J Pathol Transl Med.  2020 Jan;54(1):20-33. 10.4132/jptm.2019.10.08.

Tumor immune response and immunotherapy in gastric cancer

Affiliations
  • 1Department of Pathology, Seoul National University Hospital, Seoul, Korea
  • 2Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
  • 3Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
  • 4Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
  • 5Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea

Abstract

Remarkable developments in immuno-oncology have changed the landscape of gastric cancer (GC) treatment. Because immunotherapy intervenes with tumor immune response rather than directly targeting tumor cells, it is important to develop a greater understanding of tumor immunity. This review paper summarizes the tumor immune reaction and immune escape mechanisms while focusing on the role of T cells and their co-inhibitory signals, such as the immune checkpoint molecules programmed death-1 and programmed deathligand 1 (PD-L1). This paper also describes past clinical trials of immunotherapy for patients with GC and details their clinical implications. Strong predictive markers are essential to improve response to immunotherapy. Microsatellite instability, Epstein-Barr virus, PD-L1 expression, and tumor mutational burden are now regarded as potent predictive markers for immunotherapy in patients with GC. Novel immunotherapy and combination therapy targeting new immune checkpoint molecules such as lymphocyte-activation gene 3, T cell immunoglobulin, and mucin domain containing-3, and indoleamine 2,3-dioxygenase have been suggested, and trials are ongoing to evaluate their safety and efficacy. Immunotherapy is an important treatment option for patients with GC and has great potential for improving patient outcome, and further research in immuno-oncology should be carried out.

Keyword

Stomach neoplasms; Immunotherapy; Programmed cell death-ligand 1; Microsatellite instability; Epstein-Barr virus; Tumor mutational burden; Tumor-infiltrating lymphocytes; Biomarker

Figure

  • Fig. 1. Cell-mediated tumor immunity. (A) Altered proteins are produced following genetic mutation of tumor cells or viral genes in tumor cells. Neoantigens can arise from these altered tumor proteins and be presented on tumor cell surface via major histocompatibility complex. Newly formed antigens on tumor cell surfaces are recognized by the immune system, and the tumor immune reaction is initiated. (B) T cell responses are generated by two signals. The first signal is binding between neoantigen presented on major histocompatibility complex (MHC) molecule and T cell receptor (TCR). The second signal is co-inhibitory or co-stimulatory and determines whether T cells will be activated or not. Programmed cell death protein-1 (PD-1), lymphocyte-activation gene 3, and T cell immunoglobulin, and mucin domain containing-3 are well known co-inhibitory receptors that bind specific ligands, such as programmed death-ligand 1 (PD-L1) or PD-L2. Binding between co-inhibitory receptors and their ligands induces T cell inactivation. Blockade of these co-inhibitory signals is the basic strategy for cancer immunotherapy.

  • Fig. 2. Representative figure of PD-L1 22C3 PharmDx assay. Most tumor cells show membranous staining. Some immune cells adjacent to tumor cells also had immunoreactivity to programmed death-ligand 1.

  • Fig. 3. Representative figure of Epstein-Barr virus (EBV) in situ hybridization. This case was diagnosed as gastric carcinoma with lymphoid stroma. EBV-positive cells highlight tumor cell clusters that form vague glandular structures.


Cited by  1 articles

A standardized pathology report for gastric cancer: 2nd edition
Young Soo Park, Myeong-Cherl Kook, Baek-hui Kim, Hye Seung Lee, Dong-Wook Kang, Mi-Jin Gu, Ok Ran Shin, Younghee Choi, Wonae Lee, Hyunki Kim, In Hye Song, Kyoung-Mee Kim, Hee Sung Kim, Guhyun Kang, Do Youn Park, So-Young Jin, Joon Mee Kim, Yoon Jung Choi, Hee Kyung Chang, Soomin Ahn, Mee Soo Chang, Song-Hee Han, Yoonjin Kwak, An Na Seo, Sung Hak Lee, Mee-Yon Cho
J Pathol Transl Med. 2023;57(1):1-27.    doi: 10.4132/jptm.2022.12.23.


Reference

1. Kaufman HL, Atkins MB, Subedi P, et al. The promise of Immunooncology: implications for defining the value of cancer treatment. J Immunother Cancer. 2019; 7:129.
Article
2. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013; 39:1–10.
Article
3. Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol. 2016; 27:1492–504.
Article
4. Melief CJ, van Hall T, Arens R, Ossendorp F, van der Burg SH. Therapeutic cancer vaccines. J Clin Invest. 2015; 125:3401–12.
Article
5. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013; 14:1014–22.
Article
6. van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer. 2016; 16:219–33.
Article
7. Welters MJ, Kenter GG, Piersma SJ, et al. Induction of tumor-specific CD4+ and CD8+ T-cell immunity in cervical cancer patients by a human papillomavirus type 16 E6 and E7 long peptides vaccine. Clin Cancer Res. 2008; 14:178–87.
Article
8. Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014; 14:135–46.
Article
9. Dranoff G. Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 2004; 4:11–22.
Article
10. Houghton AN, Guevara-Patiño JA. Immune recognition of self in immunity against cancer. J Clin Invest. 2004; 114:468–71.
Article
11. Schwartz RH. T cell anergy. Annu Rev Immunol. 2003; 21:305–34.
Article
12. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012; 12:252–64.
Article
13. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017; 541:321–30.
Article
14. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017; 27:109–18.
Article
15. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 2011; 29:235–71.
Article
16. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004; 22:329–60.
Article
17. Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012; 4:127ra37.
Article
18. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016; 39:98–106.
19. Schildberg FA, Klein SR, Freeman GJ, Sharpe AH. Coinhibitory pathways in the B7-CD28 ligand-receptor family. Immunity. 2016; 44:955–72.
Article
20. Huard B, Prigent P, Tournier M, Bruniquel D, Triebel F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol. 1995; 25:2718–21.
Article
21. Liu B, Wang M, Wang X, et al. Liver sinusoidal endothelial cell lectin inhibits CTL-dependent virus clearance in mouse models of viral hepatitis. J Immunol. 2013; 190:4185–95.
Article
22. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016; 44:989–1004.
Article
23. Parham P, Ohta T. Population biology of antigen presentation by MHC class I molecules. Science. 1996; 272:67–74.
Article
24. Garrido F, Algarra I. MHC antigens and tumor escape from immune surveillance. Adv Cancer Res. 2001; 83:117–58.
Article
25. del Campo AB, Kyte JA, Carretero J, et al. Immune escape of cancer cells with beta2-microglobulin loss over the course of metastatic melanoma. Int J Cancer. 2014; 134:102–13.
Article
26. Kumar V, Abbas A, Aster J. Robbins and Cotran pathologic basis of disease. 9th ed. Philadelphia: Elsevier;2014.
27. Jiang W, Liu K, Guo Q, et al. Tumor-infiltrating immune cells and prognosis in gastric cancer: a systematic review and meta-analysis. Oncotarget. 2017; 8:62312–29.
Article
28. Zheng X, Song X, Shao Y, et al. Prognostic role of tumor-infiltrating lymphocytes in gastric cancer: a meta-analysis. Oncotarget. 2017; 8:57386–98.
Article
29. Lee HE, Chae SW, Lee YJ, et al. Prognostic implications of type and density of tumour-infiltrating lymphocytes in gastric cancer. Br J Cancer. 2008; 99:1704–11.
Article
30. Haas M, Dimmler A, Hohenberger W, Grabenbauer GG, Niedobitek G, Distel LV. Stromal regulatory T-cells are associated with a favourable prognosis in gastric cancer of the cardia. BMC Gastroenterol. 2009; 9:65.
Article
31. Shen Z, Zhou S, Wang Y, et al. Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer. J Cancer Res Clin Oncol. 2010; 136:1585–95.
Article
32. Kim HI, Kim H, Cho HW, et al. The ratio of intra-tumoral regulatory T cells (Foxp3+)/helper T cells (CD4+) is a prognostic factor and associated with recurrence pattern in gastric cardia cancer. J Surg Oncol. 2011; 104:728–33.
Article
33. Kim KJ, Lee KS, Cho HJ, et al. Prognostic implications of tumor-infiltrating FoxP3+ regulatory T cells and CD8+ cytotoxic T cells in microsatellite-unstable gastric cancers. Hum Pathol. 2014; 45:285–93.
Article
34. Li K, Zhu Z, Luo J, et al. Impact of chemokine receptor CXCR3 on tumor-infiltrating lymphocyte recruitment associated with favorable prognosis in advanced gastric cancer. Int J Clin Exp Pathol. 2015; 8:14725–32.
35. Liu K, Yang K, Wu B, et al. Tumor-infiltrating immune cells are associated with prognosis of gastric cancer. Medicine (Baltimore). 2015; 94:e1631.
Article
36. Hennequin A, Derangere V, Boidot R, et al. Tumor infiltration by Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer patients. Oncoimmunology. 2016; 5:e1054598.
Article
37. Kim JW, Nam KH, Ahn SH, et al. Prognostic implications of immunosuppressive protein expression in tumors as well as immune cell infiltration within the tumor microenvironment in gastric cancer. Gastric Cancer. 2016; 19:42–52.
Article
38. Giampieri R, Maccaroni E, Mandolesi A, et al. Mismatch repair deficiency may affect clinical outcome through immune response activation in metastatic gastric cancer patients receiving first-line chemotherapy. Gastric Cancer. 2017; 20:156–63.
Article
39. Kawazoe A, Kuwata T, Kuboki Y, et al. Clinicopathological features of programmed death ligand 1 expression with tumor-infiltrating lymphocyte, mismatch repair, and Epstein-Barr virus status in a large cohort of gastric cancer patients. Gastric Cancer. 2017; 20:407–15.
40. Koh J, Ock CY, Kim JW, et al. Clinicopathologic implications of immune classification by PD-L1 expression and CD8-positive tumor-infiltrating lymphocytes in stage II and III gastric cancer patients. Oncotarget. 2017; 8:26356–67.
Article
41. Pernot S, Terme M, Radosevic-Robin N, et al. Infiltrating and peripheral immune cell analysis in advanced gastric cancer according to the Lauren classification and its prognostic significance. Gastric Cancer. 2019; Jul. 2. [Epub]. https://doi.org/10.1007/s10120-019-00983-3.
Article
42. Kim JY, Kim WG, Kwon CH, Park DY. Differences in immune contextures among different molecular subtypes of gastric cancer and their prognostic impact. Gastric Cancer. 2019; 22:1164–75.
Article
43. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015; 65:87–108.
Article
44. Charalampakis N, Economopoulou P, Kotsantis I, et al. Medical management of gastric cancer: a 2017 update. Cancer Med. 2018; 7:123–33.
Article
45. Kiyozumi Y, Iwatsuki M, Yamashita K, Koga Y, Yoshida N, Baba H. Update on targeted therapy and immune therapy for gastric cancer, 2018. J Cancer Metastasis Treat. 2018; 4:31.
Article
46. Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017; 390:2461–71.
Article
47. Fashoyin-Aje L, Donoghue M, Chen H, et al. FDA approval summary: pembrolizumab for recurrent locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma expressing PD-L1. Oncologist. 2019; 24:103–9.
Article
48. Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res. 2019; 25:3753–8.
Article
49. Muro K, Chung HC, Shankaran V, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol. 2016; 17:717–26.
Article
50. Fuchs CS, Doi T, Jang RW, et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 Trial. JAMA Oncol. 2018; 4:e180013.
51. Shitara K, Özgürog˘lu M, Bang YJ, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018; 392:123–33.
52. Tabernero J, Van Custem E, Bang YJ, et al. Pembrolizumab with or without chemotherapy versus chemotherapy for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma: the phase III KEYNOTE-062 study. J Clin Oncol. 2019; 37:LBA4007.
53. Janjigian YY, Bendell J, Calvo E, et al. CheckMate-032 study: efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer. J Clin Oncol. 2018; 36:2836–44.
Article
54. Boku N, Ryu MH, Kato K, et al. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4). Ann Oncol. 2019; 30:250–8.
Article
55. Janjigian YY, Adenis A, Aucoin JS, et al. Checkmate 649: a randomized, multicenter, open-label, phase 3 study of nivolumab (Nivo) plus ipilimumab (Ipi) versus oxaliplatin plus fluoropyrimidine in patients (Pts) with previously untreated advanced or metastatic gastric (G) or gastroesophageal junction (GEJ) cancer. J Clin Oncol. 2017; 35:TPS213.
Article
56. Kelly RJ, Lee J, Bang YJ, et al. Safety and efficacy of durvalumab in combination with tremelimumab, durvalumab monotherapy, and tremelimumab monotherapy in patients with advanced gastric cancer. J Clin Oncol. 2018; 36:4031.
Article
57. Bang YJ, Ruiz EY, Van Cutsem E, et al. Phase III, randomised trial of avelumab versus physician's choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: primary analysis of JAVELIN Gastric 300. Ann Oncol. 2018; 29:2052–60.
Article
58. Moehler M, Ryu MH, Dvorkin M, et al. Maintenance avelumab versus continuation of first-line chemotherapy in gastric cancer: JAVELIN Gastric 100 study design. Future Oncol. 2019; 15:567–77.
Article
59. A phase 1b/2 study of MEDI4736 with tremelimumab, MEDI4736 or tremelimumab monotherapy in gastric or GEJ adenocarcinoma. Bethesda: ClincalTrials.gov, 2015 [cited 2019 Jul 21]. Available from: https://clinicaltrials.gov/ct2/show/NCT02340975.
60. An investigational immuno-therapy study to assess the safety, tolerability and effectiveness of anti-LAG-3 with and without antiPD-1 in the treatment of solid tumors [Internet]. Bethesda: ClincalTrials.gov, 2013 [cited 2019 Jul 21]. Available from: https://clinicaltrials.gov/ct2/show/NCT01968109.
61. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992; 11:3887–95.
Article
62. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012; 366:2517–9.
Article
63. Qing Y, Li Q, Ren T, et al. Upregulation of PD-L1 and APE1 is associated with tumorigenesis and poor prognosis of gastric cancer. Drug Des Devel Ther. 2015; 9:901–9.
64. Wu C, Zhu Y, Jiang J, Zhao J, Zhang XG, Xu N. Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem. 2006; 108:19–24.
Article
65. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014; 513:202–9.
66. Interpretation manual: gastric or gastroesophageal junction adenocarcinoma. PD-L1 IHC 22C3 pharmDx interpretation manual: gastric or gastroesophageal juction adenocarcinoma [Internet]. Santa Clara: DAKO Agilent Technologies, 2018 [cited 2019 Jul 21]. Available from: https://www.agilent.com/cs/library/usermanuals/public/29219_pd-l1-ihc-22C3-pharmdx-gastric-interpretation-manual_us.pdf.
67. Batenchuk C, Albitar M, Zerba K, et al. A real-world, comparative study of FDA-approved diagnostic assays PD-L1 IHC 28-8 and 22C3 in lung cancer and other malignancies. J Clin Pathol. 2018; 71:1078–83.
Article
68. Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015; 21:449–56.
Article
69. Ratti M, Lampis A, Hahne JC, Passalacqua R, Valeri N. Microsatellite instability in gastric cancer: molecular bases, clinical perspectives, and new treatment approaches. Cell Mol Life Sci. 2018; 75:4151–62.
Article
70. Polom K, Marano L, Marrelli D, et al. Meta-analysis of microsatellite instability in relation to clinicopathological characteristics and overall survival in gastric cancer. Br J Surg. 2018; 105:159–67.
Article
71. Kim JY, Shin NR, Kim A, et al. Microsatellite instability status in gastric cancer: a reappraisal of its clinical significance and relationship with mucin phenotypes. Korean J Pathol. 2013; 47:28–35.
Article
72. Setia N, Agoston AT, Han HS, et al. A protein and mRNA expression-based classification of gastric cancer. Mod Pathol. 2016; 29:772–84.
Article
73. Ahn S, Lee SJ, Kim Y, et al. High-throughput protein and mRNA expression-based classification of gastric cancers can identify clinically distinct subtypes, concordant with recent molecular classifications. Am J Surg Pathol. 2017; 41:106–15.
Article
74. Cho J, Chang YH, Heo YJ, et al. Four distinct immune microenvironment subtypes in gastric adenocarcinoma with special reference to microsatellite instability. ESMO Open. 2018; 3:e000326.
Article
75. Kim ST, Cristescu R, Bass AJ, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 2018; 24:1449–58.
Article
76. Lee J, Kim KM. Biomarkers for gastric cancer: molecular classification revisited. Precis Future Med. 2017; 1:59–68.
Article
77. Lee HS, Kim WH, Kwak Y, et al. Molecular testing for gastrointestinal cancer. J Pathol Transl Med. 2017; 51:103–21.
Article
78. Murphy KM, Zhang S, Geiger T, et al. Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn. 2006; 8:305–11.
Article
79. Campanella NC, Berardinelli GN, Scapulatempo-Neto C, et al. Optimization of a pentaplex panel for MSI analysis without control DNA in a Brazilian population: correlation with ancestry markers. Eur J Hum Genet. 2014; 22:875–80.
Article
80. Bae YS, Kim H, Noh SH, Kim H. Usefulness of immunohistochemistry for microsatellite instability screening in gastric cancer. Gut Liver. 2015; 9:629–35.
Article
81. Normanno N, Rachiglio AM, Lambiase M, et al. Heterogeneity of KRAS, NRAS, BRAF and PIK3CA mutations in metastatic colorectal cancer and potential effects on therapy in the CAPRI GOIM trial. Ann Oncol. 2015; 26:1710–4.
82. Vanderwalde A, Spetzler D, Xiao N, Gatalica Z, Marshall J. Microsatellite instability status determined by next-generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med. 2018; 7:746–56.
Article
83. Lee HS, Chang MS, Yang HK, Lee BL, Kim WH. Epstein-barr viruspositive gastric carcinoma has a distinct protein expression profile in comparison with epstein-barr virus-negative carcinoma. Clin Cancer Res. 2004; 10:1698–705.
Article
84. Kang BW, Seo AN, Yoon S, et al. Prognostic value of tumor-infiltrating lymphocytes in Epstein-Barr virus-associated gastric cancer. Ann Oncol. 2016; 27:494–501.
Article
85. Cho J, Kang MS, Kim KM. Epstein-Barr virus-associated gastric carcinoma and specific features of the accompanying immune response. J Gastric Cancer. 2016; 16:1–7.
Article
86. Derks S, Liao X, Chiaravalli AM, et al. Abundant PD-L1 expression in Epstein-Barr virus-infected gastric cancers. Oncotarget. 2016; 7:32925–32.
Article
87. Naseem M, Barzi A, Brezden-Masley C, et al. Outlooks on Epstein-Barr virus associated gastric cancer. Cancer Treat Rev. 2018; 66:15–22.
Article
88. Yau TO, Tang CM, Yu J. Epigenetic dysregulation in Epstein-Barr virus-associated gastric carcinoma: disease and treatments. World J Gastroenterol. 2014; 20:6448–56.
Article
89. Ambinder RF, Mann RB. Epstein-Barr-encoded RNA in situ hybridization: diagnostic applications. Hum Pathol. 1994; 25:602–5.
Article
90. Gulley ML. Molecular diagnosis of Epstein-Barr virus-related diseases. J Mol Diagn. 2001; 3:1–10.
Article
91. Chen XZ, Chen H, Castro FA, Hu JK, Brenner H. Epstein-Barr virus infection and gastric cancer: a systematic review. Medicine (Baltimore). 2015; 94:e792.
92. Melendez B, Van Campenhout C, Rorive S, Remmelink M, Salmon I, D’Haene N. Methods of measurement for tumor mutational burden in tumor tissue. Transl Lung Cancer Res. 2018; 7:661–7.
93. Van Allen EM, Wagle N, Stojanov P, et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat Med. 2014; 20:682–8.
Article
94. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015; 348:124–8.
95. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014; 371:2189–99.
Article
96. Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018; 378:2093–104.
Article
97. Steuer CE, Ramalingam SS. Tumor mutation burden: leading immunotherapy to the era of precision medicine? J Clin Oncol. 2018; 36:631–2.
Article
98. Wang F, Wei XL, Wang FH, et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemorefractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol. 2019; 30:1479–86.
Article
99. Griffith M, Miller CA, Griffith OL, et al. Optimizing cancer genome sequencing and analysis. Cell Syst. 2015; 1:210–23.
Article
100. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017; 9:34.
Article
101. He Y, Rivard CJ, Rozeboom L, et al. Lymphocyte-activation gene-3, an important immune checkpoint in cancer. Cancer Sci. 2016; 107:1193–7.
Article
102. Woo SR, Turnis ME, Goldberg MV, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012; 72:917–27.
Article
103. Coutzac C, Pernot S, Chaput N, Zaanan A. Immunotherapy in advanced gastric cancer, is it the future? Crit Rev Oncol Hematol. 2019; 133:25–32.
Article
104. Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005; 6:1245–52.
Article
105. Yanai H, Ban T, Wang Z, et al. HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature. 2009; 462:99–103.
Article
106. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010; 207:2187–94.
Article
107. Prendergast GC, Smith C, Thomas S, et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol Immunother. 2014; 63:721–35.
Article
108. Hornyak L, Dobos N, Koncz G, et al. The Role of Indoleamine-2,3-Dioxygenase in Cancer Development, Diagnostics, and Therapy. Front Immunol. 2018; 9:151.
Article
109. Terness P, Bauer TM, Röse L, et al. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J Exp Med. 2002; 196:447–57.
110. Spranger S, Koblish HK, Horton B, Scherle PA, Newton R, Gajewski TF. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. 2014; 2:3.
Article
111. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016; 375:819–29.
Article
112. Chowell D, Morris LG, Grigg CM, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018; 359:582–7.
Article
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr