Vaccination against RhoC induces long-lasting immune responses in patients with prostate cancer: results from a phase I/II clinical trial

Juliane Schuhmacher, Sonja Heidu, Torben Balchen, Jennifer Rebecca Richardson, Camilla Schmeltz, Jesper Sonne, Jonas Schweiker, Hans-Georg Rammensee, Per Thor Straten, Martin Andreas Røder, Klaus Brasso, Cécile Gouttefangeas, Juliane Schuhmacher, Sonja Heidu, Torben Balchen, Jennifer Rebecca Richardson, Camilla Schmeltz, Jesper Sonne, Jonas Schweiker, Hans-Georg Rammensee, Per Thor Straten, Martin Andreas Røder, Klaus Brasso, Cécile Gouttefangeas

Abstract

Background: Peptide-based vaccination is a rational option for immunotherapy of prostate cancer. In this first-in-man phase I/II study, we assessed the safety, tolerability and immunological impact of a synthetic long peptide vaccine targeting Ras homolog gene family member C (RhoC) in patients with prostate cancer. RhoC is a small GTPase overexpressed in advanced solid cancers, metastases and cancer stem cells.

Methods: Twenty-two patients who had previously undergone radical prostatectomy received subcutaneous injections of 0.1 mg of a single RhoC-derived 20mer peptide emulsified in Montanide ISA-51 every 2 weeks for the first six times, then five times every 4 weeks for a total treatment time of 30 weeks. The drug safety and vaccine-specific immune responses were assessed during treatment and thereafter within a 13-month follow-up period. Serum level of prostate-specific antigen was measured up to 26 months postvaccination.

Results: Most patients (18 of 21 evaluable) developed a strong CD4 T cell response against the vaccine, which lasted at least 10 months following the last vaccination. Three promiscuouslypresented HLA-class II epitopes were identified. Vaccine-specific CD4 T cells were polyfunctional and effector memory T cells that stably expressed PD-1 (CD279) and OX-40 (CD134), but not LAG-3 (CD223). One CD8 T cell response was detected in addition. The vaccine was well tolerated and no treatment-related adverse events of grade ≥3 were observed.

Conclusion: Targeting of RhoC induced a potent and long-lasting T cell immunity in the majority of the patients. The study demonstrates an excellent safety and tolerability profile. Vaccination against RhoC could potentially delay or prevent tumor recurrence and metastasis formation.

Trial registration number: NCT03199872.

Keywords: T-Lymphocytes; immunotherapy; prostatic neoplasms; translational medical research; vaccination.

Conflict of interest statement

Competing interests: CG and H-GR receive a research grant from RhoVac ApS. PTS is consultant at RhoVac ApS. CS is employee of DanTrials. TB is owner and CEO of DanTrials.

© 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
RV001-specific T cells are induced after RhoC vaccination. (A) Vaccination and monitoring schedule. Patients were vaccinated 11 times. For immunoassays, blood was taken prevaccination, three times during the vaccination phase (vaccination) and four times postvaccination (follow-up 1 and follow-up 2) (blood drops). PBMCs were prestimulated with the RV001 and expanded for 12 days before IFN-γ ELISpot testing (0.2×106 cells/well, except for Patient 21 visits 2–13, and Patient 012 visits 16+17: 0.17×106 cells/well). (B) Exemplary result of an ELISpot (Patient 011). ddH2O and PHA were used as negative and positive control, respectively. (C–E) RV001-specific mean spot counts per analyzed time and normalized to 0.2×106 cells/well. Three independent ELISpot experiments were performed (indicated by the gaps). The sums of RV001-specific mean spot numbers (V6+8+13) are shown for strong- (C; n=7; ≥2500 spots), intermediate- (D; n=7; ≥1500–2500 spots), and weak/non- (E; n=7; 0–1500 spots) responders. (F) RV001-specific mean spot counts per patient and visit normalized to 0.2×106 cells/well. Light green indicates a statistical significance according to the DFR(2x) permutation test. n=21 patients. ddH2O, bidistilled water; IFN-γ, interferon-γ; na, cells not available; PHA, phytohemagglutinin; RhoC, Ras homolog gene family member C.
Figure 2
Figure 2
RV001-responding cells are multifunctional. ddH2O stimulated cells harvested from the ELISpot were restimulated with RV001 for 12 hours. Expression of CD107a, CD154, IL-2, TNF, and IFN-γ was examined by ICS on live CD4 and CD8 lymphocytes and the % of RV001-specific cells calculated for each of the 5 markers within CD4 or CD8 cell subsets. (A) Overview of CD4 T cell responses during vaccination (n=18 patients). (B) Mean+95% CI of cumulative marker expression on RV001-specific CD4 T cells for strong- (n=7), intermediate- (n=7) and weak- (n=4) responders. Kruskal-Wallis test with Dunn’s post-test. (C) Min to max percentages of RV001-specific CD4 T cells expressing one to five markers simultaneously, classified per strong- (n=7), intermediate- (n=7), and weak- (n=4) responders. Median values are indicated. Two-way ANOVA with Tukey’s post-test. (D) Mean+95% CI of RV001 specific CD4 T cells expressing each of the five activation markers or combinations thereof (all patients, n=18). (E) 12 day-cultured PBMCs from Patient 004 at visit 14 were restimulated with ddH2O (upper dot-plot panel) or RV001 (lower dot-plot panel). The activation marker expression was examined on living CD4 (upper rows) and CD8 (lower rows) lymphocytes. Percentages of marker+ cells within CD4 or CD8 cells are given. *P≤0.05, **p≤0.01, ***p≤0.001. Responder groups are defined based on the ELISpot results. ANOVA, analysis of variance; ddH2O, deionized water; IFN-γ, interferon-γ; IL-2, interleukin-2; TNF, tumor necrosis factor.
Figure 3
Figure 3
RV001-specific T cells are effector memory T cells. PBMCs from Patient 005 (visits 6–15) and Patients 009 and 018 (visits 6–17) were thawed, rested and stimulated either with RV001 or ddH2O for 12 hours. Live RV001-specific CD4 lymphocytes were identified by TNF expression and examined for the expression of CD45RA, CCR7, PD-1, OX-40, and LAG-3. (A) Exemplary results (Patient 009): CD4+TNF+ cells (black) are overlaid on the whole CD4 cell population (gray). Numbers indicate CD4+TNF+ cell counts. (B) Expression profile of PD-1 (upper row) and OX-40 (lower row) for n=3 patients at visit 6 and visit 15. Numbers indicate MFI ratios of the two receptors between CD4+TNF+CD45RA-CCR7- cells (dark gray) and CD4+TNF-CD45RA-CCR7- cells (light gray). Histograms show event counts normalized to mode. (C–E) Assessment of Tregs (CD3+CD8-CD4+CD127-CD25+Foxp3+) from 5 patients (4 visits). (C) Mean+95% CI of total CD4 cells (left-axis, black curves) and Tregs (right axis, blue curves) within lymphocytes. (D) Kinetics of Treg percentages within CD4 T lymphocytes for each of the 4 patients. (E) Exemplary dot-plots for Patient 003, percentages of CD3+CD8-CD4+CD127-CD25+Foxp3+ cells are indicated. ddH2O, deionized water; MFI, median fluorescence intensity; TNF, tumor necrosis factor.
Figure 4
Figure 4
The RV001 sequence comprises several promiscuous HLA-class II epitopes and one HLA-B*27:05 restricted epitope. The expression of CD107a, CD154, IL-2, TNF and IFN-γ was examined on live CD4 or CD8 cells. (A) Cells were restimulated with RV001 or with RV001-derived 15mer peptides (ATR15, AGL15, LQV15) for 12 hours. Shown are the percentage or mean+SD percentage (n=2 repeated measurements) of peptide-specific CD4 cells expressing each activation marker for three patients at visit 16. (B) LCLs were preloaded with RV001, ATR15, or AGL15 and incubated with HLA-matched patient cells at 1:2 ratio for 12 hours. Shown are the specific percentages of CD4 cells expressing the indicated activation markers. (C) Cells were restimulated either with the RV001 peptide alone or with RV001-preloaded C1R or C1R-HLA-B*27:05 cells for 12 hours in the ICS. Shown are the percentages of specific marker expression on CD8 cells. IFN-γ, interferon-γ; IL-2, interleukin 2; LCLs, lymphoblastic cell lines; TNF, tumor necrosis factor.

References

    1. Ott PA, Hu Z, Keskin DB, et al. . An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017;547:217–21. 10.1038/nature22991
    1. Sahin U, Derhovanessian E, Miller M, et al. . Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017;547:222–6. 10.1038/nature23003
    1. Hilf N, Kuttruff-Coqui S, Frenzel K, et al. . Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 2019;565:240–5. 10.1038/s41586-018-0810-y
    1. Cassell D, Forman J. Linked recognition of helper and cytotoxic antigenic determinants for the generation of cytotoxic T lymphocytes. Ann N Y Acad Sci 1988;532:51–60. 10.1111/j.1749-6632.1988.tb36325.x
    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. Sun JC, Bevan MJ. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 2003;300:339–42. 10.1126/science.1083317
    1. Mumberg D, Monach PA, Wanderling S, et al. . CD4(+) T cells eliminate MHC class II-negative cancer cells in vivo by indirect effects of IFN-gamma. Proc Natl Acad Sci U S A 1999;96:8633–8. 10.1073/pnas.96.15.8633
    1. Müller-Hermelink N, Braumüller H, Pichler B, et al. . TNFR1 signaling and IFN-γ signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 2008;13:507–18. 10.1016/j.ccr.2008.04.001
    1. Quezada SA, Simpson TR, Peggs KS, et al. . Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J Exp Med 2010;207:637–50. 10.1084/jem.20091918
    1. Perez-Diez A, Joncker NT, Choi K, et al. . CD4 cells can be more efficient at tumor rejection than CD8 cells. Blood 2007;109:5346–54. 10.1182/blood-2006-10-051318
    1. Slingluff CL, Lee S, Zhao F, et al. . A randomized phase II trial of multiepitope vaccination with melanoma peptides for cytotoxic T cells and helper T cells for patients with metastatic melanoma (E1602). Clin Cancer Res 2013;19:4228–38. 10.1158/1078-0432.CCR-13-0002
    1. Kenter GG, Welters MJP, Valentijn ARPM, et al. . Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009;361:1838–47. 10.1056/NEJMoa0810097
    1. Rosalia RA, Quakkelaar ED, Redeker A, et al. . Dendritic cells process synthetic long peptides better than whole protein, improving antigen presentation and T-cell activation. Eur J Immunol 2013;43:2554–65. 10.1002/eji.201343324
    1. Thomas P, Pranatharthi A, Ross C, et al. . RhoC: a fascinating journey from a cytoskeletal organizer to a cancer stem cell therapeutic target. J Exp Clin Cancer Res 2019;38:328. 10.1186/s13046-019-1327-4
    1. Lang S, Busch H, Boerries M, et al. . Specific role of RhoC in tumor invasion and metastasis. Oncotarget 2017;8:87364–78. 10.18632/oncotarget.20957
    1. Ridley AJ. Rhoa, RhoB and RhoC have different roles in cancer cell migration. J Microsc 2013;251:242–9. 10.1111/jmi.12025
    1. Kleer CG, van Golen KL, Zhang Y, et al. . Characterization of RhoC expression in benign and malignant breast disease: a potential new marker for small breast carcinomas with metastatic ability. Am J Pathol 2002;160:579–84. 10.1016/S0002-9440(10)64877-8
    1. Iiizumi M, Bandyopadhyay S, Pai SK, et al. . RhoC promotes metastasis via activation of the Pyk2 pathway in prostate cancer. Cancer Res 2008;68:7613–20. 10.1158/0008-5472.CAN-07-6700
    1. Islam M, Sharma S, Teknos TN. RhoC regulates cancer stem cells in head and neck squamous cell carcinoma by overexpressing IL-6 and phosphorylation of STAT3. PLoS One 2014;9:e88527. 10.1371/journal.pone.0088527
    1. Rosenthal DT, Zhang J, Bao L, et al. . RhoC impacts the metastatic potential and abundance of breast cancer stem cells. PLoS One 2012;7:e40979. 10.1371/journal.pone.0040979
    1. Gu G, Yuan J, Wills M, et al. . Prostate cancer cells with stem cell characteristics reconstitute the original human tumor in vivo. Cancer Res 2007;67:4807–15. 10.1158/0008-5472.CAN-06-4608
    1. Miyake H, Hara I, Kurahashi T, et al. . Quantitative detection of micrometastases in pelvic lymph nodes in patients with clinically localized prostate cancer by real-time reverse transcriptase-PCR. Clin Cancer Res 2007;13:1192–7. 10.1158/1078-0432.CCR-05-2706
    1. Wenandy L, Sørensen RB, Svane IM, et al. . RhoC a new target for therapeutic vaccination against metastatic cancer. Cancer Immunol Immunother 2008;57:1871–8. 10.1007/s00262-008-0517-2
    1. Memorial Sloan Kettering cancer center. Available: [Accessed 26 Mar 2020].
    1. Widenmeyer M, Griesemann H, Stevanović S, et al. . Promiscuous survivin peptide induces robust CD4+ T-cell responses in the majority of vaccinated cancer patients. Int J Cancer 2012;131:140–9. 10.1002/ijc.26365
    1. Moodie Z, Price L, Gouttefangeas C, et al. . Response definition criteria for ELISPOT assays revisited. Cancer Immunol Immunother 2010;59:1489–501. 10.1007/s00262-010-0875-4
    1. Thomsen FB, Brasso K, Berg KD, et al. . Association between PSA kinetics and cancer-specific mortality in patients with localised prostate cancer: analysis of the placebo arm of the SPCG-6 study. Ann Oncol 2016;27:460–6. 10.1093/annonc/mdv607
    1. Fuessel S, Meye A, Schmitz M, et al. . Vaccination of hormone-refractory prostate cancer patients with peptide cocktail-loaded dendritic cells: results of a phase I clinical trial. Prostate 2006;66:811–21. 10.1002/pros.20404
    1. Feyerabend S, Stevanovic S, Gouttefangeas C, et al. . Novel multi-peptide vaccination in HLA-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate 2009;69:917–27. 10.1002/pros.20941
    1. Noguchi M, Koga N, Igawa T, et al. . Clinical development of immunotherapy for prostate cancer. Int J Urol 2017;24:675–80. 10.1111/iju.13397
    1. Antonarakis ES, Kibel AS, Yu EY, et al. . Sequencing of sipuleucel-T and androgen deprivation therapy in men with hormone-sensitive biochemically recurrent prostate cancer: a phase II randomized trial. Clin Cancer Res 2017;23:2451–9. 10.1158/1078-0432.CCR-16-1780
    1. van Doorn E, Liu H, Huckriede A, et al. . Safety and tolerability evaluation of the use of Montanide ISA™51 as vaccine adjuvant: a systematic review. Hum Vaccin Immunother 2016;12:159–69. 10.1080/21645515.2015.1071455
    1. Tay NQ, Lee DCP, Chua YL, et al. . CD40L Expression Allows CD8+ T Cells to Promote Their Own Expansion and Differentiation through Dendritic Cells. Front Immunol 2017;8:1484. 10.3389/fimmu.2017.01484
    1. Butterfield LH, Ribas A, Dissette VB, et al. . Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res 2003;9:998–1008.
    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. Schoenberger SP, Toes REM, van der Voort EIH, et al. . T-Cell help for cytotoxic T lymphocytes is mediated by CD40–CD40L interactions. Nature 1998;393:480–3. 10.1038/31002
    1. Rivoltini L, Radrizzani M, Accornero P, et al. . Human melanoma-reactive CD4+ and CD8+ CTL clones resist Fas ligand-induced apoptosis and use Fas/Fas ligand-independent mechanisms for tumor killing. J Immunol 1998;161:1220–30.
    1. Yunger S, Bar El A, Zeltzer L-A, et al. . Tumor-Infiltrating lymphocytes from human prostate tumors reveal anti-tumor reactivity and potential for adoptive cell therapy. Oncoimmunology 2019;8:e1672494. 10.1080/2162402X.2019.1672494
    1. Younger AR, Amria S, Jeffrey WA, et al. . HLA class II antigen presentation by prostate cancer cells. Prostate Cancer Prostatic Dis 2008;11:334–41. 10.1038/sj.pcan.4501021
    1. Fauskanger M, Haabeth OAW, Skjeldal FM, et al. . Tumor Killing by CD4+ T Cells Is Mediated via Induction of Inducible Nitric Oxide Synthase-Dependent Macrophage Cytotoxicity. Front Immunol 2018;9:1684. 10.3389/fimmu.2018.01684
    1. Gjertsen MK, Buanes T, Rosseland AR, et al. . Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: clinical and immunological responses in patients with pancreatic adenocarcinoma. Int J Cancer 2001;92:441–50. 10.1002/ijc.1205
    1. Kreiter S, Vormehr M, van de Roemer N, et al. . Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015;520:692–6. 10.1038/nature14426
    1. Hunder NN, Wallen H, Cao J, et al. . Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med 2008;358:2698–703. 10.1056/NEJMoa0800251
    1. Tran E, Turcotte S, Gros A, et al. . Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641–5. 10.1126/science.1251102
    1. Adusumilli PS, Cherkassky L, Villena-Vargas J, et al. . Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med 2014;6:ra151. 10.1126/scitranslmed.3010162
    1. Wang D, Aguilar B, Starr R, et al. . Glioblastoma-targeted CD4+ CAR T cells mediate superior antitumor activity. JCI Insight 2018;3:99048. 10.1172/jci.insight.99048
    1. Thomsen FB, Christensen IJ, Brasso K, et al. . Prostate-Specific antigen doubling time as a progression criterion in an active surveillance programme for patients with localized prostate cancer. BJU Int 2014;113:E98–105. 10.1111/bju.12367
    1. Diaz M, Peabody JO, Kapoor V, et al. . Oncologic outcomes at 10 years following robotic radical prostatectomy. Eur Urol 2015;67:1168–76. 10.1016/j.eururo.2014.06.025
    1. Boorjian SA, Thompson RH, Tollefson MK, et al. . Long-Term risk of clinical progression after biochemical recurrence following radical prostatectomy: the impact of time from surgery to recurrence. Eur Urol 2011;59:893–9. 10.1016/j.eururo.2011.02.026
    1. Røder MA, Berg KD, Loft MD, et al. . The CPC risk calculator: a new APP to predict prostate-specific antigen recurrence during follow-up after radical prostatectomy. Eur Urol Focus 2018;4:360–8. 10.1016/j.euf.2016.11.014

Source: PubMed

3
订阅