Phase 2 trial of T-cell activation using MVI-816 and pembrolizumab in patients with metastatic, castration-resistant prostate cancer (mCRPC)

Douglas G McNeel, Jens C Eickhoff, Ellen Wargowski, Laura E Johnson, Christos E Kyriakopoulos, Hamid Emamekhoo, Joshua M Lang, Mary Jane Brennan, Glenn Liu, Douglas G McNeel, Jens C Eickhoff, Ellen Wargowski, Laura E Johnson, Christos E Kyriakopoulos, Hamid Emamekhoo, Joshua M Lang, Mary Jane Brennan, Glenn Liu

Abstract

Background: We previously reported a trial using a DNA vaccine encoding prostatic acid phosphatase (MVI-816, pTVG-HP), given over 12 weeks concurrently or sequentially with pembrolizumab, in patients with mCRPC. We report the final analysis of this trial following two additional treatment arms in which patients with mCRPC continued concurrent treatment until progression.

Materials and methods: Patients with mCRPC were treated with MVI-816 and pembrolizumab every 3 weeks (arm 3, n=20) or MVI-816 every 2 weeks and pembrolizumab every 4 weeks (arm 4, n=20). The primary objectives were safety, 6-month progression-free survival (PFS), median time to radiographic progression, and objective response rates. Secondary objectives included immunological evaluations.

Results: In 25 patients with measurable disease, there were no complete response and one confirmed partial response in a patient who subsequently found to have an MSIhi tumor. 4/40 patients (10%) had a prostate-specific antigen decline >50%. The estimated overall radiographic PFS rate at 6 months was 47.2% (44.4% arm 3, 61.5% arm 4). Accounting for all off-study events, overall median time on treatment was 5.6 months (95% CI: 5.4 to 10.8 months), 5.6 months for arm 3 and 8.1 months for arm 4 (p=0.64). Thirty-two per cent of patients remained on trial beyond 6 months without progression. Median overall survival was 22.9 (95% CI: 16.2 to 25.6) months. One grade 4 event (hyperglycemia) was observed. Immune-related adverse events (irAEs) >grade 1 were observed in 42% of patients overall. Interferon-γ and/or granzyme B immune response to prostatic acid phosphatase was detected in 2/20 patients in arm 3 and 6/20 patients in arm 4. Plasma cytokines associated with immune activation and CD8+ T-cell recruitment were augmented at weeks 6 and 12. The development of irAE was significantly associated with a prolonged time on treatment (HR=0.42, p=0.003). Baseline DNA homologous recombination repair mutations were not associated with longer time to progression.

Conclusions: Findings here demonstrate that combining programmed cell death 1 blockade with MVI-816 is safe, can augment tumor-specific T cells, and can result in a favorable 6-month disease control rate. Correlative studies suggest T-cell activation by vaccination is critical to the mechanism of action of this combination. Future randomized clinical trials are needed to validate these findings.

Trial registration number: NCT02499835.

Keywords: clinical trials, phase II as topic; costimulatory and inhibitory T-cell receptors; immunogenicity, vaccine; prostatic neoplasms.

Conflict of interest statement

Competing interests: DGM has ownership interest, has received research support, and serves as consultant to Madison Vaccines, which has licensed material described in this manuscript and supported this trial.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Treatment schema for the original study arms evaluating treatment sequence (arms 1 and 2), and the arms evaluating treatment until progression (arms 3 and 4). pTVG-HP, DNA vaccine encoding prostatic acid phosphatase.
Figure 2
Figure 2
Immunological response: interferon-γ (IFN-γ) and granzyme B (GrzB) fluorescent ELISpot. Peripheral blood mononuclear cells (PBMCs) were collected from subjects (n=19, arm 3; n=20, arm 4) at baseline, 6 weeks, 12 weeks, and 24 weeks and evaluated for antigen-specific IFN-γ or GrzB secretion by fluorescent ELISpot. Shown are the spot-forming units (SFUs) for IFN-γ secretion (top panels) or GrzB secretion (bottom panels) following stimulation with a prostatic acid phosphatase (PAP) peptide library, PSA peptide library (non-specific control), or tetanus (positive control) for each patient. Patients treated in arm 3 are colored red, and patients treated in arm 4 are colored blue.
Figure 3
Figure 3
Treatment elicits increases in serum cytokines and chemokines. Plasma obtained from patients prior to treatment, and at weeks 6 and 12 of treatment, were evaluated for 35 cytokines and chemokines by multiplex Luminex assay. Shown are the changes in concentration from baseline for each patient, grouped by study arm (arm 1=green, arm 2=purple, arm 3=red, arm 4=blue), for those cytokines with significant change over time (by mixed-effects model): (A) G-CSF, (B) CXCL9, (C) CXCL10, (D) IFNα, (E) IL-2R, (F) IL-12, (G) MCP-1 (CCL2), (H) IL-1β, and (I) IL-2. (J) IFN-γ was below the level of detection for this multiplex assay and was separately measured from pretreatment to week 6 or pretreatment to week 12 by ELISA, with statistical comparison by paired t-test.
Figure 4
Figure 4
Clinical effects. (A) Serum PSA values were collected from all individuals prior to treatment and over the course of treatment. Shown are the best percentage change in serum PSA from day 1. Red lines show individual patients treated in arm 3, and blue lines show individual patients treated in arm 4. The horizontal line indicates 50% decrease from baseline. Asterisks indicate those individuals with immune response to prostatic acid phosphatase (PAP). (B) Shown are the best percentage change in tumor volume for 25 individuals in all treatment arms who had measurable disease, with the horizontal line indicating 30% decline. Asterisks indicate those individuals with immune response to PAP. (C) Swimmer plot showing the time on trial for all study patients. Asterisks indicate time of PSA progression, and open circles indicate radiographic progression. Triangles indicate those individuals with baseline DNA homologous recombination repair (HRR) mutations (n=10) or MSI-high tumors (n=2). The vertical line indicates 6 months.
Figure 5
Figure 5
Time on trial correlative analyses. Time on trial was assessed with respect to the development of grade 2 or higher immune-related adverse events (irAEs, A), T-cell immune response to prostatic acid phosphatase (B), increases in serum interferon-IFN-γ (C), and the presence of baseline homologous recombination repair (HRR) mutations or MSI-high tumors (D). Statistical comparisons are made using a log-rank test, with p

References

    1. Siegel RL, Miller KD, Fuchs HE, et al. . Cancer statistics, 2021. CA Cancer J Clin 2021;71:7–33. 10.3322/caac.21654
    1. Petrylak DP, Tangen CM, Hussain MHA, et al. . Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med 2004;351:1513–20. 10.1056/NEJMoa041318
    1. Tannock IF, de Wit R, Berry WR, et al. . Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351:1502–12. 10.1056/NEJMoa040720
    1. de Bono JS, Oudard S, Ozguroglu M, et al. . Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010;376:1147–54. 10.1016/S0140-6736(10)61389-X
    1. Kantoff PW, Higano CS, Shore ND, et al. . Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–22. 10.1056/NEJMoa1001294
    1. de Bono JS, Logothetis CJ, Molina A, et al. . Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 2011;364:1995–2005. 10.1056/NEJMoa1014618
    1. Scher HI, Fizazi K, Saad F, et al. . Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012;367:1187–97. 10.1056/NEJMoa1207506
    1. Parker C, Nilsson S, Heinrich D, et al. . Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369:213–23. 10.1056/NEJMoa1213755
    1. Small EJ, Lance RS, Gardner TA, et al. . A randomized phase II trial of Sipuleucel-T with concurrent versus sequential abiraterone acetate plus prednisone in metastatic castration-resistant prostate cancer. Clin Cancer Res 2015;21:3862–9. 10.1158/1078-0432.CCR-15-0079
    1. Sinha M, Zhang L, Subudhi S, et al. . Pre-existing immune status associated with response to combination of sipuleucel-T and ipilimumab in patients with metastatic castration-resistant prostate cancer. J Immunother Cancer 2021;9:e002254. 10.1136/jitc-2020-002254
    1. Graff JN, Alumkal JJ, Drake CG, et al. . Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 2016;7:52810–7. 10.18632/oncotarget.10547
    1. Slovin SF, Higano CS, Hamid O, et al. . Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol 2013;24:1813–21. 10.1093/annonc/mdt107
    1. Beer TM, Kwon ED, Drake CG, et al. . Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic Chemotherapy-Naive castration-resistant prostate cancer. J Clin Oncol 2017;35:40–7. 10.1200/JCO.2016.69.1584
    1. Petrylak DP, Loriot Y, Shaffer DR, et al. . Safety and clinical activity of Atezolizumab in patients with metastatic castration-resistant prostate cancer: a phase I study. Clin Cancer Res 2021;27:3360–9. 10.1158/1078-0432.CCR-20-1981
    1. Antonarakis ES, Piulats JM, Gross-Goupil M, et al. . Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: Multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol 2019:JCO1901638.
    1. Kwon ED, Drake CG, Scher HI, et al. . Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700–12. 10.1016/S1470-2045(14)70189-5
    1. Sharma P, Pachynski RK, Narayan V, et al. . Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell 2020;38:489–99. 10.1016/j.ccell.2020.08.007
    1. Boudadi K, Suzman DL, Anagnostou V, et al. . Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer. Oncotarget 2018;9:28561–71. 10.18632/oncotarget.25564
    1. Graff JN, Beer TM, Alumkal JJ, et al. . A phase II single-arm study of pembrolizumab with enzalutamide in men with metastatic castration-resistant prostate cancer progressing on enzalutamide alone. J Immunother Cancer 2020;8:e000642. 10.1136/jitc-2020-000642
    1. McNeel DG, Dunphy EJ, Davies JG, et al. . Safety and immunological efficacy of a DNA vaccine encoding prostatic acid phosphatase in patients with stage D0 prostate cancer. J Clin Oncol 2009;27:4047–54. 10.1200/JCO.2008.19.9968
    1. McNeel DG, Becker JT, Eickhoff JC, et al. . Real-Time immune monitoring to guide plasmid DNA vaccination schedule targeting prostatic acid phosphatase in patients with castration-resistant prostate cancer. Clin Cancer Res 2014;20:3692–704. 10.1158/1078-0432.CCR-14-0169
    1. Zahm CD, Colluru VT, McNeel DG. Vaccination with high-affinity epitopes impairs antitumor efficacy by increasing PD-1 expression on CD8+ T cells. Cancer Immunol Res 2017;5:630–41. 10.1158/2326-6066.CIR-16-0374
    1. Rekoske BT, Smith HA, Olson BM, et al. . Pd-1 or PD-L1 blockade restores antitumor efficacy following SSX2 Epitope-Modified DNA vaccine immunization. Cancer Immunol Res 2015;3:946–55. 10.1158/2326-6066.CIR-14-0206
    1. McNeel DG, Eickhoff JC, Wargowski E, et al. . Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget 2018;9:25586–96. 10.18632/oncotarget.25387
    1. Johnson LE, Frye TP, Arnot AR, et al. . Safety and immunological efficacy of a prostate cancer plasmid DNA vaccine encoding prostatic acid phosphatase (PAP). Vaccine 2006;24:293–303. 10.1016/j.vaccine.2005.07.074
    1. Scher HI, Halabi S, Tannock I, et al. . Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the prostate cancer clinical trials Working group. J Clin Oncol 2008;26:1148–59. 10.1200/JCO.2007.12.4487
    1. McNeel DG, Eickhoff JC, Johnson LE, et al. . Phase II Trial of a DNA Vaccine Encoding Prostatic Acid Phosphatase (pTVG-HP [MVI-816]) in Patients With Progressive, Nonmetastatic, Castration-Sensitive Prostate Cancer. J Clin Oncol 2019;37:3507–17. 10.1200/JCO.19.01701
    1. Kyriakopoulos CE, Eickhoff JC, Ferrari AC, et al. . Multicenter phase I trial of a DNA vaccine encoding the androgen receptor ligand-binding domain (pTVG-AR, MVI-118) in patients with metastatic prostate cancer. Clin Cancer Res 2020;26:5162–71. 10.1158/1078-0432.CCR-20-0945
    1. Zahm CD, Colluru VT, McIlwain SJ, et al. . TLR Stimulation during T-cell Activation Lowers PD-1 Expression on CD8 +T Cells. Cancer Immunol Res 2018;6:1364–74. 10.1158/2326-6066.CIR-18-0243
    1. Kantoff PW, Schuetz TJ, Blumenstein BA, et al. . Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol 2010;28:1099–105. 10.1200/JCO.2009.25.0597
    1. Robert C, Hwu W-J, Hamid O, et al. . Long-term safety of pembrolizumab monotherapy and relationship with clinical outcome: a landmark analysis in patients with advanced melanoma. Eur J Cancer 2021;144:182–91. 10.1016/j.ejca.2020.11.010
    1. Eltahir M, Isaksson J, Mattsson JSM, et al. . Plasma proteomic analysis in non-small cell lung cancer patients treated with PD-1/PD-L1 blockade. Cancers 2021;13:3116. 10.3390/cancers13133116
    1. Zahm CD, Moseman JE, Delmastro LE, et al. . PD-1 and LAG-3 blockade improve anti-tumor vaccine efficacy. Oncoimmunology 2021;10:e1912892. 10.1080/2162402X.2021.1912892

Source: PubMed

3
Subscribe