1932

Abstract

The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable—and, in theory, rejectable—by the host T cell immune response. In addition, cancers often overexpress tissue-specific and developmental antigens to which immune tolerance is incomplete. Sometimes, virally transferred oncogenes drive malignant transformation and remain expressed throughout the cancer. Despite this state of antigenic sufficiency, cancer grows progressively and overcomes all efforts of the host immune system to contain it. While therapeutic cancer vaccination can mobilize high frequencies of tumor-specific T cells, these responses remain subject to intratumoral attenuation. Antibody modulation of T cell function through checkpoint blockade or costimulatory activation can restore survival, proliferation, and effector function to these tumor-infiltrating T cells and convert otherwise subtherapeutic vaccines into potentially curative cancer immunotherapeutics.

Keyword(s): 4–1BBCTLA-4immunotherapyPD-1vaccines
Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-050217-121900
2019-01-27
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/med/70/1/annurev-med-050217-121900.html?itemId=/content/journals/10.1146/annurev-med-050217-121900&mimeType=html&fmt=ahah

Literature Cited

  1. 1.  Boon T, Coulie PG, Van den Eynde BJ, van der Bruggen P 2006. Human T cell responses against melanoma. Annu. Rev. Immunol. 24:175–208
    [Google Scholar]
  2. 2.  Powell DJ Jr, Rosenberg SA 2004. Phenotypic and functional maturation of tumor antigen-reactive CD8+ T lymphocytes in patients undergoing multiple course peptide vaccination. J. Immunother. 27:36–47
    [Google Scholar]
  3. 3.  Berger TG, Haendle I, Schrama D et al. 2004. Circulation and homing of melanoma-reactive T cells to both cutaneous and visceral metastases after vaccination with monocyte-derived dendritic cells. Int. J. Cancer 111:229–37
    [Google Scholar]
  4. 4.  Chiong B, Wong R, Lee P et al. 2004. Characterization of long-term effector-memory T-cell responses in patients with resected high-risk melanoma receiving a melanoma peptide vaccine. J. Immunother. 27:368–79
    [Google Scholar]
  5. 5.  Speiser DE, Pittet MJ, Rimoldi D et al. 2003. Evaluation of melanoma vaccines with molecularly defined antigens by ex vivo monitoring of tumor-specific T cells. Semin. Cancer Biol. 13:461–72
    [Google Scholar]
  6. 6.  Rosenberg SA, Yang JC, Restifo NP 2004. Cancer immunotherapy: moving beyond current vaccines. Nat. Med. 10:909–15
    [Google Scholar]
  7. 7.  Hailemichael Y, Dai Z, Jaffarzad N et al. 2013. Persistent antigen at vaccination sites induces tumor-specific CD8+ T cell sequestration, dysfunction and deletion. Nat. Med. 19:465–72
    [Google Scholar]
  8. 8.  Leach DR, Krummel MF, Allison JP 1996. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271:1734–36
    [Google Scholar]
  9. 9.  Hui E, Cheung J, Zhu J et al. 2017. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355:1428–33
    [Google Scholar]
  10. 10.  Parry RV, Chemnitz JM, Frauwirth KA et al. 2005. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 25:9543–53
    [Google Scholar]
  11. 11.  Gordon SR, Maute RL, Dulken BW et al. 2017. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545:495–99
    [Google Scholar]
  12. 12.  Juneja VR, McGuire KA, Manguso RT et al. 2017. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 214:895–904
    [Google Scholar]
  13. 13.  Curran MA, Montalvo W, Yagita H, Allison JP 2010. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. PNAS 107:4275–80
    [Google Scholar]
  14. 14.  Quezada SA, Peggs KS, Curran MA, Allison JP 2006. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J. Clin. Investig. 116:1935–45
    [Google Scholar]
  15. 15.  Ribas A, Wolchok JD 2018. Cancer immunotherapy using checkpoint blockade. Science 359:1350–55
    [Google Scholar]
  16. 16.  Chen R, Zinzani PL, Fanale MA et al. 2017. Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. J. Clin. Oncol. 35:2125–32
    [Google Scholar]
  17. 17.  Le DT, Durham JN, Smith KN et al. 2017. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357:409–13
    [Google Scholar]
  18. 18.  Ferris RL, Blumenschein G Jr, Fayette J et al. 2016. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 375:1856–67
    [Google Scholar]
  19. 19.  Royal RE, Levy C, Turner K et al. 2010. Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 33:828–33
    [Google Scholar]
  20. 20.  Diaz LA Jr, Le DT 2015. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 373:1979
    [Google Scholar]
  21. 21.  Attarwala H 2010. TGN1412: from discovery to disaster. J. Young Pharm. 2:332–36
    [Google Scholar]
  22. 22.  Ai M, Curran MA 2015. Immune checkpoint combinations from mouse to man. Cancer Immunol. Immunother. 64:885–92
    [Google Scholar]
  23. 23.  Croft M, Benedict CA, Ware CF 2013. Clinical targeting of the TNF and TNFR superfamilies. Nat. Rev. Drug Discov. 12:147–68
    [Google Scholar]
  24. 24.  Bartkowiak T, Curran MA 2015. Accelerating cancer immunotherapy through 4–1BB activation. Front. Oncol. 5:117
    [Google Scholar]
  25. 25.  Segal NH, Logan TF, Hodi FS et al. 2016. Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody. Clin. Cancer Res. 23:1929–36
    [Google Scholar]
  26. 26.  Curran MA, Geiger TL, Montalvo W et al. 2013. Systemic 4–1BB activation induces a novel T cell phenotype driven by high expression of Eomesodermin. J. Exp. Med. 210:743–55
    [Google Scholar]
  27. 27.  Kuner R, Vogt M, Sultmann H et al. 2007. Identification of cellular targets for the human papillomavirus E6 and E7 oncogenes by RNA interference and transcriptome analyses. J. Mol. Med. 85:1253–62
    [Google Scholar]
  28. 28.  Hoover AC, Spanos WC, Harris GF et al. 2007. The role of human papillomavirus 16 E6 in anchorage-independent and invasive growth of mouse tonsil epithelium. Arch. Otolaryngol. Head Neck Surg. 133:495–502
    [Google Scholar]
  29. 29.  Lin KY, Guarnieri FG, Staveley-O'Carroll KF et al. 1996. Treatment of established tumors with a novel vaccine that enhances major histocompatibility class II presentation of tumor antigen. Cancer Res 56:21–26
    [Google Scholar]
  30. 30.  Feltkamp MC, Smits HL, Vierboom MP et al. 1993. Vaccination with cytotoxic T lymphocyte epitope-containing peptide protects against a tumor induced by human papillomavirus type 16-transformed cells. Eur. J. Immunol. 23:2242–49
    [Google Scholar]
  31. 31.  Hoffmann C, Stanke J, Kaufmann AM et al. 2010. Combining T-cell vaccination and application of agonistic anti-GITR mAb (DTA-1) induces complete eradication of HPV oncogene expressing tumors in mice. J. Immunother. 33:136–45
    [Google Scholar]
  32. 32.  Shrimali R, Ahmad S, Berrong Z et al. 2017. Agonist anti-GITR antibody significantly enhances the therapeutic efficacy of Listeria monocytogenes-based immunotherapy. J. Immunother. Cancer 5:64
    [Google Scholar]
  33. 33.  Durham NM, Holoweckyj N, MacGill RS et al. 2017. GITR ligand fusion protein agonist enhances the tumor antigen-specific CD8 T-cell response and leads to long-lasting memory. J. Immunother. Cancer 5:47
    [Google Scholar]
  34. 34.  Bartkowiak T, Singh S, Yang G et al. 2015. Unique potential of 4–1BB agonist antibody to promote durable regression of HPV+ tumors when combined with an E6/E7 peptide vaccine. PNAS 112:E5290–99
    [Google Scholar]
  35. 35.  Rice AE, Latchman YE, Balint JP et al. 2015. An HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression. Cancer Gene Ther 22:454–62
    [Google Scholar]
  36. 36.  Dorta-Estremera S, Chin RL, Sierra G et al. 2018. Mucosal HPV E6/E7 peptide vaccination in combination with immune checkpoint modulation induces regression of HPV+ oral cancers. Cancer Res 78:5327–39
    [Google Scholar]
  37. 37.  Shrimali RK, Ahmad S, Verma V et al. 2017. Concurrent PD-1 blockade negates the effects of OX40 agonist antibody in combination immunotherapy through inducing T-cell apoptosis. Cancer Immunol. Res. 5:755–66
    [Google Scholar]
  38. 38.  Curran MA, Allison JP 2009. Tumor vaccines expressing Flt3 ligand synergize with CTLA-4 blockade to reject preimplanted tumors. Cancer Res 69:7747–55
    [Google Scholar]
  39. 39.  van Elsas A, Hurwitz AA, Allison JP 1999. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 190:355–66
    [Google Scholar]
  40. 40.  Hurwitz AA, Foster BA, Kwon ED et al. 2000. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res 60:2444–48
    [Google Scholar]
  41. 41.  Soares KC, Rucki AA, Wu AA et al. 2015. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J. Immunother. 38:1–11
    [Google Scholar]
  42. 42.  Alexandrov LB, Nik-Zainal S, Wedge DC et al. 2013. Signatures of mutational processes in human cancer. Nature 500:415–21
    [Google Scholar]
  43. 43.  Castle JC, Kreiter S, Diekmann J et al. 2012. Exploiting the mutanome for tumor vaccination. Cancer Res 72:1081–91
    [Google Scholar]
  44. 44.  Gubin MM, Zhang X, Schuster H et al. 2014. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515:577–81
    [Google Scholar]
  45. 45.  Kreiter S, Vormehr M, van de Roemer N et al. 2015. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520:692–96
    [Google Scholar]
  46. 46.  Kuai R, Ochyl LJ, Bahjat KS et al. 2017. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 16:489–96
    [Google Scholar]
  47. 47.  Kim TJ, Jin HT, Hur SY et al. 2014. Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients. Nat. Commun. 5:5317
    [Google Scholar]
  48. 48.  Trimble CL, Morrow MP, Kraynyak KA et al. 2015. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet 386:2078–88
    [Google Scholar]
  49. 49.  Bagarazzi ML, Yan J, Morrow MP et al. 2012. Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci. Transl. Med. 4:155ra38
    [Google Scholar]
  50. 50.  Kenter GG, Welters MJ, Valentijn AR et al. 2009. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N. Engl. J. Med. 361:1838–47
    [Google Scholar]
  51. 51.  Massarelli E, William W, Johnson F et al. 2018. Combining immune checkpoint blockade and tumor-specific vaccine: nivolumab and ISA101 in patients with incurable human papillomavirus16-related cancers. JAMA Oncol 4: In press
    [Google Scholar]
  52. 52.  Mehra R, Seiwert TY, Gupta S et al. 2018. Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012. Br. J. Cancer 119:153–59
    [Google Scholar]
  53. 53.  Bauml J, Seiwert TY, Pfister DG et al. 2017. Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study. J. Clin. Oncol. 35:1542–49
    [Google Scholar]
  54. 54.  Brahmer JR, Tykodi SS, Chow LQ 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]
  55. 55.  Clark CE, Hingorani SR, Mick R et al. 2007. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res 67:9518–27
    [Google Scholar]
  56. 56.  Jaffee EM, Hruban RH, Biedrzycki B et al. 2001. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J. Clin. Oncol. 19:145–56
    [Google Scholar]
  57. 57.  Laheru D, Lutz E, Burke J et al. 2008. Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin. Cancer Res. 14:1455–63
    [Google Scholar]
  58. 58.  Hurwitz AA, Foster BA, Kwon ED et al. 2000. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res 60:2444–48
    [Google Scholar]
  59. 59.  Hurwitz AA, Yu TF, Leach DR, Allison JP 1998. CTLA-4 blockade synergizes with tumor-derived granulocyte-macrophage colony-stimulating factor for treatment of an experimental mammary carcinoma. PNAS 95:10067–71
    [Google Scholar]
  60. 60.  Le DT, Lutz E, Uram JN et al. 2013. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J. Immunother. 36:382–89
    [Google Scholar]
  61. 61.  Le DT, Wang-Gillam A, Picozzi V et al. 2015. Safety and survival with GVAX pancreas prime and Listeria monocytogenes–expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J. Clin. Oncol. 33:1325–33
    [Google Scholar]
  62. 62.  Le DT, Ko AH, Wainberg ZA et al. 2017. Results from a phase 2b, randomized, multicenter study of GVAX pancreas and CRS-207 compared to chemotherapy in adults with previously-treated metastatic pancreatic adenocarcinoma (ECLIPSE Study). J. Clin. Oncol. 35:345
    [Google Scholar]
  63. 63.  Hopkins AC, Yarchoan M, Durham JN et al. 2018. T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight 3: pii:122092
    [Google Scholar]
  64. 64.  Robert L, Tsoi J, Wang X et al. 2014. CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin. Cancer Res. 20:2424–32
    [Google Scholar]
  65. 65.  Robert L, Harview C, Emerson R et al. 2014. Distinct immunological mechanisms of CTLA-4 and PD-1 blockade revealed by analyzing TCR usage in blood lymphocytes. Oncoimmunology 3:e29244
    [Google Scholar]
  66. 66.  Beer TM, Kwon ED, Drake CG et al. 2017. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J. Clin. Oncol. 35:40–47
    [Google Scholar]
  67. 67.  Slovin SF, Higano CS, Hamid O et al. 2013. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann. Oncol. 24:1813–21
    [Google Scholar]
  68. 68.  Graff JN, Alumkal JJ, Drake CG et al. 2016. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 7:52810–17
    [Google Scholar]
  69. 69.  Kantoff PW, Higano CS, Shore ND et al. 2010. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 363:411–22
    [Google Scholar]
  70. 70.  van den Eertwegh AJ, Versluis J, van den Berg HP et al. 2012. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 13:509–17
    [Google Scholar]
  71. 71.  Madan RA, Mohebtash M, Arlen PM et al. 2012. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 13:501–8
    [Google Scholar]
  72. 72.  McNeel DG, Eickhoff JC, Wargowski E et al. 2018. Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget 9:25586–96
    [Google Scholar]
  73. 73.  Sahin U, Derhovanessian E, Miller M et al. 2017. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547:222–26
    [Google Scholar]
  74. 74.  Ott PA, Hu Z, Keskin DB et al. 2017. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547:217–21
    [Google Scholar]
  75. 75.  Ott PA, Govindan R, Naing A et al. 2018. Abstract CT125: a personal neoantigen vaccine, NEO-PV-01, with anti-PD1 induces broad de novo anti-tumor immunity in patients with metastatic melanoma, NSCLC, and bladder cancer. Cancer Res 78:CT125
    [Google Scholar]
  76. 76.  Michot JM, Bigenwald C, Champiat S et al. 2016. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur. J. Cancer 54:139–48
    [Google Scholar]
/content/journals/10.1146/annurev-med-050217-121900
Loading
/content/journals/10.1146/annurev-med-050217-121900
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