Phase 1 open-label trial of intravenous administration of MVA-BN-brachyury-TRICOM vaccine in patients with advanced cancer

Peter J DeMaria, Katherine Lee-Wisdom, Renee N Donahue, Ravi A Madan, Fatima Karzai, Angie Schwab, Claudia Palena, Caroline Jochems, Charalampos Floudas, Julius Strauss, Jennifer L Marté, Jason Mark Redman, Eva Dombi, Brigitte Widemann, Borys Korchin, Tatiana Adams, Cesar Pico-Navarro, Christopher Heery, Jeffrey Schlom, James L Gulley, Marijo Bilusic, Peter J DeMaria, Katherine Lee-Wisdom, Renee N Donahue, Ravi A Madan, Fatima Karzai, Angie Schwab, Claudia Palena, Caroline Jochems, Charalampos Floudas, Julius Strauss, Jennifer L Marté, Jason Mark Redman, Eva Dombi, Brigitte Widemann, Borys Korchin, Tatiana Adams, Cesar Pico-Navarro, Christopher Heery, Jeffrey Schlom, James L Gulley, Marijo Bilusic

Abstract

Background: MVA-BN-brachyury-TRICOM is a recombinant vector-based therapeutic cancer vaccine designed to induce an immune response against brachyury. Brachyury, a transcription factor overexpressed in advanced cancers, has been associated with treatment resistance, epithelial-to-mesenchymal transition, and metastatic potential. MVA-BN-brachyury-TRICOM has demonstrated immunogenicity and safety in previous clinical trials of subcutaneously administered vaccine. Preclinical studies have suggested that intravenous administration of therapeutic vaccines can induce superior CD8+ T cell responses, higher levels of systemic cytokine release, and stronger natural killer cell activation and proliferation. This is the first-in-human study of the intravenous administration of MVA-BN-brachyury-TRICOM.

Methods: Between January 2020 and March 2021, 13 patients were treated on a phase 1, open-label, 3+3 design, dose-escalation study at the National Institutes of Health Clinical Center. The study population was adults with advanced solid tumors and was enriched for chordoma, a rare sarcoma of the notochord that overexpresses brachyury. Vaccine was administered intravenously at three DLs on days 1, 22, and 43. Blood samples were taken to assess drug pharmacokinetics and immune activation. Imaging was conducted at baseline, 1 month, and 3 months post-treatment. The primary endpoint was safety and tolerability as determined by the frequency of dose-limiting toxicities; a secondary endpoint was determination of the recommended phase 2 dose.

Results: No dose-limiting toxicities were observed and no serious adverse events were attributed to the vaccine. Vaccine-related toxicities were consistent with class profile (ie, influenza-like symptoms). Cytokine release syndrome up to grade 2 was observed with no adverse outcomes. Dose-effect trend was observed for fever, chills/rigor, and hypotension. Efficacy analysis of objective response rate per RECIST 1.1 at the end of study showed one patient with a partial response, four with stable disease, and eight with progressive disease. Three patients with stable disease experienced clinical benefit in the form of improvement in pain. Immune correlatives showed T cell activation against brachyury and other tumor-associated cascade antigens.

Conclusions: Intravenous administration of MVA-BN-brachyury-TRICOM vaccine was safe and tolerable. Maximum tolerated dose was not reached. The maximum administered dose was 109 infectious units every 3 weeks for three doses. This dose was selected as the recommended phase 2 dose.

Trial registration number: NCT04134312.

Keywords: clinical trials as topic; immunogenicity; immunotherapy; sarcoma; vaccine.

Conflict of interest statement

Competing interests: BK: Bavarian Nordic (E). TA: Bavarian Nordic (E). CP-N: Bavarian Nordic (E). CH: Arcellx (E) (previously employed by Bavarian Nordic). The other authors indicate no financial relationships. (E) Employment.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Consort flow diagram. Of 14 patients officially screened for the trial, only one patient was unable to obtain imaging with MRI to confirm eligibility. The other 13 patients screened were treated (3 at DL1, 4 at DL2 and 6 at DL3). One patient (patient #7) treated at DL2 was replaced for failure to complete the DLT period due to treatment delay associated with the SARS-CoV-2 pandemic. DL, dose level; DLT, dose-limiting toxicity.
Figure 2
Figure 2
Change in vital signs within 48 hours post-vaccination. A transient increase in temperature and decrease in blood pressure were observed with a dose-effect trend. DL, dose level; Inf. U., infectious units.
Figure 3
Figure 3
Development of multifunctional antigen specific CD4+ and CD8+ T cell responses post- (vs pre-) MVA-BN-brachyury-TRICOM vaccine. TAA responses againstbrachyury and the cascade antigens CEA and MUC1 were compared in thirteen patients pre- and post-1, 2, and 3 vaccinations, where sufficient research bloods were available. The absolute number of multifunctional TAA responses (CD4+ or CD8+ T cells expressing two or more of the following: IFN-γ, TNF-α, IL-2, or CD107a) per 1×106 PBMCs plated at the start of the stimulation assay was calculated. Any background signal obtained with the HLA peptide pool was subtracted. Patients were scored as having a >3-fold (or if no cells at pre, >100/1×106 cells at post, +) or >10-fold (or if no cells at pre, >1000/1×106 cells at post, ++) increase in CD4+ or CD8+ multifunctional cells post- (vs pre) vaccine. Gray indicates where an insufficient number of viable PBMCs were recovered for analysis. BOR, best overall response; CEA, carcinoembryonic antigen; MUC, mucin; PD, progressive disease; PR, partial response; PT, patient; SD, stable disease; TAA, tumor-associated antigen.
Figure 4
Figure 4
Patient #9 (responder). Before and after treatment photos and CT imaging for patient #9, a 59-year-old man with metastatic chordoma of the lumbar spine extending through the anterior abdomen. He had previously failed multiple lines of treatment. Following three doses of the investigational intravenous brachyury-targeting vaccine (DL3) given over a 2-month treatment window, the patient’s tumor shrank 33% per RECIST 1.1% and 66% per exploratory volumetric analysis. Left top: clinic photograph from August 14, 2020, prior to first dose of DL3 vaccine. Left bottom: baseline CT from August 14, 2020. Right top: clinic photograph from October 27, 2020, after three vaccine doses. Right bottom: End-of-treatment CT from October 27, 2020. DL, dose level; RECIST, Response Evaluation Criteria in Solid Tumors.

References

    1. Fernando RI, Castillo MD, Litzinger M, et al. . Il-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Res 2011;71:5296–306. 10.1158/0008-5472.CAN-11-0156
    1. Herrmann BG, Labeit S, Poustka A, et al. . Cloning of the T gene required in mesoderm formation in the mouse. Nature 1990;343:617–22. 10.1038/343617a0
    1. Edwards YH, Putt W, Lekoape KM, et al. . The human homolog T of the mouse T(brachyury) gene; gene structure, cDNA sequence, and assignment to chromosome 6q27. Genome Res 1996;6:226–33. 10.1101/gr.6.3.226
    1. Kispert A, Hermann BG. The Brachyury gene encodes a novel DNA binding protein. Embo J 1993;12:4898–9. 10.1002/j.1460-2075.1993.tb06179.x
    1. Kispert A, Koschorz B, Herrmann BG. The T protein encoded by brachyury is a tissue-specific transcription factor. Embo J 1995;14:4763–72. 10.1002/j.1460-2075.1995.tb00158.x
    1. Palena C, Polev DE, Tsang KY, et al. . The human T-box mesodermal transcription factor brachyury is a candidate target for T-cell-mediated cancer immunotherapy. Clin Cancer Res 2007;13:2471–8. 10.1158/1078-0432.CCR-06-2353
    1. Kispert A, Herrmann BG, Leptin M, et al. . Homologs of the mouse brachyury gene are involved in the specification of posterior terminal structures in Drosophila, Tribolium, and Locusta. Genes Dev 1994;8:2137–50. 10.1101/gad.8.18.2137
    1. Wilkinson DG, Bhatt S, Herrmann BG. Expression pattern of the mouse T gene and its role in mesoderm formation. Nature 1990;343:657–9. 10.1038/343657a0
    1. Schulte-Merker S, Smith JC. Mesoderm formation in response to brachyury requires FGF signalling. Curr Biol 1995;5:62–7. 10.1016/S0960-9822(95)00017-0
    1. Yamaguchi TP, Takada S, Yoshikawa Y, et al. . T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification. Genes Dev 1999;13:3185–90. 10.1101/gad.13.24.3185
    1. Roselli M, Fernando RI, Guadagni F, et al. . Brachyury, a driver of the epithelial-mesenchymal transition, is overexpressed in human lung tumors: an opportunity for novel interventions against lung cancer. Clin Cancer Res 2012;18:3868–79. 10.1158/1078-0432.CCR-11-3211
    1. Richardson SM, Ludwinski FE, Gnanalingham KK, et al. . Notochordal and nucleus pulposus marker expression is maintained by sub-populations of adult human nucleus pulposus cells through aging and degeneration. Sci Rep 2017;7:1501. 10.1038/s41598-017-01567-w
    1. Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 2006;7:131–42. 10.1038/nrm1835
    1. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420–8. 10.1172/JCI39104
    1. Gravdal K, Halvorsen OJ, Haukaas SA, et al. . A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clin Cancer Res 2007;13:7003–11. 10.1158/1078-0432.CCR-07-1263
    1. Onder TT, Gupta PB, Mani SA, et al. . Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res 2008;68:3645–54. 10.1158/0008-5472.CAN-07-2938
    1. Fernando RI, Litzinger M, Trono P, et al. . The T-box transcription factor brachyury promotes epithelial-mesenchymal transition in human tumor cells. J Clin Invest 2010;120:533–44. 10.1172/JCI38379
    1. Vujovic S, Henderson S, Presneau N, et al. . Brachyury, a crucial regulator of notochordal development, is a novel biomarker for chordomas. J Pathol 2006;209:157–65. 10.1002/path.1969
    1. Tirabosco R, Mangham DC, Rosenberg AE, et al. . Brachyury expression in extra-axial skeletal and soft tissue chordomas: a marker that distinguishes chordoma from mixed tumor/myoepithelioma/parachordoma in soft tissue. Am J Surg Pathol 2008;32:572–80. 10.1097/PAS.0b013e31815b693a
    1. Yang XR, Ng D, Alcorta DA, et al. . T (brachyury) gene duplication confers major susceptibility to familial chordoma. Nat Genet 2009;41:1176–8. 10.1038/ng.454
    1. Sharifnia T, Wawer MJ, Chen T, et al. . Small-molecule targeting of brachyury transcription factor addiction in chordoma. Nat Med 2019;25:292–300. 10.1038/s41591-018-0312-3
    1. Sheppard HE, Dall'Agnese A, Park WD, et al. . Targeted brachyury degradation disrupts a highly specific autoregulatory program controlling chordoma cell identity. Cell Rep Med 2021;2:100188. 10.1016/j.xcrm.2020.100188
    1. Heery CR, Singh BH, Rauckhorst M, et al. . Phase I trial of a yeast-based therapeutic cancer vaccine (GI-6301) targeting the transcription factor brachyury. Cancer Immunol Res 2015;3:1248–56. 10.1158/2326-6066.CIR-15-0119
    1. DeMaria PJ, Bilusic M, Park DM, et al. . Randomized, double-blind, placebo-controlled phase II study of yeast-brachyury vaccine (GI-6301) in combination with standard-of-care radiotherapy in locally advanced, unresectable chordoma. Oncologist 2021;26:e847–58. 10.1002/onco.13720
    1. Heery CR, Palena C, McMahon S, et al. . Phase I study of a poxviral TRICOM-based vaccine directed against the transcription factor brachyury. Clin Cancer Res 2017;23:6833–45. 10.1158/1078-0432.CCR-17-1087
    1. Collins JM, Donahue RN, Tsai Y-T, et al. . Phase I trial of a modified vaccinia ankara priming vaccine followed by a fowlpox virus boosting vaccine modified to express brachyury and costimulatory molecules in advanced solid tumors. Oncologist 2020;25:560–1006. 10.1634/theoncologist.2019-0932
    1. Hanke T, Blanchard TJ, Schneider J, et al. . Immunogenicities of intravenous and intramuscular administrations of modified vaccinia virus Ankara-based multi-CTL epitope vaccine for human immunodeficiency virus type 1 in mice. J Gen Virol 1998;79:83–90. 10.1099/0022-1317-79-1-83
    1. Shen X, Wong SBJ, Buck CB, et al. . Direct priming and cross-priming contribute differentially to the induction of CD8+ CTL following exposure to vaccinia virus via different routes. J Immunol 2002;169:4222–9. 10.4049/jimmunol.169.8.4222
    1. Lauterbach H, Kassub R, Pätzold J, et al. . Immune requirements of post-exposure immunization with modified vaccinia Ankara of lethally infected mice. PLoS One 2010;5:e9659. 10.1371/journal.pone.0009659
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47. 10.1016/j.ejca.2008.10.026
    1. U.S. Department of Health and Human Services . Common terminology criteria for adverse events (CTCAE) version 5.0. Available: [Accessed 17 May 2021].
    1. Lee DW, Santomasso BD, Locke FL, et al. . ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant 2019;25:625–38. 10.1016/j.bbmt.2018.12.758
    1. Heery CR, Singh BH, Rauckhorst M, et al. . Phase I trial of a yeast-based therapeutic cancer vaccine (GI-6301) targeting the transcription factor brachyury. Cancer Immunol Res 2015;3:1248–56. 10.1158/2326-6066.CIR-15-0119
    1. Donahue RN, Lepone LM, Grenga I, et al. . Analyses of the peripheral immunome following multiple administrations of avelumab, a human IgG1 anti-PD-L1 monoclonal antibody. J Immunother Cancer 2017;5:20. 10.1186/s40425-017-0220-y
    1. Pastor DM, Lee-Wisdom K, Arai AE, et al. . Fast clearance of the SARS-CoV-2 virus in a patient undergoing vaccine immunotherapy for metastatic chordoma: a case report. Front Oncol 2020;10:603248. 10.3389/fonc.2020.603248
    1. Wimmers F, Aarntzen EHJG, Duiveman-deBoer T, et al. . Long-lasting multifunctional CD8+ T cell responses in end-stage melanoma patients can be induced by dendritic cell vaccination. Oncoimmunology 2016;5:e1067745. 10.1080/2162402X.2015.1067745
    1. Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas. With a report of ten original cases. 1893. Clin Orthop Relat Res 1991:3–11.
    1. Dillon PM, Petroni GR, Smolkin ME, 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. 10.1186/s40425-017-0295-5
    1. Melssen MM, Petroni GR, Chianese-Bullock KA, et al. . A multipeptide vaccine plus toll-like receptor agonists LPS or polyICLC in combination with incomplete Freund's adjuvant in melanoma patients. J Immunother Cancer 2019;7:163. 10.1186/s40425-019-0625-x
    1. Engelhard VH, Obeng RC, Cummings KL, et al. . MHC-restricted phosphopeptide antigens: preclinical validation and first-in-humans clinical trial in participants with high-risk melanoma. J Immunother Cancer 2020;8:e000262. 10.1136/jitc-2019-000262
    1. Liu H, Zha Y, Choudhury N, et al. . WT1 peptide vaccine in montanide in contrast to poly ICLC, is able to induce WT1-specific immune response with TCR clonal enrichment in myeloid leukemia. Exp Hematol Oncol 2018;7:1. 10.1186/s40164-018-0093-x
    1. DeMaria PJ, Bilusic M. Cancer vaccines. Hematol Oncol Clin North Am 2019;33:199–214. 10.1016/j.hoc.2018.12.001
    1. Sahin U, Oehm P, Derhovanessian E, et al. . An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 2020;585:107–12. 10.1038/s41586-020-2537-9
    1. Darrah PA, Zeppa JJ, Maiello P, et al. . Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature 2020;577:95–102. 10.1038/s41586-019-1817-8

Source: PubMed

3
订阅