A phase 2, randomized, double-blind, placebo- controlled study of chemo-immunotherapy combination using motolimod with pegylated liposomal doxorubicin in recurrent or persistent ovarian cancer: a Gynecologic Oncology Group partners study

B J Monk, M F Brady, C Aghajanian, H A Lankes, T Rizack, J Leach, J M Fowler, R Higgins, P Hanjani, M Morgan, R Edwards, W Bradley, T Kolevska, P Foukas, E M Swisher, K S Anderson, R Gottardo, J K Bryan, M Newkirk, K L Manjarrez, R S Mannel, R M Hershberg, G Coukos, B J Monk, M F Brady, C Aghajanian, H A Lankes, T Rizack, J Leach, J M Fowler, R Higgins, P Hanjani, M Morgan, R Edwards, W Bradley, T Kolevska, P Foukas, E M Swisher, K S Anderson, R Gottardo, J K Bryan, M Newkirk, K L Manjarrez, R S Mannel, R M Hershberg, G Coukos

Abstract

Background: A phase 2, randomized, placebo-controlled trial was conducted in women with recurrent epithelial ovarian carcinoma to evaluate the efficacy and safety of motolimod-a Toll-like receptor 8 (TLR8) agonist that stimulates robust innate immune responses-combined with pegylated liposomal doxorubicin (PLD), a chemotherapeutic that induces immunogenic cell death.

Patients and methods: Women with ovarian, fallopian tube, or primary peritoneal carcinoma were randomized 1 : 1 to receive PLD in combination with blinded motolimod or placebo. Randomization was stratified by platinum-free interval (≤6 versus >6-12 months) and Gynecologic Oncology Group (GOG) performance status (0 versus 1). Treatment cycles were repeated every 28 days until disease progression.

Results: The addition of motolimod to PLD did not significantly improve overall survival (OS; log rank one-sided P = 0.923, HR = 1.22) or progression-free survival (PFS; log rank one-sided P = 0.943, HR = 1.21). The combination was well tolerated, with no synergistic or unexpected serious toxicity. Most patients experienced adverse events of fatigue, anemia, nausea, decreased white blood cells, and constipation. In pre-specified subgroup analyses, motolimod-treated patients who experienced injection site reactions (ISR) had a lower risk of death compared with those who did not experience ISR. Additionally, pre-treatment in vitro responses of immune biomarkers to TLR8 stimulation predicted OS outcomes in patients receiving motolimod on study. Immune score (tumor infiltrating lymphocytes; TIL), TLR8 single-nucleotide polymorphisms, mutational status in BRCA and other DNA repair genes, and autoantibody biomarkers did not correlate with OS or PFS.

Conclusions: The addition of motolimod to PLD did not improve clinical outcomes compared with placebo. However, subset analyses identified statistically significant differences in the OS of motolimod-treated patients on the basis of ISR and in vitro immune responses. Collectively, these data may provide important clues for identifying patients for treatment with immunomodulatory agents in novel combinations and/or delivery approaches.

Trial registration: Clinicaltrials.gov, NCT 01666444.

Trial registration: ClinicalTrials.gov NCT01666444.

Keywords: Toll-like receptor 8; biomarkers; immunotherapy; motolimod; oncology; ovarian cancer.

© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology.

Figures

Figure 1.
Figure 1.
Kaplan–Meier plots of OS (A) and PFS (B) in the ITT population. No significant increase in either OS or PFS was observed between patients treated with motolimod plus PLD (blue line) and patients treated with placebo plus PLD (red line). Landmark analysis of OS in patients receiving motolimod, comparing those who experienced injection site reaction (ISR+; blue line) to those who did not (ISR−; red line) (C). Among motolimod-treated patients, those who were ISR+ had longer OS compared with those who were ISR−.
Figure 2.
Figure 2.
(A) Statistically significant (two-sided P < 0.05) upregulation of plasma analytes following subcutaneous administration of 3.0 mg/m2 of motolimod. Analytes that were prospectively identified as being responsive to TLR8 stimulation by motolimod are identified with a dark gray heading. Data shown represent log-fold changes (pre-dose to 8 h post-dose) in each analyte. P-values for each analyte are provided in supplementary Table S3, available at Annals of Oncology online. (B) In vivo upregulation of immunologically important analytes. P-values for each analyte are provided in supplementary Table S4, available at Annals of Oncology online.
Figure 3.
Figure 3.
(A) Statistically significant (two-sided P < 0.05) upregulation of analytes in vitro from baseline (pre-treatment) assessment of immune responsiveness to TLR8 stimulation. Data shown represent log-fold changes in each analyte in response to in vitro stimulation with 300 nM motolimod compared with null (no stimulant). Corresponding P-values are provided in supplementary Table S5, available at Annals of Oncology online. Analytes that were prospectively identified as being responsive to in vitro activation of TLR8 by motolimod are identified with a dark gray heading. (B) Analysis of OS by baseline in vitro response in IFN-γ (A), IL-12p40 (B), and TNF-α. (C) In vitro responses for each analyte was discretized as low, medium, or high and their association with survival was tested using a one-sided log-rank test. Within the motolimod plus PLD treatment group only, patients with higher IFN-γ (P = 0.049), TNF-α (P = 0.041), or IL-12p40 (P = 0.024) response had significantly improved survival compared to those motolimod-treated subjects who had a lower level of mediator response.

References

    1. American Cancer Society. What are the key statistics about ovarian cancer? [article on internet] 2016. (22 September 2016, date last accessed).
    1. NIH National Cancer Institute. SEER Stat Fact Sheets: Ovarian Cancer [database on internet] 2016. (9 November 2016, date last accessed).
    1. Coleman RL, Monk BJ, Sood AK, Herzog TJ.. Latest research and treatment of advanced-stage epithelial ovarian cancer. Nat Rev Clin Oncol 2013; 10: 211–224.
    1. Morgan RJ Jr, Armstrong DK, Alvarez RD. et al. Ovarian cancer, Version 1.2016, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2016; 14: 1134–1163.
    1. Buda A, Floriani I, Rossi R. et al. Randomised controlled trial comparing single agent paclitaxel vs epidoxorubicin plus paclitaxel in patients with advanced ovarian cancer in early progression after platinum-based chemotherapy: an Italian Collaborative Study from the Mario Negri Institute, Milan, G.O.N.O. (Gruppo Oncologico Nord Ovest) group and I.O.R. (Istituto Oncologico Romagnolo) group. Br J Cancer 2004; 90: 2112–2117.
    1. Vergote I, Finkler NJ, Hall JB. et al. Randomized phase III study of canfosfamide in combination with pegylated liposomal doxorubicin compared with pegylated liposomal doxorubicin alone in platinum-resistant ovarian cancer. Int J Gynecol Cancer 2010; 20: 772–780.
    1. Mutch DG, Orlando M, Goss T. et al. Randomized phase III trial of gemcitabine compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer. J Clin Oncol 2007; 25: 2811–2818.
    1. ten Bokkel Huinink W, Gore M, Carmichael J. et al. Topotecan versus paclitaxel for the treatment of recurrent epithelial ovarian cancer. J Clin Oncol 1997; 15: 2183–2193.
    1. Monk BJ, Herzog TJ, Kaye SB. et al. Trabectedin plus pegylated liposomal doxorubicin in recurrent ovarian cancer. J Clin Oncol 2010; 28: 3107–3114.
    1. Gordon AN, Fleagle JT, Guthrie D. et al. Recurrent epithelial ovarian carcinoma: a randomized phase III study of pegylated liposomal doxorubicin versus topotecan. J Clin Oncol 2001; 19: 3312–3322.
    1. Zhang L, Conejo-Garcia JR, Katsaros D. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003; 348: 203–213.
    1. Sato E, Olson SH, Ahn J. et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 2005; 102: 18538–18543.
    1. Zitvogel L, Kepp O, Senovilla L. et al. Immunogenic tumor cell death for optimal anticancer therapy: the calreticulin exposure pathway. Clin Cancer Res 2010; 16: 3100–3104.
    1. Casares N, Pequignot MO, Tesniere A. et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med 2005; 202: 1691–1701.
    1. Obeid M, Tesniere A, Ghiringhelli F. et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007; 13: 54–61.
    1. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V.. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12: 253–268.
    1. Kawai T, Akira S.. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 2010; 11: 373–384.
    1. Lu H, Dietsch GN, Matthews MA. et al. VTX-2337 is a novel TLR8 agonist that activates NK cells and augments ADCC. Clin Cancer Res 2012; 18: 499–509.
    1. Dietsch GN, Randall TD, Gottardo R. et al. Late-stage cancer patients remain highly responsive to immune activation by the selective TLR8 agonist motolimod (VTX-2337). Clin Cancer Res 2015; 21: 5445–5452.
    1. Monk BJ, Facciabene A, Brady WE. et al. Integrative development of a TLR8 agonist for ovarian cancer chemo-immunotherapy. Clin Cancer Res 2016; doi: 10.1158/1078-0432.CCR-16-1453.
    1. Dietsch GN, Lu H, Yang Y. et al. Coordinated activation of Toll-Like Receptor8 (TLR8) and NLRP3 by the TLR8 agonist, VTX-2337, ignites tumoricidal natural killer cell activity. PLoS ONE 2016; 11: e0148764..
    1. Stephenson RM, Lim CM, Matthews M. et al. TLR8 stimulation enhances cetuximab-mediated natural killer cell lysis of head and neck cancer cells and dendritic cell cross-priming of EGFR-specific CD8+ T cells. Cancer Immunol Immunother 2013; 62: 1347–1357.
    1. Bohnsack OHA, Ludajic K.. Adaptation of the immune related response criteria: irRECIST. Ann Oncol 2014; 25 (suppl 4): iv361–iv372.
    1. National Cancer Institute. Common Terminology Criteria for Adverse Events v4.0. NIH publication # 09–7473. 2009.
    1. Hwang WT, Adams SF, Tahirovic E. et al. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol 2012; 124: 192–198.
    1. Mittica G, Genta S, Aglietta M, Valabrega G.. Immune checkpoint inhibitors: a new opportunity in the treatment of ovarian cancer? IJMS 2016; 17(7). doi: 10.3390/ijms17071169.
    1. Homicsko K, Coukos G.. Targeting programmed cell death 1 in ovarian cancer. J Clin Oncol 2015; 33: 3987–3989.
    1. Lawrence MS, Stojanov P, Polak P. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013; 499: 214–218.

Source: PubMed

3
購読する