Neoadjuvant PROSTVAC prior to radical prostatectomy enhances T-cell infiltration into the tumor immune microenvironment in men with prostate cancer

Houssein Abdul Sater, Jennifer L Marté, Renee N Donahue, Beatriz Walter-Rodriguez, Christopher R Heery, Seth M Steinberg, Lisa M Cordes, Guinevere Chun, Fatima Karzai, Marijo Bilusic, Stephanie A Harmon, Ismail Baris Turkbey, Peter L Choyke, Jeffrey Schlom, William L Dahut, Ravi A Madan, Peter A Pinto, James L Gulley, Houssein Abdul Sater, Jennifer L Marté, Renee N Donahue, Beatriz Walter-Rodriguez, Christopher R Heery, Seth M Steinberg, Lisa M Cordes, Guinevere Chun, Fatima Karzai, Marijo Bilusic, Stephanie A Harmon, Ismail Baris Turkbey, Peter L Choyke, Jeffrey Schlom, William L Dahut, Ravi A Madan, Peter A Pinto, James L Gulley

Abstract

Background: Clinical trials have shown the ability of therapeutic vaccines to generate immune responses to tumor-associated antigens (TAAs). What is relatively less known is if this translates into immune-cell (IC) infiltration into the tumor microenvironment. This study examined whether neoadjuvant prostate-specific antigen (PSA)-targeted vaccination with PROSTVAC could induce T-cell immunity, particularly at the tumor site.

Methods: An open-label, phase II study of neoadjuvant PROSTVAC vaccine enrolled 27 patients with localized prostate cancer awaiting radical prostatectomy (RP). We evaluated increases in CD4 and CD8 T-cell infiltrates (RP tissue vs baseline biopsies) using a six-color multiplex immunofluorescence Opal method. Antigen-specific responses were assessed by intracellular cytokine staining after in vitro stimulation of peripheral blood mononuclear cells with overlapping 15-mer peptide pools encoding the TAAs PSA, brachyury and MUC-1.

Results: Of 27 vaccinated patients, 26 had matched prevaccination (biopsy) and postvaccination (RP) prostate samples available for non-compartmentalized analysis (NCA) and compartmentalized analysis (CA). Tumor CD4 T-cell infiltrates were significantly increased in postvaccination RP specimens compared with baseline biopsies by NCA (median 176/mm² vs 152/mm²; IQR 136-317/mm² vs 69-284/mm²; p=0.0249; median ratio 1.20; IQR 0.64-2.25). By CA, an increase in both CD4 T-cell infiltrates at the tumor infiltrative margin (median 198/mm² vs 151/mm²; IQR 123-500/mm² vs 85-256/mm²; p=0.042; median ratio 1.44; IQR 0.59-4.17) and in CD8 T-cell infiltrates at the tumor core (median 140/mm² vs 105/mm²; IQR 91-175/mm² vs 83-163/mm²; p=0.036; median ratio 1.25; IQR 0.88-2.09) were noted in postvaccination RP specimens compared with baseline biopsies. A total of 13/25 patients (52%) developed peripheral T-cell responses to any of the three tested TAAs (non-neoantigens); five of these had responses to more than one antigen of the three evaluated.

Conclusion: Neoadjuvant PROSTVAC can induce both tumor immune response and peripheral immune response.

Trial registration number: NCT02153918.

Keywords: clinical trials as topic; immunotherapy, active; tumor microenvironment; urologic neoplasms; vaccination.

Conflict of interest statement

Competing interests: The NCI has a collaborative research and development agreement (CRADA) with Bavarian Nordic, the manufacturer of the vaccine used in this study. Under this CRADA, resources to develop agents are provided, including the vaccine.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Cell densities in prevaccination biopsies (Pre) and in radical prostatectomy sections postvaccination (Post) with PROSTVAC. (A) Representative image of multiplex immunofluorescence panel. Insets show a cell expressing four markers (CD4, FOXP3, Ki67 and DAPI). (B) Immune-cell infiltrates were quantified in both prevaccination and postvaccination sections using inForm software. NCA of both CD4 and CD8 immune-cell density ratios was assessed by the Wilcoxon signed rank test. Median±IQR shown with horizontal lines. (C) Exceptional case with CD4 and CD8 immune-cell infiltrates prevaccination and postvaccination. CTL, cytotoxic T lymphocyte; NCA, non-compartmentalized analysis; PD-L1, programmed death-ligand 1; Th, T helper; TIME: tumor immune microenvironment.
Figure 2
Figure 2
Compartmental distribution of CD4 and CD8 T cells postvaccination in intraprostatic tissue. (A) Schematic representation of radical prostatectomy (RP) section with three virtually separate compartments: tumor core (CT), invasive margin (IM) and benign glands (NL). (B, C) CD4 and CD8 T-cell infiltrate average densities were quantified as previously described using inForm software in each compartment before and after treatment with PROSTVAC vaccine. CD4 and CD8 immune-cell density ratios were assessed by the Wilcoxon signed rank test. Median±IQR shown with horizontal lines.
Figure 3
Figure 3
Heterogeneity scores of immune-cell infiltrates within prostatic tumor immune microenvironment (TIME) in prevaccination and postvaccination sections. (A, B) CD4 and CD8 T-cell infiltrate peak densities were quantified as previously described using inForm software in each compartment before and after immunotherapy. CD4 and CD8 immune-cell density ratios were assessed by the Wilcoxon signed rank test. Median±IQR is shown with horizontal lines. (C, D) Mean PARIS scores within each compartment in both prevaccination and postvaccination sections are shown. CD4 and CD8 cell density ratios were assessed by the Wilcoxon signed rank test. CT, tumor core; IM, infiltrative margins; NL, benign lymph gland; PARIS, peak to average immune score.
Figure 4
Figure 4
Changes in immune-cell subsets within prostatic tissue before and after PROSTVAC vaccine therapy. All cell densities were quantified as previously described using inForm software. (A) Frequency of T-regulatory cells (Tregs) (CD4+FOXP3+) before and after PROSTVAC therapy in non-compartmentalized analysis (NCA). (B) Frequency of T helper cells (CD4+FOXP3–) before and after PROSTVAC therapy in NCA. (C, D) Activated (Ki67+) and non-activated (Ki67–) CD8 cytotoxic T lymphocytes were quantified using NCA in both prevaccination and postvaccination sections. For (A–D), Wilcoxon signed rank test was used. Median±IQR is shown with horizontal lines. (E, F) Representative images of all four immune-cell subsets (Tregs, T helper, activated cytotoxic T lymphocytes and non-proliferative cytotoxic T lymphocytes).
Figure 5
Figure 5
Antigen-specific T-cell responses assessed by intracellular cytokine staining of peripheral blood mononuclear cells following in vitro stimulation (pIR). Responses to the vaccine target antigen prostate-specific antigen and the cascade antigens MUC-1 and brachyury were measured by increases in the number of CD4+ or CD8+ lymphocytes producing cytokines (interferon-γ, tumor necrosis factor-α or interleukin-2) or positive for CD107a. An increase of ≥twofold from baseline was considered positive. pIR, peripheral immune response.

References

    1. Gulley JL, Arlen PM, Madan RA, et al. . Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother 2010;59:663–74. 10.1007/s00262-009-0782-8
    1. GuhaThakurta D, Sheikh NA, Fan L-Q, et al. . Humoral immune response against nontargeted tumor antigens after treatment with Sipuleucel-T and its association with improved clinical outcome. Clin Cancer Res 2015;21:3619–30. 10.1158/1078-0432.CCR-14-2334
    1. Fong L, Carroll P, Weinberg V, et al. . Activated lymphocyte recruitment into the tumor microenvironment following preoperative sipuleucel-T for localized prostate cancer. J Natl Cancer Inst 2014;106:1. 10.1093/jnci/dju268
    1. Madan RA, Arlen PM, Mohebtash M, et al. . Prostvac-VF: a vector-based vaccine targeting PSA in prostate cancer. Expert Opin Investig Drugs 2009;18:1001–11. 10.1517/13543780902997928
    1. Gulley J, Chen AP, Dahut W, et al. . Phase I study of a vaccine using recombinant vaccinia virus expressing PSA (rV-PSA) in patients with metastatic androgen-independent prostate cancer. Prostate 2002;53:109–17. 10.1002/pros.10130
    1. Gulley JL, Madan RA, Tsang KY, et al. . Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res 2014;2:133–41. 10.1158/2326-6066.CIR-13-0108
    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. Gulley JL, Borre M, Vogelzang NJ, et al. . Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. J Clin Oncol 2019;37:1051–61. 10.1200/JCO.18.02031
    1. Terasawa H, Tsang K-Y, Gulley J, et al. . Identification and characterization of a human agonist cytotoxic T-lymphocyte epitope of human prostate-specific antigen. Clin Cancer Res 2002;8:41–53.
    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. Jochems C, Tucker JA, Vergati M, et al. . Identification and characterization of agonist epitopes of the MUC1-C oncoprotein. Cancer Immunol Immunother 2014;63:161–74. 10.1007/s00262-013-1494-7
    1. Tucker JA, Jochems C, Boyerinas B, et al. . Identification and characterization of a cytotoxic T-lymphocyte agonist epitope of Brachyury, a transcription factor involved in epithelial to mesenchymal transition and metastasis. Cancer Immunol Immunother 2014;63:1307–17. 10.1007/s00262-014-1603-2
    1. Hamoen EHJ, de Rooij M, Witjes JA, et al. . Use of the prostate imaging reporting and data system (PI-RADS) for prostate cancer detection with multiparametric magnetic resonance imaging: a diagnostic meta-analysis. Eur Urol 2015;67:1112–21. 10.1016/j.eururo.2014.10.033
    1. Nakamura K, Joja I, Nagasaka T, et al. . The mean apparent diffusion coefficient value (ADCmean) on primary cervical cancer is a predictive marker for disease recurrence. Gynecol Oncol 2012;127:478–83. 10.1016/j.ygyno.2012.07.123
    1. Antony PA, Piccirillo CA, Akpinarli A, et al. . Cd8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. J Immunol 2005;174:2591–601. 10.4049/jimmunol.174.5.2591
    1. Janssen EM, Lemmens EE, Wolfe T, et al. . CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003;421:852–6. 10.1038/nature01441
    1. Kwek SS, Kahn J, Greaney SK, et al. . GM-CSF and ipilimumab therapy in metastatic melanoma: clinical outcomes and immunologic responses. Oncoimmunology 2016;5:e1101204. 10.1080/2162402X.2015.1101204
    1. Spitzer MH, Carmi Y, Reticker-Flynn NE, et al. . Systemic immunity is required for effective cancer immunotherapy. Cell 2017;168:487–502. 10.1016/j.cell.2016.12.022
    1. Vergati M, Cereda V, Madan RA, et al. . Analysis of circulating regulatory T cells in patients with metastatic prostate cancer pre- versus post-vaccination. Cancer Immunol Immunother 2011;60:197–206. 10.1007/s00262-010-0927-9
    1. Farsaci B, Jochems C, Grenga I, et al. . Identification by digital immunohistochemistry of intratumoral changes of immune infiltrates after vaccine in the absence of modifications of PBMC immune cell subsets. Int J Cancer 2014;135:862–70. 10.1002/ijc.28743
    1. Jochems C, Schlom J. Tumor-infiltrating immune cells and prognosis: the potential link between conventional cancer therapy and immunity. Exp Biol Med 2011;236:567–79. 10.1258/ebm.2011.011007
    1. Berthel A, Zoernig I, Valous NA, et al. . Detailed resolution analysis reveals spatial T cell heterogeneity in the invasive margin of colorectal cancer liver metastases associated with improved survival. Oncoimmunology 2017;6:e1286436. 10.1080/2162402X.2017.1286436
    1. Keren L, Bosse M, Marquez D, et al. . A structured Tumor-Immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell 2018;174:1373–87. 10.1016/j.cell.2018.08.039
    1. Angelova M, Mlecnik B, Vasaturo A, et al. . Evolution of metastases in space and time under immune selection. Cell 2018;175:751–65. 10.1016/j.cell.2018.09.018
    1. Fridman WH, Galon J, Pagès F, et al. . Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res 2011;71:5601–5. 10.1158/0008-5472.CAN-11-1316
    1. Tarhini AA, Edington H, Butterfield LH, et al. . Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One 2014;9:e87705. 10.1371/journal.pone.0087705
    1. Martin AM, Nirschl TR, Nirschl CJ, et al. . Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance. Prostate Cancer Prostatic Dis 2015;18:325–32. 10.1038/pcan.2015.39
    1. Calagua C, Russo J, Sun Y, et al. . Expression of PD-L1 in Hormone-naïve and treated prostate cancer patients receiving neoadjuvant abiraterone acetate plus prednisone and leuprolide. Clin Cancer Res 2017;23:6812–22. 10.1158/1078-0432.CCR-17-0807
    1. Gao J, Ward JF, Pettaway CA, et al. . VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med 2017;23:551–5. 10.1038/nm.4308
    1. Zhao SG, Lehrer J, Chang SL, et al. . The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. J Natl Cancer Inst 2019;111:301–10. 10.1093/jnci/djy141
    1. Redman JM, Steinberg SM, Gulley JL. Quick efficacy seeking trial (QuEST1): a novel combination immunotherapy study designed for rapid clinical signal assessment metastatic castration-resistant prostate cancer. J Immunother Cancer 2018;6:91. 10.1186/s40425-018-0409-8

Source: PubMed

3
Předplatit