Phase Ib evaluation of a self-adjuvanted protamine formulated mRNA-based active cancer immunotherapy, BI1361849 (CV9202), combined with local radiation treatment in patients with stage IV non-small cell lung cancer

Alexandros Papachristofilou, Madeleine M Hipp, Ute Klinkhardt, Martin Früh, Martin Sebastian, Christian Weiss, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geißler, Frank Griesinger, Karl-Josef Kallen, Mariola Fotin-Mleczek, Andreas Schröder, Birgit Scheel, Anke Muth, Tobias Seibel, Claudia Stosnach, Fatma Doener, Henoch S Hong, Sven D Koch, Ulrike Gnad-Vogt, Alfred Zippelius, Alexandros Papachristofilou, Madeleine M Hipp, Ute Klinkhardt, Martin Früh, Martin Sebastian, Christian Weiss, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geißler, Frank Griesinger, Karl-Josef Kallen, Mariola Fotin-Mleczek, Andreas Schröder, Birgit Scheel, Anke Muth, Tobias Seibel, Claudia Stosnach, Fatma Doener, Henoch S Hong, Sven D Koch, Ulrike Gnad-Vogt, Alfred Zippelius

Abstract

Background: Preclinical studies demonstrate synergism between cancer immunotherapy and local radiation, enhancing anti-tumor effects and promoting immune responses. BI1361849 (CV9202) is an active cancer immunotherapeutic comprising protamine-formulated, sequence-optimized mRNA encoding six non-small cell lung cancer (NSCLC)-associated antigens (NY-ESO-1, MAGE-C1, MAGE-C2, survivin, 5T4, and MUC-1), intended to induce targeted immune responses.

Methods: We describe a phase Ib clinical trial evaluating treatment with BI1361849 combined with local radiation in 26 stage IV NSCLC patients with partial response (PR)/stable disease (SD) after standard first-line therapy. Patients were stratified into three strata (1: non-squamous NSCLC, no epidermal growth factor receptor (EGFR) mutation, PR/SD after ≥4 cycles of platinum- and pemetrexed-based treatment [n = 16]; 2: squamous NSCLC, PR/SD after ≥4 cycles of platinum-based and non-platinum compound treatment [n = 8]; 3: non-squamous NSCLC, EGFR mutation, PR/SD after ≥3 and ≤ 6 months EGFR-tyrosine kinase inhibitor (TKI) treatment [n = 2]). Patients received intradermal BI1361849, local radiation (4 × 5 Gy), then BI1361849 until disease progression. Strata 1 and 3 also had maintenance pemetrexed or continued EGFR-TKI therapy, respectively. The primary endpoint was evaluation of safety; secondary objectives included assessment of clinical efficacy (every 6 weeks during treatment) and of immune response (on Days 1 [baseline], 19 and 61).

Results: Study treatment was well tolerated; injection site reactions and flu-like symptoms were the most common BI1361849-related adverse events. Three patients had grade 3 BI1361849-related adverse events (fatigue, pyrexia); there was one grade 3 radiation-related event (dysphagia). In comparison to baseline, immunomonitoring revealed increased BI1361849 antigen-specific immune responses in the majority of patients (84%), whereby antigen-specific antibody levels were increased in 80% and functional T cells in 40% of patients, and involvement of multiple antigen specificities was evident in 52% of patients. One patient had a partial response in combination with pemetrexed maintenance, and 46.2% achieved stable disease as best overall response. Best overall response was SD in 57.7% for target lesions.

Conclusion: The results support further investigation of mRNA-based immunotherapy in NSCLC including combinations with immune checkpoint inhibitors.

Trial registration: ClinicalTrials.gov identifier: NCT01915524 .

Keywords: BI1361849; CV9202; Clinical trial; Hypofractionated radiotherapy; Immunomonitoring; Non-small cell lung cancer; mRNA active cancer immunotherapy.

Conflict of interest statement

Ethics approval and consent to participate

This study was approved by local ethics committees. The names and reference numbers are provided in Additional file 11: Table S7.

All patients provided written informed consent before receiving any study-related procedures.

Consent for publication

Not applicable

Competing interests

AP declares non-financial support from CureVac during the conduct of the study. MS declares personal fees from Boehringer Ingelheim, BMS, MSD, Lilly, Pfizer, Astra Zeneca, Roche, Celgen, and Novartis and non-financial support from Boehringer Ingelheim, BMS, and Novartis outside the submitted work. MP declares personal fees from Astra Zeneca, BMS, Boehringer Ingelheim, Merck, Novartis, Pfizer, and Roche outside the submitted work. FG declares grants and personal fees from Astra Zeneca, Boehringer Ingelheim, Pfizer, Lilly, BMS, MSD, Novartis, Celgene, and Roche and personal fees from Ariad, Takeda, and Chugai outside the submitted work. AZ declares study fees from CureVac during the conduct of the study and research funding from BeyondSprings, Secarna, and Roche outside the submitted work. UK, MFM, BS, CS, AM, TS, FD and UG-V are employees of CureVac. AS, MMH, HSH and SDK were employees of CureVac during the conduct of this study. K-JK was a consultant to CureVac until 2015 and was an employee at CureVac until 2012. K-JK, MFM and UG-V jointly hold patent WO/2015/024666. MF, CW, RC, WH, GP, TW, JA, HB, and MG have no conflicts of interest to declare.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
a Study design. b Patient disposition
Fig. 2
Fig. 2
Frequencies of patients with an at least two-fold increase in antigen-specific immune responses following BI1361849 immunotherapy combined with local radiation treatment. Values displayed above the bars indicate the percentages and actual number of patients with increase in immune responses. a Summary graph showing frequencies of patients with antigen-specific T cells, antibodies or both exhibiting an at least two-fold increase compared to baseline against one or more antigens encoded by BI1361849 (any post-vaccine time point). CD4 = antigen-specific CD4+ T cells, CD8 = antigen-specific CD8+ T cells. b Frequencies of patients with an at least a two-fold increase in immune responses compared to baseline to each of the antigens encoded by BI1361849 shown as percentage of all evaluable patients; any post-vaccine time point. c Frequencies of patients with antigen-specific T cells, antibodies or both showing at least a two-fold increase compared to baseline against multiple antigens encoded by BI1361849 shown as percentage of all evaluable patients; at any post-vaccine time point
Fig. 3
Fig. 3
a Vaccine antigen-specific CD4+ and CD8+ T cells detected by ICS at baseline (n = 7 patients for panel CD4, n = 11 patients for panel CD8, n = 16 patients for panel CD4 and/or CD8), and days 19 (n = 13 patients for panel CD4, n = 15 patients for panel CD8, n = 23 patients for panel CD4 and/or CD8) and 61 (n = 7 patients for panel CD4, n = 7 patients for panel CD8, n = 10 patients for panel CD4 and/or CD8) following BI1361849 immunotherapy combined with local radiation treatment. Vaccine antigen-specific T cell responses were determined and are shown as percentages of all possible responses. The denominator for calculating the percentage was defined as the maximum of all possible at least two-fold increases over background (unstimulated cells). Example: 4 functions (IFN-γ, TNF-α, IL-2, CD107a) * 2 T cell subsets (CD4+ or CD8+ T cells) * 6 BI1361849 antigens * 25 patients = 1200 possible responses. The numbers in the key show fold-increases over the background control. b & c Magnitude of T cell responses as measured by b ICS or c IFN-γ ELISpot of patients with an at least two-fold increase in antigen-specific immune responses following BI1361849 treatment. Values are shown as the percentage of positive cells gated on both CD4+ or CD8+ populations for ICS. ELISpot results are plotted as number of spots per 1 million PBMCs after background subtraction (PBMCs that received only cell culture medium). Legends indicate patient ID, antigen, measured cytokine (only for ICS) and time point
Fig. 4
Fig. 4
Efficacy outcomes following BI1361849 immunotherapy combined with local radiation treatment. a Change in target lesion sum of longest diameters (SLD) from baseline. b Survival, progression-free survival, and response kinetics of individual patients (post hoc swimmer plot)

References

    1. Socola F, Scherfenberg N, Raez LE. Therapeutic vaccines in non-small cell lung cancer. Immunotargets Ther. 2013;2:115–124.
    1. Butts C, Socinski MA, Mitchell PL, Thatcher N, Havel L, Krzakowski M, 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.
    1. Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, 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.
    1. Berraondo P, Umansky V, Melero I. Changing the tumor microenvironment: new strategies for immunotherapy. Cancer Res. 2012;72:5159–5164. doi: 10.1158/0008-5472.CAN-12-1952.
    1. Hoerr I, Obst R, Rammensee HG, Jung G. In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies. Eur J Immunol. 2000;30:1–7. doi: 10.1002/1521-4141(200001)30:1<1::AID-IMMU1>;2-#.
    1. Kübler H, Scheel B, Gnad-Vogt U, Miller K, Schultze-Seemann W, Vom Dorp F, et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study. J Immunother Cancer. 2015;3:26. doi: 10.1186/s40425-015-0068-y.
    1. Sebastian M, Papachristofilou A, Weiss C, Früh M, Cathomas R, Hilbe W, et al. Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive®) combined with local radiation as consolidation and maintenance treatment for patients with stage IV non-small cell lung cancer. BMC Cancer. 2014;14:748. doi: 10.1186/1471-2407-14-748.
    1. Hong HS, Koch SD, Scheel B, Gnad-Vogt U, Schröder A, Kallen K-J, et al. Distinct transcriptional changes in non-small cell lung cancer patients associated with multi-antigenic RNActive® CV9201 immunotherapy. Oncoimmunology. 2016;5:e1249560. doi: 10.1080/2162402X.2016.1249560.
    1. Kallen K-J, Heidenreich R, Schnee M, Petsch B, Schlake T, Thess A, et al. A novel, disruptive vaccination technology: self-adjuvanted RNActive(®) vaccines. Hum Vaccin Immunother. 2013;9:2263–2276. doi: 10.4161/hv.25181.
    1. Sebastian M, Schröder A, Scheel B, Hong HS, von Boehmer L, Zippelius A, et al. Active cancer immunotherapy with mRNA-based CV9201 induces antigen-specific T- and B-cell responses in patients with stage IIIb/IV non-small cell lung cancer. Cancer Immunol Immunother. 2019; In press.
    1. Quoix E, Lena H, Losonczy G, Forget F, Chouaid C, Papai Z, et al. TG4010 immunotherapy and first-line chemotherapy for advanced non-small-cell lung cancer (TIME): results from the phase 2b part of a randomised, double-blind, placebo-controlled, phase 2b/3 trial. Lancet Oncol. 2016;17:212–223. doi: 10.1016/S1470-2045(15)00483-0.
    1. Cancer Genome Atlas Research Network Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489:519–525. doi: 10.1038/nature11404.
    1. Tang H, Xiao G, Behrens C, Schiller J, Allen J, Chow C-W, et al. A 12-gene set predicts survival benefits from adjuvant chemotherapy in non-small cell lung cancer patients. Clin Cancer Res. 2013;19:1577–1586. doi: 10.1158/1078-0432.CCR-12-2321.
    1. Wersäll PJ, Blomgren H, Pisa P, Lax I, Kälkner K-M, Svedman C. Regression of non-irradiated metastases after extracranial stereotactic radiotherapy in metastatic renal cell carcinoma. Acta Oncol. 2006;45:493–497. doi: 10.1080/02841860600604611.
    1. Formenti SC, Demaria S. Radiation therapy to convert the tumor into an in situ vaccine. Int J Radiat Oncol Biol Phys. 2012;84:879–880. doi: 10.1016/j.ijrobp.2012.06.020.
    1. Demaria S, Golden EB, Formenti SC. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol. 2015;1:1325–1332. doi: 10.1001/jamaoncol.2015.2756.
    1. Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. 2004;64:4328–4337. doi: 10.1158/0008-5472.CAN-04-0073.
    1. Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15:5379–5388. doi: 10.1158/1078-0432.CCR-09-0265.
    1. Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol. 2010;28:4324–4332. doi: 10.1200/JCO.2010.28.9793.
    1. Fotin-Mleczek M, Zanzinger K, Heidenreich R, Lorenz C, Kowalczyk A, Kallen K-J, et al. mRNA-based vaccines synergize with radiation therapy to eradicate established tumors. Radiat Oncol. 2014;9:180. doi: 10.1186/1748-717X-9-180.
    1. Basler L, Kowalczyk A, Heidenreich R, Fotin-Mleczek M, Tsitsekidis S, Zips D, et al. Abscopal effects of radiotherapy and combined mRNA-based immunotherapy in a syngeneic, OVA-expressing thymoma mouse model. Cancer Immunol Immunother. 2018;67:653–662. doi: 10.1007/s00262-018-2117-0.
    1. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366:925–931. doi: 10.1056/NEJMoa1112824.
    1. Golden EB, Demaria S, Schiff PB, Chachoua A, Formenti SC. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol Res. 2013;1:365–372. doi: 10.1158/2326-6066.CIR-13-0115.
    1. Daly ME, Monjazeb AM, Kelly K. Clinical trials integrating immunotherapy and radiation for non-small-cell lung cancer. J Thorac Oncol. 2015;10:1685–1693. doi: 10.1097/JTO.0000000000000686.
    1. Rodríguez-Ruiz ME, Vanpouille-Box C, Melero I, Formenti SC, Demaria S. Immunological mechanisms responsible for radiation-induced Abscopal effect. Trends Immunol. 2018;39:644–655. doi: 10.1016/j.it.2018.06.001.
    1. Theelen W, Peulen H, Lalezari F, de Vries J, De Langen J, Aerts J, et al. Randomized phase II study of pembrolizumab after stereotactic body radiotherapy (SBRT) versus pembrolizumab alone in patients with advanced non-small cell lung cancer: The PEMBRO-RT study. J Clin Oncol. 2018;36(Suppl):Abstract 9023. doi: 10.1200/JCO.2018.36.15_suppl.9023.
    1. Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys. 2012;83:1306–1310. doi: 10.1016/j.ijrobp.2011.09.049.
    1. Kudo-Saito C, Schlom J, Camphausen K, Coleman CN, Hodge JW. The requirement of multimodal therapy (vaccine, local tumor radiation, and reduction of suppressor cells) to eliminate established tumors. Clin Cancer Res. 2005;11:4533–4544. doi: 10.1158/1078-0432.CCR-04-2237.
    1. Budach W, Belka C. Palliative percutaneous radiotherapy in non-small-cell lung cancer. Lung Cancer. 2004;45(Suppl 2):S239–S245. doi: 10.1016/j.lungcan.2004.07.969.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Hodge JW, Sharp HJ, Gameiro SR. Abscopal regression of antigen disparate tumors by antigen cascade after systemic tumor vaccination in combination with local tumor radiation. Cancer Biother Radiopharm. 2012;27:12–22. doi: 10.1089/cbr.2012.1202.
    1. Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer. 2016;4:51. doi: 10.1186/s40425-016-0156-7.
    1. Hazama S, Nakamura Y, Takenouchi H, Suzuki N, Tsunedomi R, Inoue Y, et al. A phase I study of combination vaccine treatment of five therapeutic epitope-peptides for metastatic colorectal cancer; safety, immunological response, and clinical outcome. J Transl Med. 2014;12:63. doi: 10.1186/1479-5876-12-63.
    1. Neninger Vinageras E, de la Torre A, Osorio Rodríguez M, Catalá Ferrer M, Bravo I, Mendoza del Pino M, et al. Phase II randomized controlled trial of an epidermal growth factor vaccine in advanced non-small-cell lung cancer. J Clin Oncol. 2008;26:1452–1458. doi: 10.1200/JCO.2007.11.5980.
    1. Yoshitake Y, Fukuma D, Yuno A, Hirayama M, Nakayama H, Tanaka T, et al. Phase II clinical trial of multiple peptide vaccination for advanced head and neck cancer patients revealed induction of immune responses and improved OS. Clin Cancer Res. 2015;21:312–321. doi: 10.1158/1078-0432.CCR-14-0202.
    1. Wherry EJ, Teichgräber V, Becker TC, Masopust D, Kaech SM, Antia R, et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol. 2003;4:225–234. doi: 10.1038/ni889.
    1. Chudley L, McCann KJ, Coleman A, Cazaly AM, Bidmon N, Britten CM, et al. Harmonisation of short-term in vitro culture for the expansion of antigen-specific CD8(+) T cells with detection by ELISPOT and HLA-multimer staining. Cancer Immunol Immunother. 2014;63:1199–1211. doi: 10.1007/s00262-014-1593-0.
    1. Wattenberg MM, Kwilas AR, Gameiro SR, Dicker AP, Hodge JW. Expanding the use of monoclonal antibody therapy of cancer by using ionising radiation to upregulate antibody targets. Br J Cancer. 2014;110:1472–1480. doi: 10.1038/bjc.2014.79.
    1. Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–1725. doi: 10.1084/jem.20082492.
    1. Sahin U, Derhovanessian E, Miller M, Kloke B-P, Simon P, Löwer M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–226. doi: 10.1038/nature23003.
    1. Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–221. doi: 10.1038/nature22991.
    1. Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390:1511–1520. doi: 10.1016/S0140-6736(17)31665-3.
    1. 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.
    1. Lu Y-C, Robbins PF. Targeting neoantigens for cancer immunotherapy. Int Immunol. 2016;28:365–370. doi: 10.1093/intimm/dxw026.
    1. Tran E, Robbins PF, Rosenberg SA. “Final common pathway” of human cancer immunotherapy: targeting random somatic mutations. Nat Immunol. 2017;18:255–262. doi: 10.1038/ni.3682.
    1. Yarchoan M, Johnson BA, 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.

Source: PubMed

3
Abonnieren