Since the beginning of the 20th century, scientists have tried to stimulate the antitumour activities of the immune system to fight against cancer. However, the scientific effort devoted on the development of cancer immunotherapy has not been translated into the expected clinical success. On the contrary, classical antineoplastic treatments such as surgery, radiotherapy, and chemotherapy are the first line of treatment. Nevertheless, there is compelling evidence on the immunogenicity of cancer cells and the capacity of the immune system to expand cancer-specific effector cytotoxic T cells. However, the effective activation of anticancer T cell responses strongly depends on efficient tumour antigen presentation from professional antigen presenting cells such as dendritic cells (DCs). Several strategies have been used to boost DC antigen presenting functions, but at the end cancer immunotherapy is not as effective as would be expected according to preclinical models. In this review, we comment on these discrepancies, focusing our attention on the contribution of regulatory T cells and myeloid-derived suppressor cells to the lack of therapeutic success of DC-based cancer immunotherapy. 1. Introduction: Vaccines Infectious Diseases and Cancer Not so long ago, I had a conversation with a colleague of mine about our differing research interests. He asked me why I was so much interested in cancer research and not that keen on infectious diseases. This particular question made me think about my motives for working in cancer research. Cancer is in many cases an “adult” noncontagious disease (with exceptions), while infectious diseases can attack anyone at any time. In addition, infectious agents are highly contagious and new ones arise from time to time [1, 2]. He argued that compared to cancer, infectious diseases are a much higher health burden worldwide. After this conversation I quickly looked at the statistics and according to Cancer Research UK (http://www.cancerresearchuk.org), there were about 7.6 million deaths from cancer in 2008. Then I looked at the deaths caused by infectious diseases, and according to the World Health Organisation (WHO, http://www.who.int/en/) about 13 million deaths (of all ages) were caused by infectious diseases in 1998. Even though the data was not that recent, I concluded that the first premise on health burden might not be completely accurate. However, ultimately that was not the reason I was looking for. Therefore, why is cancer so fascinating that attracts so much scientific and medical efforts? Among the nonmedical
References
[1]
K. V. Holmes and L. Enjuanes, “The SARS coronavirus: a postgenomic era,” Science, vol. 300, no. 5624, pp. 1377–1378, 2003.
[2]
A. M. Zaki, S. van Boheemen, and T. M. Bestebroer, “Brief report: isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia,” The New England Journal of Medicine, vol. 369, article 39, 2013.
[3]
“Smallpox eradication: the first three years,” WHO Chronicle, vol. 24, no. 7, pp. 301–310, 1970.
[4]
C. F. Estivariz, M. A. Pallansch, A. Anand et al., “Poliovirus vaccination options for achieving eradication and securing the endgame,” Current Opinion in Virology, vol. 3, no. 3, pp. 309–315, 2013.
[5]
D. S. Saint-Victor and S. B. Omer, “Vaccine refusal and the endgame: walking the last mile first,” Philosophical Transactions of the Royal Society B, vol. 368, no. 1623, Article ID 20120148, 2013.
[6]
R. Vivancos, A. Keenan, S. Farmer et al., “An ongoing large outbreak of measles in Merseyside, England,” Eurosurveillance, vol. 17, no. 29, 2012.
[7]
K. F. Brown, S. J. Long, M. Ramsay et al., “UK parents' decision-making about measles-mumps-rubella (MMR) vaccine 10 years after the MMR-autism controversy: a qualitative analysis,” Vaccine, vol. 30, no. 10, pp. 1855–1864, 2012.
[8]
B. Taylor, E. Miller, C. P. Farrington et al., “Autism and measles, mumps, and rubella vaccine: no epidemiological evidence for a causal association,” The Lancet, vol. 353, no. 9169, pp. 2026–2029, 1999.
[9]
C. V. Ichim, “Revisiting immunosurveillance and immunostimulation: implications for cancer immunotherapy,” Journal of Translational Medicine, vol. 3, article 8, 2005.
[10]
F. M. Burnet, “The concept of immunological surveillance,” Progress in Experimental Tumor Research, vol. 13, pp. 1–27, 1970.
[11]
D. Escors and K. Breckpot, “Lentiviral vectors in gene therapy: their current status and future potential,” Archivum Immunologiae et Therapiae Experimentalis, vol. 58, no. 2, pp. 107–119, 2010.
[12]
F. Arce, G. Kochan, K. Breckpot, H. Stephenson, and D. Escors, “Selective activation of intracellular signalling pathways in dendritic cells for cancer immunotherapy,” Anti-Cancer Agents in Medicinal Chemistry, vol. 12, no. 1, pp. 29–39, 2012.
[13]
K. Breckpot and D. Escors, “Dendritic cells for active anti-cancer immunotherapy: targeting activation pathways through genetic modification,” Endocrine, Metabolic and Immune Disorders, vol. 9, no. 4, pp. 328–343, 2009.
[14]
L. Apetoh, F. Ghiringhelli, A. Tesniere et al., “Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy,” Nature Medicine, vol. 13, no. 9, pp. 1050–1059, 2007.
[15]
F. Ghiringhelli, L. Apetoh, A. Tesniere et al., “Activation of the NLRP3 inflammasome in dendritic cells induces IL-1Β-dependent adaptive immunity against tumors,” Nature Medicine, vol. 15, no. 10, pp. 1170–1178, 2009.
[16]
L. Zitvogel, L. Apetoh, F. Ghiringhelli, and G. Kroemer, “Immunological aspects of cancer chemotherapy,” Nature Reviews Immunology, vol. 8, no. 1, pp. 59–73, 2008.
[17]
F. Ghiringhelli, M. Bruchard, and L. Apetoh, “Immune effects of 5-fluorouracil: ambivalence matters,” Oncoimmunology, vol. 2, no. 3, Article ID e23139, 2013.
[18]
H. Bugaut, M. Bruchard, H. Berger et al., “Bleomycin exerts ambivalent antitumor immune effect by triggering both immunogenic cell death and proliferation of regulatory T cells,” PLoS ONE, vol. 8, no. 6, Article ID e65181, 2013.
[19]
F. Ghiringhelli and L. Apetoh, “Chemotherapy and immunomodulation: from immunogenic chemotherapies to novel therapeutic strategies,” Future Oncology, vol. 9, no. 4, pp. 469–472, 2013.
[20]
N. Sengupta, T. S. MacFie, T. T. MacDonald, D. Pennington, and A. R. Silver, “Cancer immunoediting and “spontaneous” tumor regression,” Pathology Research and Practice, vol. 206, no. 1, pp. 1–8, 2010.
[21]
M. Hasek, J. Svoboda, and P. Koldovsky, “Immunological tolerance and the cancer problem,” Philippine Journal of Cancer, vol. 33, pp. 535–539, 1963.
[22]
J. M. Cruse, W. W. Germany, and A. D. Dulaney, “The demonstration of mouse fibrosarcoma fractions as antigens by latex agglutination reactions,” Zeitschrift für Immunit?tsforschung und Experimentelle Therapie, vol. 124, pp. 419–427, 1962.
[23]
G. Pasternak and A. Graffi, “Induction of resistance against isotransplantation of virus-induced,” British Journal of Cancer, vol. 17, pp. 532–539, 1963.
[24]
J. P. Glynn, A. R. Blanco, and A. Goldin, “Studies on induced resistance against isotransplants of virus-induced,” Cancer Research, vol. 24, pp. 502–508, 1964.
[25]
I. Witz, G. Hermann, M. Pikovski, and J. Gross, “The antigenic composition of tumours, sera and urines of tumour-bearing,” British Journal of Cancer, vol. 18, pp. 397–406, 1964.
[26]
S. Decarvalho, “Preparation of antigens specific of human breast carcinoma by an immunochromatographic method,” Nature, vol. 203, no. 4950, pp. 1186–1188, 1964.
[27]
H. O. Sjoegren, “Studies on specific transplantation resistance to polyoma-virus-induced tumors. Iv. Stability of the polyoma cell antigen,” Journal of the National Cancer Institute, vol. 32, pp. 661–666, 1964.
[28]
B. Stueck, L. J. Old, and E. A. Boyse, “Occurrence of soluble antigen in the plasma of mice with virus-induced,” Proceedings of the National Academy of Sciences of the United States of, vol. 52, pp. 950–958, 1964.
[29]
B. Stück, E. A. Boyse, L. J. Old, and E. A. Carswell, “ML: a new antigen found in leuk?mias and mammary tumours of the mouse,” Nature, vol. 203, no. 4949, pp. 1033–1034, 1964.
[30]
H. S. Rosenkranz, “Viral aetiology of human tumours,” Nature, vol. 218, no. 5139, pp. 370–371, 1968.
[31]
M. S. Reitz and R. C. Gallo, “Retroviruses of human T cells: their role in the aetiology of adult T-cell leukaemia/lymphoma and the acquired immune deficiency syndrome,” Cancer Surveys, vol. 4, no. 2, pp. 313–329, 1985.
[32]
G. Klein, “Tumor antigens,” Annual Review of Microbiology, vol. 20, pp. 223–252, 1966.
[33]
M. J. Casey, G. F. Rabotti, P. S. Sarma, W. T. Lane, H. C. Turner, and R. J. Huebner, “Complement-fixing antigens in hamster tumors induced by the bryan strain of rous sarcoma virus,” Science, vol. 151, no. 3714, pp. 1086–1088, 1966.
[34]
G. G. Kenter, M. J. P. Welters, A. R. P. M. Valentijn et al., “Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia,” The New England Journal of Medicine, vol. 361, no. 19, pp. 1838–1847, 2009.
[35]
G. Haughton and D. B. Amos, “Immunology of carcinogenesis,” Cancer Research, vol. 28, no. 9, pp. 1839–1840, 1968.
[36]
H. J. Smith, “Antigenicity of carcinogen-induced and spontaneous tumours in inbred mice,” British Journal of Cancer, vol. 20, no. 4, pp. 831–837, 1966.
[37]
T. Tran, D. Burt, L. Eapen, and O. R. Keller, “Spontaneous regression of metastatic melanoma after inoculation with tetanus-diphtheria-pertussis vaccine,” Current Oncology, vol. 20, no. 3, pp. e270–e273, 2013.
[38]
F. W. Stewart, “Experiences in spontaneous regression of neoplastic disease in man,” Texas Reports on Biology and Medicine, vol. 10, no. 1, pp. 239–253, 1952.
[39]
R. Dalla Favera, E. P. Gelmann, and S. Martinotti, “Cloning and characterization of different human sequences related to the onc gene (v-myc) of avian myelocytomatosis virus (MC29),” Proceedings of the National Academy of Sciences of the United States of America, vol. 79, no. 21, pp. 6497–6501, 1982.
[40]
S. P. Goff, P. D'Eustachio, F. H. Ruddle, and D. Baltimore, “Chromosomal assignment of the endogenous proto-oncogene C-abl,” Science, vol. 218, no. 4579, pp. 1317–1319, 1982.
[41]
L. F. Parada, C. J. Tabin, C. Shih, and R. A. Weinberg, “Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene,” Nature, vol. 297, no. 5866, pp. 474–478, 1982.
[42]
B. Vennstrom, “Isolation and characterization of chicken DNA homologous to the two putative oncogenes of avian erythroblastosis virus,” Cell, vol. 28, no. 1, pp. 135–143, 1982.
[43]
B. Vennstrom, D. Sheiness, J. Zabielski, and J. M. Bishop, “Isolation and characterization of c-myc, a cellular homolog of the oncogene (v-myc) of avian myelocytomatosis virus strain 29,” Journal of Virology, vol. 42, no. 3, pp. 773–779, 1982.
[44]
I. S. Y. Chen, K. C. Wilhelmsen, and H. M. Temin, “Structure and expression of c-rel, the cellular homolog to the oncogene of reticuloendotheliosis virus strain T,” Journal of Virology, vol. 45, no. 1, pp. 104–113, 1983.
[45]
K. H. Klempnauer, G. Ramsay, and J. M. Bishop, “The product of the retroviral transforming gene v-myb is a truncated version of the protein encoded by the cellular oncogene c-myb,” Cell, vol. 33, no. 2, pp. 345–355, 1983.
[46]
U. R. Rapp, M. D. Goldsborough, and G. E. Mark, “Structure and biological activity of v-raf, a unique oncogene transduced by a retrovirus,” Proceedings of the National Academy of Sciences of the United States of America, vol. 80, no. 14, pp. 4218–4222, 1983.
[47]
E. Santos, D. Martin Zanca, and E. P. Reddy, “Malignant activation of a K-ras oncogene in lung carcinoma but not in normal tissue of the same patient,” Science, vol. 223, no. 4637, pp. 661–664, 1984.
[48]
M. Barbacid, “Oncogenes and human cancer: cause or consequence?” Carcinogenesis, vol. 7, no. 7, pp. 1037–1042, 1986.
[49]
M. Malumbres and M. Barbacid, “Cell cycle kinases in cancer,” Current Opinion in Genetics and Development, vol. 17, no. 1, pp. 60–65, 2007.
[50]
R. Iggo, K. Gatter, J. Bartek, D. Lane, and A. L. Harris, “Increased expression of mutant forms of p53 oncogene in primary lung cancer,” The Lancet, vol. 335, no. 8691, pp. 675–679, 1990.
[51]
D. J. Slamon, T. C. Boone, and R. C. Seeger, “Identification and characterization of the protein encoded by the human N-myc oncogene,” Science, vol. 232, no. 4751, pp. 768–772, 1986.
[52]
W.-H. Lee, J.-Y. Shew, F. D. Hong et al., “The retinoblastoma susceptibility gene encodes a nuclear phosphoprotein associated with DNA binding activity,” Nature, vol. 329, no. 6140, pp. 642–645, 1987.
[53]
M. A. Cheever, J. P. Allison, A. S. Ferris et al., “The prioritization of cancer antigens: a National Cancer Institute pilot project for the acceleration of translational research,” Clinical Cancer Research, vol. 15, no. 17, pp. 5323–5337, 2009.
[54]
T. B. Fitzpatrick, A. B. Lerner, E. Calkins, and W. H. Summerson, “Occurrence of tyrosinase in horse and fish melanomas,” Proceedings of the Society for Experimental Biology and Medicine, vol. 75, no. 2, pp. 394–398, 1950.
[55]
T. B. Fitzpatric, “Human melanogenesis; the tyrosinase reaction in pigment cell neoplasms, with particular reference to the malignant melanoma; preliminary report,” A. M. A. Archives of Dermatology and Syphilology, vol. 65, no. 4, pp. 379–391, 1952.
[56]
T. L. Darrow, C. L. Slingluff Jr., and H. F. Seigler, “The role of HLA class I antigens in recognition of melanoma cells by tumor-specific cytotoxic T lymphocytes. Evidence for shared tumor antigens,” Journal of Immunology, vol. 142, no. 9, pp. 3329–3335, 1989.
[57]
V. Brichard, A. Van Pel, T. Wolfel et al., “The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas,” Journal of Experimental Medicine, vol. 178, no. 2, pp. 489–495, 1993.
[58]
S. L. Topalian, L. Rivoltini, M. Mancini et al., “Human CD4+ T cells specifically recognize a shared melanoma-associated antigen encoded by the tyrosinase gene,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 20, pp. 9461–9465, 1994.
[59]
A. A. Tarhini, S. Leng, S. J. Moschos et al., “Safety and immunogenicity of vaccination with MART-1 (26–35, 27L), gp100 (209–217, 210M), and tyrosinase (368–376, 370D) in adjuvant with PF-3512676 and GM-CSF in metastatic melanoma,” Journal of Immunotherapy, vol. 35, no. 4, pp. 359–366, 2012.
[60]
A. Ribas, J. S. Weber, B. Chmielowski et al., “Intra-lymph node prime-boost vaccination against Melan A and tyrosinase for the treatment of metastatic melanoma: results of a phase 1 clinical trial,” Clinical Cancer Research, vol. 17, no. 9, pp. 2987–2996, 2011.
[61]
P. Gold and S. O. Freedman, “Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques,” The Journal of Experimental Medicine, vol. 121, pp. 439–462, 1965.
[62]
P. Gold, M. Gold, and S. O. Freedman, “Cellular location of carcinoembryonic antigens of the human digestive system,” Cancer Research, vol. 28, no. 7, pp. 1331–1334, 1968.
[63]
A. H. Rule and M. E. Kirch, “Gene activation of molecules with carcinoembryonic antigen determinants in fetal development and in adenocarcinoma of the colon,” Cancer Research, vol. 36, no. 9, pp. 3503–3509, 1976.
[64]
S. A. Halter, L. D. Fraker, M. Parmenter, and W. D. Dupont, “Carcinoembryonic antigen expression and patient survival in carcinoma of the breast,” Oncology, vol. 41, no. 5, pp. 297–302, 1984.
[65]
W. Savino, D. Durand, and M. Dardenne, “Immunohistochemical evidence for the expression of the carcinoembryonic antigen by human thymic epithelial cells in vitro and in neoplastic conditions,” American Journal of Pathology, vol. 121, no. 3, pp. 418–425, 1985.
[66]
W. H. Allum, H. J. Stokes, F. MacDonald, and J. W. L. Fielding, “Demonstration of carcinoembryonic antigen (CEA) expression in normal, chronically inflamed, and malignant pancreatic tissue by immunohistochemistry,” Journal of Clinical Pathology, vol. 39, no. 6, pp. 610–614, 1986.
[67]
J. Kim, F. J. Kaye, J. G. Henslee et al., “Expression of carcinoembryonic antigen and related genes in lung and gastrointestinal cancers,” International Journal of Cancer, vol. 52, no. 5, pp. 718–725, 1992.
[68]
K. Y. Tsang, S. Zaremba, C. A. Nieroda, M. Z. Zhu, J. M. Hamilton, and J. Schlom, “Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine,” Journal of the National Cancer Institute, vol. 87, no. 13, pp. 982–990, 1995.
[69]
A. J. A. Bremers, S. H. Van der Burg, P. J. K. Kuppen, W. M. Kast, C. J. H. Van de Velde, and C. J. M. Melief, “The use of Epstein-Barr virus-transformed B lymphocyte cell lines in a peptide-reconstitution assay: identification of CEA-related HLA-A*0301- restricted potential cytotoxic T-lymphocyte epitopes,” Journal of Immunotherapy, vol. 18, no. 2, pp. 77–85, 1995.
[70]
I. Kawashima, V. Tsai, S. Southwood, K. Takesako, A. Sette, and E. Celis, “Identification of HLA-A3-restricted cytotoxic T lymphocyte epitopes from carcinoembryonic antigen and HER-2/neu by primary in vitro immunization with peptide-pulsed dendritic cells,” Cancer Research, vol. 59, no. 2, pp. 431–435, 1999.
[71]
I. Nukaya, M. Yasumoto, T. Iwasaki et al., “Identification of HLA-A24 epitope peptides of carcinoembryonic antigen which induce tumor-reactive cytotoxic T lymphocyte,” International Journal of Cancer, vol. 80, no. 1, pp. 92–97, 1999.
[72]
E. Huarte, P. Sarobe, J. J. Lasarte et al., “Identification of HLA-B27-restricted cytotoxic T lymphocyte epitope from carcinoembryonic antigen,” International Journal of Cancer, vol. 97, no. 1, pp. 58–63, 2002.
[73]
J. Schmitz, E. Reali, J. W. Hodge et al., “Identification of an interferon-γ-inducible carcinoembryonic antigen (CEA) CD8+ T-cell epitope, which mediates tumor killing in CEA transgenic mice,” Cancer Research, vol. 62, no. 17, pp. 5058–5064, 2002.
[74]
G. J. Ullenhag, J. Fagerberg, K. Strig?rd, J.-E. Fr?din, and H. Mellstedt, “Functional HLA-DR T cell epitopes of CEA identified in patients with colorectal carcinoma immunized with the recombinant protein CEA,” Cancer Immunology, Immunotherapy, vol. 53, no. 4, pp. 331–337, 2004.
[75]
G. Campi, M. Crosti, G. Consogno et al., “CD4+ T cells from healthy subjects and colon cancer patients recognize a carcinoembryonic antigen-specific immunodominant Epitope,” Cancer Research, vol. 63, no. 23, pp. 8481–8486, 2003.
[76]
S. R. Gameiro, M. L. Jammeh, and J. W. Hodge, “Cancer vaccines targeting carcinoembryonic antigen: state-of-the-art and future promise,” Expert Review of Vaccines, vol. 12, no. 6, pp. 617–629, 2013.
[77]
C. Merrin, E. Sarcione, M. Bohne, and D. J. Albert, “Alphafetoprotein in testicular tumors,” Journal of Surgical Research, vol. 15, no. 4, pp. 309–312, 1973.
[78]
J. Hou?těk, J. Masopust, K. Kithier, and J. Rádl, “Hepatocellular carcinoma in association with a specific fetal α1-globulin, fetoprotein,” The Journal of Pediatrics, vol. 72, no. 2, pp. 186–193, 1968.
[79]
C. Li, Z. Zhang, P. Zhang, and J. Liu, “Diagnostic accuracy of des-gamma-carboxy prothrombin versus alpha-fetoprotein for hepatocellular carcinoma: a systematic review,” Hepatology Research, 2013.
[80]
A. Alisa, S. Boswell, A. A. Pathan, L. Ayaru, R. Williams, and S. Behboudi, “Human CD4+ T cells recognize an epitope within α-fetoprotein sequence and develop into TGF-β-producing CD4+ T cells,” Journal of Immunology, vol. 180, no. 7, pp. 5109–5117, 2008.
[81]
L. H. Butterfield, A. Koh, W. Meng et al., “Generation of human T-cell responses to an HLA-A2.1-restricted peptide epitope derived from α-fetoprotein,” Cancer Research, vol. 59, no. 13, pp. 3134–3142, 1999.
[82]
L. H. Butterfield, W. S. Meng, A. Koh et al., “T cell responses to HLA-A* 0201-restricted peptides derived from human α fetoprotein,” Journal of Immunology, vol. 166, no. 8, pp. 5300–5308, 2001.
[83]
W. S. Meng, L. H. Butterfield, A. Ribas et al., “Fine specificity analysis of an HLA-A2.1-restricted immunodominant T cell epitope derived from human α-fetoprotein,” Molecular Immunology, vol. 37, no. 16, pp. 943–950, 2001.
[84]
R. Thimme, M. Neagu, T. Boettler et al., “Comprehensive analysis of the α-fetoprotein-specific CD8+ T cell responses in patients with hepatocellular carcinoma,” Hepatology, vol. 48, no. 6, pp. 1821–1833, 2008.
[85]
E. Mizukoshi, Y. Nakamoto, H. Tsuji, T. Yamashita, and S. Kaneko, “Identification of α-fetoprotein-derived peptides recognized by cytotoxic T lymphocytes in HLA-A24+ patients with hepatocellular carcinoma,” International Journal of Cancer, vol. 118, no. 5, pp. 1194–1204, 2006.
[86]
L. H. Butterfield, A. Ribas, W. S. Meng et al., “T-cell responses to HLA-A*0201 immunodominant peptides derived from α-fetoprotein in patients with hepatocellular cancer,” Clinical Cancer Research, vol. 9, no. 16, pp. 5902–5908, 2003.
[87]
F. Tada, M. Abe, M. Hirooka et al., “Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma,” International Journal of Oncology, vol. 41, no. 5, pp. 1601–1609, 2012.
[88]
R. C. Bast Jr., M. Feeney, and H. Lazarus, “Reactivity of a monoclonal antibody with human ovarian carcinoma,” Journal of Clinical Investigation, vol. 68, no. 5, pp. 1331–1337, 1981.
[89]
R. C. Bast Jr., T. L. Klug, and E. St. John, “A radioimmunoassay using a monoclonal antibody to monitor the course of epithelial ovarian cancer,” The New England Journal of Medicine, vol. 309, no. 15, pp. 883–887, 1983.
[90]
A. A. Noujaim, B. C. Schultes, R. P. Baum, and R. Madiyalakan, “Induction of CA125-specific B and T cell responses in patients injected with MAb-B43.13—evidence for antibody-mediated antigen-processing and presentation of CA125 in vivo,” Cancer Biotherapy and Radiopharmaceuticals, vol. 16, no. 3, pp. 187–203, 2001.
[91]
A. N. Gordon, B. C. Schultes, H. Gallion et al., “CA125- and tumor-specific T-cell responses correlate with prolonged survival in oregovomab-treated recurrent ovarian cancer patients,” Gynecologic Oncology, vol. 94, no. 2, pp. 340–351, 2004.
[92]
S. Bellone, S. Anfossi, T. J. O'Brien et al., “Generation of CA125-specific cytotoxic T lymphocytes in human leukocyte antigen-A2.1-positive healthy donors and patients with advanced ovarian cancer,” American Journal of Obstetrics and Gynecology, vol. 200, no. 1, pp. 75.e1–75.e10, 2009.
[93]
T. A. Stamey, N. Yang, and A. R. Hay, “Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate,” The New England Journal of Medicine, vol. 317, no. 15, pp. 909–916, 1987.
[94]
B. Seamonds, B. Whitaker, and N. Yang, “Evaluation of prostate-specific antigen and prostatic acid phosphatase as prostate cancer markers,” Urology, vol. 28, no. 6, pp. 472–479, 1986.
[95]
J. W. Hodge, J. Schlom, S. J. Donohue et al., “A recombinant vaccinia virus expressing human prostate-specific antigen (PSA): safety and immunogenicity in a non-human primate,” International Journal of Cancer, vol. 63, no. 2, pp. 231–237, 1995.
[96]
N. Meidenbauer, D. T. Harris, L. E. Spitler, and T. L. Whiteside, “Generation of PSA-reactive effector cells after vaccination with a PSA-based vaccine in patients with prostate cancer,” Prostate, vol. 43, no. 2, pp. 88–100, 2000.
[97]
S. Perambakam, S. Hallmeyer, S. Reddy et al., “Induction of specific T cell immunity in patients with prostate cancer by vaccination with PSA146-154 peptide,” Cancer Immunology, Immunotherapy, vol. 55, no. 9, pp. 1033–1042, 2006.
[98]
P. Correale, K. Walmsley, C. Nieroda et al., “In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen,” Journal of the National Cancer Institute, vol. 89, no. 4, pp. 293–300, 1997.
[99]
P. Correale, K. Walmsley, S. Zaremba, M. Zhu, J. Schlom, and K. Y. Tsang, “Generation of human cytolytic T lymphocyte lines directed against prostate-specific antigen (PSA) employing a PSA oligoepitope peptide,” Journal of Immunology, vol. 161, no. 6, pp. 3186–3194, 1998.
[100]
J. M. Corman, E. E. Sercarz, and N. K. Nanda, “Recognition of prostate-specific antigenic peptide determinants by human CD4 and CD8 T cells,” Clinical and Experimental Immunology, vol. 114, no. 2, pp. 166–172, 1998.
[101]
M. Harada, K. Kobayashi, S. Matsueda, M. Nakagawa, M. Noguchi, and K. Itoh, “Prostate-specific antigen-derived epitopes capable of inducing cellular and humoral responses in HLA-A24+ prostate cancer patients,” Prostate, vol. 57, no. 2, pp. 152–159, 2003.
[102]
H. S. Lee, S. W. Kim, J. C. Hong et al., “Expression of MAGE A1-6 and the clinical characteristics of papillary thyroid carcinoma,” Anticancer Research, vol. 33, no. 4, pp. 1731–1735, 2013.
[103]
D. W. Meek and L. Marcar, “MAGE-A antigens as targets in tumour therapy,” Cancer Letters, vol. 324, no. 2, pp. 126–132, 2012.
[104]
E. De Plaen, K. Arden, C. Traversari et al., “Structure, chromosomal localization, and expression of 12 genes of the MAGE family,” Immunogenetics, vol. 40, no. 5, pp. 360–369, 1994.
[105]
P. Van Der Bruggen, C. Traversari, P. Chomez et al., “A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma,” Science, vol. 254, no. 5038, pp. 1643–1647, 1991.
[106]
C. Traversari, P. Van der Bruggen, I. F. Luescher et al., “A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E,” Journal of Experimental Medicine, vol. 176, no. 5, pp. 1453–1457, 1992.
[107]
F. Brasseur, M. Marchand, R. Vanwijck et al., “Human gene MAGE-1, which codes for a tumor-rejection antigen, is expressed by some breast tumors,” International Journal of Cancer, vol. 52, no. 5, pp. 839–841, 1992.
[108]
P. Chaux, R. Luiten, N. Demotte et al., “Identification of five MAGE-A1 epitopes recognized by cytolytic T lymphocytes obtained by in vitro stimulation with dendritic cells transduced with MAGE-A1,” Journal of Immunology, vol. 163, no. 5, pp. 2928–2936, 1999.
[109]
P. Chaux, V. Vantomme, V. Stroobant et al., “Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4+ T lymphocytes,” Journal of Experimental Medicine, vol. 189, no. 5, pp. 767–778, 1999.
[110]
M. T. Duffour, P. Chaux, C. Lurquin, G. Cornelis, T. Boon, and P. van der Bruggen, “MAGE-A4 peptide presented by HLA-A2 is recognized by cytolytic T lymphocytes,” European Journal of Immunology, vol. 29, no. 10, pp. 3329–3337, 1999.
[111]
C. Germeau, W. Ma, F. Schiavetti et al., “High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens,” Journal of Experimental Medicine, vol. 201, no. 2, pp. 241–248, 2005.
[112]
T. Cohan, R. M. Muller, Y. Tomita, and S. Shibahara, “Nucleotide sequence of the cDNA encoding human tyrosinase-related protein,” Nucleic Acids Research, vol. 18, no. 9, pp. 2807–2808, 1990.
[113]
I. J. Jackson, D. M. Chambers, K. Tsukamoto et al., “A second tyrosinase-related protein, TRP-2, maps to and is mutated at the mouse slaty locus,” EMBO Journal, vol. 11, no. 2, pp. 527–535, 1992.
[114]
R.-F. Wang, E. Appella, Y. Kawakami, X. Kang, and S. A. Rosenberg, “Identification of TRP-2 as a human tumor antigen recognized by cytotoxic T lymphocytes,” Journal of Experimental Medicine, vol. 184, no. 6, pp. 2207–2216, 1996.
[115]
R.-F. Wang, S. L. Johnston, S. Southwood, A. Sette, and S. A. Rosenberg, “Recognition of an antigenic peptide derived from tyrosinase-related protein-2 by CTL in the context of HLA-A31 and -A33,” Journal of Immunology, vol. 160, no. 2, pp. 890–897, 1998.
[116]
M. R. Parkhurst, E. B. Fitzgerald, S. Southwood, A. Sette, S. A. Rosenberg, and Y. Kawakami, “Identification of a shared HLA-A*0201-restricted T-cell epitope from the melanoma antigen tyrosinase-related protein 2 (TRP2),” Cancer Research, vol. 58, no. 21, pp. 4895–4901, 1998.
[117]
C. Castelli, P. Tarsini, A. Mazzocchi et al., “Novel HLA-Cw8-restricted T cell epitopes derived from tyrosinase-related protein-2 and gp100 melanoma antigens,” Journal of Immunology, vol. 162, no. 3, pp. 1739–1748, 1999.
[118]
C. Noppen, F. Levy, L. Burri et al., “Naturally processed and concealed HLA-A2. 1-restricted epitopes from tumor-associated antigen tyrosinase-related protein-2,” International Journal of Cancer, vol. 87, no. 2, pp. 241–246, 2000.
[119]
Y. Sun, M. Song, S. Stevanovic et al., “Identification of a new HLA-A(*)0201-restricted T-cell epitope from the tyrosinase-related protein 2 (TRP2) melanoma antigen,” International Journal of Cancer, vol. 87, no. 3, pp. 399–404, 2000.
[120]
Y. Liu, Y. Peng, M. Mi et al., “Lentivector immunization stimulates potent CD8 T cell responses against melanoma self-antigen tyrosinase-related protein 1 and generates antitumor immunity in mice,” Journal of Immunology, vol. 182, no. 10, pp. 5960–5969, 2009.
[121]
W. Osen, S. Soltek, M. Song et al., “Screening of human tumor antigens for CD4+ T cell epitopes by combination of HLA-transgenic mice, recombinant adenovirus and antigen peptide libraries,” PLoS ONE, vol. 5, no. 11, Article ID e14137, 2010.
[122]
S. R. Sierro, A. Donda, R. Perret et al., “Combination of lentivector immunization and low-dose chemotherapy or PD-1/PD-L1 blocking primes self-reactive T cells and induces anti-tumor immunity,” European Journal of Immunology, vol. 41, no. 8, pp. 2217–2228, 2011.
[123]
W. W. Overwijk, D. S. Lee, D. R. Surman et al., “Vaccination with a recombinant vaccinia virus encoding a “self” antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4+ T lymphocytes,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 6, pp. 2982–2987, 1999.
[124]
H. T. Khong and S. A. Rosenberg, “Pre-existing immunity to tyrosinase-related protein (TRP)-2, a new TRP-2 isoform, and the NY-ESO-1 melanoma antigen in a patient with a dramatic response to immunotherapy,” Journal of Immunology, vol. 168, no. 2, pp. 951–956, 2002.
[125]
V. Bronte, E. Apolloni, R. Ronca et al., “Genetic vaccination with 'self' tyrosinase-related protein 2 causes melanoma eradication but not vitiligo,” Cancer Research, vol. 60, no. 2, pp. 253–258, 2000.
[126]
C. Vennegoor, H. P. Hageman Ph., H. Van Nouhuijs et al., “A monoclonal antibody specific for cells of the melanocyte lineage,” American Journal of Pathology, vol. 130, no. 1, pp. 179–192, 1988.
[127]
A. B. H. Bakker, M. W. J. Schreurs, A. J. De Boer et al., “Melanocyte lineage-specific antigen gp100 is recognized by melanoma-derived tumor-infiltrating lymphocytes,” Journal of Experimental Medicine, vol. 179, no. 3, pp. 1005–1009, 1994.
[128]
G. J. Adema, A. J. De Boer, A. M. Vogel, W. A. M. Loenen, and C. G. Figdor, “Molecular characterization of the melanocyte lineage-specific antigen gp100,” Journal of Biological Chemistry, vol. 269, no. 31, pp. 20126–20133, 1994.
[129]
Y.-T. Chen, A. D. Boyer, C. S. Viars, S. Tsang, L. J. Old, and K. C. Arden, “Genomic cloning and localization of CTAG, a gene encoding an autoimmunogenic cancer-testis antigen NY-ESO-1, to human chromosome Xq28,” Cytogenetics and Cell Genetics, vol. 79, no. 3-4, pp. 237–240, 1997.
[130]
Y.-T. Chen, M. J. Scanlan, U. Sahin et al., “A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 5, pp. 1914–1918, 1997.
[131]
S. Eikawa, K. Kakimi, M. Isobe et al., “Induction of CD8 T-cell responses restricted to multiple HLA class I alleles in a cancer patient by immunization with a 20-mer NY-ESO-1f (NY-ESO-1 91-110) peptide,” International Journal of Cancer, vol. 132, no. 2, pp. 345–354, 2013.
[132]
Y. Mizote, T. Taniguchi, K. Tanaka et al., “Three novel NY-ESO-1 epitopes bound to DRB10803, DQB10401 and DRB1 0901 recognized by CD4 T cells from CHP-NY-ESO-1-vaccinated patients,” Vaccine, vol. 28, no. 32, pp. 5338–5346, 2010.
[133]
N. C. Robson, T. McAlpine, A. J. Knights et al., “Processing and cross-presentation of individual HLA-A, -B, or -C epitopes from NY-ESO-1 or an HLA-A epitope for Melan-A differ according to the mode of antigen delivery,” Blood, vol. 116, no. 2, pp. 218–225, 2010.
[134]
D. Escors, L. Lopes, R. Lin et al., “Targeting dendritic cell signaling to regulate the response to immunization,” Blood, vol. 111, no. 6, pp. 3050–3061, 2008.
[135]
E. J?ger, D. J?ger, J. Karbach et al., “Identification of NY-ESO-1 epitopes presented by human histocompatibility antigen (HLA)-DRB4(*)0101-0103 and recognized by CD4+ T lymphocytes of patients with NY-ESO-1-expressing melanoma,” Journal of Experimental Medicine, vol. 191, no. 4, pp. 625–630, 2000.
[136]
K. Odunsi, J. Matsuzaki, J. Karbach et al., “Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 15, pp. 5797–5802, 2012.
[137]
S. Gnjatic, H. Nishikawa, A. A. Jungbluth et al., “NY-ESO-1: review of an immunogenic tumor antigen,” Advances in Cancer Research, vol. 95, pp. 1–30, 2006.
[138]
J. Campos-Perez, J. Rice, D. Escors et al., “DNA fusion vaccine designs to induce tumor-lytic CD8+ T-cell attack via the immunodominant cysteine-containing epitope of NY-ESO 1,” International Journal of Cancer, vol. 133, no. 6, pp. 1400–1407, 2013.
[139]
P. F. Robbins, R. A. Morgan, S. A. Feldman et al., “Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1,” Journal of Clinical Oncology, vol. 29, no. 7, pp. 917–924, 2011.
[140]
Y. Kawakami, S. Eliyahu, C. H. Delgado et al., “Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 14, pp. 6458–6462, 1994.
[141]
Y. Kawakami, S. Eliyahu, K. Sakaguchi et al., “Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes,” Journal of Experimental Medicine, vol. 180, no. 1, pp. 347–352, 1994.
[142]
K. S. Lang, C. C. Caroli, A. Muhm et al., “HLA-A2 restricted, melanocyte-specific CD8+ T lymphocytes detected in vitiligo patients are related to disease activity and are predominantly directed against MelanA/MART1,” Journal of Investigative Dermatology, vol. 116, no. 6, pp. 891–897, 2001.
[143]
S. Bobisse, M. Rondina, A. Merlo et al., “Reprogramming T lymphocytes for melanoma adoptive immunotherapy by T-cell receptor gene transfer with lentiviral vectors,” Cancer Research, vol. 69, no. 24, pp. 9385–9394, 2009.
[144]
M. Coccoris, T. Straetemans, C. Govers, C. Lamers, S. Sleijfer, and R. Debets, “T cell receptor (TCR) gene therapy to treat melanoma: lessons from clinical and preclinical studies,” Expert Opinion on Biological Therapy, vol. 10, no. 4, pp. 547–562, 2010.
[145]
R. A. Morgan, M. E. Dudley, J. R. Wunderlich et al., “Cancer regression in patients after transfer of genetically engineered lymphocytes,” Science, vol. 314, no. 5796, pp. 126–129, 2006.
[146]
V. Shankaran, H. Ikeda, A. T. Bruce et al., “IFNγ, and lymphocytes prevent primary tumour development and shape tumour immunogenicity,” Nature, vol. 410, no. 6832, pp. 1107–1111, 2001.
[147]
C. Bricogne, R. Laranga, A. Padella et al., “Critical mass hypothesis of T cell responses and its application for the treatment of T cell lymphoma,” in Hodgkin's and T-Cell Lymphoma: Diagnosis, Treatment Options and Prognosis, W. K. Harvey and R. M. Jacobs, Eds., Nova Publishers, New York, NY, USA, 2012.
[148]
K. Karwacz, F. Arce, C. Bricogne, G. Kochan, and D. Escors, “PD-L1 co-stimulation, ligand-induced TCR down-modulation and anti-tumor immunotherapy,” Oncoimmunology, vol. 1, no. 1, pp. 86–88, 2012.
[149]
K. Karwacz, C. Bricogne, D. MacDonald et al., “PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8+ T cells,” EMBO Molecular Medicine, vol. 3, no. 10, pp. 581–592, 2011.
[150]
K. Trnkova, S. Pastorekova, and J. Petrik, “Novel approaches to antiviral and anticancer immunotherapy,” Acta Virologica, vol. 56, no. 4, pp. 271–282, 2012.
[151]
E. Mohit and S. Rafati, “Chemokine-based immunotherapy: delivery systems and combination therapies,” Immunotherapy, no. 8, pp. 807–840, 2012.
[152]
S. Ghafouri-Fard, “siRNA and cancer immunotherapy,” Immunotherapy, vol. 4, no. 9, pp. 907–917, 2012.
[153]
T. Liechtenstein, I. Dufait, A. Lanna, K. Breckpot, and D. Escors, “Modulating co-stimulation during antigen presentation to enhance cancer immunotherapy,” Immunology, Endocrine & Metabolic Agents in Medicinal Chemistry, vol. 12, no. 3, pp. 224–235, 2012.
[154]
T. Liechtenstein, N. Perez-Janices, C. Bricogne et al., “Immune modulation by genetic modification of dendritic cells with lentiviral vectors,” Virus Research, vol. 176, no. 1-2, pp. 1–15, 2013.
[155]
D. Escors, C. Bricogne, F. Arce, G. Kochan, and K. Karwacz, “On the Mechanism of T cell receptor down-modulation and its physiological significance,” The Journal of Bioscience and Medicine, vol. 1, no. 1, 2012.
[156]
M. R. Parkhurst, J. C. Yang, R. C. Langan et al., “T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis,” Molecular Therapy, vol. 19, no. 3, pp. 620–626, 2011.
[157]
L. A. Johnson, B. Heemskerk, D. J. Powell Jr. et al., “Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes,” Journal of Immunology, vol. 177, no. 9, pp. 6548–6559, 2006.
[158]
M. Schnurr, P. Galambos, C. Scholz et al., “Tumor cell lysate-pulsed human dendritic cells induce a T-cell response against pancreatic carcinoma cells: an in vitro model for the assessment of tumor vaccines,” Cancer Research, vol. 61, no. 17, pp. 6445–6450, 2001.
[159]
B. Bodey, B. Bodey Jr., S. E. Siegel, and H. E. Kaiser, “Failure of cancer vaccines: the significant limitations of this approach to immunotherapy,” Anticancer Research, vol. 20, no. 4, pp. 2665–2676, 2000.
[160]
R. Nurieva, S. Thomas, T. Nguyen et al., “T-cell tolerance or function is determined by combinatorial costimulatory signals,” EMBO Journal, vol. 25, no. 11, pp. 2623–2633, 2006.
[161]
T. Liechtenstein, I. Dufait, C. Bricogne et al., “PD-L1/PD-1 co-stimulation, a brake for T cell activation and a T cell differentiation signal,” Journal of Clinical & Cellular Immunology, 2012.
[162]
L. S. K. Walker, A. Chodos, M. Eggena, H. Dooms, and A. K. Abbas, “Antigen-dependent proliferation of CD4+ CD25+ regulatory T cells in vivo,” Journal of Experimental Medicine, vol. 198, no. 2, pp. 249–258, 2003.
[163]
K. Kretschmer, I. Apostolou, D. Hawiger, K. Khazaie, M. C. Nussenzweig, and H. von Boehmer, “Inducing and expanding regulatory T cell populations by foreign antigen,” Nature Immunology, vol. 6, no. 12, pp. 1219–1227, 2005.
[164]
T. Liechtenstein, N. Perez-Janices, and D. Escors, “Lentiviral vectors for cancer immunotherapy and clinical applications,” Cancers, vol. 5, no. 3, pp. 815–837, 2013.
[165]
S. Sakaguchi, T. Yamaguchi, T. Nomura, and M. Ono, “Regulatory T cells and immune tolerance,” Cell, vol. 133, no. 5, pp. 775–787, 2008.
[166]
R. M. Steinman and Z. A. Cohn, “Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution,” The Journal of Experimental Medicine, vol. 137, no. 5, pp. 1142–1162, 1973.
[167]
R. M. Steinman and M. D. Witmer, “Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 75, no. 10, pp. 5132–5136, 1978.
[168]
M. V. Dhodapkar, R. M. Steinman, J. Krasovsky, C. Munz, and N. Bhardwaj, “Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells,” Journal of Experimental Medicine, vol. 193, no. 2, pp. 233–238, 2001.
[169]
D. Hawiger, K. Inaba, Y. Dorsett et al., “Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo,” Journal of Experimental Medicine, vol. 194, no. 6, pp. 769–779, 2001.
[170]
K. Inaba, M. Inaba, N. Romani et al., “Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor,” Journal of Experimental Medicine, vol. 176, no. 6, pp. 1693–1702, 1992.
[171]
L.-J. Zhou and T. F. Tedder, “CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 6, pp. 2588–2592, 1996.
[172]
S. Tuyaerts, S. M. Noppe, J. Corthals et al., “Generation of large numbers of dendritic cells in a closed system using Cell Factories,” Journal of Immunological Methods, vol. 264, no. 1-2, pp. 135–151, 2002.
[173]
C. A. Janeway Jr. and K. Bottomly, “Signals and signs for lymphocyte responses,” Cell, vol. 76, no. 2, pp. 275–285, 1994.
[174]
P. Matzinger, “Tolerance, danger, and the extended family,” Annual Review of Immunology, vol. 12, pp. 991–1045, 1994.
[175]
M. Dullaers, K. Breckpot, S. Van Meirvenne et al., “Side-by-side comparison of lentivirally transduced and mRNA-electroporated dendritic cells: implications for cancer immunotherapy protocols,” Molecular Therapy, vol. 10, no. 4, pp. 768–779, 2004.
[176]
E. Andreakos, R. O. Williams, J. Wales, B. M. Foxwell, and M. Feldmann, “Activation of NF-κB by the intracellular expression of NF-κB-inducing kinase acts as a powerful vaccine adjuvant,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 39, pp. 14459–14464, 2006.
[177]
S. Van Lint, C. Goyvaerts, S. Maenhout et al., “Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy,” Cancer Research, vol. 72, no. 7, pp. 1661–1671, 2012.
[178]
K. Karwacz, S. Mukherjee, L. Apolonia et al., “Nonintegrating lentivector vaccines stimulate prolonged T-cell and antibody responses and are effective in tumor therapy,” Journal of Virology, vol. 83, no. 7, pp. 3094–3103, 2009.
[179]
A. Gruber, J. Kan-Mitchell, K. L. Kuhen, T. Mukai, and F. Wong-Staal, “Dendritic cells transduced by multiply deleted HIV-1 vectors exhibit normal phenotypes and functions and elicit an HIV-specific cytotoxic T-lymphocyte response in vitro,” Blood, vol. 96, no. 4, pp. 1327–1333, 2000.
[180]
J. Dyall, J.-B. Latouche, S. Schnell, and M. Sadelain, “Lentivirus-transduced human monocyte-derived dendritic cells efficiently stimulate antigen-specific cytotoxic T lymphocytes,” Blood, vol. 97, no. 1, pp. 114–121, 2001.
[181]
J.-F. Arrighi, M. Pion, M. Wiznerowicz et al., “Lentivirus-mediated RNA interference of DC-SIGN expression inhibits human immunodeficiency virus transmission from dendritic cells to T cells,” Journal of Virology, vol. 78, no. 20, pp. 10848–10855, 2004.
[182]
Q. Yu, C. Kovacs, F. Y. Yue, and M. A. Ostrowski, “The role of the p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and phosphoinositide-3-OH kinase signal transduction pathways in CD40 ligand-induced dendritic cell activation and expansion of virus-specific CD8+ T cell memory responses,” Journal of Immunology, vol. 172, no. 10, pp. 6047–6056, 2004.
[183]
J. J. Pen, B. De Keersmaecker, S. K. Maenhout et al., “Modulation of regulatory T cell function by monocyte-derived dendritic cells matured through electroporation with mRNA encoding CD40 ligand, constitutively Active TLR4, and CD70,” Journal of Immunology, vol. 191, no. 4, pp. 1976–1983, 2013.
[184]
C. Klein, H. Bueler, and R. C. Mulligan, “Comparative analysis of genetically modified dendritic cells and tumor cells as therapeutic cancer vaccines,” Journal of Experimental Medicine, vol. 191, no. 10, pp. 1699–1708, 2000.
[185]
B. Hu, B. Dai, and P. Wang, “Vaccines delivered by integration-deficient lentiviral vectors targeting dendritic cells induces strong antigen-specific immunity,” Vaccine, vol. 28, no. 41, pp. 6675–6683, 2010.
[186]
T. Akazawa, M. Shingai, M. Sasai et al., “Tumor immunotherapy using bone marrow-derived dendritic cells overexpressing Toll-like receptor adaptors,” FEBS Letters, vol. 581, no. 18, pp. 3334–3340, 2007.
[187]
K. Breckpot, C. Aerts-Toegaert, C. Heirman et al., “Attenuated expression of A20 markedly increases the efficacy of double-stranded RNA-activated dendritic cells as an anti-cancer vaccine,” Journal of Immunology, vol. 182, no. 2, pp. 860–870, 2009.
[188]
C. L.-L. Chiang, A. R. Hagemann, R. Leskowitz et al., “Day-4 myeloid dendritic cells pulsed with whole tumor lysate are highly immunogenic and elicit potent anti-tumor responses,” PLoS ONE, vol. 6, no. 12, Article ID e28732, 2011.
[189]
F. Arce, K. Breckpot, H. Stephenson et al., “Selective ERK activation differentiates mouse and human tolerogenic dendritic cells, expands antigen-specific regulatory T cells, and suppresses experimental inflammatory arthritis,” Arthritis and Rheumatism, vol. 63, no. 1, pp. 84–95, 2011.
[190]
S. Rutella, S. Danese, and G. Leone, “Tolerogenic dendritic cells: cytokine modulation comes of age,” Blood, vol. 108, no. 5, pp. 1435–1440, 2006.
[191]
D. Kabelitz, D. Wesch, and H.-H. Oberg, “Regulation of regulatory T cells: role of dendritic cells and toll-like receptors,” Critical Reviews in Immunology, vol. 26, no. 4, pp. 291–306, 2006.
[192]
M. G. Toscano, M. Delgado, W. Kong, F. Martin, M. Skarica, and D. Ganea, “Dendritic cells transduced with lentiviral vectors expressing vip differentiate into vip-secreting tolerogenic-like DCs,” Molecular Therapy, vol. 18, no. 5, pp. 1035–1045, 2010.
[193]
J. M. Ilarregui, D. O. Croci, G. A. Bianco et al., “Tolerogenic signals delivered by dendritic cells to T cells through a galectin-1-driven immunoregulatory circuit involving interleukin 27 and interleukin 10,” Nature Immunology, vol. 10, no. 9, pp. 981–991, 2009.
[194]
K. V. Tarbell, L. Petit, X. Zuo et al., “Dendritic cell-expanded, islet-specific CD4+ CD25+ CD62L+ regulatory T cells restore normoglycemia in diabetic NOD mice,” Journal of Experimental Medicine, vol. 204, no. 1, pp. 191–201, 2007.
[195]
L. N. Liu, R. Shivakumar, C. Allen, and J. C. Fratantoni, “Delivery of whole tumor lysate into dendritic cells for cancer vaccination,” Methods in Molecular Biology, vol. 423, pp. 139–153, 2008.
[196]
J. P. J. J. Hegmans, A. Hemmes, J. G. Aerts, H. C. Hoogsteden, and B. N. Lambrecht, “Immunotherapy of murine malignant mesothelioma using tumor lysate-pulsed dendritic cells,” American Journal of Respiratory and Critical Care Medicine, vol. 171, no. 10, pp. 1168–1177, 2005.
[197]
N. Herbert, A. Haferkamp, H. F. Schmitz-Winnenthal, and M. Z?ller, “Concomitant tumor and autoantigen vaccination supports renal cell carcinoma rejection,” Journal of Immunology, vol. 185, no. 2, pp. 902–916, 2010.
[198]
B. J. Gitlitz, A. S. Belldegrun, A. Zisman et al., “A pilot trial of tumor lysate-loaded dendritic cells for the treatment of metastatic renal cell carcinoma,” Journal of Immunotherapy, vol. 26, no. 5, pp. 412–419, 2003.
[199]
H. M. Zarour and J. M. Kirkwood, “Melanoma vaccines: early progress and future promises,” Seminars in Cutaneous Medicine and Surgery, vol. 22, no. 1, pp. 68–75, 2003.
[200]
M. Leslie, “Solution to vaccine mystery starts to crystallize,” Science, vol. 341, no. 6141, pp. 26–27, 2013.
[201]
T. L. Flach, G. Ng, A. Hari et al., “Alum interaction with dendritic cell membrane lipids is essential for its adjuvanticity,” Nature Medicine, vol. 17, no. 4, pp. 479–487, 2011.
[202]
H. Xiao, Y. Peng, Y. Hong et al., “Local administration of TLR ligands rescues the function of tumor-infiltrating CD8 T cells and enhances the antitumor effect of lentivector immunization,” Journal of Immunology, vol. 190, no. 11, pp. 5866–5873, 2013.
[203]
S. N. Lim, S. Kuhn, E. Hyde, and F. Ronchese, “Combined TLR stimulation with Pam3Cys and Poly I: C enhances Flt3-ligand dendritic cell activation for tumor immunotherapy,” Journal of Immunotherapy, vol. 35, no. 9, pp. 670–679, 2012.
[204]
T. Kawai and S. Akira, “Toll-like receptor and RIG-1-like receptor signaling,” Annals of the New York Academy of Sciences, vol. 1143, pp. 1–20, 2008.
[205]
B. Coornaert, I. Carpentier, and R. Beyaert, “A20: central gatekeeper in inflammation and immunity,” Journal of Biological Chemistry, vol. 284, no. 13, pp. 8217–8221, 2009.
[206]
X.-T. Song, K. E. Kabler, L. Shen, L. Rollins, X. F. Huang, and S.-Y. Chen, “A20 is an antigen presentation attenuator, and its inhibition overcomes regulatory T cell-mediated suppression,” Nature Medicine, vol. 14, no. 3, pp. 258–265, 2008.
[207]
T. Yokosuka, M. Takamatsu, W. Kobayashi-Imanishi, A. Hashimoto-Tane, M. Azuma, and T. Saito, “Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2,” Journal of Experimental Medicine, vol. 209, no. 6, pp. 1201–1217, 2012.
[208]
M. R. Junttila, S.-P. Li, and J. Westermarck, “Phosphatase-mediated crosstalk between MAPK signaling pathways in the regulation of cell survival,” FASEB Journal, vol. 22, no. 4, pp. 954–965, 2008.
[209]
Q. Zhao, X. Wang, L. D. Nelin et al., “MAP kinase phosphatase 1 controls innate immune responses and suppresses endotoxic shock,” Journal of Experimental Medicine, vol. 203, no. 1, pp. 131–140, 2006.
[210]
K. V. Salojin, I. B. Owusu, K. A. Millerchip, M. Potter, K. A. Platt, and T. Oravecz, “Essential role of MAPK phosphatase-1 in the negative control of innate immune responses,” Journal of Immunology, vol. 176, no. 3, pp. 1899–1907, 2006.
[211]
M. Hammer, J. Mages, H. Dietrich et al., “Dual specificity phosphatase 1 (DUSP1) regulates a subset of LPS-induced genes and protects mice from lethal endotoxin shock,” Journal of Experimental Medicine, vol. 203, no. 1, pp. 15–20, 2006.
[212]
S. Saccani, S. Pantano, and G. Natoli, “p38-dependent marking of inflammatory genes for increased NF-κB recruitment,” Nature Immunology, vol. 3, no. 1, pp. 69–75, 2002.
[213]
A. Shimizu, M. Baratchian, Y. Takeuchi et al., “Kaposi's sarcoma-associated herpesvirus vFLIP and human T cell lymphotropic virus type 1 Tax oncogenic proteins activate IκB kinase subunit γ by different mechanisms independent of the physiological cytokine-induced pathways,” Journal of Virology, vol. 85, no. 14, pp. 7444–7448, 2011.
[214]
N. Field, W. Low, M. Daniels et al., “KSHV vFLIP binds to IKK-γ to activate IKK,” Journal of Cell Science, vol. 116, no. 18, pp. 3721–3728, 2003.
[215]
C. Bagnéris, A. V. Ageichik, N. Cronin et al., “Crystal structure of a vFlip-IKKγ complex: insights into viral activation of the IKK signalosome,” Molecular Cell, vol. 30, no. 5, pp. 620–631, 2008.
[216]
H. M. Rowe, L. Lopes, N. Brown et al., “Expression of vFLIP in a lentiviral vaccine vector activates NF-κB, matures dendritic cells, and increases CD8+ T-cell responses,” Journal of Virology, vol. 83, no. 4, pp. 1555–1562, 2009.
[217]
I. E. Wartz, K. M. O'Rourke, H. Zhou et al., “De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-κB signalling,” Nature, vol. 430, no. 7000, pp. 694–699, 2004.
[218]
R. Beyaert, K. Heyninck, and S. Van Huffel, “A20 and A20-binding proteins as cellular inhibitors of nuclear factor- κB-dependent gene expression and apoptosis,” Biochemical Pharmacology, vol. 60, no. 8, pp. 1143–1151, 2000.
[219]
S. Q. Zhang, A. Kovalenko, G. Cantarella, and D. Wallach, “Recruitment of the IKK signalosome to the p55 TNF receptor: RIP and A20 bind to NEMO (IKKγ) upon receptor stimulation,” Immunity, vol. 12, no. 3, pp. 301–311, 2000.
[220]
K. Heyninck and R. Beyaert, “The cytokine-inducible zinc finger protein A20 inhibits IL-1-induced NF-κB activation at the level of TRAF6,” FEBS Letters, vol. 442, no. 2-3, pp. 147–150, 1999.
[221]
D. L. Boone, E. E. Turer, E. G. Lee et al., “The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses,” Nature Immunology, vol. 5, no. 10, pp. 1052–1060, 2004.
[222]
K. Sato, H. Nagayama, K. Tadokoro, T. Juji, and T. A. Takahashi, “Extracellular signal-regulated kinase, stress-activated protein kinase/c-Jun N-terminal kinase, and p38(mapk) are involved in IL-10-mediated selective repression of TNF-α-induced activation and maturation of human peripheral blood monocyte-derived dendritic cells,” Journal of Immunology, vol. 162, no. 7, pp. 3865–3872, 1999.
[223]
K. M. Ardeshna, A. R. Pizzey, S. Devereux, and A. Khwaja, “The PI3 kinase, p38 SAP kinase, and NF-κb signal transduction pathways are involved in the survival and maturation of lipopolysaccharide-stimulated human monocyte-derived dendritic cells,” Blood, vol. 96, no. 3, pp. 1039–1046, 2000.
[224]
J.-F. Arrighi, M. Rebsamen, F. Rousset, V. Kindler, and C. Hauser, “A critical role for p38 mitogen-activated protein kinase in the maturation of human blood-derived dendritic cells induced by lipopolysaccharide, TNF-α, and contact sensitizers,” Journal of Immunology, vol. 166, no. 6, pp. 3837–3845, 2001.
[225]
J. Raingeaud, A. J. Whitmarsh, T. Barrett, B. Dérijard, and R. J. Davis, “MKK3- and MKK6-regulated gene expression is mediated by the p38 mitogen-activated protein kinase signal transduction pathway,” Molecular and Cellular Biology, vol. 16, no. 3, pp. 1247–1255, 1996.
[226]
Z. Yang, X. Zhang, P. A. Darrah, and D. M. Mosser, “The regulation of Th1 responses by the p38 MAPK,” Journal of Immunology, vol. 185, no. 10, pp. 6205–6213, 2010.
[227]
H. A. Franks, Q. Wang, S. J. Lax et al., “Novel function for the p38-MK2 signalling pathway in circulating CD1c+ (BDCA-1+) myeloid dendritic cells from healthy donors and advanced cancer patients, inhibition of p38 enhances IL-12 whilst suppressing IL-10,” International Journal of Cancer, 2013.
[228]
H. Firat, F. Garcia-Pons, S. Tourdot et al., “H-2 class I knockout, HLA-A2. 1-transgenic mice: a versatile animal model for preclinical evaluation of antitumor immunotherapeutic strategies,” European Journal of Immunology, vol. 29, no. 10, pp. 3112–3121, 1999.
[229]
E. Kriehuber, W. Bauer, A.-S. Charbonnier et al., “Balance between NF-κB and JNK/AP-1 activity controls dendritic cell life and death,” Blood, vol. 106, no. 1, pp. 175–183, 2005.
[230]
T. Nakahara, H. Uchi, K. Urabe, Q. Chen, M. Furue, and Y. Moroi, “Role of c-Jun N-terminal kinase on lipopolysaccharide induced maturation of human monocyte-derived dendritic cells,” International Immunology, vol. 16, no. 12, pp. 1701–1709, 2004.
[231]
K. Lei, A. Nimnual, W.-X. Zong et al., “The Bax subfamily of Bcl2-related proteins is essential for apoptotic signal transduction by c-Jun NH2-terminal kinase,” Molecular and Cellular Biology, vol. 22, no. 13, pp. 4929–4942, 2002.
[232]
M. Dupage, C. Mazumdar, L. M. Schmidt, A. F. Cheung, and T. Jacks, “Expression of tumour-specific antigens underlies cancer immunoediting,” Nature, vol. 482, no. 7385, pp. 405–409, 2012.
[233]
A. D. Griesemer, E. C. Sorenson, and M. A. Hardy, “The role of the thymus in tolerance,” Transplantation, vol. 90, no. 5, pp. 465–474, 2010.
[234]
F. Flores-Borja, C. Mauri, and M. R. Ehrenstein, “Restoring the balance: harnessing regulatory T cells for therapy in rheumatoid arthritis,” European Journal of Immunology, vol. 38, no. 4, pp. 934–937, 2008.
[235]
R. R. Rich and C. W. Pierce, “Biological expressions of lymphocyte activation. II. Generation of a population of thymus-derived suppressor lymphocytes,” Journal of Experimental Medicine, vol. 137, no. 3, pp. 649–659, 1973.
[236]
E. Simpson and H. Cantor, “Regulation of the immune response by subclasses of T lymphocytes. II. The effect of adult thymectomy upon humoral and cellular responses in mice,” European Journal of Immunology, vol. 5, no. 5, pp. 337–343, 1975.
[237]
H. Cantor and E. Simpson, “Regulation of the immune response by subclasses of T lymphocytes. I. Interactions between pre killer T cells and regulatory T cells obtained from peripheral lymphoid tissues of mice,” European Journal of Immunology, vol. 5, no. 5, pp. 330–336, 1975.
[238]
T. Y. H. Tai You Ha, B. H. Waksman, and H. P. Treffers, “The thymic suppressor cell. I. Separation of subpopulations with suppressor activity,” Journal of Experimental Medicine, vol. 139, no. 1, pp. 13–23, 1974.
[239]
M. J. Taussig, “Demonstration of suppressor T cells in a population of 'educated' T cells,” Nature, vol. 248, no. 5445, pp. 236–238, 1974.
[240]
L. Polak and J. L. Turk, “Reversal of immunological tolerance by cyclophosphamide through inhibition of suppressor cell activity,” Nature, vol. 249, no. 5458, pp. 654–656, 1974.
[241]
S. Kan, S. Hazama, K. Maeda et al., “Suppressive effects of cyclophosphamide and gemcitabine on regulatory T-cell induction in vitro,” Anticancer Research, vol. 32, no. 12, pp. 5363–5369, 2012.
[242]
H. Kirchner, T. M. Chused, and R. B. Herberman, “Evidence of suppressor cell activity in spleens of mice bearing primary tumors induced by Moloney sarcoma virus,” Journal of Experimental Medicine, vol. 139, no. 6, pp. 1473–1487, 1974.
[243]
D. I. Gabrilovich, V. Bronte, S.-H. Chen et al., “The terminology issue for myeloid-derived suppressor cells,” Cancer Research, vol. 67, no. 1, article 425, 2007.
[244]
A. Basten, J. F. A. P. Miller, J. Sprent, and C. Cheers, “Cell to cell interaction in the immune response. X. T cell dependent suppression in tolerant mice,” Journal of Experimental Medicine, vol. 140, no. 1, pp. 199–217, 1974.
[245]
D. M. Sansom, “CD28, CTLA-4 and their ligands: who does what and to whom?” Immunology, vol. 101, no. 2, pp. 169–177, 2000.
[246]
O. S. Qureshi, Y. Zheng, K. Nakamura et al., “Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4,” Science, vol. 332, no. 6029, pp. 600–603, 2011.
[247]
L. S. K. Walker and D. M. Sansom, “The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses,” Nature Reviews Immunology, vol. 11, no. 12, pp. 852–863, 2011.
[248]
S. Hori, T. Nomura, and S. Sakaguchi, “Control of regulatory T cell development by the transcription factor Foxp3,” Science, vol. 299, no. 5609, pp. 1057–1061, 2003.
[249]
J. Schwaber and E. P. Cohen, “Human X mouse somatic cell hybrid clone secreting immunoglobulins of both parental types,” Nature, vol. 244, no. 5416, pp. 444–447, 1973.
[250]
S. Sakaguchi, “The origin of FOXP3-expressing CD4+ regulatory T cells: thymus or periphery,” Journal of Clinical Investigation, vol. 112, no. 9, pp. 1310–1312, 2003.
[251]
X. C. Li, G. Demirci, S. Ferrari-Lacraz et al., “IL-15 and IL-2: a matter of life and death for T cells in vivo,” Nature Medicine, vol. 7, no. 1, pp. 114–118, 2001.
[252]
A. Scheffold, J. Hühn, and T. H?fer, “Regulation of CD4+CD25+ regulatory T cell activity: it takes (IL-)two to tango,” European Journal of Immunology, vol. 35, no. 5, pp. 1336–1341, 2005.
[253]
J. D. Fontenot, J. P. Rasmussen, M. A. Gavin, and A. Y. Rudensky, “A function for interleukin 2 in Foxp3-expressing regulatory T cells,” Nature Immunology, vol. 6, no. 11, pp. 1142–1151, 2005.
[254]
D. Getnet, J. F. Grosso, M. V. Goldberg et al., “A role for the transcription factor Helios in human CD4+CD25+ regulatory T cells,” Molecular Immunology, vol. 47, no. 7-8, pp. 1595–1600, 2010.
[255]
P. Chappert, M. Leboeuf, P. Rameau et al., “Antigen-driven interactions with dendritic cells and expansion of Foxp3+ regulatory T cells occur in the absence of inflammatory signals,” Journal of Immunology, vol. 180, no. 1, pp. 327–334, 2008.
[256]
K. Mahnke, Y. Qian, J. Knop, and A. H. Enk, “Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells,” Blood, vol. 101, no. 12, pp. 4862–4869, 2003.
[257]
L. Wang, K. Pino-Lagos, V. C. De Vries, I. Guleria, M. H. Sayegh, and R. J. Noelle, “Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 27, pp. 9331–9336, 2008.
[258]
S. You, B. Leforban, C. Garcia, J.-F. Bach, J. A. Bluestone, and L. Chatenoud, “Adaptive TGF-β-dependent regulatory T cells control autoimmune diabetes and are a privileged target of anti-CD3 antibody treatment,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 15, pp. 6335–6340, 2007.
[259]
O. Akbari, G. J. Freeman, E. H. Meyer et al., “Antigen-specific regulatory T cells develop via the ICOS-ICOS-ligand pathway and inhibit allergen-induced airway hyperreactivity,” Nature Medicine, vol. 8, no. 9, pp. 1024–1032, 2002.
[260]
A. O'Garra, P. L. Vieira, P. Vieira, and A. E. Goldfeld, “IL-10-producing and naturally occurring CD4+Tregs: limiting collateral damage,” Journal of Clinical Investigation, vol. 114, no. 10, pp. 1372–1378, 2004.
[261]
E.-O. Glocker, D. Kotlarz, K. Boztug et al., “Inflammatory bowel disease and mutations affecting the interleukin-10 receptor,” The New England Journal of Medicine, vol. 361, no. 21, pp. 2033–2045, 2009.
[262]
S. Onizuka, I. Tawara, J. Shimizu, S. Sakaguchi, T. Fujita, and E. Nakayama, “Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor α) monoclonal antibody,” Cancer Research, vol. 59, no. 13, pp. 3128–3133, 1999.
[263]
G. Zhou, C. G. Drake, and H. I. Levitsky, “Amplification of tumor-specific regulatory T cells following therapeutic cancer vaccines,” Blood, vol. 107, no. 2, pp. 628–636, 2006.
[264]
A. M. Wolf, D. Wolf, M. Steurer, G. Gastl, E. Gunsilius, and B. Grubeck-Loebenstein, “Increase of regulatory T cells in the peripheral blood of cancer patients,” Clinical Cancer Research, vol. 9, no. 2, pp. 606–612, 2003.
[265]
H. Y. Wang, D. A. Lee, G. Peng et al., “Tumor-specific human CD4+ regulatory T cells and their ligands: implications for immunotherapy,” Immunity, vol. 20, no. 1, pp. 107–118, 2004.
[266]
M. Beyer and J. L. Schultze, “Regulatory T cells in cancer,” Blood, vol. 108, no. 3, pp. 804–811, 2006.
[267]
A. Gallimore and A. Godkin, “Regulatory T cells and tumour immunity—observations in mice and men,” Immunology, vol. 123, no. 2, pp. 157–163, 2008.
[268]
T. L. Whiteside, P. Schuler, and B. Schilling, “Induced and natural regulatory T cells in human cancer,” Expert Opinion on Biological Therapy, vol. 12, no. 10, pp. 1383–1397, 2012.
[269]
F. Ghiringhelli, P. E. Puig, S. Roux et al., “Tumor cells convert immature myeloid dendritic cells into TGF-β-secreting cells inducing CD4+CD25+ regulatory T cell proliferation,” Journal of Experimental Medicine, vol. 202, no. 7, pp. 919–929, 2005.
[270]
S. Sharma, S.-C. Yang, L. Zhu et al., “Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4+CD25+ T regulatory cell activities in lung cancer,” Cancer Research, vol. 65, no. 12, pp. 5211–5220, 2005.
[271]
E. Schlecker, A. Stojanovic, C. Eisen et al., “Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth,” Journal of Immunology, vol. 189, no. 12, pp. 5602–5611, 2012.
[272]
M. L. Belladonna, C. Orabona, U. Grohmann, and P. Puccetti, “TGF-β and kynurenines as the key to infectious tolerance,” Trends in Molecular Medicine, vol. 15, no. 2, pp. 41–49, 2009.
[273]
S. P. Cobbold, E. Adams, C. A. Farquhar et al., “Infectious tolerance via the consumption of essential amino acids and mTOR signaling,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 29, pp. 12055–12060, 2009.
[274]
F. Flores-Borja, E. C. Jury, C. Mauri, and M. R. Ehrenstein, “Defects in CTLA-4 are associated with abnormal regulatory T cell function in rheumatoid arthritis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 49, pp. 19396–19401, 2008.
[275]
A. V. Collins, D. W. Brodie, R. J. C. Gilbert et al., “The interaction properties of costimulatory molecules revisited,” Immunity, vol. 17, no. 2, pp. 201–210, 2002.
[276]
R. Nasser, M. Pelegrin, M. Plays, L. Gros, and M. Piechaczyk, “Control of regulatory T cells is necessary for vaccine-like effects of antiviral immunotherapy by monoclonal antibodies,” Blood, vol. 121, no. 7, pp. 1102–1111, 2013.
[277]
R. P. M. Sutmuller, L. M. Van Duivenvoorde, A. Van Elsas et al., “Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25+ regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses,” Journal of Experimental Medicine, vol. 194, no. 6, pp. 823–832, 2001.
[278]
J. Duraiswamy, K. M. Kaluza, G. J. Freeman, and G. Coukos, “Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores t-cell rejection function in tumors,” Cancer Research, vol. 73, no. 12, pp. 3591–3603, 2013.
[279]
D. R. Leach, M. F. Krummel, and J. P. Allison, “Enhancement of antitumor immunity by CTLA-4 blockade,” Science, vol. 271, no. 5256, pp. 1734–1736, 1996.
[280]
V. A. Trinh and W. J. Hwu, “Ipilimumab in the treatment of melanoma,” Expert Opinion on Biological Therapy, vol. 12, no. 6, pp. 773–782, 2012.
[281]
F. S. Hodi, D. A. Oble, J. Drappatz et al., “CTLA-4 blockade with ipilimumab induces significant clinical benefit in a female with melanoma metastases to the CNS,” Nature Clinical Practice Oncology, vol. 5, no. 9, pp. 557–561, 2008.
[282]
F. S. Hodi, S. J. O'Day, D. F. McDermott et al., “Improved survival with ipilimumab in patients with metastatic melanoma,” The New England Journal of Medicine, vol. 363, no. 8, pp. 711–723, 2010.
[283]
P. Tomasini, N. Khobta, L. Greillier, and F. Barlesi, “Ipilimumab: its potential in non-small cell lung cancer,” Therapeutic Advances in Medical Oncology, vol. 4, no. 2, pp. 43–50, 2012.
[284]
T. R. Simpson, F. Li, W. Montalvo-Ortiz et al., “Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma,” Journal of Experimental Medicine, vol. 210, no. 9, pp. 1695–1710, 2013.
[285]
R. E. Royal, C. Levy, K. Turner et al., “Phase 2 trial of single agent ipilimumab (Anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma,” Journal of Immunotherapy, vol. 33, no. 8, pp. 828–833, 2010.
[286]
S. A. Quezada, K. S. Peggs, M. A. Curran, and J. P. Allison, “CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells,” Journal of Clinical Investigation, vol. 116, no. 7, pp. 1935–1945, 2006.
[287]
J. H. Finke, B. Rini, J. Ireland et al., “Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients,” Clinical Cancer Research, vol. 14, no. 20, pp. 6674–6682, 2008.
[288]
H. Xin, C. Zhang, A. Herrmann, Y. Du, R. Figlin, and H. Yu, “Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells,” Cancer Research, vol. 69, no. 6, pp. 2506–2513, 2009.
[289]
C. Nishioka, T. Ikezoe, J. Yang et al., “Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene,” Leukemia Research, vol. 32, no. 6, pp. 865–872, 2008.
[290]
P. J. Roberts and C. J. Der, “Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer,” Oncogene, vol. 26, no. 22, pp. 3291–3310, 2007.
[291]
Y. Hailemichael, Z. Dai, N. Jaffarzad et al., “Persistent antigen at vaccination sites induces tumor-specific CD8+ T cell sequestration, dysfunction and deletion,” Nature Medicine, vol. 19, no. 4, pp. 465–472, 2013.
[292]
D. I. Gabrilovich and S. Nagaraj, “Myeloid-derived suppressor cells as regulators of the immune system,” Nature Reviews Immunology, vol. 9, no. 3, pp. 162–174, 2009.
[293]
T. Condamine and D. I. Gabrilovich, “Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function,” Trends in Immunology, vol. 32, no. 1, pp. 19–25, 2011.
[294]
C. Iclozan, S. Antonia, A. Chiappori, D. T. Chen, and D. Gabrilovich, “Therapeutic regulation of myeloid-derived suppressor cells and immune response to cancer vaccine in patients with extensive stage small cell lung cancer,” Cancer Immunology, Immunotherapy, vol. 62, no. 5, pp. 909–918, 2013.
[295]
G. A. Rabinovich, D. Gabrilovich, and E. M. Sotomayor, “Immunosuppressive strategies that are mediated by tumor cells,” Annual Review of Immunology, vol. 25, pp. 267–296, 2007.
[296]
M. B. Lutz, N. A. Kukutsch, M. Menges, S. Rossner, and G. Schuler, “Culture of bone marrow cells in GM-CSF plus high doses of lipopolysaccharide generates exclusively immature dendritic cells which induce alloantigen-specific CD4 T cell anergy in vitro,” European Journal of Immunology, vol. 30, no. 4, pp. 1048–1052, 2000.
[297]
P. Cheng, C. A. Corzo, N. Luetteke et al., “Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein,” Journal of Experimental Medicine, vol. 205, no. 10, pp. 2235–2249, 2008.
[298]
J.-I. Youn, S. Nagaraj, M. Collazo, and D. I. Gabrilovich, “Subsets of myeloid-derived suppressor cells in tumor-bearing mice,” Journal of Immunology, vol. 181, no. 8, pp. 5791–5802, 2008.
[299]
S. Solito, V. Bronte, and S. Mandruzzato, “Antigen specificity of immune suppression by myeloid-derived suppressor cells,” Journal of Leukocyte Biology, vol. 90, no. 1, pp. 31–36, 2011.
[300]
M. K. Srivastava, P. Sinha, V. K. Clements, P. Rodriguez, and S. Ostrand-Rosenberg, “Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine,” Cancer Research, vol. 70, no. 1, pp. 68–77, 2010.
[301]
V. Bronte, E. Apolloni, A. Cabrelle et al., “Identification of a CD11b+/Gr-1+/CD31+ myeloid progenitor capable of activating or suppressing CD8+ T cells,” Blood, vol. 96, no. 12, pp. 3838–3846, 2000.
[302]
P. Serafini, K. Meckel, M. Kelso et al., “Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function,” Journal of Experimental Medicine, vol. 203, no. 12, pp. 2691–2702, 2006.
[303]
J. K. Morales, M. Kmieciak, K. L. Knutson, H. D. Bear, and M. H. Manjili, “GM-CSF is one of the main breast tumor-derived soluble factors involved in the differentiation of CD11b-Gr1-bone marrow progenitor cells into myeloid-derived suppressor cells,” Breast Cancer Research and Treatment, vol. 123, no. 1, pp. 39–49, 2010.
[304]
L. Dolcetti, E. Peranzoni, S. Ugel et al., “Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF,” European Journal of Immunology, vol. 40, no. 1, pp. 22–35, 2010.
[305]
E. J. Small, D. M. Reese, B. Um, S. Whisenant, S. C. Dixon, and W. D. Figg, “Therapy of advanced prostate cancer with granulocyte macrophage colony-stimulating factor,” Clinical Cancer Research, vol. 5, no. 7, pp. 1738–1744, 1999.
[306]
M. J. Mastrangelo, H. C. Maguire Jr., L. C. Eisenlohr et al., “Intratumoral recombinant GM-CSF-encoding virus as gene therapy in patients with cutaneous melanoma,” Cancer Gene Therapy, vol. 6, no. 5, pp. 409–422, 1999.
[307]
G. Dranoff, “GM-CSF-secreting melanoma vaccines,” Oncogene, vol. 22, no. 20, pp. 3188–3192, 2003.
[308]
B. H. Kushner and N.-K. V. Cheung, “GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma,” Blood, vol. 73, no. 7, pp. 1936–1941, 1989.
[309]
P. R?ssner, J. Bubeník, V. Sobotá et al., “Granulocyte-macrophage colony-stimulating factor-producing tumour vaccines,” Folia Biologica, vol. 45, no. 5, pp. 173–177, 1999.
[310]
C. M. Gutschalk, A. K. Yanamandra, N. Linde, A. Meides, S. Depner, and M. M. Mueller, “GM-CSF enhances tumor invasion by elevated MMP-2, -9, and -26 expression,” Cancer Medicine, vol. 2, no. 2, pp. 117–129, 2012.
[311]
M. Martinez, N. Ono, M. Planutiene, K. Planutis, E. L. Nelson, and R. F. Holcombe, “Granulocyte-macrophage stimulating factor (GM-CSF) increases circulating dendritic cells but does not abrogate suppression of adaptive cellular immunity in patients with metastatic colorectal cancer receiving chemotherapy,” Cancer Cell International, vol. 12, article 2, 2012.
[312]
I. Marigo, E. Bosio, S. Solito et al., “Tumor-induced tolerance and immune suppression depend on the C/EBPβ transcription factor,” Immunity, vol. 32, no. 6, pp. 790–802, 2010.
[313]
M. G. Lechner, D. J. Liebertz, and A. L. Epstein., “Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells,” Journal of Immunology, vol. 185, no. 4, pp. 2273–2284, 2010.
[314]
S. L. Highfill, P. C. Rodriguez, Q. Zhou et al., “Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13,” Blood, vol. 116, no. 25, pp. 5738–5747, 2010.
[315]
J. Dulos, G. J. Carven, S. J. Van Boxtel et al., “PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer,” Journal of Immunotherapy, vol. 35, no. 2, pp. 169–178, 2012.
[316]
H. Navabi, B. Jasani, A. Reece et al., “A clinical grade poly I:C-analogue (Ampligen) promotes optimal DC maturation and Th1-type T cell responses of healthy donors and cancer patients in vitro,” Vaccine, vol. 27, no. 1, pp. 107–115, 2009.
[317]
X. Xiang, A. Poliakov, C. Liu et al., “Induction of myeloid-derived suppressor cells by tumor exosomes,” International Journal of Cancer, vol. 124, no. 11, pp. 2621–2633, 2009.
[318]
R. Valenti, V. Huber, P. Filipazzi et al., “Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-β-mediated suppressive activity on T lymphocytes,” Cancer Research, vol. 66, no. 18, pp. 9290–9298, 2006.
[319]
N. Obermajer, R. Muthuswamy, J. Lesnock, R. P. Edwards, and P. Kalinski, “Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells,” Blood, vol. 118, no. 20, pp. 5498–5505, 2011.
[320]
P. U. Emeagi, S. Maenhout, N. Dang, C. Heirman, K. Thielemans, and K. Breckpot, “Downregulation of Stat3 in melanoma: reprogramming the immune microenvironment as an anticancer therapeutic strategy,” Gene Therapy, 2013.
[321]
J. I. Youn, V. Kumar, M. Collazo et al., “Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer,” Nature Immunology, vol. 14, no. 3, pp. 211–220, 2013.
[322]
E. Peranzoni, S. Zilio, I. Marigo et al., “Myeloid-derived suppressor cell heterogeneity and subset definition,” Current Opinion in Immunology, vol. 22, no. 2, pp. 238–244, 2010.
[323]
E. Apolloni, V. Bronte, A. Mazzoni et al., “Immortalized myeloid suppressor cells trigger apoptosis in antigen-activated T lymphocytes,” Journal of Immunology, vol. 165, no. 12, pp. 6723–6730, 2000.