Immunotherapy in ovarian cancer

K Odunsi, K Odunsi

Abstract

Immunological destruction of tumors is a multistep, coordinated process that can be modulated or targeted at several critical points to elicit tumor rejection. These steps in the cancer immunity cycle include: (i) generation of sufficient numbers of effector T cells with high avidity recognition of tumor antigens in vivo; (ii) trafficking and infiltration into the tumor; (iii) overcoming inhibitory networks in the tumor microenvironment; (iv) direct recognition of tumor antigens and generation of an effector anti-tumor response; and (v) persistence of the anti-tumor T cells. In an effort to understand whether the immune system plays a role in controlling ovarian cancer, our group and others demonstrated that the presence of tumor infiltrating lymphocytes (TILs) is associated with improved clinical outcome in ovarian cancer patients. Recently, we hypothesized that the quality of infiltrating T cells could also be a critical determinant of outcome in ovarian cancer patients. In the past decade, several immune-based interventions have gained regulatory approval in many solid tumors and hematologic malignancies. These interventions include immune checkpoint blockade, cancer vaccines, and adoptive cell therapy. There are currently no approved immune therapies for ovarian cancer. Immunotherapy in ovarian cancer will have to consider the immune suppressive networks within the ovarian tumor microenvironment; therefore, a major direction is to develop biomarkers that would predict responsiveness to different types of immunotherapies, and allow for treatment selection based on the results. Moreover, such biomarkers would allow rational combination of immunotherapies, while minimizing toxicities. In this review, the current understanding of the host immune response in ovarian cancer patients will be briefly reviewed, progress in immune therapies, and future directions for exploiting immune based strategies for long lasting durable cure.

Keywords: NY-ESO-1; T cells; adoptive cell therapy; cancer vaccines; immunomodulation; immunotherapy; tumor antigens.

© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For Permissions, please email: journals.permissions@oup.com.

References

    1. Bray F, Ren JS, Masuyer E, Ferlay J.. Global estimates of cancer prevalence for 27 sites in the adult population in 2008. Int J Cancer 2013; 132: 1133–1145.
    1. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J. et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur Cancer 2013; 49: 1374–1403.
    1. Siegel R, Naishadham D, Jemal A.. Cancer statistics, 2013. Ca Cancer J Clin 2013; 63(1): 11–30.
    1. Sato E, Olson SH, Ahn J. et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 2005; 102(51): 18538–18543.
    1. Zhang L, Conejo-Garcia JR, Katsaros D. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003; 348(3): 203–213.
    1. Hwang WT, Adams SF, Tahirovic E. et al. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol 2012; 124(2): 192–198.
    1. Dunn GP, Bruce AT, Ikeda H. et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002; 3(11): 991–998.
    1. Dunn GP, Old LJ, Schreiber RD.. The three Es of cancer immunoediting. Annu Rev Immunol 2004; 22: 329–360.
    1. Dunn GP, Old LJ, Schreiber RD.. The immunobiology of cancer immunosurveillance and immunoediting. Immunity 2004; 21(2): 137–148.
    1. Nishikawa H, Jager E, Ritter G. et al. CD4+ CD25+ regulatory T cells control the induction of antigen-specific CD4+ helper T cell responses in cancer patients. Blood 2005; 106(3): 1008–1011.
    1. Qian F, Villella J, Wallace PK. et al. Efficacy of levo-1-methyl tryptophan and dextro-1-methyl tryptophan in reversing indoleamine-2,3-dioxygenase-mediated arrest of T-cell proliferation in human epithelial ovarian cancer. Cancer Res 2009; 69(13): 5498–5504.
    1. Qian F, Liao J, Villella J. et al. Effects of 1-methyltryptophan stereoisomers on IDO2 enzyme activity and IDO2-mediated arrest of human T cell proliferation. Cancer Immunol Immunother 2012. April 25 [epub ahead of print], doi:10.1007/s00262-012-1265-x.
    1. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P. et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA 2010; 107(17): 7875–7880.
    1. Huang RY, Eppolito C, Lele S. et al. LAG3 and PD1 co-inhibitory molecules collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a murine ovarian cancer model. Oncotarget 2015; 6(29): 27359–27377.
    1. Huang R, Francois A, McGray AJR. et al. Compensatory upregulation of PD-1, LAG-3 and CTLA-4 limits the efficacy of single agent checkpoint blockade in metastatic ovarian cancer. OncoImmunology 2017, .
    1. Odunsi K, Qian F, Matsuzaki J. et al. Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci USA 2007; 104(31): 12837–12842.
    1. Khan AN, Kolomeyevskaya N, Singel KL. et al. Targeting myeloid cells in the tumor microenvironment enhances vaccine efficacy in murine epithelial ovarian cancer. Oncotarget 2015; 6(13): 11310–11326.
    1. Brahmer JR, Tykodi SS, Chow LQ. et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012; 366(26): 2455–2465.
    1. Hamanishi J, Mandai M, Ikeda T. et al. Safety and antitumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer. J Clin Oncol 2015; 33(34): 4015–4022.
    1. Disis ML, Patel MR, Pant S. et al. Avelumab (MSB0010718C), an anti-PD-L1 antibody, in patients with previously treated, recurrent or refractory ovarian cancer: a phase Ib, open-label expansion trial. J Clin Oncol 2015; 33(Suppl): 5509.
    1. Varga A, Piha-Paul SA, Ott PA. et al. Antitumor activity and safety of pembrolizumab in patients (pts) with PD-L1 positive advanced ovarian cancer: Interim results from a phase Ib study. J Clin Oncol 2015; 33(Suppl): 5510.
    1. Tureci O, Sahin U, Zwick C. et al. Identification of a meiosis-specific protein as a member of the class of cancer/testis antigens. Proc Natl Acad Sci USA 1998; 95(9): 5211–5216.
    1. Old LJ. Cancer/testis (CT) antigens - a new link between gametogenesis and cancer. Cancer Immun 2001; 1: 1.
    1. Simpson AJ, Caballero OL, Jungbluth A. et al. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 2005; 5(8): 615–625.
    1. Odunsi K, Jungbluth AA, Stockert E. et al. NY-ESO-1 and LAGE-1 cancer-testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res 2003; 63(18): 6076–6083.
    1. Szender JB, Papanicolau-Sengos A, Eng KH. et al. NY-ESO-1 expression predicts an aggressive phenotype of ovarian cancer. Gynecol Oncol 2017; 145(3): 420–425.
    1. Tsuji T, Sabbatini P, Jungbluth AA. et al. Effect of montanide and poly-ICLC adjuvant on human self/tumor antigen-specific CD4+ T cells in phase I overlapping long peptide vaccine trial. Cancer Immunol Res 2013; 1(5): 340–350.
    1. Odunsi K, Matsuzaki J, Karbach J. et al. Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients. Proc Natl Acad Sci USA 2012; 109(15): 5797–5802.
    1. Odunsi K, Matsuzaki J, James SR. et al. Epigenetic potentiation of NY-ESO-1 vaccine therapy in human ovarian cancer. Cancer Immunol Res 2014; 2(1): 37–49.
    1. Dudley ME, Gross CA, Somerville RP. et al. Randomized selection design trial evaluating CD8+-enriched versus unselected tumor-infiltrating lymphocytes for adoptive cell therapy for patients with melanoma. J Clin Oncol 2013; 31(17): 2152–2159.
    1. Tumeh PC, Harview CL, Yearley JH. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014; 515(7528): 568–571.
    1. Castle JC, Kreiter S, Diekmann J. et al. Exploiting the mutanome for tumor vaccination. Cancer Res 2012; 72(5): 1081–1091.
    1. Duan F, Duitama J, Al Seesi S. et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J Exp Med 2014; 211(11): 2231–2248.
    1. Gubin MM, Zhang X, Schuster H. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014; 515(7528): 577–581.
    1. Wick DA, Webb JR, Nielsen JS. et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin Cancer Res 2014; 20(5): 1125–1134.
    1. 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(6230): 124–128.
    1. Gerlinger M, Rowan AJ, Horswell S. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012; 366(10): 883–892.
    1. Linnemann C, van Buuren MM, Bies L. et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat Med 2015; 21(1): 81–85.
    1. Lu YC, Yao X, Crystal JS. et al. Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin Cancer Res 2014; 20(13): 3401–3410.
    1. Munn DH. Indoleamine 2,3-dioxygenase, tumor-induced tolerance and counter-regulation. Curr Opin Immunol 2006; 18(2): 220–225.
    1. Fallarino F, Grohmann U, You S. et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J Immunol 2006; 176(11): 6752–6761.
    1. Curiel TJ, Coukos G, Zou L. et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10(9): 942–949.
    1. Bonifaz LC, Bonnyay DP, Charalambous A. et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J Exp Med 2004; 199(6): 815–824.
    1. Bozzacco L, Trumpfheller C, Siegal FP. et al. DEC-205 receptor on dendritic cells mediates presentation of HIV gag protein to CD8+ T cells in a spectrum of human MHC I haplotypes. Proc Natl Acad Sci USA 2007; 104(4): 1289–1294.
    1. Koneru M, O'Cearbhaill R, Pendharkar S. et al. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Transl Med 2015; 13: 102..
    1. Koneru M, Purdon TJ, Spriggs D. et al. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology 2015; 4(3): e994446.
    1. Pegram HJ, Lee JC, Hayman EG. et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 2012; 119(18): 4133–4141.
    1. Pegram HJ, Purdon TJ, van Leeuwen DG. et al. IL-12-secreting CD19-targeted cord blood-derived T cells for the immunotherapy of B-cell acute lymphoblastic leukemia. Leukemia 2015; 29(2): 415–422.
    1. Jackson HJ, Rafiq S, Brentjens RJ.. Driving CAR T-cells forward. Nat Rev Clin Oncol 2016; 13(6): 370–383.
    1. Jaspers JE, Brentjens RJ.. Development of CAR T cells designed to improve antitumor efficacy and safety. Pharmacol Ther 2017; doi:10.1016/j.pharmthera.2017.03.012.
    1. Gschweng EH, McCracken MN, Kaufman ML. et al. HSV-sr39TK positron emission tomography and suicide gene elimination of human hematopoietic stem cells and their progeny in humanized mice. Cancer Res 2014; 74(18): 5173–5183.
    1. Minagawa K, Jamil MO, Al-Obaidi M. et al. In vitro pre-clinical validation of suicide gene modified anti-CD33 redirected chimeric antigen receptor T-cells for acute myeloid leukemia. PLoS One 2017; 12(2): e0172640.
    1. Resetca D, Neschadim A, Medin JA.. Engineering hematopoietic cells for cancer immunotherapy: strategies to address safety and toxicity concerns. J Immunother 2016; 39(7): 249–259.

Source: PubMed

3
S'abonner