Skip to main content

Ovarian Cancer Immunology and Immunotherapy

  • Chapter
  • First Online:
Emerging Therapeutic Targets in Ovarian Cancer

Abstract

Epithelial ovarian cancer is a common cancer in women and prognosis from the disease remains poor. Although conventional treatments play a major role in its management, new therapies are required in order to improve disease control and survival. The immune system protects us from developing cancer but once ovarian cancer is established, it employs elaborate pathways to circumvent the immune system. Immune suppression in the form of defective antigen presentation and effector cell anergy are whole marks of ovarian cancer much of which is initiated by tumour cells or by associated suppressive antigen-presenting cells. Recently much research has been carried out to improve our understanding of these suppressive pathways. The balance between T-regulatory cells and effector cytotoxic T-cells remains a key factor in prognosis from this cancer. More recently suppressive monocytes and macrophages have been implicated in T-cell suppression mediated by a number of cell surface molecules such as B7-H1 or enzymes such as arginase. Inflammation may be important in mediating suppression or defective response to treatment. Novel immune therapies have endeavoured to address immune suppression alone or in combination with immune activating treatments such as anti-CTLA-4 antibody therapy in combination with peptide vaccination. Further research is required in order to produce safe and effective immunotherapies to complement chemotherapeutic and surgical treatments.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Shankaran V, et al. (2001). IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410(6832):1107–11.

    Article  PubMed  CAS  Google Scholar 

  2. Coppin C, et al. (2005). Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev 2005, Jan 25;(1):CD001425.

    Google Scholar 

  3. McDermott DF, et al. (2005). Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol 23(1):133–41.

    Article  PubMed  CAS  Google Scholar 

  4. Yang JC, et al. (2003). Randomized study of high-dose and low-dose interleukin-2 in patients with metastatic renal cancer. J Clin Oncol 21(16):3127–32.

    Article  PubMed  CAS  Google Scholar 

  5. Atkins MB, Regan M, McDermott D (2004). Update on the role of interleukin 2 and other cytokines in the treatment of patients with stage IV renal carcinoma. Clin Cancer Res 10(18 Pt 2):6342S–6S.

    Article  PubMed  CAS  Google Scholar 

  6. Ives NJ, et al. (2007). Chemotherapy compared with biochemotherapy for the treatment of metastatic melanoma: a meta-analysis of 18 trials involving 2,621 patients. J Clin Oncol 25(34):5426–34.

    Article  PubMed  CAS  Google Scholar 

  7. Ioannides CG, et al. (1991). Cytotoxic T cell clones isolated from ovarian tumor-infiltrating lymphocytes recognize multiple antigenic epitopes on autologous tumor cells. J Immunol 146(5):1700–7.

    PubMed  CAS  Google Scholar 

  8. Tomsova M, et al. (2008). Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol 108(2):415–20.

    Article  PubMed  CAS  Google Scholar 

  9. Sato E, et al. (2005). 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 102(51):18538–43.

    Article  PubMed  CAS  Google Scholar 

  10. Wolf D, et al. (2005). The expression of the regulatory T cell-specific Forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer. Clin Cancer Res 11(23):8326–31.

    Article  PubMed  CAS  Google Scholar 

  11. Curiel TJ, et al. (2004). Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10(9):942–9.

    Article  PubMed  CAS  Google Scholar 

  12. Zou W (2005). Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 5(4):263–74.

    Article  PubMed  CAS  Google Scholar 

  13. Nelson BH (2008). The impact of T-cell immunity on ovarian cancer outcomes. Immunol Rev 222:101–16.

    Article  PubMed  CAS  Google Scholar 

  14. Zhang L, et al. (2003). Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 348(3):203–13.

    Article  PubMed  CAS  Google Scholar 

  15. Schneiderman D, et al. (1999). Sustained suppression of Fas ligand expression in cisplatin-resistant human ovarian surface epithelial cancer cells. Apoptosis 4(4):271–81.

    PubMed  CAS  Google Scholar 

  16. Yu P, et al. (2006). The role of stroma in immune recognition and destruction of well-established solid tumors. Curr Opin Immunol 18(2):226–31.

    Article  PubMed  CAS  Google Scholar 

  17. Vitale M, et al. (2005). HLA class I antigen down-regulation in primary ovary carcinoma lesions: association with disease stage. Clin Cancer Res 11(1):67–72.

    PubMed  CAS  Google Scholar 

  18. Steinman RM, Banchereau J (2007). Taking dendritic cells into medicine. Nature 449(7161):419–26.

    Article  PubMed  CAS  Google Scholar 

  19. Wei S, et al. (2005). Plasmacytoid dendritic cells induce CD8+ regulatory T cells in human ovarian carcinoma. Cancer Res 65(12):5020–6.

    Article  PubMed  CAS  Google Scholar 

  20. Huarte E, et al. (2008). Depletion of dendritic cells delays ovarian cancer progression by boosting antitumor immunity. Cancer Res 68(18):7684–91.

    Article  PubMed  CAS  Google Scholar 

  21. Melichar B, et al. (1998). Lineage-negative human leukocyte antigen-DR+ cells with the phenotype of undifferentiated dendritic cells in patients with carcinoma of the abdomen and pelvis. Clin Cancer Res 4(3):799–809.

    PubMed  CAS  Google Scholar 

  22. Pan PY, et al. (2009). Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res 70(1):99–108

    Article  PubMed  CAS  Google Scholar 

  23. Rodriguez PC, Ochoa AC (2008). Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev 222:180–91.

    Article  PubMed  CAS  Google Scholar 

  24. Bingle L, Brown NJ, Lewis CE (2002). The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol 196(3):254–65.

    Article  PubMed  CAS  Google Scholar 

  25. Pollard JW (2004). Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer 4(1):71–8.

    Article  PubMed  CAS  Google Scholar 

  26. Hagemann T, Balkwill F, Lawrence T (2007). Inflammation and cancer: a double-edged sword. Cancer Cell 12(4):300–1.

    Article  PubMed  CAS  Google Scholar 

  27. van der Bij GJ, et al. (2005). The role of macrophages in tumor development. Cell Oncol 27(4):203–13.

    PubMed  Google Scholar 

  28. Agarwal R, et al. (2007). Macrophage migration inhibitory factor expression in ovarian cancer. Am J Obstet Gynecol 196(4):348 e1–5.

    Article  PubMed  CAS  Google Scholar 

  29. Duluc D, et al. (2007). Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 110(13):4319–30.

    Article  PubMed  CAS  Google Scholar 

  30. Kacinski BM (1995). CSF-1 and its receptor in ovarian, endometrial and breast cancer. Ann Med 27(1):79–85.

    Article  PubMed  CAS  Google Scholar 

  31. Krockenberger M, et al. (2008). Macrophage migration inhibitory factor contributes to the immune escape of ovarian cancer by down-regulating NKG2D. J Immunol 180(11):7338–48.

    PubMed  CAS  Google Scholar 

  32. Hagemann T, et al. (2006). Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype. J Immunol 176(8):5023–32.

    PubMed  CAS  Google Scholar 

  33. Wyckoff J, et al. (2004). A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res 64(19):7022–9.

    Article  PubMed  CAS  Google Scholar 

  34. Lin EY, et al. (2001). Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med 193(6):727–40.

    Article  PubMed  CAS  Google Scholar 

  35. Nowicki A, et al. (1996). Impaired tumor growth in colony-stimulating factor 1 (CSF-1)-deficient, macrophage-deficient op/op mouse: evidence for a role of CSF-1-dependent macrophages in formation of tumor stroma. Int J Cancer 65(1):112–9.

    Article  PubMed  CAS  Google Scholar 

  36. Huang S, et al. (2002). Contributions of stromal metalloproteinase-9 to angiogenesis and growth of human ovarian carcinoma in mice. J Natl Cancer Inst 94(15):1134–42.

    PubMed  CAS  Google Scholar 

  37. Hagemann T, et al. (2008). “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med 205(6):1261–8.

    Article  PubMed  CAS  Google Scholar 

  38. Hagemann T, et al. (2005). Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. J Immunol 175(2):1197–205.

    PubMed  CAS  Google Scholar 

  39. Kryczek I, et al. (2007). Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma. Cancer Res 67(18):8900–5.

    Article  PubMed  CAS  Google Scholar 

  40. Kulbe H, et al. (2007). The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res 67(2):585–92.

    Article  PubMed  CAS  Google Scholar 

  41. Yang J, et al. (2009). Reciprocal regulation of 17beta-estradiol, interleukin-6 and interleukin-8 during growth and progression of epithelial ovarian cancer. Cytokine 46(3):382–91.

    Article  PubMed  CAS  Google Scholar 

  42. Obata NH, et al. (1997). Effects of interleukin-6 on in vitro cell attachment, migration and invasion of human ovarian carcinoma. Anticancer Res 17(1A):337–42.

    PubMed  CAS  Google Scholar 

  43. Li X, et al. (2007). Human ovarian carcinoma cells generate CD4(+)CD25(+) regulatory T cells from peripheral CD4(+)CD25(−) T cells through secreting TGF-beta. Cancer Lett 253(1):144–53.

    Article  PubMed  CAS  Google Scholar 

  44. Keir ME, Francisco LM, Sharpe AH (2007). PD-1 and its ligands in T-cell immunity. Curr Opin Immunol 19(3):309–14.

    Article  PubMed  CAS  Google Scholar 

  45. Keir ME, Freeman GJ, Sharpe AH (2007). PD-1 regulates self-reactive CD8+ T cell responses to antigen in lymph nodes and tissues. J Immunol 179(8):5064–70.

    PubMed  CAS  Google Scholar 

  46. Hamanishi J, et al. (2007). Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA 104(9):3360–5.

    Article  PubMed  CAS  Google Scholar 

  47. Curiel TJ, et al. (2003). Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 9(5):562–7.

    Article  PubMed  CAS  Google Scholar 

  48. Liu Y, et al. (2008). B7-H1 on myeloid-derived suppressor cells in immune suppression by a mouse model of ovarian cancer. Clin Immunol 129(3):471–81.

    Article  PubMed  CAS  Google Scholar 

  49. Dong H, et al. (2002). Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 8(8):793–800.

    PubMed  CAS  Google Scholar 

  50. Wang L, et al. (2008). Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells. Proc Natl Acad Sci USA 105(27):9331–6.

    Article  PubMed  Google Scholar 

  51. Liu Y, et al. (2009). Regulation of arginase I activity and expression by both PD-1 and CTLA-4 on the myeloid-derived suppressor cells. Cancer Immunol Immunother 58(5):687–97.

    Article  PubMed  CAS  Google Scholar 

  52. Yang YF, et al. (1997). Enhanced induction of antitumor T-cell responses by cytotoxic T lymphocyte-associated molecule-4 blockade: the effect is manifested only at the restricted tumor-bearing stages. Cancer Res 57(18):4036–41.

    PubMed  CAS  Google Scholar 

  53. Weber J (2007). Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events. Oncologist 12(7):864–72.

    Article  PubMed  CAS  Google Scholar 

  54. Patel S, Chiplunkar S (2009). Host immune responses to cervical cancer. Curr Opin Obstet Gynecol 21(1):54–9.

    Article  PubMed  Google Scholar 

  55. Yi HJ, et al. (2008). Defect in TCR-CD3zeta signaling mediates T cell hypo-responsiveness in mesenteric lymph node. Mol Immunol 45(14):3748–55.

    Article  PubMed  CAS  Google Scholar 

  56. Macian F, et al. (2004). T-cell anergy. Curr Opin Immunol 16(2):209–16.

    Article  PubMed  CAS  Google Scholar 

  57. Nagaraj S, Gabrilovich DI (2008). Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Res 68(8):2561–3.

    Article  PubMed  CAS  Google Scholar 

  58. Hanson EM, et al. (2009). Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J Immunol 183(2):937–44.

    Article  PubMed  CAS  Google Scholar 

  59. Nash MA, et al. (1999). The role of cytokines in both the normal and malignant ovary. Endocr Relat Cancer 6(1):93–107.

    Article  PubMed  CAS  Google Scholar 

  60. Giuntoli RL 2nd, et al. (2009). Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res 29(8):2875–84.

    PubMed  CAS  Google Scholar 

  61. Brandenburg S, et al. (2008). IL-2 induces in vivo suppression by CD4(+)CD25(+)Foxp3(+) regulatory T cells. Eur J Immunol 38(6):1643–53.

    Article  PubMed  CAS  Google Scholar 

  62. Campbell JD, et al. (2001). Suppression of IL-2-induced T cell proliferation and phosphorylation of STAT3 and STAT5 by tumor-derived TGF beta is reversed by IL-15. J Immunol 167(1):553–61.

    PubMed  CAS  Google Scholar 

  63. Charles KA, et al. (2009). The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest 119(10):3011–23.

    Article  PubMed  CAS  Google Scholar 

  64. Brown ER, et al. (2008). A clinical study assessing the tolerability and biological effects of infliximab, a TNF-alpha inhibitor, in patients with advanced cancer. Ann Oncol 19(7):1340–6.

    Article  PubMed  CAS  Google Scholar 

  65. Nelson BH (2009). IDO and outcomes in ovarian cancer. Gynecol Oncol 115(2):179–80.

    Article  PubMed  Google Scholar 

  66. Inaba T, et al. (2009). Role of the immunosuppressive enzyme indoleamine 2,3-dioxygenase in the progression of ovarian carcinoma. Gynecol Oncol 115(2):185–92.

    Article  PubMed  CAS  Google Scholar 

  67. Lob S, Konigsrainer A (2008). Is IDO a key enzyme bridging the gap between tumor escape and tolerance induction? Langenbecks Arch Surg 393(6):995–1003.

    Article  PubMed  Google Scholar 

  68. Baniyash M (2004). TCR zeta-chain downregulation: curtailing an excessive inflammatory immune response. Nat Rev Immunol 4(9):675–87.

    Article  PubMed  CAS  Google Scholar 

  69. Bansal V, Ochoa JB (2003). Arginine availability, arginase, and the immune response. Curr Opin Clin Nutr Metab Care 6(2):223–8.

    Article  PubMed  CAS  Google Scholar 

  70. Kropf P, et al. (2007). Arginase activity mediates reversible T cell hyporesponsiveness in human pregnancy. Eur J Immunol 37(4):935–45.

    Article  PubMed  CAS  Google Scholar 

  71. Munder M, et al. (2006). Suppression of T-cell functions by human granulocyte arginase. Blood 108(5):1627–34.

    Article  PubMed  CAS  Google Scholar 

  72. Rodriguez PC, et al. (2004). Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res 64(16):5839–49.

    Article  PubMed  CAS  Google Scholar 

  73. Taylor DD, et al. (2009). Characterization of humoral responses of ovarian cancer patients: antibody subclasses and antigenic components. Gynecol Oncol 116(2):213–21.

    Article  PubMed  CAS  Google Scholar 

  74. Williamson NA, Rossjohn J, Purcell AW (2006). Tumors reveal their secrets to cytotoxic T cells. Proc Natl Acad Sci USA 103(40):14649–50.

    Article  PubMed  CAS  Google Scholar 

  75. Disis ML, et al. (2004). Humoral epitope-spreading following immunization with a HER-2/neu peptide based vaccine in cancer patients. J Clin Immunol 24(5):571–8.

    Article  PubMed  CAS  Google Scholar 

  76. Disis ML, et al. (2002). Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol 20(11):2624–32.

    Article  PubMed  CAS  Google Scholar 

  77. Vlad AM, et al. (2004). MUC1 immunobiology: from discovery to clinical applications. Adv Immunol 82:249–93.

    Article  PubMed  CAS  Google Scholar 

  78. Markert S, et al. (2008). Alpha-folate receptor expression in epithelial ovarian carcinoma and non-neoplastic ovarian tissue. Anticancer Res 28(6A):3567–72.

    PubMed  Google Scholar 

  79. Chang K, Pastan I (1996). Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers. Proc Natl Acad Sci USA 93(1):136–40.

    Article  PubMed  CAS  Google Scholar 

  80. Odunsi K, et al. (2003). NY-ESO-1 and LAGE-1 cancer-testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res 63(18):6076–83.

    PubMed  CAS  Google Scholar 

  81. Sandmaier BM, et al. (1999). Evidence of a cellular immune response against sialyl-Tn in breast and ovarian cancer patients after high-dose chemotherapy, stem cell rescue, and immunization with Theratope STn-KLH cancer vaccine. J Immunother 22(1):54–66.

    Article  PubMed  CAS  Google Scholar 

  82. Behrens MD, et al. (2008). The endogenous danger signal, crystalline uric acid, signals for enhanced antibody immunity. Blood 111(3):1472–9.

    Article  PubMed  CAS  Google Scholar 

  83. Salazar LG, et al. (2007). Kinetics of tumor-specific T-cell response development after active immunization in patients with HER-2/neu overexpressing cancers. Clin Immunol 125(3):275–80.

    Article  PubMed  CAS  Google Scholar 

  84. Bookman MA, et al. (2003). Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J Clin Oncol 21(2):283–90.

    Article  PubMed  CAS  Google Scholar 

  85. Seiden MV, et al. (2007). A phase II trial of EMD72000 (matuzumab), a humanized anti-EGFR monoclonal antibody, in patients with platinum-resistant ovarian and primary peritoneal malignancies. Gynecol Oncol 104(3):727–31.

    Article  PubMed  CAS  Google Scholar 

  86. Burger RA (2007). Experience with bevacizumab in the management of epithelial ovarian cancer. J Clin Oncol 25(20):2902–8.

    Article  PubMed  CAS  Google Scholar 

  87. Burger RA, et al. (2007). Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group Study. J Clin Oncol 25(33):5165–71.

    Article  PubMed  CAS  Google Scholar 

  88. Micha JP, et al. (2007). A phase II study of outpatient first-line paclitaxel, carboplatin, and bevacizumab for advanced-stage epithelial ovarian, peritoneal, and fallopian tube cancer. Int J Gynecol Cancer 17(4):771–6.

    Article  PubMed  CAS  Google Scholar 

  89. Schultes BC, et al. (1999). Immunotherapy of human ovarian carcinoma with OvaRex MAb-B43.13 in a human-PBL-SCID/BG mouse model. Hybridoma 18(1):47–55.

    Article  PubMed  CAS  Google Scholar 

  90. Ehlen TG, et al. (2005). A pilot phase 2 study of oregovomab murine monoclonal antibody to CA125 as an immunotherapeutic agent for recurrent ovarian cancer. Int J Gynecol Cancer 15(6):1023–34.

    Article  PubMed  CAS  Google Scholar 

  91. Berek JS, et al. (2004). Randomized, placebo-controlled study of oregovomab for consolidation of clinical remission in patients with advanced ovarian cancer. J Clin Oncol 22(17):3507–16.

    Article  PubMed  CAS  Google Scholar 

  92. Berek JS, Taylor PT, Nicodemus CF (2008). CA125 velocity at relapse is a highly significant predictor of survival post relapse: results of a 5-year follow-up survey to a randomized placebo-controlled study of maintenance oregovomab immunotherapy in advanced ovarian cancer. J Immunother 31(2):207–14.

    Article  PubMed  Google Scholar 

  93. Gordon AN, et al. (2004). CA125- and tumor-specific T-cell responses correlate with prolonged survival in oregovomab-treated recurrent ovarian cancer patients. Gynecol Oncol 94(2):340–51.

    Article  PubMed  CAS  Google Scholar 

  94. Wagner U, et al. (2001). Immunological consolidation of ovarian carcinoma recurrences with monoclonal anti-idiotype antibody ACA125: immune responses and survival in palliative treatment. See The biology behind: K. A. Foon and M. Bhattacharya-Chatterjee, Are solid tumor anti-idiotype vaccines ready for prime time? Clin Cancer Res 7:1112–1115, 2001. Clin Cancer Res 7(5):1154–62.

    PubMed  CAS  Google Scholar 

  95. Reinartz S, et al. (2004). Vaccination of patients with advanced ovarian carcinoma with the anti-idiotype ACA125: immunological response and survival (phase Ib/II). Clin Cancer Res 10(5):1580–7.

    Article  PubMed  CAS  Google Scholar 

  96. van Zanten-Przybysz I, et al. (2002). Cellular and humoral responses after multiple injections of unconjugated chimeric monoclonal antibody MOv18 in ovarian cancer patients: a pilot study. J Cancer Res Clin Oncol 128(9):484–92.

    Article  PubMed  CAS  Google Scholar 

  97. Canevari S, et al. (1995). Regression of advanced ovarian carcinoma by intraperitoneal treatment with autologous T lymphocytes retargeted by a bispecific monoclonal antibody. J Natl Cancer Inst 87(19):1463–9.

    Article  PubMed  CAS  Google Scholar 

  98. Lamers CH, et al. (1997). Local but no systemic immunomodulation by intraperitoneal treatment of advanced ovarian cancer with autologous T lymphocytes re-targeted by a bi-specific monoclonal antibody. Int J Cancer 73(2):211–9.

    Article  PubMed  CAS  Google Scholar 

  99. Agus DB, et al. (2005). Phase I clinical study of pertuzumab, a novel HER dimerization inhibitor, in patients with advanced cancer. J Clin Oncol 23(11):2534–43.

    Article  PubMed  CAS  Google Scholar 

  100. Epenetos AA, et al. (2000). Long term survival of patients with advanced ovarian cancer treated with intraperitoneal radioimmunotherapy. Int J Gynecol Cancer 10(S1):44–46.

    Article  PubMed  Google Scholar 

  101. Nicholson S, et al. (1998). Radioimmunotherapy after chemotherapy compared to chemotherapy alone in the treatment of advanced ovarian cancer: a matched analysis. Oncol Rep 5(1):223–6.

    PubMed  CAS  Google Scholar 

  102. Nicholson S, et al. (2000). A randomised phase III trial of adjuvant intraperitoneal radioimmunotherapy in ovarian cancer. J Clin Oncol (1):(suppl 1), ASCO, Abstract 1514.

    Google Scholar 

  103. Verheijen RH, et al. (2006). Phase III trial of intraperitoneal therapy with yttrium-90-labeled HMFG1 murine monoclonal antibody in patients with epithelial ovarian cancer after a surgically defined complete remission. J Clin Oncol 24(4):571–8.

    Article  PubMed  CAS  Google Scholar 

  104. Chianese-Bullock KA, et al. (2008). A multipeptide vaccine is safe and elicits T-cell responses in participants with advanced stage ovarian cancer. J Immunother 31(4):420–30.

    Article  PubMed  CAS  Google Scholar 

  105. Odunsi K, et al. (2007). 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 104(31):12837–42.

    Article  PubMed  CAS  Google Scholar 

  106. Qian F, et al. (2004). Th1/Th2 CD4+ T cell responses against NY-ESO-1 in HLA-DPB1*0401/0402 patients with epithelial ovarian cancer. Cancer Immun 4:12.

    PubMed  Google Scholar 

  107. Jager E, et al. (2000). Induction of primary NY-ESO-1 immunity: CD8+ T lymphocyte and antibody responses in peptide-vaccinated patients with NY-ESO-1+ cancers. Proc Natl Acad Sci USA 97(22):12198–203.

    Article  PubMed  CAS  Google Scholar 

  108. Leffers N, et al. (2010). Antigen-specific active immunotherapy for ovarian cancer. Cochrane Database Syst Rev 2010, Jan 20;(1):CD007287.

    Google Scholar 

  109. Brossart P, et al. (2000). Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood 96(9):3102–8.

    PubMed  CAS  Google Scholar 

  110. Gong J, et al. (2000). Fusions of human ovarian carcinoma cells with autologous or allogeneic dendritic cells induce antitumor immunity. J Immunol 165(3):1705–11.

    PubMed  CAS  Google Scholar 

  111. Hernando JJ, et al. (2002). Vaccination with autologous tumour antigen-pulsed dendritic cells in advanced gynaecological malignancies: clinical and immunological evaluation of a phase I trial. Cancer Immunol Immunother 51(1):45–52.

    Article  PubMed  CAS  Google Scholar 

  112. Dobrzanski MJ, et al. (2009). Autologous MUC1-specific Th1 effector cell immunotherapy induces differential levels of systemic TReg cell subpopulations that result in increased ovarian cancer patient survival. Clin Immunol 133(3):333–52.

    Article  PubMed  CAS  Google Scholar 

  113. Govers C, et al. (2010). T cell receptor gene therapy: strategies for optimizing transgenic TCR pairing. Trends Mol Med 16(2):77–87.

    Article  PubMed  CAS  Google Scholar 

  114. Maher J, et al. (2002). Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor. Nat Biotechnol 20(1):70–5.

    Article  PubMed  CAS  Google Scholar 

  115. Maher J, Wilkie S (2009). CAR mechanics: driving T cells into the MUC of cancer. Cancer Res 69(11):4559–62.

    Article  PubMed  CAS  Google Scholar 

  116. Barber A, et al. (2007). Chimeric NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. Cancer Res 67(10):5003–8.

    Article  PubMed  CAS  Google Scholar 

  117. Carpenito C, et al. (2009). Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci USA 106(9):3360–5.

    Article  PubMed  Google Scholar 

  118. Litzinger MT, et al. (2007). IL-2 immunotoxin denileukin diftitox reduces regulatory T cells and enhances vaccine-mediated T-cell immunity. Blood 110(9):3192–201.

    Article  PubMed  CAS  Google Scholar 

  119. Hodi FS, et al. (2003). Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA 100(8):4712–7.

    Article  PubMed  CAS  Google Scholar 

  120. Phan GQ, Weber JS, Sondak VK (2008). CTLA-4 blockade with monoclonal antibodies in patients with metastatic cancer: surgical issues. Ann Surg Oncol 15(11):3014–21.

    Article  PubMed  Google Scholar 

  121. ten Berge RJ, et al. (1984). Combination chemotherapy and immune capacity in advanced ovarian carcinoma. Eur J Cancer Clin Oncol 20(1):91–8.

    Article  PubMed  Google Scholar 

  122. Tsuda N, et al. (2007). Taxol increases the amount and T cell activating ability of self-immune stimulatory multimolecular complexes found in ovarian cancer cells. Cancer Res 67(17):8378–87.

    Article  PubMed  CAS  Google Scholar 

  123. Mitchell MS, Kohorn EI (1976). Cell-mediated immunity and blocking factor in ovarian carcinoma. Obstet Gynecol 48(5):590–7.

    PubMed  CAS  Google Scholar 

  124. Muranski P, et al. (2006). Increased intensity lymphodepletion and adoptive immunotherapy – how far can we go? Nat Clin Pract Oncol 3(12):668–81.

    Article  PubMed  CAS  Google Scholar 

  125. Thistlethwaite FC, et al. (2008). Adoptive transfer of T(reg) depleted autologous T cells in advanced renal cell carcinoma. Cancer Immunol Immunother 57(5):623–34.

    Article  PubMed  Google Scholar 

  126. Larmonier N, et al. (2008). Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCR-ABL− tumors. J Immunol 181(10):6955–63.

    PubMed  CAS  Google Scholar 

  127. Brownlow N, et al. (2009). Dasatinib is a potent inhibitor of tumour-associated macrophages, osteoclasts and the FMS receptor. Leukemia 23(3):590–4.

    Article  PubMed  CAS  Google Scholar 

  128. Knutson KL, et al. (2003). Immunologic principles and immunotherapeutic approaches in ovarian cancer. Hematol Oncol Clin North Am 17(4):1051–73.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sadaf Ghaem-Maghami .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2011 Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Ghaem-Maghami, S., Gore, M. (2011). Ovarian Cancer Immunology and Immunotherapy. In: Kaye, S., Brown, R., Gabra, H., Gore, M. (eds) Emerging Therapeutic Targets in Ovarian Cancer. Springer, New York, NY. https://doi.org/10.1007/978-1-4419-7216-3_10

Download citation

Publish with us

Policies and ethics