Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models

Zebin Wang, Kaiming Sun, Yonghong Xiao, Bin Feng, Keith Mikule, XiaoYan Ma, Ningping Feng, Christopher P Vellano, Lorenzo Federico, Joseph R Marszalek, Gordon B Mills, Jeffrey Hanke, Sridhar Ramaswamy, Jing Wang, Zebin Wang, Kaiming Sun, Yonghong Xiao, Bin Feng, Keith Mikule, XiaoYan Ma, Ningping Feng, Christopher P Vellano, Lorenzo Federico, Joseph R Marszalek, Gordon B Mills, Jeffrey Hanke, Sridhar Ramaswamy, Jing Wang

Abstract

PARP inhibitors have been proven clinically efficacious in platinum-responsive ovarian cancer regardless of BRCA1/2 status and in breast cancers with germline BRCA1/2 mutation. However, resistance to PARP inhibitors may preexist or evolve during treatment in many cancer types and may be overcome by combining PARP inhibitors with other therapies, such as immune checkpoint inhibitors, which confer durable responses and are rapidly becoming the standard of care for multiple tumor types. This study investigated the therapeutic potential of combining niraparib, a highly selective PARP1/2 inhibitor, with anti-PD-1 immune checkpoint inhibitors in preclinical tumor models. Our results indicate that niraparib treatment increases the activity of the type I (alpha) and type II (gamma) interferon pathways and enhances the infiltration of CD8+ cells and CD4+ cells in tumors. When coadministered in immunocompetent models, the combination of niraparib and anti-PD-1 demonstrated synergistic antitumor activities in both BRCA-proficient and BRCA-deficient tumors. Interestingly, mice with tumors cured by niraparib monotherapy completely rejected tumor growth upon rechallenge with the same tumor cell line, suggesting the potential establishment of immune memory in animals treated with niraparib monotherapy. Taken together, our findings uncovered immunomodulatory effects of niraparib that may sensitize tumors to immune checkpoint blockade therapies.

Conflict of interest statement

Z. Wang., K. Sun., Y. Xiao., B. Fen., K. Mikule., J. Hanke., S. Ramaswamy. and J. Wang. are employees and/or shareholders of TESARO, Inc. G. B. Mills receives sponsored research support from TESARO, Inc.

Figures

Figure 1
Figure 1
Interferon response signature genes were significantly enriched in niraparib-treated tumors. (A) Tumor growth curve for the SK6005 syngeneic model with control or 50 mg/kg niraparib (QD) treatment. (B) Genes identified with DEseq as significantly upregulated upon niraparib treatment (p <=0.05, fold change >=1.5) were subjected to enrichment analysis of pathway gene sets. Gene Set Enrichment Analysis (GSEA) demonstrated a significant enrichment of interferon gamma signature (C) and interferon alpha signature (D) genes in niraparib-treated samples. (E) Tumor growth curve for the MDA-MB-436 NOG-EXL humanized model treated with control or 35 mg/kg niraparib daily for 5 days on and 2 days off for 4 weeks (QD × 5 × 4). (F) Significantly upregulated genes identified with a two-sample t test (p <=0.05, fold change >=1.5) were subjected to enrichment analysis of pathway gene sets. Gene Set Enrichment Analysis (GSEA) demonstrated a significant enrichment of interferon gamma signature (G) and interferon alpha signature (H) genes in niraparib-treated samples.
Figure 2
Figure 2
Niraparib promoted tumor immune cell infiltration in both the BRCA-proficient SK6005 syngeneic and BRCA-deficient MDA-MB-436 NOG-EXL humanized tumor models (A) Representative images of CD4 and CD8 immunohistochemical staining in control- and niraparib-treated BRCA-proficient SK6005 tumors. (BD) Quantification of the number of CD4+ cells, CD8+ cells and FoxP3+ cells per field upon niraparib treatment in BRCA-proficient SK6005 tumors. (E-G) Percentage of Ki67-positive CD4+ cells, CD8+ cells and FoxP3+ cells among the total CD3+ population by flow cytometry in humanized NOG-EXL MDA-MB-436 tumors. **p-value is less than 0.05 by Student’s t test.
Figure 3
Figure 3
Niraparib-induced interferon activation is present in xenograft tumors established in immunocompromised mice and in cultured tumor cells (A) Tumor growth curve for MDA-MB-436 tumors in an immunodeficient NOG model treated with control or 50 mg/kg niraparib (QD). (B) Genes identified with a two-sample t-test as significantly upregulated upon niraparib treatment (p <= 0.05, fold change >=1.5) in MDA-MB-436 tumors in immune-deficient NOG mice were subjected to enrichment analysis of pathway gene sets. (C) Genes identified with a paired two-sample t test as significantly upregulated upon niraparib treatment (p <=0.05, fold change >=1.5) in 8 niraparib sensitive PDX models were subjected to enrichment analysis of pathway gene sets. (D) mRNA expression of IFNB1 upon niraparib treatment (300 nM, 24 h and 48 h), etoposide treatment (50 µM, 18 h), or dA:dT transfection (0.5 µg/ml) in MDA-MB-436 cells. (E) Protein expression of p-STING (Ser366), STING, p-TBK1 (Ser172), TBK1, p-NF-κB p65 (Ser536) and NF-κB p65 upon 1 µM niraparib treatment (48 h) by western blotting from the same MDA-MB-436 lysate run on different gels indicated by the divider lines. (F) DAPI staining of nuclear structures following DMSO or 300 nM niraparib treatment in MDA-MB-436 cells cultured in vitro with arrows indicating micronuclei formation in niraparib-treated cells in vitro.
Figure 4
Figure 4
Combination therapy with niraparib and anti-PD-1 augmented antitumor activity and conferred durable responses in BRCA-deficient tumor models (A) Tumor growth in the BRCA-deficient MDA-MB-436 model in huNOG-EXL mice treated with 200 mg anti-PD-1 (pembrolizumab) on days 0, 4, 9, 13, 18, 22, and 28; 35 mg/kg niraparib daily for 5 days on and 2 days off for 4 weeks; and the combination of these agents. (B) The BRCA1-null ovarian cancer mouse syngeneic model was treated with 30 mg/kg niraparib QD, 10 mg/kg anti-PD-1 (BioXCell RMP1-14) BIW, and the combination of these agents for 21 days (day 9–29). Tumor regrowth was monitored post treatment (days 29–64). (C) The BRCA1-null ovarian cancer mouse syngeneic model was treated with niraparib 50 mg/kg QD, anti-PD-1 10 mg/kg BIW, and the combination of these agents for 21 days (days 9–29). Tumor regrowth was monitored post treatment (days 29–64). (D) Table summarizing the ratio of mice with palpable tumors on day 29 (last treatment day) and the ratio of mice with tumor growth observed during the drug-free, posttreatment observation period (days 30–64). (E) Growth curves for rechallenge with BRCA1-null ovarian cancer cells implanted on day 65 in the tumor-free mice from (D) and age-matched treatment-naïve mice.
Figure 5
Figure 5
Combination therapy with niraparib and an anti-PD-(L)1 antibody demonstrated significantly enhanced antitumor activity in a BRCA-proficient mouse syngeneic model (A) Tumor growth in the SK6005 skin syngeneic transplant model treated with anti-PD-1 (BioXCell RMP1–14), niraparib, and the combination of these agents. Niraparib was administered orally at 25 mg/kg daily, and the anti-PD-1 antibody was administered intraperitoneally at 5 mg/kg twice weekly. (B) Tumor growth in the MMTV-LPA1-T22 syngeneic transplant models treated with anti-PD-1 (2C4), niraparib, and the combination of these agents. Niraparib was administered orally at 50 mg/kg daily, and the anti-PD-1 antibody was administered intraperitoneally at 10 mg/kg twice weekly. (C) Tumor growth in the SA9003 sarcoma syngeneic model treated with anti-PD-1 (BioXCell RMP1–14), niraparib, and the combination of these agents. Niraparib was administered orally at 50 mg/kg daily, and the anti-PD-1 antibody was administered intraperitoneally at 10 mg/kg twice weekly. (D) Tumor growth in the KLN205 lung syngeneic model treated with anti-PD-1 (2C4), niraparib, and the combination of these agents. Niraparib was administered orally at 50 mg/kg daily, and the anti-PD-1 antibody was administered intraperitoneally at 5 mg/kg twice weekly. (E) Tumor growth in the MC38 colon syngeneic model treated with anti-PD-1 (BioXCell RMP1–14), niraparib, and the combination of these agents. Niraparib was administered orally at 50 mg/kg daily, and the anti-PD-1 antibody was administered intraperitoneally at 0.5 mg/kg twice weekly. (F) Tumor growth in the BL6078 bladder syngeneic model treated with anti-PD-L1 (BioXCell 10 f.9G2), niraparib, and the combination of these agents. Niraparib was administered orally at 50 mg/kg daily, and the anti-PD-L1 antibody was administered intraperitoneally at 10 mg/kg twice weekly.

References

    1. Vyas S, Chang P. New PARP targets for cancer therapy. Nat Rev Cancer. 2014;14:502–509. doi: 10.1038/nrc3748.
    1. Pommier Y, O’Connor MJ, de Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med. 2016;8:362ps317. doi: 10.1126/scitranslmed.aaf9246.
    1. Bryant HE, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434:913–917. doi: 10.1038/nature03443.
    1. Murai J, et al. Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. Cancer Res. 2012;72:5588–5599. doi: 10.1158/0008-5472CAN-12-2753.
    1. Mirza MR, et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N Engl J Med. 2016;375:2154–2164. doi: 10.1056/NEJMoa1611310.
    1. Pujade-Lauraine E, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. The Lancet. Oncology. 2017;18:1274–1284. doi: 10.1016/S1470-2045(17)30469-2.
    1. Coleman RL, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390:1949–1961. doi: 10.1016/S0140-6736(17)32440-6.
    1. Robson M, et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med. 2017;377:523–533. doi: 10.1056/NEJMoa1706450.
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465. doi: 10.1056/NEJMoa1200694.
    1. Gettinger S, Herbst RS. B7-H1/PD-1 blockade therapy in non-small cell lung cancer: current status and future direction. Cancer J. 2014;20:281–289. doi: 10.1097/PPO.0000000000000063.
    1. Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci. 2017;24:26. doi: 10.1186/s12929-017-0329-9.
    1. Philips GK, Atkins M. Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Int Immunol. 2015;27:39–46. doi: 10.1093/intimm/dxu095.
    1. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present. and future. J Clin Invest. 2015;125:3384–3391. doi: 10.1172/JCI80011.
    1. Feng M, et al. PD-1/PD-L1 and immunotherapy for pancreatic cancer. Cancer Lett. 2017;407:57–65. doi: 10.1016/j.canlet.2017.08.006.
    1. Mathew M, Enzler T, Shu CA, Rizvi NA. Combining chemotherapy with PD-1 blockade in NSCLC. Pharmacol Ther. 2018 doi: 10.1016/j.pharmthera.2018.01.003.
    1. Gotwals P, et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer. 2017;17:286–301. doi: 10.1038/nrc.2017.17.
    1. Huang J, et al. The PARP1 inhibitor BMN 673 exhibits immunoregulatory effects in a Brca1(−/−) murine model of ovarian cancer. Biochem Biophys Res Commun. 2015;463:551–556. doi: 10.1016/j.bbrc.2015.05.083.
    1. Jiao S, et al. PARP Inhibitor Upregulates PD-L1 Expression and Enhances Cancer-Associated Immunosuppression. Clin Cancer Res. 2017;23:3711–3720. doi: 10.1158/1078-0432.CCR-16-3215.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323. doi: 10.1186/1471-2105-12-323.
    1. Elstrodt F, et al. BRCA1 mutation analysis of 41 human breast cancer cell lines reveals three new deleterious mutants. Cancer Res. 2006;66:41–45. doi: 10.1158/0008-5472.CAN-05-2853.
    1. Ito R, et al. Establishment of a human allergy model using human IL-3/GM-CSF-transgenic NOG mice. J Immunol. 2013;191:2890–2899. doi: 10.4049/jimmunol.1203543.
    1. Farmer H, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917–921. doi: 10.1038/nature03445.
    1. Higuchi T, et al. CTLA-4 Blockade Synergizes Therapeutically with PARP Inhibition in BRCA1-Deficient Ovarian Cancer. Cancer Immunol Res. 2015;3:1257–1268. doi: 10.1158/2326-6066.CIR-15-0044.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Liberzon A, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–425. doi: 10.1016/j.cels.2015.12.004.
    1. Minn AJ, Wherry EJ. Combination Cancer Therapies with Immune Checkpoint Blockade: Convergence on Interferon Signaling. Cell. 2016;165:272–275. doi: 10.1016/j.cell.2016.03.031.
    1. Parker BS, Rautela J, Hertzog PJ. Antitumour actions of interferons: implications for cancer therapy. Nat Rev Cancer. 2016;16:131–144. doi: 10.1038/nrc.2016.14.
    1. Sistigu A, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20:1301–1309. doi: 10.1038/nm.3708.
    1. Ermolaeva MA, et al. DNA damage in germ cells induces an innate immune response that triggers systemic stress resistance. Nature. 2013;501:416–420. doi: 10.1038/nature12452.
    1. Brown, J. S., Sundar, R. & Lopez, J. Combining DNA damaging therapeutics with immunotherapy: more haste, less speed. Br J Cancer, 10.1038/bjc.2017.376 (2017).
    1. Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–1149. doi: 10.1038/ni.3558.
    1. Konno H, Konno K, Barber GN. Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling. Cell. 2013;155:688–698. doi: 10.1016/j.cell.2013.09.049.
    1. Liu S, et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science. 2015;347:aaa2630. doi: 10.1126/science.aaa2630.
    1. De Oliveira Mann CC, Kranzusch PJ. cGAS Conducts Micronuclei DNA Surveillance. Trends Cell Biol. 2017;27:697–698. doi: 10.1016/j.tcb.2017.08.007.
    1. Harding SM, et al. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature. 2017;548:466–470. doi: 10.1038/nature23470.
    1. Mackenzie KJ, et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature. 2017;548:461–465. doi: 10.1038/nature23449.
    1. Hartlova A, et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity. 2015;42:332–343. doi: 10.1016/j.immuni.2015.01.012.
    1. Orsulic S, et al. Induction of ovarian cancer by defined multiple genetic changes in a mouse model system. Cancer Cell. 2002;1:53–62. doi: 10.1016/S1535-6108(01)00002-2.
    1. Xing D, Orsulic S. A mouse model for the molecular characterization of brca1-associated ovarian carcinoma. Cancer Res. 2006;66:8949–8953. doi: 10.1158/0008-5472.CAN-06-1495.
    1. Liu S, et al. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell. 2009;15:539–550. doi: 10.1016/j.ccr.2009.03.027.
    1. Federico L, et al. A murine preclinical syngeneic transplantation model for breast cancer precision medicine. Sci Adv. 2017;3:e1600957. doi: 10.1126/sciadv.1600957.
    1. Homet Moreno B, et al. Response to Programmed Cell Death-1 Blockade in a Murine Melanoma Syngeneic Model Requires Costimulation, CD4, and CD8 T Cells. Cancer Immunol Res. 2016;4:845–857. doi: 10.1158/2326-6066.CIR-16-0060.
    1. Deng L, et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity. 2014;41:843–852. doi: 10.1016/j.immuni.2014.10.019.
    1. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15:760–770. doi: 10.1038/nri3921.
    1. Oyler-Yaniv J, et al. Catch and Release of Cytokines Mediated by Tumor Phosphatidylserine Converts Transient Exposure into Long-Lived Inflammation. Mol Cell. 2017;66:635–647 e637. doi: 10.1016/j.molcel.2017.05.011.
    1. Hosking MP, Flynn CT, Whitton JL. Antigen-specific naive CD8+T cells produce a single pulse of IFN-gamma in vivo within hours of infection, but without antiviral effect. J Immunol. 2014;193:1873–1885. doi: 10.4049/jimmunol.1400348.
    1. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75:163–189. doi: 10.1189/jlb.0603252.
    1. Takaoka A, et al. Cross talk between interferon-gamma and -alpha/beta signaling components in caveolar membrane domains. Science. 2000;288:2357–2360. doi: 10.1126/science.288.5475.2357.
    1. Vanpouille-Box C, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. doi: 10.1038/ncomms15618.
    1. Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative mean? J Exp Med. 2016;213:2835–2840. doi: 10.1084/jem.20161462.
    1. Luke JJ, Flaherty KT, Ribas A, Long GV. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol. 2017;14:463–482. doi: 10.1038/nrclinonc.2017.43.
    1. Daud AI, et al. Programmed Death-Ligand 1 Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J Clin Oncol. 2016;34:4102–4109. doi: 10.1200/JCO.2016.67.2477.
    1. Nowicki TS, et al. Infiltration of CD8 T Cells and Expression of PD-1 and PD-L1 in Synovial Sarcoma. Cancer Immunol Res. 2017;5:118–126. doi: 10.1158/2326-6066.CIR-16-0148.
    1. Strickland KC, et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget. 2016;7:13587–13598. doi: 10.18632/oncotarget.7277.

Source: PubMed

3
Subscribe