAbl as IgG4; (c) aglycosylated mAbl as IgG4; or (d) aglycosylated mAbl as IgGl.

As shown in FIGs. 13A and 13B, aglycosylated IgG4 and IgGl isotypes of the parental mAbl antibody had reduced effect on tumor volume in comparison to mAbl. However, efficacy was not completely abolished. Accordingly, these results indicated that while the anti-tumor efficacy of mAbl is not entirely Fc-dependent, it is enhanced by Fc receptor binding.

Example 14: Cross-Species Affinity of Anti-CD137 Antibodies

The anti-CD 137 antibodies were further tested for their binding to CD 137 from multiple species. Specifically, mAbl, mAb8 and mAblO were analyzed for binding to human, mouse, cynomolgus and canine CD 137. Kinetic experiments were performed on Octet HTX (ForteBio) in kinetics buffer (lx PBS, pH 7.4, 0.1 mg/ml BSA, and 0.002% Tween 20). Fc-, mouse IgG2a-, or His- tagged CD 137 (human, mouse, cyno or canine) were loaded for 5 minutes on pre-hydrated biosensors, AHC, AMC or NTA respectively. The sensors were then dipped into Fabs (0, 5.12, 12.8, 32, 80, 200 and 500 nM) for 5 minutes of association, following by 15 minutes of dissociation. Results were analyzed with ForteBio Data Analysis 9.0 and fit globally to a 1: 1 binding model to determine the apparent KD. KD for human and mouse CD 137 binding were confirmed by using antigens from different sources (ACRO Biosystems, Sino Biological and internal). The results are shown in Table 2 below.

Table 2: Cross-Species Affinity Example 15: Effect of Size of Tumor on Anti-Tumor Efficacy of Anti-CD137 Antibodies

To further characterize the anti-tumor efficacy of the anti-CDl37 antibodies, the anti tumor efficacy against large tumors was assessed. CT26 tumors were allowed to grow to approximately 500mm 3 prior to treatment. Parental mAbl, and affinity matured mAb8 and mAblO antibodies were administered at l50pg/mouse (n=6 mice/treatment group) on days 0, 3, 6 and 9 post tumor-establishment. The IgG4 isotype control antibody was used as a comparator.

As shown in FIGs. 14A-14C, the parental mAbl as well as the affinity-matured mAb8 and mAblO reduced tumor volume (FIGs. 14A-14B) and increased mouse survival (FIG. 14C) relative to the isotype control. mAb8 resulted in significantly greater anti-tumor efficacy compared to mAbl and mAblO. A separate study was conducted comparing the efficacy of mAb8 and 3H3 against large tumors using the same study design, except 25pg of the antibodies were administered on days 0, 7 and 14. FIG. 14D provides the results, showing 3H3 had no efficacy against large tumors, whereas mAb8 induced tumor regression.

As described in Example 14, mAb8 has an affinity for mouse CD137 that is comparable with the affinity of mAbl for human CD 137. While the disclosure is not bound by any particular theory or mechanism of action, it is believed that agonist anti-CD 137 antibodies with intermediate affinity may be even more useful for treating cancer.

Mice with no palpable tumor at day 70 were considered cured and re-challenged with subcutaneous injection of CT26 cells in the opposite flank. Specifically, mice with eradicated tumors were injected again with lxlO 5 CT26 cells in the left flank and tumor volume was calculated twice weekly (Length*(Width A 2)/2) using dial calipers. Five non-immunized (naive) mice were injected in the same manner as a control, respectively. Results of the re-challenge experiment are shown in FIG. 15. Eighty days after the subcutaneous injection of CT26 cells, none of the re-challenged mice formed tumors. In contrast, all of the naive mice that were injected with the same cells formed tumors. Therefore, all mice that were considered cured rejected CT26 tumors suggesting that mAbl can induce long-term protective memory immunity.

Example 16: Toxicity of Anti-CD137 Antibodies in Tumor-Bearing Mice

To determine the effect of anti-CD 137 antibodies (i.e., 3H3 and mAbl) on the level of intrahepatic T cells in tumor-bearing mice, mice from Example 7 were analyzed. Liver lymphocytes were collected and analyzed via flow cytometry. Specifically, single cell suspensions from the liver were obtained using the liver dissociation kit (Miltenyi cat# 130-105-807) and the gentle MACS Dissociator (Miltenyi). Cell suspensions were treated with complete medium to inactivate the enzymes and then passed through a 40 pm cell strainer. Red blood cells were lysed using ACK buffer. Cells were stained with antibodies against CD45, CD8 and CD4, and analyzed as described in Example 3.

FIGs. 16A and 16B show the number of CD4+ and CD8+ T cells, as a percentage of CD45+ cells, found in the livers of treated mice. The results indicated mAbl did not induce infiltration of intrahepatic T cells, demonstrating lower toxicity relative to antibody 3H3.

Example 17: Toxicity of Affinity-Matured Anti-CD137 Antibodies in Tumor-Bearing Mice

To assess toxicity-related effects mediated by anti-CDl37 antibodies (i.e., 3H3, mAbl, and mAb7-mAbl2), the cellular composition of spleens and livers of tumor-bearing mice from Example 8 were analyzed following antibody administration.

Intrahepatic (liver) and intrasplenic (spleen) T cells in tumor-bearing mice from Example 8 were collected and analyzed via flow cytometry. CD45+ cells from livers and spleens were assessed for CD3+, CD4+, or CD8+ expression following administration of anti-CDl37 antibodies or the isotype control antibody, as indicated. Results are shown in FIGs. 17A (splenic) and 17B (liver). The results indicated that the administration of parental mAbl as well as the affinity- matured antibodies (mAb7-mAbl2) had little to no effect on the percentage of intrahepatic or intrasplenic T cells relative to administration of the isotype control antibody. In contrast, administration of the 3H3 antibody resulted in elevated T cells in both the spleens and livers relative to the isotype control antibody, particularly CD3+ T cells and CD8+ T cells.

Further, CD45+ CD8+ T cells and CD45+CD4+ T cells from the livers and spleens of treated mice were assessed for expression of TIGIT, PD-l, or LAG-3 co-inhibitory receptors, as indicators of T cell activation or exhaustion, following administration of anti-CD 137 antibodies or the isotype control antibody. Levels of TIGIT, PD-l, and LAG-3 expression on CD8+ T cells and CD4+ T cells were measured by flow cytometry as described in previous Examples. FIGs. 18A- 18B and 19A-19B show that administration of the 3H3 antibody caused a significant increase in expression of these co-inhibitory receptor in both CD8+ T cells and CD4+ T cells, whereas administration of the parental mAbl or affinity-matured mAb7-mAbl2 antibodies resulted in expression of TIGIT, PD-l, or LAG-3 to a similar extent as seen after administration of the isotype control antibody. These results indicated the affinity matured antibodies did not induce systemic CD8+ T cell or CD4+ T cell activation.

Taken together, these results indicate that the parental mAbl and affinity-matured mAb7- mAbl2 antibodies exhibit lower potential for in vivo toxicity relative to the 3H3 comparator antibody. Absence of systemic T cell activation and expansion, particularly in the liver, after treatment with mAbl and affinity-matured mAb7-mAbl2 antibodies might translate into lower possibility of hep ato toxicity (transaminitis) in patients.

Example 18: Toxicity of Multiple Doses of Anti-CD137 Antibodies in Tumor-Bearing Mice

To confirm the lack of toxicity induced by mAbl, a repeated-dose toxicity study was conducted. Specifically, mice were administered anti-CDl37 antibodies mAbl, mAb8, or 3H3 weekly, for 4 weeks. mAbl and mAb8 were administered at either 10, 20, 40 or 80 mg/kg, whereas 3H3 was administered at either 10 or 80 mg/kg. On day 35, alanine aminotransaminase (ALT) levels in the plasma was determined using a fluorometric activity assay (Sigma, cat# MAK052), CD8+ T cells in the liver was determined using flowcytometry (as described above), and concentration of TNFoc in the plasma was determined using an electrochemiluminscence assay (Meso Scale Discovery, custom kit) according to manufacturer’s instructions.

FIG. 20A shows low levels of CD8+ T cells in the livers of mice administered mAbl and mAb8 at all 4 doses, whereas 3H3 induced high levels of CD8+ T cells at both the low (10 mg/kg) and high (80 mg/kg) doses. FIG. 20B shows low levels of ALT activity in the plasma of mice administered mAbl and mAb8 at all 4 doses, whereas 3H3 induced high levels of ALT at the 80 mg/kg dose. FIG. 20C shows low levels of TNFoc in the plasma of mice administered mAbl mAb8 at both low (10 mg/kg) and high (80 mg/kg) doses, whereas 3H3 induced high levels of TNFoc at both low (10 mg/kg) and high (80 mg/kg) doses.

In addition, livers from treated mice that received 80 mg/kg of the anti-CD 137 agonistic antibodies were sectioned and stained with H&E. From each animal, half of a liver lobe was embedded in OCT and fresh frozen in liquid nitrogen. Sectioning and H&E staining was performed by a histopathology laboratory (Mass Histology Service, Inc) according to standard procedures. FIG. 21 provides the results, which show inflammatory centrilobular foci in mice that received 3H3 (see arrows), but not mAbl or affinity-matured mAbl. Example 19: Immune Reprogramming with Anti-CD137 Antibodies

To determine the role of anti-CD 137 antibodies on immune cells in the tumor microenvironment, the CT26 tumor model was utilized. Specifically, CT26 tumors were established as described in Example 7. mAb8 was administered to mice on days 0, 3, 6 and 9 at a dose of 25ug. Tumors were analyzed on day 11 as described in Example 16.

Overall infiltration of immune cells into the tumor microenvironment was determined by measuring the quantity of CD45+ live cells. As shown in FIG. 22A, mAb8 significantly increased infiltration of immune cells into the tumor microenvironment.

The level of Treg cells in the tumor microenvironment was determined by measuring the quantity of CD25+ FOXP-3+ CD4+ tumor infiltrating lymphocytes. As shown in FIG. 22B, mAb8 significantly reduced the level of Tregs in the tumor microenvironment.

The effect of mAb8 on T-cell exhaustion was determined by measuring the level of PD- 1+TIGIT+ expression on CD8+ or CD4+ tumor infiltrating lymphocytes (TILs). FIG. 22C shows the results for CD8+ TILs, wherein PD-1+TIGIT+ cells were reduced in the tumor microenvironment when mAb8 was administered. Similar results were observed for CD4+ TILs (data not shown). These results indicate mAb8 protects and/or reverses T-cell exhaustion.

In addition, the effect of mAb8 on tumor-associated macrophages was analyzed. Specifically, F4/80+CDl lb+CD45+ cells were measured and a reduction in tumor-associated macrophages was observed with treatment of mAb8.

In a separate study, the effect of anti-CDl37 antibodies (/. <? ., mAbl and 3H3) on peripheral immune cells was assessed. Specifically, spleens from CT26 tumor-bearing mice treated with mAbl or 3H3 on days 0 and 3 at a dose of l50ug, were analyzed on day 7. As shown in FIG. 23, anti-CD 137 antibody 3H3 induced TIGIT and PD-l expression on CD8+ and CD4+ T cells, as well as increased CD8+CD25+ and CD4+Foxp3+ cells. In addition, 3H3 induced both CD8+ and CD4+ effector memory cells. In contrast, anti-CD 137 antibody mAbl did not significantly induce CD8+TIGIT+PD-1+, CD8+CD25+, and CD4+Foxp3+ T cells. Further, mAbl did not induce CD8+ or CD4+ effector memory cells.

Overall, these results indicate anti-CD 137 antibodies mAbl and mAb8 induce dramatic immune reprogramming within the tumor microenvironment and has less of an effect, if any, on peripheral immune cells. Example 20: Enhancement of Murine T Cell Activation by Anti-CD137 Antibodies

The agonistic activity of the anti-CDl37 antibodies was further analyzed by assessing the stimulation of IL-2 production in a murine ovalbumin stimulation assay. In a 96-well plate, JAWS- II dendritic cell-like cells were plated at 10 4 cells/well and incubated overnight in the presence of murine IFNy (lOng/mL). Cells were incubated with 2pg/mL OVA/A2 peptide and incubated for 2 hours at 37°C, followed by incubation with 10 5 CD8+ T cells isolated from OT-I mouse spleen, which express OVA. Antibodies were added simultaneously. Atezolizumab (anti-PD-Ll antibody) and a mouse anti-PD-l antibody (RMP1-14), along with an IgG4 isotype control, were used as comparators. IL-2 concentration was determined by Meso Scale Discovery (MSD).

As shown in FIG. 24, mAb8 and mAblO significantly enhanced IL-2 production.

In addition to measuring IL-2 production, the percentages of CD25+CD8+ T cells and TIGIT+CD8+ T cells were analyzed using the same murine ovalbumin stimulation assay. Antibody 3H3 was included as a comparator. FIGs. 25A and 25B show that mAb8 and mAblO enhanced the expression of CD25, an activation marker, and spared the induction of TIGIT, an exhaustion marker. In contrast, 3H3 enhanced the expression of TIGIT.

Example 21: Effect of Anti-CD137 Antibodies on Cytokine Induction

To determine the effect of anti-CDl37 antibodies on cytokine induction by T cells, plate- bound antibodies were utilized. Three antibodies were used as comparators: mAb4, corresponding to urelumab (Bristol-Myers Squibb; CAS Number: 934823-49-1), a fully human IgG4-S228P agonistic antibody that targets the extracellular domain of CD137, but does not block ligand binding; mAb5, corresponding to utomilumab (Pfizer; CAS Number: 1417318-27-4), a fully human IgG2-S228P agonistic antibody that targets the extracellular domain of CD 137 and blocks ligand binding; and mAb6, a fully human IgG4-S228P agonistic antibody selected from the same library as mAbl and targets the extracellular domain of CD 137. The mAb6 antibody does not block ligand binding.

Human CD3+ T cells were isolated via negative selection and added to plates bound with anti-CDl37 antibodies and 1 pg/ml of anti-CD3. Anti-CDl37 antibodies were added at either lnM, lOnM, 50nM or lOOnM. Antibodies were coated overnight at 4°C.

72 hours after addition of the T cells, levels of IL-2, IFNy, TNFoc and IL-13 were assessed by Luminex kits (Luminex Corporation, Austin, TX) following the manufacturer’s instructions. Soluble anti-CD28 (2mg/mL) was used as a T cell activation control and the activation baseline was set using the plate bound anti-CD3. FIG. 26 shows the fold change in each cytokine level as it relates to the activation baseline. mAb4 (urelumab) showed the highest level of induction of each cytokine, with mAb 1 showing a lower level of induction but higher relative to mAb5 (utomilumab) and mAb6. These results indicate mAbl agonizes CD137 less than mAb4 (urelumab) at the same concentrations.

Example 22: Induction of Interferon-gamma (IFNy) by Anti-CD137 Antibodies

To further assess the agonistic activity of the anti-CD 137 antibodies, IFNy production was analyzed in a mixed lymphocyte reaction (MLR). mAb2, mAb4 (urelumab), mAb5 (utomilumab) and Keytruda, a humanized antibody that blocks PD-l (Merck) and is known to induce IFNy production, were used as comparators.

Peripheral blood mononuclear cells (PBMCs) were isolated from leukopaks (HemaCare, Van Nuys, CA) derived from three independent human donors (D985, D7603, and D5004). Total T cells were enriched from PBMC by negative selection using immunomagnetic cell separation (EasySep™; Stemcell Technologies, Vancouver BC). Monocytes were isolated from PBMCs using immunomagnetic cell separation (EasySep™; Stemcell Technologies, Vancouver BC). T cells were resuspended in complete RPMI at lxlO 6 cells/ml concentration and monocytes were adjusted to 5xl0 5 cells/ml respectively. In a 96-well plate, IOOmI of media containing T cells were plated at lxlO 5 cells/well density followed by adding IOOmI of monocyte cell suspension (E:T ratio 2: 1). Next, 50 pl of media containing various dilutions of CD137 antibodies was added. Plates were incubated at 37°C in a C0 2 incubator for five days. At the end of incubation period, culture supernatants were collected and IFNy levels were analyzed by MSD assay (Mesoscale Diagnostics, Rockville, MD). FIGs. 27A-27C show the concentration of IFNy as pg/mL at the final concentrations of antibodies tested, as indicated. These results indicated mAbl agonizes CD 137 less than mAb4 (urelumab), but to a similar extent as mAb5 (utomilumab) at the same concentrations.

In a separate study, IFNy induction was measured by utilizing CHO cells engineered to express CD32 (FCyRIIb) (CHO-CD32 cells). Specifically, CHO-CD32 cells were co-cultured with human T cells in the presence of soluble anti-CD3 and anti-CDl37 antibodies mAbl, mAb8, mAb4 and mAb5. Frozen PBMCs were thawed and rested overnight in T cell media (TCM) in a humidified 37°C 5% C02 incubator. The following day, CD3+ T cells were isolated with an untouched CD3 T cell isolation kit (Stemcell # 17951) before being mixed together with CHO cells (Gibco # A29127) transduced to express human CD32 (CHO-CD32), 250 ng/ml anti-CD3 (clone OKT3), and the anti-CDl37 or control antibodies. 100,000 T cells were mixed together with 50,000 CHO- CD32 cells. After incubation at 37°C for 3 days, supernatants were collected for analysis of secreted interferon-gamma (IFNy) via MesoScale Discovery (MSD).

FIG. 28 provides the results, showing mAb4 induced IFNy to the highest level and at low doses. In contrast, mAb5 induced almost no product of IFNy. Notably, mAbl and mAb8 provided a dose-dependent response and induced IFNy production between the levels induced by mAb4 and mAb5. Overall, these results indicate that mAb4 has superagonist activity, mAb5 has weaker activity, and mAbl and mAb8 have an intermediate activity compared to mAb4 and mAb5.

Example 23: Effect of Anti-CD137 Antibodies on Treg Cells

To further characterize the mechanism of action for anti-CDl37 antibodies, the effect of the antibodies on Treg cells was determined. Human Tregs were isolated using EasySep™ Human CD4+CDl27lowCD25+ Regulatory T Cell Isolation Kit (Stemcell Technologies, Cat #18063) and expanded for 13 days by immunocult anti-CD3/28 (Stemcell # 10971) in complete T cell media with 10% FBS. Specifically, the CHO-CD32 cells described in Example 21 were co-cultured with expanded human Treg cells, which were labeled with Cell-trace violet dye (Thermo Fisher, Cat #C34557) in the presence of soluble anti-CD3 (clone OKT3) and anti-CDl37 antibodies mAbl, mAb8, mAb4, mAb5 and isotype control. Proliferation of Treg cells was determined on Day 4.

FIG. 29 provides the results, showing mAb4 strongly induced Treg proliferation, even at low concentrations. In contrast, mAb5 had a very weak effect on Treg proliferation. Notably, mAbl and mAb8 showed moderate increases in Treg proliferation. Overall, these results confirm that mAb4 has superagonist activity, mAb5 has weak activity, and mAbl and mAb8 have an intermediate activity.

Example 24: Effect of Anti-CD137 Antibodies on Intracellular Signaling

To further assess the differences between anti-CD 137 agonistic antibodies, intracellular signaling was assessed in vitro. Specifically, CCL-119 T cells (ATCC; Cat# ATCC CCL-119) lentifected with NFk (Qiagen; Cat# CLS-013L-1) or SRF (Qiagen; Cat# CLS-010L-1) were stimulated with 250 ng/mL of plate-bound anti-CD3 (clone OKT3) in conjunction with varying concentrations of plate-bound mAbl, mAb8, mAb4, mAb5 and isotype control. After stimulation for 16 hours in RPMI media without additives, cells were lysed in lucif erase buffer (Promega; Cat# E263B) and relative light units (RLUs) were acquired on a BioTek Synergy Hl microplate reader (Cat# 11-120-533). Raw RLU data was then exported to Microsoft Excel and fold-induction was calculated by dividing RLUs from stimulated conditions over unstimulated controls.

FIG. 30 provides the results, showing minimal NI¾b and SRF activity of mAb4 and mAb5 relative to mAbl and its affinity-matured variant, mAb8. Overall, these results indicate mAbl induces intracellular signaling differently than mAb4 and mAb5.

Example 25: Effect of Anti-CD137 Antibodies on Macrophage Activation and Differentiation

It has previously been shown the hepatotoxicity induced by anti-mCDl37 agonistic antibody 3H3 was associated with expansion of macrophages and CD8+ T cells in the livers, and increased cytokine levels and ALT activity in the serum. Further, antibody 3H3 has been characterized as having similar properties as urelumab. As described herein, mAbl does not induce hepatotoxicity. Accordingly, anti-CD 137 agonistic antibodies were analyzed for their effect on macrophage activation in vitro.

Specifically, murine bone marrow-derived mouse macrophages were established from 10- week old female C57BL/6 mice (Charles River Laboratories). The femur and tibia bones were extracted from the musculature of the mice and bone marrow was flushed with PBS into 15 mL conical tubes on ice. The cells were centrifuged at 1500 rpm for 5 minutes and the supernatant was discarded. The cell pellet was broken and culture media (RPMI, 20% FBS, 50 pg/mL M-CSF (Shenandoah Biotechnology, Inc. ; Cat# 200-08-100), and pen/strep) was added. Cells were filtered on 40-micron mesh filter and plated into non-tissue culture treated petri dishes. After 3 days 10 mL of media was added to each petri dish. On day 7 of culture, media was removed and cells were washed with PBS (10 mL) twice. MACS buffer (PBS, 2 mM EDTA, and 0.5% FBS) was added to each dish and incubated at 37°C for 10 minutes. Cells were collected from the petri dishes and centrifuged at 1500 rpm for 5 minutes. These bone marrow derived macrophages were then stimulated with TLR9 agonist CpG in the presence of 50nm of anti-CD 137 antibodies mAbl, 3H3, or LOB 12.3 (mouse specific CD137 agonist antibody). Production of IL-6, TNFoc and IL-27 by murine bone marrow-derived macrophages was assessed from culture supernatants after 48 hours using an electrochemiluminscence assay (Meso Scale Discovery, custom kit) according to manufacturer’s instructions. FIG. 31 provides the results, which indicate mAbl did not induce secretion of proinflammatory cytokines by macrophages, whereas antibodies 3H3 and LOB 12.3 did.

The human monocyte-derived macrophages were generated by magnetically separating CDl4 + cells using anti-CDl4 microbeads (Miltenyi Biotech, Cat# 130-050-201) and maturing 7 days in the presence of 50ng/mL m-CSF. Human monocyte-derived macrophages were than stimulated with lOng/mL LPS in the presence of 5nm of anti-CDl37 antibodies mAbl, mAb4 or mAb5. Production of TNFoc was assessed after 48 hours using an electrochemiluminscence assay (Meso Scale Discovery, custom kit) according to manufacturer’s instructions. FIG. 32 provides the results, which indicate mAb4 and mAb5 induced macrophage activation significantly more than mAbl.

Further, THP1 monocytes were differentiated to macrophages with 2 mM

phorbol l2-myristate l3-acetate (PMA; Sigma; P1585) overnight. The macrophages were than cultured in the presence of 50nm of anti-CD 137 antibodies mAbl, mAb4 or mAb5 and CD64 expression was measured 48 hours later using flow cytometry (APC anti-human CD64 antibody clone 10.1; BioLegend; Cat#3050l3). FIG. 33 provides the results, which indicate mAb4 and mAb5 induced macrophage differentiation significantly more than mAbl.

While the disclosure is not bound by any particular theory or mechanism of action, overall, these results suggest mAbl spares hepatic toxicity due to reduced potential for macrophage activation.

Example 26: Expansion of human CD8+ T cells in vivo by anti-CD137 agonistic antibodies

To test the effect of CD137 agonistic antibodies on human cells in vivo, human PBMCs (7xl0 6 ) were intravenously injected to immunocompromised NSG mice (NOD.Cg- Prkdc scld Il2rg tmlw i l l SzJ; Jackson Laboratory; Cat# 005557). The mice were randomized to groups of 8 and received CD137 antibodies (200 pg/mouse) or vehicle control on days 0, 7 and 14. Peripheral blood from each mouse was collected on days 10, 20 and 29 for determination of human CD45 + (FITC anti-human CD45 clone HI30; BioLegend; Cat# 304038), CD8 + (Alexa Fluor® 647 anti-human CD8a clone HIT8a; Bio Legend; Cat#3009l8), and CD4 + (APC-Cy7 anti-human CD4 clone RPA-T4; Bd; Cat# 557871) engraftment using flow cytometry.

FIGs. 34A-34C show overall increase in numbers of hCD45 + cells and systemic hyper expansion of human CD8+ T cells in mice that received mAb4 at the expense of human CD4+ T cells. Notably, mAbl did not induce over activation of human T cells. Reduced potential of mAbl to activate human T cells in the periphery might contribute to reduced potential for toxicity.

Example 27: Characterization of B2M and/or MHC I Expression in Various Cancer Cell Lines

The expression of beta-2 microglobulin (B2M) and/or major histocompatibility complex I (MHC I) was measured using flow cytometry of various cancer cell lines included in Table 3.

TABLE 3: CANCER CELL LINES AND TYPES

FIGs. 35A, 35B, 36A, and 36B show histograms of CT26 cells and B 16-F10 cells which have been sorted using flow cytometry. The CT26 cells and B 16-F10 cancer cell lines were stained with fluorescently-labeled anti-B2M antibody (Biolegend, #154506), anti-MHC I antibody ebioscience, #12-5998-82), or isotype control. The anti-MHC I antibody used reacts with mouse H-2Kd and H-2Dd, and cross-reacts with H-2Kb, H-2Ks, H-2Kr. H-2Kq, and H-2Kp. The results demonstrate that while B2M and MHC I is expressed in CT26 cancer cells, there is no expression of B2M or MHC I in B 16-F10 cancer cells.

FIG. 37 shows flow cytometric histograms of CT26 cells, EMT6 cells, A20 cells, MC38 cells, RENCA cells, MB49 cells, 4T1 cells, ID-8 cells, B 16-F10 cells, PANC02 cells, HEPA1-6 cells, and Rl. l cells which have been sorted using flow cytometry. Prior to sorting, a sample from each cell line was stained with either fluorescently-labeled anti-MHC I antibody or fluorescently-labeled isotype control. As shown in FIG. 37, cell samples stained with anti-MHC I antibody are depicted in blue, and cell samples incubated with isotype control are depicted in red, with the exception of4Tl and ID-8, which are reversed (i.e., red indicates antibody and blue indicates isotype control). Level of expression of MHC I was determined relative to isotype control for each sample using flow cytometry.

The results demonstrate that some cancer cell lines express MHC I. In particular, CT26 cells, EMT6 cells, A20 cells, MC38 cells, 4T1 cells, ID-8 cells, and RENCA cells express MHC I. However, other cancer cells lines, such as MB49 cells, B 16-F10 cells, PANC02 cells, HEPA1-6 cells, and Rl.l cells do not express MHC I or express low levels of MHC I.

Example 28: Efficacy of Agonistic Anti-CD137 Antibodies in Tumor-Bearing Mice and the Effect MHC-I Expression in Tumors

The effect of MHC I expression on tumor size reduction with agonistic anti-CD 137 antibodies was evaluated in tumor-bearing mice that had been inoculated with one of the following cell lines: CT26 cells, EMT6 cells, A20 cells, MC38 cells, B 16-F10 cells, PANC02 cells, or 4T1 cells. To determine whether or not each of the cells lines express MHC I, the cells were stained with a fluorescent antibody against beta-2 microglobulin (B2M) or isotype control, and sorted using flow cytometry as described above in Example 27. Groups of the tumor-bearing mice were inoculated with a particular cancer cell line, and the effect of an anti-CD 137 antibody was assessed. Specifically, the tumor cells (passage 3) were maintained under aseptic conditions in DMEM Medium (Gibco cat# 11965-092), containing 10% 56°C -heat inactivated FBS (Gibco 10438-034), 1 mM sodium pyruvate (Gibco cat. # 11360-070), IX NEAA (Gibco cat# 11140-050) and IX MEM Vitamin solution (Gibco cat#l 1120-052). Cells were maintained at 37°C and 5% C0 2 . ETpon reaching 50-70% confluence, cells were passaged at a ratio of 1: 10, for a total of two passages, prior to in vivo implantation. Cells were harvested and counted using a Hemacytometer (Hausser Scientific Bright-Line #1492).

Balb/c female mice were purchased from Charles River Laboratories and were nine weeks old at the start of study. The tumor cells (1 xlO 5 cells per mouse in 0.1 mL PBS) were injected subcutaneously into the right flank of each mouse, and tumor volume was calculated twice weekly (Length* (Width A 2)/2) using dial calipers. On day 7 post-tumor inoculation, tumor-inoculated animals were sorted into separate, randomized groups of eight, and antibody treatments and control administrations were initiated. Body weights were recorded three times per week for the duration of the study.

The agonistic anti-CD 137 antibodies were administered at varying dosages and administration schedules. The treatment, dosage, and administration schedules for each cell line can be found in Table 4.

TABLE 4: CANCER CELL LINES TREATMENT, DOSAGE, AND ADMINISTRATION SCHEDULES

Dosing for the weekly dosing was on day 0, 7, 14, 21 and 28, if the experiment continued for this duration. For example, for a l5-day experiment dosing would be day 0, 7, 14. For the other three models, doses were administered on the indicated days only. The mice were sacrificed individually when their tumors reached to 2000 mm 3 .

Individual tumor volumes were measured and are shown for the anti-CD 137 antibody treated groups (middle panels) and control groups (lower panels) in Figures 38A, 38B, 38C, 38D, 39A, 39B, and 39C. Agonistic anti-CDl37 therapies resulted in inhibition of tumor growth compared to the control group in CT26 cancer cells, EMT6 cancer cells, A20 cancer cells, and MC38 cancer cells, all of which were shown to express MHC I.

Example 29: Efficacy of CD137 agonist antibody against CT26 tumors that lack B2M

The role of antigen presentation by tumor cells in response to CD 137 agonist antibody mAb8 was investigated in a beta-2 microglobulin knock out (b2M ko ) CT26 colorectal cancer model. b2M is a structural component of major histocompatibility complex (MHC) class I that stabilizes the tertiary structure of MHC-I. As shown in FIG. 40 b2M expression is absent in the knockout cell lines. In addition, MHC Class I is also absent in the knockout cell lines. There is no change in expression of MHC Class I in the knockout cell lines even in the presence of IFNy.

CT26- b2M ko cells were generated using ribonucleoprotein (RNP) based CRISPR-Cas9 (Synthego Corp., Menlo Park, CA) gene editing system. Cas9 nuclease (20 pMols) and sgRNA- 873 or sgRNA-882 (180 pmols) complex were prepared and transfected to CT26 cells (5xl0 5 ) using SF Cell Line 4D-NUCLEOFECTOR® X Kit (catalog# V4XC-2012). Transfected cells were grown in complete media for a week and b2hT ne MHC-T ve cells were sorted by FACS (BD FACSARIA Fusion). A second round of sorting was performed to improve the purity of the b2M knocked-out cell lines to more than 98%. Sequencing confirmed deletions in the b2M locus at the sites of sgRNA binding. As shown herein, two different CT26- b2M ko cell lines (873 CNT and 882 CNT) were generated with two different CRISPR-Cas9 ribonucleoprotein complexes that targeted different portions of the b2M sequence.

b2M, MHC-I and PD-L1 are known to be up regulated in response to IFNy stimulation. To further confirm complete b2M knock-out, CT26 parental (Parental CNT) and CT26- b2M ko cells were treated with 100 pg/ml IFNy (Catalog # 200-16, Shenandoah Biotetchnology) overnight and expression of b2M (Biolegend Cat. #154506), MHC-I (ebioscience Cat. #12-5998-82), and PD- Ll (Biolegend Cat. # 124313) were analyzed by flow cytometry. As can be seen in Figure 40, CT26 parental cells upregulated the expression of b2M, MHC-I and PD-L1, whereas the CT26- B2M KO cells were only able to upregulate PD-L1, but not b2M or MHC-I. This demonstrates that the CT26^2M KO cells are responsive to IFNy, but cannot upregulate b2M or MHC-I due to CRISPR mediated knock out.

Anti-tumor activity of CD 137 agonist antibody mAb8 was tested in mice carrying either CT26 parental or CT- b2M ko tumors. B alb/c female mice were purchased from Charles River Laboratories and were seven weeks old at the start of study. CT26 parental or CT26- b2M ko cells (CT26 B2M KO 882 and CT26 B2 KO 883 ) (lxlO 5 per mouse) generated with two different CRISPR- Cas9 ribonucleoprotein complexes were suspended in sterile PBS and injected subcutaneously into the right flank. Ten days after inoculation, when the tumors had reached a mean volume of -100 mm 3 , mice were randomized to receive isotype control or mAb8 (25 pg) intraperitoneally on days 0, 7, 14, and 21. Tumor width and length were measured using dial calipers and tumor volumes were calculated by the formula Length*(Width A 2)/2. Body weights were recorded two times per week for the duration of the study. All mice in the control groups developed tumors that grew rapidly, whereas treatment with mAb8 resulted in complete tumor regressions in 3 out of 6 mice in the CT26 parental tumors, while no cures were observed in mice carrying CT26- b2M ko tumors (Figure 41A-41C).

These data demonstrate that treatment with an anti-CD 137 antibody results in the inhibition and/or regression of tumor growth in mice carrying tumors expressing MHC I, as well as prolonged survival.

TABLE 5: ANTIBODY COMBINATION TABLE

TABLE 6: ALTERNATIVE ANTIBODY COMBINATION TABLE

TABLE 7: SEQUENCE LISTING

TABLE 8: SEQUENCE LISTING (continued)