Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN Solid Tumor study

Luc Y Dirix, Istvan Takacs, Guy Jerusalem, Petros Nikolinakos, Hendrik-Tobias Arkenau, Andres Forero-Torres, Ralph Boccia, Marc E Lippman, Robert Somer, Martin Smakal, Leisha A Emens, Borys Hrinczenko, William Edenfield, Jayne Gurtler, Anja von Heydebreck, Hans Juergen Grote, Kevin Chin, Erika P Hamilton, Luc Y Dirix, Istvan Takacs, Guy Jerusalem, Petros Nikolinakos, Hendrik-Tobias Arkenau, Andres Forero-Torres, Ralph Boccia, Marc E Lippman, Robert Somer, Martin Smakal, Leisha A Emens, Borys Hrinczenko, William Edenfield, Jayne Gurtler, Anja von Heydebreck, Hans Juergen Grote, Kevin Chin, Erika P Hamilton

Abstract

Purpose: Agents targeting programmed death receptor 1 (PD-1) or its ligand (PD-L1) have shown antitumor activity in the treatment of metastatic breast cancer (MBC). The aim of this study was to assess the activity of avelumab, a PD-L1 inhibitor, in patients with MBC.

Methods: In a phase 1 trial (JAVELIN Solid Tumor; NCT01772004), patients with MBC refractory to or progressing after standard-of-care therapy received avelumab intravenously 10 mg/kg every 2 weeks. Tumors were assessed every 6 weeks by RECIST v1.1. Adverse events (AEs) were graded by NCI-CTCAE v4.0. Membrane PD-L1 expression was assessed by immunohistochemistry (Dako PD-L1 IHC 73-10 pharmDx).

Results: A total of 168 patients with MBC, including 58 patients with triple-negative breast cancer (TNBC), were treated with avelumab for 2-50 weeks and followed for 6-15 months. Patients were heavily pretreated with a median of three prior therapies for metastatic or locally advanced disease. Grade ≥ 3 treatment-related AEs occurred in 13.7% of patients, including two treatment-related deaths. The confirmed objective response rate (ORR) was 3.0% overall (one complete response and four partial responses) and 5.2% in patients with TNBC. A trend toward a higher ORR was seen in patients with PD-L1+ versus PD-L1- tumor-associated immune cells in the overall population (16.7% vs. 1.6%) and in the TNBC subgroup (22.2% vs. 2.6%).

Conclusion: Avelumab showed an acceptable safety profile and clinical activity in a subset of patients with MBC. PD-L1 expression in tumor-associated immune cells may be associated with a higher probability of clinical response to avelumab in MBC.

Keywords: Avelumab; Metastatic breast cancer; PD-L1; Second-line; Triple-negative breast cancer.

Conflict of interest statement

GJ Reports honoria from Novartis, Celgene, and Roche; consulting/advisory roles for Novartis and Celgene; research funding from Novartis, MSD, and Roche; travel, accommodations, expenses from Novartis, Roche, and GSK. AFT reports research funding (to institution) from Pfizer. RB reports honoraria from Amgen, Celgene, AstraZeneca, and Tesaro; consulting/advisory role, speaker’s bureau fees, and travel, accommodations, and expenses for Heron Therapeutics. ML reports a leadership role and stock or other ownership for Seattle Genetics, Oncology analytics, and Geneyus. LE reports travel, accommodations, or expenses from Bayer Pharmaceuticals; consulting/advisory roles for Celgene, Vaccinex, Amgen, AstraZeneca, Syndax, Peregrine, eTHeRNA, Bayer, Molecuvax, and Gritstone; research funding from Genentech, Roche, EMD Serono, MaxCyte, MSD, AstraZeneca, Aduro Biotech, and Corvus; and is an SGE to the FDA. BH reports consulting/advisory roles for Bayer, Gilead, and Biotheranostics and research funding from EMD Serono, MedImmune, and AstraZeneca. WE participated in speakers’ bureau for Astellas and Medivation. JG reports research funding from EMD Serono. AvH and HJG are employees at Merck KGaA, Darmstadt, Germany. KC is an employee of EMD Serono, Inc., a subsidiary of Merck KGaA, Darmstadt, Germany. EH reports research funding (to institution) from Sarah Cannon Research Institute. All other authors have nothing to disclose.

Figures

Fig. 1
Fig. 1
Time to and duration of response for patients with confirmed or unconfirmed responses
Fig. 2
Fig. 2
Best change in target lesions from baseline in 140 evaluable patients with baseline tumor assessment and ≥ 1 post-baseline assessment
Fig. 3
Fig. 3
Percent change in target lesions from baseline in 46 evaluable patients with TNBC with baseline tumor assessment and ≥ 1 post-baseline assessment

References

    1. O’Shaughnessy J. Extending survival with chemotherapy in metastatic breast cancer. Oncologist. 2005;10(Suppl 3):20–29. doi: 10.1634/theoncologist.10-90003-20.
    1. National Cancer Institute . SEER cancer stat facts: female breast cancer. Bethesda: National Cancer Institute; 2016. p. 2016.
    1. National Comprehensive Cancer Network (V2 2017) NCCN clinical practice guidelines in oncology: breast cancer. Orlando: National Comprehensive Cancer Network; 2017.
    1. Dent R, Trudeau M, Pritchard KI, et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007;13:4429–4434. doi: 10.1158/1078-0432.CCR-06-3045.
    1. Anders CK, Abramson V, Tan T, et al. The evolution of triple-negative breast cancer: from biology to novel therapeutics. Am Soc Clin Oncol Educ Book. 2016;35:34–42. doi: 10.14694/EDBK_159135.
    1. Andre F, Zielinski CC. Optimal strategies for the treatment of metastatic triple-negative breast cancer with currently approved agents. Ann Oncol. 2012;23:46–51. doi: 10.1093/annonc/mdr047.
    1. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol. 2015;33:1974–1982. doi: 10.1200/JCO.2014.59.4358.
    1. Mittendorf EA, Philips AV, Meric-Bernstam F, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res. 2014;2:361–370. doi: 10.1158/2326-6066.CIR-13-0127.
    1. Schalper KA, Velcheti V, Carvajal D, et al. In situ tumor PD-L1 mRNA expression is associated with increased TILs and better outcome in breast carcinomas. Clin Cancer Res. 2014;20:2773–2782. doi: 10.1158/1078-0432.CCR-13-2702.
    1. Wimberly H, Brown JR, Schalper K, et al. PD-L1 expression correlates with tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy in breast cancer. Cancer Immunol Res. 2015;3:326–332. doi: 10.1158/2326-6066.CIR-14-0133.
    1. Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730.
    1. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. doi: 10.1084/jem.192.7.1027.
    1. Iwai Y, Ishida M, Tanaka Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 2002;99:12293–12297. doi: 10.1073/pnas.192461099.
    1. Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer. 2016;4:59. doi: 10.1186/s40425-016-0165-6.
    1. Cimino-Mathews A, Thompson E, Taube JM, et al. PD-L1 (B7-H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. Hum Pathol. 2016;47:52–63. doi: 10.1016/j.humpath.2015.09.003.
    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–461. doi: 10.1016/j.ccell.2015.03.001.
    1. Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol. 2016;34:2460–2467. doi: 10.1200/JCO.2015.64.8931.
    1. Emens L, Braiteh F, Cassier P, et al. Inhibition of PD-L1 by MPDL3280A leads to clinical activity in patients with metastatic triple-negative breast cancer (TNBC) Cancer Res. 2015;75:2859. doi: 10.1158/1538-7445.AM2015-2859.
    1. Schmid P, Cruz C, Braiteh FS, et al. Atezolizumab in metastatic TNBC (mTNBC): long-term clinical outcomes and biomarker analysis. Cancer Res. 2017;77:2986. doi: 10.1158/1538-7445.AM2017-2986.
    1. Heery CR, O’Sullivan-Coyne G, Madan RA, et al. Avelumab for metastatic or locally advanced previously treated solid tumours (JAVELIN Solid Tumor): a phase 1a, multicohort, dose-escalation trial. Lancet Oncol. 2017;18:587–597. doi: 10.1016/S1470-2045(17)30239-5.
    1. Boyerinas B, Jochems C, Fantini M, et al. Antibody-dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res. 2015;3:1148–1157. doi: 10.1158/2326-6066.CIR-15-0059.
    1. Fujii R, Friedman ER, Richards J, et al. Enhanced killing of chordoma cells by antibody-dependent cell-mediated cytotoxicity employing the novel anti-PD-L1 antibody avelumab. Oncotarget. 2016;7:33498–33511.
    1. Grenga I, Donahue RN, Lepone LM, et al. A fully human IgG1 anti-PD-L1 MAb in an in vitro assay enhances antigen-specific T-cell responses. Clin Transl Immunol. 2016;5:e83. doi: 10.1038/cti.2016.27.
    1. Kaufman HL, Russell J, Hamid O, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. 2016;17:1374–1385. doi: 10.1016/S1470-2045(16)30364-3.
    1. Gulley JL, Rajan A, Spigel DR, et al. Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN Solid Tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial. Lancet Oncol. 2017;18:599–610. doi: 10.1016/S1470-2045(17)30240-1.
    1. Apolo AB, Infante JR, Balmanoukian A, et al. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, phase Ib study. J Clin Oncol. 2017;35:2117–2124. doi: 10.1200/JCO.2016.71.6795.
    1. Bavencio (avelumab) injection [package insert] Darmstadt. Darmstadt: Merck KGaA; 2017.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Donahue RN, Lepone LM, Grenga I, et al. Analyses of the peripheral immunome following multiple administrations of avelumab, a human IgG1 anti-PD-L1 monoclonal antibody. J Immunother Cancer. 2017;5:13. doi: 10.1186/s40425-017-0220-y.
    1. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–856. doi: 10.1158/1535-7163.MCT-14-0983.
    1. Wang X, Teng F, Kong L, et al. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016;9:5023–5039. doi: 10.2147/OTT.S105862.
    1. Hirsch FR, McElhinny A, Stanforth D, et al. PD-L1 Immunohistochemistry assays for lung cancer: results from phase 1 of the “blueprint PD-L1 IHC assay comparison project”. J Thorac Oncol. 2016;12:208–222. doi: 10.1016/j.jtho.2016.11.2228.
    1. Adams S, Diamond J, Hamiton E, et al. Phase Ib trial of atezolizumab in combination with nab-paclitaxel in patients with metastatic triple-negative breast cancer (mTNBC) J Clin Oncol. 2016;34:1009.
    1. Tolaney S, Savulsky C, Aktan G, et al. Phase 1b/2 study to evaluate eribulin mesylate in combination with pembrolizumab in patients with metastatic triple-negative breast cancer. Eur J Cancer. 2017;72:S16. doi: 10.1016/S0959-8049(17)30131-4.

Source: PubMed

3
Tilaa