Randomized phase II trial of autologous dendritic cell vaccines versus autologous tumor cell vaccines in metastatic melanoma: 5-year follow up and additional analyses

Robert O Dillman, Andrew N Cornforth, Gabriel I Nistor, Edward F McClay, Thomas T Amatruda, Carol Depriest, Robert O Dillman, Andrew N Cornforth, Gabriel I Nistor, Edward F McClay, Thomas T Amatruda, Carol Depriest

Abstract

Background: Despite improved survival following checkpoint inhibitors, there is still a potential role for anti-cancer therapeutic vaccines. Because of biological heterogeneity and neoantigens resulting from each patient's mutanome, autologous tumor may be the best source of tumor-associated antigens (TAA) for vaccines. Ex vivo loading of autologous dendritic cells with TAA may be associated with superior clinical outcome compared to injecting irradiated autologous tumor cells. We conducted a randomized phase II trial to compare autologous tumor cell vaccines (TCV) and autologous dendritic cell vaccines (DCV) loaded with autologous TAA.

Methods: Short-term autologous tumor cell lines were established from metastatic tumor. Vaccines were admixed with 500 micrograms of GM-CSF and injected weekly for 3 weeks, then at weeks 8, 12,16, 20, and 24. The primary endpoint was overall survival. Secondary objectives were identification of adverse events, and results of delayed type hypersensitivity (DTH) reactions to intradermal tumor cell injections.

Results: Forty-two patients were randomized. All were followed from randomization until death or for five years; none were lost to follow-up. DCV was associated with longer survival: median 43.4 versus 20.5 months (95% CI, 18.6 to > 60 versus 9.3 to 32.3 months) and a 70% reduction in the risk of death (hazard ratio = 0.304, p = 0.0053, 95% CI, 0.131 to 0.702). Tumor DTH reactions were neither prognostic nor predictive. The most common treatment-related adverse events were mild to moderate local injection site reactions and flu-like symptoms; but grade 2 treatment-related adverse events were more frequent with TCV. Serum marker analyses at week-0 and week-4 showed that serum markers were similar at baseline in each arm, but differed after vaccination.

Conclusions: This is the only human clinical trial comparing DCV and TCV as platforms for autologous TAA presentation. DCV was associated with minimal toxicity and long-term survival in patients with metastatic melanoma. DTH to autologous tumor cells was neither prognostic for survival nor predictive of benefit for either vaccine.

Trial registration: Clinical trials.gov NCT00948480 retrospectively registered 28 July 2009.

Keywords: Autologous tumor cell lines; Dendritic-cell vaccines; Metastatic melanoma; Patient-specific vaccines; Tumor-cell vaccines.

Conflict of interest statement

Authors’ information

ROD is a former president of the Society for Immunotherapy of Cancer (2000–2002, then called the International Society for the Biotherapy of Cancer) after serving two previous terms on its Board of Directors. For more than a decade he was Chairman of the Cancer Biotherapy Research Group. From 1989 to 2011 he was Medical Director of the Hoag Cancer Center, where this trial was conducted, and Scientific Director for the Hoag Cell Biology Laboratory, where the investigational products used in this trial were manufactured.

Ethics approval and consent to participate

The MACVAC clinical trial was conducted per the Declaration of Helsinki after approval by the Western Institutional Review Board (Seattle, WA), and in accord with assurances filed with and approved by the Department of Health and Human Services. All patients gave written informed consent prior to randomization. In addition to Institutional Review Board oversight, conduct of the trial was monitored by an external consultant, and Hoag Hospital’s institutional clinical research oversight committee. The protocol and manufacturing procedures were reviewed by the US Food and Drug Administration in association with BB-IND 5838 for the tumor cell vaccine, 8554 for the dendritic cell vaccine. The trial was registered on ClinicalTrials.gov (NCT00436930) on 28 July 2009.

Consent for publication

Not applicable

Competing interests

ROD is an employ of AIVITA Biomedical, Inc. ANC is an employee of TCR2. GIN is an employ of AVITIA Biomedical, Inc. ROD, ANC, and GIN are former employees of Caladrius Biosciences, Inc. and retain stock in that company. None of the other authors declared any conflicts as related to this trial.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
a Overall Survival by treatment arm. Median OS was 43.4 months versus 20.5 months for DCV and TCV respectively (18.6 to > 60 vs 9.3 to 32.3 months, 95% CI) (p = 0.194 Mantel-Haenzsel; p = 0.088 Gehan’s Wilcoxon). Adjusted Cox proportional hazard model revealed a 70% reduction in risk of death in the DCV arm (HR = 0.304, 95% CI 0.131 to 0.702, p = 0.0053, Wald test). Variables in multivariate analysis included age, stage, LDH, performance status, gender, M1 category, whether patient had measurable disease, treatment with dendritic cell vaccine, and whether patient lived outside California (see Additional file 4: Table S3. b Progression free survival (PFS) by treatment arm. Median PFS was 5.4 months in the DCV arm and 3.7 months in the TCV arm (4.0 to 8.0 vs 1.0 to 5.0 months, 95% CI p = 0.498)
Fig. 2
Fig. 2
a Baseline analysis of serum cytokines. In order to summarize data for all tests, data is expressed as change in relation to values for set of assays compared to 3 normal control volunteers. There was substantial variation in markers among patients. At baseline most markers were elevated compared to normals, especially in the DCV arm. b The post treatment analysis of cytokines one week after the third weekly vaccine injection, showing changes compared to baseline. The changes associated with the two vaccines were quite different. Levels increased for nearly all markers in the TCV arm, but decreased for eight of the 17 groupings in the DCV arm

References

    1. Ugurel S, Röhmel J, Ascierto PA, Flaherty KT, Grob JJ, Hasuchild A, et al. Survival of patients with advanced metastatic melanoma: the impact of novel therapies. Eur J Cancer. 2016;53:125–134. doi: 10.1016/j.ejca.2015.09.013.
    1. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, et al. Combined vemurfenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;13(37):1867–1876. doi: 10.1056/NEJMoa1408868.
    1. Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroiakovski D, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30–39. doi: 10.1056/NEJMoa1412690.
    1. Schadendorf D, Hodi S, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 2015;33(17):1889–1894. doi: 10.1200/JCO.2014.56.2736.
    1. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–2532. doi: 10.1056/NEJMoa1503093.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34. doi: 10.1056/NEJMoa1504030.
    1. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–1356. doi: 10.1056/NEJMoa1709684.
    1. 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(3):355–366. doi: 10.1084/jem.190.3.355.
    1. Li B, VanRoey M, Wang C, Chen TH, Korman A, Jooss K. Anti-programmed death-1 synergies with granulocytes macrophage colony-stimulating factor-secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors. Clin Cancer Res. 2009;15(5):1623–1634. doi: 10.1158/1078-0432.CCR-08-1825.
    1. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10. doi: 10.1016/j.immuni.2013.07.012.
    1. Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6(8):827–837. doi: 10.1158/-15-1545.
    1. Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33(25):2780–2788. doi: 10.1200/JCO.2014.58.3377.
    1. de Rosa F, Ridolfi L, Fiammenghi L, Petrini M, Granato AM, Ancarani V, et al. Dendritic cell vaccination for metastatic melanoma: a 14 year monoinstitutional experience. Melanoma Res. 2017;27(4):351–357. doi: 10.1097/CMR.0000000000000356.
    1. Carreno BM, Magrini V, Becker-Hapak, Kaabinejadian S, Hundal J, Petti AA, et al. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science. 2015;348(6236):803–808. doi: 10.1126/science.aaa3828.
    1. Dillman RO. An update on the relevance of vaccine research for the treatment of metastatic melanoma. Melanoma Manage. 2017;4:203–15.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74. doi: 10.1126/science.aaa4971.
    1. Türeci Ö, Vormehr M, Diken M, Kreiter S, Huber C, Sahin U. Mutanome directed cancer immunotherapy. Curr Opin Immunol. 2016;39:14–22. doi: 10.1016/j.coi.2015.12.001.
    1. Dillman RO, Nayak SK, Beutel L. Establishing in vitro cultures of autologous tumor cells for use in active specific immunotherapy. J Immunother Emphasis Tumor Immunol. 1993;14(1):65–69. doi: 10.1097/00002371-199307000-00009.
    1. Dillman RO, Cornforth AN, Nistor G. Cancer stem cell antigen-based vaccines: the preferred strategy for active specific immunotherapy of metastatic melanoma? Expert Opin Biol Ther. 2013;13(5):643–656. doi: 10.1517/14712598.2013.759556.
    1. Dillman RO, DePriest C, DeLeon C, Barth NM, Schwartzberg LS, Beutel LD, et al. Patient-specific vaccines derived from autologous tumor cell lines as active specific immunotherapy: results of exploratory phase I/II trials in patients with metastatic melanoma. Cancer Biother Radiopharm. 2007;22(3):309–321. doi: 10.1089/cbr.2007.345.
    1. Dillman RO, Selvan SR, Schiltz PM, McClay EF, Barth NM, Depriest C, et al. Phase II trial of dendritic cells loaded with antigens from self-renewing, proliferating autologous tumor cells as patient-specific anti-tumor vaccines in patients with metastatic melanoma: final report. Cancer Biother Radiopharm. 2009;24(3):311–319. doi: 10.1089/cbr.2008.0599.
    1. Dillman RO, Cornforth AN, Depriest C, McClay EF, Amatruda TT, de Leon C, et al. Tumor stem cell antigens as consolidative active specific immunotherapy: a randomized phase II trial of dendritic cells versus tumor cells in patients with metastatic melanoma. J Immunother. 2012;35(8):641–649. doi: 10.1097/CJI.0b013e31826f79c8.
    1. Selvan SR, Carbonell DJ, Fowler AW, Beatty AR, Ravindranath MH, Dillman RO. Establishment of stable cell lines for personalized melanoma cell vaccine. Melanoma Res. 2010;20(4):280–292. doi: 10.1097/CMR.0b013e3283390696.
    1. Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27(36):6199–6206. doi: 10.1200/JCO.2009.23.4799.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92(3):205–216. doi: 10.1093/jnci/92.3.205.
    1. Dillman RO, Nanci AA, Williams ST, Kim RB, Hafer RL, Coleman CL, et al. Durable complete response of refractory, progressing metastatic melanoma after treatment with a patient-specific vaccine. Cancer Biother Radiopharm. 2010;25(5):553–557. doi: 10.1089/cbr.2010.0819.
    1. Dillman RO. Cancer vaccines: can they improve survival? Cancer Biother Radiopharm. 2015;30(4):147–151. doi: 10.1089/cbr.2014.1805.
    1. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–422. doi: 10.1056/NEJMoa1001294.
    1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466.
    1. Flanigan RC, Polcari AJ, Shore ND, Price TH, Sims RB, Maher JC, et al. An analysis of leukapheresis and central venous catheter use in the randomized, placebo controlled, phase 3 IMPACT trial of sipuleucel-T for metastatic castrate resistant prostate cancer. J Urol. 2013;189(2):521–526. doi: 10.1016/j.juro.2012.09.029.
    1. Berd D, Maguire HC. Ur, McCue P, Mastrangelo MJ. Treatment of metastatic melanoma with an autologous tumor-cell vaccine: clinical and immunologic results in 64 patients. J Clin Oncol. 1990;8(11):1858–1867. doi: 10.1200/JCO.1990.8.11.1858.
    1. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, et al. Vaccination of melanoma patients with peptide-or tumor lysate-pulsed cells. Nat Med. 1998;4(3):328–332. doi: 10.1038/nm0398-328.
    1. Dillman RO, Beutel LD, Barth NM, de Leon C, O’Connor AA, Depriest C, Nayak SK. Irradiated cells from autologous tumor cell lines as patient-specific vaccine therapy in 125 patients with metastatic cancer: induction of delayed-type hypersensitivity to autologous tumor is associated with improved survival. Cancer Biother Radiopharm. 2002;17(1):51–66. doi: 10.1089/10849780252824073.
    1. Dillman RO, Cornforth AN, Nestor GI. Dendritic cell vaccines for melanoma: past, present, and future. Melanoma Manag. 2016;3:267–283. doi: 10.2217/mmt-2016-0021.
    1. Dillman RO. Is there a role for therapeutic cancer vaccines in the age of checkpoint inhibitors? Human Vaccine Immunother. 2017;13(3):528–532. doi: 10.1080/21645515.2016.1244149.
    1. Dillman RO, Depriest C, McClure SE. High-dose IL-2 in metastatic melanoma: better survival in patients immunized with antigens from autologous tumor cell lines. Cancer Biother Radiopharm. 2014;29(2):53–57. doi: 10.1089/cbr.2013.1565.
    1. Puzanov I, Milhem MM, Minor D, Hamid O, Li A, Chen L, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol. 2016;34:2619–2626. doi: 10.1200/JCO.2016.67.1529.
    1. Vreeland TJ, Clifton GT, Herbert GS, Hale DF, Jackson DO, Berry JS, Peoples GE. Gaining ground on a cure through synergy: combining checkpoint inhibitors with cancer vaccines. Expert Rev Clin Immunol. 2016;12:1347–1357. doi: 10.1080/1744666X.2016.1202114.

Source: PubMed

3
Se inscrever