490
Views
4
CrossRef citations to date
0
Altmetric
Review

Methods for improving the immunogenicity and efficacy of cancer vaccines

, & ORCID Icon
Pages 765-784 | Received 28 Feb 2018, Accepted 04 Jun 2018, Published online: 17 Jun 2018

References

  • Coley WB. The treatment of inoperable sarcoma by bacterial toxins (the mixed toxins of the Streptococcus erysipelas and the Bacillus prodigiosus). Proc R Soc Med. 1910;3:1–48.
  • van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254:1643–1647.
  • Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011;480:480–489.
  • Tran E, Rosenberg SA. T-cell therapy against cancer mutations. Oncotarget. 2014;5:4579–4580.
  • Gubin MM, Artyomov MN, Mardis ER, et al. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125:3413–3421.
  • Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128.
  • Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.
  • Parmiani G, Russo V, Maccalli C, et al. Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother. 2014;10:3175–3178.
  • Seremet T, Brasseur F, Coulie PG. Tumor-specific antigens and immunologic adjuvants in cancer immunotherapy. Cancer J. 2011;17:325–330.
  • Rosenberg SA. Progress in the development of immunotherapy for the treatment of patients with cancer. J Intern Med. 2001;250:462–475.
  • Simpson AJ, Caballero OL, Jungbluth A, et al. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer. 2005;5:615–625.
  • Segal NH, Parsons DW, Peggs KS, et al. Epitope landscape in breast and colorectal cancer. Cancer Res. 2008;68:889–892.
  • Pilla L, Rivoltini L, Patuzzo R, et al. Multipeptide vaccination in cancer patients. Expert Opin Biol Ther. 2009;9:1043–1055.
  • Melero I, Gaudernack G, Gerritsen W, et al. Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol. 2014;11:509–524.
  • Parkhurst MR, Salgaller ML, Southwood S, et al. Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues. J Immunol. 1996;157:2539–2548.
  • Zaremba S, Barzaga E, Zhu M, et al. Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen. Cancer Res. 1997;57:4570–4577.
  • Valmori D, Fonteneau JF, Lizana CM, et al. Enhanced generation of specific tumor-reactive CTL in vitro by selected Melan-A/MART-1 immunodominant peptide analogues. J Immunol. 1998;160:1750–1758.
  • Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med. 1998;4:321–327.
  • Geynisman DM, Zha Y, Kunnavakkam R, et al. A randomized pilot phase I study of modified carcinoembryonic antigen (CEA) peptide (CAP1-6D)/montanide/GM-CSF-vaccine in patients with pancreatic adenocarcinoma. J Immunother Cancer. 2013;1:8.
  • Fauquembergue E, Toutirais O, Tougeron D, et al. HLA-A*0201-restricted CEA-derived peptide CAP1 is not a suitable target for T-cell-based immunotherapy. J Immunother. 2010;33:402–413.
  • Filipazzi P, Pilla L, Mariani L, et al. Limited induction of tumor cross-reactive T cells without a measurable clinical benefit in early melanoma patients vaccinated with human leukocyte antigen class I-modified peptides. Clin Cancer Res. 2012;18:6485–6496.
  • Buhrman JD, Slansky JE. Improving T cell responses to modified peptides in tumor vaccines. Immunol Res. 2013;55:34–47.
  • Schwartzentruber DJ, Lawson DH, Richards JM, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119–2127.
  • Chauvin JM, Larrieu P, Sarrabayrouse G, et al. HLA anchor optimization of the melan-A-HLA-A2 epitope within a long peptide is required for efficient cross-priming of human tumor-reactive T cells. J Immunol. 2012;188:2102–2110.
  • Trajanoski Z, Maccalli C, Mennonna D, et al. Somatically mutated tumor antigens in the quest for a more efficacious patient-oriented immunotherapy of cancer. Cancer Immunol Immunother. 2015;64:99–104.
  • Mennonna D, Maccalli C, Romano MC, et al. T cell neoepitope discovery in colorectal cancer by high throughput profiling of somatic mutations in expressed genes. Gut. 2017 Mar;66(3):454-463. doi: 10.1136/gutjnl-2015-309453. Epub 2015 Dec 17.
  • Robbins PF, Lu YC, El-Gamil M, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med. 2013;19:747–752.
  • Britten CM, Singh-Jasuja H, Flamion B, et al. The regulatory landscape for actively personalized cancer immunotherapies. Nat Biotechnol. 2013;31:880–882.
  • Rahma OE, Hamilton JM, Wojtowicz M, et al. The immunological and clinical effects of mutated ras peptide vaccine in combination with IL-2, GM-CSF, or both in patients with solid tumors. J Transl Med. 2014;12:55.
  • Sampson JH, Heimberger AB, Archer GE, et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28:4722–4729.
  • Vonderheide RH, Nathanson KL. Immunotherapy at large: the road to personalized cancer vaccines. Nat Med. 2013;19:1098–1100.
  • Toubaji A, Achtar M, Provenzano M, et al. Pilot study of mutant ras peptide-based vaccine as an adjuvant treatment in pancreatic and colorectal cancers. Cancer Immunol Immunother. 2008;57:1413–1420.
  • Efremova M, Finotello F, Rieder D, et al. Neoantigens generated by individual mutations and their role in cancer immunity and immunotherapy. Front Immunol. 2017;8:1679.
  • Yarchoan M, Johnson BA 3rd, Lutz ER, et al. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer. 2017;17:209–222.
  • Robbins PF. Tumor-infiltrating lymphocyte therapy and neoantigens. Cancer J. 2017;23:138–143.
  • van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol. 2013;31:e439–e442.
  • Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–2199.
  • Kvistborg P, Philips D, Kelderman S, et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med. 2014;6:254ra128.
  • Ulmer JB, Geall AJ. Recent innovations in mRNA vaccines. Curr Opin Immunol. 2016;41:18–22.
  • Rice J, Dossett ML, Ohlen C, et al. DNA fusion gene vaccination mobilizes effective anti-leukemic cytotoxic T lymphocytes from a tolerized repertoire. Eur J Immunol. 2008;38:2118–2130.
  • Calarota SA, Weiner DB. Enhancement of human immunodeficiency virus type 1-DNA vaccine potency through incorporation of T-helper 1 molecular adjuvants. Immunol Rev. 2004;199:84–99.
  • Dell’Agnola C, Biragyn A. Clinical utilization of chemokines to combat cancer: the double-edged sword. Expert Rev Vaccines. 2007;6:267–283.
  • Heath WR, Belz GT, Behrens GM, et al. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol Rev. 2004;199:9–26.
  • Radcliffe JN, Roddick JS, Friedmann PS, et al. Prime-boost with alternating DNA vaccines designed to engage different antigen presentation pathways generates high frequencies of peptide-specific CD8+ T cells. J Immunol. 2006;177:6626–6633.
  • Jin B, Sun T, Yu XH, et al. Immunomodulatory effects of dsRNA and its potential as vaccine adjuvant. J Biomed Biotechnol. 2010;2010:690438.
  • McNamara MA, Nair SK, Holl EK. RNA-based vaccines in cancer immunotherapy. J Immunol Res. 2015;2015:794528.
  • Li L, Saade F, Petrovsky N. The future of human DNA vaccines. J Biotechnol. 2012;162:171–182.
  • McNeel DG, Dunphy EJ, Davies JG, et al. Safety and immunological efficacy of a DNA vaccine encoding prostatic acid phosphatase in patients with stage D0 prostate cancer. J Clin Oncol. 2009;27:4047–4054.
  • Becker JT, Olson BM, Johnson LE, et al. DNA vaccine encoding prostatic acid phosphatase (PAP) elicits long-term T-cell responses in patients with recurrent prostate cancer. J Immunother. 2010;33:639–647.
  • Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–226.
  • Smola S, Trimble C, Stern PL. Human papillomavirus-driven immune deviation: challenge and novel opportunity for immunotherapy. Ther Adv Vaccines. 2017;5:69–82.
  • Trimble CL, Morrow MP, Kraynyak KA, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386:2078–2088.
  • Butterfield LH, Economou JS, Gamblin TC, et al. Alpha fetoprotein DNA prime and adenovirus boost immunization of two hepatocellular cancer patients. J Transl Med. 2014;12:86.
  • Butterfield LH, Comin-Anduix B, Vujanovic L, et al. Adenovirus MART-1-engineered autologous dendritic cell vaccine for metastatic melanoma. J Immunother. 2008;31:294–309.
  • Kirk CJ, Mule JJ. Gene-modified dendritic cells for use in tumor vaccines. Hum Gene Ther. 2000;11:797–806.
  • Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422.
  • Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther. 2007;15:651–659.
  • Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012;30:658–670.
  • Liu TC, Thorne SH, Kirn DH. Oncolytic adenoviruses for cancer gene therapy. Methods Mol Biol. 2008;433:243–258.
  • Breitbach CJ, Arulanandam R, De Silva N, et al. Oncolytic vaccinia virus disrupts tumor-associated vasculature in humans. Cancer Res. 2013;73:1265–1275.
  • Lichty BD, Breitbach CJ, Stojdl DF, et al. Going viral with cancer immunotherapy. Nat Rev Cancer. 2014;14:559–567.
  • Diaz RM, Galivo F, Kottke T, et al. Oncolytic immunovirotherapy for melanoma using vesicular stomatitis virus. Cancer Res. 2007;67:2840–2848.
  • Liu BL, Robinson M, Han ZQ, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10:292–303.
  • Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33:2780–2788.
  • Ribas A, Dummer R, Puzanov I, et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2017;170:1109–19 e10.
  • Garber K. China approves world’s first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst. 2006;98:298–300.
  • Steinman RM, Nussenzweig MC. Dendritic cells: features and functions. Immunol Rev. 1980;53:127–147.
  • Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252.
  • Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immunity. 2013;39:38–48.
  • Gross S, Erdmann M, Haendle I, et al. Twelve-year survival and immune correlates in dendritic cell-vaccinated melanoma patients. JCI Insight.2017Apr 20;2(8). pii: 91438. doi: 10.1172/jci.insight.91438. [Epub ahead of print].
  • Boudewijns S, Bol KF, Schreibelt G, et al. Adjuvant dendritic cell vaccination induces tumor-specific immune responses in the majority of stage III melanoma patients. Oncoimmunology. 2016;5:e1191732.
  • Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol. 2009;27:83–117.
  • Wolkers MC, Stoetter G, Vyth-Dreese FA, et al. Redundancy of direct priming and cross-priming in tumor-specific CD8+ T cell responses. J Immunol. 2001;167:3577–3584.
  • Thomas AM, Santarsiero LM, Lutz ER, et al. Mesothelin-specific CD8(+) T cell responses provide evidence of in vivo cross-priming by antigen-presenting cells in vaccinated pancreatic cancer patients. J Exp Med. 2004;200:297–306.
  • Nouri-Shirazi M, Banchereau J, Bell D, et al. Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J Immunol. 2000;165:3797–3803.
  • Huang AY, Golumbek P, Ahmadzadeh M, et al. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science. 1994;264:961–965.
  • Kovalcsik E, John J, Turner M, et al. Differential expression of melanoma-associated antigens and molecules involved in antigen processing and presentation in three cell lines established from a single patient. Melanoma Res. 2004;14:463–471.
  • Mendez R, Serrano A, Jager E, et al. Analysis of HLA class I expression in different metastases from two melanoma patients undergoing peptide immunotherapy. Tissue Antigens. 2001;57:508–519.
  • Souberbielle BE, Westby M, Ganz S, et al. Comparison of four strategies for tumour vaccination in the B16-F10 melanoma model. Gene Ther. 1998;5:1447–1454.
  • Knight BC, Souberbielle BE, Rizzardi GP, et al. Allogeneic murine melanoma cell vaccine: a model for the development of human allogeneic cancer vaccine. Melanoma Res. 1996;6:299–306.
  • Hrouda D, Todryk SM, Perry MJ, et al. Allogeneic whole-tumour cell vaccination in the rat model of prostate cancer. BJU Int. 2000;86:742–748.
  • Michael A, Ball G, Quatan N, et al. Delayed disease progression after allogeneic cell vaccination in hormone-resistant prostate cancer and correlation with immunologic variables. Clin Cancer Res. 2005;11:4469–4478.
  • Hsueh EC, Gupta RK, Qi K, et al. Correlation of specific immune responses with survival in melanoma patients with distant metastases receiving polyvalent melanoma cell vaccine. J Clin Oncol. 1998;16:2913–2920.
  • Hsueh EC, Gupta RK, Yee R, et al. Does endogenous immune response determine the outcome of surgical therapy for metastatic melanoma? Ann Surg Oncol. 2000;7:232–238.
  • Vilella R, Benitez D, Mila J, et al. Treatment of patients with progressive unresectable metastatic melanoma with a heterologous polyvalent melanoma whole cell vaccine. Int J Cancer. 2003;106:626–631.
  • Dols A, Smith JW 2nd, Meijer SL, et al. Vaccination of women with metastatic breast cancer, using a costimulatory gene (CD80)-modified, HLA-A2-matched, allogeneic, breast cancer cell line: clinical and immunological results. Hum Gene Ther. 2003;14:1117–1123.
  • Raez LE, Cassileth PA, Schlesselman JJ, et al. Allogeneic vaccination with a B7.1 HLA-A gene-modified adenocarcinoma cell line in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2004;22:2800–2807.
  • Arienti F, Sule-Suso J, Melani C, et al. Interleukin-2 gene-transduced human melanoma cells efficiently stimulate MHC-unrestricted and MHC-restricted autologous lymphocytes. Hum Gene Ther. 1994;5:1139–1150.
  • Belli F, Mascheroni L, Gallino G, et al. Active immunization of metastatic melanoma patients with IL-2 or IL-4 gene transfected, allogeneic melanoma cells. Adv Exp Med Biol. 1998;451:543–545.
  • Melani C, Sule-Suso J, Arienti F, et al. A human melanoma cell line transduced with an interleukin-4 gene by a retroviral vector releases biologically active IL-4 and maintains the original tumor antigenic phenotype. Hum Gene Ther. 1995;6:1427–1436.
  • Parmiani G, Pilla L, Maccalli C, et al. Autologous versus allogeneic cell-based vaccines? Cancer J. 2011;17:331–336.
  • Mach N, Dranoff G. Cytokine-secreting tumor cell vaccines. Curr Opin Immunol. 2000;12:571–575.
  • Thomas MC, Greten TF, Pardoll DM, et al. Enhanced tumor protection by granulocyte-macrophage colony-stimulating factor expression at the site of an allogeneic vaccine. Hum Gene Ther. 1998;9:835–843.
  • Chang EY, Chen CH, Ji H, et al. Antigen-specific cancer immunotherapy using a GM-CSF secreting allogeneic tumor cell-based vaccine. Int J Cancer. 2000;86:725–730.
  • Jaffee EM, Hruban RH, Biedrzycki B, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J Clin Oncol. 2001;19:145–156.
  • Simons JW, Sacks N. Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: the GVAX vaccine for prostate cancer. Urol Oncol. 2006;24:419–424.
  • Salcedo M, Bercovici N, Taylor R, et al. Vaccination of melanoma patients using dendritic cells loaded with an allogeneic tumor cell lysate. Cancer Immunol Immunother. 2006;55:819–829.
  • Sondak VK, Sosman JA. Results of clinical trials with an allogenic melanoma tumor cell lysate vaccine: melacine. Semin Cancer Biol. 2003;13:409–415.
  • Quezada SA, Peggs KS, Curran MA, et al. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116:1935–1945.
  • Parmiani G, Maccalli C, Maio M. Integrating immune checkpoint blockade with anti-neo/mutated antigens reactivity to increase the clinical outcome of immunotherapy. Vaccines (Basel). 2015;3:420–428.
  • Mendillo ML, Santagata S, Koeva M, et al. HSF1 drives a transcriptional program distinct from heat shock to support highly malignant human cancers. Cell. 2012;150:549–562.
  • Srivastava PK, DeLeo AB, Old LJ. Tumor rejection antigens of chemically induced sarcomas of inbred mice. Proc Natl Acad Sci U S A. 1986;83:3407–3411.
  • Udono H, Srivastava PK. Comparison of tumor-specific immunogenicities of stress-induced proteins gp96, hsp90, and hsp70. J Immunol. 1994;152:5398–5403.
  • Chandawarkar RY, Wagh MS, Srivastava PK. The dual nature of specific immunological activity of tumor-derived gp96 preparations. J Exp Med. 1999;189:1437–1442.
  • Mazzaferro V, Coppa J, Carrabba MG, et al. Vaccination with autologous tumor-derived heat-shock protein gp96 after liver resection for metastatic colorectal cancer. Clin Cancer Res. 2003;9:3235–3245.
  • Parmiani G, Castelli C, Pilla L, et al. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann Oncol. 2007;18:226–232.
  • Parmiani G, Testori A, Maio M, et al. Heat shock proteins and their use as anticancer vaccines. Clin Cancer Res. 2004;10:8142–8146.
  • Guzhova IV, Margulis BA. HSP70-based anti-cancer immunotherapy. Hum Vaccin Immunother. 2016;12:2529–2535.
  • Castelli C, Rivoltini L, Rini F, et al. Heat shock proteins: biological functions and clinical application as personalized vaccines for human cancer. Cancer Immunol Immunother. 2004;53:227–233.
  • Manjili MH, Wang XY, Park J, et al. Immunotherapy of cancer using heat shock proteins. Front Biosci. 2002;7:d43–d52.
  • Ampie L, Choy W, Lamano JB, et al. Heat shock protein vaccines against glioblastoma: from bench to bedside. J Neurooncol. 2015;123:441–448.
  • Pilla L, Squarcina P, Coppa J, et al. Natural killer and NK-like T-cell activation in colorectal carcinoma patients treated with autologous tumor-derived heat shock protein 96. Cancer Res. 2005;65:3942–3949.
  • Yuan J, Kashiwagi S, Reeves P, et al. A novel mycobacterial Hsp70-containing fusion protein targeting mesothelin augments antitumor immunity and prolongs survival in murine models of ovarian cancer and mesothelioma. J Hematol Oncol. 2014;7:15.
  • Bloch O, Crane CA, Fuks Y, et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro Oncol. 2014;16:274–279.
  • Bloch O, Lim M, Sughrue ME, et al. Autologous heat shock protein peptide vaccination for newly diagnosed glioblastoma: impact of peripheral PD-L1 expression on response to therapy. Clin Cancer Res. 2017;23:3575–3584.
  • Poskitt DC, Jean-Francois MJ, Turnbull S, et al. Internal image (Ab2 beta) anti-idiotype vaccines. Theoretical and practical aspects. Vaccine. 1991;9:792–796.
  • Pasquali JL, Knapp AM, Farradji A, et al. Mapping of four light chain-associated idiotopes of a human monoclonal rheumatoid factor. J Immunol. 1987;139:818–823.
  • Parhami-Seren B, Wysocki LJ, Margolies MN, et al. H chain somatic mutations shared by anti-p-azophenylarsonate antibodies confer enhanced affinity and ablate the cross-reactive idiotype. J Immunol. 1990;145:2340–2346.
  • Singhal A, Hakomori S. Molecular changes in carbohydrate antigens associated with cancer. Bioessays. 1990;12:223–230.
  • Hakomori S, Zhang Y. Glycosphingolipid antigens and cancer therapy. Chem Biol. 1997;4:97–104.
  • Kobata A, Amano J. Altered glycosylation of proteins produced by malignant cells, and application for the diagnosis and immunotherapy of tumours. Immunol Cell Biol. 2005;83:429–439.
  • Guo Z, Wang Q. Recent development in carbohydrate-based cancer vaccines. Curr Opin Chem Biol. 2009;13:608–617.
  • Cazet A, Julien S, Bobowski M, et al. Tumour-associated carbohydrate antigens in breast cancer. Breast Cancer Res. 2010;12:204.
  • Fernandez LE, Gabri MR, Guthmann MD, et al. NGcGM3 ganglioside: a privileged target for cancer vaccines. Clin Dev Immunol. 2010;2010:814397.
  • Foon KA, Bhattacharya-Chatterjee M. Idiotype vaccines in the clinic. Nat Med. 1998;4:870.
  • Samonigg H, Wilders-Truschnig M, Kuss I, et al. A double-blind randomized-phase II trial comparing immunization with antiidiotype goat antibody vaccine SCV 106 versus unspecific goat antibodies in patients with metastatic colorectal cancer. J Immunother. 1999;22:481–488.
  • Wagner U, Kohler S, Reinartz S, et al. Immunological consolidation of ovarian carcinoma recurrences with monoclonal anti-idiotype antibody ACA125: immune responses and survival in palliative treatment. See the biology behind: K. A. Foon and M. Bhattacharya-Chatterjee, are solid tumor anti-idiotype vaccines ready for prime time?. Clin Cancer Res. 2001;7:1112–1115, Clin Cancer Res 2001;7:1154-62
  • Mittelman A, Chen ZJ, Liu CC, et al. Kinetics of the immune response and regression of metastatic lesions following development of humoral anti-high molecular weight-melanoma associated antigen immunity in three patients with advanced malignant melanoma immunized with mouse antiidiotypic monoclonal antibody MK2-23. Cancer Res. 1994;54:415–421.
  • Mittelman A, Wang X, Matsumoto K, et al. Antiantiidiotypic response and clinical course of the disease in patients with malignant melanoma immunized with mouse antiidiotypic monoclonal antibody MK2-23. Hybridoma. 1995;14:175–181.
  • Wang X, Ko EC, Peng L, et al. Human high molecular weight melanoma-associated antigen mimicry by mouse anti-idiotypic monoclonal antibody MK2-23: enhancement of immunogenicity of anti-idiotypic monoclonal antibody MK2-23 by fusion with interleukin 2. Cancer Res. 2005;65:6976–6983.
  • Murray JL, Gillogly M, Kawano K, et al. Fine specificity of high molecular weight-melanoma-associated antigen-specific cytotoxic T lymphocytes elicited by anti-idiotypic monoclonal antibodies in patients with melanoma. Cancer Res. 2004;64:5481–5488.
  • Ladjemi MZ. Anti-idiotypic antibodies as cancer vaccines: achievements and future improvements. Front Oncol. 2012;2:158.
  • Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010;33:492–503.
  • Pradere JP, Dapito DH, Schwabe RF. The Yin and Yang of Toll-like receptors in cancer. Oncogene. 2014;33:3485–3495.
  • Del Vecchio M, Bajetta E, Canova S, et al. Interleukin-12: biological properties and clinical application. Clin Cancer Res. 2007;13:4677–4685.
  • Teng MW, Westwood JA, Darcy PK, et al. Combined natural killer T-cell based immunotherapy eradicates established tumors in mice. Cancer Res. 2007;67:7495–7504.
  • Marciani DJ. Vaccine adjuvants: role and mechanisms of action in vaccine immunogenicity. Drug Discov Today. 2003;8:934–943.
  • Hailemichael Y, Dai Z, Jaffarzad N, et al. Persistent antigen at vaccination sites induces tumor-specific CD8(+) T cell sequestration, dysfunction and deletion. Nat Med. 2013;19:465–472.
  • Bald T, Landsberg J, Lopez-Ramos D, et al. Immune cell-poor melanomas benefit from PD-1 blockade after targeted type I IFN activation. Cancer Discov. 2014;4:674–687.
  • Dubensky TW Jr., Kanne DB, Leong ML. Rationale, progress and development of vaccines utilizing STING-activating cyclic dinucleotide adjuvants. Ther Adv Vaccines. 2013;1:131–143.
  • Barber GN. Cytoplasmic DNA innate immune pathways. Immunol Rev. 2011;243:99–108.
  • Fu J, Kanne DB, Leong M, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7:283ra52.
  • Barrios K, Celis E. TriVax-HPV: an improved peptide-based therapeutic vaccination strategy against human papillomavirus-induced cancers. Cancer Immunol Immunother. 2012;61:1307–1317.
  • Kaufman HL, Ruby CE, Hughes T, et al. Current status of granulocyte-macrophage colony-stimulating factor in the immunotherapy of melanoma. J Immunother Cancer. 2014;2:11.
  • Lee P, Wang F, Kuniyoshi J, et al. Effects of interleukin-12 on the immune response to a multipeptide vaccine for resected metastatic melanoma. J Clin Oncol. 2001;19:3836–3847.
  • Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465.
  • Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828–833.
  • Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454.
  • Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571.
  • Teng MW, Ngiow SF, Ribas A, et al. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res. 2015;75:2139–2145.
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264.
  • Fridman WH, Galon J, Dieu-Nosjean MC, et al. Immune infiltration in human cancer: prognostic significance and disease control. Curr Top Microbiol Immunol. 2011;344:1–24.
  • Fridman WH, Dieu-Nosjean MC, Pages F, et al. The immune microenvironment of human tumors: general significance and clinical impact. Cancer Microenviron. 2013;6:117–122.
  • Bindea G, Mlecnik B, Angell HK, et al. The immune landscape of human tumors: implications for cancer immunotherapy. Oncoimmunology. 2014;3:e27456.
  • Donnem T, Kilvaer TK, Andersen S, et al. Strategies for clinical implementation of TNM-Immunoscore in resected nonsmall-cell lung cancer. Ann Oncol. 2016;27:225–232.
  • Duraiswamy J, Kaluza KM, Freeman GJ, et al. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013;73:3591–3603.
  • Soares KC, Rucki AA, Wu AA, et al. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J Immunother. 2015;38:1–11.
  • Antonios JP, Soto H, Everson RG, et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight. 2016 Jul 7;1(10). pii: e87059.
  • Yuan J, Ginsberg B, Page D, et al. CTLA-4 blockade increases antigen-specific CD8(+) T cells in prevaccinated patients with melanoma: three cases. Cancer Immunol Immunother. 2011;60:1137–1146.
  • Smyth MJ, Ngiow SF, Ribas A, et al. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol. 2016;13:143–158.
  • Melero I, Berman DM, Aznar MA, et al. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. 2015;15:457–472.
  • Bjoern J, Iversen TZ, Nitschke NJ, et al. Safety, immune and clinical responses in metastatic melanoma patients vaccinated with a long peptide derived from indoleamine 2,3-dioxygenase in combination with ipilimumab. Cytotherapy. 2016;18:1043–1055.
  • Ly LV, Sluijter M, Versluis M, et al. Peptide vaccination after T-cell transfer causes massive clonal expansion, tumor eradication, and manageable cytokine storm. Cancer Res. 2010;70:8339–8346.
  • Kohlmeyer J, Cron M, Landsberg J, et al. Complete regression of advanced primary and metastatic mouse melanomas following combination chemoimmunotherapy. Cancer Res. 2009;69:6265–6274.
  • Rapoport AP, Aqui NA, Stadtmauer EA, et al. Combination immunotherapy after ASCT for multiple myeloma using MAGE-A3/Poly-ICLC immunizations followed by adoptive transfer of vaccine-primed and costimulated autologous T cells. Clin Cancer Res. 2014;20:1355–1365.
  • Ruffell B, Coussens LM. Macrophages and therapeutic resistance in cancer. Cancer Cell. 2015;27:462–472.
  • Young A, Mittal D, Stagg J, et al. Targeting cancer-derived adenosine: new therapeutic approaches. Cancer Discov. 2014;4:879–888.
  • Munn DH. Blocking IDO activity to enhance anti-tumor immunity. Front Biosci (Elite Ed). 2012;4:734–745.
  • Parmiani G, Pilla L, Corti A, et al. A pilot phase I study combining peptide-based vaccination and NGR-hTNF vessel targeting therapy in metastatic melanoma. Oncoimmunology. 2014;3:e963406.
  • Galluzzi L, Vitale I, Abrams JM, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012;19:107–120.
  • Kroemer G, Galluzzi L, Kepp O, et al. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72.
  • Pol J, Vacchelli E, Aranda F, et al. Trial Watch: immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology. 2015;4:e1008866.
  • Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195.
  • Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425:516–521.
  • Bianchi ME, Agresti A. HMG proteins: dynamic players in gene regulation and differentiation. Curr Opin Genet Dev. 2005;15:496–506.
  • Zitvogel L, Kepp O, Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol. 2011;8:151–160.
  • Denkert C, Loibl S, Noske A, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28:105–113.
  • Kroemer G, Galluzzi L. Combinatorial immunotherapy with checkpoint blockers solves the problem of metastatic melanoma-an exclamation sign with a question mark. Oncoimmunology. 2015;4:e1058037.
  • Ghiringhelli F, Larmonier N, Schmitt E, et al. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol. 2004;34:336–344.
  • Rettig L, Seidenberg S, Parvanova I, et al. Gemcitabine depletes regulatory T-cells in human and mice and enhances triggering of vaccine-specific cytotoxic T-cells. Int J Cancer. 2011;129:832–838.
  • Emens LA, Tuohy VK, Stanton SE. Immunotherapy for breast cancer: is it feasible? Immunotherapy. 2015;7:1135–1143.
  • Antonarakis ES, Carducci MA. Combining low-dose cyclophosphamide with GM-CSF-secreting prostate cancer immunotherapy enhances antitumor immune effects. Expert Opin Investig Drugs. 2010;19:311–314.
  • Butts C, Socinski MA, Mitchell PL, et al. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15:59–68.
  • Springett GM. Novel pancreatic cancer vaccines could unleash the army within. Cancer Control. 2014;21:242–246.
  • Prise KM, O’Sullivan JM. Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer. 2009;9:351–360.
  • Rees GJ. Abscopal regression in lymphoma: a mechanism in common with total body irradiation? Clin Radiol. 1981;32:475–480.
  • Horiuhi T, Nomura J, Okuda M, et al. [Abscopal effect of small intestinal NK/T-cell lymphoma]. Rinsho Ketsueki. 2003;44:940–945.
  • Stamell EF, Wolchok JD, Gnjatic S, et al. The abscopal effect associated with a systemic anti-melanoma immune response. Int J Radiat Oncol Biol Phys. 2013;85:293–295.
  • Vacchelli E, Vitale I, Tartour E, et al. Trial watch: anticancer radioimmunotherapy. Oncoimmunology. 2013;2:e25595.
  • Dovedi SJ, Melis MH, Wilkinson RW, et al. Systemic delivery of a TLR7 agonist in combination with radiation primes durable antitumor immune responses in mouse models of lymphoma. Blood. 2013;121:251–259.
  • Kim YH, Gratzinger D, Harrison C, et al. In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: a phase 1/2 study. Blood. 2012;119:355–363.
  • Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447:425–432.
  • Stallcup MR. Role of protein methylation in chromatin remodeling and transcriptional regulation. Oncogene. 2001;20:3014–3020.
  • Marazzi I, Greenbaum BD, Low DHP, et al. Chromatin dependencies in cancer and inflammation. Nat Rev Mol Cell Biol. 2018 Apr;19(4):245-261. doi: 10.1038/nrm.2017.113. Epub2017Nov 29. Review.
  • Toh TB, Lim JJ, Chow EK. Epigenetics in cancer stem cells. Mol Cancer. 2017;16:29.
  • Chiappinelli KB, Zahnow CA, Ahuja N, et al. Combining epigenetic and immunotherapy to combat cancer. Cancer Res. 2016;76:1683–1689.
  • Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic approach. Ann Oncol. 2017;28:1776–1787.
  • Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol. 2014;6:a018713.
  • Aspeslagh S, Morel D, Soria JC, et al. Epigenetic modifiers as new immunomodulatory therapies in solid tumours. Ann Oncol. 2018; 1;29(4):812-824. doi: 10.1093/annonc/mdy050.
  • Suarez-Alvarez B, Baragano Raneros A, Ortega F, et al. Epigenetic modulation of the immune function: a potential target for tolerance. Epigenetics. 2013;8:694–702.
  • Christiansen AJ, West A, Banks KM, et al. Eradication of solid tumors using histone deacetylase inhibitors combined with immune-stimulating antibodies. Proc Natl Acad Sci U S A. 2011;108:4141–4146.
  • West AC, Christiansen AJ, Smyth MJ, et al. The combination of histone deacetylase inhibitors with immune-stimulating antibodies has potent anti-cancer effects. Oncoimmunology. 2012;1:377–379.
  • Gandhi L, Ou SI, Shaw AT, et al. Efficacy of alectinib in central nervous system metastases in crizotinib-resistant ALK-positive non-small-cell lung cancer: comparison of RECIST 1.1 and RANO-HGG criteria. Eur J Cancer. 2017;82:27–33.
  • Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967–970.
  • Paulos CM, Kaiser A, Wrzesinski C, et al. Toll-like receptors in tumor immunotherapy. Clin Cancer Res. 2007;13:5280–5289.
  • Berman D, Parker SM, Siegel J, et al. Blockade of cytotoxic T-lymphocyte antigen-4 by ipilimumab results in dysregulation of gastrointestinal immunity in patients with advanced melanoma. Cancer Immun. 2010;10:11.
  • Vetizou M, Pitt JM, Daillere R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–1084.
  • Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–1089.
  • Gammon JM, Dold NM, Jewell CM. Improving the clinical impact of biomaterials in cancer immunotherapy. Oncotarget. 2016;7:15421–15443.
  • Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle vaccine delivery systems. Pharm Res. 2014;31:2563–2582.
  • Sunshine JC, Perica K, Schneck JP, et al. Particle shape dependence of CD8+ T cell activation by artificial antigen presenting cells. Biomaterials. 2014;35:269–277.
  • Hardy CL, Lemasurier JS, Mohamud R, et al. Differential uptake of nanoparticles and microparticles by pulmonary APC subsets induces discrete immunological imprints. J Immunol. 2013;191:5278–5290.
  • Kumar S, Anselmo AC, Banerjee A, et al. Shape and size-dependent immune response to antigen-carrying nanoparticles. J Control Release. 2015;220:141–148.
  • Moyano DF, Goldsmith M, Solfiell DJ, et al. Nanoparticle hydrophobicity dictates immune response. J Am Chem Soc. 2012;134:3965–3967.
  • Shahbazi MA, Fernandez TD, Makila EM, et al. Surface chemistry dependent immunostimulative potential of porous silicon nanoplatforms. Biomaterials. 2014;35:9224–9235.
  • Andorko JI, Hess KL, Pineault KG, et al. Intrinsic immunogenicity of rapidly-degradable polymers evolves during degradation. Acta Biomater. 2016;32:24–34.
  • Wen CY, Tang M, Hu J, et al. Determination of the absolute number concentration of nanoparticles and the active affinity sites on their surfaces. Anal Chem. 2016;88:10134–10142.
  • Ali OA, Huebsch N, Cao L, et al. Infection-mimicking materials to program dendritic cells in situ. Nat Mater. 2009;8:151–158.
  • Bencherif SA, Warren Sands R, Ali OA, et al. Injectable cryogel-based whole-cell cancer vaccines. Nat Commun. 2015;6:7556.
  • Luo M, Wang H, Wang Z, et al. A STING-activating nanovaccine for cancer immunotherapy. Nat Nanotechnol. 2017;12:648–654.
  • Kuai R, Ochyl LJ, Bahjat KS, et al. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat Mater. 2017;16:489–496.
  • Kosmides AK, Meyer RA, Hickey JW, et al. Biomimetic biodegradable artificial antigen presenting cells synergize with PD-1 blockade to treat melanoma. Biomaterials. 2017;118:16–26.
  • Grabbe S, Haas H, Diken M, et al. Translating nanoparticulate-personalized cancer vaccines into clinical applications: case study with RNA-lipoplexes for the treatment of melanoma. Nanomedicine (Lond). 2016;11:2723–2734.
  • Kranz LM, Diken M, Haas H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534:396–401.
  • Chen Q, Xu L, Liang C, et al. Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy. Nat Commun. 2016;7:13193.
  • Stephan SB, Taber AM, Jileaeva I, et al. Biopolymer implants enhance the efficacy of adoptive T-cell therapy. Nat Biotechnol. 2015;33:97–101.
  • Itano AA, McSorley SJ, Reinhardt RL, et al. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity. 2003;19:47–57.
  • Johansen P, Haffner AC, Koch F, et al. Direct intralymphatic injection of peptide vaccines enhances immunogenicity. Eur J Immunol. 2005;35:568–574.
  • Maloy KJ, Erdmann I, Basch V, et al. Intralymphatic immunization enhances DNA vaccination. Proc Natl Acad Sci U S A. 2001;98:3299–3303.
  • Liu H, Moynihan KD, Zheng Y, et al. Structure-based programming of lymph-node targeting in molecular vaccines. Nature. 2014;507:519–522.
  • Smirnov D, Schmidt JJ, Capecchi JT, et al. Vaccine adjuvant activity of 3M-052: an imidazoquinoline designed for local activity without systemic cytokine induction. Vaccine. 2011;29:5434–5442.
  • Marabelle A, Kohrt H, Levy R. Intratumoral anti-CTLA-4 therapy: enhancing efficacy while avoiding toxicity. Clin Cancer Res. 2013;19:5261–5263.
  • Schulze HJ, Cribier B, Requena L, et al. Imiquimod 5% cream for the treatment of superficial basal cell carcinoma: results from a randomized vehicle-controlled phase III study in Europe. Br J Dermatol. 2005;152:939–947.
  • Zamarin D, Holmgaard RB, Subudhi SK, et al. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci Transl Med. 2014;6:226ra32.
  • Egilmez NK, Jong YS, Sabel MS, et al. In situ tumor vaccination with interleukin-12-encapsulated biodegradable microspheres: induction of tumor regression and potent antitumor immunity. Cancer Res. 2000;60:3832–3837.
  • Kumamoto T, Huang EK, Paek HJ, et al. Induction of tumor-specific protective immunity by in situ Langerhans cell vaccine. Nat Biotechnol. 2002;20:64–69.
  • Maccalli C, De Maria R. Cancer stem cells: perspectives for therapeutic targeting. Cancer Immunol Immunother. 2015;64:91–97.
  • Maccalli C, Parmiani G, Ferrone S. Immunomodulating and immunoresistance properties of cancer-initiating cells: implications for the clinical success of immunotherapy. Immunol Invest. 2017;46:221–238.
  • Codony-Servat J, Rosell R. Cancer stem cells and immunoresistance: clinical implications and solutions. Transl Lung Cancer Res. 2015;4:689–703.
  • Ali HR, Glont SE, Blows FM, et al. PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes. Ann Oncol. 2015;26:1488–1493.
  • Takeda N, Jain R, LeBoeuf MR, et al. Interconversion between intestinal stem cell populations in distinct niches. Science. 2011;334:1420–1424.
  • Finocchiaro G, Pellegatta S. Immunotherapy with dendritic cells loaded with glioblastoma stem cells: from preclinical to clinical studies. Cancer Immunol Immunother. 2016;65:101–109.
  • Kano M, Tsukahara T, Emori M, et al. Autologous CTL response against cancer stem-like cells/cancer-initiating cells of bone malignant fibrous histiocytoma. Cancer Sci. 2011;102:1443–1447.
  • Ning N, Pan Q, Zheng F, et al. Cancer stem cell vaccination confers significant antitumor immunity. Cancer Res. 2012;72:1853–1864.
  • Vik-Mo EO, Nyakas M, Mikkelsen BV, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62:1499–1509.
  • Todaro M, Gaggianesi M, Catalano V, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14:342–356.
  • Rolih V, Barutello G, Iussich S, et al. CSPG4: a prototype oncoantigen for translational immunotherapy studies. J Transl Med. 2017;15:151.
  • Maciag PC, Seavey MM, Pan ZK, et al. Cancer immunotherapy targeting the high molecular weight melanoma-associated antigen protein results in a broad antitumor response and reduction of pericytes in the tumor vasculature. Cancer Res. 2008;68:8066–8075.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.