Safety, tolerability, pharmacokinetics, and pharmacodynamics of the afucosylated, humanized anti-EPHA2 antibody DS-8895a: a first-in-human phase I dose escalation and dose expansion study in patients with advanced solid tumors

Kohei Shitara, Taroh Satoh, Satoru Iwasa, Kensei Yamaguchi, Kei Muro, Yoshito Komatsu, Tomohiro Nishina, Taito Esaki, Jun Hasegawa, Yasuyuki Kakurai, Emi Kamiyama, Tomoko Nakata, Kota Nakamura, Hayato Sakaki, Ichinosuke Hyodo, Kohei Shitara, Taroh Satoh, Satoru Iwasa, Kensei Yamaguchi, Kei Muro, Yoshito Komatsu, Tomohiro Nishina, Taito Esaki, Jun Hasegawa, Yasuyuki Kakurai, Emi Kamiyama, Tomoko Nakata, Kota Nakamura, Hayato Sakaki, Ichinosuke Hyodo

Abstract

Background: Erythropoietin-producing hepatocellular receptor A2 (EPHA2) is overexpressed on the cell surface in many cancers and predicts poor prognosis. DS-8895a is a humanized anti-EPHA2 IgG1 monoclonal antibody afucosylated to enhance antibody-dependent cellular cytotoxicity activity. We conducted a two-step, phase I, multicenter, open-label study to determine the safety, tolerability, and pharmacokinetics of DS-8895a in patients with advanced solid tumors.

Methods: Step 1 was a dose escalation cohort in advanced solid tumor patients (six dose levels, 0.1-20 mg/kg) to determine Step 2 dosing. Step 2 was a dose expansion cohort in EPHA2-positive esophageal and gastric cancer patients. DS-8895a was intravenously administered every 2 weeks for the duration of the study, with a 28-day period to assess dose-limiting toxicity (DLT). Safety, pharmacokinetics, tumor response, and potential biomarkers were evaluated.

Results: Thirty-seven patients (Step 1: 22, Step 2: 15 [9: gastric cancer, 6: esophageal cancer]) were enrolled. Although one DLT (Grade 4 platelet count decreased) was observed in Step 1 (dose level 6, 20 mg/kg), the maximum tolerated dose was not reached; the highest dose (20 mg/kg) was used in Step 2. Of the 37 patients, 24 (64.9%) experienced drug-related adverse events (AEs) including three (8.1%) with Grade ≥ 3 AEs. Infusion-related reactions occurred in 19 patients (51.4%) but were manageable. All patients discontinued the study (evident disease progression, 33; AEs, 4). Maximum and trough serum DS-8895a concentrations increased dose-dependently. One gastric cancer patient achieved partial response and 13 patients achieved stable disease. Serum inflammatory cytokines transiently increased at completion of and 4 h after the start of DS-8895a administration. The proportion of CD16-positive natural killer (NK) cells (CD3-CD56+CD16+) decreased 4 h after the start of DS-8895a administration, and the ratio of CD3-CD56+CD137+ to CD3-CD56+CD16+ cells increased on day 3.

Conclusions: Twenty mg/kg DS-8895a infused intravenously every 2 weeks was generally safe and well tolerated in patients (n = 21) with advanced solid tumors. The exposure of DS-8895a seemed to increase dose-dependently and induce activated NK cells.

Trial registration: Phase 1 Study of DS-8895a in patients with advanced solid tumors ( NCT02004717 ; 7 November 2013 to 2 February 2017); retrospectively registered on 9 December 2013.

Keywords: Advanced solid tumors; Antibody-dependent cellular cytotoxicity; DS-8895a; Erythropoietin-producing hepatocellular receptor A2; Esophageal cancer; Gastric cancer; Phase I study.

Conflict of interest statement

Kohei Shitara has received consultation fees from Astellas Pharma, Lilly, Bristol-Myers Squibb, Takeda Pharmaceutical, Pfizer, and Ono Pharmaceutical; honoraria from Novartis, Abbvie, and Yakult Honsha; and research funding from Dainippon Sumitomo Pharma, Eli Lilly, MSD, Daiichi Sankyo, Taiho Pharmaceutical, Chugai Pharmaceutical, and Ono Pharmaceutical.

Taroh Satoh has received consultation fees from Bayer Yakuhin, Ltd., Eli Lilly, Ono Pharmaceutical, Takara Bio, and Merck Serono; honoraria from Chugai Pharmaceutical, Merck Serono, Bristol-Myers Squibb, Takeda Pharmaceutical, Yakult Honsha, Eli Lilly, Bayer Yakuhin, Ono Pharmaceutical, Merck, and Astellas Pharma; and research funding from Yakult Honsha, Chugai Pharmaceutical, Ono Pharmaceutical, Sanofi, Eli Lilly, Daiichi Sankyo, Merck, Merck Serono, Gilead Sciences, and Dainippon Sumitomo Pharma.

Satoru Iwasa has received honoraria from Taiho Pharmaceutical, Lilly Japan and Chugai Pharma; and research funding from Daiichi Sankyo, Chugai Pharma, Bristol-Myers Squibb, Lilly Japan, Eisai, Novartis, Merck Serono, Otsuka and Bayer.

Kensei Yamaguchi has received consultation fees from Bristol-Myers Squibb Japan and Daiichi Sankyo; speaker’s bureau funds from Chugai Pharma, Merck Serono, Bristol-Myers Squibb Japan, Takeda, Taiho Pharmaceutical, Lilly, Yakult Honsha, Ono Pharmaceutical and Sanofi; and research funding from MSD Oncology, Ono Pharmaceutical, Dainippon Sumitomo Pharma, Taiho Pharmaceutical, Daiichi Sankyo, Lilly, Gilead Sciences and Yakult Honsha.

Kei Muro has received honoraria from Chugai Pharma, Merck Serono, Takeda, Taiho Pharmaceutical, Lilly, Yakult Honsha, Ono Pharmaceutical and Bayer; and research funding from Ono Pharmaceutical, MSD, Daiichi Sankyo, Shionogi, Kyowa Hakko Kirin, Gilead Sciences, Merck Serono, Pfizer and Sanofi.

Yoshito Komatsu has received speaker’s bureau funds from Taiho Pharmaceutical, Lilly, Chugai Pharma, Merck Serono, Novartis, Pfizer, Bayer, Ono Pharmaceutical, Yakult Honsha, Takeda, Sanofi, Bristol-Myers Squibb Japan, Daiichi Sankyo, TOWA and Nipro Corporation; honoraria from Novartis, Pfizer and Bayer; and research funding from Taiho Pharmaceutical, Lilly, MSD, Ono Pharmaceutical, Novartis, Bayer, Chugai Pharma, Yakult, Takeda, Dainippon Sumitomo Pharma, Boehringer Ingelheim, Sysmex, Sanofi, Astellas Pharma and Eisai.

Tomohiro Nishina has received honoraria from Taiho Pharmaceutical, Chugai Pharma, Lilly, Merck Serono and Takeda; and research funding from Taiho Pharmaceutical, Chugai Pharma, Dainippon Sumitomo Pharma, Lilly Japan, Merck Serono, MSD, Daiichi Sankyo, and Ono Pharmaceutical.

Taito Esaki has received consultation fees from Chugai Pharma; speakers’ bureau funds from Taiho Pharmaceutical, Bristol-Myers Squibb Japan, Lilly, Eisai, Daiichi Sankyo, Merck Serono, Chugai Pharma, Ono Pharmaceutical and Takeda; honoraria from Lilly and Kyowa Hakko Kirin; and research funding from Daiichi Sankyo, Merck Serono, Taiho Pharmaceutical, MSD, Novartis, Dainippon Sumitomo Pharma and Ono Pharmaceutical.

Jun Hasegawa, Yasuyuki Kakurai, Emi Kamiyama, Tomoko Nakata, Kota Nakamura and Hayato Sakaki are employees of Daiichi Sankyo.

Ichinosuke Hyodo has received honoraria from Chugai Pharma, Taiho Pharmaceutical, Daiichi Sankyo, Lilly, Ono Pharmaceutical and Bristol-Myers Squibb; and research funding from Bristol-Myers Squibb Japan, Chugai Pharma, Kyowa Hakko Kirin, Takeda, Merck Serono, Yakult Honsha, Taiho Pharmaceutical, Sanofi, Daiichi Sankyo and Ono Pharmaceutical.

Figures

Fig. 1
Fig. 1
Best (minimum) percent change from baseline in sum of diameters (%) of target lesions. Baseline is defined as the last measurement prior to administration of the first dose of DS-8895a. Each vertical bar represents the best (minimum) percent change from baseline for an individual patient
Fig. 2
Fig. 2
Changes in CD16-positive natural killer (NK) cells. Time-course of circulating CD16-positive NK cells (CD3−CD56+CD16+) after DS-8895a treatment in a) Cycle 1 of Step 1 and b) Cycle 1 of Step 2, and c) the ratio of CD3−CD56+CD137+ cells to CD3−CD56+CD16+ cells in Cycle 1 of Step 2

References

    1. Tandon M, Vemula SV, Mittal SK. Emerging strategies for EphA2 receptor targeting for cancer therapeutics. Expert Opin Ther Targets. 2011;15:31–51. doi: 10.1517/14728222.2011.538682.
    1. Miao H, Wang B. EphA receptor signaling-complexity and emerging themes. Semin Cell Dev Biol. 2012;23:16–25. doi: 10.1016/j.semcdb.2011.10.013.
    1. Pasquale EB. Eph-ephrin bidirectional signaling in physiology and disease. Cell. 2008;133:38–52. doi: 10.1016/j.cell.2008.03.011.
    1. Hafner Christian, Becker Bernd, Landthaler Michael, Vogt Thomas. Expression profile of Eph receptors and ephrin ligands in human skin and downregulation of EphA1 in nonmelanoma skin cancer. Modern Pathology. 2006;19(10):1369–1377. doi: 10.1038/modpathol.3800660.
    1. Hou F, Yuan W, Huang J, et al. Overexpression of EphA2 correlates with epithelial–mesenchymal transition-related proteins in gastric cancer and their prognostic importance for postoperative patients. Med Oncol. 2012;29:2691–2700. doi: 10.1007/s12032-011-0127-2.
    1. Hafner C, Schmitz G, Meyer S, et al. Differential gene expression of Eph receptors and ephrins in benign human tissues and cancers. Clin Chem. 2004;50(3):490–499. doi: 10.1373/clinchem.2003.026849.
    1. Saito T, Masuda N, Miyazaki T, et al. Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol Rep. 2004;11:605–611.
    1. Abraham S, Knapp DW, Cheng L, et al. Expression of EphA2 and Ephrin A-1 in carcinoma of the urinary bladder. Clin Cancer Res. 2006;12:353–360. doi: 10.1158/1078-0432.CCR-05-1505.
    1. Dunne PD, Dasgupta S, Blayney JK, et al. EphA2 expression is a key driver of migration and invasion and a poor prognostic marker in colorectal cancer. Clin Cancer Res. 2016;22:230–242. doi: 10.1158/1078-0432.CCR-15-0603.
    1. Zelinski DP, Zantek ND, Stewart JC, Irizarry AR, Kinch MS. EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res. 2001;61:2301–2306.
    1. Easty DJ, Guthrie BA, Maung K, et al. Protein B61 as a new growth factor: expression of B61 and up-regulation of its receptor epithelial cell kinase during melanoma progression. Cancer Res. 1995;55:2528–2532.
    1. Wykosky J, Gibo DM, Stanton C, Debinski W. EphA2 as a novel molecular marker and target in glioblastoma multiforme. Mol Cancer Res. 2005;3:541–551. doi: 10.1158/1541-7786.MCR-05-0056.
    1. Brannan JM, Dong W, Prudkin L, et al. Expression of the receptor tyrosine kinase EphA2 is increased in smokers and predicts poor survival in non-small cell lung cancer. Clin Cancer Res. 2009;15:4423–4430. doi: 10.1158/1078-0432.CCR-09-0473.
    1. Miyazaki Tatsuya, Kato Hiroyuki, Fukuchi Minoru, Nakajima Masanobu, Kuwano Hiroyuki. EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. International Journal of Cancer. 2002;103(5):657–663. doi: 10.1002/ijc.10860.
    1. Biao-xue RONG, Xi-guang CAI, Shuan-ying YANG, Wei LI, Zong-juan MING. EphA2-Dependent Molecular Targeting Therapy for Malignant Tumors. Current Cancer Drug Targets. 2011;11(9):1082–1097. doi: 10.2174/156800911798073050.
    1. Merritt William M., Kamat Aparna A., Hwang Jee-Young, Bottsford-Miller Justin, Lu Chunhua, Lin Yvonne G., Coffey Donna, Spannuth Whitney A., Nugent Elizabeth, Han Liz Y., Landen Charles N., Nick Alpa M., Stone Rebecca L., Coffman Karen, Bruckheimer Elizabeth, Broaddus Russell R., Gershenson David M., Coleman Robert L., Sood Anil K. Clinical and biological impact of EphA2 overexpression and angiogenesis in endometrial cancer. Cancer Biology & Therapy. 2010;10(12):1306–1314. doi: 10.4161/cbt.10.12.13582.
    1. Yang P, Yuan W, He J, et al. Overexpression of EphA2, MMP-9, and MVD-CD34 in hepatocellular carcinoma: implications for tumor progression and prognosis. Hepatol Res. 2009;39:1169–1177. doi: 10.1111/j.1872-034X.2009.00563.x.
    1. Neill T, Buraschi S, Goyal A, et al. EphA2 is a functional receptor for the growth factor progranulin. J Cell Biol. 2016;215:687–703. doi: 10.1083/jcb.201603079.
    1. Zhou Y, Sakurai H. Emerging and diverse functions of the EphA2 noncanonical pathway in cancer progression. Biol Pharm Bull. 2017;40:1616–1624. doi: 10.1248/bpb.b17-00446.
    1. Macrae M, Neve RM, Rodriguez-Viciana P, et al. A conditional feedback loop regulates Ras activity through EphA2. Cancer Cell. 2005;8:111–118. doi: 10.1016/j.ccr.2005.07.005.
    1. Kubota Tsuguo, Niwa Rinpei, Satoh Mitsuo, Akinaga Shiro, Shitara Kenya, Hanai Nobuo. Engineered therapeutic antibodies with improved effector functions. Cancer Science. 2009;100(9):1566–1572. doi: 10.1111/j.1349-7006.2009.01222.x.
    1. Lieberman J. The ABCs of granule-mediated cytotoxicity: new weapons in the arsenal. Nat Rev Immunol. 2003;3:361–370. doi: 10.1038/nri1083.
    1. Hasegawa J, Sue M, Yamato M, et al. Novel anti-EPHA2 antibody, DS-8895a for cancer treatment. Cancer Biol Ther. 2016;17:1158–1167. doi: 10.1080/15384047.2016.1235663.
    1. Velders MP, van Rhijn CM, Oskam E, Fleuren GJ, Warnaar SO, Litvinov SV. The impact of antigen density and antibody affinity on antibody-dependent cellular cytotoxicity: relevance for immunotherapy of carcinomas. British Journal of Cancer. 1998;78(4):478–483. doi: 10.1038/bjc.1998.518.
    1. Ministry of Education, Culture, Sports, Science and Technology, Ministry of Health, Labour and Welfare, and Ministry of Economy, Trade and Industry. Ethical Guidelines for Human Genome/Gene Analysis Research (issued on March 29, 2001, fully revised on December 28, 2004, partially revised on June 29, 2005, partially revised on December 1, 2008, and fully revised on February 8, 2013). . Accessed 11 Dec 2018.
    1. Ministry of Health, Labour and Welfare. Ethical Guideline for Clinical Research (issued on July 30, 2003, fully revised on December 28, 2004, and fully revised on July 31, 2008).
    1. Delgado DC, Hank JA, Kolesar J, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcγ receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res. 2010;70(23):9554–9561. doi: 10.1158/0008-5472.CAN-10-2211.
    1. Guidelines for the clinical evaluation of anti-cancer drugs, notification no. 1101001 of the evaluation and licensing division, pharmaceutical and food safety bureau (November 1, 2005).
    1. Paz-Ares LG, Gomez-Roca C, Delord JP, et al. Phase I pharmacokinetic and pharmacodynamic dose-escalation study of RG7160 (GA201), the first glycoengineered monoclonal antibody against the epidermal growth factor receptor, in patients with advanced solid tumors. J Clin Oncol. 2011;29:3783–3790. doi: 10.1200/JCO.2011.34.8888.
    1. Delord JP, Tabernero J, García-Carbonero R, et al. Open-label, multicenter expansion cohort to evaluate imgatuzumab in pre-treated patients with KRAS-mutant advanced colorectal carcinoma. Eur J Cancer. 2014;50:496–505. doi: 10.1016/j.ejca.2013.10.015.
    1. Duvic M, Pinter-Brown LC, Foss FM, et al. Phase 1/2 study of mogamulizumab, a defucosylated anti-CCR4 antibody, in previously treated patients with cutaneous T-cell lymphoma. Blood. 2015;125(2):1883–1889. doi: 10.1182/blood-2014-09-600924.
    1. Shitara K, Ueha S, Shichino S, et al. First-in-human phase 1 study of IT1208, a defucosylated humanized anti-CD4 depleting antibody, in patients with advanced solid tumors. Society for Immunotherapy of Cancer 2018. Poster #P300 presented at SITC 2018. Washington, D.C; 2018.
    1. Kohrt HE, Houot R, Weiskopf K, et al. Simulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer. J Clin Invest. 2012;122:1066–1075. doi: 10.1172/JCI61226.
    1. Kohrt HE, Houot R, Goldstein MJ, et al. CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies. Blood. 2011;117:2423–2432. doi: 10.1182/blood-2010-08-301945.
    1. Wang Lin, Wei Yingfeng, Fang Weijia, Lu Chong, Chen Jianing, Cui Guangying, Diao Hongyan. Cetuximab Enhanced the Cytotoxic Activity of Immune Cells during Treatment of Colorectal Cancer. Cellular Physiology and Biochemistry. 2017;44(3):1038–1050. doi: 10.1159/000485404.
    1. Tarek N, Le Luduec JB, Gallagher MM, et al. Unlicensed NK cells target neuroblastoma following anti-GD2 antibody treatment. J Clin Invest. 2012;122:3260–3270. doi: 10.1172/JCI62749.

Source: PubMed

3
订阅