Phase 1b study of a small molecule antagonist of human chemokine (C-C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma

Investigational New Drugs - Tập 38 Số 3 - Trang 800-811 - 2020
Marcus Smith Noel1, Eileen M. O’Reilly2, Brian M. Wolpin3, David P. Ryan4, Andrea J. Bullock5, Carolyn D. Britten6, David C. Linehan7, Brian A. Belt8, Éric Gamelin9, Bishu Ganguly9, Dong Yin9, Tenshang Joh9, Ira Jacobs9, Carrie Turich Taylor9, Maeve A. Lowery10
1Department of Medicine, Division of Hematology/Oncology, University of Rochester Medical Center School of Medicine & Dentistry, Rochester, NY, USA
2Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
3Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
4MGH Cancer Center, Division of Hematogy-Oncology, Massachusetts General Hospital, Boston, MA, USA
5Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
6Division of Hematology/ Oncology, Medical University of South Carolina, Charleston, SC, USA
7Department of Surgery, University of Rochester Medical Center School of Medicine & Dentistry, Rochester, NY, USA
8Department of Surgery, University of Rochester, Rochester, NY, USA
9Early Oncology Development and Clinical Research, Pfizer Inc, 219 East 42nd Street, New York, NY, 10017, USA
10Trinity St James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland

Tóm tắt

SummaryBackground In pancreatic ductal adenocarcinoma (PDAC), the chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-C motif) receptor 2 (CCR2) axis plays a key role in immunosuppressive properties of the tumor microenvironment, patient prognosis, and chemoresistance. This phase Ib study assessed the effects of the orally administered CCR2 inhibitor PF-04136309 in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic PDAC. Methods Patients received PF-04136309 twice daily (BID) continuously plus nab-paclitaxel (125 mg/m2) and gemcitabine (1000 mg/m2) administered on days 1, 8, and 15 of each 28-day cycle. The primary objectives were to evaluate safety and tolerability, characterize dose-limiting toxicities (DLTs), and determine the recommended phase II dose (RP2D) of PF-04136309. Results In all, 21 patients received PF-04136309 at a starting dose of 500 mg or 750 mg BID. The RP2D was identified to be 500 mg BID. Of 17 patients treated at the 500 mg BID starting dose, three (17.6%) experienced a total of four DLTs, including grade 3 dysesthesia, diarrhea, and hypokalemia and one event of grade 4 hypoxia. Relative to the small number of patients (n = 21), a high incidence (24%) of pulmonary toxicity was observed in this study. The objective response rate for 21 patients was 23.8% (95% confidence interval: 8.2–47.2%). Levels of CD14 + CCR2+ inflammatory monocytes (IM) decreased in the peripheral blood, but did not accumulate in the bone marrow. Conclusions PF-04136309 in combination with nab-paclitaxel plus gemcitabine had a safety profile that raises concern for synergistic pulmonary toxicity and did not show an efficacy signal above nab-paclitaxel and gemcitabine. ClinicalTrials.gov identifier: NCT02732938.

Từ khóa


Tài liệu tham khảo

Ryan DP, Hong TS, Bardeesy N (2014) Pancreatic adenocarcinoma. N Engl J Med 371:2140–2141. https://doi.org/10.1056/NEJMc1412266

Hidalgo M (2010) Pancreatic cancer. N Engl J Med 362:1605–1617. https://doi.org/10.1056/NEJMra0901557

Von Hoff DD, Ervin T, Arena FP et al (2013) Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369:1691–1703. https://doi.org/10.1056/NEJMoa1304369

Conroy T, Desseigne F, Ychou M et al (2011) FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364:1817–1825. https://doi.org/10.1056/NEJMoa1011923

Adamska A, Domenichini A, Falasca M (2017) Pancreatic ductal adenocarcinoma: current and evolving therapies. Int J Mol Sci 18. https://doi.org/10.3390/ijms18071338

Rodriguez D, Silvera R, Carrio R et al (2013) Tumor microenvironment profoundly modifies functional status of macrophages: peritoneal and tumor-associated macrophages are two very different subpopulations. Cell Immunol 283:51–60. https://doi.org/10.1016/j.cellimm.2013.06.008

Rolinski J, Hus I (2014) Breaking immunotolerance of tumors: a new perspective for dendritic cell therapy. J Immunotoxicol 11:311–318. https://doi.org/10.3109/1547691X.2013.865094

Sanford DE, Belt BA, Panni RZ et al (2013) Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res 19:3404–3415. https://doi.org/10.1158/1078-0432.CCR-13-0525

Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ (2009) Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother 58:49–59. https://doi.org/10.1007/s00262-008-0523-4

Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP (2007) Amino-biphosphonate-mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res 67:11438–11446. https://doi.org/10.1158/0008-5472.CAN-07-1882

Yang L, DeBusk LM, Fukuda K et al (2004) Expansion of myeloid immune suppressor gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 6:409–421. https://doi.org/10.1016/j.ccr.2004.08.031

Bunt SK, Yang L, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S (2007) Reduced inflammation in the tumor microenvironment delays the accumulation of myeloid-derived suppressor cells and limits tumor progression. Cancer Res 67:10019–10026. https://doi.org/10.1158/0008-5472.CAN-07-2354

Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand-Rosenberg S (2007) Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol 179:977–983

Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S (2010) Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 70:68–77. https://doi.org/10.1158/0008-5472.CAN-09-2587

Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9:162–174. https://doi.org/10.1038/nri2506

Marx J (2008) Cancer immunology. Cancer's bulwark against immune attack: MDS cells. Science 319:154–156. https://doi.org/10.1126/science.319.5860.154

Ostrand-Rosenberg S, Sinha P (2009) Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol 182:4499–4506. https://doi.org/10.4049/jimmunol.0802740

Ostrand-Rosenberg S (2010) Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol Immunother 59:1593–1600. https://doi.org/10.1007/s00262-010-0855-8

Huang B, Lei Z, Zhao J et al (2007) CCL2/CCR2 pathway mediates recruitment of myeloid suppressor cells to cancers. Cancer Lett 252:86–92. https://doi.org/10.1016/j.canlet.2006.12.012

Huang B, Pan PY, Li Q et al (2006) Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res 66:1123–1131. https://doi.org/10.1158/0008-5472.CAN-05-1299

Koga M, Kai H, Egami K et al (2008) Mutant MCP-1 therapy inhibits tumor angiogenesis and growth of malignant melanoma in mice. Biochem Biophys Res Commun 365:279–284. https://doi.org/10.1016/j.bbrc.2007.10.182

Nywening TM, Wang-Gillam A, Sanford DE et al (2016) Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol 17:651–662. https://doi.org/10.1016/S1470-2045(16)00078-4

Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM (2014) Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 74:2913–2921. https://doi.org/10.1158/0008-5472.CAN-14-0155

Pokorny AMJ, Chin VT, Nagrial AM, Yip D, Chantrill LA (2018) Metastatic pancreatic ductal adenocarcinoma: diagnosis and treatment with a view to the future. Intern Med J 48:637–644. https://doi.org/10.1111/imj.13810

Mitchem JB, Brennan DJ, Knolhoff BL et al (2013) Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res 73:1128–1141. https://doi.org/10.1158/0008-5472.CAN-12-2731

Chiorean EG, Coveler AL (2015) Pancreatic cancer: optimizing treatment options, new, and emerging targeted therapies. Drug Des Devel Ther 9:3529–3545. https://doi.org/10.2147/DDDT.S60328

Von Hoff DD, Ramanathan RK, Borad MJ et al (2011) Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 29:4548–4554. https://doi.org/10.1200/JCO.2011.36.5742

Sahin IH, Geyer AI, Kelly DW, O'Reilly EM (2016) Gemcitabine-related pneumonitis in pancreas adenocarcinoma–an infrequent event: elucidation of risk factors and management implications. Clin Colorectal Cancer 15:24–31. https://doi.org/10.1016/j.clcc.2015.08.003

Nywening TM, Belt BA, Cullinan DR et al (2018) Targeting both tumour-associated CXCR2(+) neutrophils and CCR2(+) macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 67:1112–1123. https://doi.org/10.1136/gutjnl-2017-313738