1932

Abstract

Molecularly targeted therapy and immunotherapy have dramatically changed the landscape of available treatment options for patients with advanced cancer. Improved understanding of the molecular and genomic features of cancers over the last decade has led to the development of successful targeted therapies and the field of precision cancer medicine. As a result of these advances, patients whose tumors harbor select molecular alterations are eligible for treatment with targeted therapies active against the unique molecular aberration. Concurrently, advances in tumor immunology have led to the development of immunomodulatory antibodies targeting T cell coinhibitory receptors CTLA-4 and PD-1 (programmed death–1) that have shown activity in several cancer histologies, reinvigorating antitumor immune responses in a subset of patients. These immunomodulatory antibodies offer the promise of durable disease control. However, discrete genomic determinants of response to cancer immunotherapy, unlike molecularly targeted therapies, have remained elusive, and robust biomarkers are lacking. Recent advances in tumor profiling have begun to identify novel genomic features that may influence response and resistance to cancer immunotherapy, including tumor mutational burden (e.g., microsatellite instability), copy-number alterations, and specific somatic alterations that influence immune recognition and response. Further investigation into the molecular and genomic features of response and resistance to cancer immunotherapy will be needed. We review the recent advances in understanding the molecular and genomic determinants of response to cancer immunotherapy, with an emphasis on immune checkpoint inhibitors.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-060116-022926
2018-01-29
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/med/69/1/annurev-med-060116-022926.html?itemId=/content/journals/10.1146/annurev-med-060116-022926&mimeType=html&fmt=ahah

Literature Cited

  1. Hanahan D, Weinberg RA. 1.  2000. The hallmarks of cancer. Cell 100:57–70 [Google Scholar]
  2. Hanahan D, Coussens LM. 2.  2012. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21:309–22 [Google Scholar]
  3. Palucka AK, Coussens LM. 3.  2016. The basis of oncoimmunology. Cell 164:1233–47 [Google Scholar]
  4. Pardoll DM. 4.  2012. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12:252–64 [Google Scholar]
  5. Camidge DR, Pao W, Sequist LV. 5.  2014. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat. Rev. Clin. Oncol. 11:473–81 [Google Scholar]
  6. Carlino MS, Long GV, Kefford RF, Rizos H. 6.  2015. Targeting oncogenic BRAF and aberrant MAPK activation in the treatment of cutaneous melanoma. Crit. Rev. Oncol. Hematol. 96:385–98 [Google Scholar]
  7. Dienstmann R, Jang IS, Bot B. 7.  et al. 2015. Database of genomic biomarkers for cancer drugs and clinical targetability in solid tumors. Cancer Discov 5:118–23 [Google Scholar]
  8. Tannock IF, Hickman JA. 8.  2016. Limits to personalized cancer medicine. N. Engl. J. Med. 375:1289–94 [Google Scholar]
  9. O'Donnell JS, Long GV, Scolyer RA. 9.  et al. 2017. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat Rev 52:71–81 [Google Scholar]
  10. Pitt JM, Vetizou M, Daillere R. 10.  et al. 2016. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity 44:1255–69 [Google Scholar]
  11. Carbognin L, Pilotto S, Milella M. 11.  et al. 2015. Differential activity of nivolumab, pembrolizumab and MPDL3280a according to the tumor expression of programmed death-ligand-1 (PD-L1): sensitivity analysis of trials in melanoma, lung and genitourinary cancers. PLOS ONE 10:e0130142 [Google Scholar]
  12. Kluger HM, Zito CR, Turcu G. 13.  et al. 2017. PD-L1 studies across tumor types, its differential expression and predictive value in patients treated with immune checkpoint inhibitors. Clin. Cancer Res. 23:154270–79 [Google Scholar]
  13. Reck M, Rodriguez-Abreu D, Robinson AG. 12.  et al. 2016. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 375:1823–33 [Google Scholar]
  14. Lau J, Cheung J, Navarro A. 14.  et al. 2017. Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice. Nat. Commun. 8:14572 [Google Scholar]
  15. Topalian SL, Hodi FS, Brahmer JR. 15.  et al. 2012. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366:2443–54 [Google Scholar]
  16. Brahmer JR, Tykodi SS, Chow LQ. 16.  et al. 2012. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366:2455–65 [Google Scholar]
  17. Hodi FS, O'Day SJ, McDermott DF. 17.  et al. 2010. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363:711–23 [Google Scholar]
  18. Schadendorf D, Hodi FS, Robert C. 18.  et al. 2015. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J. Clin. Oncol. 33:1889–94 [Google Scholar]
  19. Ribas A, Hamid O, Daud A. 19.  et al. 2016. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA 315:1600–9 [Google Scholar]
  20. Larkin J, Hodi FS, Wolchok JD. 20.  2015. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373:1270–71 [Google Scholar]
  21. Postow MA, Chesney J, Pavlick AC. 21.  et al. 2015. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N. Engl. J. Med. 372:2006–17 [Google Scholar]
  22. De Velasco G, Je Y, Bosse D. 22.  et al. 2017. Comprehensive meta-analysis of key immune-related adverse events from CTLA-4 and PD-1/PD-L1 inhibitors in cancer patients. Cancer Immunol. Res. 5:312–18 [Google Scholar]
  23. Topalian SL, Drake CG, Pardoll DM. 23.  2015. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27:450–61 [Google Scholar]
  24. Tumeh PC, Harview CL, Yearley JH. 24.  et al. 2014. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515:568–71 [Google Scholar]
  25. Taube JM, Young GD, McMiller TL. 25.  et al. 2015. Differential expression of immune-regulatory genes associated with PD-L1 display in melanoma: implications for PD-1 pathway blockade. Clin. Cancer Res. 21:3969–76 [Google Scholar]
  26. Teng MW, Ngiow SF, Ribas A, Smyth MJ. 26.  2015. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 75:2139–45 [Google Scholar]
  27. Gotwals P, Cameron S, Cipolletta D. 27.  et al. 2017. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat. Rev. Cancer 17:286–301 [Google Scholar]
  28. Buque A, Bloy N, Aranda F. 28.  et al. 2015. Trial watch: immunomodulatory monoclonal antibodies for oncological indications. Oncoimmunology 4:e1008814 [Google Scholar]
  29. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. 29.  2008. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26:677–704 [Google Scholar]
  30. Buchbinder EI, Desai A. 30.  2016. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. 39:98–106 [Google Scholar]
  31. Veras E, Kurman RJ, Wang TL, Shih IM. 31.  2017. PD-L1 expression in human placentas and gestational trophoblastic diseases. Int. J. Gynecol. Pathol. 36:146–53 [Google Scholar]
  32. Schreiber RD, Old LJ, Smyth MJ. 32.  2011. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 331:1565–70 [Google Scholar]
  33. Chen DS, Mellman I. 33.  2017. Elements of cancer immunity and the cancer-immune set point. Nature 541:321–30 [Google Scholar]
  34. Harty JT, Badovinac VP. 34.  2008. Shaping and reshaping CD8+ T-cell memory. Nat. Rev. Immunol. 8:107–19 [Google Scholar]
  35. Farber DL, Yudanin NA, Restifo NP. 35.  2014. Human memory T cells: generation, compartmentalization and homeostasis. Nat. Rev. Immunol. 14:24–35 [Google Scholar]
  36. Ribas A, Shin DS, Zaretsky J. 36.  et al. 2016. PD-1 blockade expands intratumoral memory T Cells. Cancer Immunol. Res. 4:194–203 [Google Scholar]
  37. Restifo NP, Smyth MJ, Snyder A. 37.  2016. Acquired resistance to immunotherapy and future challenges. Nat. Rev. Cancer 16:121–26 [Google Scholar]
  38. Joyce JA, Fearon DT. 38.  2015. T cell exclusion, immune privilege, and the tumor microenvironment. Science 348:74–80 [Google Scholar]
  39. Gubin MM, Zhang X, Schuster H. 39.  et al. 2014. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515:577–81 [Google Scholar]
  40. Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. 40.  2014. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat. Rev. Cancer 14:135–46 [Google Scholar]
  41. Lawrence MS, Stojanov P, Polak P. 41.  et al. 2013. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499:214–18 [Google Scholar]
  42. Mouw KW, Goldberg MS, Konstantinopoulos PA, D'Andrea AD. 42.  2017. DNA Damage and repair biomarkers of immunotherapy response. Cancer Discov 7:7675–93 [Google Scholar]
  43. Le DT, Uram JN, Wang H. 43.  et al. 2015. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372:2509–20 [Google Scholar]
  44. Le DT, Durham JN, Smith KN. 44.  et al. 2017. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357:6349409–13 [Google Scholar]
  45. Rizvi NA, Hellmann MD, Snyder A. 45.  et al. 2015. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348:124–28 [Google Scholar]
  46. Snyder A, Makarov V, Merghoub T. 46.  et al. 2014. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371:2189–99 [Google Scholar]
  47. Van Allen EM, Miao D, Schilling B. 47.  et al. 2015. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350:207–11 [Google Scholar]
  48. Higuchi T, Flies DB, Marjon NA. 48.  et al. 2015. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer. Cancer Immunol. Res. 3:1257–68 [Google Scholar]
  49. Strickland KC, Howitt BE, Shukla SA. 49.  et al. 2016. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 7:13587–98 [Google Scholar]
  50. Wagle N, Grabiner BC, Van Allen EM. 50.  et al. 2014. Response and acquired resistance to everolimus in anaplastic thyroid cancer. N. Engl. J. Med. 371:1426–33 [Google Scholar]
  51. Van Allen EM, Golay HG, Liu Y. 51.  et al. 2015. Long-term benefit of PD-L1 blockade in lung cancer associated with JAK3 activation. Cancer Immunol. Res. 3:855–63 [Google Scholar]
  52. Johanns TM, Miller CA, Dorward IG. 52.  et al. 2016. Immunogenomics of hypermutated glioblastoma: a patient with germline POLE deficiency treated with checkpoint blockade immunotherapy. Cancer Discov 6:1230–36 [Google Scholar]
  53. Bouffet E, Larouche V, Campbell BB. 53.  et al. 2016. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J. Clin. Oncol. 34:2206–11 [Google Scholar]
  54. Chen DS, Mellman I. 54.  2013. Oncology meets immunology: the cancer-immunity cycle. Immunity 39:1–10 [Google Scholar]
  55. Miao D, Van Allen EM. 55.  2016. Genomic determinants of cancer immunotherapy. Curr. Opin. Immunol. 41:32–38 [Google Scholar]
  56. Schumacher TN, Schreiber RD. 56.  2015. Neoantigens in cancer immunotherapy. Science 348:69–74 [Google Scholar]
  57. McGranahan N, Furness AJ, Rosenthal R. 57.  et al. 2016. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351:1463–69 [Google Scholar]
  58. Anagnostou V, Smith KN, Forde PM. 58.  et al. 2016. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov 7:3264–76 [Google Scholar]
  59. Hugo W, Zaretsky JM, Sun L. 59.  et al. 2016. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165:35–44 [Google Scholar]
  60. Zaretsky JM, Garcia-Diaz A, Shin DS. 60.  et al. 2016. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375:9819–29 [Google Scholar]
  61. Shin DS, Zaretsky JM, Escuin-Ordinas H. 61.  et al. 2017. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov 7:188–201 [Google Scholar]
  62. Peng W, Chen JQ, Liu C. 62.  et al. 2016. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov 6:202–16 [Google Scholar]
  63. George S, Miao D, Demetri GD. 63.  et al. 2017. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity 46:197–204 [Google Scholar]
  64. Spranger S, Bao R, Gajewski TF. 64.  2015. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 523:231–35 [Google Scholar]
  65. Davoli T, Uno H, Wooten EC, Elledge SJ. 65.  2017. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355:1–14 [Google Scholar]
  66. Roh W, Chen PL, Reuben A. 66.  et al. 2017. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 9:1–11 [Google Scholar]
  67. Gao J, Shi LZ, Zhao H. 67.  et al. 2016. Loss of IFN-gamma pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167:397–404.e9 [Google Scholar]
  68. Benci JL, Xu B, Qiu Y. 68.  et al. 2016. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell 167:1540–54.e12 [Google Scholar]
  69. Heninger E, Krueger TE, Lang JM. 69.  2015. Augmenting antitumor immune responses with epigenetic modifying agents. Front. Immunol. 6: 29:1–14 [Google Scholar]
  70. Peng D, Kryczek I, Nagarsheth N. 70.  et al. 2015. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 527:249–53 [Google Scholar]
  71. Downey RF, Sullivan FJ, Wang-Johanning F. 71.  et al. 2015. Human endogenous retrovirus K and cancer: innocent bystander or tumorigenic accomplice?. Int. J. Cancer 137:1249–57 [Google Scholar]
  72. Li J, Hu L, Liu Y. 72.  et al. 2015. DDX19A senses viral RNA and mediates NLRP3-dependent inflammasome activation. J. Immunol. 195:5732–49 [Google Scholar]
  73. Nissen KK, Laska MJ, Hansen B. 73.  et al. 2013. Endogenous retroviruses and multiple sclerosis—new pieces to the puzzle. BMC Neurol 13:111 [Google Scholar]
  74. Suntsova M, Garazha A, Ivanova A. 74.  et al. 2015. Molecular functions of human endogenous retroviruses in health and disease. Cell Mol. Life Sci. 72:3653–75 [Google Scholar]
  75. Volkman HE, Stetson DB. 75.  2014. The enemy within: endogenous retroelements and autoimmune disease. Nat. Immunol. 15:415–22 [Google Scholar]
  76. Chiappinelli KB, Strissel PL, Desrichard A. 76.  et al. 2015. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 162:974–86 [Google Scholar]
  77. Roulois D, Loo Yau H, Singhania R. 77.  et al. 2015. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 162:961–73 [Google Scholar]
  78. Rooney MS, Shukla SA, Wu CJ. 78.  et al. 2015. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160:48–61 [Google Scholar]
  79. Iglesia MD, Parker JS, Hoadley KA. 79.  et al. 2016. Genomic analysis of immune cell infiltrates across 11 tumor types. J. Natl. Cancer Inst. 108:1–4 [Google Scholar]
  80. Newman AM, Liu CL, Green MR. 80.  et al. 2015. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12:453–57 [Google Scholar]
  81. Gentles AJ, Newman AM, Liu CL. 81.  et al. 2015. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21:938–45 [Google Scholar]
  82. Li B, Severson E, Pignon JC. 82.  et al. 2016. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biol 17:174 [Google Scholar]
  83. Charoentong P, Finotello F, Angelova M. 83.  et al. 2017. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep 18:248–62 [Google Scholar]
  84. De Henau O, Rausch M, Winkler D. 84.  et al. 2016. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kgamma in myeloid cells. Nature 539:443–47 [Google Scholar]
  85. Kaneda MM, Messer KS, Ralainirina N. 85.  et al. 2016. PI3Kgamma is a molecular switch that controls immune suppression. Nature 539:437–42 [Google Scholar]
  86. Gebhardt C, Sevko A, Jiang H. 86.  et al. 2015. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin. Cancer Res. 21:5453–59 [Google Scholar]
  87. Meyer C, Cagnon L, Costa-Nunes CM. 87.  et al. 2014. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol. Immunother. 63:247–57 [Google Scholar]
  88. Arce Vargas F, Furness AJS, Solomon I. 88.  et al. 2017. Fc-optimized anti-CD25 depletes tumor-infiltrating regulatory T cells and synergizes with PD-1 blockade to eradicate established tumors. Immunity 46:577–86 [Google Scholar]
  89. Shang B, Liu Y, Jiang SJ, Liu Y. 89.  2015. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci. Rep. 5:15179 [Google Scholar]
  90. Tashiro H, Brenner MK. 90.  2017. Immunotherapy against cancer-related viruses. Cell Res 27:59–73 [Google Scholar]
  91. Urbani S, Amadei B, Tola D. 91.  et al. 2006. PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. J. Virol. 80:11398–403 [Google Scholar]
  92. Wherry EJ, Kurachi M. 92.  2015. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15:486–99 [Google Scholar]
  93. Kahan SM, Wherry EJ, Zajac AJ. 93.  2015. T cell exhaustion during persistent viral infections. Virology 479–480:180–93 [Google Scholar]
  94. Mandal R, Senbabaoglu Y, Desrichard A. 94.  et al. 2016. The head and neck cancer immune landscape and its immunotherapeutic implications. JCI Insight 1:e89829 [Google Scholar]
  95. Gao G, Smith DI. 95.  2016. Human papillomavirus and the development of different cancers. Cytogenet. Genome Res. 150:185–93 [Google Scholar]
  96. Ferris RL, Blumenschein G Jr., Fayette J. 96.  et al. 2016. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 375:1856–67 [Google Scholar]
  97. Seiwert TY, Burtness B, Mehra R. 97.  et al. 2016. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol 17:956–65 [Google Scholar]
  98. Nghiem PT, Bhatia S, Lipson EJ. 98.  et al. 2016. PD-1 blockade with pembrolizumab in advanced Merkel-cell carcinoma. N. Engl. J. Med. 374:2542–52 [Google Scholar]
  99. Green MR, Rodig S, Juszczynski P. 99.  et al. 2012. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin. Cancer Res. 18:1611–18 [Google Scholar]
  100. Green MR, Monti S, Rodig SJ. 100.  et al. 2010. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 116:3268–77 [Google Scholar]
  101. Ansell SM, Lesokhin AM, Borrello I. 101.  et al. 2015. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N. Engl. J. Med. 372:311–19 [Google Scholar]
  102. Ma J, Li J, Hao Y. 102.  et al. 2017. Differentiated tumor immune microenvironment of Epstein-Barr virus-associated and negative gastric cancer: implication in prognosis and immunotherapy. Oncotarget 8:4067094–103 [Google Scholar]
  103. El-Khoueiry AB, Sangro B, Yau T. 103.  et al. 2017. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 389:2492–502 [Google Scholar]
  104. Goh G, Walradt T, Markarov V. 104.  et al. 2016. Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 7:3403–15 [Google Scholar]
  105. Harms PW, Vats P, Verhaegen ME. 105.  et al. 2015. The distinctive mutational spectra of polyomavirus-negative Merkel cell carcinoma. Cancer Res 75:3720–27 [Google Scholar]
  106. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. 106.  2017. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat. Rev. Clin. Oncol. 88:1–14 [Google Scholar]
  107. Koyama S, Akbay EA, Li YY. 107.  et al. 2016. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 7:10501 [Google Scholar]
  108. Gao J, Ward JF, Pettaway CA. 108.  et al. 2017. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat. Med. 23:551–55 [Google Scholar]
  109. Ascierto ML, Makohon-Moore A, Lipson EJ. 109.  et al. 2017. Transcriptional mechanisms of resistance to anti-PD-1 therapy. Clin. Cancer Res. 23:3168–80 [Google Scholar]
  110. Friedman AA, Letai A, Fisher DE, Flaherty KT. 110.  2015. Precision medicine for cancer with next-generation functional diagnostics. Nat. Rev. Cancer 15:747–56 [Google Scholar]
/content/journals/10.1146/annurev-med-060116-022926
Loading
/content/journals/10.1146/annurev-med-060116-022926
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error