Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
A THERAPEUTIC COMBINATION COMPRISING A T1GIT ANTAGONIST, A PD-1 ANTAGONIST, AND A CHEMOTHERAPEUTIC AGENT(S)
Document Type and Number:
WIPO Patent Application WO/2023/018677
Kind Code:
A1
Abstract:
Provided herein are methods of treating cancer, which comprise administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) one or more chemotherapeutic agents. Also provided are kits containing such agents and uses of therapeutic combinations of such agents for the treatment of cancer.

Inventors:
HEALY JANE ANNE (US)
SHRAWANKUMAR JHA SUJATA (US)
KEENAN TANYA (US)
SIEGEL ABBY (US)
PINHEIRO ELAINE (US)
SNYDER ALEXANDRA (US)
CAI MINGMEI (US)
Application Number:
PCT/US2022/039767
Publication Date:
February 16, 2023
Filing Date:
August 09, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK SHARP & DOHME LLC (US)
HEALY JANE ANNE (US)
SHRAWANKUMAR JHA SUJATA (US)
KEENAN TANYA (US)
SIEGEL ABBY (US)
PINHEIRO ELAINE M (US)
SNYDER ALEXANDRA (US)
CAI MINGMEI (US)
International Classes:
A61K31/00; A61K39/395; A61P35/00; C07K16/28; A61K31/136; A61K31/337; A61K31/473
Domestic Patent References:
WO2021154761A12021-08-05
WO2020096915A12020-05-14
Attorney, Agent or Firm:
MERCK SHARP & DOHME LLC (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS: 1. A method of treating cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I) and I); sented by Formula (II) (II). 2. The method of claim 1, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). 3. The method of claim 2, wherein the cancer is esophageal cancer. 4. The method of claim 3, wherein the esophageal cancer is advanced esophageal cancer. 5. The method of claim 3, wherein the esophageal cancer is metastatic esophageal cancer. 6. The method of claim 3, wherein the esophageal cancer is locally advanced unresectable. 7. The method of claim 3, wherein the esophageal cancer is metastatic adenocarcinoma.

8. The method of claim 3, wherein the esophageal cancer is squamous cell carcinoma of the esophagus (ESCC). 9. The method of claim 3, wherein the esophageal cancer is advanced Siewert type 1 adenocarcinoma of gastroesophageal junction (GEJ). 10. The method of claim 3, wherein the esophageal cancer is metastatic Siewert type 1 adenocarcinoma of GEJ. 11. A kit comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). 12. The kit of claim 11, further comprising instructions for administering to a human patient in need thereof the TIGIT antagonist, the PD-1 antagonist, 5-fluorouracil, and cisplatin. 13. Use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). 14. The use of claim 13, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). 15. The use of claim 14, wherein the cancer is esophageal cancer. 16. The use of claim 15, wherein the esophageal cancer is advanced esophageal cancer.

17. The use of claim 15, wherein the esophageal cancer is metastatic esophageal cancer. 18. The use of claim 15, wherein the esophageal cancer is locally advanced unresectable. 19. The use of claim 15, wherein the esophageal cancer is metastatic adenocarcinoma. 20. The use of claim 15, wherein the esophageal cancer is squamous cell carcinoma of the esophagus (ESCC). 21. The use of claim 15, wherein the esophageal cancer is advanced Siewert type 1 adenocarcinoma of gastroesophageal junction (GEJ). 22. The use of claim 15, wherein the esophageal cancer is metastatic Siewert type 1 adenocarcinoma of GEJ. 23. The method, kit, or use of any one of claims 1-22, wherein the PD-1 antagonist and the TIGIT antagonist are formulated separately. 24. The method, kit, or use of any one of claims 1-22, wherein the PD-1 antagonist and the TIGIT antagonist are in a fixed dose combination. 25. The method, kit, or use of any one of claims 1-22, wherein the PD-1 antagonist and the TIGIT antagonist are co-formulated. 26. The method, kit, or use of any one of claims 1-25, wherein the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. 27. The method, kit, or use of any one of claims 1-25, wherein the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. 28. The method, kit, or use of claim 26, wherein the anti-human PD-1 monoclonal antibody is a humanized antibody. 29. The method, kit, or use of claim 26, wherein the anti-human PD-1 monoclonal antibody is a human antibody.

30. The method, kit, or use of any one of claims 1-29, wherein the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. 31. The method, kit, or use of claim 30, wherein the anti-human TIGIT monoclonal antibody is a humanized antibody. 32. The method, kit, or use of claim 30, wherein the anti-human TIGIT monoclonal antibody is a human antibody. 33. The method, kit, or use of claim 26, wherein the anti-human PD-1 monoclonal antibody is pembrolizumab. 34. The method, kit, or use of claim 26, wherein the anti-human PD-1 monoclonal antibody is nivolumab. 35. The method, kit, or use of claim 26, wherein the anti-human PD-1 monoclonal antibody is cemiplimab. 36. The method, kit, or use of claim 30, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 37. The method, kit, or use of claim 36, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 38. The method, kit, or use of claim 37, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 39. The method, kit, or use of any one of claims 1-25, wherein: (a) the PD-1 antagonist is pembrolizumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 40. The method, kit, or use of any one of claims 1-25, wherein: (a) the PD-1 antagonist is nivolumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 41. The method, kit, or use of any one of claims 1-25, wherein: (a) the PD-1 antagonist is cemiplimab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 42. The method, kit or use of claims 39-41, wherein the TIGIT antagonist comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 43. The method, kit or use of claim 42, wherein the TIGIT antagonist comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 44. The method of claims 39, 42, or 43, wherein the human patient is administered about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks. 45. The method of claim 44, wherein the human patient is administered about 200 mg pembrolizumab once every three weeks. 46. The method of claim 40, wherein the human patient is administered about 240 mg or about 3 mg/kg nivolumab once every two weeks, or about 480 mg nivolumab once every four weeks. 47. The method of claim 41, wherein the human patient is administered about 350 mg cemiplimab once every three weeks. 48. The method of any one of claims 39-43, wherein the human patient is administered from about 100 mg to about 500 mg of the TIGIT antagonist once every three weeks. 49. The method of claim 48, wherein the human patient is administered about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, or about 300 mg of the TIGIT antagonist once every three weeks. 50. The method of claim 49, wherein the human patient is administered about 200 mg of the TIGIT antagonist once every three weeks. 51. The method of claims 39-43, wherein the human patient is administered from about 400 mg of the TIGIT antagonist once every six weeks.

52. The method of any one of claims 1-5 and 25-51, wherein the human patient is administered a total of about 3000 mg/m2 to about 4000 mg/m25-fluorouracil per 3 week cycle. 53. The method of claim 52, wherein the human patient is administered about 4000 mg/m25-fluorouracil per 3 week cycle. 54. The method of claim 53, wherein the human patient is administered about 800 or about 1000 mg/m25-fluorouracil a day. 55. The method of claim 52, wherein the human patient is administered a total of about 3000 mg/m25-fluorouracil per 3 week cycle. 56. The method of claim 55, wherein the human patient is administered about 600 mg/m2 5-fluorouracil a day. 57. The method of claims 1-5 and 25-55, wherein the human patient is administered about 40 mg/m2 to about 100 mg/m2 cisplatin once every three weeks. 58. The method of claim 57, wherein the human patient is administered about 80 mg/m2 cisplatin once every three weeks. 59. The method of claim 57, wherein the human patient is administered about 60 mg/m2 cisplatin once every three weeks. 60. A method of treating esophageal cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising: (a) about 200 mg pembrolizumab; (b) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110; (c) a total of about 3000 mg/m2 or 4000 mg/m25-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m2 or 80 mg/m2 cisplatin represented by Formula (II). 61. The method of claim 60, wherein each of (a), (b), and (d) is administered once every three weeks. 62. The method of claims 60-61, wherein 5-fluorouracil is administered five times every three weeks. 63. The method of claims 60-62, wherein (a) and (b) are administered on the same day, and wherein (a) and (b) are administered sequentially or concurrently. 64. The method of claim 63, wherein (a) and (b) are co-formulated. 65. The method of claim 63, wherein (a) and (b) are in a fixed dose combination. 66. The method of claims 60-65, wherein the esophageal cancer is advanced esophageal cancer. 67. The method of claims 60-65, wherein the esophageal cancer is metastatic esophageal cancer. 68. The method of claims 60-65, wherein the esophageal cancer is locally advanced unresectable. 69. The method of claims 60-65, wherein the esophageal cancer is metastatic adenocarcinoma. 70. The method of claims 60-65, wherein the esophageal cancer is squamous cell carcinoma of the esophagus (ESCC). 71. A method of treating cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. I) 72. The method of claim 71, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). 73. The method of claim 72, wherein the cancer is TNBC. 74. The method of claim 73, wherein the TNBC is recurrent unresectable. 75. The method of claim 73, wherein the TNBC is metastatic TNBC. 76. A kit comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. 77. The kit of claim 76, further comprising instructions for administering to a human patient in need thereof the TIGIT antagonist, the PD-1 antagonist, and paclitaxel. 78. Use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. 79. The use of claim 78, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). 80. The use of claim 79, wherein the cancer is TNBC. 81. The use of claim 80, wherein the TNBC is recurrent unresectable. 82. The use of claim 80, wherein the TNBC is metastatic TNBC. 83. The method, kit, or use of any one of claims 71-82, wherein the PD-1 antagonist and the TIGIT antagonist are in a fixed dose combination. 84. The method, kit, or use of any one of claims 71-82, wherein the PD-1 antagonist and the TIGIT antagonist are formulated separately. 85. The method, kit, or use of any one of claims 71-82, wherein the PD-1 antagonist and the TIGIT antagonist are co-formulated. 86. The method, kit, or use of any one of claims 71-83, wherein the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. 87. The method, kit, or use of any one of claims 71-83, wherein the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. 88. The method, kit, or use of claim 86, wherein the anti-human PD-1 monoclonal antibody is a humanized antibody. 89. The method, kit, or use of claim 86, wherein the anti-human PD-1 monoclonal antibody is a human antibody.

90. The method, kit, or use of any one of claims 71-89, wherein the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. 91. The method, kit, or use of claim 90, wherein the anti-human TIGIT monoclonal antibody is a humanized antibody. 92. The method, kit, or use of claim 90, wherein the anti-human TIGIT monoclonal antibody is a human antibody. 93. The method, kit, or use of claim 86, wherein the anti-human PD-1 monoclonal antibody is pembrolizumab. 94. The method, kit, or use of claim 86, wherein the anti-human PD-1 monoclonal antibody is nivolumab. 95. The method, kit, or use of claim 86, wherein the anti-human PD-1 monoclonal antibody is cemiplimab. 96. The method, kit, or use of claim 90, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 97. The method, kit, or use of claim 96, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 98. The method, kit, or use of claim 97, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 99. The method, kit, or use of any one of claims 71-83, wherein: (a) the PD-1 antagonist is pembrolizumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 100. The method, kit, or use of any one of claims 71-83, wherein: (a) the PD-1 antagonist is nivolumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 101. The method, kit, or use of any one of claims 71-83, wherein: (a) the PD-1 antagonist is cemiplimab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 102. The method, kit or use of claims 99-101, wherein the TIGIT antagonist comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 103. The method, kit or use of claim 103, wherein the TIGIT antagonist comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 104. The method of claims 99, 102, or 103, wherein the human patient is administered about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks. 105. The method of claim 104, wherein the human patient is administered about 200 mg pembrolizumab once every three weeks. 106. The method of claim 100, wherein the human patient is administered about 240 mg or about 3 mg/kg nivolumab once every two weeks, or about 480 mg nivolumab once every four weeks. 107. The method of claim 101, wherein the human patient is administered about 350 mg cemiplimab once every three weeks. 108. The method of any one of claims 99-103, wherein the human patient is administered from about 100 mg to about 500 mg of the TIGIT antagonist once every three weeks. 109. The method of claim 108, wherein the human patient is administered about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, or about 300 mg of the TIGIT antagonist once every three weeks. 110. The method of claim 109, wherein the human patient is administered about 200 mg of the TIGIT antagonist once every three weeks. 111. The method of any one of claims 71-75 and 83-110, wherein the human patient is administered about 70 mg/m2 to about 100 mg/m2 paclitaxel once every week.

112. The method of claim 111, wherein the human patient is administered about 90 mg/m2 paclitaxel once every week. 113. The method of claim 111, wherein the human patient is administered about 80 mg/m2 paclitaxel once every week. 114. A method of treating TNBC, comprising administering to a human patient in need thereof a therapeutic combination comprising: (a) about 200 mg pembrolizumab; (b) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110; and (c) about 80 mg/m2 or 90 mg/m2 paclitaxel represented by Formula (III). 115. The method of claim 114, wherein each of (a), and (b) is administered once every three weeks. 116. The method of claims 114-115, wherein (c) is administered once every week. 117. The method of claim 114-116, wherein (a) and (b) are administered on the same day, and wherein (a) and (b) are administered sequentially or concurrently. 118. The method of claim 114-117, wherein the TNBC is recurrent unresectable. 119. The method of claim 114-117, wherein the TNBC is metastatic. 120. The method of claim 114-116, wherein (a) and (b) are co-formulated. 121. The method of claim 114-116, wherein (a) and (b) are in a fixed dose combination.

122. A method of treating cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and ); by Formula (II) (II). 123. The method of claim 122, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). 124. The method of claim 123, wherein the cancer is biliary cancer. 125. The method of claim 124, wherein the biliary cancer is metastatic biliary cancer. 126. The method of claim 124, wherein the esophageal cancer is locally recurrent unresectable. 127. A kit comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). 128. The kit of claim 127, further comprising instructions for administering to a human patient in need thereof the TIGIT antagonist, the PD-1 antagonist, gemcitabine, and cisplatin. 129. Use of a therapeutic combination of for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). 130. The use of claim 129, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). 131. The use of claim 130, wherein the cancer is biliary cancer. 132. The use of claim 131, wherein the biliary cancer is metastatic biliary cancer. 133. The use of claim 131, wherein the biliary cancer is locally advanced unresectable. 134. The method, kit, or use of any one of claims 122-133, wherein the PD-1 antagonist and the TIGIT antagonist are formulated separately. 135. The method, kit, or use of any one of claims 122-133, wherein the PD-1 antagonist and the TIGIT antagonist are in a fixed dose combination. 136. The method, kit, or use of any one of claims 122-133, wherein the PD-1 antagonist and the TIGIT antagonist are co-formulated.

137. The method, kit, or use of any one of claims 122-136, wherein the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. 138. The method, kit, or use of any one of claims 122-136, wherein the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. 139. The method, kit, or use of claim 137, wherein the anti-human PD-1 monoclonal antibody is a humanized antibody. 140. The method, kit, or use of claim 137, wherein the anti-human PD-1 monoclonal antibody is a human antibody. 141. The method, kit, or use of any one of claims 122-140, wherein the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. 142. The method, kit, or use of claim 141, wherein the anti-human TIGIT monoclonal antibody is a humanized antibody. 143. The method, kit, or use of claim 141, wherein the anti-human TIGIT monoclonal antibody is a human antibody. 144. The method, kit, or use of claim 137, wherein the anti-human PD-1 monoclonal antibody is pembrolizumab. 145. The method, kit, or use of claim 137, wherein the anti-human PD-1 monoclonal antibody is nivolumab. 146. The method, kit, or use of claim 137, wherein the anti-human PD-1 monoclonal antibody is cemiplimab. 147. The method, kit, or use of claim 137, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 148. The method, kit, or use of claim 137, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 149. The method, kit, or use of claim 137, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 150. The method, kit, or use of any one of claims 122-143, wherein: (a) the PD-1 antagonist is pembrolizumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 151. The method, kit, or use of any one of claims 122-143, wherein: (a) the PD-1 antagonist is nivolumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110.

152. The method, kit, or use of any one of claims 122-143, wherein: (a) the PD-1 antagonist is cemiplimab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 153. The method, kit or use of claims 150-152, wherein the TIGIT antagonist comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 154. The method, kit or use of claim 153, wherein the TIGIT antagonist comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 155. The method of claims 150, 153, or 154, wherein the human patient is administered about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks. 156. The method of claim 150, wherein the human patient is administered about 200 mg pembrolizumab once every three weeks. 157. The method of claim 151, wherein the human patient is administered about 240 mg or about 3 mg/kg nivolumab once every two weeks, or about 480 mg nivolumab once every four weeks. 158. The method of claim 152, wherein the human patient is administered about 350 mg cemiplimab once every three weeks. 159. The method of any one of claims 150-158, wherein the human patient is administered from about 100 mg to about 500 mg of the TIGIT antagonist once every three weeks. 160. The method of claim 159, wherein the human patient is administered about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, or about 300 mg of the TIGIT antagonist once every three weeks. 161. The method of claim 160, wherein the human patient is administered about 200 mg of the TIGIT antagonist once every three weeks. 162. The method of claims 150-158, wherein the human patient is administered from about 400 mg of the TIGIT antagonist once every six weeks. 163. The method of any one of claims 122-126 and 134-162, wherein the human patient is administered about 800 mg/m2 to about 1000 mg/m2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks. 164. The method of claim 163, wherein the human patient is administered about 1000 mg/m2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks. 165. The method of claim 163, wherein the human patient is administered about 800 mg/m2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks. 166. The method of claims 122-126 and 134-165, wherein the human patient is administered about 20 mg/m2 to about 25 mg/m2 cisplatin once every three weeks. 167. The method of claim 166, wherein the human patient is administered about 25 mg/m2 cisplatin once every three weeks. 168. The method of claim 166, wherein the human patient is administered about 20 mg/m2 cisplatin once every three weeks. 169. A method of treating biliary cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising: (a) about 200 mg pembrolizumab; (b) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110; (c) about 800 mg/m2 or 1000 mg/m2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 60 mg/m2 or 80 mg/m2 cisplatin represented by Formula (II). 170. The method of claim 169, wherein each of (a), (b), (c) and (d) is administered once every three weeks. 171. The method of claims 169-170, wherein (a) and (b) are administered on the same day, and wherein (a) and (b) are administered sequentially or concurrently. 172. The method of claim 171, wherein (a) and (b) are co-formulated. 173. The method of claim 171, wherein (a) and (b) are in a fixed dose combination. 174. The method of claims 169-173, wherein the biliary cancer is metastatic biliary cancer. 175. The method of claims 169-173, wherein the biliary cancer is locally recurrent unresectable. 176. A method of treating cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and ); ula (VI) I). 177. The method of claim 176, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC), and gastric cancer. 178. The method of claim 177, wherein the cancer is gastric cancer. 179. The method of claim 178, wherein the gastric cancer is advanced or GEJ adenocarcinoma. 180. The method of claim 178, wherein the gastric cancer is HER2 negative. 181. The method of claim 178, wherein the gastric cancer is previously untreated. 182. A kit comprising an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). 183. The kit of claim 183, further comprising instructions for administering to a human patient in need thereof the TIGIT antagonist, the PD-1 antagonist, capecitabine, and oxaliplatin. 184. Use of a therapeutic combination of for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). 185. The use of claim 184, wherein the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC), and gastric cancer. 186. The use of claim 185, wherein the cancer is gastric cancer. 187. The use of claim 186, wherein the gastric cancer is advanced or GEJ adenocarcinoma. 188. The use of claim 186, wherein the gastric cancer is HER2 negative. 189. The use of claim 186, wherein the gastric cancer is previously untreated. 190. The method, kit, or use of any one of claims 176-189, wherein the PD-1 antagonist and the TIGIT antagonist are formulated separately. 191. The method, kit, or use of any one of claims 176-189, wherein the PD-1 antagonist and the TIGIT antagonist are in a fixed dose combination. 192. The method, kit, or use of any one of claims 176-189, wherein the PD-1 antagonist and the TIGIT antagonist are co-formulated. 193. The method, kit, or use of any one of claims 176-192, wherein the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. 194. The method, kit, or use of any one of claims 176-192, wherein the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. 195. The method, kit, or use of claim 193, wherein the anti-human PD-1 monoclonal antibody is a humanized antibody. 196. The method, kit, or use of claim 193, wherein the anti-human PD-1 monoclonal antibody is a human antibody. 197. The method, kit, or use of any one of claims 176-196, wherein the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. 198. The method, kit, or use of claim 197, wherein the anti-human TIGIT monoclonal antibody is a humanized antibody. 199. The method, kit, or use of claim 197, wherein the anti-human TIGIT monoclonal antibody is a human antibody. 200. The method, kit, or use of claim 196, wherein the anti-human PD-1 monoclonal antibody is pembrolizumab. 201. The method, kit, or use of claim 196, wherein the anti-human PD-1 monoclonal antibody is nivolumab. 202. The method, kit, or use of claim 196, wherein the anti-human PD-1 monoclonal antibody is cemiplimab. 203. The method, kit, or use of claim 193, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 204. The method, kit, or use of claim 193, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 205. The method, kit, or use of claim 193, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 206. The method, kit, or use of any one of claims 176-192, wherein: (a) the PD-1 antagonist is pembrolizumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 207. The method, kit, or use of any one of claims 176-192, wherein: (a) the PD-1 antagonist is nivolumab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 208. The method, kit, or use of any one of claims 176-192, wherein: (a) the PD-1 antagonist is cemiplimab; and (b) the TIGIT antagonist comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 209. The method, kit or use of claims 206-208, wherein the TIGIT antagonist comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 210. The method, kit or use of claim 209, wherein the TIGIT antagonist comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. 211. The method of claims 206, 209, or 210, wherein the human patient is administered about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks. 212. The method of claim 211, wherein the human patient is administered about 200 mg pembrolizumab once every three weeks. 213. The method of claim 207, wherein the human patient is administered about 240 mg or about 3 mg/kg nivolumab once every two weeks, or about 480 mg nivolumab once every four weeks. 214. The method of claim 208, wherein the human patient is administered about 350 mg cemiplimab once every three weeks. 215. The method of any one of claims 176-214, wherein the human patient is administered from about 100 mg to about 500 mg of the TIGIT antagonist once every three weeks.

216. The method of claim 215, wherein the human patient is administered about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, or about 300 mg of the TIGIT antagonist once every three weeks. 217. The method of claim 216, wherein the human patient is administered about 200 mg of the TIGIT antagonist once every three weeks. 218. The method of claims 176-214, wherein the human patient is administered from about 400 mg of the TIGIT antagonist once every six weeks. 219. The method of any one of claims 176-181 and 190-218, wherein the human patient is administered about 500 mg/m2 to about 1500 mg/m2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. 220. The method of claim 163, wherein the human patient is administered about 750 mg/m2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. 221. The method of claim 163, wherein the human patient is administered about 1000 mg/m2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. 222. The method of claims 176-181 and 190-221, wherein the human patient is administered about 80 mg/m2 to about 150 mg/m2 oxaliplatin once every three weeks. 223. The method of claim 166, wherein the human patient is administered about 100 mg/m2 oxaliplatin once every three weeks. 224. The method of claim 166, wherein the human patient is administered about 130 mg/m2 oxaliplatin once every three weeks. 225. A method of treating gastric cancer, comprising administering to a human patient in need thereof a therapeutic combination comprising an effective amount of : (a) about 200 mg pembrolizumab; (b) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110; (c) about 750 mg/m2 or 1000 mg/m2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m2 or 130 mg/m2 oxaliplatin represented by Formula (VI). 226. The method of claim 226, wherein each of (a), (b) and (d) is administered once every three weeks and (c) is administered twice a day. 227. The method of claims 225-226, wherein (a) and (b) are administered on the same day, and wherein (a) and (b) are administered sequentially or concurrently. 228. The method of claim 227, wherein (a) and (b) are co-formulated. 229. The method of claim 227, wherein (a) and (b) are in a fixed dose combination. 230. The method of claims 225-229, wherein the gastric cancer is advanced or GEJ adenocarcinoma. 231. The method of claims 225-229, wherein the gastric cancer is HER2 negative. 232. The method of claims 225-229, wherein the gastric cancer is previously untreated. 233. A composition or kit comprising a PD-1 antagonist and/or TIGIT antagonist and a hyaluronan degrading enzyme. 234. The composition or kit of claim 233, comprising a PD-1 antagonist and a TIGIT antagonist, and further comprising 5-fluorouracil and cisplatin. 235. The composition or kit of claim 233, comprising a PD-1 antagonist and a TIGIT antagonist, and further comprising paclitaxel or a pharmaceutically acceptable salt thereof. 236. The composition or kit of claim 233, comprising a PD-1 antagonist and a TIGIT antagonist, and further comprising gemcitabine or a pharmaceutically acceptable salt thereof and cisplatin. 237. The composition or kit of claim 233, comprising a PD-1 antagonist and a TIGIT antagonist, and further comprising capecitabine or a pharmaceutically acceptable salt thereof and oxaliplatin. 238. A method comprising administering a PD-1 antagonist and/or a TIGIT antagonist and a hyaluronan degrading enzyme. 239. The method of claim 238, comprising administering a PD-1 antagonist and a TIGIT antagonist, and further comprising administering 5-fluorouracil and cisplatin. 240. The method of claim 238, comprising administering a PD-1 antagonist and a TIGIT antagonist, and further comprising administering paclitaxel or a pharmaceutically acceptable salt thereof. 241. The method of claim 238, comprising administering a PD-1 antagonist and a TIGIT antagonist, and further comprising administering gemcitabine or a pharmaceutically acceptable salt thereof and cisplatin. 242. The method of claim 238, comprising administering a PD-1 antagonist and a TIGIT antagonist, and further comprising administering capecitabine or a pharmaceutically acceptable salt there and oxaliplatin 243. The composition or kit of any one of claims 233-237 comprising a PD-1 antagonist and a TIGIT antagonist, or the method of any one of claims 238-242 comprising administering a PD-1 antagonist and a TIGIT antagonist; wherein the PD-1 antagonist and the TIGIT antagonist are formulated separately. 244. The composition or kit of any one of claims 233-237 comprising a PD-1 antagonist and a TIGIT antagonist, or the method of any one of claims 238-242 comprising administering a PD-1 antagonist and a TIGIT antagonist; wherein the PD-1 antagonist and the TIGIT antagonist are in a fixed dose combination. 245. The composition, or kit of any one of claims 233-237 comprising a PD-1 antagonist and a TIGIT antagonist, or the method of any one of claims 238-242 comprising administering a PD-1 antagonist and a TIGIT antagonist; wherein the PD-1 antagonist and the TIGIT antagonist are co-formulated. 246. The composition or kit of any one of claims 233-237 comprising a PD-1 antagonist and a TIGIT antagonist, or the method of any one of claims 238-245 comprising administering a PD-1 antagonist and a TIGIT antagonist; wherein the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. 247. The composition or kit of any one of claims 233-237 comprising a PD-1 antagonist and a TIGIT antagonist, or the method of any one of claims 238-245 comprising administering a PD-1 antagonist and a TIGIT antagonist; wherein the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. 248. The composition, kit, or method of claim 246, wherein the anti-human PD-1 monoclonal antibody is a humanized antibody. 249. The composition, kit, or method of claim 246, wherein the anti-human PD-1 monoclonal antibody is a human antibody. 250. The composition or kit of any one of claims 233-237 comprising a PD-1 antagonist and a TIGIT antagonist, or the method of any one of claims 238-249 comprising administering a PD-1 antagonist and a TIGIT antagonist; wherein the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. 251. The composition, kit, or method of claim 250, wherein the anti-human TIGIT monoclonal antibody is a humanized antibody. 252. The composition, kit, or method of claim 250, wherein the anti-human TIGIT monoclonal antibody is a human antibody.

253. The composition, kit, or method of claim 246, wherein the anti-human PD-1 monoclonal antibody is pembrolizumab. 254. The composition, kit, or method of claim 246, wherein the anti-human PD-1 monoclonal antibody is nivolumab. 255. The composition, kit, or method of claim 246, wherein the anti-human PD-1 monoclonal antibody is cemiplimab. 256. The composition, kit, or method of claim 250, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising an amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising an amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising an amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising an amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising an amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising an amino acid sequence as set forth in SEQ ID NO: 110. 257. The composition, kit, or method of claim 250, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 152. 258. The composition, kit, or method of claim 250, wherein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295.

Description:
A THERAPEUTIC COMBINATION COMPRISING A T1GIT ANTAGONIST, A PD-1 ANTAGONIST, AND A CHEMOTHERAPEUTIC AGENT(S) 1

FIELD

Provided herein are methods for treating cancer, an infectious disease, or an infection using a combination of (a) a T cell immunoreceptor with Ig and ITIM domains (TIGIT) antagonist, (b) a programmed death I protein (PD-1) antagonist, and (c) one or more chemotherapeutic agents.

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Patent Application No, 63/231,525, filed August 10, 2021 and U.S. Provisional Patent Application No. 63/327,070, filed April 4, 2022, each of which is incorporated by reference herein in its entirety.

REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY

The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety’. The XML file, created on August 1, 2022, is named 25305-WO-PCT_SL.XML and is 352 KB in size.

BACKGROUND OF THE INVENTION

TIGIT is an immunomodulatory receptor expressed primarily on activated T cells and NK cells. TIGIT is also blown as VSIG9, VSTM3, and WUCAM. Its structure shows one extracellular immunoglobulin domain, a type 1 transmembrane region and two ITIM motifs. TIGIT forms part of a co-stimulatoty network that consists of positive (CD226) and negative (TIGIT) immunomodulatory' receptors on T cells, and ligands expressed on APCs (CD155 and CD 112).

An important feature in the structure of TIGIT is the presence of an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic tail domain. As with PD-1, the ITIM domain in the cytoplasmic region of TIGIT is predicted to recruit tyrosine phosphatases, such as SHP-1 and SHP-2, and subsequent de-phosphorylation of tyrosine residues within the immunoreceptor tyrosine-base activation motifs (IT AM) on T cell receptor (TCR) subunits. Hence, ligation of TIGIT by receptor-ligands CD155 and CD112 expressed by tumor cells or TAMS may contribute to the suppression of TCR-signaling and T cell activation, which is essential for mounting effective anti-tumor immunity. Thus, an antagonist antibody specific for TIGIT could inhibit the CD155 and CD112 induced suppression of T cell responses and enhance anti-tumor immunity. PD-1 is recognized as an important player in immune regulation and the maintenance of peripheral tolerance. Immune checkpoint therapies targeting PD-1 or its ligand (e.g., PD- L1) have resulted in groundbreaking improvements in clinical response in multiple human cancer types (Brahmer et al., N Engl J Med, 366: 2455-2465 (2012); Garon et al., N Engl J Med, 372:2018-2028 (2015); Hamid et al., N Engl J Med, 369:134-144 (2013); Robert et al., Lancet, 384:1109-1117 (2014); Robert et al., N Engl J Med, 372: 2521-2532 (2015); Robert et al., N Engl J Med, 372:320-330 (2015); Topalian et al., N Engl J Med, 366:2443-2454 (2012); Topalian et al., J Clin Oncol, 32:1020-1030 (2014); Wolchok et al., N Engl J Med, 369:122-133 (2013)). Immune therapies targeting the PD-1 axis include monoclonal antibodies directed to the PD-1 receptor (e.g., KEYTRUDA ® (pembrolizumab), Merck and Co., Inc., Kenilworth, NJ; OPDIVO ® (nivolumab), Bristol-Myers Squibb Company, Princeton, NJ) and those that bind to the PD-L1 ligand (e.g., TECENTRIQ ® (atezolizumab), Genentech, San Francisco, CA). Chemotherapeutic agents typically provide non-specific usage of intracellular poisons to inhibit mitosis or induce DNA damage. For example, chemotherapeutic agents are cytotoxic by means of interfering with cell division (mitosis) but cancer cells vary widely in their susceptibility to these agents. Chemotherapy can be thought of as a way to damage or stress cells, which may then lead to cell death if apoptosis is initiated. SUMMARY The present disclosure provides methods, pharmaceutical compositions, uses and kits of treating a cancer, an infectious disease, or an infection using a combination of therapeutic agents, e.g., a combination of antibodies or antigen binding fragments thereof. The present disclosure provides methods of treating a cancer, an infectious disease, or an infection using a combination of a TIGIT antagonist (e.g., antibody (e.g., monoclonal antibody) or antigen binding fragment thereof), a PD-1 antagonist (e.g., antibody (e.g., monoclonal antibody) or antigen binding fragment thereof), and one or more chemotherapeutic agents (e.g., alkylating agents, antimetabolites, anti-microtubule agents, etc.). Among other things, the present disclosure encompasses insights that certain combinations of immune checkpoint inhibitors (e.g., a TIGIT antagonist and a PD-1 antagonist) in combination with one or more chemotherapeutic agents (e.g., alkylating agents (e.g., cisplatin), antimetabolites (e.g., 5-fluorouracil, gemcitabine, capecitabine), anti- microtubule agents (e.g., paclitaxel), etc.) as provided herein may enhance the efficacy without significant added toxicity as compared with existing treatments. While it has been proposed that the efficacy of anti-TIGIT antagonistic antibodies and anti-PD-1 antagonistic antibodies might be enhanced if administered in combination with other approved or experimental cancer therapies, there are no clear guidelines as to which agent combined with the anti-TIGIT antagonistic antibodies and anti-PD-1 antagonistic antibodies may be effective or in which patients the combination may enhance the efficacy of treatment. The present disclosure provides methods of treating cancer (e.g., esophageal cancer, triple negative breast cancer (TNBC), biliary cancer, gastric cancer, etc.) using a combination of a TIGIT antagonist, a PD-1 antagonist, 5-fluorouracil represented by Formula (I), and cisplatin represented by Formula (II). I) I) osure provides methods of treating cancer (e.g., esophageal cancer, TNBC, biliary cancer, gastric cancer etc.) using a combination of a TIGIT antagonist, a PD-1 antagonist, and paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. I) of treating cancer (e.g., esophageal cancer, TNBC, biliary cancer, etc.) using a combination of a TIGIT antagonist, a PD-1 antagonist, gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof, and cisplatin represented by Formula (II). V) rovides methods of treating cancer (e.g., esophageal cancer, TNBC, biliary cancer, etc.) using a combination of a TIGIT antagonist, a PD-1 antagonist, capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof, and oxaliplatin represented by Formula (VI). V) ides kits including a TIGIT antagonist, a PD-1 antagonist, 5-fluorouracil and cisplatin. The present disclosure further provides kits including a TIGIT antagonist, a PD-1 antagonist, and paclitaxel or a pharmaceutically acceptable salt thereof. The present disclosure further provides kits including a TIGIT antagonist, a PD-1 antagonist, gemcitabine or pharmaceutically acceptable salt thereof and cisplatin. The present disclosure further provides kits including a TIGIT antagonist, a PD-1 antagonist, capecitabine or pharmaceutically acceptable salt thereof and oxaliplatin. Also provided herein are uses of a therapeutic combination for treating cancer (e.g., esophageal cancer), wherein the therapeutic combination includes a TIGIT antagonist, a PD-1 antagonist, 5-fluorouracil and cisplatin. Also provided herein are uses of a therapeutic combination for treating cancer (e.g., TNBC), wherein the therapeutic combination includes a TIGIT antagonist, a PD-1 antagonist, and paclitaxel or a pharmaceutically acceptable salt thereof. Also provided herein are uses of a therapeutic combination for treating cancer (e.g., biliary cancer), wherein the therapeutic combination includes a TIGIT antagonist, a PD-1 antagonist, gemcitabine or pharmaceutically acceptable salt thereof and cisplatin. Also provided herein are uses of a therapeutic combination for treating cancer (e.g., gastric cancer), wherein the therapeutic combination includes a TIGIT antagonist, a PD-1 antagonist, capecitabine or pharmaceutically acceptable salt thereof and oxaliplatin. In one aspect, provided herein is a method of treating cancer, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil; and (d) cisplatin. In one aspect, provided herein are therapeutic combinations for use in treating cancer, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one aspect, provided herein is a method of treating cancer, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine or pharmaceutically acceptable salt thereof; and (d) cisplatin. In one aspect, provided herein is a method of treating cancer, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine or pharmaceutically acceptable salt thereof; and (d) oxaliplatin. In some embodiments, the cancer is selected from the group consisting of osteosarcoma, rhabdomyosarcoma, neuroblastoma, kidney cancer, leukemia, renal transitional cell cancer, bladder cancer, Wilm’s cancer, ovarian cancer, pancreatic cancer, breast cancer, prostate cancer, bone cancer, lung cancer (e.g., non-small cell lung cancer), gastric cancer, colorectal cancer, cervical cancer, synovial sarcoma, head and neck cancer, squamous cell carcinoma, lymphoma (e.g., diffuse large B-cell lymphoma (DLBCL) or non- Hodgkin lymphoma (NHL)), multiple myeloma, renal cell cancer, retinoblastoma, hepatoblastoma, hepatocellular carcinoma, melanoma, rhabdoid tumor of the kidney, Ewing's sarcoma, chondrosarcoma, brain cancer, glioblastoma, meningioma, pituitary adenoma, vestibular schwannoma, primitive neuroectodermal tumor, medulloblastoma, astrocytoma, anaplastic astrocytoma, oligodendroglioma, ependymoma, choroid plexus papilloma, polycythemia vera, thrombocythemia, idiopathic myelfibrosis, soft tissue sarcoma, thyroid cancer, endometrial cancer, and carcinoid cancer. In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC) and gastric cancer. In some embodiments, the cancer is esophageal cancer, TNBC, biliary cancer or gastric cancer. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In certain embodiments, the cancer is resectable. In another aspect, provided herein is a kit comprising: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil; and (d) cisplatin. In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, 5-fluorouracil, and cisplatin. In another aspect, provided herein is a kit comprising: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, and paclitaxel or a pharmaceutically acceptable salt thereof. In another aspect, provided herein is a kit comprising: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine or pharmaceutically acceptable salt thereof; and (d) cisplatin. In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, gemcitabine or a pharmaceutically acceptable salt thereof and cisplatin. In another aspect, provided herein is a kit comprising: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine or pharmaceutically acceptable salt thereof; and (d) oxaliplatin. In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, capecitabine or a pharmaceutically acceptable salt thereof and oxaliplatin. In still another aspect, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil; and (d) cisplatin. In still another aspect, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In still another aspect, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine or pharmaceutically acceptable salt thereof; and (d) cisplatin. In still another aspect, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine or pharmaceutically acceptable salt thereof; and (d) oxaliplatin. In certain embodiments of the methods, pharmaceutical compositions, kits, uses, or the combinations for use provided herein, the subject is a human patient. In certain embodiments the methods, pharmaceutical compositions, kits, uses, or the combinations for use provided herein are for treating cancer. In certain embodiments of the methods, pharmaceutical compositions, kits, uses or the combinations for use provided herein, the cancer is esophageal cancer, TNBC, biliary cancer, or gastric cancer. In certain embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In other embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. In some embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody is a humanized antibody. In other embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody is a human antibody. In some embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-L1 monoclonal antibody is a humanized antibody. In other embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-L1 monoclonal antibody is a human antibody. In certain embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human TIGIT monoclonal antibody is a humanized antibody. In other embodiments of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human TIGIT monoclonal antibody is a human antibody. In still other embodiments of the methods, pharmaceutical compositions, kits, uses provided herein, the anti-PD-1 antibody is independently selected from pembrolizumab, nivolumab, cemiplimab, sintilimab, tislelizumab, camrelizumab and toripalimab. In one embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody is pembrolizumab. In another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody is nivolumab. In another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody is cemiplimab. In yet another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is pidilizumab (U.S. Pat. No.7,332,582). In yet another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is AMP-514 (MedImmune LLC, Gaithersburg, MD). In yet another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is PDR001 (U.S. Pat. No.9,683,048). In yet another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is BGB-A317 (U.S. Pat. No.8,735,553). In yet another embodiment of the methods, pharmaceutical compositions, kits, or uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is MGA012 (MacroGenics, Rockville, MD). In certain embodiments of the methods, kits, or uses provided herein, the anti-human TIGIT monoclonal antibody comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of the methods, kits, or uses provided herein the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In other embodiments of the methods, kits, or uses provided herein, the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295. In one specific embodiment of the methods, kits, or uses provided herein, the PD-1 antagonist is pembrolizumab; and the TIGIT antagonist is a monoclonal antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In one specific embodiment of the methods, kits, or uses provided herein, the PD-1 antagonist is nivolumab; and the TIGIT antagonist is a monoclonal antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In one specific embodiment of the methods, kits, or uses provided herein, the PD-1 antagonist is cemiplimab; and the TIGIT antagonist is a monoclonal antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In another aspect, provided herein is a method of enhancing T cell activity, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (c) 5-fluorouracil; and (d) cisplatin. In another aspect, provided herein is a method of enhancing T cell activity, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In another aspect, provided herein is a method of enhancing T cell activity, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (c) gemcitabine or a pharmaceutically acceptable salt thereof; and (d) cisplatin. In another aspect, provided herein is a method of enhancing T cell activity, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (c) capecitabine or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin. In some embodiments, the enhancement of T cell activity occurs in vitro. In other embodiments, the enhancement of T cell activity occurs in vivo. For example, the enhancement is in a subject including but not limited to a human subject or human patient. In certain embodiments, the enhancement of T cell activity is measured by increased cytokine production. In other embodiments, the enhancement of T cell activity is measured by increased cell proliferation. In some embodiments, provided herein is a method of increasing cytokine production of T cells, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (c) 5-fluorouracil; and (d) cisplatin. In some embodiments, provided herein is a method of increasing cytokine production of T cells, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is a method of increasing cytokine production of T cells, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (c) gemcitabine or a pharmaceutically acceptable salt thereof; and (d) cisplatin. In some embodiments, provided herein is a method of increasing cytokine production of T cells, comprising contacting the T cells with: (a) an anti-human TIGIT antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (b) an anti-human PD-1 antibody (e.g., monoclonal antibody) or antigen binding fragment thereof; (c) capecitabine or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin. In some embodiments, the increased cytokine production of T cells occurs in vitro. In other embodiments, the increased cytokine production of T cells occurs in vivo. In some embodiments of the methods, kits, or uses described herein, the human patient is administered about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab, and pembrolizumab is administered once every three weeks. In one embodiment, the human patient is administered about 200 mg pembrolizumab once every three weeks. In one embodiment, the human patient is administered about 240 mg pembrolizumab once every three weeks. In one embodiment, the human patient is administered about 2 mg/kg pembrolizumab once every three weeks. In certain embodiments of the methods, kits, or uses described herein, the human patient is administered about 400 mg pembrolizumab, and pembrolizumab is administered once every six weeks. In other embodiments of the methods, kits, or uses described herein, the human patient is administered about 240 mg or about 3 mg/kg nivolumab once every two weeks, or about 480 mg nivolumab once every four weeks. In one specific embodiment, the human patient is administered about 240 mg nivolumab once every two weeks. In one specific embodiment, the human patient is administered about 3 mg/kg nivolumab once every two weeks. In one specific embodiment, the human patient is administered about 480 mg nivolumab once every four weeks. In yet other embodiments of the methods, kits, or uses described herein, the human patient is administered about 350 mg cemiplimab, and cemiplimab is administered once every three weeks. In some embodiments of the methods, kits, or uses described herein, the human patient is administered about 200 mg, about 240 mg, or about 2 mg/kg of the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295, and the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof is administered once every three weeks. In one embodiment, the human patient is administered about 200 mg of the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof once every three weeks. In one embodiment, the human patient is administered about 240 mg of the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof once every three weeks. In one embodiment, the human patient is administered about 2 mg/kg of the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof once every three weeks. In certain embodiments of the methods, kits, or uses described herein, the human patient is administered about 400 mg of the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295, and e anti-human TIGIT monoclonal antibody or antigen binding fragment thereof is administered once every six weeks. In yet still other embodiments of the methods, kits, or uses described herein, the human patient is administered about 3000 mg/m 2 to about 4000 mg/m 2 5-fluorouracil, wherein the 5-fluorouracil is administered five times every three weeks. In some embodiments, the human patient is administered about 600 or about 800 mg/m 2 5- fluorouracil a day. In some embodiments, the human patient is administered about 40 mg/m 2 to about 100 mg/m 2 cisplatin, wherein the cisplatin is administered once every three weeks. In some embodiments, the human patient is administered about 60 or about 80 mg/m 2 cisplatin, wherein the cisplatin is administered once every three weeks. In some embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three or six weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 240 mg pembrolizumab once every three weeks; (b) about 240 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 2 mg/kg pembrolizumab once every three weeks; (b) about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 400 mg pembrolizumab once every six weeks; (b) about 400 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every six weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In a specific embodiment, provided herein is a method of treating esophageal cancer, comprising administering to a human patient in need thereof: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil per cycle (e.g., about 600 mg/m 2 or 800 mg/m 2 per day five times every three weeks); and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin once every three weeks. In some embodiments, the human patient is administered about 70 mg/m 2 to about 100 mg/m 2 paclitaxel, wherein the paclitaxel is administered once every week. In some embodiments, the human patient is administered about 80 mg/m 2 or about 90 mg/m 2 paclitaxel, wherein the paclitaxel is administered once every week. In some embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three or six weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In certain embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In certain embodiments, the human patient is administered: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In certain embodiments, the human patient is administered: (a) about 240 mg pembrolizumab once every three weeks; (b) about 240 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In certain embodiments, the human patient is administered: (a) about 2 mg/kg pembrolizumab once every three weeks; (b) about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In certain embodiments, the human patient is administered: (a) about 400 mg pembrolizumab once every six weeks; (b) about 400 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every six weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In a specific embodiment, provided herein is a method of treating TNBC, comprising administering to a human patient in need thereof: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel once every week. In yet still other embodiments of the methods, kits, or uses described herein, the human patient is administered about 500 mg/m 2 to about 1500 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof, wherein the gemcitabine or a pharmaceutically acceptable salt thereof is administered every three weeks. In some embodiments, the human patient is administered about 800 mg/m 2 to about 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof, wherein the gemcitabine or a pharmaceutically acceptable salt thereof is administered every three weeks. In some embodiments, the human patient is administered about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof, wherein the gemcitabine or a pharmaceutically acceptable salt thereof is administered every three weeks. In some embodiments, the human patient is administered about 10 mg/m 2 to about 50 mg/m 2 cisplatin, wherein the cisplatin is administered once every three weeks. In some embodiments, the human patient is administered about 20 mg/m 2 to about 25 mg/m 2 cisplatin, wherein the cisplatin is administered once every three weeks. In some embodiments, the human patient is administered about 20 or about 25 mg/m 2 cisplatin, wherein the cisplatin is administered once every three weeks. In some embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three or six weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 240 mg pembrolizumab once every three weeks; (b) about 240 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 2 mg/kg pembrolizumab once every three weeks; (b) about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 400 mg pembrolizumab once every six weeks; (b) about 400 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every six weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In a specific embodiment, provided herein is a method of treating biliary cancer, comprising administering to a human patient in need thereof: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three weeks; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin once every three weeks. In some embodiments of the methods, kits, or uses described herein, the human patient is administered about 500 mg/m 2 to about 1500 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof, wherein the capecitabine or a pharmaceutically acceptable salt thereof is administered twice a day. In some embodiments, the human patient is administered about 750 mg/m 2 to about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof, wherein the capecitabine or a pharmaceutically acceptable salt thereof is administered twice a day. In some embodiments, the human patient is administered about 750 mg/m 2 or about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof, wherein the capecitabine or a pharmaceutically acceptable salt thereof is administered twice a day. In some embodiments, the human patient is administered about 80 mg/m 2 to about 150 mg/m 2 oxaliplatin, wherein the oxaliplatin is administered once every three weeks. In some embodiments, the human patient is administered about 100 mg/m 2 to about 130 mg/m 2 oxaliplatin, wherein the oxaliplatin is administered once every three weeks. In some embodiments, the human patient is administered about 100 or about 130 mg/m 2 oxaliplatin, wherein the oxaliplatin is administered once every three weeks. In some embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three or six weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 200 mg, about 240 mg, or about 2 mg/kg pembrolizumab once every three weeks; (b) about 200 mg, about 240 mg, or about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 240 mg pembrolizumab once every three weeks; (b) about 240 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 2 mg/kg pembrolizumab once every three weeks; (b) about 2 mg/kg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In certain embodiments, the human patient is administered: (a) about 400 mg pembrolizumab once every six weeks; (b) about 400 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every six weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In a specific embodiment, provided herein is a method of treating gastric cancer, comprising administering to a human patient in need thereof: (a) about 200 mg pembrolizumab once every three weeks; (b) about 200 mg of an anti-human TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 once every three weeks; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin once every three weeks. In certain embodiments of the methods, kits and uses provided herein, the anti-human TIGIT monoclonal antibody and the anti-human PD-1 monoclonal antibody are administered on the same day. In some embodiments, the anti-human TIGIT monoclonal antibody and the anti-human PD-1 monoclonal antibody are administered sequentially. In other embodiments, the anti-human TIGIT monoclonal antibody and the anti-human PD-1 monoclonal antibody are administered concurrently. In some embodiments, the anti-human TIGIT monoclonal antibody and the anti-human PD-1 monoclonal antibody are co-formulated. BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows tumor volume size as a function of time for one or more cancer treatments, including the combination treatment of a PD-1 antagonist, a TIGIT antagonist, 5- Fluorouracil (5-FU) and cisplatin. Figure 2 shows changes in tumor volume at Day 17 for one or more cancer treatments, including the combination treatment of a PD-1 antagonist, a TIGIT antagonist, 5- FU and cisplatin. Figure 3 depicts tumor volume size as a function of time for one or more cancer treatments, including the combination treatment of a PD-1 antagonist, a TIGIT antagonist, gemcitabine and cisplatin. Figure 4 depicts best overall response shown as percentage change in tumor volume at the end of study for one or more cancer treatments, including the combination treatment of a PD-1 antagonist, a TIGIT antagonist, gemcitabine and cisplatin. DETAILED DESCRIPTION OF THE INVENTION Definitions Certain technical and scientific terms are specifically defined below. Unless specifically defined elsewhere in this document, all other technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which this disclosure relates. “About” when used to modify a numerically defined parameter (e.g., the dose of an anti-TIGIT antibody or antigen binding fragment thereof, an anti-PD-1 antibody or antigen binding fragment thereof, 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin twice a day or the length of treatment time with a combination therapy described herein) means that the parameter is within 20%, within 15%, within 10%, within 9%, within 8%, within 7%, within 6%, within 5%, within 4%, within 3%, within 2%, within 1%, or less of the stated numerical value or range for that parameter; where appropriate, the stated parameter may be rounded to the nearest whole number. For example, a dose of about 5 mg/kg may vary between 4.5 mg/kg and 5.5 mg/kg. As used herein, including the appended claims, the singular forms of words such as “a,” “an,” and “the,” include their corresponding plural references unless the context clearly dictates otherwise. The terms “administration” or “administer” refers to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g., an anti-TIGIT antibody, an anti-PD-1 antibody, 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin as described herein) into a patient, such as by oral, mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery, and/or any other methods of physical delivery described herein or known in the art. As used herein, the term “antibody” refers to any form of immunoglobulin molecule that exhibits the desired biological or binding activity. Thus, it is used in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized, fully human antibodies, and chimeric antibodies. “Parental antibodies” are antibodies obtained by exposure of an immune system to an antigen prior to modification of the antibodies for an intended use, such as humanization of an antibody for use as a human therapeutic. As used herein, the term “antibody” encompasses not only intact polyclonal or monoclonal antibodies, but also, unless otherwise specified, fusion proteins comprising an antigen binding portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen binding fragment thereof that competes with the intact antibody for specific binding. In general, the basic antibody structural unit comprises a tetramer. Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The variable regions of each light/heavy chain pair form the antibody binding site. Thus, in general, an intact antibody has two binding sites. The carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function. Typically, human light chains are classified as kappa and lambda light chains. Furthermore, human heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody’s isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids. See generally, Fundamental Immunology Ch.7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989). “Variable regions” or “V region” or “V chain” as used herein means the segment of IgG chains which is variable in sequence between different antibodies. A “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. The variable region of the heavy chain may be referred to as “VH.” The variable region of the light chain may be referred to as “V L .” Typically, the variable regions of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope. In general, from N-terminal to C-terminal, both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. As referred to herein the light chain CDRs are CDRL1, CDRL2 and CDRL3, respectively, and the heavy chain CDRs are CDRH1, CDRH2 and CDRH3, respectively. The assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al.; National Institutes of Health, Bethesda, Md.; 5th ed.; NIH Publ. No.91-3242 (1991); Kabat (1978) Adv. Prot. Chem.32:1-75; Kabat, et al., (1977) J. Biol. Chem.252:6609-6616; Chothia, et al., (1987) J Mol. Biol.196:901-917 or Chothia, et al., (1989) Nature 342:878- 883. A “CDR” refers to one of three hypervariable regions (H1, H2, or H3) within the non- framework region of the antibody VH β-sheet framework, or one of three hypervariable regions (L1, L2, or L3) within the non-framework region of the antibody V L β-sheet framework. Accordingly, CDRs are variable region sequences interspersed within the framework region sequences. CDR regions are well known to those skilled in the art and have been defined by, for example, Kabat as the regions of most hypervariability within the antibody variable domains. CDR region sequences also have been defined structurally by Chothia as those residues that are not part of the conserved b-sheet framework, and thus are able to adapt to different conformation. Both terminologies are well recognized in the art. CDR region sequences have also been defined by AbM, Contact, and IMGT. The positions of CDRs within a canonical antibody variable region have been determined by comparison of numerous structures (Al-Lazikani et al., 1997, J. Mol. Biol.273:927-48; Morea et al., 2000, Methods 20:267-79). Because the number of residues within a hypervariable region varies in different antibodies, additional residues relative to the canonical positions are conventionally numbered with a, b, c and so forth next to the residue number in the canonical variable region numbering scheme (Al-Lazikani et al., supra). Such nomenclature is similarly well known to those skilled in the art. Correspondence between the numbering system, including, for example, the Kabat numbering and the IMGT unique numbering system, is well known to one skilled in the art and shown below in Table 1. In some embodiments, the CDRs are as defined by the Kabat numbering system. In other embodiments, the CDRs are as defined by the IMGT numbering system. In yet other embodiments, the CDRs are as defined by the AbM numbering system. In still other embodiments, the CDRs are as defined by the Chothia numbering system. In yet other embodiments, the CDRs are as defined by the Contact numbering system. Table 1. Correspondence between the CDR Numbering Systems Kabat + IMGT Kabat AbM Chothia Contact Chothia V H CDR2 50-65 56-65 50-65 50-58 52-56 47-58 V H CDR3 95-102 105-117 95-102 95-102 95-102 93-101 V L CDR1 24-34 27-38 24-34 24-34 24-34 30-36 V L CDR2 50-56 56-65 50-56 50-56 50-56 46-55 V L CDR3 89-97 105-117 89-97 89-97 89-97 89-96 “Chimeric antibody” refers to an antibody in which a portion of the heavy and/or light chain contains sequences derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity. “Human antibody” refers to an antibody that comprises human immunoglobulin protein sequences or derivatives thereof. A human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell. Similarly, “mouse antibody” or “rat antibody” refer to an antibody that comprises only mouse or rat immunoglobulin sequences or derivatives thereof, respectively. “Humanized antibody” refers to forms of antibodies that contain sequences from non- human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non- human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. The prefix “hum”, “hu” or “h” may be added to antibody clone designations when necessary to distinguish humanized antibodies from parental rodent antibodies. The humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parental rodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability of the humanized antibody, or for other reasons. “Monoclonal antibody” or “mAb” or “Mab”, as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present disclosure may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No.4,816,567). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol.222: 581- 597, for example. See also Presta (2005) J. Allergy Clin. Immunol.116:731. As used herein, unless otherwise indicated, “antibody fragment” or “antigen binding fragment” refers to a fragment of an antibody that retains the ability to bind specifically to the antigen, e.g., fragments that retain one or more CDR regions and the ability to bind specifically to the antigen. An antibody that “specifically binds to” TIGIT or PD-1 is an antibody that exhibits preferential binding to TIGIT or PD-1 (as appropriate) as compared to other proteins, but this specificity does not require absolute binding specificity. An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g., without producing undesired results such as false positives. Antibodies, or binding fragments thereof, will bind to the target protein with an affinity that is at least two-fold greater, preferably at least ten times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins. Antigen binding portions include, for example, Fab, Fab’, F(ab’)2, Fd, Fv, fragments including CDRs, and single chain variable fragment antibodies (scFv), and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the antigen (e.g., TIGIT or PD-1). An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant region of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. The heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three- dimensional configurations of different classes of immunoglobulins are well known. As used herein, the terms “at least one” item or “one or more” item each include a single item selected from the list as well as mixtures of two or more items selected from the list. As used herein, the term “immune response” relates to any one or more of the following: specific immune response, non-specific immune response, both specific and non- specific response, innate response, primary immune response, adaptive immunity, secondary immune response, memory immune response, immune cell activation, immune cell- proliferation, immune cell differentiation, and cytokine expression. The term “subject” (alternatively “patient”) as used herein refers to a mammal that has been the object of treatment, observation, or experiment. The mammal may be male or female. The mammal may be one or more selected from the group consisting of humans, bovine (e.g., cows), porcine (e.g., pigs), ovine (e.g., sheep), capra (e.g., goats), equine (e.g., horses), canine (e.g., domestic dogs), feline (e.g., house cats), lagomorph (e.g., rabbits), rodent (e.g., rats or mice), Procyon lotor (e.g., raccoons). In particular embodiments, the subject is human. The term “subject in need thereof” as used herein refers to a subject diagnosed with or suspected of having cancer or an infectious disease as defined herein. The therapeutic agents and compositions provided by the present disclosure can be administered via any suitable enteral route or parenteral route of administration. The term “enteral route” of administration refers to the administration via any part of the gastrointestinal tract. Examples of enteral routes include oral, mucosal, buccal, and rectal route, or intragastric route. “Parenteral route” of administration refers to a route of administration other than enteral route. Examples of parenteral routes of administration include intravenous, intramuscular, intradermal, intraperitoneal, intratumor, intravesical, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, transtracheal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal, subcutaneous, or topical administration. The therapeutic agents and compositions of the disclosure can be administered using any suitable method, such as by oral ingestion, nasogastric tube, gastrostomy tube, injection, infusion, implantable infusion pump, and osmotic pump. The suitable route and method of administration may vary depending on a number of factors such as the specific therapeutic agent being used, the rate of absorption desired, specific formulation or dosage form used, type or severity of the disorder being treated, the specific site of action, and conditions of the patient, and can be readily selected by a person skilled in the art. The term “variant” when used in relation to an antibody (e.g., an anti-TIGIT antibody or an anti-PD-1 antibody) or an amino acid region within the antibody may refer to a peptide or polypeptide comprising one or more (such as, for example, about 1 to about 25, about 1 to about 20, about 1 to about 15, about 1 to about 10, or about 1 to about 5) amino acid sequence substitutions, deletions, and/or additions as compared to a native or unmodified sequence. For example, a variant of an anti-PD-1 antibody may result from one or more (such as, for example, about 1 to about 25, about 1 to about 20, about 1 to about 15, about 1 to about 10, or about 1 to about 5) changes to an amino acid sequence of a native or previously unmodified anti-PD-1 antibody. Variants may be naturally occurring or may be artificially constructed. Polypeptide variants may be prepared from the corresponding nucleic acid molecules encoding the variants. In specific embodiments, an antibody variant (e.g., an anti- TIGIT antibody variant or an anti-PD-1 antibody variant) at least retains the antibody functional activity. In specific embodiments, an anti-TIGIT antibody variant binds to TIGIT and/or is antagonistic to TIGIT activity. In some embodiments, an anti-PD-1 antibody variant binds to PD-1 and/or is antagonistic to PD-1 activity. “Conservatively modified variants” or “conservative substitution” refers to substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g., charge, side-chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity or other desired property of the protein, such as antigen affinity and/or specificity. Those of skill in this art recognize that, in general, single amino acid substitutions in non- essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity. Exemplary conservative substitutions are set forth in Table 2 below. Table 2. Exemplary Conservative Amino Acid Substitutions Original residue Conservative substitution Al (A) Gl S r Original residue Conservative substitution “Homolog y re ers o sequence s m ar y e ween wo po ypep de sequences when they are optimally aligned. When a position in both of the two compared sequences is occupied by the same amino acid monomer subunit, e.g., if a position in a light chain CDR of two different Abs is occupied by alanine, then the two Abs are homologous at that position. The percent of homology is the number of homologous positions shared by the two sequences divided by the total number of positions compared × 100. For example, if 8 of 10 of the positions in two sequences are matched when the sequences are optimally aligned then the two sequences are 80% homologous. Generally, the comparison is made when two sequences are aligned to give maximum percent homology. For example, the comparison can be performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences. The following references relate to BLAST algorithms often used for sequence analysis: BLAST ALGORITHMS: Altschul, S.F., et al., (1990) J. Mol. Biol.215:403-410; Gish, W., et al., (1993) Nature Genet.3:266-272; Madden, T.L., et al., (1996) Meth. Enzymol.266:131-141; Altschul, S.F., et al., (1997) Nucleic Acids Res.25:3389-3402; Zhang, J., et al., (1997) Genome Res.7:649-656; Wootton, J.C., et al., (1993) Comput. Chem.17:149-163; Hancock, J.M. et al., (1994) Comput. Appl. Biosci.10:67-70; ALIGNMENT SCORING SYSTEMS: Dayhoff, M.O., et al., “A model of evolutionary change in proteins.” in Atlas of Protein Sequence and Structure, (1978) vol.5, suppl.3. M.O. Dayhoff (ed.), pp.345-352, Natl. Biomed. Res. Found., Washington, DC; Schwartz, R.M., et al., “Matrices for detecting distant relationships.” in Atlas of Protein Sequence and Structure, (1978) vol.5, suppl.3.” M.O. Dayhoff (ed.), pp.353-358, Natl. Biomed. Res. Found., Washington, DC; Altschul, S.F., (1991) J. Mol. Biol.219:555-565; States, D.J., et al., (1991) Methods 3:66-70; Henikoff, S., et al., (1992) Proc. Natl. Acad. Sci. USA 89:10915-10919; Altschul, S.F., et al., (1993) J. Mol. Evol.36:290-300; ALIGNMENT STATISTICS: Karlin, S., et al., (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268; Karlin, S., et al., (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877; Dembo, A., et al., (1994) Ann. Prob.22:2022-2039; and Altschul, S.F. “Evaluating the statistical significance of multiple distinct local alignments.” in Theoretical and Computational Methods in Genome Research (S. Suhai, ed.), (1997) pp.1- 14, Plenum, New York. “RECIST 1.1 Response Criteria” as used herein means the definitions set forth in Eisenhauer, E.A. et al., Eur. J. Cancer 45:228-247 (2009) for target lesions or nontarget lesions, as appropriate based on the context in which response is being measured. “Sustained response” means a sustained therapeutic effect after cessation of treatment as described herein. In some embodiments, the sustained response has a duration that is at least the same as the treatment duration, or at least 1.5, 2.0, 2.5 or 3 times longer than the treatment duration. As used herein, the term "treat" or "treating" means to administer a therapeutic combination of a PD-1 antagonist, a TIGIT antagonist, and one or more chemotherapeutic agents (e.g., an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin) or pharmaceutically acceptable salts thereof) to a subject or patient having one or more disease symptoms as provided herein. Typically, the agents of the therapeutic combination are administered in an amount effective to alleviate one or more disease symptoms in the treated subject or population, whether by inducing the regression of or inhibiting the progression of such symptom(s) by any clinically measurable degree. The amount of the agents of the therapeutic combination that is effective to alleviate any particular disease symptom may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapeutic combination to elicit a desired response in the subject. Whether a disease symptom has been alleviated can be assessed by any clinical measurement typically used by physicians or other skilled healthcare providers to assess the severity or progression status of that symptom. “Treat” or “treating” cancer as used herein means to administer a therapeutic combination of a PD-1 antagonist, a TIGIT antagonist, and one or more chemotherapeutic agents or pharmaceutically acceptable salts thereof (e.g., an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, and one or more chemotherapeutic agents (e.g., 5- fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin)) to a subject having cancer or diagnosed with cancer to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastasis or tumor growth, comprising administration by oral, mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery, and/or any other methods of physical delivery described herein or known in the art. In specific embodiments, the therapeutic combination is a therapeutic combination of an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, 5-fluorouracil, and cisplatin; a therapeutic combination of an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, and paclitaxel or a pharmaceutically acceptable salt thereof; a therapeutic combination of an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, gemcitabine or a pharmaceutically acceptable salt thereof, and cisplatin; or a therapeutic combination of an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, capecitabine or a pharmaceutically acceptable salt thereof, and oxaliplatin. “Treatment” may include one or more of the following: inducing/increasing an antitumor immune response, decreasing the number of one or more tumor markers, halting or delaying the growth of a tumor or blood cancer or progression of disease such as cancer, stabilization of disease, inhibiting the growth or survival of tumor cells, eliminating or reducing the size of one or more cancerous lesions or tumors, decreasing the level of one or more tumor markers, ameliorating or abrogating the clinical manifestations of disease, reducing the severity or duration of the clinical symptoms, prolonging the survival or patient relative to the expected survival in a similar untreated patient, and inducing complete or partial remission of a cancerous condition, wherein the disease is cancer, and in certain embodiments wherein the cancer is selected from the group consisting of endometrial cancer cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, bladder cancer, breast cancer, triple negative breast cancer (TNBC), non-small cell lung cancer (NSCLC), colorectal cancer (CRC), renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), and melanoma. The amount of a therapeutic agent that is effective to alleviate any particular disease symptom may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the drug to elicit a desired response in the subject. Whether a disease symptom has been alleviated can be assessed by any clinical measurement typically used by physicians or other skilled healthcare providers to assess the severity or progression status of that symptom. Positive therapeutic effects in cancer can be measured in a number of ways (See, W. A. Weber, J. Nucl. Med.50:1S-10S (2009)). For example, with respect to tumor growth inhibition, according to NCI standards, a T/C أ 42% is the minimum level of anti-tumor activity. A T/C < 10% is considered a high anti-tumor activity level, with T/C (%) = Median tumor volume of the treated/Median tumor volume of the control × 100. In some embodiments, the treatment achieved by a combination therapy of the disclosure is any of PR, CR, OR, PFS, DFS, and OS. PFS, also referred to as “Time to Tumor Progression” indicates the length of time during and after treatment that the cancer does not grow, and includes the amount of time patients have experienced a CR or PR, as well as the amount of time patients have experienced SD. DFS refers to the length of time during and after treatment that the patient remains free of disease. OS refers to a prolongation in life expectancy as compared to naive or untreated individuals or patients. In some embodiments, response to a combination therapy of the disclosure is any of PR, CR, PFS, DFS, or OR that is assessed using RECIST 1.1 response criteria. The treatment regimen for a combination therapy of the disclosure that is effective to treat a cancer patient may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the subject. While an embodiment of any of the aspects of the disclosure may not be effective in achieving a positive therapeutic effect in every subject, it should do so in a statistically significant number of subjects as determined by any statistical test known in the art such as the Student’s t-test, the chi 2 -test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test. “Treat” or “treating” an infectious disease or an infection as used herein means to administer a therapeutic combination of a PD-1 antagonist, a TIGIT antagonist, and one or more chemotherapeutic agents or pharmaceutically acceptable salts thereof (e.g., an anti- human PD-1 monoclonal antibody or antigen binding fragment thereof, an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin) or pharmaceutically acceptable salts thereof), to a subject having an infectious disease or an infection (e.g., caused by many pathogens, including bacteria, viruses, fungi) to achieve at least one positive therapeutic effect. The term “co-formulation” refers to a formulation comprising two or more of therapeutic agents. In some embodiments, co-formulation comprises a TIGIT antagonist and a PD-1 antagonist. The term “pharmaceutically acceptable carrier” refers to any inactive substance that is suitable for use in a formulation for the delivery of a therapeutic agent. A carrier may be an anti-adherent, binder, coating, disintegrant, filler or diluent, preservative (such as antioxidant, antibacterial, or antifungal agent), sweetener, absorption delaying agent, wetting agent, emulsifying agent, buffer, and the like. Examples of suitable pharmaceutically acceptable carriers include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), dextrose, vegetable oils (such as olive oil), saline, buffer, buffered saline, and isotonic agents such as sugars, polyalcohols, sorbitol, and sodium chloride. As used herein, the terms “combination,” “combination therapy,” and “therapeutic combination” refer to treatments in which at least one PD-1 antagonist, at least one TIGIT antagonist, and one or more chemotherapeutic agents or pharmaceutically acceptable salts thereof, and optionally additional therapeutic agents, each are administered to a patient in a coordinated manner, either sequentially, simultaneously or separately, over an overlapping period of time. In specific embodiments, the treatments are a combination of at least one anti- human TIGIT monoclonal antibody or antigen-binding fragment thereof, at least one anti- human PD-1 monoclonal antibody or antigen-binding fragment thereof, 5-fluorouracil and cisplatin; a combination of at least one anti-human TIGIT monoclonal antibody or antigen- binding fragment thereof, at least one anti-human PD-1 monoclonal antibody or antigen- binding fragment thereof, and paclitaxel or a pharmaceutically acceptable salt thereof; a combination of at least one anti-human TIGIT monoclonal antibody or antigen-binding fragment thereof, at least one anti-human PD-1 monoclonal antibody or antigen-binding fragment thereof, gemcitabine or a pharmaceutically acceptable salt thereof and cisplatin; or a combination of at least one anti-human TIGIT monoclonal antibody or antigen-binding fragment thereof, at least one anti-human PD-1 monoclonal antibody or antigen-binding fragment thereof, capecitabine or a pharmaceutically acceptable salt thereof and oxaliplatin; each optionally having additional therapeutic agents. The period of treatment with the at least one TIGIT antagonist (the “anti-TIGIT treatment”) is the period of time that a patient undergoes treatment with the TIGIT antagonist, e.g., an anti-human TIGIT monoclonal antibody (or antigen-binding fragment thereof); that is, the period of time from the initial dosing with the TIGIT antagonist through the final day of a treatment cycle. Similarly, the period of treatment with the at least one PD-1 antagonist (the “anti- PD-1 treatment”) is the period of time that a patient undergoes treatment with the PD-1 antagonist, e.g., an anti- human PD-1 monoclonal antibody (or antigen-binding fragment thereof); that is, the period of time from the initial dosing with the PD-1 antagonist through the final day of a treatment cycle. The period of treatment with 5-fluorouracil and/or cisplatin (the “5-FU/cisplatin treatment”) is the period of time that a patient undergoes treatment with 5-fluorouracil and/or cisplatin; that is, the period of time from the initial dosing with 5-fluorouracil and/or cisplatin through the final day of a treatment cycle. In the methods, uses, and therapeutic combinations described herein, the anti-TIGIT treatment overlaps by at least one day with the anti-PD-1 treatment and overlaps by at least one day with the 5-FU/cisplatin treatment. In certain embodiments, the anti-TIGIT treatment, the anti-PD-1 treatment, and the 5-FU/cisplatin treatment are the same period of time. In some embodiments, the anti-TIGIT treatment begins prior to the anti-PD-1 and/or the 5-FU/cisplatin treatment. In other embodiments, the anti- TIGIT treatment begins after the anti-PD-1 and/or the 5-FU/cisplatin treatment. In yet other embodiments, the anti-PD-1 treatment begins prior to the anti-TIGIT and/or the 5- FU/cisplatin treatment. In still other embodiments, the anti- PD-1 treatment begins after the anti-TIGIT and/or the 5-FU/cisplatin treatment. In some embodiments, the 5-FU/cisplatin treatment begins prior to the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the 5-FU/cisplatin treatment begins after the anti-PD-1 and/or the anti-TIGIT treatment. In certain embodiments, the anti-TIGIT treatment is terminated prior to termination of the anti-PD-1 and/or the 5-FU/cisplatin treatment. In other embodiments, the anti-TIGIT treatment is terminated after termination of the anti-PD-1 and/or the 5- FU/cisplatin treatment. In yet other embodiments, the anti-PD-1 treatment is terminated prior to termination of the anti-TIGIT and/or the 5-FU/cisplatin treatment. In still other embodiments, the anti-PD-1 treatment is terminated after termination of the anti-TIGIT and/or the 5-FU/cisplatin treatment. In certain embodiments, the 5-FU/cisplatin treatment is terminated prior to termination of the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the 5-FU/cisplatin treatment is terminated after termination of the anti-PD-1 and/or the anti-TIGIT treatment. The period of treatment with paclitaxel or a pharmaceutically acceptable salt thereof (the “paclitaxel treatment”) is the period of time that a patient undergoes treatment with paclitaxel or a pharmaceutically acceptable salt thereof; that is, the period of time from the initial dosing with paclitaxel or a pharmaceutically acceptable salt thereof through the final day of a treatment cycle. In the methods, uses, and therapeutic combinations described herein, the anti-TIGIT treatment overlaps by at least one day with the anti-PD-1 treatment and overlaps by at least one day with the paclitaxel treatment. In certain embodiments, the anti-TIGIT treatment, the anti-PD-1 treatment, and the paclitaxel treatment are the same period of time. In some embodiments, the anti-TIGIT treatment begins prior to the anti-PD-1 and/or the paclitaxel treatment. In other embodiments, the anti-TIGIT treatment begins after the anti-PD-1 and/or the paclitaxel treatment. In yet other embodiments, the anti-PD-1 treatment begins prior to the anti-TIGIT and/or the paclitaxel treatment. In still other embodiments, the anti- PD-1 treatment begins after the anti-TIGIT and/or the paclitaxel treatment. In some embodiments, the paclitaxel treatment begins prior to the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the paclitaxel treatment begins after the anti- PD-1 and/or the anti-TIGIT treatment. In certain embodiments, the anti-TIGIT treatment is terminated prior to termination of the anti-PD-1 and/or the paclitaxel treatment. In other embodiments, the anti-TIGIT treatment is terminated after termination of the anti-PD-1 and/or the paclitaxel treatment. In yet other embodiments, the anti-PD-1 treatment is terminated prior to termination of the anti-TIGIT and/or the paclitaxel treatment. In still other embodiments, the anti-PD-1 treatment is terminated after termination of the anti-TIGIT and/or the paclitaxel treatment. In certain embodiments, the paclitaxel treatment is terminated prior to termination of the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the paclitaxel treatment is terminated after termination of the anti-PD-1 and/or the anti-TIGIT treatment. The period of treatment with gemcitabine or a pharmaceutically acceptable salt thereof and/or cisplatin (the “gemcitabine/cisplatin treatment”) is the period of time that a patient undergoes treatment with gemcitabine or a pharmaceutically acceptable salt thereof and/or cisplatin; that is, the period of time from the initial dosing with gemcitabine or a pharmaceutically acceptable salt thereof and/or cisplatin through the final day of a treatment cycle. In the methods, uses, and therapeutic combinations described herein, the anti-TIGIT treatment overlaps by at least one day with the anti-PD-1 treatment and overlaps by at least one day with the gemcitabine/cisplatin treatment. In certain embodiments, the anti-TIGIT treatment, the anti-PD-1 treatment, and the gemcitabine/cisplatin treatment are the same period of time. In some embodiments, the anti-TIGIT treatment begins prior to the anti-PD-1 and/or the gemcitabine/cisplatin treatment. In other embodiments, the anti-TIGIT treatment begins after the anti-PD-1 and/or the gemcitabine/cisplatin treatment. In yet other embodiments, the anti-PD-1 treatment begins prior to the anti-TIGIT and/or the gemcitabine/cisplatin treatment. In still other embodiments, the anti- PD-1 treatment begins after the anti-TIGIT and/or the gemcitabine/cisplatin treatment. In some embodiments, the gemcitabine/cisplatin treatment begins prior to the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the gemcitabine/cisplatin treatment begins after the anti-PD-1 and/or the anti-TIGIT treatment. In certain embodiments, the anti-TIGIT treatment is terminated prior to termination of the anti-PD-1 and/or the gemcitabine/cisplatin treatment. In other embodiments, the anti-TIGIT treatment is terminated after termination of the anti-PD-1 and/or the gemcitabine/cisplatin treatment. In yet other embodiments, the anti-PD-1 treatment is terminated prior to termination of the anti-TIGIT and/or the gemcitabine/cisplatin treatment. In still other embodiments, the anti-PD-1 treatment is terminated after termination of the anti-TIGIT and/or the gemcitabine/cisplatin treatment. In certain embodiments, the gemcitabine/cisplatin treatment is terminated prior to termination of the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the gemcitabine/cisplatin treatment is terminated after termination of the anti-PD-1 and/or the anti-TIGIT treatment. The period of treatment with capecitabine or a pharmaceutically acceptable salt thereof and/or oxaliplatin (the “capecitabine/oxaliplatin treatment”) is the period of time that a patient undergoes treatment with capecitabine or a pharmaceutically acceptable salt thereof and/or oxaliplatin; that is, the period of time from the initial dosing with capecitabine or a pharmaceutically acceptable salt thereof and/or oxaliplatin through the final day of a treatment cycle. In the methods, uses, and therapeutic combinations described herein, the anti-TIGIT treatment overlaps by at least one day with the anti-PD-1 treatment and overlaps by at least one day with the capecitabine/oxaliplatin treatment. In certain embodiments, the anti-TIGIT treatment, the anti-PD-1 treatment, and the capecitabine/oxaliplatin treatment are the same period of time. In some embodiments, the anti-TIGIT treatment begins prior to the anti-PD-1 and/or the capecitabine/oxaliplatin treatment. In other embodiments, the anti- TIGIT treatment begins after the anti-PD-1 and/or the capecitabine/oxaliplatin treatment. In yet other embodiments, the anti-PD-1 treatment begins prior to the anti-TIGIT and/or the capecitabine/oxaliplatin treatment. In still other embodiments, the anti- PD-1 treatment begins after the anti-TIGIT and/or the capecitabine/oxaliplatin treatment. In some embodiments, the capecitabine/oxaliplatin treatment begins prior to the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the capecitabine/oxaliplatin treatment begins after the anti-PD-1 and/or the anti-TIGIT treatment. In certain embodiments, the anti-TIGIT treatment is terminated prior to termination of the anti-PD-1 and/or the capecitabine/oxaliplatin treatment. In other embodiments, the anti-TIGIT treatment is terminated after termination of the anti-PD-1 and/or the capecitabine/oxaliplatin treatment. In yet other embodiments, the anti-PD-1 treatment is terminated prior to termination of the anti- TIGIT and/or the capecitabine/oxaliplatin treatment. In still other embodiments, the anti-PD- 1 treatment is terminated after termination of the anti-TIGIT and/or the capecitabine/oxaliplatin treatment. In certain embodiments, the capecitabine/oxaliplatin treatment is terminated prior to termination of the anti-PD-1 and/or the anti-TIGIT treatment. In other embodiments, the capecitabine/oxaliplatin treatment is terminated after termination of the anti-PD-1 and/or the anti-TIGIT treatment. The terms “treatment regimen,” “dosing protocol,” and “dosing regimen” are used interchangeably to refer to the dose and timing of administration of each therapeutic agent in a combination therapy of the disclosure. The terms “cancer”, “cancerous”, or “malignant” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma. More particular examples of such cancers include, but are not limited to, squamous cell carcinoma, myeloma, small-cell lung cancer, non-small cell lung cancer, glioma, Hodgkin lymphoma, non-hodgkin's lymphoma, acute myeloid leukemia (AML), multiple myeloma, gastrointestinal (tract) cancer, renal cancer, ovarian cancer, liver cancer, lymphoblastic leukemia, lymphocytic leukemia, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, melanoma, chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, brain cancer, stomach cancer, bladder cancer, hepatoma, hepatocellular carcinoma, biliary cancer, esophageal cancer, breast cancer, triple negative breast cancer, colon carcinoma, and head and neck cancer. “Tumor” as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size, and includes primary tumors and secondary neoplasms. Non-limiting examples of tumors include solid tumor (e.g., sarcoma (such as chondrosarcoma), carcinoma (such as colon carcinoma), blastoma (such as hepatoblastoma), etc.) and blood tumor (e.g., leukemia (such as acute myeloid leukemia (AML)), lymphoma (such as DLBCL), multiple myeloma (MM), etc.). “Tumor burden” also referred to as “tumor load”, refers to the total amount of tumor material distributed throughout the body. Tumor burden refers to the total number of cancer cells or the total size of tumor(s), throughout the body, including lymph nodes and bone narrow. Tumor burden can be determined by a variety of methods known in the art, such as, e.g., by measuring the dimensions of tumor(s) upon removal from the subject, e.g., using calipers, or while in the body using imaging techniques, e.g., ultrasound, bone scan, computed tomography (CT) or magnetic resonance imaging (MRI) scans. The term “tumor volume” or “tumor size” refers to the total size of the tumor which can be measured as the length and width of a tumor. Tumor size may be determined by a variety of methods known in the art, such as, e.g., by measuring the dimensions of tumor(s) upon removal from the subject, e.g., using calipers, or while in the body using imaging techniques, e.g., bone scan, ultrasound, CT or MRI scans. As used herein, the term "effective amount" refer to an amount of a PD-1 antagonist, a TIGIT antagonist, and one or more chemotherapeutic agents or pharmaceutically acceptable salts thereof (e.g., an anti-TIGIT antibody or antigen binding fragment, an anti-PD-1 antibody or antigen binding fragment of the invention, and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin)) that, when administered alone or in combination with an additional therapeutic agent to a cell, tissue, or subject, is effective to cause a measurable improvement in one or more symptoms of an infection or a disease, for example cancer or the progression of cancer. An effective amount further refers to that amount of the antibody or fragment sufficient to result in at least partial amelioration of symptoms, e.g., tumor shrinkage or elimination, lack of tumor growth, increased survival time. When applied to an individual active ingredient administered alone, an effective amount refers to that ingredient alone. When applied to a combination, an effective amount refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously. An effective amount of a therapeutic may result in an improvement of a diagnostic measure or parameter by at least 10%; usually by at least 20%; preferably at least about 30%; more preferably at least 40%, and most preferably by at least 50%. An effective amount can also result in an improvement in a subjective measure in cases where subjective measures are used to assess disease severity. Toxicity and therapeutic efficacy of the antibodies or antigen binding fragments of the invention, administered alone or in combination with another therapeutic agent, can be determined by any number of systems or means. For example, the toxicity and therapeutic efficacy of the antibodies or antigen binding fragments or compounds of the invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, 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 (LD50/ ED50). The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration. It is understood that wherever embodiments are described herein with the language “comprising,” otherwise analogous embodiments described in terms of “consisting of” and/or “consisting essentially of” are also provided. “Consists essentially of,” and variations such as “consist essentially of” or “consisting essentially of,” as used throughout the specification and claims, indicate the inclusion of any recited elements or group of elements, and the optional inclusion of other elements, of similar or different nature than the recited elements, that do not materially change the basic or novel properties of the specified dosage regimen, method, or composition. Unless expressly stated to the contrary, all ranges cited herein are inclusive; i.e., the range includes the values for the upper and lower limits of the range as well as all values in between. As an example, temperature ranges, percentages, ranges of equivalents, and the like described herein include the upper and lower limits of the range and any value in the continuum there between. Numerical values provided herein, and the use of the term “about”, may include variations of ± 1%, ± 2%, ±3%, ± 4%, ± 5%, ± 10%, ± 15%, and ± 20% and their numerical equivalents. All ranges also are intended to include all included sub-ranges, although not necessarily explicitly set forth. For example, a range of 3 to 7 days is intended to include 3, 4, 5, 6, and 7 days. In addition, the term “or,” as used herein, denotes alternatives that may, where appropriate, be combined; that is, the term “or” includes each listed alternative separately as well as their combination. Where aspects or embodiments of the disclosure are described in terms of a Markush group or other grouping of alternatives, the present disclosure encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group, but also the main group absent one or more of the group members. The present disclosure also envisages the explicit exclusion of one or more of any of the group members in the claims. 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 relates. In case of conflict, the present specification, including definitions, will control. Throughout this specification and claims, the word “comprise,” or variations such as “comprises” or “comprising” will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers. Unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Any example(s) following the term “e.g.” or “for example” is not meant to be exhaustive or limiting. Exemplary methods and materials are described herein, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure. The materials, methods, and examples are illustrative only and not intended to be limiting. Abbreviations with expanded term Table 3: Abbreviations Abbreviation Expanded Term 1L Fi li DNA deoxyribonucleic acid i f mL Milliliter illi Q7D dose every 7 days g Provided herein are PD-1 antagonists or anti-human PD-1 monoclonal antibodies that can be used in any of the methods, compositions, kits, and uses disclosed herein, including any chemical compound or biological molecule that blocks binding of PD-L1 to PD-1 and preferably also blocks binding of PD-L2 to PD-1. Any monoclonal antibodies that bind to a PD-1 polypeptide, a PD-1 polypeptide fragment, a PD-1 peptide, or a PD-1 epitope and block the interaction between PD-1 and its ligand PD-L1 or PD-L2 can be used. In some embodiments, the anti-human PD-1 monoclonal antibody binds to a PD-1 polypeptide, a PD-1 polypeptide fragment, a PD-1 peptide, or a PD-1 epitope and blocks the interaction between PD-1 and PD-L1. In other embodiments, the anti-human PD-1 monoclonal antibody binds to a PD-1 polypeptide, a PD- 1 polypeptide fragment, a PD-1 peptide, or a PD-1 epitope and blocks the interaction between PD-1 and PD-L2. In yet other embodiments, the anti-human PD-1 monoclonal antibody binds to a PD-1 polypeptide, a PD-1 polypeptide fragment, a PD-1 peptide, or a PD-1 epitope and blocks the interaction between PD-1 and PD-L1 and the interaction between PD-1 and PD- L2. Any monoclonal antibodies that bind to a PD-L1 polypeptide, a PD-L1 polypeptide fragment, a PD-L1 peptide, or a PD-L1 epitope and block the interaction between PD-L1 and PD-1 can also be used. In certain embodiments, the anti-human PD-1 monoclonal antibody is selected from the group consisting of pembrolizumab, nivolumab, cemiplimab, pidilizumab (U.S. Pat. No. 7,332,582), AMP-514 (MedImmune LLC, Gaithersburg, MD), PDR001 (U.S. Pat. No. 9,683,048), BGB-A317 (U.S. Pat. No.8,735,553), and MGA012 (MacroGenics, Rockville, MD). In one embodiment, the anti-human PD-1 monoclonal antibody is pembrolizumab. In one embodiment, the anti-human PD-1 monoclonal antibody is pembrolizumab. In another embodiment, the anti-human PD-1 monoclonal antibody is nivolumab. In another embodiment, the anti-human PD-1 monoclonal antibody is cemiplimab. In yet another embodiment, the anti-human PD-1 monoclonal antibody is pidilizumab. In one embodiment, the anti-human PD-1 monoclonal antibody is AMP-514. In another embodiment, the anti- human PD-1 monoclonal antibody is PDR001. In yet another embodiment, the anti-human PD-1 monoclonal antibody is BGB-A317. In still another embodiment, the anti-human PD-1 monoclonal antibody is MGA012. In some embodiments, an anti-human PD-1 antibody or antigen binding fragment thereof for use in the methods, kits, uses and co-formulations of the invention comprises three light chain CDRs of CDRL1, CDRL2 and CDRL3 and/or three heavy chain CDRs of CDRH1, CDRH2 and CDRH3. In one embodiment of the invention, CDRL1 comprises the amino acid sequence as set forth in SEQ ID NO:1 or a variant of the amino acid sequence as set forth in SEQ ID NO:1, CDRL2 comprises the amino acid sequence as set forth in SEQ ID NO:2 or a variant of the amino acid sequence as set forth in SEQ ID NO:2, and CDRL3 comprises the amino acid sequence as set forth in SEQ ID NO:3 or a variant of the amino acid sequence as set forth in SEQ ID NO:3. In one embodiment, CDRH1 comprises the amino acid sequence as set forth in SEQ ID NO:6 or a variant of the amino acid sequence as set forth in SEQ ID NO:6, CDRH2 comprises the amino acid sequence as set forth in SEQ ID NO: 7 or a variant of the amino acid sequence as set forth in SEQ ID NO:7, and CDRH3 comprises the amino acid sequence as set forth in SEQ ID NO:8 or a variant of the amino acid sequence as set forth in SEQ ID NO:8. In one embodiment, the three light chain CDRs have the amino acid sequences as set forth in SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3 and the three heavy chain CDRs have the amino acid sequences as set forth in SEQ ID NO:6, SEQ ID NO:7 and SEQ ID NO:8. In an alternative embodiment of the invention, CDRL1 comprises the amino acid sequence as set forth in SEQ ID NO:11 or a variant of the amino acid sequence as set forth in SEQ ID NO:11, CDRL2 comprises the amino acid sequence as set forth in SEQ ID NO:12 or a variant of the amino acid sequence as set forth in SEQ ID NO:12, and CDRL3 comprises the amino acid sequence as set forth in SEQ ID NO:13 or a variant of the amino acid sequence as set forth in SEQ ID NO:13. In one embodiment, CDRH1 comprises the amino acid sequence as set forth in SEQ ID NO:16 or a variant of the amino acid sequence as set forth in SEQ ID NO:16, CDRH2 comprises the amino acid sequence as set forth in SEQ ID NO:17 or a variant of the amino acid sequence as set forth in SEQ ID NO:17, and CDRH3 comprises the amino acid sequence as set forth in SEQ ID NO:18 or a variant of the amino acid sequence as set forth in SEQ ID NO:18. In one embodiment, the three light chain CDRs have the amino acid sequences as set forth in SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3 and the three heavy chain CDRs have the amino acid sequences as set forth in SEQ ID NO:6, SEQ ID NO:7 and SEQ ID NO:8. In an alternative embodiment, the three light chain CDRs have the amino acid sequences as set forth in SEQ ID NO:11, SEQ ID NO:12, and SEQ ID NO:13 and the three heavy chain CDRs have the amino acid sequences as set forth in SEQ ID NO:16, SEQ ID NO:17 and SEQ ID NO:18. In a further embodiment of the invention, CDRL1 comprises the amino acid sequence as set forth in SEQ ID NO:21 or a variant of the amino acid sequence as set forth in SEQ ID NO:21, CDRL2 comprises the amino acid sequence as set forth in SEQ ID NO:22 or a variant of the amino acid sequence as set forth in SEQ ID NO:22, and CDRL3 comprises the amino acid sequence as set forth in SEQ ID NO:23 or a variant of the amino acid sequence as set forth in SEQ ID NO:23. In yet another embodiment, CDRH1 comprises the amino acid sequence as set forth in SEQ ID NO:24 or a variant of the amino acid sequence as set forth in SEQ ID NO:24, CDRH2 comprises the amino acid sequence as set forth in SEQ ID NO: 25 or a variant of the amino acid sequence as set forth in SEQ ID NO:25, and CDRH3 comprises the amino acid sequence as set forth in SEQ ID NO:26 or a variant of the amino acid sequence as set forth in SEQ ID NO:26. In another embodiment, the three light chain CDRs have the amino acid sequences as set forth in SEQ ID NO:21, SEQ ID NO:22, and SEQ ID NO:23 and the three heavy chain CDRs have the amino acid sequences as set forth in SEQ ID NO:24, SEQ ID NO:25 and SEQ ID NO:26. Some anti-human PD-1 antibody and antigen binding fragments of the invention comprise a light chain variable region and a heavy chain variable region. In some embodiments, the light chain variable region comprises the amino acid sequence as set forth in SEQ ID NO:4 or a variant of the amino acid sequence as set forth in SEQ ID NO:4, and the heavy chain variable region comprises the amino acid sequence as set forth in SEQ ID NO:9 or a variant of the amino acid sequence as set forth in SEQ ID NO:9. In further embodiments, the light chain variable region comprises the amino acid sequence as set forth in SEQ ID NO:14 or a variant of the amino acid sequence as set forth in SEQ ID NO:14, and the heavy chain variable region comprises the amino acid sequence as set forth in SEQ ID NO:19 or a variant of the amino acid sequence as set forth in SEQ ID NO:19. In further embodiments, the heavy chain variable region comprises the amino acid sequence as set forth in SEQ ID NO:27 or a variant of the amino acid sequence as set forth in SEQ ID NO:27 and the light chain variable region comprises the amino acid sequence as set forth in SEQ ID NO:28 or a variant of the amino acid sequence as set forth in SEQ ID NO:28, SEQ ID NO:29 or a variant of the amino acid sequence as set forth in SEQ ID NO:29, or SEQ ID NO:30 or a variant of the amino acid sequence as set forth in SEQ ID NO:30. In such embodiments, a light chain variable region or heavy chain variable region sequence is identical to the reference sequence except having one, two, three, four or five amino acid substitutions. In some embodiments, the substitutions are in the framework region (i.e., outside of the CDRs). In some embodiments, one, two, three, four or five of the amino acid substitutions are conservative substitutions. In one embodiment of the co-formulations, methods, kits or uses of the invention, the anti-human PD-1 antibody or antigen binding fragment comprises a light chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:4 and a heavy chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:9. In a further embodiment, the anti-human PD-1 antibody or antigen binding fragment comprises a light chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:14 and a heavy chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:19. In one embodiment of the formulations of the invention, the anti-human PD-1 antibody or antigen binding fragment comprises a light chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:28 and a heavy chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:27. In a further embodiment, the anti-human PD-1 antibody or antigen binding fragment comprises a light chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:29 and a heavy chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:27. In another embodiment, the antibody or antigen binding fragment comprises a light chain variable region comprising or consisting of the amino acid sequence as set forth in SEQ ID NO:30 and a heavy chain variable region comprising or consisting the the amino acid sequence as set forth in SEQ ID NO:27. In another embodiment, the co-formulations, methods, kits or uses of the invention comprise an anti-human PD-1 antibody or antigen binding protein that has a V L domain and/or a V H domain with at least 95%, 90%, 85%, 80%, 75% or 50% sequence homology to one of the V L domains or V H domains described above, and exhibits specific binding to PD-1. In another embodiment, the anti-human PD-1 antibody or antigen binding protein of the co- formulations of the invention comprises V L and V H domains having up to 1, 2, 3, 4, or 5 or more amino acid substitutions, and exhibits specific binding to PD-1. In any of the embodiments above, the PD-1 antagonist may be a full-length anti-PD-1 antibody or an antigen binding fragment thereof that specifically binds human PD-1. In certain embodiments, the PD-1 antagonist is a full-length anti-PD-1 antibody selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA, and IgE. Preferably, the antibody is an IgG antibody. Any isotype of IgG can be used, including IgG 1 , IgG 2 , IgG 3 , and IgG 4 . Different constant domains may be appended to the V L and V H regions provided herein. For example, if a particular intended use of an antibody (or fragment) of the present invention were to call for altered effector functions, a heavy chain constant domain other than IgG1 may be used. Although IgG1 antibodies provide for long half-life and for effector functions, such as complement activation and antibody-dependent cellular cytotoxicity, such activities may not be desirable for all uses of the antibody. In such instances an IgG4 constant domain, for example, may be used. In embodiments of the invention, the PD-1 antagonist is an anti-PD-1 antibody comprising a light chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:5 and a heavy chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:10. In alternative embodiments, the PD-1 antagonist is an anti-PD-1 antibody comprising a light chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:15 and a heavy chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:20. In further embodiments, the PD-1 antagonist is an anti-PD-1 antibody comprising a light chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:32 and a heavy chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:31. In additional embodiments, the PD-1 antagonist is an anti-PD-1 antibody comprising a light chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:33 and a heavy chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:31. In yet additional embodiments, the PD-1 antagonist is an anti-PD-1 antibody comprising a light chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:34 and a heavy chain comprising or consisting of a sequence of amino acid residues as set forth in SEQ ID NO:31. In some co-formulations, methods, kits or uses of the invention, the PD-1 antagonist is pembrolizumab or a pembrolizumab biosimilar. In some co-formulations, methods, kits or uses of the invention, the PD-1 antagonist is nivolumab or a nivolumab biosimilar. Ordinarily, amino acid sequence variants of the anti-PD-1 antibodies and antigen binding fragments of the invention and the anti-TIGIT antibodies and antigen binding fragments will have an amino acid sequence having at least 75% amino acid sequence identity with the amino acid sequence of a reference antibody or antigen binding fragment (e.g. heavy chain, light chain, V H , V L , or humanized sequence), more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, and most preferably at least 95, 98, or 99%. Identity or homology with respect to a sequence is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the reference antibody or antigen binding fragment residues, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. None of N- terminal, C-terminal, or internal extensions, deletions, or insertions into the antibody sequence shall be construed as affecting sequence identity or homology. Sequence identity refers to the degree to which the amino acids of two polypeptides are the same at equivalent positions when the two sequences are optimally aligned. Sequence identity can be determined using a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences. The following references relate to BLAST algorithms often used for sequence analysis: BLAST ALGORITHMS: Altschul, S.F., et al., (1990) J. Mol. Biol.215:403-410; Gish, W., et al., (1993) Nature Genet.3:266-272; Madden, T.L., et al., (1996) Meth. Enzymol.266:131-141; Altschul, S.F., et al., (1997) Nucleic Acids Res. 25:3389-3402; Zhang, J., et al., (1997) Genome Res.7:649-656; Wootton, J.C., et al., (1993) Comput. Chem.17:149-163; Hancock, J.M. et al., (1994) Comput. Appl. Biosci.10:67-70; ALIGNMENT SCORING SYSTEMS: Dayhoff, M.O., et al., "A model of evolutionary change in proteins." in Atlas of Protein Sequence and Structure, (1978) vol.5, suppl.3. M.O. Dayhoff (ed.), pp.345-352, Natl. Biomed. Res. Found., Washington, DC; Schwartz, R.M., et al., "Matrices for detecting distant relationships." in Atlas of Protein Sequence and Structure, (1978) vol.5, suppl.3." M.O. Dayhoff (ed.), pp.353-358, Natl. Biomed. Res. Found., Washington, DC; Altschul, S.F., (1991) J. Mol. Biol.219:555-565; States, D.J., et al., (1991) Methods 3:66-70; Henikoff, S., et al., (1992) Proc. Natl. Acad. Sci. USA 89:10915-10919; Altschul, S.F., et al., (1993) J. Mol. Evol.36:290-300; ALIGNMENT STATISTICS: Karlin, S., et al., (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268; Karlin, S., et al., (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877; Dembo, A., et al., (1994) Ann. Prob.22:2022-2039; and Altschul, S.F. "Evaluating the statistical significance of multiple distinct local alignments." in Theoretical and Computational Methods in Genome Research (S. Suhai, ed.), (1997) pp.1- 14, Plenum, New York. Likewise, either class of light chain can be used in the compositions and methods herein. Specifically, kappa, lambda, or variants thereof are useful in the present compositions and methods. Table 4. Exemplary PD-1 Antibody Sequences Antibody Amino Acid Sequence SEQ ID Feature NO. Antibody Amino Acid Sequence SEQ ID Feature NO. Formulations, Compositions, Methods, Kits and Uses of the Invention. A. Antibodies and antigen binding fragments comprising light and heavy chain CDRs of hPD- 1.08A in WO2008/156712 eavy c a Q : L ight chain VR SEQ ID NO:28 or SEQ ID NO:29 or SEQ ID NO:30 Light chain SEQ ID NO:32 or SEQ ID NO:33 or SEQ ID NO:34 TIGIT Antagonists Provided herein are anti-human TIGIT monoclonal antibodies or antigen binding fragments thereof that can be used in the methods, pharmaceutical compositions, kits, and uses disclosed herein. Any monoclonal antibodies that bind to a TIGIT polypeptide, a TIGIT polypeptide fragment, a TIGIT peptide, or a TIGIT epitope and block the interaction between TIGIT and its ligand CD155 and/or CD112 can be used. In some embodiments, the anti- human TIGIT monoclonal antibody binds to a TIGIT polypeptide, a TIGIT polypeptide fragment, a TIGIT peptide, or a TIGIT epitope and blocks the interaction between TIGIT and CD155. In other embodiments, the anti-human TIGIT monoclonal antibody binds to a TIGIT polypeptide, a TIGIT polypeptide fragment, a TIGIT peptide, or a TIGIT epitope and blocks the interaction between TIGIT and CD112. In yet other embodiments, the anti-human TIGIT monoclonal antibody binds to a TIGIT polypeptide, a TIGIT polypeptide fragment, a TIGIT peptide, or a TIGIT epitope and blocks the interaction between TIGIT and CD155 and the interaction between TIGIT and CD112. In some embodiments the human constant region is selected from the group consisting of IgG1, IgG2, IgG3 and IgG4 constant regions, and in preferred embodiments, the human constant region is an IgG1 or IgG4 constant region. Exemplary anti-TIGIT antibody sequences are set forth below in Tables 6 and 7. Table 6. Exemplary anti-TIGIT antibodies Description SEQ SEQUENCE ID QPPGKGLEWIGFITYSGSTSYNPSLKSRITISRDTSKNQ FSLKLSSVTAADTAVYYCARMPSFITLASLSTWEGYF QKPGKAPKFLIYYANSLQTGIPSRFSGSGSGTDFTLTIS SLQPEDFATYYCQQYYSGWTFGGGTKVEIK QKPGKAPKLLIYYANSLQTGVPSRFSGSGSGTDFTLTI LB155.14A6.G2.A8_VL3 SSLQPEDFATYFCQQYYSGWTFGGGTKVEIK 31C6 H – CDR2 VARIANT (D56Y) 121 YIDPYNyGAKYNEKFG LQPEDFATYYCQHHFGSPLTFGQGTRLEIK 31C6_Humz_L2 151 DIQMTQSPSSLSASVGDRVTITCRASEHIYSYLSWYQQ X24= D or A X25= T or V X 6 = A or V X7 = F or V X 15 = W, A, D, E, F, G, I, K, N, Q, R, S, T, V or Y 31C6 VL 171 DIQMTQSPX 1 SLSASVGX 2 X 3 VTITCRASEHIYSYLSWY Vibostolimab, heavy chain 295 EVQLVQSGAEVKKPGSSVKVSCKASGYTFSSYVMHW VRQAPGQGLEWIGYIDPYNDGAKYAQKFQGRVTLTS , y g g g comprises three light chain CDRs of CDRL1, CDRL2, and CDRL3 and/or three heavy chain CDRs of CDRH1, CDRH2, and CDRH3. In one embodiment, the anti- TIGIT antibody or antigen binding fragment thereof comprises a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:35, a CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO:36, a CDRH3 comprising any of the amino acid sequences as set forth in SEQ ID NOs:37, 103, 104, 105, 106, 107, or 160, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:38, a CDRL2 comprising any of the amino acid sequences as set forth in SEQ ID NOs:39, 89, 90, 91, 92, 93, 94, 95, 96, 97, or 161, and a CDRL3 comprising any of the amino acid sequences as set forth in SEQ ID NOs:40, 98, 99, 100, 101, 102, or 162. In another embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:81, a CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO:82, a CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO:83, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:84, a CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO:85, and a CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO:86. In another embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:108, a CDRH2 comprising any of the amino acid sequences as set forth in SEQ ID NOs:109, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 154, 155 or 167, a CDRH3 comprising any of the amino acid sequences as set forth in SEQ ID NOs:110, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186 or 187, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:111, a CDRL2 comprising any of the amino acid sequences as set forth in SEQ ID NOs:112, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142 or 168, and a CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO:113. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:35, a CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO:36, a CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO:37, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:38, a CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO:39, and a CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO:40. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:108, a CDRH2 comprising any one of the amino acid sequences as set forth in SEQ ID NO:109, 154 or 145, a CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO:110, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:111, a CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO:112, and a CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO:113. In another embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:108, a CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, a CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO:110, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:111, a CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO:112, and a CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO:113. In some embodiments, the anti- TIGIT antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region and a variable light chain variable region. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:41 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:42. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:87 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:88. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO: 114 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:115. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 43-58, 65-75 and 87 and a variable light chain region comprising any one of the amino acid sequences as set forth in SEQ ID NOs: 59-64, 76-80 and 88. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 144-149 and a variable light chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 150-153. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:148 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:152. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:147 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:150. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:148 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:153. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:163 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:165. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:169 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:171. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:164 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:166. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:170 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:172. Table 7. Exemplary sequences of anti-TIGIT antibodies Description SEQ ID SEQUENCE NO X 8 = V, I Hu14D7V L1 201 EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTS chain) IK 14D7 H – CDR3 220 ALYEGFYFDY Hu14D7V L1 242 EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTS (Humanized VL TLASGVPARFSGSGSGTDYTLTISSLEPEDFAVYYCHQGSSFPYTFGQGTKLE Hu26B10 VH1 265 EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGG (Humanized VH LKPDDGGISYNQKFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAY chain)(N55G) YRYDADYWGQGTLVTVSS Hu26B10 VH3 286 EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGG n one em o ment, t e ant - ant o y or ant gen n ng ragment compr ses a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:188, a CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO:189, a CDRH3 comprising any of the amino acid sequences as set forth in SEQ ID NOs:190, 220, 221, or 222, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:191, a CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO:192, and a CDRL3 comprising any of the amino acid sequences as set forth in SEQ ID NOs:193, 232, 233, 234, 235, 236, or 237. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO:204, a CDRH2 comprising any of the amino acid sequences as set forth in SEQ ID NOs: 205, 256, 257, 258, 259, 260, 261, 262, or 263, a CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO:206, a CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO:207, a CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO:208, and a CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO:209. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region and a variable light chain variable region. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:194 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:195. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO:196 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:200. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO: 210 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:211. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising the amino acid sequence as set forth in SEQ ID NO: 212 and a variable light chain region comprising the amino acid sequence as set forth in SEQ ID NO:216. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 197, 198, 199, 223, 224, 225, 226, 227, 228, 229, 230, and 231 and a variable light chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 201, 202, 203, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, and 255. In one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof comprises a variable heavy chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 213, 214, 215, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, and 286 and a variable light chain region comprising any of the amino acid sequences as set forth in SEQ ID NOs: 217, 218, and 219. Additional anti-TIGIT antibodies which may be used in the formulations described herein include those disclosed, for example, in PCT International Application No. WO 2016/106302; WO 2016/011264; and WO 2009/126688. Table 8. Exemplary Heavy Chain Sequences Heavy chain 291 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHT constant domain– FPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCD I 1 KTHT PP PAPELL P FLFPPKPKDTLMI RTPE T D HEDPE K In any of the above mentioned embodiments, the anti-TIGIT antibody or antigen binding fragment thereof is an antibody comprising any of the variable heavy chains described above and any human heavy chain constant domain. In one embodiment, the antibody or antigen binding fragment thereof of the invention is of the IgG isotype, and comprises a human IgG1, IgG2, IgG3 or IgG4 human heavy chain constant domain. In one embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human heavy chain IgG1 constant domain (SEQ ID NO: 291) or a variant thereof, wherein the variant comprises up to 20 modified amino acid substitutions. In one embodiment, the antibody or antigen binding fragment thereof of the invention is an antibody comprising a human heavy chain IgG1 constant domain comprising the amino acid sequence as set forth in SEQ ID NO: 291. In one embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human heavy chain IgG1 constant domain wherein the IgG1 constant domain is afucosylated. In one embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human heavy chain IgG4 constant domain or a variant thereof, wherein the variant comprises up to 20 modified amino acid substitutions. In another embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human heavy chain IgG4 constant domain, wherein the amino acid at position 228 (using EU numbering scheme) has been substituted from Ser to Pro. In one embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human heavy chain IgG4 constant domain comprising the amino acid sequence as set forth in SEQ ID NO: 292. In any of the above mentioned embodiments, the anti-TIGIT antibody or antigen binding fragment thereof can comprise any of the variable light chains described above and human light chain constant domain. In one embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human kappa light chain constant domain or a variant thereof, wherein the variant comprises up to 20 modified amino acid substitutions. In another embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human lambda light chain constant domain or a variant thereof, wherein the variant comprises up to 20 modified amino acid substitutions. In one embodiment, the antibody or antigen binding fragment thereof of the invention comprises a human kappa light chain constant domain comprising the amino acid sequence as set forth in SEQ ID NO: 293. Methods of Treating Cancer Using a Combination of a PD-1 Antagonist, a TIGIT Antagonist, 5-fluorouracil, and Cisplatin In another aspect, provided herein are methods of treating cancer (e.g., esophageal cancer) using a combination of a TIGIT antagonist, a PD-1 antagonist, 5-fluorouracil and cisplatin. In certain embodiments, the TIGIT antagonist is an anti-TIGIT antibody or antigen binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody or antigen binding fragment thereof. In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I); and I) sented by Formula (II). I) ments, the cancer is selected from the group consisting of: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma) colorectal; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome); hematopoietic tumors of the lymphoid lineage, include leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, mantle cell lymphoma, myeloma, and Burkett’s lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; and other tumors, including melanoma, skin (non- melanomal) cancer, mesothelioma (cells), seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi’s sarcoma. In some embodiments, the cancer is selected from the group consisting of anal cancer, biliary tract cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer (CRC), esophageal cancer, gastrointestinal cancer, glioblastoma, glioma, head and neck cancer (HNSCC), hepatocellular carcinoma (HCC), lung cancer, liver cancer, lymphoma, melanoma, mesothelioma, multiple myeloma, non-small cell lung cancer (NSCLC), ovarian cancer, pancreatic cancer, prostate cancer (e.g. hormone refractory prostate adenocarcinoma), renal cell carcinoma (RCC), salivary cancer, thyroid cancer, and other neoplastic malignancies. In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). In some embodiments, the cancer is esophageal cancer. In some embodiments, the esophageal cancer is locally advanced unresectable. In some embodiments, the esophageal cancer is metastatic adenocarcinoma. In some embodiments, the esophageal cancer is squamous cell carcinoma of the esophagus (ESCC). In some embodiments, the esophageal cancer is advanced Siewert type 1 adenocarcinoma of the gastroesophageal junction (GEJ). In some embodiments, the esophageal cancer is metastatic Siewert type 1 adenocarcinoma of the gastroesophageal junction (GEJ). In some embodiments, a combination of a TIGIT antagonist, a PD-1 antagonist, 5- fluorouracil and cisplatin may provide enhanced efficacy as compared with existing treatments. For example, TIGIT and PD-L1 are tightly co-expressed in adenocarcinoma and ESCC. A combination of a TIGIT antagonist, a PD-1 antagonist may provide added benefit over single-agent checkpoint blockade. Without wishing to be bound by any particular theory, chemotherapeutic agents may augment antitumor immune responses by (i) inducing immunogenic cell death, (ii) enhancing the maturation and activation of dendritic cells, (iii) increasing T-cell penetrance and function in the tumor, (iv) improving the presentation of tumor antigens, and (v) eliminating immunosuppressive cells. In certain embodiments, the tolerability of vibostolimab (an anti-TIGIT antibody) is a potential added benefit for combination with one or more chemotherapeutic agents and a PD-1 antagonist. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In certain embodiments, provided herein is a method of treating esophageal cancer (e.g., locally advanced unresectable, metastatic adenocarcinoma, ESCC, advanced Siewert type 1 adenocarcinoma of GEJ, or metastatic Siewert type 1 adenocarcinoma of GEJ), comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist as disclosed in Section titled TIGIT Antagonists; (b) a PD-1 antagonist as disclosed in Section titled PD-1 Antagonists; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-L1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-L1 monoclonal antibody is a humanized antibody. In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. Thus, in certain embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In some embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In other embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In one embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is pembrolizumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is nivolumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is cemiplimab. In certain embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 of comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In certain embodiments, the anti- TIGIT antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating esophageal cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating esophageal cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating esophageal cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). Methods of Treating Cancer Using a Combination of a PD-1 Antagonist, a TIGIT Antagonist, and Paclitaxel or a Pharmaceutically Acceptable Salt Thereof In another aspect, provided herein are methods of treating cancer (e.g., TNBC) using a combination of a TIGIT antagonist, a PD-1 antagonist, and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. In certain embodiments, the TIGIT antagonist is an anti-TIGIT antibody or antigen binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody or antigen binding fragment thereof. In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. In some embodiments, the cancer is selected from the group consisting of: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma) colorectal; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome); hematopoietic tumors of the lymphoid lineage, include leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, mantle cell lymphoma, myeloma, and Burkett’s lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; and other tumors, including melanoma, skin (non- melanomal) cancer, mesothelioma (cells), seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi’s sarcoma. In some embodiments, the cancer is selected from the group consisting of anal cancer, biliary tract cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer (CRC), esophageal cancer, gastrointestinal cancer, glioblastoma, glioma, head and neck cancer (HNSCC), hepatocellular carcinoma (HCC), lung cancer, liver cancer, lymphoma, melanoma, mesothelioma, multiple myeloma, non-small cell lung cancer (NSCLC), ovarian cancer, pancreatic cancer, prostate cancer (e.g. hormone refractory prostate adenocarcinoma), renal cell carcinoma (RCC), salivary cancer, thyroid cancer, and other neoplastic malignancies. In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). In some embodiments, the cancer is TNBC. In some embodiments, the TNBC is recurrent unresectable. In some embodiments, the TNBC is metastatic. In some embodiments, a combination of a TIGIT antagonist, a PD-1 antagonist, and paclitaxel, or a pharmaceutically acceptable salt thereof may provide enhanced efficacy as compared with existing treatments. For example, TNBC may be an immune-reactive disease amenable to immune modulation of the tumor microenvironment, and expresses TIGIT and PD-L1 highly. Without wishing to be bound by any particular theory, chemotherapeutic agents may augment antitumor immune responses by (i) inducing immunogenic cell death, (ii) enhancing the maturation and activation of dendritic cells, (iii) increasing T-cell penetrance and function in the tumor, (iv) improving the presentation of tumor antigens, and (v) eliminating immunosuppressive cells. In certain embodiments, the tolerability of vibostolimab is a potential added benefit for combination with chemotherapy and a PD-1 antagonist. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In certain embodiments, provided herein is a method of treating TNBC, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is a method of treating hepatocellular carcinoma, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist as disclosed in Section titled TIGIT Antagonists; (b) a PD-1 antagonist as disclosed in Section titled PD-1 Antagonists; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-L1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-L1 monoclonal antibody is a humanized antibody. In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. Thus, in certain embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In other embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is pembrolizumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is nivolumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is cemiplimab. In certain embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In certain embodiments, the anti- TIGIT antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment of the methods provided herein, the method for treating TNBC comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment of the methods provided herein, the method for treating TNBC comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment of the methods provided herein, the method for treating TNBC comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. Methods of Treating Cancer Using a Combination of a PD-1 Antagonist, a TIGIT Antagonist, Gemcitabine, and Cisplatin In another aspect, provided herein are methods of treating cancer (e.g., biliary cancer) using a combination of a TIGIT antagonist, a PD-1 antagonist, gemcitabine and cisplatin. In certain embodiments, the TIGIT antagonist is an anti-TIGIT antibody or antigen binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody or antigen binding fragment thereof. In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In some embodiments, the cancer is selected from the group consisting of: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma) colorectal; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome); hematopoietic tumors of the lymphoid lineage, include leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, mantle cell lymphoma, myeloma, and Burkett’s lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; and other tumors, including melanoma, skin (non- melanomal) cancer, mesothelioma (cells), seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi’s sarcoma. In some embodiments, the cancer is selected from the group consisting of anal cancer, biliary tract cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer (CRC), esophageal cancer, gastrointestinal cancer, glioblastoma, glioma, head and neck cancer (HNSCC), hepatocellular carcinoma (HCC), lung cancer, liver cancer, lymphoma, melanoma, mesothelioma, multiple myeloma, non-small cell lung cancer (NSCLC), ovarian cancer, pancreatic cancer, prostate cancer (e.g. hormone refractory prostate adenocarcinoma), renal cell carcinoma (RCC), salivary cancer, thyroid cancer, and other neoplastic malignancies. In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, and triple negative breast cancer (TNBC). In some embodiments, the cancer is biliary cancer. In some embodiments, the biliary cancer is locally recurrent unresectable. In some embodiments, the biliary cancer is metastatic. In some embodiments, a combination of a TIGIT antagonist, a PD-1 antagonist, gemcitabine and cisplatin may provide enhanced efficacy as compared with existing treatments. For example, TIGIT and PD-L1 are tightly co-expressed in adenocarcinoma and ESCC. A combination of a TIGIT antagonist, a PD-1 antagonist may provide added benefit over single-agent checkpoint blockade. Without wishing to be bound by any particular theory, chemotherapeutic agents may augment antitumor immune responses by (i) inducing immunogenic cell death, (ii) enhancing the maturation and activation of dendritic cells, (iii) increasing T-cell penetrance and function in the tumor, (iv) improving the presentation of tumor antigens, and (v) eliminating immunosuppressive cells. In certain embodiments, the tolerability of vibostolimab is a potential added benefit for combination with one or more chemotherapeutic agents and a PD-1 antagonist. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In certain embodiments, provided herein is a method of treating biliary cancer (e.g., locally recurrent unresectable, or metastatic), comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist as disclosed in Section titled TIGIT Antagonists; (b) a PD-1 antagonist as disclosed in Section titled PD-1 Antagonists; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-L1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-L1 monoclonal antibody is a humanized antibody. In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. Thus, in certain embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In some embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In other embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In one embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is pembrolizumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is nivolumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is cemiplimab. In certain embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In certain embodiments, the anti- TIGIT antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating biliary cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating biliary cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In one specific embodiment of the methods provided herein, the method for treating biliary cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). Methods of Treating Cancer Using a Combination of a PD-1 Antagonist, a TIGIT Antagonist, Capecitabine, and Oxaliplatin In another aspect, provided herein are methods of treating cancer (e.g., gastric cancer) using a combination of a TIGIT antagonist, a PD-1 antagonist, capecitabine or a pharmaceutically acceptable salt thereof and oxaliplatin. In certain embodiments, the TIGIT antagonist is an anti-TIGIT antibody or antigen binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody or antigen binding fragment thereof. In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In some embodiments, the cancer is selected from the group consisting of: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma) colorectal; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome); hematopoietic tumors of the lymphoid lineage, include leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, mantle cell lymphoma, myeloma, and Burkett’s lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; and other tumors, including melanoma, skin (non- melanomal) cancer, mesothelioma (cells), seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi’s sarcoma. In some embodiments, the cancer is selected from the group consisting of anal cancer, biliary tract cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer (CRC), esophageal cancer, gastrointestinal cancer, glioblastoma, glioma, head and neck cancer (HNSCC), hepatocellular carcinoma (HCC), lung cancer, liver cancer, lymphoma, melanoma, mesothelioma, multiple myeloma, non-small cell lung cancer (NSCLC), ovarian cancer, pancreatic cancer, prostate cancer (e.g. hormone refractory prostate adenocarcinoma), renal cell carcinoma (RCC), salivary cancer, thyroid cancer, and other neoplastic malignancies. In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC) and gastric cancer. In some embodiments, the cancer is gastric cancer. In some embodiments, the gastric cancer is advanced or GEJ adenocarcinoma. In some embodiments, the gastric cancer is HER2 negative. In some embodiments, the gastric cancer is previously untreated. In some embodiments, a combination of a TIGIT antagonist, a PD-1 antagonist, gemcitabine and cisplatin may provide enhanced efficacy as compared with existing treatments. For example, TIGIT and PD-L1 are tightly co-expressed in adenocarcinoma and ESCC. A combination of a TIGIT antagonist, a PD-1 antagonist may provide added benefit over single-agent checkpoint blockade. Without wishing to be bound by any particular theory, chemotherapeutic agents may augment antitumor immune responses by (i) inducing immunogenic cell death, (ii) enhancing the maturation and activation of dendritic cells, (iii) increasing T-cell penetrance and function in the tumor, (iv) improving the presentation of tumor antigens, and (v) eliminating immunosuppressive cells. In certain embodiments, the tolerability of vibostolimab is a potential added benefit for combination with one or more chemotherapeutic agents and a PD-1 antagonist. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In certain embodiments, provided herein is a method of treating gastric cancer, comprising administering to a human patient in need thereof: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In certain embodiments, the method of treating cancer comprises administering to a human patient in need thereof: (a) a TIGIT antagonist as disclosed in Section titled TIGIT Antagonists; (b) a PD-1 antagonist as disclosed in Section titled PD-1 Antagonists; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-L1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-L1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-L1 monoclonal antibody is a humanized antibody. In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. Thus, in certain embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In some embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In other embodiments, provided herein is a method for treating cancer, comprising administering to a human patient in need thereof: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is pembrolizumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is nivolumab. In another embodiment of the methods, compositions, kits and uses provided herein, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof is cemiplimab. In certain embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of the methods, compositions, kits and uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In certain embodiments, the anti- TIGIT antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment of the methods provided herein, the method for treating cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment of the methods provided herein, the method for treating gastric cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment of the methods provided herein, the method for treating gastric cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment of the methods provided herein, the method for treating gastric cancer comprises administering to a human patient in need thereof: (a) an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). Dosing and Administration Further provided herein are dosing regimens and routes of administration for treating cancer (e.g., esophageal cancer, TNBC, biliary cancer, gastric cancer) using a combination of a TIGIT antagonist (e.g., an anti-TIGIT monoclonal antibody or antigen binding fragment thereof), a PD-1 antagonist (e.g., an anti-PD-1 monoclonal antibody or antigen binding fragment thereof), and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin). The TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof), the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof), or one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin) disclosed herein may be administered by doses administered, e.g., daily, 1-7 times per week, weekly, bi-weekly, tri-weekly, every four weeks, every five weeks, every 6 weeks, monthly, bimonthly, quarterly, semiannually, annually, etc. Doses may be administered, e.g., intravenously, subcutaneously, topically, orally, nasally, rectally, intramuscular, intracerebrally, intraspinally, or by inhalation. In certain embodiments, the doses are administered intravenously. In certain embodiments, the doses are administered subcutaneously. In certain embodiments, the doses are administered orally. In some embodiments, the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti- TIGIT monoclonal antibody) or antigen binding fragment thereof) is administered subcutaneously or intravenously, on a weekly, biweekly, triweekly, every 4 weeks, every 5 weeks, every 6 weeks, every 8 weeks, every 9 weeks, every 10 weeks, every 12 weeks, monthly, bimonthly, or quarterly basis at about 10, about 20, about 50, about 80, about 100, about 200, about 300, about 400, about 500, about 1000 or about 2500 mg/subject. In some specific methods, the dose of the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) is from about 0.01 mg/kg to about 50 mg/kg, from about 0.05 mg/kg to about 25 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, from about 0.2 mg/kg to about 9 mg/kg, from about 0.3 mg/kg to about 8 mg/kg, from about 0.4 mg/kg to about 7 mg/kg, from about 0.5 mg/kg to about 6 mg/kg, from about 0.6 mg/kg to about 5 mg/kg, from about 0.7 mg/kg to about 4 mg/kg, from about 0.8 mg/kg to about 3 mg/kg, from about 0.9 mg/kg to about 2 mg/kg, from about 1.0 mg/kg to about 1.5 mg/kg, from about 1.0 mg/kg to about 2.0 mg/kg, from about 1.0 mg/kg to about 3.0 mg/kg, or from about 2.0 mg/kg to about 4.0 mg/kg. In some specific methods, the dose of the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof) is from about 10 mg to about 500 mg, from about 25 mg to about 500 mg, from about 50 mg to about 500 mg, from about 100 mg to about 500 mg, from about 200 mg to about 500 mg, from about 150 mg to about 250 mg, from about 175 mg to about 250 mg, from about 200 mg to about 250 mg, from about 150 mg to about 240 mg, from about 175 mg to about 240 mg, or from about 200 mg to about 240 mg. In some embodiments, the dose of the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof) is about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 240 mg, about 250 mg, about 300 mg, about 400 mg, or about 500 mg. In some embodiments, the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof) is administered subcutaneously or intravenously, on a weekly, biweekly, triweekly, every 4 weeks, every 5 weeks, every 6 weeks, every 8 weeks, every 9 weeks, every 10 weeks, every 12 weeks, monthly, bimonthly, or quarterly basis at about 10, about 20, about 50, about 80, about 100, about 200, about 300, about 400, about 500, about 1000 or about 2500 mg/subject. In some specific methods, the dose of the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof) is from about 0.01 mg/kg to about 50 mg/kg, from about 0.05 mg/kg to about 25 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, from about 0.2 mg/kg to about 9 mg/kg, from about 0.3 mg/kg to about 8 mg/kg, from about 0.4 mg/kg to about 7 mg/kg, from about 0.5 mg/kg to about 6 mg/kg, from about 0.6 mg/kg to about 5 mg/kg, from about 0.7 mg/kg to about 4 mg/kg, from about 0.8 mg/kg to about 3 mg/kg, from about 0.9 mg/kg to about 2 mg/kg, from about 1.0 mg/kg to about 1.5 mg/kg, from about 1.0 mg/kg to about 2.0 mg/kg, from about 1.0 mg/kg to about 3.0 mg/kg, or from about 2.0 mg/kg to about 4.0 mg/kg. In some specific methods, the dose of the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof) is from about 10 mg to about 500 mg, from about 25 mg to about 500 mg, from about 50 mg to about 500 mg, from about 100 mg to about 500 mg, from about 200 mg to about 500 mg, from about 150 mg to about 250 mg, from about 175 mg to about 250 mg, from about 200 mg to about 250 mg, from about 150 mg to about 240 mg, from about 175 mg to about 240 mg, or from about 200 mg to about 240 mg. In some embodiments, the dose of the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof) is about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 240 mg, about 250 mg, about 300 mg, about 400 mg, or about 500 mg. In some embodiments of the methods described herein, the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110, and the human patient is administered about 100 mg, about 150 mg, about 200 mg, about 240 mg, about 400 mg, about 480 mg, or about 720 mg, or about 2 mg/kg of the TIGIT antagonist, wherein the TIGIT antagonist is administered once every three or six weeks. In one embodiment, the human patient is administered about 200 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every three weeks. In one embodiment, the human patient is administered 240 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every three weeks. In one embodiment, the human patient is administered 2 mg/kg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every three weeks. In one embodiment, the human patient is administered 400 mg a TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every three weeks. In certain embodiments of the methods described herein, the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110, the human patient is administered 400 mg anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody), and the anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) is administered once every six weeks. In some embodiments of the methods described herein, the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110, and the human patient is administered 200 mg, 240 mg, 400 mg, 480 mg, 720 mg, or 2 mg/kg of the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof), wherein the TIGIT antagonist is administered once every six weeks. In one embodiment, the human patient is administered 200 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every six weeks. In one embodiment, the human patient is administered 240 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every six weeks. In one embodiment, the human patient is administered 400 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every six weeks. In one embodiment, the human patient is administered about 480 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every six weeks. In one embodiment, the human patient is administered 720 mg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every six weeks. In one embodiment, the human patient is administered 2 mg/kg TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof) once every six weeks. In some embodiments of the methods described herein, the PD-1 antagonist, anti-PD- 1 antibody or anti-PD-1 monoclonal antibody is pembrolizumab, the human patient is administered about 200 mg, about 240 mg, about 400 mg, about 480 mg, about 720 mg, or about 2 mg/kg pembrolizumab, and pembrolizumab is administered once every three or six weeks. In one embodiment, the human patient is administered about 200 mg pembrolizumab once every three weeks. In one embodiment, the human patient is administered about 240 mg pembrolizumab once every three weeks. In one embodiment, the human patient is administered 2 mg/kg pembrolizumab once every three weeks. In one embodiment, the human patient is administered 400 mg pembrolizumab once every three weeks. In certain embodiments of the methods described herein, the PD-1 antagonist, anti- PD-1 antibody or anti-PD-1 monoclonal antibody is pembrolizumab, the human patient is administered 400 mg pembrolizumab, and pembrolizumab is administered once every six weeks. In some embodiments of the methods described herein, the PD-1 antagonist, anti-PD- 1 antibody or anti-PD-1 monoclonal antibody is pembrolizumab, the human patient is administered about 200 mg, about 240 mg, about 400 mg, about 480 mg, about 720 mg, or about 2 mg/kg pembrolizumab, and pembrolizumab is administered once every six weeks. In one embodiment, the human patient is administered about 200 mg pembrolizumab once every six weeks. In one embodiment, the human patient is administered about 240 mg pembrolizumab once every six weeks. In one embodiment, the human patient is administered about 400 mg pembrolizumab once every six weeks. In one embodiment, the human patient is administered 480 mg pembrolizumab once every six weeks. In one embodiment, the human patient is administered 720 mg pembrolizumab once every six weeks. In one embodiment, the human patient is administered 2 mg/kg pembrolizumab once every six weeks. In some embodiments of the invention, the TIGIT antagonist (e.g., anti-TIGIT antibody, or antigen binding fragment thereof), and the PD-1 antagonist (e.g., anti-PD-1 antibody, or antigen binding fragment thereof), are administered to the patient once every approximately six weeks for 12 weeks or more. In other embodiments, the TIGIT antagonist (e.g., anti-TIGIT antibody, or antigen binding fragment) and the PD-1 antagonist (e.g., anti- PD-1 antibody, or antigen binding fragment thereof), are administered to the patient once every six weeks for 18 weeks or more, 24 weeks or more, 30 weeks or more, 36 weeks or more, 42 weeks or more, 48 weeks or more, 54 weeks or more, 60 weeks or more, 66 weeks or more, 72 weeks or more, 78 weeks or more, 84 weeks or more, or 90 weeks or more. In one embodiment, the administration occurs on the same day. In a sub-embodiment, the TIGIT antagonist (e.g., anti-TIGIT antibody, or antigen binding fragment thereof), and the PD-1 antagonist (e.g., anti-PD-1 antibody, or antigen binding fragment thereof), are administered on the same day sequentially, simultaneously or separately (e.g., in a single formulation, a co-formulation, or concurrently as separate formulations). In an alternative embodiment, the anti-TIGIT antibody or antigen binding fragment thereof and the anti-PD-1 antibody or antigen binding fragment thereof are administered sequentially on the same day (e.g., as separate formulations), in either order. In one embodiment of same day sequential administration, the anti-TIGIT antibody or antigen binding fragment thereof is administered first. In another embodiment of same day sequential administration, the anti-PD-1 antibody or antigen binding fragment thereof is administered first. In certain embodiments of the methods compositions, kits and uses described herein, the TIGIT antagonist (e.g., anti- human TIGIT antibody or antigen binding fragment thereof) comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110, the human patient is administered 200 mg of the TIGIT antagonist, and the TIGIT antagonist is administered once every three weeks. In certain embodiments of the methods compositions, kits and uses described herein, the TIGIT antagonist (e.g., anti- human TIGIT antibody or antigen binding fragment thereof) comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110, and the human patient is administered about 400 mg of the TIGIT antagonist, and the TIGIT antagonist is administered once every six weeks. In certain embodiments of the methods compositions, kits and uses described herein, the PD-1 antagonist or anti-human PD-1 monoclonal antibody is pembrolizumab, the human patient is administered about 200 mg pembrolizumab, and pembrolizumab is administered once every three weeks. In certain embodiments of the methods compositions, kits and uses described herein, the PD-1 antagonist or anti-human PD-1 monoclonal antibody is pembrolizumab, the human patient is administered about 400 mg pembrolizumab, and pembrolizumab is administered once every six weeks. In other embodiments of the methods compositions, kits and uses described herein, the PD-antagonist or anti-human PD-1 monoclonal antibody is nivolumab, the human patient is administered about 240 mg or about 3 mg/kg nivolumab, and nivolumab is administered once every two weeks. In one specific embodiment, the human patient is administered about 240 mg nivolumab once every two weeks. In one specific embodiment, the human patient is administered about 3 mg/kg nivolumab once every two weeks. In other embodiments of the methods described herein, the PD-1 antagonist or anti-human PD-1 monoclonal antibody is nivolumab, the human patient is administered about 480 mg nivolumab, and nivolumab is administered once every four weeks. In yet other embodiments of the methods compositions, kits and uses described herein, the PD-1 antagonist (e.g, anti-human PD-1 monoclonal antibody or antigen binding fragment thereof) is cemiplimab, the human patient is administered about 350 mg cemiplimab, and cemiplimab is administered once every three weeks. In some embodiments, the TIGIT antagonist (e.g., an anti-TIGIT antibody) and the PD-1 antagonist (e.g, anti-PD-1 antibody) are co-formulated. In one embodiment, a co- formulated product with about 200 mg pembrolizumab or a pembrolizumab variant and 200 mg of an antibody comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 is used for intravenous infusion. In one embodiment, a co-formulated product with 200 mg pembrolizumab or a pembrolizumab variant and 300 mg of antibody comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 is used for intravenous infusion. In one embodiment, a co-formulated product with about 200 mg pembrolizumab or a pembrolizumab variant and about 400 mg of antibody comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 is used for intravenous infusion. In another embodiment, a co-formulated product with about 200 mg of pembrolizumab or a pembrolizumab variant and about 500 mg of antibody comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 is used for intravenous infusion. In another embodiment, a co-formulated product with of about 200 mg pembrolizumab or a pembrolizumab variant and about 600 mg of antibody comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 is used for intravenous infusion. In another embodiment, a co-formulated product with about 200 mg of pembrolizumab or a pembrolizumab variant and about 700 mg of antibody comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 is used for intravenous infusion. In certain embodiments, 5-fluorouracil is administered intravenously (e.g., as a continuous IV infusion). In certain embodiments, 5-fluorouracil is administered five times every three weeks. In some embodiments, a human patient is administered a total of about 3000 mg/m 2 or about 4000 mg/m 2 5-fluorouracil per cycle. In some embodiments, a human patient is administered about 500 mg/m 2 , about 600 mg/m 2 , about 650 mg/m 2 , about 700 mg/m 2 , about 750 mg/m 2 , about 800 mg/m 2 , or about 900 mg/m 2 5-fluorouracil per day wherein the 5-fluorouracil is administered five times every three weeks. In some embodiments, 5-fluorouracil is administered on the first five days of a 3-week cycle. In certain embodiments, cisplatin is administered intravenously (e.g., a 60 minutes to 120 minutes IV infusion). In certain embodiments, cisplatin is administered once every three weeks. In some embodiments, a human patient is administered about 40 mg/m 2 to about 100 mg/m 2 cisplatin once every three weeks. In some embodiments, a human patient is administered about 60 mg/m 2 to about 80 mg/m 2 cisplatin once every three weeks. Thus, in some embodiments of the methods compositions, kits and uses provided herein, the human patient is administered: (a) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg pembrolizumab; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three or six weeks, and wherein (c) is administered five times every three or six week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL pembrolizumab; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) a total of about 3000 mg/m 2 or 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m 2 or 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) a total of about 3000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; (c) a total of about 3000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; (c) a total of about 3000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 60 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 240 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 240 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is are administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 2 mg/kg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), and (d) is administered once every three weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 400 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a) and (b) is administered once every six weeks, wherein (c) is administered five times every three weeks, and wherein (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a) and (b) is administered once every six weeks, wherein (c) is administered five times every three weeks, and wherein (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (d) is administered once every three weeks, wherein (b) is administered once every six weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) a total of about 4000 mg/m 2 5-fluorouracil represented by Formula (I) per cycle; and (d) about 80 mg/m 2 cisplatin represented by Formula (II), wherein each of (a) and(d) is administered once every three weeks, wherein (b) is administered once every six weeks, and wherein (c) is administered five times every three weeks. In certain embodiments of the methods, compositions, kits and uses described herein, a co-formulation of pembrolizumab and of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113 and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 are administered by IV infusion. In some embodiments, the co-formulation is administered for about 30 minutes every three weeks. In some embodiments, the co- formulation is administered for from about 25 to about 40 minutes every three weeks. In certain embodiments, paclitaxel or a pharmaceutically acceptable salt thereof is administered intravenously. In certain embodiments, paclitaxel or a pharmaceutically acceptable salt thereof is administered once every week. In some embodiments, a human patient is administered about 70 mg/m 2 to about 100 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof once every week. In some embodiments, a human patient is administered about 80 mg/m 2 to about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof once every week. In some embodiments, a human patient is administered about 70 mg/m 2 , about 80 mg/m 2 , about 90 mg/m 2 , or about 100 mg/m 2 , paclitaxel or a pharmaceutically acceptable salt thereof once every week. In some embodiments, a human patient is administered about 80 mg/m 2 , or about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof once every week. Thus, in some embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg pembrolizumab; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three or six weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; and (b) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL pembrolizumab; and (c) about 80 mg/m 2 to about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; and (c) about 80 mg/m 2 or about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; and (c) about 80 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQcomprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQcomprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; and (c) about 80 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; and (c) about 80 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 240 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 240 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 2 mg/kg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every three weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 400 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every six weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein each of (a) and (b) is administered once every six weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein (a) is administered once every three weeks, wherein (b) is administered once every six weeks, and wherein (c) is administered once a week. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; and (c) about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, wherein (a) is administered once every three weeks, wherein (b) is administered once every six weeks, and wherein (c) is administered once a week. In certain embodiments, gemcitabine or a pharmaceutically acceptable salt thereof is administered intravenously (e.g., as a continuous IV infusion). In certain embodiments, gemcitabine or a pharmaceutically acceptable salt thereof is administered once every three weeks. In some embodiments, a human patient is administered about 500 mg/m 2 to about 1500 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three week. In some embodiments, a human patient is administered about 800 mg/m 2 to about 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three week. In some embodiments, a human patient is administered about 500 mg/m 2 , about 600 mg/m 2 , about 700 mg/m 2 , about 800 mg/m 2 , about 900 mg/m 2 , about 1000 mg/m 2 , about 1100 mg/m 2 , about 1200 mg/m 2 , about 1300 mg/m 2 , about 1400 mg/m 2 , or about 1500 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three week. In some embodiments, a human patient is administered about 800 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three week. In some embodiments, a human patient is administered about 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof once every three week. In certain embodiments, cisplatin is administered intravenously (e.g., a 60 minutes to 120 minutes IV infusion). In certain embodiments, cisplatin is administered once every three weeks. In some embodiments, a human patient is administered about 10 mg/m 2 to about 50 mg/m 2 cisplatin once every three weeks. In some embodiments, a human patient is administered about 20 mg/m 2 to about 25 mg/m 2 cisplatin once every three weeks. In some embodiments, a human patient is administered about 10 mg/m 2 , about 15 mg/m 2 , about 20 mg/m 2 , about 25 mg/m 2 , about 30 mg/m 2 , about 35 mg/m 2 , about 40 mg/m 2 , about 45 mg/m 2 , or about 50 mg/m 2 cisplatin once every three weeks. Thus, in some embodiments of the methods compositions, kits and uses provided herein, the human patient is administered: (a) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg pembrolizumab; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three or six weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL pembrolizumab; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) about 800 mg/m 2 or 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 20 mg/m 2 or 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) about 800 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 20 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; (c) about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; (c) about 800 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 20 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; (c) about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks.. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; (c) about 800 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 20 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 240 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 240 mg pembrolizumab; (c) about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 200 mg/m 2 or about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 2 mg/kg pembrolizumab; (c) about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 200 mg/m 2 or about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 400 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 200 mg/m 2 or about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a) and (b) is administered once every six weeks, and each of (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 200 mg/m 2 or about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (b), (c) and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine represented by Formula (IV) or a pharmaceutically acceptable salt thereof; and (d) about 200 mg/m 2 or about 25 mg/m 2 cisplatin represented by Formula (II), wherein each of (a), (c) and (d) is administered once every three weeks, and (b) is administered once every six weeks. In certain embodiments, capecitabine or a pharmaceutically acceptable salt thereof is administered orally. In certain embodiments, capecitabine or a pharmaceutically acceptable salt thereof is administered twice a day. In some embodiments, a human patient is administered about 500 mg/m 2 to about 1500 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. In some embodiments, a human patient is administered about 750 mg/m 2 to about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. In some embodiments, a human patient is administered about 750 mg/m 2 , about 800 mg/m 2 , about 850 mg/m 2 , about 900 mg/m 2 , about 950 mg/m 2 , or about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. In some embodiments, a human patient is administered about 750 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. In some embodiments, a human patient is administered about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof twice a day. In certain embodiments, oxaliplatin is administered intravenously (e.g., a 60 minutes to 120 minutes IV infusion). In certain embodiments, oxaliplatin is administered once every three weeks. In some embodiments, a human patient is administered about 80 mg/m 2 to about 150 mg/m 2 oxaliplatin once every three weeks. In some embodiments, a human patient is administered about 100 mg/m 2 to about 130 mg/m 2 cisplatin once every three weeks. In some embodiments, a human patient is administered about 100 mg/m 2 , about 120 mg/m 2 , or about 130 mg/m 2 oxaliplatin once every three weeks. Thus, in some embodiments of the methods compositions, kits and uses provided herein, the human patient is administered: (a) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 100 mg, about 150 mg, about 200 mg, about 250 mg, or about 400 mg, or about 2 mg/kg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), and (b) is administered once every three or six weeks, (c) is administered twice a day, and (d) is administered once every three weeks. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg, about 240 mg, about 2 mg/kg or about 22 mg/mL pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 200 mg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152; (b) about 200 mg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 295; (b) about 200 mg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 240 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 240 mg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 2 mg/kg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 2 mg/kg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 400 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a), (b), and (d) is administered once every three weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses provided herein, the human patient is administered: (a) about 200 mg of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) about 400 mg pembrolizumab; (c) about 750 mg/m 2 or 1000 mg/m 2 capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) about 100 mg/m 2 or 130 mg/m 2 oxaliplatin represented by Formula (VI), wherein each of (a) and (d) is administered once every three weeks, (b) is administered once every six weeks, and (c) is administered twice a day. In certain embodiments of the methods, compositions, kits and uses described herein, a co-formulation of pembrolizumab and of an anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113 and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110 are administered by IV infusion. In some embodiments, the co-formulation is administered for about 30 minutes every three weeks. In some embodiments, the co- formulation is administered for from about 25 to about 40 minutes every three weeks. In some embodiments, at least one of the therapeutic agents (e.g., the anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or binding fragment thereof, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or binding fragment thereof, 5-fluorouracil, cisplatin, or paclitaxel) in the combination therapy is administered using the same dosage regimen (dose, frequency, and duration of treatment) that is typically employed when the agent is used as monotherapy for treating the same condition. In other embodiments, the patient receives a lower total amount of at least one of the therapeutic agents (e.g., the anti- TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or binding fragment thereof, the anti- PD-1monoclonal antibody or binding fragment thereof, 5-fluorouracil, cisplatin, or paclitaxel) in the combination therapy than when the agent is used as monotherapy, e.g., smaller doses, less frequent doses, and/or shorter treatment duration. A combination therapy disclosed herein may be used prior to or following surgery to remove a tumor and may be used prior to, during, or after radiation treatment. In some embodiments, a combination therapy disclosed herein is administered to a patient who has not previously been treated with a biotherapeutic or chemotherapeutic agent, i.e., is treatment-naïve. In other embodiments, the combination therapy is administered to a patient who failed to achieve a sustained response after prior therapy with the biotherapeutic or chemotherapeutic agent, i.e., is treatment-experienced. The therapeutic combination disclosed herein may be used in combination with one or more other active agents, including but not limited to, other anti-cancer agents that are used in the prevention, treatment, control, amelioration, or reduction of risk of a particular disease or condition (e.g., cancer). Such other active agents may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with one or more of the therapeutic agents in the combinations disclosed herein. The one or more additional active agents may be co-administered with the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof), the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof), or one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, or paclitaxel). The additional active agent(s) can be administered in a single dosage form with one or more co-administered agent selected from the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof), the PD-1 antagonist, e.g., anti-PD-1 antibody (e.g., anti- PD-1 monoclonal antibody) or antigen binding fragment thereof), and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, or paclitaxel). The additional active agent(s) can also be administered in separate dosage form(s) from the dosage forms containing the TIGIT antagonist (e.g., anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof), the PD-1 antagonist (e.g., anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof), or one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin). Administration of a PD-1 antagonist and/or a TIGIT antagonist can be by any suitable route, and can be facilitated by agents such as hyaluronan degrading enzymes, including hyaluronidases, including soluble PH20 polypeptides, and variants thereof. For systemic administration, the facilitating agents can be modified to increase pharmacological properties, such as serum half-life, by modifying the agents, such as with polymers. See, e.g., U.S. Patent Nos.7,767,429, 8,431,380, 7,871,607, International Publication No. WO 2020/022791, U.S. Patent Publication No. US2006/0104968 and European Patent 1858926, and in numerous other patents and publications. Exemplary of such agents is the known agent PEGPH20 or rHuPH20. Accordingly, specific embodiments cover pharmaceutical compositions comprising a PD-1 antagonist and/or a TIGIT antagonist and any one of a hyaluronan degrading enzyme, hyaluronidase, soluble hyaluronidase, soluble PH20 polypeptide, or a variant of any of the foregoing enzymes. In particular embodiments, the pharmaceutical composition comprises a PD-1 antagonist and a soluble PH20 polypeptide or a variant thereof. In another particular embodiments, the pharmaceutical composition comprises a TIGIT antagonist and a soluble PH20 polypeptide or a variant thereof. In some embodiments, the pharmaceutical composition comprises a PD-1 antagonist, a TIGIT antagonist and a soluble PH20 polypeptide or a variant thereof. In some embodiments, the pharmaceutical composition comprises a PD-1 antagonist, a TIGIT antagonist, 5-fluorouracil, cisplatin, and a soluble PH20 polypeptide or a variant thereof. In some embodiments, the pharmaceutical composition comprises a PD-1 antagonist, a TIGIT antagonist, paclitaxel or a pharmaceutically acceptable salt thereof, and a soluble PH20 polypeptide or a variant thereof. In some embodiments, the pharmaceutical composition comprises a PD-1 antagonist, a TIGIT antagonist, gemcitabine or a pharmaceutically acceptable salt thereof, cisplatin and a soluble PH20 polypeptide or a variant thereof. In some embodiments, the pharmaceutical composition comprises a PD-1 antagonist, a TIGIT antagonist, capecitabine or a pharmaceutically acceptable salt thereof, oxaliplatin and a soluble PH20 polypeptide or a variant thereof. Pharmaceutical Compositions In yet another aspect, provided herein are pharmaceutical compositions comprising the therapeutic agents disclosed herein (e.g., a TIGIT antagonist, a PD-1 antagonist, and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin)). In certain embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier. The pharmaceutical compositions comprising an anti-human TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof, an anti-human PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof, and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin), can be prepared for storage by mixing the antibodies or compounds having the desired degree of purity with optionally physiologically acceptable carriers, excipients, or stabilizers (see, e.g., Remington, Remington’s Pharmaceutical Sciences (18 th ed.1980)) in the form of aqueous solutions or lyophilized or other dried forms. The pharmaceutically acceptable carriers, excipients, or stabilizers are non-toxic to the cell or mammalian being exposed thereto at the dosage and concentrations employed. Often the pharmaceutically acceptable carrier is an aqueous pH buffered solution. Examples of pharmaceutically acceptable carriers include buffers, such as phosphate, citrate, acetate, and other organic acids; antioxidants, such as ascorbic acid; low molecular weight (e.g., fewer than about 10 amino acid residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulin; hydrophilic polymers, such as polyvinylpyrrolidone; amino acids, such as glycine, glutamine, asparagine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates, including glucose, mannose, or dextrins; chelating agents, such as EDTA; sugar alcohols, such as mannitol or sorbitol; salt-forming counterions, such as sodium; and/or nonionic surfactants, such as TWEEN TM , polyethylene glycol (PEG), and PLURONICS TM . The pharmaceutically acceptable carriers can also refer to a diluent, adjuvate (e.g., Freund’s adjuvate (complete or incomplete)), excipient, or vehicle. Such carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. Water is an exemplary carrier when a composition (e.g., a pharmaceutical composition) is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable excipients (e.g., pharmaceutical excipients) include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol, and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. Compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations, and the like. Kits In still another aspect, provided herein are kits comprising the therapeutic agents disclosed herein (e.g., a TIGIT antagonist, a PD-1 antagonist, and one or more chemotherapeutic agents (e.g., 5-fluorouracil, cisplatin, paclitaxel or a pharmaceutically acceptable salt thereof, gemcitabine or a pharmaceutically acceptable salt thereof, capecitabine or a pharmaceutically acceptable salt thereof, and/or oxaliplatin)) or pharmaceutical compositions thereof, packaged into suitable packaging material. A kit optionally includes a label or packaging insert that include a description of the components or instructions for use in vitro, in vivo, or ex vivo, of the components therein. In some embodiments, the kit comprises (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I) and (d) cisplatin represented by Formula (II). In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, 5-fluorouracil and cisplatin. In one embodiment, the kit comprises: (a) one or more dosages of an anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof; (b) one or more dosages of an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof; (c) one or more dosages of 5-fluorouracil; (d) one or more dosages of cisplatin; and (e) instructions for administering to a human patient the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, 5-fluorouracil and cisplatin. In some embodiments, the TIGIT antagonist is an anti-TIGIT antibody (e.g., anti- TIGIT monoclonal antibody) or antigen-binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen- binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-L1 monoclonal antibody or antigen-binding fragment thereof. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. The dosages for the anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody), the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody), 5-fluorouracil or cisplatin described herein can be used in various kits herein. In some embodiments, a kit comprises dosages of each component sufficient for a certain period of treatment (e.g., 3, 6, 12, or 24 weeks, etc.). For example, a kit can comprise a dosage of about 200 mg pembrolizumab, a dosage of about 200 mg anti-TIGIT antibody, 5 dosages of about 600 mg/m 2 or about 800 mg/m 2 5-fluorouracil, and a dosage of about 60 or 80 mg/m 2 cisplatin, which are sufficient for a 3-week treatment. Or, a kit can also comprise a dosage of about 400 mg pembrolizumab, 1 dosage of about 400 mg anti- TIGIT antibody, 10 dosages of about 600 mg/m 2 or about 800 mg/m 2 5-fluorouracil, and 2 dosages of about 60 or 80 mg/m 2 cisplatin, which are sufficient for a 6-week treatment. In some embodiments, the kit comprises (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel represented by Formula (III) or a pharmaceutically acceptable salt thereof. In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, and paclitaxel or a pharmaceutically acceptable salt thereof. In one embodiment, the kit comprises: (a) one or more dosages of an anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof; (b) one or more dosages of an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof; (c) one or more dosages of paclitaxel or a pharmaceutically acceptable salt thereof; and (d) instructions for administering to a human patient the anti- human TIGIT monoclonal antibody or antigen binding fragment thereof, the anti-human PD- 1 monoclonal antibody or antigen binding fragment thereof, and paclitaxel or a pharmaceutically acceptable salt thereof. In some embodiments, the TIGIT antagonist is an anti-TIGIT antibody (e.g., anti- TIGIT monoclonal antibody) or antigen-binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen- binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-L1 monoclonal antibody or antigen-binding fragment thereof. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. The dosages for the anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody), the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody), or paclitaxel or a pharmaceutically acceptable salt thereof described herein can be used in various kits herein. In some embodiments, a kit comprises dosages of each component sufficient for a certain period of treatment (e.g., 3, 6, 12, or 24 weeks, etc.). For example, a kit can comprise a dosage of about 200 mg pembrolizumab, a dosage of about 200 mg anti-TIGIT antibody, and a dosage of about 80 mg/m 2 or about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, which are sufficient for a 3-week treatment. Or, a kit can also comprise a dosage of about 400 mg pembrolizumab, a dosage of about 400 mg anti- TIGIT antibody, 2 dosages of about 80 mg/m 2 or about 90 mg/m 2 paclitaxel or a pharmaceutically acceptable salt thereof, which are sufficient for a 6-week treatment. In some embodiments, the kit comprises (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and (d) cisplatin represented by Formula (II). In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, gemcitabine or pharmaceutically acceptable salt thereof and cisplatin. In one embodiment, the kit comprises: (a) one or more dosages of an anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof; (b) one or more dosages of an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof; (c) one or more dosages of gemcitabine or a pharmaceutically acceptable salt thereof; (d) one or more dosages of cisplatin; and (e) instructions for administering to a human patient the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, gemcitabine or a pharmaceutically acceptable salt thereof, and cisplatin. In some embodiments, the TIGIT antagonist is an anti-TIGIT antibody (e.g., anti- TIGIT monoclonal antibody) or antigen-binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen- binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-L1 monoclonal antibody or antigen-binding fragment thereof. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. The dosages for the anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody), the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody), gemcitabine or a pharmaceutically acceptable salt thereof or cisplatin described herein can be used in various kits herein. In some embodiments, a kit comprises dosages of each component sufficient for a certain period of treatment (e.g., 3, 6, 12, or 24 weeks, etc.). For example, a kit can comprise a dosage of about 200 mg pembrolizumab, a dosage of about 200 mg anti-TIGIT antibody, a dosage of about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof, and a dosage of about 20 mg/m 2 to about 25 mg/m 2 cisplatin, which are sufficient for a 3-week treatment. Or, a kit can also comprise a dosage of about 400 mg pembrolizumab, 1 dosage of about 400 mg anti- TIGIT antibody, 2 dosages of about 800 mg/m 2 or about 1000 mg/m 2 gemcitabine or a pharmaceutically acceptable salt thereof, and about 20 mg/m 2 to about 25 mg/m 2 cisplatin, which are sufficient for a 6-week treatment. In some embodiments, the kit comprises (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof and (d) oxaliplatin represented by Formula (VI). In certain embodiments, the kit further comprises instructions for administering to a human patient the TIGIT antagonist, the PD-1 antagonist, capecitabine or a pharmaceutically acceptable salt and oxaliplatin. In one embodiment, the kit comprises: (a) one or more dosages of an anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody) or antigen binding fragment thereof; (b) one or more dosages of an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof; (c) one or more dosages of capecitabine or a pharmaceutically acceptable salt; (d) one or more dosages of oxaliplatin; and (e) instructions for administering to a human patient the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof, the anti-human PD-1 monoclonal antibody or antigen binding fragment thereof, capecitabine or a pharmaceutically acceptable salt and oxaliplatin. In some embodiments, the TIGIT antagonist is an anti-TIGIT antibody (e.g., anti- TIGIT monoclonal antibody) or antigen-binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen- binding fragment thereof. In some embodiments, the PD-1 antagonist is an anti-PD-L1 monoclonal antibody or antigen-binding fragment thereof. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. The dosages for the anti-TIGIT antibody (e.g., anti-TIGIT monoclonal antibody), the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody), capecitabine or a pharmaceutically acceptable salt or oxaliplatin described herein can be used in various kits herein. In some embodiments, a kit comprises dosages of each component sufficient for a certain period of treatment (e.g., 3, 6, 12, or 24 weeks, etc.). For example, a kit can comprise a dosage of about 200 mg pembrolizumab, a dosage of about 200 mg anti-TIGIT antibody, 62 dosages of about 750 or about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof, and a dosage of about 100 or about 130 mg/m 2 oxaliplatin, which are sufficient for a 3-week treatment. Or, a kit can also comprise a dosage of about 400 mg pembrolizumab, 1 dosage of about 400 mg anti- TIGIT antibody, 124 dosages of about 750 or about 1000 mg/m 2 capecitabine or a pharmaceutically acceptable salt thereof, and 2 dosages of about 100 or about 130 mg/m 2 oxaliplatin, which are sufficient for a 6-week treatment. In some embodiments, the kit comprises means for separately retaining the components, such as a container, divided bottle, or divided foil packet. A kit of this disclosure can be used for administration of different dosage forms, for example, oral and parenteral, for administration of the separate compositions at different dosage intervals, or for titration of the separate compositions against one another. Uses of a Therapeutic Combination for Treating Cancer In still another aspect, provided herein are uses of a therapeutic combination for treating cancer (e.g., esophageal cancer) in a human patient, wherein the therapeutic combination comprises: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC) and gastric cancer. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In one embodiment, the cancer is esophageal cancer. In one embodiment, provided herein is use of a therapeutic combination for treating esophageal cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In still other embodiments, provided herein is use of a therapeutic combination for treating cancer in a patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist as disclosed in the present disclosure; (b) a PD-1 antagonist as disclosed in the present disclosure; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In some embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In other embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) 5-fluorouracil represented by Formula (I); and (d) cisplatin represented by Formula (II). In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. In certain embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In other embodiments of various uses provided herein, the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. Thus, in one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) 5-fluorouracil represented by Formula (I) and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) 5-fluorouracil represented by Formula (I) and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) 5-fluorouracil represented by Formula (I) and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating esophageal cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) 5-fluorouracil represented by Formula (I) and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating esophageal cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) 5-fluorouracil represented by Formula (I) and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating esophageal cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) 5-fluorouracil represented by Formula (I) and; (d) cisplatin represented by Formula (II). In still another aspect, provided herein are uses of a therapeutic combination for treating cancer (e.g., TNBC) in a human patient, wherein the therapeutic combination comprises: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC) and gastric cancer. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In one embodiment, the cancer is TNBC. In one embodiment, provided herein is use of a therapeutic combination for treating TNBC in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In still other embodiments, provided herein is use of a therapeutic combination for treating cancer in a patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist as disclosed in the present disclosure; (b) a PD-1 antagonist as disclosed in the present disclosure; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In other embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. In certain embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In other embodiments of various uses provided herein, the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. Thus, in one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment, provided herein is use of a therapeutic combination for treating TNBC in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment, provided herein is use of a therapeutic combination for treating TNBC in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In one specific embodiment, provided herein is use of a therapeutic combination for treating TNBC in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; and (c) paclitaxel or a pharmaceutically acceptable salt thereof. In still another aspect, provided herein are uses of a therapeutic combination for treating cancer (e.g., biliary cancer) in a human patient, wherein the therapeutic combination comprises: (c) a TIGIT antagonist; (d) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC) and gastric cancer. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In one embodiment, the cancer is biliary cancer. In one embodiment, provided herein is use of a therapeutic combination for treating biliary cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In still other embodiments, provided herein is use of a therapeutic combination for treating cancer in a patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist as disclosed in the present disclosure; (b) a PD-1 antagonist as disclosed in the present disclosure; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In some embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In other embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof; and (d) cisplatin represented by Formula (II). In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. In certain embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In other embodiments of various uses provided herein, the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. Thus, in one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating biliary cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating biliary cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and; (d) cisplatin represented by Formula (II). In one specific embodiment, provided herein is use of a therapeutic combination for treating biliary cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) gemcitabine represented by Formula (IV) or pharmaceutically acceptable salt thereof and; (d) cisplatin represented by Formula (II). In still another aspect, provided herein are uses of a therapeutic combination for treating cancer (e.g., biliary cancer) in a human patient, wherein the therapeutic combination comprises: (e) a TIGIT antagonist; (f) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In some embodiments, the cancer is selected from the group consisting of endometrial cancer, hepatocellular carcinoma (HCC), cervical cancer, head and neck cancer (HNSCC), biliary cancer, esophageal cancer, triple negative breast cancer (TNBC) and gastric cancer. In certain embodiments, the cancer is metastatic. In some embodiments, the cancer is relapsed. In other embodiments, the cancer is refractory. In yet other embodiments, the cancer is relapsed and refractory. In one embodiment, the cancer is gastric cancer. In some embodiments, the gastric cancer is advanced or GEJ adenocarcinoma. In some embodiments, the gastric cancer is HER2 negative. In some embodiments, the gastric cancer is previously untreated. In one embodiment, provided herein is use of a therapeutic combination for treating biliary cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist; (b) a PD-1 antagonist; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In still other embodiments, provided herein is use of a therapeutic combination for treating cancer in a patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a TIGIT antagonist as disclosed in the present disclosure; (b) a PD-1 antagonist as disclosed in the present disclosure; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In certain embodiments, the TIGIT antagonist is an anti-human TIGIT monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human TIGIT monoclonal antibody is a human antibody. In other embodiments, the anti-human TIGIT monoclonal antibody is a humanized antibody. In certain embodiments, the PD-1 antagonist is an anti-human PD-1 monoclonal antibody or antigen binding fragment thereof. In some embodiments, the anti-human PD-1 monoclonal antibody is a human antibody. In other embodiments, the anti-human PD-1 monoclonal antibody is a humanized antibody. In certain embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human or humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human or humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In some embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a human anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a human anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (V). In other embodiments, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) a humanized anti-human TIGIT monoclonal antibody or antigen binding fragment thereof; (b) a humanized anti-human PD-1 monoclonal antibody or antigen binding fragment thereof; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is pembrolizumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is nivolumab. In some embodiments of various uses provided herein, the anti-PD-1 antibody (e.g., anti-PD-1 monoclonal antibody) or antigen binding fragment thereof is cemiplimab. In certain embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110. In some embodiments of various uses provided herein, the anti-TIGIT antibody or antigen binding fragment thereof comprises a heavy chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 148 and a light chain variable region comprising the amino acid sequence as set forth in SEQ ID NO: 152. In other embodiments of various uses provided herein, the anti-human TIGIT monoclonal antibody or antigen binding fragment thereof comprises a light chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:294 and a heavy chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO:295. Thus, in one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment, provided herein is use of a therapeutic combination for treating cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment, provided herein is use of a therapeutic combination for treating gastric cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) pembrolizumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment, provided herein is use of a therapeutic combination for treating gastric cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) nivolumab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). In one specific embodiment, provided herein is use of a therapeutic combination for treating gastric cancer in a human patient in need thereof, wherein the therapeutic combination comprises an effective amount of: (a) anti-TIGIT antibody or antigen binding fragment thereof comprising three light chain CDRs: CDRL1 comprising the amino acid sequence as set forth in SEQ ID NO: 111, CDRL2 comprising the amino acid sequence as set forth in SEQ ID NO: 112, and CDRL3 comprising the amino acid sequence as set forth in SEQ ID NO: 113, and three heavy chain CDRs: CDRH1 comprising the amino acid sequence as set forth in SEQ ID NO: 108, CDRH2 comprising the amino acid sequence as set forth in SEQ ID NO: 154, and CDRH3 comprising the amino acid sequence as set forth in SEQ ID NO: 110; (b) cemiplimab; (c) capecitabine represented by Formula (V) or a pharmaceutically acceptable salt thereof; and (d) oxaliplatin represented by Formula (VI). A number of embodiments of the invention have been described. It will be understood that various modifications may be made without departing from the spirit and scope of the invention. It will be further understood that each embodiment may be combined with one or more other embodiments, to the extent that such a combination is consistent with the description of the embodiments. General Methods Standard methods in molecular biology are described Sambrook, Fritsch and Maniatis (1982 & 19892 nd Edition, 20013 rd Edition) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Sambrook and Russell (2001) Molecular Cloning, 3 rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Wu (1993) Recombinant DNA, Vol.217, Academic Press, San Diego, CA). Standard methods also appear in Ausbel, et al. (2001) Current Protocols in Molecular Biology, Vols.1- 4, John Wiley and Sons, Inc. New York, NY, which describes cloning in bacterial cells and DNA mutagenesis (Vol.1), cloning in mammalian cells and yeast (Vol.2), glycoconjugates and protein expression (Vol.3), and bioinformatics (Vol.4). Methods for protein purification including immunoprecipitation, chromatography, electrophoresis, centrifugation, and crystallization are described (Coligan, et al. (2000) Current Protocols in Protein Science, Vol.1, John Wiley and Sons, Inc., New York). Chemical analysis, chemical modification, post-translational modification, production of fusion proteins, glycosylation of proteins are described (see, e.g., Coligan, et al. (2000) Current Protocols in Protein Science, Vol.2, John Wiley and Sons, Inc., New York; Ausubel, et al. (2001) Current Protocols in Molecular Biology, Vol.3, John Wiley and Sons, Inc., NY, NY, pp.16.0.5-16.22.17; Sigma-Aldrich, Co. (2001) Products for Life Science Research, St. Louis, MO; pp.45-89; Amersham Pharmacia Biotech (2001) BioDirectory, Piscataway, N.J., pp.384-391). Production, purification, and fragmentation of polyclonal and monoclonal antibodies are described (Coligan, et al. (2001) Current Protocols in Immunology, Vol.1, John Wiley and Sons, Inc., New York; Harlow and Lane (1999) Using Antibodies, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Harlow and Lane, supra). Standard techniques for characterizing ligand/receptor interactions are available (see, e.g., Coligan, et al. (2001) Current Protocols in Immunology, Vol.4, John Wiley, Inc., New York). Monoclonal, polyclonal, and humanized antibodies can be prepared (see, e.g., Sheperd and Dean (eds.) (2000) Monoclonal Antibodies, Oxford Univ. Press, New York, NY; Kontermann and Dubel (eds.) (2001) Antibody Engineering, Springer-Verlag, New York; Harlow and Lane (1988) Antibodies A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, pp.139-243; Carpenter, et al. (2000) J. Immunol.165:6205; He, et al. (1998) J. Immunol.160:1029; Tang et al. (1999) J. Biol. Chem.274:27371-27378; Baca et al. (1997) J. Biol. Chem.272:10678-10684; Chothia et al. (1989) Nature 342:877- 883; Foote and Winter (1992) J. Mol. Biol.224:487-499; U.S. Pat. No.6,329,511). An alternative to humanization is to use human antibody libraries displayed on phage or human antibody libraries in transgenic mice (Vaughan et al. (1996) Nature Biotechnol. 14:309-314; Barbas (1995) Nature Medicine 1:837-839; Mendez et al. (1997) Nature Genetics 15:146-156; Hoogenboom and Chames (2000) Immunol. Today 21:371-377; Barbas et al. (2001) Phage Display: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York; Kay et al. (1996) Phage Display of Peptides and Proteins: A Laboratory Manual, Academic Press, San Diego, CA; de Bruin et al. (1999) Nature Biotechnol.17:397-399). Purification of antigen is not necessary for the generation of antibodies. Animals can be immunized with cells bearing the antigen of interest. Splenocytes can then be isolated from the immunized animals, and the splenocytes can fused with a myeloma cell line to produce a hybridoma (see, e.g., Meyaard et al. (1997) Immunity 7:283-290; Wright et al. (2000) Immunity 13:233-242; Preston et al., supra; Kaithamana et al. (1999) J. Immunol. 163:5157-5164). Antibodies can be conjugated, e.g., to small drug molecules, enzymes, liposomes, polyethylene glycol (PEG). Antibodies are useful for therapeutic, diagnostic, kit or other purposes, and include antibodies coupled, e.g., to dyes, radioisotopes, enzymes, or metals, e.g., colloidal gold (see, e.g., Le Doussal et al. (1991) J. Immunol.146:169-175; Gibellini et al. (1998) J. Immunol.160:3891-3898; Hsing and Bishop (1999) J. Immunol.162:2804-2811; Everts et al. (2002) J. Immunol.168:883-889). Methods for flow cytometry, including fluorescence activated cell sorting (FACS), are available (see, e.g., Owens, et al. (1994) Flow Cytometry Principles for Clinical Laboratory Practice, John Wiley and Sons, Hoboken, NJ; Givan (2001) Flow Cytometry, 2 nd ed.; Wiley- Liss, Hoboken, NJ; Shapiro (2003) Practical Flow Cytometry, John Wiley and Sons, Hoboken, NJ). Fluorescent reagents suitable for modifying nucleic acids, including nucleic acid primers and probes, polypeptides, and antibodies, for use, e.g., as diagnostic reagents, are available (Molecular Probesy (2003) Catalogue, Molecular Probes, Inc., Eugene, OR; Sigma-Aldrich (2003) Catalogue, St. Louis, MO). Standard methods of histology of the immune system are described (see, e.g., Muller- Harmelink (ed.) (1986) Human Thymus: Histopathology and Pathology, Springer Verlag, New York, NY; Hiatt, et al. (2000) Color Atlas of Histology, Lippincott, Williams, and Wilkins, Phila, PA; Louis, et al. (2002) Basic Histology: Text and Atlas, McGraw-Hill, New York, NY). Software packages and databases for determining, e.g., antigenic fragments, leader sequences, protein folding, functional domains, glycosylation sites, and sequence alignments, are available (see, e.g., GenBank, Vector NTI® Suite (Informax, Inc, Bethesda, MD); GCG Wisconsin Package (Accelrys, Inc., San Diego, CA); DeCypher® (TimeLogic Corp., Crystal Bay, Nevada); Menne, et al. (2000) Bioinformatics 16: 741-742; Menne, et al. (2000) Bioinformatics Applications Note 16:741-742; Wren, et al. (2002) Comput. Methods Programs Biomed.68:177-181; von Heijne (1983) Eur. J. Biochem.133:17-21; von Heijne (1986) Nucleic Acids Res.14:4683-4690). Analytical Methods Analytical methods suitable for evaluating the product stability include size exclusion chromatography (SEC), dynamic light scattering test (DLS), differential scanning calorimetery (DSC), iso-asp quantification, potency, UV at 340 nm, UV spectroscopy, and Fourier-transform infrared spectroscopy (FTIR). SEC (J. Pharm. Scien., 83:1645-1650, (1994); Pharm. Res., 11:485 (1994); J. Pharm. Bio. Anal., 15:1928 (1997); J. Pharm. Bio. Anal., 14:1133-1140 (1986)) measures percent monomer in the product and gives information of the amount of soluble aggregates. DSC (Pharm. Res., 15:200 (1998); Pharm. Res., 9:109 (1982)) gives information of protein denaturation temperature and glass transition temperature. DLS (American Lab., November (1991)) measures mean diffusion coefficient, and gives information of the amount of soluble and insoluble aggregates. UV at 340 nm measures scattered light intensity at 340 nm and gives information about the amounts of soluble and insoluble aggregates. UV spectroscopy measures absorbance at 278 nm and gives information of protein concentration. FTIR (Eur. J. Pharm. Biopharm., 45:231 (1998); Pharm. Res., 12:1250 (1995); J. Pharm. Scien., 85:1290 (1996); J. Pharm. Scien., 87:1069 (1998)) measures IR spectrum in the amide one region, and gives information of protein secondary structure. The iso-asp content in the samples is measured using the Isoquant Isoaspartate Detection System (Promega). The kit uses the enzyme Protein Isoaspartyl Methyltransferase (PIMT) to specifically detect the presence of isoaspartic acid residues in a target protein. PIMT catalyzes the transfer of a methyl group from S-adenosyl-L-methionine to isoaspartic acid at the .alpha.-carboxyl position, generating S-adenosyl-L-homocysteine (SAH) in the process. This is a relatively small molecule, and can usually be isolated and quantitated by reverse phase HPLC using the SAH HPLC standards provided in the kit. The potency or bioidentity of an antibody can be measured by its ability to bind to its antigen. The specific binding of an antibody to its antigen can be quantitated by any method known to those skilled in the art, for example, an immunoassay, such as ELISA (enzyme- linked immunosorbant assay). All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing methodologies and materials that might be used in connection with the present invention. Having described different embodiments of the invention herein with reference to the accompanying drawings, it is to be understood that the invention is not limited to those precise embodiments, and that various changes and modifications may be effected therein by one skilled in the art without departing from the scope or spirit of the invention as defined in the appended claims. EXAMPLES The examples in this section are offered by way of illustration, and not by way of limitation. Example 1: Clinical Trial of Administering a Combination of an Anti-TIGIT Antibody and an Anti-PD-1 Antibody with One or More Chemotherapeutic Agents in Esophageal Cancer, TNBC or Biliary Cancer Patients This study is a multicenter, open-label phase 2 study of an anti-TIGIT antagonist and an anti-PD-1 antagonist with or without one or more chemotherapeutic agents for the treatment of esophageal cancer or TNBC. For esophageal cancer, approximately 40 participants with locally advanced unresectable or metastatic adenocarcinoma or squamous cell carcinoma of the esophagus (ESCC) or advanced/metastatic Siewert type 1 adenocarcinoma of the gastroesophageal junction (GEJ) will be allocated by nonrandom assignment to Composition A (coformulation of pembrolizumab and vibostolimab; vibostolimab comprising heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 295 and light chain comprising the amino acid sequence as set forth in SEQ ID NO: 294) + 5-fluorouracil (5-FU) + cisplatin. A total of 40 participants will be allocated to receive Composition A plus the RP2D for 5- fluorouracil/cisplatin. For TNBC, approximately 40 participants with locally recurrent unresectable or metastatic TNBC, will be allocated by nonrandom assignment to Composition A + paclitaxel. A total of 40 participants will be allocated to receive Composition A plus the RP2D for paclitaxel. For biliary cancer, approximately 40 participants with locally recurrent unresectable or metastatic biliary cancer will be allocated by nonrandom assignment to Composition A. Approximately 10 to 14 participants with locally recurrent unresectable or metastatic biliary cancer will be allocated by nonrandom assignment to Composition A plus gemcitabine/cisplatin. A total of 10 participants will be allocated to receive Composition A plus the RP2D for gemcitabine/cisplatin. For gastric cancer, approximately 40 participants with previously untreated, HER2 negative, advanced gastric or GEJ adenocarcinoma will be allocated by nonrandom assignment to Composition A in combination with capecitabine/oxaliplatin. Objective and Endpoints A listing of objectives and endpoints for the clinical trial substudy are shown in the table below. Table 9. Objectives and endpoints for the clinical trial Objectives Endpoints Objectives Endpoints Objectives Endpoints x Esophageal cancer A modified Toxicity Probability Interval (mTPI) design with a target dose-limiting toxicity (DLT) rate of 30% will be used to identify the preliminary recommended Phase 2 dose (RP2D) of 5-FU/cisplatin to use in combination with Composition A for the treatment of participants with esophageal cancer. If de-escalation is called for by mTPI, the dose of 5- FU/cisplatin will be reduced while the dose of Composition A will remain fixed. There are 2 predetermined dose levels of 5-FU/cisplatin that may be explored in the mTPI design. If the mTPI calls for an additional de-escalation, enrollment will be closed. Treatment with Composition A will continue for a total of 35 administrations (approximately 2 years) or until any of the discontinuation criteria are met. Treatment with the RP2D of 5-FU and cisplatin will continue until any of the discontinuation criteria are met. There are 2 predetermined dose levels that may be explored in the mTPI design (Table 10). If the mTPI calls for an additional de-escalation, enrollment will be put on hold while the Sponsor evaluates the observed DLTs. x TNBC An mTPI design with a target DLT rate of 30% will be used to identify the preliminary RP2D of paclitaxel to use in combination with Composition A for the treatment of participants with TNBC. If de-escalation is called for by mTPI, the dose of paclitaxel will be reduced while the dose of Composition A will remain fixed. There are 2 predetermined dose levels of paclitaxel that may be explored in the mTPI design (Table 10). If the mTPI calls for an additional de-escalation, enrollment into Cohort F will be closed. Treatment with Composition A will continue for a total of 35 administrations (approximately 2 years) or until any of the discontinuation criteria are met. Treatment with the RP2D of paclitaxel will continue until any of the discontinuation criteria are met. x Biliary Cancer A total of 10 participants will be allocated to receive Composition A plus the RP2D for gemcitabine/cisplatin (Table 10). An mTPI design with a target DLT rate of 30% will be used to identify the preliminary RP2D of gemcitabine/cisplatin to use in combination with Composition A for the treatment of participants with biliary cancer. If de-escalation is called for by mTPI, the dose of gemcitabine/cisplatin will be reduced while the dose of Composition A will remain fixed. There are 2 predetermined dose levels of gemcitabine/cisplatin that may be explored in the mTPI design. x Gastric cancer A mTPI design with a target DLT rate of 30% will be used to identify the preliminary RP2D of capecitabine/oxaliplatin to use in combination with Composition A for the treatment of participants with gastric cancer. If de-escalation is called for by mTPI, the dose of capecitabine/oxaliplatin will be reduced while the dose of Composition A will remain fixed. There are 2 predetermined dose levels of capecitabine/oxaliplatin that may be explored in the mTPI design (Table 10). Treatment with MK‑7684A will continue for a total of 35 administrations (approximately 2 years) or until any of the discontinuation criteria are met. Table 10. Predetermined Dose Levels of 5-FU, Cisplatin, Paclitaxel, Gemcitabine, Capecitabine and Oxaliplatin for Determining the Recommended Phase 2 Dose of Chemotherapy Indication Study Intervention Dose Level 0 Dose Level –1 Participants are planned at the starting doses of chemotherapeutic agents as listed in Table 10. However, depending on the accrual rate, the completion of DLT evaluation period and number of DLTs observed, fewer participants may be enrolled at the starting dose. Participants will be closely followed for DLTs for the first cycle after the first dose of study intervention (the DLT evaluation period). The dose will be evaluated on an ongoing basis as participants complete the DLT evaluation period. In Table 11, the columns indicate the numbers of participants treated at the current dose level, and the rows indicate the numbers of participants experiencing a DLT. The entries of the table are the dose-finding decisions: E, S, D, and DU represent escalating the dose, staying at the same dose, de-escalating the dose, and excluding the dose from the study due to unacceptable toxicity, respectively. For example, if 2 out of 3 participants at this dose level develop a DLT, the dose will be de-escalated to the next lower dose level but may be re-escalated at a later time if the lower dose is well tolerated. If 3 out of 3 participants develop a DLT, this indicates an unacceptable toxicity at this dose. The dose should be de-escalated, if allowed per-protocol, and the current dose will not be explored further. Table 11. Dose-finding Rules per mTPI Design (Target Toxicity Rate up to 30%) Number of Participants Evaluable for DLT at Current Dose Number of participants 3 4 5 6 7 8 9 10 p p have completed enrollment at the starting dose, the enrollment at this dose level will be halted and a new set of participants will be enrolled and treated at the next lower dose level. This process will continue, with up to 10 DLT-evaluable participants enrolled at each dose explored, until 10 participants have been enrolled at any of the tested doses and a dosing decision of “S” (for staying at current dose) is made for the 10th participant evaluated. If, at any point, the dosing decision is “E” after evaluation of all enrolled patients at a dose, the dose may be escalated to a higher predetermined dose provided that the higher dose has not previously been discontinued due to a “DU” decision. A “D” or “DU” decision at the lowest dose level will stop the evaluation of the combination under study. An “E” decision at the highest dose level will result in staying at that level. During dose-finding, it may be acceptable to de-escalate or escalate to an intermediate dose that was not predefined and not previously studied if evaluation of toxicity at such a dose is desired. The maximum dose/exposure allowed in this study is 200 mg vibostolimab / 200 mg pembrolizumab for up to 2 years (35 treatment cycles) of initial treatment/first course. The maximum dose/exposure of 5-FU allowed is 4000 mg/m 2 /cycle (21-day cycle) for up to 2 years (35 treatment cycles) of initial treatment. The maximum dose/exposure of cisplatin allowed is 80 mg/m 2 for up to 6 cycles. Paclitaxel will be administered as per the approved product label and applicable institutional standards. Based on global clinical practice patterns, gemcitabine at 1000 mg/m 2 on Day 1 and Day 8 of a 21-Day cycle will be administered until PD or unacceptable toxicity. Cisplatin at 25 mg/m 2 will be administered on Day 1 and Day 8 of a 21-Day cycle for a maximum of 8 cycles. The capecitabine/oxaliplatin dose regimen will be oxaliplatin (130 mg/m 2 IV) on Day 1 of each treatment cycle (Q3W) plus capecitabine (1000 mg/m 2 orally twice daily) on Days 1 to 14 of each treatment cycle (Q3W). Duration of oxaliplatin treatment may be capped at 6 cycles as per local country guidelines. Dose modifications in response to treatment-related AEs are permitted to keep the participant on study medication, when appropriate. In general, Composition A, 5-FU, cisplatin, paclitaxel, and/or gemcitabine may be interrupted together or separately at the Investigator’s discretion to determine which treatment component is the source for a given toxicity. Action may be taken independently for each component of study treatment (e.g., chemotherapy should be discontinued for prohibitive toxicity that is chemotherapy-related, but Composition A may continue if well tolerated). chemotherapy may be interrupted, dose reduced or discontinued. Composition A may only be interrupted or discontinued. AEs associated with Composition A exposure may represent an immune-related response. These immune-related AEs (irAEs) may occur shortly after the first dose or several months after the last dose of pembrolizumab monotherapy, coformulation, or immune- oncology (IO) combination treatment and may affect more than one body system simultaneously. Therefore, early recognition and initiation of treatment is critical to reduce complications. Based on existing clinical study data, most irAEs were reversible and could be managed with interruptions of pembrolizumab monotherapy, coformulation, or IO combination administration of corticosteroids and/or other supportive care. For suspected irAEs, ensure adequate evaluation to confirm etiology or exclude other causes. Additional procedures or tests such as bronchoscopy, endoscopy, skin biopsy may be included as part of the evaluation. Table 12. Dose Modification and Toxicity Management Guidelines for Immune-related Adverse Events Associated with Pembrolizumab Monotherapy, Coformulations or IO Combinations General instructions: 1. Severe and life-threatening irAEs should be treated with IV corticosteroids followed by oral steroids. Action With Pembrolizum Action With Pembrolizum capecitabine, or oxaliplatin) for more than 6 weeks will require Sponsor approval before the treatment can be resumed. For participants experiencing an AE requiring dose modification of study intervention, the next treatment period with this(these) drug(s) may be delayed, if the scheduled off-drug periods are not adequate to allow for recovery of the AE to ≤Grade 1 or the baseline status of the participant. The investigator may attribute each toxicity event to 5-FU, cisplatin, or Composition A and use a stepwise approach to dose reduction of 5-FU and cisplatin (Table 10). Participants may have a maximum of 2 dose reductions to each of the components throughout the course of the study. Once the dose has been reduced, it may not be re-escalated. If a participant experiences several toxicities and there are conflicting recommendations, follow the most conservative dose adjustment recommended (i.e., dose reduction appropriate to the most severe toxicity). Participants who require a third dose modification to any component will have that agent discontinued. Reduction of one chemotherapy agent and not the other is appropriate if, in the opinion of the investigator, the toxicity is clearly related to one of the agents. If the toxicity is related to the combination of both chemotherapy agents, both drugs should be reduced according to recommended dose modifications. If the toxicity is related to the combination of all study intervention administered, chemotherapy should be reduced, interrupted, or discontinued, and Composition A should be interrupted or discontinued according to the recommended dose modifications. Participants may discontinue either 5-FU or cisplatin and continue receiving the other component in combination with Composition A. Participants may discontinue both 5-FU and cisplatin and continue receiving Composition A. Dose modification criteria for 5-FU and cisplatin are provided in Error! Reference source not found. and Error! Reference source not found., respectively. Table 13. Dose Modification Guidelines for 5-FU-related Adverse Events Hold 5-FU Timing for Dose for Discontinue Treatment Restarting 5-FU Restarting 5-FU e e e Hold 5-FU Timing for Dose for Treatment Restarting 5-FU Restarting 5-FU Discontinue e e e e . p Timing for Dose for Hold Restarting Restarting Cisplatin Timing for Dose for Hold Restarting Restarting Cis latin 4 e ves ga o ay a ue eac o c y eve o pac a e o o pos o a use a stepwise approach to dose reduction of paclitaxel. Participants may have a maximum of 1 dose reduction to paclitaxel throughout the course of the study. Once the dose has been reduced, it may not be re-escalated. If a participant experiences several toxicities and there are conflicting recommendations, follow the most conservative dose adjustment recommended (i.e., dose reduction appropriate to the most severe toxicity). If the toxicity is related to the combination of all study intervention administered, paclitaxel should be reduced, interrupted, or discontinued, and Composition A should be interrupted or discontinued according to the recommended dose modifications. Participants may discontinue paclitaxel and continue receiving Composition A. If paclitaxel is discontinued, the participants does not need to come to the clinic on dates when only the chemotherapy would have been administered. Dose modification criteria for paclitaxel are provided in Table 15. Table 15. Dose Modification Guidelines for Paclitaxel-related Adverse Events T oxicities Grade or a ctual value Paclitaxel Toxicities Grade or a ctual value Paclitaxel significant renal or hepatic impairment as there is insufficient information from clinical studies to allow clear dose recommendation for these patient populations. For significant renal impairment defined as creatinine clearance (CrCl) <30 mL/min, consider dose reduction and discuss with Sponsor. For serum bilirubin >1.6 mg/dL, consider dose reduction of gemcitabine to 800 mg/m 2 . Monitor carefully and discontinue gemcitabine if any drug-related worsening occurs. Gemcitabine should be discontinued at the first signs of any evidence of microangiopathic hemolytic anemia, such as rapidly falling hemoglobin with concomitant thrombocytopenia, elevation of serum bilirubin, serum creatinine, blood urea nitrogen, or LDH, which may indicate development of hemolytic uremic syndrome. Renal failure may not be reversible, even with discontinuation of therapy, and dialysis may be required. Maximal dose reduction permitted is up to 40% of starting dose of cisplatin (10 mg/m 2 ) and gemcitabine (400 mg/m 2 ). Doses below this level are not considered therapeutic and the participant will be discontinued from the related chemotherapy. Dose modification criteria for gemcitabine/cisplatin are provided in Table 16. Table 16 Dose Modification Guidelines for Gemcitabine/Cisplatin-related Adverse Events Timing for C ategory Toxicity Grade Gem Cis d ose dose Restarting Study Subsequent Dose Int r nti n Timing for Category Toxicity Grade Gem Cis d ose dose Restarting Study Subsequent Dose I i Dose modifications for capecitabine/oxaliplatin combination, oxaliplatin, and capecitabine are detailed in Table 1, Table 2, and Error! Reference source not found., respectively. In participants with unrecognized dihydropyrimidine dehydrogenase deficiency, acute, life-threatening toxicity may occur when participants are treated with capecitabine. It is recommended that dihydropyrimidine dehydrogenase testing be performed as appropriate per local guidelines Table 1 Dose Modification Guidelines for CAPOX Drug-related Adverse Events Dose Level 0 Dose Level -1 Dose Level -2 Dose Level -3 Dose Level -4 Table 2 Dose Modification Guidelines for Oxaliplatin Drug-related Adverse Events Hold Timing for Dose for Oxaliplatin Restarting Restarting Table 3 Dose Modification Guidelines for Capecitabine Drug-related Adverse Events Hold Capecitabine Timing for Restarting Dose for Restarting T C i bi C i bi Di i i f Composition A co-formulation administration Composition A co-formulation will be administered using a 30-minute IV infusion every 3 weeks. Sites should make every effort to target infusion timing to be as close to 30 minutes as possible. However, given the variability of infusion pumps from site to site, a window of -5 minutes and +10 minutes is permitted (i.e., infusion time is 25 to 40 minutes). 5-Fluorouracil + cisplatin Administration 5-FU will be administered as a continuous IV infusion of 800 mg/m 2 /day on each of Days 1 to 5 Q3W according to approved prescribing information in each country/region or per local standard for 5-FU administration duration as long as total dose of 4000 mg/m 2 per 3-week cycle is followed (e.g., 1000 mg/m 2 /day on Days 1 to 4 Q3W is also acceptable).5- FU will be administered after Composition A and cisplatin administration. Treatment with 5- FU may follow 1 to 2 days after Composition A (e.g., Day 2 or Day 3) as needed per local standard of care. Duration of 5-FU treatment will not exceed 35 cycles. Cisplatin 80 mg/m 2 will be administered as a 60- or 120-minute IV infusion (or per site’s standard practice) Q3W on Day 1 of each treatment cycle after Composition A administration according to approved prescribing information in each country/region or local institutional guidelines. Duration of cisplatin treatment will be capped at 6 doses; cisplatin dosing may occur after Cycle 6 if cisplatin was withheld for 1 or more cycles during Cycles 2 to 6. Treatment with cisplatin may follow 1 to 2 days after Composition A (e.g., Day 2 or Day 3) as needed per local standard of care. Paclitaxel Administration Paclitaxel will be administered as a dose of 90 mg/m 2 Q3W on Day 1, 8, and 15 of each 28-day treatment cycle according to approved prescribing information in each country/region or local institutional guidelines. When Composition A and paclitaxel are administered on the same day, paclitaxel will be administered after Composition A administration. Gemcitabine + Cisplatin Administration Cisplatin will be administered as an IV infusion on Day 1 and Day 8 of a 3 week cycle and should be immediately preceded and followed by hydration procedures and administered according to local guidelines/product label procedures. Cisplatin may be administered on Day 2, if required per local guidelines; however, Day 1 is the preferred day for intervention administration. Gemcitabine will be administered as an IV infusion on Days 1 and 8 of a 3 week cycle, according to local guidelines and product label procedures. Premedication with antiemetic therapy can be given prior to cisplatin and gemcitabine administration. Consider following Multinational Association of Supportive Care in Cancer (MASCC) guidelines (http://www.mascc.org/antiemetic-guidelines) and, including a 5-HT3 receptor antagonist, dexamethasone (or equivalent) and aprepitant (or equivalent) as per the MASCC guidelines. Based on data showing potential impact of steroid use on efficacy of PD-1 and PD-L1 blockade, systemic steroid use as anti-nausea medication should be limited as deemed safe by the investigator. Capecitabine + Oxaliplatin Administration Oxaliplatin 130 mg/m 2 will be administered as a 60- to 120 minute IV infusion or per the site’s standard practice on Day 1 of each treatment cycle. Capecitabine will be administered orally as a 1000 mg/m 2 dose twice daily from Day 1 to Day 14 of each treatment cycle. The evening dose of capecitabine should be taken approximately 12 hours after the morning dose. Capecitabine should be taken with food, or within 30 minutes after food/meal, with approximately 200 mL of water. Sites may follow local practice patterns regarding calculation of capecitabine milligram dose. Refer to the product label for additional guidance on administration procedures for capecitabine. If the participant is enrolled later in the day, it is acceptable for the first dose to be taken on Day 1, and twice daily dosing can continue on Days 2 to 14; the final dose will be taken in the morning of Day 15. Investigators are advised to counsel participants assigned to receive capecitabine about the risk of photosensitivity and to take sun protection measures accordingly. Participants receiving capecitabine should be carefully monitored for ophthalmological complications and skin reactions. Efficacy Endpoint This study will use objective response rate as the primary efficacy endpoint. Objective response rate is defined as the proportion of participants who have best response as CR or PR. Responses are based on blinded independent central review (BICR) using RECIST 1.1 (modified to follow a maximum of 10 target lesions and a maximum of 5 target lesions per organ). Objective response rate is an appropriate endpoint to evaluate the antitumor activity of investigational treatment arms. For the randomized cohort, ORR is defined as the percentage of participants who achieve a confirmed CR or PR per RECIST 1.1 as assessed by BICR. For the single-arm cohorts, ORR is defined as the percentage of participants who achieve a confirmed CR or PR per RECIST 1.1 as assessed by investigator. For the randomized cohort, PFS is defined as the time from randomization to the first documented disease progression per RECIST 1.1 by BICR or death due to any cause, whichever occurs first. This study will use PFS as a secondary efficacy endpoint. Progression-free survival is defined as the time from date of randomization/allocation until the first date of disease progression or death from any cause, whichever comes first, based on RECIST 1.1 criteria as assessed by BICR. For the single-arm cohorts, PFS is defined as the time from the first dose of study intervention to the first documented disease progression per RECIST 1.1 by investigator or death due to any cause, whichever occurs first. Additionally, this study will use OS as an exploratory efficacy endpoint. Overall survival in this study is defined as the time from date of randomization/allocation to death from any cause. For the randomized cohort, OS is defined as the time from randomization to death due to any cause. For the single-arm cohorts, OS is defined as the time from the first dose of study intervention to death due to any cause. Inclusion criteria All participants will be given a participant identification card identifying them as participants in a research study. The card will contain study-site contact information (including direct telephone numbers) to be used in the event of an emergency. The investigator or qualified designee will provide the participant with a participant identification card immediately after the participant provides documented informed consent. At the time of intervention allocation/randomization, site personnel will add the treatment/randomization number to the participant identification card. The participant ID card also contains contact information for the emergency unblinding call center so that a health care provider can obtain information about study intervention in emergency situations where the investigator is not available. A medical history will be obtained by the investigator or qualified designee. The medical history will collect all active conditions and any condition diagnosed within the prior 10 years that the investigator considers to be clinically important. Details regarding the disease for which the participant has enrolled in this study will be recorded separately and not listed as medical history. If a medical condition is diagnosed at the time of screening due to the physical examination, laboratory tests, radiologic assessment, other assessment, and/or a combination of these evaluations, the medical condition is to be recorded as a baseline condition along with the participant’s other medical history unless due to any protocol-specified intervention (e.g., procedure, washout or run-in treatment including placebo run-in). Comprehensive details regarding the participant’s cancer history will be recorded separately and not listed as medical history. These details include, but are not limited to, stage at diagnosis, histopathology, location(s) of tumor burden, investigator-determined tumor size per RECIST 1.1, and all prior treatment (including prior radiation, prior chemotherapy, and prior surgery). The investigator or qualified designee will review before medication use, including any protocol-specified washout requirement, and record prior medication taken by the participant within 28 days before the first dose of study intervention. The investigator or qualified designee will record medication, if any, taken by the participant during the study. All consented participants will be given a unique screening number that will be used to identify the participant for all procedures that occur before randomization/intervention allocation. Each participant will be assigned only 1 screening number. Screening numbers must not be reused for different participants. Any participant who is screened multiple times will retain the original screening number assigned at the initial screening Visit. All eligible participants will be allocated, by nonrandom assignment, and will receive a treatment/randomization number. The treatment/randomization number identifies the participant for all procedures occurring after treatment allocation/randomization. Once a treatment/randomization number is assigned to a participant, it can never be reassigned to another participant. A single participant cannot be assigned more than 1 treatment/randomization number. Exclusion criteria The participant must be excluded from the study if the participant: 1. Has a history of a second malignancy, unless potentially curative treatment has been completed with no evidence of malignancy for 3 years. The time requirement does not apply to participants who underwent successful definitive resection of basal cell carcinoma of the skin, squamous cell carcinoma of the skin, in-situ cervical cancer, or other in-situ cancers. For gastric cancer, has a known additional malignancy that is progressing or has required active treatment within the past 5 years. 2. Human immunodeficiency virus (HIV)-infected participants with a history of Kaposi’s sarcoma and/or Multicentric Castleman’s Disease. 3. Has had previous systemic therapy for advanced (metastatic) or unresectable (locally advanced) biliary tract cancer (intra-or extra hepatic cholangiocarcinoma or gallbladder cancer), with the exception of neoadjuvant/adjuvant therapy which is allowed. Neoadjuvant/adjuvant therapy should have been completed at least 6 months prior to diagnosis of advanced and/or unresectable disease, and participants should not have received gemcitabine and/or cisplatin in the neoadjuvant/adjuvant setting. Participants who received prior neoadjuvant/adjuvant therapy with R2 postoperative pathology of the oncologic resection are excluded. 4. Has squamous cell or undifferentiated gastric cancer. 5. Has preexisting peripheral neuropathy >Grade 1. 6. Has received prior therapy with an anti-PD-1, anti-PD-L1, anti-PD-L2, or anti- TIGIT agent. 7. Has received prior systemic anticancer therapy including investigational agents within 4 weeks before randomization/allocation. 8. Has received prior radiotherapy within 2 weeks of start of study intervention or radiation-related toxicities requiring corticosteroids. Two weeks or fewer of palliative radiotherapy for non- central nervous system (CNS) disease, with a 1-week washout, is permitted. 9. Has received a live or live-attenuated vaccine within 30 days before the first dose of study intervention. Administration of killed vaccines are allowed. 10. Has received prior anticancer therapy (e.g., transarterial chemoembolization (TACE), palliative surgery) for advanced unresectable biliary tract cancer (intra-or extra hepatic cholangiocarcinoma or gallbladder cancer), including investigational agents within 4 weeks prior to allocation. 11. Has had previous therapy for locally advanced, unresectable or metastatic gastric/GEJ cancer. Participants may have received prior neoadjuvant and/or adjuvant therapy as long as it was completed at least 6 months prior to randomization. 12. Has received prior therapy with an agent directed to a stimulatory or coinhibitory T-cell receptor (e.g., CTLA-4, OX 40, CD137). 13. Has received an investigational agent or has used an investigational device within 4 prior to study intervention administration. 14. Has a diagnosis of immunodeficiency or is receiving chronic systemic steroid therapy (dosing exceeding 10 mg daily of prednisone equivalent) or any other form of immunosuppressive therapy within 7 days before the first dose of study medication. 15. Has known active CNS metastases and/or carcinomatous meningitis. Participants with previously treated brain metastases may participate provided they are radiologically stable, (i.e., without evidence of progression) for at least 4 weeks by repeat imaging (The repeat imaging should be performed during study screening), clinically stable, and without requirement of steroid treatment for at least 14 days before the first dose of study intervention. Participants with known untreated, asymptomatic brain metastases (i.e., no neurological symptoms, no requirement for corticosteroids, no or minimal surrounding edema, and no lesion >1.5 cm) may participate but will require regular imaging of the brain as a site of disease. 16. Known severe hypersensitivity (≥Grade 3) to Composition A or chemotherapy for the cohort to which the participant is allocated (including dihydropyrimidine dehydrogenase deficiency for 5-FU and capecitabine, respectively) and/or any of their excipients. 17. Has an active autoimmune disease that has required systemic treatment in past 2 years except replacement therapy (e.g., thyroxine, insulin, or physiologic corticosteroid). 18. Has a history of (noninfectious) pneumonitis/interstitial lung disease that required steroids or has current pneumonitis/interstitial lung disease. 19. Has an active infection requiring systemic therapy. 20. Has a history or current evidence of any condition, therapy, or laboratory abnormality that might confound the results of the study, interfere with the participant's participation for the full duration of the study, or is not in the best interest of the participant to participate, in the opinion of the treating investigator. 21. Has a known psychiatric or substance abuse disorder that would interfere with the participant's ability to cooperate with the requirements of the study. 22. Has present or progressive accumulation of pleural, ascitic, or pericardial fluid requiring drainage or diuretic drugs within 2 weeks before randomization/allocation. 23. Has concurrent active hepatitis B (defined as hepatitis B surface antigen (HBsAg) positive and /or detectable hepatitis B virus (HBV) DNA) and hepatitis C virus (defined as anti- hepatitis C virus (HCV) Ab positive and detectable HCV RNA) infection. Hepatitis B and C screening tests are not required unless: known history of HBV and HCV infection, or as mandated by local health authority. 24. For gastric cancer, has a known history of active tuberculosis (TB; Bacillus tuberculosis). 25. For gastric cancer, participants with hypokalemia (serum potassium less than the lower limit of normal). 26. For gastric cancer, participants with hypomagnesemia (serum magnesium less than the lower limit of normal). 27. For gastric cancer, participants with hypocalcemia (serum calcium less than the lower limit of normal). 28. Participants who have not adequately recovered from major surgery or have ongoing surgical complications. 29. Participant, in the judgment of the investigator, is unlikely to comply with the study procedures, restrictions, and requirements of the study. 30. Has had an allogenic tissue/solid organ transplant. 31. For participants who will receive paclitaxel, has a history of class II-IV congestive heart failure or myocardial infarction within 6 months of allocation. Dose Limiting Toxicity All toxicities will be graded using National Cancer Institute (NCI) CTCAE Version 5.0 based on the investigator assessment. The DLT window of observation will be during Cycle 1 for each dose level. The occurrence of any of the following toxicities during Cycle 1 of each dose level will be considered a DLT, if assessed by the investigator to be related to study intervention administration: a. Grade 4 nonhematologic toxicity (not laboratory). b. Grade 4 hematologic toxicity lasting ≥7 days, except thrombocytopenia: • Grade 4 thrombocytopenia of any duration • Grade 3 thrombocytopenia associated with clinically significant bleeding c. Any nonhematologic AE ≥Grade 3 in severity should be considered a DLT, with the following exceptions: Grade 3 fatigue lasting ≤3 days; Grade 3 diarrhea, nausea, or vomiting without use of antiemetics or antidiarrheals per standard of care; Grade 3 rash without use of corticosteroids or anti-inflammatory agents per standard of care d. Any Grade 3 or Grade 4 nonhematologic laboratory value if: • Clinically significant medical intervention is required to treat the participant, or • The abnormality leads to hospitalization, or • The abnormality persists for >1 week • The abnormality results in a drug-induced liver injury (DILI) Exceptions: Clinically nonsignificant, treatable, or reversible laboratory abnormalities including liver function tests, uric acid, etc. e. Febrile neutropenia Grade 3 or Grade 4: • Grade 3 is defined as absolute neutrophil count (ANC) <1000/mm 3 with a single temperature of >38.3 degrees C (101 degrees F) or a sustained temperature of ≥38 degrees C (100.4 degrees F) for more than 1 hour. • Grade 4 is defined as ANC <1000/mm 3 with a single temperature of >38.3 degrees C (101 degrees F) or a sustained temperature of ≥38 degrees C (100.4 degrees F) for more than 1 hour, with life-threatening consequences and urgent intervention indicated. f. Prolonged delay (>2 weeks) in initiating Cycle 2 due to intervention-related toxicity. g. Any intervention-related toxicity that causes the participant to discontinue intervention during Cycle 1. h. Grade 5 toxicity. Assessment at Screening and Prior to RECIST 1.1 Progression Until radiographic disease progression based on RECIST 1.1, there is no distinct iRECIST assessment. Assessment and Decision at RECIST 1.1 Progression For participants who show radiological disease progression by RECIST 1.1, the investigator will decide whether to continue a participant on study intervention until repeat scans 4 to 8 weeks later are obtained. Tumor flare may manifest as any factor causing radiographic progression per RECIST 1.1, including: x Increase in the sum of diameters of target lesion(s) identified at baseline to ≥20% and ≥5 mm from nadir. The Response Evaluation Criteria in Solid Tumors 1.1 for immune-based therapeutics (iRECIST) publication uses the terminology “sum of measurements,” but “sum of diameters” will be used in this protocol, consistent with the original RECIST 1.1 terminology. x Unequivocal progression of nontarget lesion(s) identified at baseline x Development of new lesion(s) iRECIST defines new response categories, including iUPD (unconfirmed progressive disease) and iCPD (confirmed progressive disease). For purposes of iRECIST assessment, the first visit showing progression according to RECIST 1.1 will be assigned a visit (overall) response of iUPD, regardless of which factors caused the progression. At this visit, target and nontarget lesions identified at baseline by RECIST 1.1 will be assessed as usual. New lesions will be classified as measurable or nonmeasurable, using the same size thresholds and rules as for baseline lesion assessment in RECIST 1.1. From measurable new lesions, up to 5 lesions total (up to 2 per organ), may be selected as New Lesions – Target. The sum of diameters of these lesions will be calculated and kept distinct from the sum of diameters for target lesions at baseline. All other new lesions will be followed qualitatively as New Lesions – Nontarget. Assessment at the Confirmatory Imaging On the confirmatory scans, the participant will be classified as progression confirmed (with an overall response of iCPD), or as showing persistent unconfirmed progression (with an overall response of iUPD), or as showing disease stability or response (stable disease by iRECIST (iSD) / partial response by iRECIST (iPR) / complete response by iRECIST (iCR)). Confirmation of Progression Progression is considered confirmed, and the overall response will be confirmed progressive disease by iRECIST (iCPD), if ANY of the following occurs: x Any of the factors that were the basis for the iUPD at the previous visit show worsening o For target lesions, worsening is a further increase in the sum of diameters of ≥5 mm, compared with any prior iUPD time point o For nontarget lesions, worsening is any significant growth in lesions overall, compared with a prior iUPD time point; this does not have to meet the “unequivocal” standard of RECIST 1.1 o For new lesions, worsening is any of these: ^ An increase in the new lesion sum of diameters by ≥5 mm from a prior iUPD time point ^ Visible growth of new nontarget lesions ^ The appearance of additional new lesions x Any new factor appears that would have triggered disease progression by RECIST 1.1 Persistent iUPD Progression is considered not confirmed, and the overall response remains iUPD, if: x None of the progression-confirming factors identified above occurs AND x The target lesion sum of diameters (initial target lesions) remains above the initial disease progression threshold (by RECIST 1.1) Additional scans for confirmation are to be scheduled 4 to 8 weeks from the scans on which iUPD is seen. This may correspond to the next visit in the original visit schedule. The assessment of the subsequent confirmation scan proceeds in an identical manner, with possible outcomes of iCPD, iUPD, and iSD/iPR/iCR. Resolution of iUPD Progression is considered not confirmed, and the overall response becomes iSD/iPR/iCR, if: x None of the progression-confirming factors identified above occurs, AND x The target lesion sum of diameters (initial target lesions) is not above the disease progression threshold. The response is classified as iSD or iPR (depending on the sum of diameters of the target lesions), or iCR if all lesions resolve. In this case, the initial iUPD is considered to be pseudoprogression, and the level of suspicion for progression is “reset.” This means that the next visit that shows radiographic progression, whenever it occurs, is again classified as iUPD by iRECIST, and the confirmation process is repeated before a response of iCPD can be assigned. Management Following the Confirmatory Imaging If repeat scans do not confirm disease progression, and the participant continues to be clinically stable, study intervention is to continue. The regular scan schedule is to be followed. If disease progression is confirmed, participants may be discontinued from study intervention. If a participant has confirmed radiographic progression (iCPD) and clinically meaningful benefit, study intervention may be continued after consultation with the Sponsor. If study intervention is continued, tumor scans are to be performed after the intervals. Detection of Progression at Visits After Pseudo-progression Resolves After resolution of pseudoprogression (i.e., after iSD/iPR/iCR), another instance of progression (another iUPD) is indicated by any of the following events: x Target lesions o Sum of diameters reaches the disease progression threshold (≥20% and ≥5 mm increase from nadir) either for the first time, or after resolution of previous pseudoprogression. The nadir is always the smallest sum of diameters seen during the entire study, either before or after an instance of pseudoprogression. x Nontarget lesions o If nontarget lesions have never shown unequivocal progression, their doing so for the first-time results in iUPD. o If nontarget lesions have shown previous unequivocal progression, and this progression has not resolved, iUPD results from any significant further growth of nontarget lesions, taken as a whole. o New lesions o New lesions appear for the first time o Additional new lesions appear o Previously identified new target lesions show an increase of ≥5 mm in the new lesion sum of diameters, from the nadir value of that sum o Previously identified nontarget lesions show any significant growth If any of the events above occur, the overall response for that visit is iUPD, and the iUPD evaluation process (see Assessment at the Confirmatory Scan above) is repeated. Progression must be confirmed before iCPD can occur. The decision process on the subsequent iUPD is identical to the iUPD confirmation process for the initial disease progression, with one exception, which can occur if new lesions had occurred at a prior instance of iUPD, had not resolved, then worsened (increase in size or number) leading to the second iUPD. If new lesion worsening has not resolved at the confirmatory scan, then iUPD cannot resolve to iSD or iPR. It will remain iUPD until either a decrease in the new lesion burden allows resolution to iSD or iPR, or until new or worsening cause of progression indicates iCPD. Discontinuation Discontinuation of study intervention does not represent withdrawal from the study. As certain data on clinical events beyond study intervention discontinuation may be important to the study, they must be collected through the participant’s last scheduled follow- up, even if the participant has discontinued study intervention. Therefore, all participants who discontinue study intervention before completion of the protocol-specified treatment period will still continue to be monitored in the study and participate in the study visits and procedures unless the participant has withdrawn from the study. Participants may discontinue study intervention at any time for any reason or be discontinued from the study intervention at the discretion of the investigator should any untoward effect occur. In addition, a participant may be discontinued from study intervention by the investigator or the Sponsor if study intervention is inappropriate, the study plan is violated, or for administrative and/or other safety reasons. A participant must be discontinued from study intervention, but continue to be monitored in the study for any of the following reasons: x The participant or participant’s legally acceptable representative requests to discontinue study intervention. x Any prolonged interruption of study intervention beyond the permitted periods, for irAE or AE management or other allowed dose interruptions, require Sponsor consultation before restarting treatment. If treatment will not be restarted, the participant will continue to be monitored in the study and the reason for discontinuation of study intervention will be recorded in the medical record. x The participant has a medical condition or personal circumstance, which in the opinion of the investigator and/or Sponsor, placed the participant at unnecessary risk from continued administration of study intervention. x The participant has a confirmed positive serum pregnancy test. x Radiographic disease progression (after obtaining informed consent addendum and Sponsor communication, the investigator may elect to continue treatment beyond imaging CRO (iCRO)-verified disease progression). x Any progression or recurrence of malignancy, or any occurrence of another malignancy that requires active treatment. x Any study intervention-related toxicity specified as a reason for permanent discontinuation as defined in the guidelines for dose modification due to AEs. x The participant requires any of the prohibited concomitant medication listed x Recurrence of a severe or life-threatening event, or of any of the laboratory abnormalities listed above, that are presumed to be immune-related. A participant must be withdrawn from the study if the participant or participant’s legally acceptable representative withdraws consent from the study. If a participant withdraws from the study, they will no longer receive study intervention or be followed at scheduled protocol visits. If a participant fails to return to the clinic for a required study visit and/or if the site is unable to contact the participant, the following procedures are to be performed: x The site must attempt to contact the participant and reschedule the missed visit. If the participant is contacted, the participant should be counseled on the importance of maintaining the protocol-specified visit schedule. x The investigator or designee must make every effort to regain contact with the participant at each missed visit (e.g., telephone calls and/or a certified letter to the participant’s last known mailing address or locally equivalent methods). These contact attempts should be documented in the participant’s medical record. x Note: A participant is not considered lost to follow-up until the last scheduled visit for the individual participant. The missing data for the participant will be managed via the prespecified statistical data handling and analysis guidelines. Informed Consent The investigator or medically qualified designee (consistent with local requirements) must obtain documented informed consent from each potential participant (or their legally acceptable representative) prior to participating in this clinical study If there are changes to the participant’s status during the study (e.g., health or age of majority requirements), the investigator or medically qualified designee must ensure the appropriate documented informed consent is in place. Tumor Imaging and Assessment of Disease The term “scan” refers to any medical imaging data used to assess tumor burden and may include cross-sectional imaging (such as CT or MRI), medical photography, or other methods as specified in this protocol. In addition to survival, efficacy will be assessed based on evaluation of scan changes in tumor burden over time, until the participant is discontinued from the study or goes into survival follow-up. The process for scan collection and transmission to the iCRO can be found in the Site Imaging Manual (SIM). Tumor scans by CT are strongly preferred. For the abdomen and pelvis, contrast-enhanced MRI may be used when CT with iodinated contrast is contraindicated, or when mandated by local practice. For the purposes of assessing tumor scans, the term “investigator” refers to the local investigator at the site and/or the radiological reviewer at the site or at an offsite facility. If brain scans are performed, MRI is preferred; however, CT imaging will be acceptable, if MRI is medically contraindicated. Bone scans may be performed to evaluate bone metastases. Any supplemental scans performed to support a positive or negative bone scan, such as plain X-rays acquired for correlation, should also be submitted to the iCRO. Other imaging modalities that may be collected, submitted to the iCRO, and included in the response assessment and those that should not be submitted to the iCRO and will not be included in response assessment are defined in the SIM. At screening, participant eligibility will require radiographic documentation of at least 1 lesion that meets the requirements for selection as a target lesion, before participant allocation. All scheduled scans for participants will be submitted to the iCRO. In addition, a scan that is obtained at an unscheduled time point, for any reason (including suspicion of progression or other clinical reason), should also be submitted to the iCRO if it shows disease progression, or if it is used to support a response assessment. All scans acquired within the protocol-specified window of time around a scheduled scan visit are to be classified as pertaining to that visit. Initial Tumor Imaging Initial tumor scans at screening must be performed within 28 days before the date of randomization/allocation. Any scans obtained after Cycle 1 Day 1 cannot be included in the screening assessment. The site must review screening scans to confirm the participant has measurable disease per RECIST 1.1. Tumor scans performed as part of routine clinical management are acceptable for screening if they are of acceptable diagnostic quality and performed within 28 days of randomization and can be assessed by the iCRO. If brain scans are required to document the stability of existing metastases, the brain MRI should be acquired during screening. Bone scans are required at screening for participants with a history of bone metastases and/or for those participants with indicative clinical signs/symptoms such as bone pain or elevated alkaline phosphatase levels. Tumor Imaging During the Study The first on study scan should be performed at 9 weeks (63 days +7 days) from the date of randomization/allocation. Subsequent tumor scans should be performed every 9 weeks (63 days ±7 days) or more frequently if clinically indicated. After 54 weeks (378 days ±7 days), participants who remain on treatment will have scans performed every 12 weeks (84 days ±7 days). Scan timing should follow calendar days and should not be adjusted for delays in cycle starts. Scans are to be performed until disease progression is identified by the investigator or notification by the Sponsor, or until the start of new anticancer treatment, withdrawal of consent, or death, whichever occurs first. Or should be confirmed by a repeat scan performed at least 4 weeks after the first indication of a response is observed. Participants will then return to the regular scan schedule, starting with the next scheduled time point. Participants who receive additional scans for confirmation do not need to undergo the next scheduled scan if it is fewer than 4 weeks later; scans may resume at the subsequent scheduled time point. On study brain or bone scans should be performed if clinically indicated or to confirm CR (if other lesions indicate CR and brain or bone lesions existed at baseline). When radiological disease progression is identified by the investigator in clinically stable participants, disease progression is to be confirmed by another set of scans performed 4 to 8 weeks later. If disease progression is not confirmed, clinically stable participants are to continue study intervention until progression is confirmed. Participants are to return to their regular scan schedule. If the next scheduled scan will occur in less than 4 weeks, this scheduled scan may be skipped. If disease progression is confirmed, study intervention will be discontinued. Safety Endpoints The safety endpoints include AEs, SAEs, and study intervention discontinuation due to AEs. Safety and tolerability will be assessed by clinical review of all relevant parameters including AEs, laboratory tests and vital signs. For cohorts with the safety lead-in phase, the safety endpoints also include the incidence of DLTs. Adverse Event (AE) An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a study intervention. For purposes of AE definition, study intervention (also referred to as Sponsor’s product) includes any pharmaceutical product, biological product, vaccine, diagnostic agent, medical device, combination product, or protocol specified procedure whether investigational or marketed (including placebo, active comparator product, or run-in intervention), manufactured by, licensed by, provided by, or distributed by the Sponsor for human use in this study. The following are included as AEs: x Any abnormal laboratory test results (hematology, clinical chemistry, or urinalysis) or other safety assessments (e.g., ECG, radiological scans, vital signs measurements), including those that worsen from baseline, considered clinically significant in the medical and scientific judgment of the investigator. x Exacerbation of a chronic or intermittent preexisting condition including either an increase in frequency and/or intensity of the condition. x New conditions detected or diagnosed after study intervention administration even though it may have been present before the start of the study. x Signs, symptoms, or the clinical sequelae of a suspected drug-drug interaction. x Signs, symptoms, or the clinical sequelae of a suspected overdose of either study intervention or a concomitant medication. x For all reports of overdose (whether accidental or intentional) with an associated AE, the AE term should reflect the clinical symptoms or abnormal test result. An overdose without any associated clinical symptoms or abnormal laboratory results is reported using the terminology “accidental or intentional overdose without adverse effect.” x Any new cancer (that is not a condition of the study). Progression of the cancer under study is not a reportable event. The following events do not meet the AE definition for the purpose of this study: x Medical or surgical procedure (e.g., endoscopy, appendectomy): the condition that leads to the procedure is the AE. x Situations in which an untoward medical occurrence did not occur (social and/or convenience admission to a hospital). x Anticipated day-to-day fluctuations of preexisting disease(s) or condition(s) present or detected at the start of the study that do not worsen. x Surgical procedure(s) planned prior to informed consent to treat a preexisting condition that has not worsened. Serious Adverse Event (SAE) If an event is not an AE per the above, then it cannot be an SAE even if serious conditions are met. An SAE is defined as any untoward medical occurrence that, at any dose: x Results in death. x Is life-threatening. The term “life-threatening” in the definition of “serious” refers to an event in which the participant was at risk of death at the time of the event. It does not refer to an event, which hypothetically might have caused death, if it were more severe. x Requires inpatient hospitalization or prolongation of existing hospitalization. Hospitalization is defined as an inpatient admission, regardless of length of stay, even if the hospitalization is a precautionary measure for continued observation. (Note: Hospitalization for an elective procedure to treat a pre-existing condition that has not worsened is not an SAE. A pre-existing condition is a clinical condition that is diagnosed prior to the use of the combinations disclosed herein and is documented in the participant’s medical history. x Results in persistent or significant disability/incapacity. The term disability means a substantial disruption of a person’s ability to conduct normal life functions. This definition is not intended to include experiences of relatively minor medical significance such as uncomplicated headache, nausea, vomiting, diarrhea, influenza, and accidental trauma (e.g., sprained ankle) that may interfere with or prevent everyday life functions but do not constitute a substantial disruption. x Is a congenital anomaly/birth defect. In offspring of participant taking the product regardless of time to diagnosis. x Other important medical events. Medical or scientific judgment should be exercised in deciding whether SAE reporting is appropriate in other situations such as important medical events that may not be immediately life-threatening or result in death or hospitalization but may jeopardize the participant or may require medical or surgical intervention to prevent 1 of the other outcomes listed in the above definition. These events should usually be considered serious. Examples of such events include invasive or malignant cancers, intensive treatment in an emergency room or at home for allergic bronchospasm, blood dyscrasias or convulsions that do not result in hospitalization, or development of drug dependency or drug abuse. The study treatments are outlined below in Tables 20-23. Table 20. Investigation summary – initial treatment of esophageal cancer, biliary cancer and gastric cancer Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes

Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes . Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes

Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes - . . Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes r Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes r Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes

Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes f t Initial Treatment/First Course – esophageal cancer, biliary cancer, g astric cancer Post-treatment Visits Notes a e . ves ga o su ay – a ea e o Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are r e . Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are r Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are a Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are y l Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are d . y Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are y e s Initial Treatment/First Course – TNBC Post-treatment Visits Notes Effic Survi All procedures are . g y p g , y cancer and gastric cancer Second Course – esophageal cancer, biliary End-of- Study Period: cancer, gastric cancer Treatm Post-Treatment Visits Notes Treatment Cycle = 21 days ent Second Course – esophageal cancer, biliary End-of- Study Period: cancer, gastric cancer Treatm Post-Treatment Visits Notes Treatment Cycle = 21 days ent e Second Course – esophageal cancer, biliary End-of- Study Period: cancer, gastric cancer Treatm Post-Treatment Visits Notes Treatment Cycle = 21 days ent t Second Course – esophageal cancer, biliary End-of- Study Period: cancer, gastric cancer Treatm Post-Treatment Visits Notes Treatment Cycle = 21 days ent . Second Course – esophageal cancer, biliary End-of- Study Period: cancer, gastric cancer Treatm Post-Treatment Visits Notes Treatment Cycle = 21 days ent n , Second Course – esophageal cancer, biliary End-of- Study Period: cancer, gastric cancer Treatm Post-Treatment Visits Notes Treatment Cycle = 21 days ent . g y Second Course – TNBC Post-treatment Visits Notes E Survi Second Course – TNBC Post-treatment Visits Notes E Survi s

Second Course – TNBC Post-treatment Visits Notes E Survi t

Second Course – TNBC Post-treatment Visits Notes E Survi Second Course – TNBC Post-treatment Visits Notes E Survi Second Course – TNBC Post-treatment Visits Notes E Survi p The present Example describes anti-tumor effects of a TIGIT antagonist and a PD-1 antagonist in combination with 5-Fluorouracil (5-FU) and Cisplatin chemotherapy, using mouse syngeneic models. Female C57BL/6J mice aged 7 weeks weighing between 18 to 21 grams were anesthetized and injected subcutaneously into the rear flank with 0.5 x 10 6 log-phase sub- confluent MC38 cells. Mice were pair-matched into treatment groups consisting of 10 mice per group when the mean tumor volume of inoculated MC38 tumors reached approximately 340mm 3 . Antibody reagents in this Example include: x Anti-PD-1 mIgG1 antibody, which is a murinized version of a rat anti–mouse PD1 Ab with a mouse IgG1 Fc; x Anti-TIGIT mIgG2a antibody having a mouse Fc region of mIgG2a subtype that is the murine counterpart isotype to human isotype IgG1; x Isotype mIgG1 antibody, which is a mouse IgG1 isotype-matched control monoclonal antibody specific for adenoviral hexon 25; and x Isotype mIgG2a antibody, which is a mouse IgG2a isotype-matched control monoclonal antibody specific for adenoviral hexon 25. The amino acid sequences of antibody reagents are listed in Table 24. Table 24. Sequences of antibodies used in Examples 2 and 3. Sequences SEQ ID NO. Anti-TIGIT DIQMTQSPSLLSASVGDRVTLNCIASQNIYKSLAWYQLKLGEAPKLLIYNA 303 mIgG2a NSLQAGIPSRFSGSGSGTDFALTISGLQPEDVATYFCQQYSGGYTFGAGTKL reatment groups consste o : 1) Isotype mouse IgG1 antibody (mIgG1) + isotype mouse IgG2a antibody (mIgG2a) + 0.9% saline; 2) Anti-PD-1 mIgG1 antibody (murinized pembrolizumab) + isotype mIgG2a antibody + 0.9% saline; 3) Anti-TIGIT mIgG2a antibody (murinized vibostolimab) + isotype mIgG1 antibody + 0.9% saline; 4) Anti-PD-1 mIgG1 antibody + anti-TIGIT mIgG2a antibody + 0.9% saline; 5) 5-FU + cisplatin + isotype mIgG1 antibody + isotype mIgG2a antibody; and 6) Anti-PD-1 mIgG1 antibody + anti-TIGIT mIgG2a antibody + 5-FU + cisplatin. Isotype mIgG1 control, a mouse monoclonal antibody specific for adenoviral hexon of the isotype IgG1, and anti-PD-1 antibodies were dosed intraperitoneally every 5 days at 5 mg/kg body weight. Isotype mIgG2a control, a mouse monoclonal antibody specific for adenoviral hexon of the isotype IgG2a, and anti-TIGIT antibodies were dosed intraperitoneally every 5 days at 10 mg/kg body weight.5-FU was dosed intraperitoneally once weekly at 60 mg/kg body weight. Cisplatin was dosed intraperitoneally once weekly at 4 mg/kg body weight. Start of treatments was considered Day 0 and dosing based on schedules continued as described until Day 38. Caliper measurements of tumors and body weights were captured twice weekly. Statistical analyses were performed by one-way ANOVA with Kruskal-Wallis multiple comparison tests at Day 17 specified when isotype group reached endpoints. The combination treatment of the anti-PD-1 antibody, the anti-TIGIT antibody, 5- FUm and cisplatin demonstrated the greatest anti-tumor efficacy such that 9 out of 10 animals achieved complete tumor regressions (CRs) where no measurable tumor was detected by end of study, as shown in Figures 1 and 2. In the anti-PD-1 antibody + anti-TIGIT antibody group, 8 out of 10 animals achieved CRs while the anti-PD-1 antibody monotherapy group achieved 2 out of 10 CRs. None of the other treatment group had CRs, as shown in Figure 2. A summary of the best overall response as measured by the change in tumor volume is depicted in Figure 2. The mean anti-tumor response of the quadruple combination treatment (i.e., Group 6) was significantly greater than the anti-tumor response observed with either isotype-treated animals (p<0.001) or the 5-FU + cisplatin chemotherapy group (p=0.007) as shown in Table 25. A summary of relevant mean tumor volume comparisons at Day 17 when isotype treated group exited the study is shown in Table 25. There was no significant body weight loss or adverse events observed in any animals treated with the above therapies, indicating the treatments were well tolerated. Table 25. Summary of 1-way ANOVA comparisons of mean tumor volumes used to determine significance between treatment groups. Analysis performed based on Day 17 when isotype treated animals exited study. Comparison P-value Example 3 The present Example describes anti-tumor effects of a TIGIT antagonist and a PD-1 antagonist in combination with gemcitabine and cisplatin chemotherapy, using mouse syngeneic models. Female C57BL/6J mice aged 7 weeks weighing between 18 to 21 grams were anesthetized and injected subcutaneously into the rear flank with 0.5 x 10 6 log-phase sub- confluent MC38 cells. Mice were pair-matched into treatment groups consisting of 10 mice per group when the mean tumor volume of inoculated MC38 tumors reached approximately 335mm 3 . Antibody reagents in this Example include: x Anti-PD-1 mIgG1 antibody, which is a murinized version of a rat anti–mouse PD1 Ab with a mouse IgG1 Fc; x Anti-TIGIT mIgG2a antibody having a mouse Fc region of mIgG2a subtype that is the murine counterpart isotype to human isotype IgG1; x Isotype mIgG1 antibody, which is a mouse IgG1 isotype-matched control monoclonal antibody specific for adenoviral hexon 25; and x Isotype mIgG2a antibody, which is a mouse IgG2a isotype-matched control monoclonal antibody specific for adenoviral hexon 25. The amino acid sequences of antibody reagents are listed in Table 24. Treatment groups consisted of: 1) Isotype mIgG1 antibody + Isotype mIgG2a antibody + 0.9% saline; 2) Anti-PD-1 mIgG1 antibody + Isotype mIgG2a antibody + 0.9% saline; 3) Anti-TIGIT mIgG2a antibody + Isotype mIgG1 antibody + 0.9% saline; 4) Anti-PD-1 mIgG1 antibody + anti-TIGIT mIgG2a antibody + 0.9% saline; 5) Gemcitabine + cisplatin + Isotype mIgG1 antibody + Isotype mIgG2a antibody; and 6) Anti-PD-1 mIgG1 antibody + anti-TIGIT mIgG2a antibody + gemcitabine + cisplatin. Isotype mIgG1 and anti-PD-1 antibodies were dosed intraperitoneally every 5 days at 5 mg/kg body weight. Isotype mIgG2a and anti-TIGIT antibodies were dosed intraperitoneally every 5 days at 10 mg/kg body weight. Gemcitabine was dosed intraperitoneally once weekly at 100 mg/kg body weight for four total doses. Cisplatin was dosed intraperitoneally once weekly at 4 mg/kg body weight for four total doses. Start of treatments was considered Day 0 and dosing based on schedules continued as described until Day 55. Caliper measurements of tumors and body weights were captured twice weekly. Statistical analyses were performed by one-way ANOVA with Kruskal-Wallis multiple comparison tests at Day 14 specified when control isotype group reached endpoints. Quadruple combination treatment with anti-PD-1 + anti-TIGIT + gemcitabine + cisplatin demonstrated the greatest anti-tumor efficacy as shown in Figures 3 and 4. 10 out of 10 animals achieved partial tumor regressions (PRs) where the final tumor volume was smaller than when treatment began. The result is summarized in Table 26. In the anti-PD-1 + anti-TIGIT group, 6 out of 10 animals achieved PRs while the anti-PD-1 monotherapy group achieved 2 out of 10 PRs. None of the treatment groups had complete tumor regressions. A summary of the best overall response as measured by the change in tumor volume is depicted in Figure 4. The mean anti-tumor response of the quadruple combination treatment (Group 6) was significantly greater than the anti-tumor response observed with either isotype-treated animals (p<0.001) or the gemcitabine + cisplatin chemotherapy group (p=0.032) as shown in Table 27. Although the quadruple combination group had more animals achieving PRs than the anti-PD-1 + anti-TIGIT groups, there was no statistically significant difference in tumor volumes, largely due to the variability in the latter group. A summary of relevant mean tumor volume comparisons at Day 14 when isotype treated group exited the study is shown in Table 27. There was no significant body weight loss or adverse events observed in any animals treated with the above therapies, indicating the treatments were well tolerated. Table 26. Summary of tumor growth inhibition (TGI) % at Day 14 when control animals exited study and the total number of partial tumor regressions observed at end of study (Day 55). T reatment Tumor growth Number of partial tumor regressions at Table 27. Summary of 1-way ANOVA comparisons of mean tumor volumes used to determine significance between treatment groups. Analysis performed based on Day 14 when isotype treated animals exited study. Comparison P-value