Skip to main content
Top
Published in: Cancer and Metastasis Reviews 1/2011

01-03-2011

Tumor immune surveillance and ovarian cancer

Lessons on immune mediated tumor rejection or tolerance

Authors: Lana E. Kandalaft, Gregory T. Motz, Jaikumar Duraiswamy, George Coukos

Published in: Cancer and Metastasis Reviews | Issue 1/2011

Login to get access

Abstract

In the past few years, cancer immunotherapies have produced promising results. Although traditionally considered unresponsive to immune therapy, increasing evidence indicates that ovarian cancers are, in fact, immunogenic tumors. This evidence comes from diverse epidemiologic and clinical data comprising evidence of spontaneous antitumor immune response and its association with longer survival in a proportion of ovarian cancer patients; evidence of tumor immune evasion mechanisms and their association with short survival in some ovarian cancer patients; and finally pilot data supporting the efficacy of immune therapy. Below we will discuss lessons learned on the biology underlying ovarian cancer immune rejection or tolerance and we will discuss its association with clinical outcome. We will discuss the role of angiogenesis and the tumor endothelium on regulation of the antitumor immune response with a special emphasis on the role of vascular endothelial growth factor (VEGF) in the suppression of immunological processes, which control tumor progression and its unique crosstalk with endothelin systems, and how their interactions may shape the antitumor immune response. In addition, we will discuss mechanisms of tumor tolerance through the suppression or exhaustion of effector cells and how these could be countered in the clinic. We believe that understanding these pathways in the tumor microenvironment will lead to novel strategies for enhancing ovarian cancer immunotherapy.
Literature
1.
go back to reference Bukowski, R. M., Ozols, R. F., & Markman, M. (2007). The management of recurrent ovarian cancer. Seminars in Oncology, 34(2 Suppl 2), S1–S15.PubMed Bukowski, R. M., Ozols, R. F., & Markman, M. (2007). The management of recurrent ovarian cancer. Seminars in Oncology, 34(2 Suppl 2), S1–S15.PubMed
2.
go back to reference Ozols, R. F. (2006). Systemic therapy for ovarian cancer: current status and new treatments. Seminars in Oncology, 33(2 Suppl 6), S3–S11.PubMed Ozols, R. F. (2006). Systemic therapy for ovarian cancer: current status and new treatments. Seminars in Oncology, 33(2 Suppl 6), S3–S11.PubMed
3.
go back to reference Gajewski, T. F. (2006). Identifying and overcoming immune resistance mechanisms in the melanoma tumor microenvironment. Clinical Cancer Research, 12(7 Pt 2), 2326s–2330s.PubMed Gajewski, T. F. (2006). Identifying and overcoming immune resistance mechanisms in the melanoma tumor microenvironment. Clinical Cancer Research, 12(7 Pt 2), 2326s–2330s.PubMed
4.
go back to reference Gajewski, T. F., et al. (2006). Immune resistance orchestrated by the tumor microenvironment. Immunological Reviews, 213, 131–145.PubMed Gajewski, T. F., et al. (2006). Immune resistance orchestrated by the tumor microenvironment. Immunological Reviews, 213, 131–145.PubMed
5.
go back to reference Yigit, R., et al. (2010). Ovarian cancer creates a suppressive microenvironment to escape immune elimination. Gynecologic Oncology, 117(2), 366–372.PubMed Yigit, R., et al. (2010). Ovarian cancer creates a suppressive microenvironment to escape immune elimination. Gynecologic Oncology, 117(2), 366–372.PubMed
6.
go back to reference Nagaraj, S., & Gabrilovich, D. I. (2008). Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Research, 68(8), 2561–2563.PubMed Nagaraj, S., & Gabrilovich, D. I. (2008). Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Research, 68(8), 2561–2563.PubMed
7.
go back to reference Hamanishi, J., et al. (2007). Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proceedings of the National Academy of Sciences of the United States of America, 104(9), 3360–3365.PubMed Hamanishi, J., et al. (2007). Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proceedings of the National Academy of Sciences of the United States of America, 104(9), 3360–3365.PubMed
8.
go back to reference Schlienger, K., et al. (2003). TRANCE- and CD40 ligand-matured dendritic cells reveal MHC class I-restricted T cells specific for autologous tumor in late-stage ovarian cancer patients. Clinical Cancer Research, 9(4), 1517–1527.PubMed Schlienger, K., et al. (2003). TRANCE- and CD40 ligand-matured dendritic cells reveal MHC class I-restricted T cells specific for autologous tumor in late-stage ovarian cancer patients. Clinical Cancer Research, 9(4), 1517–1527.PubMed
9.
go back to reference Zhang, L., et al. (2003). Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. The New England Journal of Medicine, 348(3), 203–213.PubMed Zhang, L., et al. (2003). Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. The New England Journal of Medicine, 348(3), 203–213.PubMed
10.
go back to reference Adams, S. F., et al. (2009). Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer, 115(13), 2891–2902.PubMed Adams, S. F., et al. (2009). Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer, 115(13), 2891–2902.PubMed
11.
go back to reference Clarke, B., et al. (2008). Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type and BRCA1 loss. Modern Pathology (in press) Clarke, B., et al. (2008). Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type and BRCA1 loss. Modern Pathology (in press)
12.
go back to reference 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. Proceedings of the National Academy of Sciences of the United States of America, 102(51), 18538–18543.PubMed 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. Proceedings of the National Academy of Sciences of the United States of America, 102(51), 18538–18543.PubMed
13.
go back to reference Shah, C. A., et al. (2008). Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer. Gynecol Oncol. Shah, C. A., et al. (2008). Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer. Gynecol Oncol.
14.
go back to reference Tomsova, M., et al. (2008). Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecologic Oncology, 108(2), 415–420.PubMed Tomsova, M., et al. (2008). Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecologic Oncology, 108(2), 415–420.PubMed
15.
go back to reference Stumpf, M., et al. (2009). Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes. British Journal of Cancer, 101(9), 1513–1521.PubMed Stumpf, M., et al. (2009). Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes. British Journal of Cancer, 101(9), 1513–1521.PubMed
16.
go back to reference Milne, K., et al. (2009). Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS ONE, 4(7), e6412.PubMed Milne, K., et al. (2009). Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS ONE, 4(7), e6412.PubMed
17.
go back to reference Albert, M. L., et al. (1998). Tumor-specific killer cells in paraneoplastic cerebellar degeneration. Natural Medicines, 4(11), 1321–1324. Albert, M. L., et al. (1998). Tumor-specific killer cells in paraneoplastic cerebellar degeneration. Natural Medicines, 4(11), 1321–1324.
18.
go back to reference Chiriva-Internati, M., et al. (2010). Cancer testis antigen vaccination affords long-term protection in a murine model of ovarian cancer. PLoS ONE, 5(5), e10471.PubMed Chiriva-Internati, M., et al. (2010). Cancer testis antigen vaccination affords long-term protection in a murine model of ovarian cancer. PLoS ONE, 5(5), e10471.PubMed
19.
go back to reference Coukos, G., et al. (2005). Immunotherapy for gynaecological malignancies. Expert Opinion on Biological Therapy, 5(9), 1193–1210.PubMed Coukos, G., et al. (2005). Immunotherapy for gynaecological malignancies. Expert Opinion on Biological Therapy, 5(9), 1193–1210.PubMed
20.
go back to reference Gordan, J. D., & Vonderheide, R. H. (2002). Universal tumor antigens as targets for immunotherapy. Cytotherapy, 4(4), 317–327.PubMed Gordan, J. D., & Vonderheide, R. H. (2002). Universal tumor antigens as targets for immunotherapy. Cytotherapy, 4(4), 317–327.PubMed
21.
go back to reference Vonderheide, R. H., et al. (1999). The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity, 10(6), 673–679.PubMed Vonderheide, R. H., et al. (1999). The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity, 10(6), 673–679.PubMed
22.
go back to reference Counter, C. M., et al. (1994). Telomerase activity in human ovarian carcinoma. Proceedings of the National Academy of Sciences of the United States of America, 91(8), 2900–2904.PubMed Counter, C. M., et al. (1994). Telomerase activity in human ovarian carcinoma. Proceedings of the National Academy of Sciences of the United States of America, 91(8), 2900–2904.PubMed
23.
go back to reference Freedman, R. S., & Platsoucas, C. D. (1996). Immunotherapy for peritoneal ovarian carcinoma metastasis using ex vivo expanded tumor infiltrating lymphocytes. Cancer Treatment and Research, 82, 115–146.PubMed Freedman, R. S., & Platsoucas, C. D. (1996). Immunotherapy for peritoneal ovarian carcinoma metastasis using ex vivo expanded tumor infiltrating lymphocytes. Cancer Treatment and Research, 82, 115–146.PubMed
24.
go back to reference Pertl, U., et al. (2001). IFN-gamma-inducible protein-10 is essential for the generation of a protective tumor-specific CD8 T cell response induced by single-chain IL-12 gene therapy. Journal of Immunology, 166(11), 6944–6951. Pertl, U., et al. (2001). IFN-gamma-inducible protein-10 is essential for the generation of a protective tumor-specific CD8 T cell response induced by single-chain IL-12 gene therapy. Journal of Immunology, 166(11), 6944–6951.
25.
go back to reference Nehme, A., et al. (1994). Modulation of cisplatin cytotoxicity by human recombinant interferon-gamma in human ovarian cancer cell lines. European Journal of Cancer, 30A(4), 520–525.PubMed Nehme, A., et al. (1994). Modulation of cisplatin cytotoxicity by human recombinant interferon-gamma in human ovarian cancer cell lines. European Journal of Cancer, 30A(4), 520–525.PubMed
26.
go back to reference Melichar, B., et al. (2003). rIFN-gamma-mediated growth suppression of platinum-sensitive and -resistant ovarian tumor cell lines not dependent upon arginase inhibition. Journal of Translational Medicine, 1(1), 5.PubMed Melichar, B., et al. (2003). rIFN-gamma-mediated growth suppression of platinum-sensitive and -resistant ovarian tumor cell lines not dependent upon arginase inhibition. Journal of Translational Medicine, 1(1), 5.PubMed
27.
go back to reference Wall, L., et al. (2003). The anti-proliferative activity of interferon-gamma on ovarian cancer: in vitro and in vivo. Gynecologic Oncology, 88(1 Pt 2), S149–S151.PubMed Wall, L., et al. (2003). The anti-proliferative activity of interferon-gamma on ovarian cancer: in vitro and in vivo. Gynecologic Oncology, 88(1 Pt 2), S149–S151.PubMed
28.
go back to reference Freedman, R. S., et al. (2000). Clinical and biological effects of intraperitoneal injections of recombinant interferon-gamma and recombinant interleukin 2 with or without tumor-infiltrating lymphocytes in patients with ovarian or peritoneal carcinoma. Clinical Cancer Research, 6(6), 2268–2278.PubMed Freedman, R. S., et al. (2000). Clinical and biological effects of intraperitoneal injections of recombinant interferon-gamma and recombinant interleukin 2 with or without tumor-infiltrating lymphocytes in patients with ovarian or peritoneal carcinoma. Clinical Cancer Research, 6(6), 2268–2278.PubMed
29.
go back to reference Kooi, S., et al. (1996). HLA class I expression on human ovarian carcinoma cells correlates with T-cell infiltration in vivo and T-cell expansion in vitro in low concentrations of recombinant interleukin-2. Cellular Immunology, 174(2), 116–128.PubMed Kooi, S., et al. (1996). HLA class I expression on human ovarian carcinoma cells correlates with T-cell infiltration in vivo and T-cell expansion in vitro in low concentrations of recombinant interleukin-2. Cellular Immunology, 174(2), 116–128.PubMed
30.
go back to reference Coughlin, C. M., et al. (1998). Tumor cell responses to IFNgamma affect tumorigenicity and response to IL-12 therapy and antiangiogenesis. Immunity, 9(1), 25–34.PubMed Coughlin, C. M., et al. (1998). Tumor cell responses to IFNgamma affect tumorigenicity and response to IL-12 therapy and antiangiogenesis. Immunity, 9(1), 25–34.PubMed
31.
go back to reference Maheshwari, R. K., et al. (1991). Differential effects of interferon gamma and alpha on in vitro model of angiogenesis. Journal of Cellular Physiology, 146(1), 164–169.PubMed Maheshwari, R. K., et al. (1991). Differential effects of interferon gamma and alpha on in vitro model of angiogenesis. Journal of Cellular Physiology, 146(1), 164–169.PubMed
32.
go back to reference Sidky, Y. A., & Borden, E. C. (1987). Inhibition of angiogenesis by interferons: effects on tumor- and lymphocyte-induced vascular responses. Cancer Research, 47(19), 5155–5161.PubMed Sidky, Y. A., & Borden, E. C. (1987). Inhibition of angiogenesis by interferons: effects on tumor- and lymphocyte-induced vascular responses. Cancer Research, 47(19), 5155–5161.PubMed
33.
go back to reference Sgadari, C., et al. (1997). Mig, the monokine induced by interferon-gamma, promotes tumor necrosis in vivo. Blood, 89(8), 2635–2643.PubMed Sgadari, C., et al. (1997). Mig, the monokine induced by interferon-gamma, promotes tumor necrosis in vivo. Blood, 89(8), 2635–2643.PubMed
34.
go back to reference Strieter, R. M., et al. (1995). The role of CXC chemokines as regulators of angiogenesis. Shock, 4(3), 155–160.PubMed Strieter, R. M., et al. (1995). The role of CXC chemokines as regulators of angiogenesis. Shock, 4(3), 155–160.PubMed
35.
go back to reference Stewart, R. J., Kashour, T. S., & Marsden, P. A. (1996). Vascular endothelial platelet endothelial adhesion molecule-1 (PECAM-1) expression is decreased by TNF-alpha and IFN-gamma. Evidence for cytokine-induced destabilization of messenger ribonucleic acid transcripts in bovine endothelial cells. Journal of Immunology, 156(3), 1221–1228. Stewart, R. J., Kashour, T. S., & Marsden, P. A. (1996). Vascular endothelial platelet endothelial adhesion molecule-1 (PECAM-1) expression is decreased by TNF-alpha and IFN-gamma. Evidence for cytokine-induced destabilization of messenger ribonucleic acid transcripts in bovine endothelial cells. Journal of Immunology, 156(3), 1221–1228.
36.
go back to reference Ruegg, C., Dormond, O., & Foletti, A. (2002). Suppression of tumor angiogenesis through the inhibition of integrin function and signaling in endothelial cells: which side to target? Endothelium, 9(3), 151–160.PubMed Ruegg, C., Dormond, O., & Foletti, A. (2002). Suppression of tumor angiogenesis through the inhibition of integrin function and signaling in endothelial cells: which side to target? Endothelium, 9(3), 151–160.PubMed
37.
go back to reference Qin, Z., et al. (2003). A critical requirement of interferon gamma-mediated angiostasis for tumor rejection by CD8+ T cells. Cancer Research, 63(14), 4095–4100.PubMed Qin, Z., et al. (2003). A critical requirement of interferon gamma-mediated angiostasis for tumor rejection by CD8+ T cells. Cancer Research, 63(14), 4095–4100.PubMed
38.
go back to reference Edwards, R. P., et al. (1997). Comparison of toxicity and survival following intraperitoneal recombinant interleukin-2 for persistent ovarian cancer after platinum: twenty-four-hour versus 7-day infusion. Journal of Clinical Oncology, 15(11), 3399–3407.PubMed Edwards, R. P., et al. (1997). Comparison of toxicity and survival following intraperitoneal recombinant interleukin-2 for persistent ovarian cancer after platinum: twenty-four-hour versus 7-day infusion. Journal of Clinical Oncology, 15(11), 3399–3407.PubMed
39.
go back to reference Vlad, A.M., et al., A phase II trial of intraperitoneal interleukin-2 in patients with platinum-resistant or platinum-refractory ovarian cancer. Cancer Immunol Immunother, 2009. Vlad, A.M., et al., A phase II trial of intraperitoneal interleukin-2 in patients with platinum-resistant or platinum-refractory ovarian cancer. Cancer Immunol Immunother, 2009.
40.
go back to reference Hodi, F. S., et al. (2008). Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proceedings of the National Academy of Sciences of the United States of America, 105(8), 3005–3010.PubMed Hodi, F. S., et al. (2008). Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proceedings of the National Academy of Sciences of the United States of America, 105(8), 3005–3010.PubMed
41.
go back to reference Hodi, F. S., et al. (2003). Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proceedings of the National Academy of Sciences of the United States of America, 100(8), 4712–4717.PubMed Hodi, F. S., et al. (2003). Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proceedings of the National Academy of Sciences of the United States of America, 100(8), 4712–4717.PubMed
42.
go back to reference Pujade-Lauraine, E., et al. (1996). Intraperitoneal recombinant interferon gamma in ovarian cancer patients with residual disease at second-look laparotomy. Journal of Clinical Oncology, 14(2), 343–350.PubMed Pujade-Lauraine, E., et al. (1996). Intraperitoneal recombinant interferon gamma in ovarian cancer patients with residual disease at second-look laparotomy. Journal of Clinical Oncology, 14(2), 343–350.PubMed
43.
go back to reference Chen, J. T., Hasumi, K., & Masubuchi, K. (1992). Interferon-alpha, interferon-gamma and sizofiran in the adjuvant therapy in ovarian cancer–a preliminary trial. Biotherapy, 5(4), 275–280.PubMed Chen, J. T., Hasumi, K., & Masubuchi, K. (1992). Interferon-alpha, interferon-gamma and sizofiran in the adjuvant therapy in ovarian cancer–a preliminary trial. Biotherapy, 5(4), 275–280.PubMed
44.
go back to reference Colombo, N., et al. (1992). Anti-tumor and immunomodulatory activity of intraperitoneal IFN-gamma in ovarian carcinoma patients with minimal residual tumor after chemotherapy. International Journal of Cancer, 51(1), 42–46. Colombo, N., et al. (1992). Anti-tumor and immunomodulatory activity of intraperitoneal IFN-gamma in ovarian carcinoma patients with minimal residual tumor after chemotherapy. International Journal of Cancer, 51(1), 42–46.
45.
go back to reference Schmeler, K. M., et al. (2009). A phase II study of GM-CSF and rIFN-gamma1b plus carboplatin for the treatment of recurrent, platinum-sensitive ovarian, fallopian tube and primary peritoneal cancer. Gynecologic Oncology, 113(2), 210–215.PubMed Schmeler, K. M., et al. (2009). A phase II study of GM-CSF and rIFN-gamma1b plus carboplatin for the treatment of recurrent, platinum-sensitive ovarian, fallopian tube and primary peritoneal cancer. Gynecologic Oncology, 113(2), 210–215.PubMed
46.
go back to reference Berek, J. S. (2000). Interferon plus chemotherapy for primary treatment of ovarian cancer. Lancet, 356(9223), 6–7.PubMed Berek, J. S. (2000). Interferon plus chemotherapy for primary treatment of ovarian cancer. Lancet, 356(9223), 6–7.PubMed
47.
go back to reference Windbichler, G. H., et al. (2000). Interferon-gamma in the first-line therapy of ovarian cancer: a randomized phase III trial. British Journal of Cancer, 82(6), 1138–1144.PubMed Windbichler, G. H., et al. (2000). Interferon-gamma in the first-line therapy of ovarian cancer: a randomized phase III trial. British Journal of Cancer, 82(6), 1138–1144.PubMed
48.
go back to reference Alberts, D. S., et al. (2008). Randomized phase 3 trial of interferon gamma-1b plus standard carboplatin/paclitaxel versus carboplatin/paclitaxel alone for first-line treatment of advanced ovarian and primary peritoneal carcinomas: results from a prospectively designed analysis of progression-free survival. Gynecologic Oncology, 109(2), 174–181.PubMed Alberts, D. S., et al. (2008). Randomized phase 3 trial of interferon gamma-1b plus standard carboplatin/paclitaxel versus carboplatin/paclitaxel alone for first-line treatment of advanced ovarian and primary peritoneal carcinomas: results from a prospectively designed analysis of progression-free survival. Gynecologic Oncology, 109(2), 174–181.PubMed
49.
go back to reference 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. Proceedings of the National Academy of Sciences of the United States of America, 104(31), 12837–12842.PubMed 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. Proceedings of the National Academy of Sciences of the United States of America, 104(31), 12837–12842.PubMed
50.
go back to reference Oei, A. L., et al. (2008). The use of monoclonal antibodies for the treatment of epithelial ovarian cancer (review). International Journal of Oncology, 32(6), 1145–1157.PubMed Oei, A. L., et al. (2008). The use of monoclonal antibodies for the treatment of epithelial ovarian cancer (review). International Journal of Oncology, 32(6), 1145–1157.PubMed
51.
go back to reference 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. Clinical Cancer Research, 11(23), 8326–8331.PubMed 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. Clinical Cancer Research, 11(23), 8326–8331.PubMed
52.
go back to reference Curiel, T. J., et al. (2004). Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Research, 64(16), 5535–5538.PubMed Curiel, T. J., et al. (2004). Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Research, 64(16), 5535–5538.PubMed
53.
go back to reference Oei, A. L., et al. (2007). Induction of IgG antibodies to MUC1 improves survival in patients with epithelial ovarian cancer. Gynecologic Oncology, 104(3), S11–S11. Oei, A. L., et al. (2007). Induction of IgG antibodies to MUC1 improves survival in patients with epithelial ovarian cancer. Gynecologic Oncology, 104(3), S11–S11.
54.
go back to reference Oei, A. L. M., et al. (2008). Induction of IgG antibodies to MUC1 and survival in patients with epithelial ovarian cancer. International Journal of Cancer, 123(8), 1848–1853. Oei, A. L. M., et al. (2008). Induction of IgG antibodies to MUC1 and survival in patients with epithelial ovarian cancer. International Journal of Cancer, 123(8), 1848–1853.
55.
go back to reference Takikawa, O. (2005). Biochemical and medical aspects of the indoleamine 2, 3-dioxygenase-initiated L-tryptophan metabolism. Biochemical and Biophysical Research Communications, 338(1), 12–19.PubMed Takikawa, O. (2005). Biochemical and medical aspects of the indoleamine 2, 3-dioxygenase-initiated L-tryptophan metabolism. Biochemical and Biophysical Research Communications, 338(1), 12–19.PubMed
56.
go back to reference Woo, E. Y., et al. (2001). Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Research, 61(12), 4766–4772.PubMed Woo, E. Y., et al. (2001). Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Research, 61(12), 4766–4772.PubMed
57.
go back to reference Curiel, T. J., et al. (2004). Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Natural Medicines, 10(9), 942–949. Curiel, T. J., et al. (2004). Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Natural Medicines, 10(9), 942–949.
58.
go back to reference Bluestone, J. A., & Abbas, A. K. (2003). Natural versus adaptive regulatory T cells. Nature Reviews. Immunology, 3(3), 253–257.PubMed Bluestone, J. A., & Abbas, A. K. (2003). Natural versus adaptive regulatory T cells. Nature Reviews. Immunology, 3(3), 253–257.PubMed
59.
go back to reference Sakaguchi, S. (2004). Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annual Review of Immunology, 22, 531–562.PubMed Sakaguchi, S. (2004). Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annual Review of Immunology, 22, 531–562.PubMed
60.
go back to reference Read, S., & Powrie, F. (2001). CD4(+) regulatory T cells. Current Opinion in Immunology, 13(6), 644–649.PubMed Read, S., & Powrie, F. (2001). CD4(+) regulatory T cells. Current Opinion in Immunology, 13(6), 644–649.PubMed
61.
go back to reference 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. Clinical Cancer Research, 11(23), 8326–8331.PubMed 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. Clinical Cancer Research, 11(23), 8326–8331.PubMed
62.
go back to reference Leffers, N., et al. (2009). Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunology, Immunotherapy, 58(3), 449–459.PubMed Leffers, N., et al. (2009). Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunology, Immunotherapy, 58(3), 449–459.PubMed
63.
go back to reference Wherry, E. J., & Ahmed, R. (2004). Memory CD8 T-cell differentiation during viral infection. Journal of Virology, 78(11), 5535–5545.PubMed Wherry, E. J., & Ahmed, R. (2004). Memory CD8 T-cell differentiation during viral infection. Journal of Virology, 78(11), 5535–5545.PubMed
64.
go back to reference Sharpe, A. H., & Freeman, G. J. (2002). The B7-CD28 superfamily. Nature Reviews. Immunology, 2(2), 116–126.PubMed Sharpe, A. H., & Freeman, G. J. (2002). The B7-CD28 superfamily. Nature Reviews. Immunology, 2(2), 116–126.PubMed
65.
go back to reference Chen, L. (2004). Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nature Reviews. Immunology, 4(5), 336–347.PubMed Chen, L. (2004). Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nature Reviews. Immunology, 4(5), 336–347.PubMed
66.
go back to reference Okazaki, T., & Honjo, T. (2006). The PD-1-PD-L pathway in immunological tolerance. Trends in Immunology, 27(4), 195–201.PubMed Okazaki, T., & Honjo, T. (2006). The PD-1-PD-L pathway in immunological tolerance. Trends in Immunology, 27(4), 195–201.PubMed
67.
go back to reference Barber, D. L., et al. (2006). Restoring function in exhausted CD8 T cells during chronic viral infection. Nature, 439(7077), 682–687.PubMed Barber, D. L., et al. (2006). Restoring function in exhausted CD8 T cells during chronic viral infection. Nature, 439(7077), 682–687.PubMed
68.
go back to reference Day, C. L., et al. (2006). PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature, 443(7109), 350–354.PubMed Day, C. L., et al. (2006). PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature, 443(7109), 350–354.PubMed
69.
go back to reference Trautmann, L., et al. (2006). Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Natural Medicines, 12(10), 1198–1202. Trautmann, L., et al. (2006). Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Natural Medicines, 12(10), 1198–1202.
70.
go back to reference Freeman, G. J., et al. (2006). Reinvigorating exhausted HIV-specific T cells via PD-1-PD-1 ligand blockade. The Journal of Experimental Medicine, 203(10), 2223–2227.PubMed Freeman, G. J., et al. (2006). Reinvigorating exhausted HIV-specific T cells via PD-1-PD-1 ligand blockade. The Journal of Experimental Medicine, 203(10), 2223–2227.PubMed
71.
go back to reference Kim, P. S., & Ahmed, R. (2010). Features of responding T cells in cancer and chronic infection. Curr Opin Immunol, 22(2), 223–230.PubMed Kim, P. S., & Ahmed, R. (2010). Features of responding T cells in cancer and chronic infection. Curr Opin Immunol, 22(2), 223–230.PubMed
73.
go back to reference Chemnitz, J. M., et al. (2004). SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. Journal of Immunology, 173(2), 945–954. Chemnitz, J. M., et al. (2004). SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. Journal of Immunology, 173(2), 945–954.
74.
go back to reference Nishimura, H., et al. (2001). Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science, 291(5502), 319–322.PubMed Nishimura, H., et al. (2001). Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science, 291(5502), 319–322.PubMed
75.
go back to reference Dong, H., et al. (2002). Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Natural Medicines, 8(8), 793–800. Dong, H., et al. (2002). Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Natural Medicines, 8(8), 793–800.
76.
go back to reference Inman, B. A., et al. (2007). PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer, 109(8), 1499–1505.PubMed Inman, B. A., et al. (2007). PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer, 109(8), 1499–1505.PubMed
77.
go back to reference Nomi, T., et al. (2007). Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical Cancer Research, 13(7), 2151–2157.PubMed Nomi, T., et al. (2007). Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical Cancer Research, 13(7), 2151–2157.PubMed
78.
go back to reference Thompson, R. H., Dong, H., & Kwon, E. D. (2007). Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clinical Cancer Research, 13(2 Pt 2), 709s–715s.PubMed Thompson, R. H., Dong, H., & Kwon, E. D. (2007). Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clinical Cancer Research, 13(2 Pt 2), 709s–715s.PubMed
79.
go back to reference Ahmadzadeh, M., et al. (2009). Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood, 114(8), 1537–1544.PubMed Ahmadzadeh, M., et al. (2009). Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood, 114(8), 1537–1544.PubMed
80.
go back to reference Matsuzaki, J., 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 U S A, 107(17), 7875–7880. Matsuzaki, J., 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 U S A, 107(17), 7875–7880.
81.
go back to reference Ercolini, A. M., et al. (2005). Recruitment of latent pools of high-avidity CD8(+) T cells to the antitumor immune response. The Journal of Experimental Medicine, 201(10), 1591–1602.PubMed Ercolini, A. M., et al. (2005). Recruitment of latent pools of high-avidity CD8(+) T cells to the antitumor immune response. The Journal of Experimental Medicine, 201(10), 1591–1602.PubMed
82.
go back to reference Thompson, R. H., et al. (2007). PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clinical Cancer Research, 13(6), 1757–1761.PubMed Thompson, R. H., et al. (2007). PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clinical Cancer Research, 13(6), 1757–1761.PubMed
83.
go back to reference Quezada, S. A., et al. (2006). CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. The Journal of Clinical Investigation, 116(7), 1935–1945.PubMed Quezada, S. A., et al. (2006). CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. The Journal of Clinical Investigation, 116(7), 1935–1945.PubMed
84.
go back to reference Li, B., et al. (2009). Anti-programmed death-1 synergizes with granulocyte macrophage colony-stimulating factor–secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors. Clinical Cancer Research, 15(5), 1623–1634.PubMed Li, B., et al. (2009). Anti-programmed death-1 synergizes with granulocyte macrophage colony-stimulating factor–secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors. Clinical Cancer Research, 15(5), 1623–1634.PubMed
85.
go back to reference Curran, M.A. and J.P. Allison, Tumor Vaccines Expressing Flt3 Ligand Synergize with CTLA-4 Blockade to Reject Preimplanted Tumors. Cancer Res, 2009. Curran, M.A. and J.P. Allison, Tumor Vaccines Expressing Flt3 Ligand Synergize with CTLA-4 Blockade to Reject Preimplanted Tumors. Cancer Res, 2009.
86.
go back to reference Li, Q., et al. (2007). Adjuvant effect of anti-4-1BB mAb administration in adoptive T cell therapy of cancer. International Journal of Biological Sciences, 3(7), 455–462.PubMed Li, Q., et al. (2007). Adjuvant effect of anti-4-1BB mAb administration in adoptive T cell therapy of cancer. International Journal of Biological Sciences, 3(7), 455–462.PubMed
87.
go back to reference Brahmer, J.R., et al., Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 28(19): p. 3167-75. Brahmer, J.R., et al., Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 28(19): p. 3167-75.
88.
go back to reference Butte, M. J., et al. (2007). Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity, 27(1), 111–122.PubMed Butte, M. J., et al. (2007). Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity, 27(1), 111–122.PubMed
89.
go back to reference Ohigashi, Y., et al. (2005). Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clinical Cancer Research, 11(8), 2947–2953.PubMed Ohigashi, Y., et al. (2005). Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clinical Cancer Research, 11(8), 2947–2953.PubMed
90.
go back to reference Francisco, L.M., P.T. Sage, and A.H. Sharpe, The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 236: p. 219-42. Francisco, L.M., P.T. Sage, and A.H. Sharpe, The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 236: p. 219-42.
91.
go back to reference Frumento, G., et al. (2002). Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2, 3-dioxygenase. The Journal of Experimental Medicine, 196(4), 459–468.PubMed Frumento, G., et al. (2002). Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2, 3-dioxygenase. The Journal of Experimental Medicine, 196(4), 459–468.PubMed
92.
go back to reference Terness, P., et al. (2002). Inhibition of allogeneic T cell proliferation by indoleamine 2, 3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. The Journal of Experimental Medicine, 196(4), 447–457.PubMed Terness, P., et al. (2002). Inhibition of allogeneic T cell proliferation by indoleamine 2, 3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. The Journal of Experimental Medicine, 196(4), 447–457.PubMed
93.
go back to reference Della Chiesa, M., et al. (2006). The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood, 108(13), 4118–4125. Della Chiesa, M., et al. (2006). The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood, 108(13), 4118–4125.
94.
go back to reference Munn, D. H., & Mellor, A. L. (2007). Indoleamine 2, 3-dioxygenase and tumor-induced tolerance. The Journal of Clinical Investigation, 117(5), 1147–1154.PubMed Munn, D. H., & Mellor, A. L. (2007). Indoleamine 2, 3-dioxygenase and tumor-induced tolerance. The Journal of Clinical Investigation, 117(5), 1147–1154.PubMed
95.
go back to reference Katz, J. B., Muller, A. J., & Prendergast, G. C. (2008). Indoleamine 2, 3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunological Reviews, 222, 206–221.PubMed Katz, J. B., Muller, A. J., & Prendergast, G. C. (2008). Indoleamine 2, 3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunological Reviews, 222, 206–221.PubMed
96.
go back to reference Sharma, M. D., et al. (2007). Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2, 3-dioxygenase. The Journal of Clinical Investigation, 117(9), 2570–2582.PubMed Sharma, M. D., et al. (2007). Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2, 3-dioxygenase. The Journal of Clinical Investigation, 117(9), 2570–2582.PubMed
97.
go back to reference Chen, W., Liang, X., Peterson, A. J., et al. (2008). The indoleamine 2, 3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. Journal of Immunology, 181(8), 5396–5404. Chen, W., Liang, X., Peterson, A. J., et al. (2008). The indoleamine 2, 3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. Journal of Immunology, 181(8), 5396–5404.
98.
go back to reference Brandacher, G., et al. (2006). Prognostic value of indoleamine 2, 3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clinical Cancer Research, 12(4), 1144–1151.PubMed Brandacher, G., et al. (2006). Prognostic value of indoleamine 2, 3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clinical Cancer Research, 12(4), 1144–1151.PubMed
99.
go back to reference Pan, K., et al. (2008). Expression and prognosis role of indoleamine 2, 3-dioxygenase in hepatocellular carcinoma. Journal of Cancer Research and Clinical Oncology, 134(11), 1247–1253.PubMed Pan, K., et al. (2008). Expression and prognosis role of indoleamine 2, 3-dioxygenase in hepatocellular carcinoma. Journal of Cancer Research and Clinical Oncology, 134(11), 1247–1253.PubMed
100.
go back to reference Ino, K., et al. (2006). Indoleamine 2, 3-dioxygenase is a novel prognostic indicator for endometrial cancer. British Journal of Cancer, 95(11), 1555–1561.PubMed Ino, K., et al. (2006). Indoleamine 2, 3-dioxygenase is a novel prognostic indicator for endometrial cancer. British Journal of Cancer, 95(11), 1555–1561.PubMed
101.
go back to reference Ino, K., et al. (2008). Inverse correlation between tumoral indoleamine 2, 3-dioxygenase expression and tumor-infiltrating lymphocytes in endometrial cancer: its association with disease progression and survival. Clinical Cancer Research, 14(8), 2310–2317.PubMed Ino, K., et al. (2008). Inverse correlation between tumoral indoleamine 2, 3-dioxygenase expression and tumor-infiltrating lymphocytes in endometrial cancer: its association with disease progression and survival. Clinical Cancer Research, 14(8), 2310–2317.PubMed
102.
go back to reference Inaba, T., et al. (2010). Indoleamine 2, 3-dioxygenase expression predicts impaired survival of invasive cervical cancer patients treated with radical hysterectomy. Gynecologic Oncology, 117(3), 423–428.PubMed Inaba, T., et al. (2010). Indoleamine 2, 3-dioxygenase expression predicts impaired survival of invasive cervical cancer patients treated with radical hysterectomy. Gynecologic Oncology, 117(3), 423–428.PubMed
103.
go back to reference Okamoto, A., et al. (2005). Indoleamine 2, 3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clinical Cancer Research, 11(16), 6030–6039.PubMed Okamoto, A., et al. (2005). Indoleamine 2, 3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clinical Cancer Research, 11(16), 6030–6039.PubMed
104.
go back to reference Takao, M. (2007). O.A., Nikaido T, et al, Increased synthesis of indoleamine-2, 3-dioxygenase protein is positively associated with impaired survival in patients with serous-type, but not with other types of ovarian cancer. Oncology Reports, 17, 1333–1339.PubMed Takao, M. (2007). O.A., Nikaido T, et al, Increased synthesis of indoleamine-2, 3-dioxygenase protein is positively associated with impaired survival in patients with serous-type, but not with other types of ovarian cancer. Oncology Reports, 17, 1333–1339.PubMed
105.
go back to reference Inaba, T., et al. (2009). Role of the immunosuppressive enzyme indoleamine 2, 3-dioxygenase in the progression of ovarian carcinoma. Gynecologic Oncology, 115(2), 185–192.PubMed Inaba, T., et al. (2009). Role of the immunosuppressive enzyme indoleamine 2, 3-dioxygenase in the progression of ovarian carcinoma. Gynecologic Oncology, 115(2), 185–192.PubMed
106.
go back to reference Qian, F., et al. (2009). 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 Research, 69(13), 5498–5504.PubMed Qian, F., et al. (2009). 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 Research, 69(13), 5498–5504.PubMed
107.
go back to reference Yoshida, N., et al. (2008). Overexpression of indoleamine 2, 3-dioxygenase in human endometrial carcinoma cells induces rapid tumor growth in a mouse xenograft model. Clinical Cancer Research, 14(22), 7251–7259.PubMed Yoshida, N., et al. (2008). Overexpression of indoleamine 2, 3-dioxygenase in human endometrial carcinoma cells induces rapid tumor growth in a mouse xenograft model. Clinical Cancer Research, 14(22), 7251–7259.PubMed
108.
go back to reference Dudley, M. E., et al. (2002). Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science, 298(5594), 850–854.PubMed Dudley, M. E., et al. (2002). Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science, 298(5594), 850–854.PubMed
109.
go back to reference Peggs, K. S., et al. (2009). Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. The Journal of Experimental Medicine, 206(8), 1717–1725.PubMed Peggs, K. S., et al. (2009). Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. The Journal of Experimental Medicine, 206(8), 1717–1725.PubMed
110.
go back to reference Shrimali, R. K., et al. (2010). Antiangiogenic Agents Can Increase Lymphocyte Infiltration into Tumor and Enhance the Effectiveness of Adoptive Immunotherapy of Cancer. Cancer Research, 70(15), 6171–6180.PubMed Shrimali, R. K., et al. (2010). Antiangiogenic Agents Can Increase Lymphocyte Infiltration into Tumor and Enhance the Effectiveness of Adoptive Immunotherapy of Cancer. Cancer Research, 70(15), 6171–6180.PubMed
111.
go back to reference Buckanovich, R. J., et al. (2008). Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Natural Medicines, 14(1), 28–36. Buckanovich, R. J., et al. (2008). Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Natural Medicines, 14(1), 28–36.
112.
go back to reference Mazanet, M. M., & Hughes, C. C. W. (2002). B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. Journal of Immunology, 169(7), 3581–3588. Mazanet, M. M., & Hughes, C. C. W. (2002). B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. Journal of Immunology, 169(7), 3581–3588.
113.
go back to reference Rodig, N., et al. (2003). Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. European Journal of Immunology, 33(11), 3117–3126.PubMed Rodig, N., et al. (2003). Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. European Journal of Immunology, 33(11), 3117–3126.PubMed
114.
go back to reference Sata, M., & Walsh, K. (1998). Oxidized LDL activates fas-mediated endothelial cell apoptosis. The Journal of Clinical Investigation, 102(9), 1682–1689.PubMed Sata, M., & Walsh, K. (1998). Oxidized LDL activates fas-mediated endothelial cell apoptosis. The Journal of Clinical Investigation, 102(9), 1682–1689.PubMed
115.
go back to reference Secchiero, P., et al. (2006). Systemic tumor necrosis factor-related apoptosis-inducing ligand delivery shows antiatherosclerotic activity in apolipoprotein E-null diabetic mice. Circulation, 114(14), 1522–1530.PubMed Secchiero, P., et al. (2006). Systemic tumor necrosis factor-related apoptosis-inducing ligand delivery shows antiatherosclerotic activity in apolipoprotein E-null diabetic mice. Circulation, 114(14), 1522–1530.PubMed
116.
go back to reference Hernandez-Alcoceba, R., et al. (2001). Evaluation of a new dual-specificity promoter for selective induction of apoptosis in breast cancer cells. Cancer Gene Therapy, 8(4), 298–307.PubMed Hernandez-Alcoceba, R., et al. (2001). Evaluation of a new dual-specificity promoter for selective induction of apoptosis in breast cancer cells. Cancer Gene Therapy, 8(4), 298–307.PubMed
117.
go back to reference Pirtskhalaishvili, G. (2000). Transduction of dendritic cells with Bcl-xL increases their resistance to prostate cancer-induced apoptosis and antitumor effect in mice. Journal of Immunology, 165, 1956–1964. Pirtskhalaishvili, G. (2000). Transduction of dendritic cells with Bcl-xL increases their resistance to prostate cancer-induced apoptosis and antitumor effect in mice. Journal of Immunology, 165, 1956–1964.
118.
go back to reference Batista, F. D., & Harwood, N. (2009). The who, how and where of antigen presentation to B cells. Nature Reviews. Immunology, 9(1), 15–27.PubMed Batista, F. D., & Harwood, N. (2009). The who, how and where of antigen presentation to B cells. Nature Reviews. Immunology, 9(1), 15–27.PubMed
119.
go back to reference Grant, K., Loizidou, M., & Taylor, I. (2003). Endothelin-1: a multifunctional molecule in cancer. British Journal of Cancer, 88(2), 163–166.PubMed Grant, K., Loizidou, M., & Taylor, I. (2003). Endothelin-1: a multifunctional molecule in cancer. British Journal of Cancer, 88(2), 163–166.PubMed
120.
go back to reference Kedzierski, R. M., & Yanagisawa, M. (2001). Endothelin system: the double-edged sword in health and disease. Annual Review of Pharmacology and Toxicology, 41, 851–876.PubMed Kedzierski, R. M., & Yanagisawa, M. (2001). Endothelin system: the double-edged sword in health and disease. Annual Review of Pharmacology and Toxicology, 41, 851–876.PubMed
121.
go back to reference Yanagisawa, H., et al. (1998). Role of Endothelin-1/Endothelin-A receptor-mediated signaling pathway in the aortic arch patterning in mice. The Journal of Clinical Investigation, 102(1), 22–33.PubMed Yanagisawa, H., et al. (1998). Role of Endothelin-1/Endothelin-A receptor-mediated signaling pathway in the aortic arch patterning in mice. The Journal of Clinical Investigation, 102(1), 22–33.PubMed
122.
go back to reference Meidan, R., & Levy, N. (2007). The ovarian endothelin network: an evolving story. Trends in Endocrinology and Metabolism, 18(10), 379–385.PubMed Meidan, R., & Levy, N. (2007). The ovarian endothelin network: an evolving story. Trends in Endocrinology and Metabolism, 18(10), 379–385.PubMed
123.
go back to reference Nelson, J., et al. (2003). The endothelin axis: emerging role in cancer. Nature Reviews. Cancer, 3(2), 110–116.PubMed Nelson, J., et al. (2003). The endothelin axis: emerging role in cancer. Nature Reviews. Cancer, 3(2), 110–116.PubMed
124.
go back to reference Bagnato, A., & Rosano, L. (2008). The endothelin axis in cancer. The International Journal of Biochemistry & Cell Biology, 40(8), 1443–1451. Bagnato, A., & Rosano, L. (2008). The endothelin axis in cancer. The International Journal of Biochemistry & Cell Biology, 40(8), 1443–1451.
125.
go back to reference Yanagisawa, M., & Masaki, T. (1989). Molecular biology and biochemistry of the endothelins. Trends in Pharmacological Sciences, 10(9), 374–378.PubMed Yanagisawa, M., & Masaki, T. (1989). Molecular biology and biochemistry of the endothelins. Trends in Pharmacological Sciences, 10(9), 374–378.PubMed
126.
go back to reference Saida, K., Mitsui, Y., & Ishida, N. (1989). A novel peptide, vasoactive intestinal contractor, of a new (endothelin) peptide family. Molecular cloning, expression, and biological activity. The Journal of Biological Chemistry, 264(25), 14613–14616.PubMed Saida, K., Mitsui, Y., & Ishida, N. (1989). A novel peptide, vasoactive intestinal contractor, of a new (endothelin) peptide family. Molecular cloning, expression, and biological activity. The Journal of Biological Chemistry, 264(25), 14613–14616.PubMed
127.
go back to reference Valdenaire, O., Rohrbacher, E., & Mattei, M. G. (1995). Organization of the gene encoding the human endothelin-converting enzyme (ECE-1). The Journal of Biological Chemistry, 270(50), 29794–29798.PubMed Valdenaire, O., Rohrbacher, E., & Mattei, M. G. (1995). Organization of the gene encoding the human endothelin-converting enzyme (ECE-1). The Journal of Biological Chemistry, 270(50), 29794–29798.PubMed
128.
go back to reference Frommer, K. W., & Muller-Ladner, U. (2008). Expression and function of ETA and ETB receptors in SSc. Rheumatology (Oxford), 47(Suppl 5), v27–v28. Frommer, K. W., & Muller-Ladner, U. (2008). Expression and function of ETA and ETB receptors in SSc. Rheumatology (Oxford), 47(Suppl 5), v27–v28.
129.
go back to reference Luscher, T. F., & Barton, M. (2000). Endothelins and endothelin receptor antagonists: therapeutic considerations for a novel class of cardiovascular drugs. Circulation, 102(19), 2434–2440.PubMed Luscher, T. F., & Barton, M. (2000). Endothelins and endothelin receptor antagonists: therapeutic considerations for a novel class of cardiovascular drugs. Circulation, 102(19), 2434–2440.PubMed
130.
go back to reference Salani, D., et al. (2000). Endothelin-1 induces an angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. The American Journal of Pathology, 157(5), 1703–1711.PubMed Salani, D., et al. (2000). Endothelin-1 induces an angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. The American Journal of Pathology, 157(5), 1703–1711.PubMed
131.
go back to reference Jesmin, S., et al. (2006). Down-regulated VEGF expression in the diabetic heart is normalized by an endothelin ETA receptor antagonist. European Journal of Pharmacology, 542(1–3), 184–185.PubMed Jesmin, S., et al. (2006). Down-regulated VEGF expression in the diabetic heart is normalized by an endothelin ETA receptor antagonist. European Journal of Pharmacology, 542(1–3), 184–185.PubMed
132.
go back to reference Kato, T., et al. (2001). Angiogenesis as a predictor of long-term survival for 377 Japanese patients with breast cancer. Breast Cancer Research and Treatment, 70(1), 65–74.PubMed Kato, T., et al. (2001). Angiogenesis as a predictor of long-term survival for 377 Japanese patients with breast cancer. Breast Cancer Research and Treatment, 70(1), 65–74.PubMed
133.
go back to reference Spinella, F., et al. (2004). Endothelin-1 Stimulates Cyclooxygenase-2 Expression in Ovarian Cancer Cells Through Multiple Signaling Pathways: Evidence for Involvement of Transactivation of the Epidermal Growth Factor Receptor. Journal of Cardiovascular Pharmacology, 44, S140–S143.PubMed Spinella, F., et al. (2004). Endothelin-1 Stimulates Cyclooxygenase-2 Expression in Ovarian Cancer Cells Through Multiple Signaling Pathways: Evidence for Involvement of Transactivation of the Epidermal Growth Factor Receptor. Journal of Cardiovascular Pharmacology, 44, S140–S143.PubMed
134.
go back to reference Spinella, F., et al. (2007). Endothelin-1 and endothelin-3 promote invasive behavior via hypoxia-inducible factor-1alpha in human melanoma cells. Cancer Research, 67(4), 1725–1734.PubMed Spinella, F., et al. (2007). Endothelin-1 and endothelin-3 promote invasive behavior via hypoxia-inducible factor-1alpha in human melanoma cells. Cancer Research, 67(4), 1725–1734.PubMed
135.
go back to reference Rosano, L., et al. (2003). Endothelin receptor blockade inhibits molecular effectors of Kaposi's sarcoma cell invasion and tumor growth in vivo. The American Journal of Pathology, 163(2), 753–762.PubMed Rosano, L., et al. (2003). Endothelin receptor blockade inhibits molecular effectors of Kaposi's sarcoma cell invasion and tumor growth in vivo. The American Journal of Pathology, 163(2), 753–762.PubMed
136.
go back to reference Rosano, L., et al. (2003). Therapeutic targeting of the endothelin a receptor in human ovarian carcinoma. Cancer Research, 63(10), 2447–2453.PubMed Rosano, L., et al. (2003). Therapeutic targeting of the endothelin a receptor in human ovarian carcinoma. Cancer Research, 63(10), 2447–2453.PubMed
137.
go back to reference Spinella, F., et al. (2002). Endothelin-1 induces vascular endothelial growth factor by increasing hypoxia-inducible factor-1alpha in ovarian carcinoma cells. The Journal of Biological Chemistry, 277(31), 27850–27855.PubMed Spinella, F., et al. (2002). Endothelin-1 induces vascular endothelial growth factor by increasing hypoxia-inducible factor-1alpha in ovarian carcinoma cells. The Journal of Biological Chemistry, 277(31), 27850–27855.PubMed
138.
go back to reference Spinella, F., et al. (2004). Inhibition of cyclooxygenase-1 and -2 expression by targeting the endothelin a receptor in human ovarian carcinoma cells. Clinical Cancer Research, 10(14), 4670–4679.PubMed Spinella, F., et al. (2004). Inhibition of cyclooxygenase-1 and -2 expression by targeting the endothelin a receptor in human ovarian carcinoma cells. Clinical Cancer Research, 10(14), 4670–4679.PubMed
139.
go back to reference Arnett, B., et al. (2003). Expression of CAAT enhancer binding protein beta (C/EBP beta) in cervix and endometrium. Molecular Cancer, 2, 21.PubMed Arnett, B., et al. (2003). Expression of CAAT enhancer binding protein beta (C/EBP beta) in cervix and endometrium. Molecular Cancer, 2, 21.PubMed
140.
go back to reference Merogi, A. J., et al. (1997). Tumor-host interaction: analysis of cytokines, growth factors, and tumor-infiltrating lymphocytes in ovarian carcinomas. Human Pathology, 28(3), 321–331.PubMed Merogi, A. J., et al. (1997). Tumor-host interaction: analysis of cytokines, growth factors, and tumor-infiltrating lymphocytes in ovarian carcinomas. Human Pathology, 28(3), 321–331.PubMed
141.
go back to reference Wanecek, M., et al. (2000). The endothelin system in septic and endotoxin shock. European Journal of Pharmacology, 407(1–2), 1–15.PubMed Wanecek, M., et al. (2000). The endothelin system in septic and endotoxin shock. European Journal of Pharmacology, 407(1–2), 1–15.PubMed
142.
go back to reference Sampaio, A. L., Rae, G. A., & Henriques, M. G. (2004). Effects of endothelin ETA receptor antagonism on granulocyte and lymphocyte accumulation in LPS-induced inflammation. Journal of Leukocyte Biology, 76(1), 210–216.PubMed Sampaio, A. L., Rae, G. A., & Henriques, M. G. (2004). Effects of endothelin ETA receptor antagonism on granulocyte and lymphocyte accumulation in LPS-induced inflammation. Journal of Leukocyte Biology, 76(1), 210–216.PubMed
143.
go back to reference Sampaio, A. L., Rae, G. A., & Henriques, M. M. (2000). Role of endothelins on lymphocyte accumulation in allergic pleurisy. Journal of Leukocyte Biology, 67(2), 189–195.PubMed Sampaio, A. L., Rae, G. A., & Henriques, M. M. (2000). Role of endothelins on lymphocyte accumulation in allergic pleurisy. Journal of Leukocyte Biology, 67(2), 189–195.PubMed
144.
go back to reference Rajeshkumar, N. V., Rai, A., & Gulati, A. (2005). Endothelin B receptor agonist, IRL 1620, enhances the anti-tumor efficacy of paclitaxel in breast tumor rats. Breast Cancer Research and Treatment, 94(3), 237–247.PubMed Rajeshkumar, N. V., Rai, A., & Gulati, A. (2005). Endothelin B receptor agonist, IRL 1620, enhances the anti-tumor efficacy of paclitaxel in breast tumor rats. Breast Cancer Research and Treatment, 94(3), 237–247.PubMed
145.
go back to reference Wulfing, P., et al. (2003). Expression of endothelin-1, endothelin-A, and endothelin-B receptor in human breast cancer and correlation with long-term follow-up. Clinical Cancer Research, 9(11), 4125–4131.PubMed Wulfing, P., et al. (2003). Expression of endothelin-1, endothelin-A, and endothelin-B receptor in human breast cancer and correlation with long-term follow-up. Clinical Cancer Research, 9(11), 4125–4131.PubMed
146.
go back to reference Grimshaw, M. J., et al. (2004). A role for endothelin-2 and its receptors in breast tumor cell invasion. Cancer Research, 64(7), 2461–2468.PubMed Grimshaw, M. J., et al. (2004). A role for endothelin-2 and its receptors in breast tumor cell invasion. Cancer Research, 64(7), 2461–2468.PubMed
147.
go back to reference Balint, K., et al. (2008). Role of vascular leukocytes in ovarian cancer neovascularization. Advances in Experimental Medicine and Biology, 622, 273–280.PubMed Balint, K., et al. (2008). Role of vascular leukocytes in ovarian cancer neovascularization. Advances in Experimental Medicine and Biology, 622, 273–280.PubMed
148.
go back to reference Halcox, J. P., et al. (2007). Endogenous endothelin in human coronary vascular function: differential contribution of endothelin receptor types A and B. Hypertension, 49(5), 1134–1141.PubMed Halcox, J. P., et al. (2007). Endogenous endothelin in human coronary vascular function: differential contribution of endothelin receptor types A and B. Hypertension, 49(5), 1134–1141.PubMed
149.
go back to reference Cardillo, C., et al. (1999). Role of endothelin in the increased vascular tone of patients with essential hypertension. Hypertension, 33(2), 753–758.PubMed Cardillo, C., et al. (1999). Role of endothelin in the increased vascular tone of patients with essential hypertension. Hypertension, 33(2), 753–758.PubMed
150.
go back to reference Cowburn, P. J., et al. (2005). Comparison of selective ET(A) and ET(B) receptor antagonists in patients with chronic heart failure. European Journal of Heart Failure, 7(1), 37–42.PubMed Cowburn, P. J., et al. (2005). Comparison of selective ET(A) and ET(B) receptor antagonists in patients with chronic heart failure. European Journal of Heart Failure, 7(1), 37–42.PubMed
151.
go back to reference Hodi, F. S., & Dranoff, G. (2006). Combinatorial cancer immunotherapy. Advances in Immunology, 90, 341–368.PubMed Hodi, F. S., & Dranoff, G. (2006). Combinatorial cancer immunotherapy. Advances in Immunology, 90, 341–368.PubMed
152.
go back to reference Cemazar, M., et al. (2005). The endothelin B (ETB) receptor agonist IRL 1620 is highly vasoconstrictive in two syngeneic rat tumour lines: potential for selective tumour blood flow modification. British Journal of Cancer, 93(1), 98–106.PubMed Cemazar, M., et al. (2005). The endothelin B (ETB) receptor agonist IRL 1620 is highly vasoconstrictive in two syngeneic rat tumour lines: potential for selective tumour blood flow modification. British Journal of Cancer, 93(1), 98–106.PubMed
Metadata
Title
Tumor immune surveillance and ovarian cancer
Lessons on immune mediated tumor rejection or tolerance
Authors
Lana E. Kandalaft
Gregory T. Motz
Jaikumar Duraiswamy
George Coukos
Publication date
01-03-2011
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 1/2011
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-011-9289-9

Other articles of this Issue 1/2011

Cancer and Metastasis Reviews 1/2011 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine