6,630
Views
103
CrossRef citations to date
0
Altmetric
Review

Trial watch: Peptide-based vaccines in anticancer therapy

ORCID Icon, ORCID Icon, , , , , , ORCID Icon & show all
Article: e1511506 | Received 10 Aug 2018, Published online: 06 Sep 2018

References

  • Hoos A. Development of immuno-oncology drugs - from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov. 2016;15:235–247. doi:10.1038/nrd.2015.35.
  • June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–1365. doi:10.1126/science.aar6711.
  • Palucka AK, Coussens LM. The basis of oncoimmunology. Cell. 2016;164:1233–1247. doi:10.1016/j.cell.2016.01.049.
  • Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv Immunol. 2006;90:1–50. doi:10.1016/S0065-2776(06)90001-7.
  • Berraondo P, Labiano S, Minute L, Etxeberria I, Vasquez M, Sanchez-Arraez A, Teijeira A, Melero I. Cellular immunotherapies for cancer. Oncoimmunology. 2017;6:e1306619. doi:10.1080/2162402X.2017.1306619.
  • Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S, Formenti SC. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer. 2018;18:313–322. doi:10.1038/nrc.2018.6.
  • Thomas S, Prendergast GC. Cancer vaccines: a brief overview. Methods Mol Biol. 2016;1403:755–761. doi:10.1007/978-1-4939-3387-7_43.
  • Galluzzi L, Vacchelli E, Bravo-San Pedro JM, Buque A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P, et al. Classification of current anticancer immunotherapies. Oncotarget. 2014;5:12472–12508. doi:10.18632/oncotarget.2998.
  • Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–330. doi:10.1038/nature21349.
  • Hanoteau A, Moser M. Chemotherapy and immunotherapy: A close interplay to fight cancer? Oncoimmunology. 2016;5:e1190061. doi:10.1080/2162402X.2016.1190061.
  • Yamazaki T, Galluzzi L. Blinatumomab bridges the gap between leukemia and immunity. Oncoimmunology. 2017;6:e1358335. doi:10.1080/2162402X.2017.1358335.
  • Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14:561–584. doi:10.1038/nrd4591.
  • Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, et al. Trial watch: immune checkpoint blockers for cancer therapy. Oncoimmunology. 2017;6:e1373237. doi:10.1080/2162402X.2017.1373237.
  • Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. doi:10.1126/science.aaa8172.
  • Derosa L, Routy B, Kroemer G, Zitvogel L. The intestinal microbiota determines the clinical efficacy of immune checkpoint blockers targeting PD-1/PD-L1. Oncoimmunology. 2018;7:e1434468. doi:10.1080/2162402X.2018.1434468.
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. doi:10.1038/nrc3239.
  • Vanella V, Festino L, Strudel M, Simeone E, Grimaldi AM, Ascierto PA. PD-L1 inhibitors in the pipeline: promise and progress. Oncoimmunology. 2017;7:e1365209. doi:10.1080/2162402X.2017.1365209.
  • Simon S, Labarriere N. PD-1 expression on tumor-specific T cells: friend or foe for immunotherapy? Oncoimmunology. 2017;7:e1364828. doi:10.1080/2162402X.2017.1364828.
  • Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, Hodi FS, Martin-Algarra S, Mandal R, Sharfman WH, et al. Tumor and microenvironment evolution during immunotherapy with Nivolumab. Cell. 2017;171:934–949 e915. doi:10.1016/j.cell.2017.09.028.
  • Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111. doi:10.1038/nri.2016.107.
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi:10.1146/annurev-immunol-032712-100008.
  • Garg AD, More S, Rufo N, Mece O, Sassano ML, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology. 2017;6:e1386829. doi:10.1080/2162402X.2017.1386829.
  • Yang M, Li C, Zhu S, Cao L, Kroemer G, Zeh H, Tang D, Kang R. TFAM is a novel mediator of immunogenic cancer cell death. Oncoimmunology. 2018;7:e1431086. doi:10.1080/2162402X.2018.1431086.
  • Montico B, Nigro A, Casolaro V, Dal Col J. Immunogenic apoptosis as a novel tool for anticancer vaccine development. Int J Mol Sci. 2018;19. doi:10.3390/ijms19020594.
  • Bezu L, Sauvat A, Humeau J, Leduc M, Kepp O, Kroemer G. eIF2alpha phosphorylation: A hallmark of immunogenic cell death. Oncoimmunology. 2018;7:e1431089. doi:10.1080/2162402X.2018.1431089.
  • Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Nat Rev Immunol. 2015;15:405–414. doi:10.1038/nri3845.
  • Eggermont AM, Spatz A, Robert C. Cutaneous melanoma. Lancet. 2014;383:816–827. doi:10.1016/S0140-6736(13)60802-8.
  • Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology. 2017;6:e1371896. doi:10.1080/2162402X.2017.1371896.
  • Sanmamed MF, Pastor F, Rodriguez A, Perez-Gracia JL, Rodriguez-Ruiz ME, Jure-Kunkel M, Melero I. Agonists of co-stimulation in cancer immunotherapy directed against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol. 2015;42:640–655. doi:10.1053/j.seminoncol.2015.05.014.
  • Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15:47–62. doi:10.1038/nrclinonc.2017.148.
  • Brudno JN, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for lymphoma. Nat Rev Clin Oncol. 2018;15:31–46. doi:10.1038/nrclinonc.2017.128.
  • Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68. doi:10.1126/science.aaa4967.
  • Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer. 2016;16:566–581. doi:10.1038/nrc.2016.97.
  • Zhang Q, Zhang Z, Peng M, Fu S, Xue Z, Zhang R. CAR-T cell therapy in gastrointestinal tumors and hepatic carcinoma: from bench to bedside. Oncoimmunology. 2016;5:e1251539. doi:10.1080/2162402X.2016.1251539.
  • Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13:273–290. doi:10.1038/nrclinonc.2016.25.
  • Atanackovic D, Steinbach M, Radhakrishnan SV, Luetkens T. Immunotherapies targeting CD38 in multiple myeloma. Oncoimmunology. 2016;5:e1217374. doi:10.1080/2162402X.2016.1217374.
  • Adams JL, Smothers J, Srinivasan R, Hoos A. Big opportunities for small molecules in immuno-oncology. Nat Rev Drug Discov. 2015;14:603–622. doi:10.1038/nrd4596.
  • Buque A, Bloy N, Aranda F, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Spisek R, Tartour E, et al. Trial watch-small molecules targeting the immunological tumor microenvironment for cancer therapy. Oncoimmunology. 2016;5:e1149674. doi:10.1080/2162402X.2016.1149674.
  • Iyer VV. Small molecules for immunomodulation in cancer: a review. Anticancer Agents Med Chem. 2015;15:433–452.
  • Teulings HE, Tjin EPM, Willemsen KJ, Van Der Kleij S, Ter Meulen S, Kemp EH, Krebbers G, Van Noesel CJM, Franken C, Drijfhout JW, et al. Anti-Melanoma immunity and local regression of cutaneous metastases in melanoma patients treated with monobenzone and imiquimod; a phase 2 a trial. Oncoimmunology. 2018;7:e1419113. doi:10.1080/2162402X.2017.1419113.
  • Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–2199. doi:10.1056/NEJMoa1406498.
  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. doi:10.1056/NEJMoa1200690.
  • Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372:2018–2028. doi:10.1056/NEJMoa1501824.
  • Rosenberg JE, Hoffman-Censits J, Powles T, Van Der Heijden MS, Balar AV, Necchi A, Dawson N, O’Donnell PH, Balmanoukian A, Loriot Y, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387:1909–1920. doi:10.1016/S0140-6736(16)00561-4.
  • Fuca G, De Braud F, Di Nicola M. Immunotherapy-based combinations: an update. Curr Opin Oncol. 2018. doi:10.1097/CCO.0000000000000466.
  • Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439–448. doi:10.1056/NEJMoa1709866.
  • Batlevi CL, Matsuki E, Brentjens RJ, Younes A. Novel immunotherapies in lymphoid malignancies. Nat Rev Clin Oncol. 2016;13:25–40. doi:10.1038/nrclinonc.2015.187.
  • Suryadevara CM, Desai R, Abel ML, Riccione KA, Batich KA, Shen SH, Chongsathidkiet P, Gedeon PC, Elsamadicy AA, Snyder DJ, et al. Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma. Oncoimmunology. 2018;7:e1434464. doi:10.1080/2162402X.2018.1434464.
  • Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214. doi:10.1016/j.cell.2015.03.030.
  • Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, Chung CH, Hernandez-Aya L, Lim AM, Chang ALS, et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med. 2018. doi:10.1056/NEJMoa1805131.
  • Vacchelli E, Bloy N, Aranda F, Buque A, Cremer I, Demaria S, Eggermont A, Formenti SC, Fridman WH, Fucikova J, et al. Trial watch: immunotherapy plus radiation therapy for oncological indications. Oncoimmunology. 2016;5:e1214790. doi:10.1080/2162402X.2016.1214790.
  • Butts C, Socinski MA, Mitchell PL, Thatcher N, Havel L, Krzakowski M, Nawrocki S, Ciuleanu TE, Bosquee L, Trigo JM, 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. doi:10.1016/S1470-2045(13)70510-2.
  • Ruckert M, Deloch L, Fietkau R, Frey B, Hecht M, Gaipl US. Immune modulatory effects of radiotherapy as basis for well-reasoned radioimmunotherapies. Strahlenther Onkol. 2018;194:509–519. doi:10.1007/s00066-018-1287-1.
  • Fumet JD, Isambert N, Hervieu A, Zanetta S, Guion JF, Hennequin A, Rederstorff E, Bertaut A, Ghiringhelli F. Phase Ib/II trial evaluating the safety, tolerability and immunological activity of durvalumab (MEDI4736) (anti-PD-L1) plus tremelimumab (anti-CTLA-4) combined with FOLFOX in patients with metastatic colorectal cancer. ESMO Open. 2018;3:e000375. doi:10.1136/esmoopen-2018-000375.
  • Coulie PG, Van Den Eynde BJ, Van Der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135–146. doi:10.1038/nrc3670.
  • Ilyas S, Yang JC. Landscape of tumor antigens in T cell immunotherapy. J Immunol. 2015;195:5117–5122. doi:10.4049/jimmunol.1501657.
  • Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi:10.1126/science.aaa1348.
  • Boon T, Cerottini JC, Van Den Eynde B, Van Der Bruggen P, Van Pel A. Tumor antigens recognized by T lymphocytes. Annu Rev Immunol. 1994;12:337–365. doi:10.1146/annurev.iy.12.040194.002005.
  • Boon T. van der Bruggen P. Human tumor antigens recognized by T lymphocytes. J Exp Med. 1996;183:725–729.
  • Van Pel A, Van Der Bruggen P, Coulie PG, Brichard VG, Lethe B, Van Den Eynde B, Uyttenhove C, Renauld JC, Boon T. Genes coding for tumor antigens recognized by cytolytic T lymphocytes. Immunol Rev. 1995;145:229–250.
  • Blander JM. Regulation of the cell biology of antigen cross-presentation. Annu Rev Immunol. 2018;36:717–753. doi:10.1146/annurev-immunol-041015-055523.
  • Kloetzel PM, Ossendorp F. Proteasome and peptidase function in MHC-class-I-mediated antigen presentation. Curr Opin Immunol. 2004;16:76–81.
  • Gerner MY, Casey KA, Kastenmuller W, Germain RN. Dendritic cell and antigen dispersal landscapes regulate T cell immunity. J Exp Med. 2017;214:3105–3122. doi:10.1084/jem.20170335.
  • Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21:938–945. doi:10.1038/nm.3909.
  • Van Den Eynde BJ, Boon T. Tumor antigens recognized by T lymphocytes. Int J Clin Lab Res. 1997;27:81–86.
  • Menez-Jamet J, Gallou C, Rougeot A, Kosmatopoulos K. Optimized tumor cryptic peptides: the basis for universal neo-antigen-like tumor vaccines. Ann Transl Med. 2016;4:266. doi:10.21037/atm.2016.05.15.
  • Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125:3413–3421. doi:10.1172/JCI80008.
  • Wang RF, Wang HY. Immune targets and neoantigens for cancer immunotherapy and precision medicine. Cell Res. 2017;27:11–37. doi:10.1038/cr.2016.155.
  • Kahles A, Lehmann KV, Toussaint NC, Huser M, Stark SG, Sachsenberg T, Stegle O, Kohlbacher O, Sander C, Cancer Genome Atlas Research N et al. Comprehensive analysis of alternative splicing across tumors from 8,705 patients. Cancer Cell. 2018. doi:10.1016/j.ccell.2018.07.001.
  • Hamada T, Soong TR, Masugi Y, Kosumi K, Nowak JA, Da Silva A, Mu XJ, Twombly TS, Koh H, Yang J, et al. TIME (Tumor Immunity in the MicroEnvironment) classification based on tumor CD274 (PD-L1) expression status and tumor-infiltrating lymphocytes in colorectal carcinomas. Oncoimmunology. 2018;7:e1442999. doi:10.1080/2162402X.2018.1442999.
  • Martin SD, Wick DA, Nielsen JS, Little N, Holt RA, Nelson BH. A library-based screening method identifies neoantigen-reactive T cells in peripheral blood prior to relapse of ovarian cancer. Oncoimmunology. 2017;7:e1371895. doi:10.1080/2162402X.2017.1371895.
  • Dang E, Yang S, Song C, Jiang D, Li Z, Fan W, Sun Y, Tao L, Wang J, Liu T, et al. BAP31, a newly defined cancer/testis antigen, regulates proliferation, migration, and invasion to promote cervical cancer progression. Cell Death Dis. 2018;9:791. doi:10.1038/s41419-018-0824-2.
  • Mufson RA. Tumor antigen targets and tumor immunotherapy. Front Biosci. 2006;11:337–343.
  • Schietinger A, Philip M, Schreiber H. Specificity in cancer immunotherapy. Semin Immunol. 2008;20:276–285. doi:10.1016/j.smim.2008.07.001.
  • Chang AY, Dao T, Gejman RS, Jarvis CA, Scott A, Dubrovsky L, Mathias MD, Korontsvit T, Zakhaleva V, Curcio M, et al. A therapeutic T cell receptor mimic antibody targets tumor-associated PRAME peptide/HLA-I antigens. J Clin Invest. 2017;127:2705–2718. doi:10.1172/JCI92335.
  • Schumacher TN, Hacohen N. Neoantigens encoded in the cancer genome. Curr Opin Immunol. 2016;41:98–103. doi:10.1016/j.coi.2016.07.005.
  • Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi:10.1126/science.aaa4971.
  • Verdegaal EM, De Miranda NF, Visser M, Harryvan T, Van Buuren MM, Andersen RS, Hadrup SR, Van Der Minne CE, Schotte R, Spits H, et al. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature. 2016;536:91–95. doi:10.1038/nature18945.
  • Cha SC, Qin H, Sakamaki I, Kwak L. Cloning variable region genes of clonal lymphoma immunoglobulin for generating patient-specific idiotype DNA vaccine. Methods Mol Biol. 2014;1139:289–303. doi:10.1007/978-1-4939-0345-0_24.
  • Kurth R, Fenyo EM, Klein E, Essex M. Cell-surface antigens induced by RNA tumour viruses. Nature. 1979;279:197–201.
  • Wang C, Dickie J, Sutavani RV, Pointer C, Thomas GJ, Savelyeva N. Targeting head and neck cancer by vaccination. Front Immunol. 2018;9:830. doi:10.3389/fimmu.2018.00830.
  • Qin Y, Ekmekcioglu S, Forget MA, Szekvolgyi L, Hwu P, Grimm EA, Jazaeri AA, Roszik J. Cervical cancer neoantigen landscape and immune activity is associated with human papillomavirus master regulators. Front Immunol. 2017;8:689. doi:10.3389/fimmu.2017.00689.
  • Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing and optimizing new generation vaccines. Nat Rev Immunol. 2001;1:209–219. doi:10.1038/35105075.
  • Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–915. doi:10.1038/nm1100.
  • Schumacher T, Bunse L, Pusch S, Sahm F, Wiestler B, Quandt J, Menn O, Osswald M, Oezen I, Ott M, et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature. 2014;512:324–327. doi:10.1038/nature13387.
  • McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, Jamal-Hanjani M, Wilson GA, Birkbak NJ, Hiley CT, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–1469. doi:10.1126/science.aaf1490.
  • Jager E, Jager D, Knuth A. Clinical cancer vaccine trials. Curr Opin Immunol. 2002;14:178–182.
  • Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol. 2010;10:787–796. doi:10.1038/nri2868.
  • Melief CJ, Van Hall T, Arens R, Ossendorp F, van der Burg SH. Therapeutic cancer vaccines. J Clin Invest. 2015;125:3401–3412. doi:10.1172/JCI80009.
  • Sawada Y, Yoshikawa T, Ofuji K, Yoshimura M, Tsuchiya N, Takahashi M, Nobuoka D, Gotohda N, Takahashi S, Kato Y, et al. Phase II study of the GPC3-derived peptide vaccine as an adjuvant therapy for hepatocellular carcinoma patients. Oncoimmunology. 2016;5:e1129483. doi:10.1080/2162402X.2015.1129483.
  • Melero I, Gaudernack G, Gerritsen W, Huber C, Parmiani G, Scholl S, Thatcher N, Wagstaff J, Zielinski C, Faulkner I, et al. Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol. 2014;11:509–524. doi:10.1038/nrclinonc.2014.111.
  • Bobisse S, Foukas PG, Coukos G, Harari A. Neoantigen-based cancer immunotherapy. Ann Transl Med. 2016;4:262. doi:10.21037/atm.2016.06.17.
  • Chiang CL, Coukos G, Kandalaft LE. Whole tumor antigen vaccines: where are we? Vaccines (Basel). 2015;3:344–372. doi:10.3390/vaccines3020344.
  • Zhu G, Lynn GM, Jacobson O, Chen K, Liu Y, Zhang H, Ma Y, Zhang F, Tian R, Ni Q, et al. Albumin/vaccine nanocomplexes that assemble in vivo for combination cancer immunotherapy. Nat Commun. 2017;8:1954. doi:10.1038/s41467-017-02191-y.
  • Butterfield LH, Zhao F, Lee S, Tarhini AA, Margolin KA, White RL, Atkins MB, Cohen GI, Whiteside TL, Kirkwood JM, et al. Immune correlates of GM-CSF and melanoma peptide vaccination in a randomized trial for the adjuvant therapy of resected high-risk melanoma (E4697). Clin Cancer Res. 2017;23:5034–5043. doi:10.1158/1078-0432.CCR-16-3016.
  • Li F, Chen C, Ju T, Gao J, Yan J, Wang P, Xu Q, Hwu P, Du X, Lizee G. Rapid tumor regression in an Asian lung cancer patient following personalized neo-epitope peptide vaccination. Oncoimmunology. 2016;5:e1238539. doi:10.1080/2162402X.2016.1238539.
  • Bijker MS, Melief CJ, Offringa R, van der Burg SH. Design and development of synthetic peptide vaccines: past, present and future. Expert Rev Vaccines. 2007;6:591–603. doi:10.1586/14760584.6.4.591.
  • Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, Gailani F, Riley L, Conlon K, Pockaj B, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119–2127. doi:10.1056/NEJMoa1012863.
  • Gouttefangeas C, Rammensee HG. Personalized cancer vaccines: adjuvants are important, too. Cancer Immunol Immunother. 2018. doi:10.1007/s00262-018-2158-4.
  • Silva AL, Rosalia RA, Sazak A, Carstens MG, Ossendorp F, Oostendorp J, Jiskoot W. Optimization of encapsulation of a synthetic long peptide in PLGA nanoparticles: low-burst release is crucial for efficient CD8(+) T cell activation. Eur J Pharm Biopharm. 2013;83:338–345. doi:10.1016/j.ejpb.2012.11.006.
  • Silva JM, Videira M, Gaspar R, Preat V, Florindo HF. Immune system targeting by biodegradable nanoparticles for cancer vaccines. J Control Release. 2013;168:179–199. doi:10.1016/j.jconrel.2013.03.010.
  • Varypataki EM, Silva AL, Barnier-Quer C, Collin N, Ossendorp F, Jiskoot W. Synthetic long peptide-based vaccine formulations for induction of cell mediated immunity: A comparative study of cationic liposomes and PLGA nanoparticles. J Control Release. 2016;226:98–106. doi:10.1016/j.jconrel.2016.02.018.
  • Avigan D, Rosenblatt J. Vaccine therapy in hematologic malignancies. Blood. 2018;131:2640–2650. doi:10.1182/blood-2017-11-785873.
  • Bloy N, Garcia P, Laumont CM, Pitt JM, Sistigu A, Stoll G, Yamazaki T, Bonneil E, Buque A, Humeau J, et al. Immunogenic stress and death of cancer cells: contribution of antigenicity vs adjuvanticity to immunosurveillance. Immunol Rev. 2017;280:165–174. doi:10.1111/imr.12582.
  • Galluzzi L, Zitvogel L, Kroemer G. Immunological mechanisms underneath the efficacy of cancer therapy. Cancer Immunol Res. 2016;4:895–902. doi:10.1158/2326-6066.CIR-16-0197.
  • Klein L, Hinterberger M, Wirnsberger G, Kyewski B. Antigen presentation in the thymus for positive selection and central tolerance induction. Nat Rev Immunol. 2009;9:833–844. doi:10.1038/nri2669.
  • Kroemer G, Senovilla L, Galluzzi L, Andre F, Zitvogel L. Natural and therapy-induced immunosurveillance in breast cancer. Nat Med. 2015;21:1128–1138. doi:10.1038/nm.3944.
  • Arrieta VA, Cacho-Diaz B, Zhao J, Rabadan R, Chen L, Sonabend AM. The possibility of cancer immune editing in gliomas. A Critical Review. Oncoimmunology. 2018;7:e1445458. doi:10.1080/2162402X.2018.1445458.
  • Terry S, Buart S, Tan TZ, Gros G, Noman MZ, Lorens JB, Mami-Chouaib F, Thiery JP, Chouaib S. Acquisition of tumor cell phenotypic diversity along the EMT spectrum under hypoxic pressure: consequences on susceptibility to cell-mediated cytotoxicity. Oncoimmunology. 2017;6:e1271858. doi:10.1080/2162402X.2016.1271858.
  • Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. The determinants of tumour immunogenicity. Nat Rev Cancer. 2012;12:307–313. doi:10.1038/nrc3246.
  • Pietrocola F, Bravo-San Pedro JM, Galluzzi L, Kroemer G. Autophagy in natural and therapy-driven anticancer immunosurveillance. Autophagy. 2017;13:2163–2170. doi:10.1080/15548627.2017.1310356.
  • Bui JD, Schreiber RD. Cancer immunosurveillance, immunoediting and inflammation: independent or interdependent processes? Curr Opin Immunol. 2007;19:203–208. doi:10.1016/j.coi.2007.02.001.
  • Shime H, Maruyama A, Yoshida S, Takeda Y, Matsumoto M, Seya T. Toll-like receptor 2 ligand and interferon-gamma suppress anti-tumor T cell responses by enhancing the immunosuppressive activity of monocytic myeloid-derived suppressor cells. Oncoimmunology. 2017;7:e1373231. doi:10.1080/2162402X.2017.1373231.
  • Chaoul N, Tang A, Desrues B, Oberkampf M, Fayolle C, Ladant D, Sainz-Perez A, Leclerc C. Lack of MHC class II molecules favors CD8(+) T-cell infiltration into tumors associated with an increased control of tumor growth. Oncoimmunology. 2018;7:e1404213. doi:10.1080/2162402X.2017.1404213.
  • Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6:836–848. doi:10.1038/nri1961.
  • Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–998. doi:10.1038/ni1102-991.
  • Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, Staehler M, Brugger W, Dietrich PY, Mendrzyk R, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18:1254–1261. doi:10.1038/nm.2883.
  • Pol J, Bloy N, Buque A, Eggermont A, Cremer I, Sautes-Fridman C, Galon J, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: peptide-based anticancer vaccines. Oncoimmunology. 2015;4:e974411. doi:10.4161/2162402X.2014.974411.
  • Romero P, Banchereau J, Bhardwaj N, Cockett M, Disis ML, Dranoff G, Gilboa E, Hammond SA, Hershberg R, Korman AJ, et al. The human vaccines project: a roadmap for cancer vaccine development. Sci Transl Med. 2016;8:334–339. doi:10.1126/scitranslmed.aaf0685.
  • Hos BJ, Tondini E, Van Kasteren SI, Ossendorp F. Approaches to improve chemically defined synthetic peptide vaccines. Front Immunol. 2018;9:884. doi:10.3389/fimmu.2018.00884.
  • Arens R, Van Hall T, Van Der Burg SH, Ossendorp F, Melief CJ. Prospects of combinatorial synthetic peptide vaccine-based immunotherapy against cancer. Semin Immunol. 2013;25:182–190. doi:10.1016/j.smim.2013.04.008.
  • Drug and Device News. P T. 2017;42:554–593.
  • Agosti JM, Goldie SJ. Introducing HPV vaccine in developing countries–key challenges and issues. N Engl J Med. 2007;356:1908–1910. doi:10.1056/NEJMp078053.
  • Paavonen J, Naud P, Salmeron J, Wheeler CM, Chow SN, Apter D, Kitchener H, Castellsague X, Teixeira JC, Skinner SR, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009;374:301–314. doi:10.1016/S0140-6736(09)61248-4.
  • Group FIS. Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med. 2007;356:1915–1927. doi:10.1056/NEJMoa061741.
  • Zhai L, Tumban E. Gardasil-9: A global survey of projected efficacy. Antiviral Res. 2016;130:101–109. doi:10.1016/j.antiviral.2016.03.016.
  • Cuzick J. Gardasil 9 joins the fight against cervix cancer. Expert Rev Vaccines. 2015;14:1047–1049. doi:10.1586/14760584.2015.1051470.
  • Van Poelgeest MI, Welters MJ, Van Esch EM, Stynenbosch LF, Kerpershoek G, Van Persijn Van Meerten EL, Van Den Hende M, Lowik MJ, Berends-Van Der Meer DM, Fathers LM, et al. HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial. J Transl Med. 2013;11:88. doi:10.1186/1479-5876-11-88.
  • Van Poelgeest MI, Welters MJ, Vermeij R, Stynenbosch LF, Loof NM, Berends-Van Der Meer DM, Lowik MJ, Hamming IL, Van Esch EM, Hellebrekers BW, et al. Vaccination against oncoproteins of HPV16 for noninvasive vulvar/vaginal lesions: lesion clearance is related to the strength of the T-cell response. Clin Cancer Res. 2016;22:2342–2350. doi:10.1158/1078-0432.CCR-15-2594.
  • Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-Van Der Meer DM, Vloon AP, Essahsah F, Fathers LM, Offringa R, Drijfhout JW, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838–1847. doi:10.1056/NEJMoa0810097.
  • Obeid J, Hu Y, Slingluff CL Jr. Vaccines, adjuvants, and dendritic cell activators–current status and future challenges. Semin Oncol. 2015;42:549–561. doi:10.1053/j.seminoncol.2015.05.006.
  • Moynihan KD, Opel CF, Szeto GL, Tzeng A, Zhu EF, Engreitz JM, Williams RT, Rakhra K, Zhang MH, Rothschilds AM, et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med. 2016;22:1402–1410. doi:10.1038/nm.4200.
  • Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J, Arthur CD, Krebber WJ, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–581. doi:10.1038/nature13988.
  • Hailemichael Y, Woods A, Fu T, He Q, Nielsen MC, Hasan F, Roszik J, Xiao Z, Vianden C, Khong H, et al. Cancer vaccine formulation dictates synergy with CTLA-4 and PD-L1 checkpoint blockade therapy. J Clin Invest. 2018;128:1338–1354. doi:10.1172/JCI93303.
  • Popovic A, Jaffee EM, Zaidi N. Emerging strategies for combination checkpoint modulators in cancer immunotherapy. J Clin Invest. 2018;128:3209–3218. doi:10.1172/JCI120775.
  • Nagaoka K, Hosoi A, Iino T, Morishita Y, Matsushita H, Kakimi K. Dendritic cell vaccine induces antigen-specific CD8(+) T cells that are metabolically distinct from those of peptide vaccine and is well-combined with PD-1 checkpoint blockade. Oncoimmunology. 2018;7:e1395124. doi:10.1080/2162402X.2017.1395124.
  • Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. doi:10.1056/NEJMoa1003466.
  • Fournier C, Martin F, Zitvogel L, Kroemer G, Galluzzi L, Apetoh L. Trial watch: adoptively transferred cells for anticancer immunotherapy. Oncoimmunology. 2017;6:e1363139. doi:10.1080/2162402X.2017.1363139.
  • Pierini S, Perales-Linares R, Uribe-Herranz M, Pol JG, Zitvogel L, Kroemer G, Facciabene A, Galluzzi L. Trial watch: DNA-based vaccines for oncological indications. Oncoimmunology. 2017;6:e1398878. doi:10.1080/2162402X.2017.1398878.
  • Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, Jassem J, Yoshimura M, Dahabreh J, Nakayama H, et al. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2016;17:822–835. doi:10.1016/S1470-2045(16)00099-1.
  • Higgins M, Curigliano G, Dieras V, Kuemmel S, Kunz G, Fasching PA, Campone M, Bachelot T, Krivorotko P, Chan S, et al. Safety and immunogenicity of neoadjuvant treatment using WT1-immunotherapeutic in combination with standard therapy in patients with WT1-positive Stage II/III breast cancer: a randomized Phase I study. Breast Cancer Res Treat. 2017;162:479–488. doi:10.1007/s10549-017-4130-y.
  • Obara W, Eto M, Mimata H, Kohri K, Mitsuhata N, Miura I, Shuin T, Miki T, Koie T, Fujimoto H, et al. A phase I/II study of cancer peptide vaccine S-288310 in patients with advanced urothelial carcinoma of the bladder. Ann Oncol. 2017;28:798–803. doi:10.1093/annonc/mdw675.
  • Obara W, Sato F, Takeda K, Kato R, Kato Y, Kanehira M, Takata R, Mimata H, Sugai T, Nakamura Y, et al. Phase I clinical trial of cell division associated 1 (CDCA1) peptide vaccination for castration resistant prostate cancer. Cancer Sci. 2017;108:1452–1457. doi:10.1111/cas.13278.
  • Slingluff CL Jr., Petroni GR, Olson WC, Smolkin ME, Chianese-Bullock KA, Mauldin IS, Smith KT, Deacon DH, Varhegyi NE, Donnelly SB, et al. A randomized pilot trial testing the safety and immunologic effects of a MAGE-A3 protein plus AS15 immunostimulant administered into muscle or into dermal/subcutaneous sites. Cancer Immunol Immunother. 2016;65:25–36. doi:10.1007/s00262-015-1770-9.
  • Gutzmer R, Rivoltini L, Levchenko E, Testori A, Utikal J, Ascierto PA, Demidov L, Grob JJ, Ridolfi R, Schadendorf D, et al. Safety and immunogenicity of the PRAME cancer immunotherapeutic in metastatic melanoma: results of a phase I dose escalation study. ESMO Open. 2016;1:e000068. doi:10.1136/esmoopen-2016-000068.
  • Saiag P, Gutzmer R, Ascierto PA, Maio M, Grob JJ, Murawa P, Dreno B, Ross M, Weber J, Hauschild A, et al. Prospective assessment of a gene signature potentially predictive of clinical benefit in metastatic melanoma patients following MAGE-A3 immunotherapeutic (PREDICT). Ann Oncol. 2016;27:1947–1953. doi:10.1093/annonc/mdw291.
  • Pujol JL, Vansteenkiste JF, De Pas TM, Atanackovic D, Reck M, Thomeer M, Douillard JY, Fasola G, Potter V, Taylor P, et al. Safety and Immunogenicity of MAGE-A3 cancer immunotherapeutic with or without adjuvant chemotherapy in patients with resected stage IB to III MAGE-A3-positive non-small-cell lung cancer. J Thorac Oncol. 2015;10:1458–1467. doi:10.1097/JTO.0000000000000653.
  • Stanton SE, Eary JF, Marzbani EA, Mankoff D, Salazar LG, Higgins D, Childs J, Reichow J, Dang Y, Disis ML. Concurrent SPECT/PET-CT imaging as a method for tracking adoptively transferred T-cells in vivo. J Immunother Cancer. 2016;4:27. doi:10.1186/s40425-016-0131-3.
  • Berinstein NL, Karkada M, Oza AM, Odunsi K, Villella JA, Nemunaitis JJ, Morse MA, Pejovic T, Bentley J, Buyse M, et al. Survivin-targeted immunotherapy drives robust polyfunctional T cell generation and differentiation in advanced ovarian cancer patients. Oncoimmunology. 2015;4:e1026529. doi:10.1080/2162402X.2015.1026529.
  • Eckert F, Gaipl US, Niedermann G, Hettich M, Schilbach K, Huber SM, Zips D. Beyond checkpoint inhibition - Immunotherapeutical strategies in combination with radiation. Clin Transl Radiat Oncol. 2017;2:29–35. doi:10.1016/j.ctro.2016.12.006.
  • Brayer J, Lancet JE, Powers J, List A, Balducci L, Komrokji R, Pinilla-Ibarz J. WT1 vaccination in AML and MDS: A pilot trial with synthetic analog peptides. Am J Hematol. 2015;90:602–607. doi:10.1002/ajh.24014.
  • Okada H, Butterfield LH, Hamilton RL, Hoji A, Sakaki M, Ahn BJ, Kohanbash G, Drappatz J, Engh J, Amankulor N, et al. Induction of robust type-I CD8+ T-cell responses in WHO grade 2 low-grade glioma patients receiving peptide-based vaccines in combination with poly-ICLC. Clin Cancer Res. 2015;21:286–294. doi:10.1158/1078-0432.CCR-14-1790.
  • Weller M, Butowski N, Tran DD, Recht LD, Lim M, Hirte H, Ashby L, Mechtler L, Goldlust SA, Iwamoto F, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017;18:1373–1385. doi:10.1016/S1470-2045(17)30517-X.
  • Wood LV, Fojo A, Roberson BD, Hughes MS, Dahut W, Gulley JL, Madan RA, Arlen PM, Sabatino M, Stroncek DF, et al. TARP vaccination is associated with slowing in PSA velocity and decreasing tumor growth rates in patients with stage D0 prostate cancer. Oncoimmunology. 2016;5:e1197459. doi:10.1080/2162402X.2016.1197459.
  • Mauldin IS, Wages NA, Stowman AM, Wang E, Olson WC, Deacon DH, Smith KT, Galeassi N, Teague JE, Smolkin ME, et al. Topical treatment of melanoma metastases with imiquimod, plus administration of a cancer vaccine, promotes immune signatures in the metastases. Cancer Immunol Immunother. 2016;65:1201–1212. doi:10.1007/s00262-016-1880-z.
  • Mauldin IS, Wages NA, Stowman AM, Wang E, Smolkin ME, Olson WC, Deacon DH, Smith KT, Galeassi NV, Chianese-Bullock KA, et al. Intratumoral interferon-gamma increases chemokine production but fails to increase T cell infiltration of human melanoma metastases. Cancer Immunol Immunother. 2016;65:1189–1199. doi:10.1007/s00262-016-1881-y.
  • Nitschke NJ, Bjoern J, Iversen TZ, Andersen MH, Svane IM. Indoleamine 2,3-dioxygenase and survivin peptide vaccine combined with temozolomide in metastatic melanoma. Stem Cell Investig. 2017;4:77. doi:10.21037/sci.2017.08.06.
  • Sabado RL, Pavlick A, Gnjatic S, Cruz CM, Vengco I, Hasan F, Spadaccia M, Darvishian F, Chiriboga L, Holman RM, et al. Resiquimod as an immunologic adjuvant for NY-ESO-1 protein vaccination in patients with high-risk melanoma. Cancer Immunol Res. 2015;3:278–287. doi:10.1158/2326-6066.CIR-14-0202.
  • Kobayashi Y, Sakura T, Miyawaki S, Toga K, Sogo S, Heike Y. A new peptide vaccine OCV-501: in vitro pharmacology and phase 1 study in patients with acute myeloid leukemia. Cancer Immunol Immunother. 2017;66:851–863. doi:10.1007/s00262-017-1981-3.
  • Sakamoto S, Yamada T, Terazaki Y, Yoshiyama K, Sugawara S, Takamori S, Matsueda S, Shichijo S, Yamada A, Noguchi M, et al. Feasibility study of personalized peptide vaccination for advanced small cell lung cancer. Clin Lung Cancer. 2017;18:e385–e394. doi:10.1016/j.cllc.2017.03.011.
  • Mittendorf EA, Ardavanis A, Litton JK, Shumway NM, Hale DF, Murray JL, Perez SA, Ponniah S, Baxevanis CN, Papamichail M, et al. Primary analysis of a prospective, randomized, single-blinded phase II trial evaluating the HER2 peptide GP2 vaccine in breast cancer patients to prevent recurrence. Oncotarget. 2016;7:66192–66201. doi:10.18632/oncotarget.11751.
  • Baumgaertner P, Costa Nunes C, Cachot A, Maby-El Hajjami H, Cagnon L, Braun M, Derre L, Rivals JP, Rimoldi D, Gnjatic S, et al. Vaccination of stage III/IV melanoma patients with long NY-ESO-1 peptide and CpG-B elicits robust CD8(+) and CD4(+) T-cell responses with multiple specificities including a novel DR7-restricted epitope. Oncoimmunology. 2016;5:e1216290. doi:10.1080/2162402X.2016.1216290.
  • Hasegawa K, Ikeda Y, Kunugi Y, Kurosaki A, Imai Y, Kohyama S, Nagao S, Kozawa E, Yoshida K, Tsunoda T, et al. Phase I study of multiple epitope peptide vaccination in patients with recurrent or persistent cervical cancer. J Immunother. 2018;41:201–207. doi:10.1097/CJI.0000000000000214.
  • Yutani S, Shirahama T, Muroya D, Matsueda S, Yamaguchi R, Morita M, Shichijo S, Yamada A, Sasada T, Itoh K. Feasibility study of personalized peptide vaccination for hepatocellular carcinoma patients refractory to locoregional therapies. Cancer Sci. 2017;108:1732–1738. doi:10.1111/cas.13301.
  • Shirahama T, Muroya D, Matsueda S, Yamada A, Shichijo S, Naito M, Yamashita T, Sakamoto S, Okuda K, Itoh K, et al. A randomized phase II trial of personalized peptide vaccine with low dose cyclophosphamide in biliary tract cancer. Cancer Sci. 2017;108:838–845. doi:10.1111/cas.13193.
  • Kohrt HE, Muller A, Baker J, Goldstein MJ, Newell E, Dutt S, Czerwinski D, Lowsky R, Strober S. Donor immunization with WT1 peptide augments antileukemic activity after MHC-matched bone marrow transplantation. Blood. 2011;118:5319–5329. doi:10.1182/blood-2011-05-356238.
  • Kimura T, Egawa S, Uemura H. Personalized peptide vaccines and their relation to other therapies in urological cancer. Nat Rev Urol. 2017;14:501–510. doi:10.1038/nrurol.2017.77.
  • Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–221. doi:10.1038/nature22991.
  • Melief CJM. Cancer: precision T-cell therapy targets tumours. Nature. 2017;547:165–167. doi:10.1038/nature23093.
  • Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Borresen-Dale AL, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi:10.1038/nature12477.
  • Gall VA, Philips AV, Qiao N, Clise-Dwyer K, Perakis AA, Zhang M, Clifton GT, Sukhumalchandra P, Ma Q, Reddy SM, et al. Trastuzumab increases HER2 uptake and cross-presentation by dendritic cells. Cancer Res. 2017;77:5374–5383. doi:10.1158/0008-5472.CAN-16-2774.
  • Tsuruta M, Ueda S, Yew PY, Fukuda I, Yoshimura S, Kishi H, Hamana H, Hirayama M, Yatsuda J, Irie A, et al. Bladder cancer-associated cancer-testis antigen-derived long peptides encompassing both CTL and promiscuous HLA class II-restricted Th cell epitopes induced CD4(+) T cells expressing converged T-cell receptor genes in vitro. Oncoimmunology. 2018;7:e1415687. doi:10.1080/2162402X.2017.1415687.
  • Petrizzo A, Mauriello A, Luciano A, Rea D, Barbieri A, Arra C, Maiolino P, Tornesello M, Gigantino V, Botti G, et al. Inhibition of tumor growth by cancer vaccine combined with metronomic chemotherapy and anti-PD-1 in a pre-clinical setting. Oncotarget. 2018;9:3576–3589. doi:10.18632/oncotarget.23181.
  • Tanaka H, Hazama S, Iida M, Tsunedomi R, Takenouchi H, Nakajima M, Tokumitsu Y, Kanekiyo S, Shindo Y, Tomochika S, et al. miR-125b-1 and miR-378a are predictive biomarkers for the efficacy of vaccine treatment against colorectal cancer. Cancer Sci. 2017;108:2229–2238. doi:10.1111/cas.13390.
  • Huang L, Wang Z, Liu C, Xu C, Mbofung RM, McKenzie JA, Khong H, Hwu P, Peng W. CpG-based immunotherapy impairs antitumor activity of BRAF inhibitors in a B-cell-dependent manner. Oncogene. 2017;36:4081–4086. doi:10.1038/onc.2017.35.
  • Yarchoan M, Johnson BA 3rd, Lutz ER, Laheru DA, Jaffee EM. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer. 2017;17:209–222. doi:10.1038/nrc.2016.154.
  • Purcell AW, McCluskey J, Rossjohn J. More than one reason to rethink the use of peptides in vaccine design. Nat Rev Drug Discov. 2007;6:404–414. doi:10.1038/nrd2224.
  • Melief CJ, van der Burg SH. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat Rev Cancer. 2008;8:351–360. doi:10.1038/nrc2373.
  • Goodridge HS, Ahmed SS, Curtis N, Kollmann TR, Levy O, Netea MG, Pollard AJ, Van Crevel R, Wilson CB. Harnessing the beneficial heterologous effects of vaccination. Nat Rev Immunol. 2016;16:392–400. doi:10.1038/nri.2016.43.
  • Pashine A, Valiante NM, Ulmer JB. Targeting the innate immune response with improved vaccine adjuvants. Nat Med. 2005;11:S63–68. doi:10.1038/nm1210.
  • Chiang CL, Kandalaft LE, Coukos G. Adjuvants for enhancing the immunogenicity of whole tumor cell vaccines. Int Rev Immunol. 2011;30:150–182. doi:10.3109/08830185.2011.572210.
  • McElrath MJ. Adjuvants: tailoring humoral immune responses. Curr Opin HIV AIDS. 2017;12:278–284. doi:10.1097/COH.0000000000000365.
  • Chianese-Bullock KA, Pressley J, Garbee C, Hibbitts S, Murphy C, Yamshchikov G, Petroni GR, Bissonette EA, Neese PY, Grosh WW, et al. MAGE-A1-, MAGE-A10-, and gp100-derived peptides are immunogenic when combined with granulocyte-macrophage colony-stimulating factor and montanide ISA-51 adjuvant and administered as part of a multipeptide vaccine for melanoma. J Immunol. 2005;174:3080–3086.
  • Verdijk RM, Mutis T, Esendam B, Kamp J, Melief CJ, Brand A, Goulmy E. Polyriboinosinic polyribocytidylic acid (poly(I:C)) induces stable maturation of functionally active human dendritic cells. J Immunol. 1999;163:57–61.
  • Van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med. 1999;190:355–366.
  • Van Den Eertwegh AJ, Versluis J, Van Den Berg HP, Santegoets SJ, Van Moorselaar RJ, Van Der Sluis TM, Gall HE, Harding TC, Jooss K, Lowy I, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:509–517. doi:10.1016/S1470-2045(12)70007-4.
  • Lawson DH, Lee S, Zhao F, Tarhini AA, Margolin KA, Ernstoff MS, Atkins MB, Cohen GI, Whiteside TL, Butterfield LH, et al. Randomized, placebo-controlled, phase III trial of yeast-derived granulocyte-macrophage colony-stimulating factor (GM-CSF) versus peptide vaccination versus GM-CSF plus peptide vaccination versus placebo in patients with no evidence of disease after complete surgical resection of locally advanced and/or stage IV melanoma: a trial of the eastern cooperative oncology group-american college of radiology imaging network cancer research group (E4697). J Clin Oncol. 2015;33:4066–4076. doi:10.1200/JCO.2015.62.0500.
  • Van Der Sluis TC, Van Duikeren S, Huppelschoten S, Jordanova ES, Beyranvand Nejad E, Sloots A, Boon L, Smit VT, Welters MJ, Ossendorp F, et al. Vaccine-induced tumor necrosis factor-producing T cells synergize with cisplatin to promote tumor cell death. Clin Cancer Res. 2015;21:781–794. doi:10.1158/1078-0432.CCR-14-2142.
  • Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8:59–73. doi:10.1038/nri2216.
  • Dranoff G. Tailor-made renal cell carcinoma vaccines. Cancer Cell. 2012;22:287–289. doi:10.1016/j.ccr.2012.08.021.
  • Tagliamonte M, Petrizzo A, Napolitano M, Luciano A, Arra C, Maiolino P, Izzo F, Tornesello ML, Aurisicchio L, Ciliberto G, et al. Novel metronomic chemotherapy and cancer vaccine combinatorial strategy for hepatocellular carcinoma in a mouse model. Cancer Immunol Immunother. 2015;64:1305–1314. doi:10.1007/s00262-015-1698-0.
  • Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, Formenti SC. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58:862–870. doi:10.1016/j.ijrobp.2003.09.012.
  • Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, Barcellos-Hoff MH, Demaria S. TGFbeta is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res. 2015;75:2232–2242. doi:10.1158/0008-5472.CAN-14-3511.
  • Wennerberg E, Lhuillier C, Vanpouille-Box C, Pilones KA, Garcia-Martinez E, Rudqvist NP, Formenti SC, Demaria S. Barriers to radiation-induced in situ tumor vaccination. Front Immunol. 2017;8:229. doi:10.3389/fimmu.2017.00229.
  • Cadena A, Cushman TR, Anderson C, Barsoumian HB, Welsh JW, Cortez MA. Radiation and anti-cancer vaccines: a winning combination. Vaccines (Basel). 2018;6. doi:10.3390/vaccines6010009.
  • Yamaue H, Tsunoda T, Tani M, Miyazawa M, Yamao K, Mizuno N, Okusaka T, Ueno H, Boku N, Fukutomi A, et al. Randomized phase II/III clinical trial of elpamotide for patients with advanced pancreatic cancer: PEGASUS-PC study. Cancer Sci. 2015;106:883–890. doi:10.1111/cas.12674.
  • Melssen M, Slingluff CL Jr. Vaccines targeting helper T cells for cancer immunotherapy. Curr Opin Immunol. 2017;47:85–92. doi:10.1016/j.coi.2017.07.004.
  • Keefe DM, Bateman EH. Tumor control versus adverse events with targeted anticancer therapies. Nat Rev Clin Oncol. 2011;9:98–109. doi:10.1038/nrclinonc.2011.192.
  • Lopez JS, Banerji U. Combine and conquer: challenges for targeted therapy combinations in early phase trials. Nat Rev Clin Oncol. 2017;14:57–66. doi:10.1038/nrclinonc.2016.96.
  • Schmidt C. The benefits of immunotherapy combinations. Nature. 2017;552:S67–S69. doi:10.1038/d41586-017-08702-7.
  • Versteven M, Jmj VDB, Marcq E, Smits ELJ, Van Tendeloo VFI, Hobo W, Lion E. Dendritic cells and programmed death-1 blockade: a joint venture to combat cancer. Front Immunol. 2018;9:394. doi:10.3389/fimmu.2018.00394.
  • Jacquelot N, Roberti MP, Enot DP, Rusakiewicz S, Ternes N, Jegou S, Woods DM, Sodre AL, Hansen M, Meirow Y, et al. Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun. 2017;8:592. doi:10.1038/s41467-017-00608-2.
  • Smyth MJ, Ngiow SF, Ribas A, Teng MW. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol. 2016;13:143–158. doi:10.1038/nrclinonc.2015.209.
  • Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAS, Andrews MC, Sharma P, Wang J, Wargo JA, Pe’er D, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell. 2017;170:1120–1133 e1117. doi:10.1016/j.cell.2017.07.024.
  • Liu P, Zhao L, Loos F, Iribarren K, Kepp O, Kroemer G. Epigenetic anticancer agents cause HMGB1 release in vivo. Oncoimmunology. 2018;7:e1431090. doi:10.1080/2162402X.2018.1431090.
  • Pfirschke C, Engblom C, Rickelt S, Cortez-Retamozo V, Garris C, Pucci F, Yamazaki T, Poirier-Colame V, Newton A, Redouane Y, et al. Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity. 2016;44:343–354. doi:10.1016/j.immuni.2015.11.024.
  • Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi:10.1038/nature13954.
  • Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology. 2012;1:1223–1225. doi:10.4161/onci.21335.
  • Kang J, Galluzzi L. PD-L1 blockade for urothelial carcinoma. Oncoimmunology. 2017;6:e1334028. doi:10.1080/2162402X.2017.1334028.
  • Siefker-Radtke A, Curti B. Immunotherapy in metastatic urothelial carcinoma: focus on immune checkpoint inhibition. Nat Rev Urol. 2018;15:112–124. doi:10.1038/nrurol.2017.190.
  • Jelinek T, Hajek R. PD-1/PD-L1 inhibitors in multiple myeloma: the present and the future. Oncoimmunology. 2016;5:e1254856. doi:10.1080/2162402X.2016.1254856.
  • Sharma P, Logothetis C. Prostate cancer: combination of vaccine plus ipilimumab–safety and toxicity. Nat Rev Urol. 2012;9:302–303. doi:10.1038/nrurol.2012.103.
  • Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–1084. doi:10.1126/science.aad1329.
  • Sidaway P. Immunotherapy: local chemotherapy synergizes with CTLA-4 inhibition. Nat Rev Clin Oncol. 2018;15:202. doi:10.1038/nrclinonc.2018.22.
  • Hopkins AC, Yarchoan M, Durham JN, Yusko EC, Rytlewski JA, Robins HS, Laheru DA, Le DT, Lutz ER, Jaffee EM. T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight. 2018;3. doi:10.1172/jci.insight.122092.
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology. 2014;3:e27297. doi:10.4161/onci.27297.
  • Chester C, Sanmamed MF, Wang J, Melero I. Immunotherapy targeting 4-1BB: mechanistic rationale, clinical results, and future strategies. Blood. 2018;131:49–57. doi:10.1182/blood-2017-06-741041.
  • Enhancing PD-1. Blockade in solid tumors. Cancer Discov. 2016;6:OF2. doi:10.1158/2159-8290.CD-NB2016-086.
  • Turaj AH, Hussain K, Cox KL, Rose-Zerilli MJJ, Testa J, Dahal LN, Chan HTC, James S, Field VL, Carter MJ, et al. Antibody tumor targeting is enhanced by CD27 agonists through myeloid recruitment. Cancer Cell. 2017;32:777–791 e776. doi:10.1016/j.ccell.2017.11.001.
  • Wei SM, Fei JX, Tao F, Pan HL, Shen Q, Wang L, Wu YJ, Zhou L, Zhu SX, Liao WB, et al. Anti-CD27 antibody potentiates antitumor effect of dendritic cell-based vaccine in prostate cancer-bearing mice. Int Surg. 2015;100:155–163. doi:10.9738/INTSURG-D-14-00147.1.
  • Vo MC, Jung SH, Chu TH, Lee HJ, Lakshmi TJ, Park HS, Kim HJ, Rhee JH, Lee JJ. Lenalidomide and programmed death-1 blockade synergistically enhances the effects of dendritic cell vaccination in a model of murine myeloma. Front Immunol. 2018;9:1370. doi:10.3389/fimmu.2018.01370.
  • Henry JY, Labarthe MC, Meyer B, Dasgupta P, Dalgleish AG, Galustian C. Enhanced cross-priming of naive CD8+ T cells by dendritic cells treated by the IMiDs(R) immunomodulatory compounds lenalidomide and pomalidomide. Immunology. 2013;139:377–385. doi:10.1111/imm.12087.
  • Petrylak DP, Vogelzang NJ, Budnik N, Wiechno PJ, Sternberg CN, Doner K, Bellmunt J, Burke JM, De Olza MO, Choudhury A, et al. Docetaxel and prednisone with or without lenalidomide in chemotherapy-naive patients with metastatic castration-resistant prostate cancer (MAINSAIL): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2015;16:417–425. doi:10.1016/S1470-2045(15)70025-2.
  • Sakamaki I, Kwak LW, Cha SC, Yi Q, Lerman B, Chen J, Surapaneni S, Bateman S, Qin H. Lenalidomide enhances the protective effect of a therapeutic vaccine and reverses immune suppression in mice bearing established lymphomas. Leukemia. 2014;28:329–337. doi:10.1038/leu.2013.177.
  • Yamada A, Sasada T, Noguchi M, Itoh K. Next-generation peptide vaccines for advanced cancer. Cancer Sci. 2013;104:15–21. doi:10.1111/cas.12050.
  • Sasada T, Yamada A, Noguchi M, Itoh K. Personalized peptide vaccine for treatment of advanced cancer. Curr Med Chem. 2014;21:2332–2345.
  • Roden RBS, Stern PL. Opportunities and challenges for human papillomavirus vaccination in cancer. Nat Rev Cancer. 2018;18:240–254. doi:10.1038/nrc.2018.13.
  • Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, 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. doi:10.1016/S0140-6736(15)00239-1.
  • Killock D. Therapeutic HPV vaccine holds promise. Nat Rev Clin Oncol. 2015;12:686. doi:10.1038/nrclinonc.2015.180.
  • Doyle HA, Koski RA, Bonafe N, Bruck RA, Tagliatela SM, Gee RJ, Mamula MJ. Epidermal growth factor receptor peptide vaccination induces cross-reactive immunity to human EGFR, HER2, and HER3. Cancer Immunol Immunother. 2018. doi:10.1007/s00262-018-2218-9.
  • Palladini A, Thrane S, Janitzek CM, Pihl J, Clemmensen SB, De Jongh WA, Clausen TM, Nicoletti G, Landuzzi L, Penichet ML, et al. Virus-like particle display of HER2 induces potent anti-cancer responses. Oncoimmunology. 2018;7:e1408749. doi:10.1080/2162402X.2017.1408749.
  • Dillon PM, Petroni GR, Smolkin ME, Brenin DR, Chianese-Bullock KA, Smith KT, Olson WC, Fanous IS, Nail CJ, Brenin CM, et al. A pilot study of the immunogenicity of a 9-peptide breast cancer vaccine plus poly-ICLC in early stage breast cancer. J Immunother Cancer. 2017;5:92. doi:10.1186/s40425-017-0295-5.
  • Arab A, Nicastro J, Slavcev R, Razazan A, Barati N, Nikpoor AR, Brojeni AAM, Mosaffa F, Badiee A, Jaafari MR, et al. Lambda phage nanoparticles displaying HER2-derived E75 peptide induce effective E75-CD8(+) T response. Immunol Res. 2018;66:200–206. doi:10.1007/s12026-017-8969-0.
  • Arab A, Behravan J, Razazan A, Gholizadeh Z, Nikpoor AR, Barati N, Mosaffa F, Badiee A, Jaafari MR. A nano-liposome vaccine carrying E75, a HER-2/neu-derived peptide, exhibits significant antitumour activity in mice. J Drug Target. 2018;26:365–372. doi:10.1080/1061186X.2017.1387788.
  • Behravan J, Razazan A, Behravan G. Towards Breast Cancer Vaccines, Progress and Challenges. Curr Drug Discov Technol. 2018. doi:10.2174/1570163815666180502164652.
  • Takeoka T, Nagase H, Kurose K, Ohue Y, Yamasaki M, Takiguchi S, Sato E, Isobe M, Kanazawa T, Matsumoto M, et al. NY-ESO-1 protein cancer vaccine with poly-ICLC and OK-432: rapid and Strong Induction of NY-ESO-1-specific immune responses by poly-ICLC. J Immunother. 2017. doi:10.1097/CJI.0000000000000162.
  • Schneble EJ, Berry JS, Trappey FA, Clifton GT, Ponniah S, Mittendorf E, Peoples GE. The HER2 peptide nelipepimut-S (E75) vaccine (NeuVax) in breast cancer patients at risk for recurrence: correlation of immunologic data with clinical response. Immunotherapy. 2014;6:519–531. doi:10.2217/imt.14.22.
  • Andersen MH. thor SP. Survivin–a universal tumor antigen. Histol Histopathol. 2002;17:669–675. doi:10.14670/HH-17.669.
  • Shima H, Kutomi G, Satomi F, Imamura M, Kimura Y, Mizuguchi T, Watanabe K, Takahashi A, Murai A, Tsukahara T, et al. Case report: long-term survival of a pancreatic cancer patient immunized with an SVN-2B peptide vaccine. Cancer Immunol Immunother. 2018. doi:10.1007/s00262-018-2217-x.
  • Chiang CY, Chen YJ, Wu CC, Liu SJ, Leng CH, Chen HW. Efficient uptake of recombinant lipidated survivin by antigen-presenting cells initiates antigen cross-presentation and antitumor immunity. Front Immunol. 2018;9:822. doi:10.3389/fimmu.2018.00822.
  • Berzofsky JA, Terabe M, Trepel JB, Pastan I, Stroncek DF, Morris JC, Wood LV. Cancer vaccine strategies: translation from mice to human clinical trials. Cancer Immunol Immunother. 2017. doi:10.1007/s00262-017-2084-x.
  • Lowenfeld L, Zaheer S, Oechsle C, Fracol M, Datta J, Xu S, Fitzpatrick E, Roses RE, Fisher CS, McDonald ES, et al. Addition of anti-estrogen therapy to anti-HER2 dendritic cell vaccination improves regional nodal immune response and pathologic complete response rate in patients with ER(pos)/HER2(pos) early breast cancer. Oncoimmunology. 2017;6:e1207032. doi:10.1080/2162402X.2016.1207032.
  • Cui N, Shi J, Yang C. HER2-Based Immunotherapy for Breast Cancer. Cancer Biother Radiopharm. 2018;33:169–175. doi:10.1089/cbr.2017.2327.
  • Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, Decock JNY-ESO-1. Based Immunotherapy of Cancer: current Perspectives. Front Immunol. 2018;9:947. doi:10.3389/fimmu.2018.00947.
  • Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer. 2005;5:615–625. doi:10.1038/nrc1669.
  • Zeng G, Wang X, Robbins PF, Rosenberg SA, Wang RF. CD4(+) T cell recognition of MHC class II-restricted epitopes from NY-ESO-1 presented by a prevalent HLA DP4 allele: association with NY-ESO-1 antibody production. Proc Natl Acad Sci U S A. 2001;98:3964–3969. doi:10.1073/pnas.061507398.
  • Wang Y, Zhang J, Wu Y, Zy D, Xm L, Liu J, Zhong WN, Deng GH, Xia XY, Deng YT, et al. Mannan-modified adenovirus targeting TERT and VEGFR-2: A universal tumour vaccine. Sci Rep. 2015;5:11275. doi:10.1038/srep11275.
  • Lilleby W, Gaudernack G, Brunsvig PF, Vlatkovic L, Schulz M, Mills K, Hole KH, Inderberg EM. Phase I/IIa clinical trial of a novel hTERT peptide vaccine in men with metastatic hormone-naive prostate cancer. Cancer Immunol Immunother. 2017;66:891–901. doi:10.1007/s00262-017-1994-y.
  • Kim H, Seo EH, Lee SH, Kim BJ. The telomerase-derived anticancer peptide vaccine GV1001 as an extracellular heat shock protein-mediated cell-penetrating peptide. Int J Mol Sci. 2016;17. doi:10.3390/ijms17122054.
  • Fenoglio D, Parodi A, Lavieri R, Kalli F, Ferrera F, Tagliamacco A, Guastalla A, Lamperti MG, Giacomini M, Filaci G. Immunogenicity of GX301 cancer vaccine: four (telomerase peptides) are better than one. Hum Vaccin Immunother. 2015;11:838–850. doi:10.1080/21645515.2015.1012032.
  • Lee YK, Nata’atmaja BS, Kim BH, Pak CS, Heo CY. Protective effect of telomerase-based 16-mer peptide vaccine (GV1001) on inferior epigastric island skin flap survivability in ischaemia-reperfusion injury rat model. J Plast Surg Hand Surg. 2017;51:210–216. doi:10.1080/2000656X.2016.1235046.
  • Kailashiya C, Sharma HB, Kailashiya J. Telomerase based anticancer immunotherapy and vaccines approaches. Vaccine. 2017;35:5768–5775. doi:10.1016/j.vaccine.2017.09.011.
  • Shinde R, Shimoda M, Chaudhary K, Liu H, Mohamed E, Bradley J, Kandala S, Li X, Liu K, McGaha TL, et al. IDO1 Regulates Humoral Immunity to T Cell-Independent Antigens. J Immunol. 2015;195:2374–2382. doi:10.4049/jimmunol.1402854.
  • Muller S, Agnihotri S, Shoger KE, Myers MI, Smith N, Chaparala S, Villanueva CR, Chattopadhyay A, Lee AV, Butterfield LH, et al. Peptide vaccine immunotherapy biomarkers and response patterns in pediatric gliomas. JCI Insight. 2018:3. doi:10.1172/jci.insight.98791.
  • Zhai L, Spranger S, Binder DC, Gritsina G, Lauing KL, Giles FJ, Wainwright DA. Molecular pathways: targeting IDO1 and other tryptophan dioxygenases for cancer immunotherapy. Clin Cancer Res. 2015;21:5427–5433. doi:10.1158/1078-0432.CCR-15-0420.
  • Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ. Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res. 2017;77:6795–6811. doi:10.1158/0008-5472.CAN-17-2285.
  • Mitchell P, Thatcher N, Socinski MA, Wasilewska-Tesluk E, Horwood K, Szczesna A, Martin C, Ragulin Y, Zukin M, Helwig C, et al. Tecemotide in unresectable stage III non-small-cell lung cancer in the phase III START study: updated overall survival and biomarker analyses. Ann Oncol. 2015;26:1134–1142. doi:10.1093/annonc/mdv104.
  • Kruit WH, Suciu S, Dreno B, Mortier L, Robert C, Chiarion-Sileni V, Maio M, Testori A, Dorval T, Grob JJ, et al. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European organisation for research and treatment of cancer melanoma group in metastatic melanoma. J Clin Oncol. 2013;31:2413–2420. doi:10.1200/JCO.2012.43.7111.
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial watch: monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi:10.4161/onci.1.1.17938.
  • Seton-Rogers S. Immunotherapy: switching off immune suppression. Nat Rev Cancer. 2017;17:1. doi:10.1038/nrc.2016.144.
  • Martins I, Galluzzi L, Kroemer G. Hormesis, cell death and aging. Aging (Albany NY). 2011;3:821–828. doi:10.18632/aging.100380.
  • Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M. Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother. 2014;10:3175–3178. doi:10.4161/hv.29418.
  • Garg AD, Agostinis P. Cell death and immunity in cancer: from danger signals to mimicry of pathogen defense responses. Immunol Rev. 2017;280:126–148. doi:10.1111/imr.12574.
  • Garg AD, Galluzzi L, Apetoh L, Baert T, Birge RB, Bravo-San Pedro JM, Breckpot K, Brough D, Chaurio R, Cirone M, et al. Molecular and translational classifications of DAMPs in immunogenic cell death. Front Immunol. 2015;6:588. doi:10.3389/fimmu.2015.00588.
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570. doi:10.1126/science.1203486.
  • Gross ET, Han S, Vemu P, Peinado CD, Marsala M, Ellies LG, Bui JD. Immunosurveillance and immunoediting in MMTV-PyMT-induced mammary oncogenesis. Oncoimmunology. 2017;6:e1268310. doi:10.1080/2162402X.2016.1268310.
  • Lu L, Barbi J, Pan F. The regulation of immune tolerance by FOXP3. Nat Rev Immunol. 2017;17:703–717. doi:10.1038/nri.2017.75.
  • Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486–499. doi:10.1038/nri3862.
  • Carter CA, Oronsky BT, Roswarski J, Oronsky AL, Oronsky N, Scicinski J, Lybeck H, Kim MM, Lybeck M, Reid TR. No patient left behind: the promise of immune priming with epigenetic agents. Oncoimmunology. 2017;6:e1315486. doi:10.1080/2162402X.2017.1315486.
  • Bommareddy PK, Shettigar M, Kaufman HL. Integrating oncolytic viruses in combination cancer immunotherapy. Nat Rev Immunol. 2018. doi:10.1038/s41577-018-0014-6.
  • Van Der Burg SH, Arens R, Ossendorp F, Van Hall T, Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer. 2016;16:219–233. doi:10.1038/nrc.2016.16.
  • Hirayama M, Nishimura Y. The present status and future prospects of peptide-based cancer vaccines. Int Immunol. 2016;28:319–328. doi:10.1093/intimm/dxw027.
  • Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2018;18:168–182. doi:10.1038/nri.2017.131.
  • Finn OJ. The dawn of vaccines for cancer prevention. Nat Rev Immunol. 2018;18:183–194. doi:10.1038/nri.2017.140.
  • Guo Y, Lei K, Tang L. Neoantigen vaccine delivery for personalized anticancer immunotherapy. Front Immunol. 2018;9:1499. doi:10.3389/fimmu.2018.01499.