A phase 1b study of andecaliximab in combination with S-1 plus platinum in Japanese patients with gastric adenocarcinoma

Akira Ooki, Taroh Satoh, Kei Muro, Atsuo Takashima, Shigenori Kadowaki, Daisuke Sakai, Takashi Ichimura, Seiichiro Mitani, Toshihiro Kudo, Keisho Chin, Shigehisa Kitano, Dung Thai, Marianna Zavodovskaya, JieJane Liu, Narikazu Boku, Kensei Yamaguchi, Akira Ooki, Taroh Satoh, Kei Muro, Atsuo Takashima, Shigenori Kadowaki, Daisuke Sakai, Takashi Ichimura, Seiichiro Mitani, Toshihiro Kudo, Keisho Chin, Shigehisa Kitano, Dung Thai, Marianna Zavodovskaya, JieJane Liu, Narikazu Boku, Kensei Yamaguchi

Abstract

Andecaliximab (ADX) is a monoclonal antibody that inhibits matrix metalloproteinase 9 (MMP9), an extracellular enzyme involved in matrix remodeling, tumor growth, and metastasis. In preclinical models, MMP9 inhibitors have been shown to enhance the cytotoxic effects of chemotherapeutic agents and to suppress distant metastasis. In this phase Ib, multicenter study, the safety and efficacy of ADX combined with S-1 plus cisplatin (SP) or S-1 plus oxaliplatin (SOX) as a first-line treatment were evaluated in Japanese patients with advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma. ADX was administrated at a dose of 800 mg every 2 weeks for the SP cohort and 1200 mg every three weeks for the SOX cohort. As of December 2019, 16 patients were enrolled (six patients in the SP cohort and 10 patients in the SOX cohort). Peripheral sensory neuropathy (69%), anorexia (63%), nausea (56%), and decreased neutrophil counts (44%) were the most common adverse events (AEs). The grade 3 or higher AEs attributed to ADX were stomatitis and abnormal hepatic function (each one patient) in the SP cohort and decreased neutrophil counts (two patients) in the SOX cohort. The objective response rate in 11 patients with measurable target lesions was 73% (8/11), based on the investigator's evaluation. Median progression-free survival was11.9 months (90% confidence interval, 5.6-16.6), and median overall survival was not reached. In conclusion, ADX combined with S-1 plus platinum demonstrated a manageable safety profile and promising clinical activity in the first-line treatment of patients with advanced gastric or GEJ adenocarcinoma.Clinical Trial Registration information: ClinicalTrials.gov Identifier: NCT02862535 (11/08/2016) and protocol ID: GS-US-296-1884.

Conflict of interest statement

KY received speaker honoraria from Chugai Pharmaceutical Co. Ltd., Bristol-Myers Squibb, Merck Serono, Taiho Pharmaceutical Co., Takeda, and Eli Lilly; a consultant fee from Takeda Pharmaceutical Co. Ltd.; honoraria from Tsumura Co. Ltd., Nihon Kayaku Co. Ltd., and Chugai Pharmaceutical Co. Ltd; research grants from Sumitomo Dainippon Pharma, Gilead Sciences, MSD, and Boehringer Ingelheim, Daiichi Sankyo, and Chugai Pharmaceutical Co. Ltd; Speaker honoraria, research grants, and scholarship grant from Ono Pharmaceutical, Yakult Honsha Co., Ltd., and Sanofi. AO received speaker honoraria from Merck Serono, Chugai, Takeda Pharmaceutical Co. Ltd., Taiho Pharmaceutical Co., Bristol Myers-Squib, Daiichi Sankyo, and Ono Pharmaceutical. TS belonged to Endosed Department of Yakult Honsha Co., Ltd., Chugai Pharmaceutical Co., Ltd., and Ono pharmaceutical Co., Ltd.received speaker honoraria from Chugai Pharmaceutical Co. Ltd.Daiichi Sankyo Ono Pharmaceutical. Bristol-Myers Squibb, Merck Serono and Elli Lilly, a consultant fee from Takara Bio. TK research fund from Chugai Pharmaceutical Co. Ltd. Daiichi Sankyo Ono Pharmaceutical. Bristol-Myers Squibb, Parexell, Hutchmed, Giliad, BeiGene and Elli Lilly. NB has received research grant from Ono pharmaceutical company and Takeda pharmaceutical company, and honorarium from Ono pharmaceutical company, Taiho, ONO Pharmaceutical. Co. Ltd and Bristol Myers Squibb. KM received grants (to his institution and himself) from Astellas, Amgen, Daiichi Sankyo, Merck Sharp & Dohme Corp., Merck Serono, Ono Pharmaceutical Co., Ltd, Parexel International, Pfizer, Sanofi, Solasia Pharma, and Taiho Pharmaceutical Co; consulting fees from Amgen, AstraZeneca, and Ono Pharmaceutical Co., Ltd; honoraria from Bayer, Bristol-Myers Squibb, Chugai, Eli Lilly, Ono Pharmaceutical Co., Ltd, Sanofi, Taiho Pharmaceutical Co, and Takeda. SK received honoraria for lectures from Eli Lilly, Taiho Pharmaceutical Co., ONO Pharmaceutical. Co. Ltd, Bristol Myers-Squib, Chugai Pharmaceutical Co. Ltd, Bayer, Merck Serono, Eisai, and Daiichi-Sankyo; departmental research grants from Taiho Pharmaceutical Co., Eli Lilly, MSD, Chugai Pharmaceutical Co. Ltd, Nobelpharma, Daiichi Sankyo, ONO Pharmaceutical. Co. Ltd, and Yansen. AT received personal fees from Eli Lilly, Taiho Pharmaceutical, Chugai Pharma, and Merck Serono; grants from Merck Sharp & Dohme, Eisai, Bayer Yakuhin, Bristol-Myers Squibb, and BeiGene Japan; grants and personal fees from Ono Pharmaceutical and Takeda. TK belonged to a donated fund laboratory of Yakult Honsha Co., Ltd., Chugai Pharmaceutical Co., Ltd., and Ono pharmaceutical Co., Ltd. SM received speaker honoraria from Taiho Pharmaceutical Co., Takeda Pharmaceutical Co., and Ono Pharmaceutical Co, and grants from Taiho Pharmaceutical Co. SK received personal fees from Astra Zeneca, Pfizer, Taiho, Novartis, Sumitomo Dainippon Pharma, Bristol-Myers Squibb, AYUMI Pharmaceutical Corporation, Rakuten Medical, GSK, ImmuniT Research Inc., and PMDA(Pharmaceuticals and Medical Devices Agency) outside the submitted work; grants and personal fees from Gilead Sciences, Chugai, Boehringer Ingelheim, MSD, Eisai, Ono Pharmaceutical Co., Ltd, REGENERON, and Daiichi Sankyo; grants from Astellas, PACT Pharma, Takara Bio Inc., grants from AMED (Japan Agency for Medical Research and Development), and JSPS (Japan Society for the Promotion of Science).

© 2022. The Author(s).

Figures

Figure 1
Figure 1
Kaplan–Meier curve of PFS when cohorts 2 and 3 were combined. Kaplan–Meier curve was estimated from cut-off data of December 2018. The median PFS was 12.5 months (90% CI 5.6–14.7). After follow up at the data cut-off date (December, 2019), the median PFS was 11.9 months (90% CI 5.6–16.6).
Figure 2
Figure 2
Kaplan–Meier curve of OS when cohorts 2 and 3 were combined.

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018;68:394–424. doi: 10.3322/caac.21492.
    1. Balakrishnan M, George R, Sharma A, Graham DY. Changing trends in stomach cancer throughout the world. Curr. Gastroenterol. Rep. 2017;19:36. doi: 10.1007/s11894-017-0575-8.
    1. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology, Gastric Cancer, Version 2. (2022). .
    1. Muro K, Van Cutsem E, Narita Y, et al. Pan-Asian adapted ESMO Clinical Practice Guidelines for the management of patients with metastatic gastric cancer: A JSMO-ESMO initiative endorsed by CSCO, KSMO, MOS, SSO and TOS. Ann. Oncol. 2019;30:19–33. doi: 10.1093/annonc/mdy502.
    1. Wagner AD, Syn NL, Moehler M, et al. Chemotherapy for advanced gastric cancer. Cochrane Database Syst. Rev. 2017;8:004064.
    1. Moreira AM, Pereira J, Melo S, et al. The extracellular matrix: An accomplice in gastric cancer development and progression. Cells. 2020;9:394. doi: 10.3390/cells9020394.
    1. Yang Q, Ye ZY, Zhang JX, Tao HQ, Li SG, Zhao ZS. Expression of matrix metalloproteinase-9 mRNA and vascular endothelial growth factor protein in gastric carcinoma and its relationship to its pathological features and prognosis. Anat. Rec. 2010;293:2012–2019. doi: 10.1002/ar.21071.
    1. Shah MA, Starodub A, Sharma S, et al. Andecaliximab/GS-5745 alone and combined with mFOLFOX6 in advanced gastric and gastroesophageal junction adenocarcinoma: results from a phase I study. Clin. Cancer Res. 2018;24:3829–3837. doi: 10.1158/1078-0432.CCR-17-2469.
    1. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: Structure, function, and biochemistry. Circ. Res. 2003;92:827–839. doi: 10.1161/01.RES.0000070112.80711.3D.
    1. Bramhall SR, Hallissey MT, Whiting J, et al. Marimastat as maintenance therapy for patients with advanced gastric cancer: A randomised trial. Br. J. Cancer. 2002;86:1864–1870. doi: 10.1038/sj.bjc.6600310.
    1. Baragaño Raneros A, Suarez-Álvarez B, López-Larrea C. Secretory pathways generating immunosuppressive NKG2D ligands: New targets for therapeutic intervention. Oncoimmunology. 2014;3:e28497. doi: 10.4161/onci.28497.
    1. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell. 2010;141:52–67. doi: 10.1016/j.cell.2010.03.015.
    1. Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP. 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. 2007;67:11438–11446. doi: 10.1158/0008-5472.CAN-07-1882.
    1. Liu L, Ye Y, Zhu X. MMP-9 secreted by tumor associated macrophages promoted gastric cancer metastasis through a PI3K/AKT/Snail pathway. Biomed. Pharmacother. 2019;117:109096. doi: 10.1016/j.biopha.2019.109096.
    1. Oliveira MJ, Costa AC, Costa AM, et al. Helicobacter pylori induces gastric epithelial cell invasion in a c-Met and type IV secretion system-dependent manner. J. Biol. Chem. 2006;281:34888–34896. doi: 10.1074/jbc.M607067200.
    1. Kitadai Y, Sasaki A, Ito M, et al. Helicobacter pylori infection influences expression of genes related to angiogenesis and invasion in human gastric carcinoma cells. Biochem. Biophys. Res. Commun. 2003;311:809–814. doi: 10.1016/j.bbrc.2003.10.077.
    1. Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 1984;1:1311–1315. doi: 10.1016/S0140-6736(84)91816-6.
    1. Chen J, Chen LJ, Zhou HC, et al. Prognostic value of matrix metalloproteinase-9 in gastric cancer: A meta-analysis. Hepatogastroenterology. 2014;61:518–524.
    1. Jia X, Lu M, Rui C, Xiao Y. Consensus-expressed CXCL8 and MMP9 identified by meta-analyzed perineural invasion gene signature in gastric cancer microarray data. Front. Genet. 2019;10:851. doi: 10.3389/fgene.2019.00851.
    1. Yao Z, Yuan T, Wang H, et al. MMP-2 together with MMP-9 overexpression correlated with lymph node metastasis and poor prognosis in early gastric carcinoma. Tumour Biol. 2017;39:1010428317700411.
    1. Wroblewski LE, Pritchard DM, Carter S, Varro A. Gastrin-stimulated gastric epithelial cell invasion: The role and mechanism of increased matrix metalloproteinase 9 expression. Biochem. J. 2002;365:873–879. doi: 10.1042/bj20020068.
    1. Chang X, Xu X, Xue X, et al. NDRG1 controls gastric cancer migration and invasion through regulating MMP-9. Pathol. Oncol. Res. 2016;22:789–796. doi: 10.1007/s12253-016-0071-8.
    1. Marshall DC, Lyman SK, McCauley S, et al. Selective allosteric inhibition of MMP9 is efficacious in preclinical models of ulcerative colitis and colorectal cancer. PLoS ONE. 2015;10:e0127063. doi: 10.1371/journal.pone.0127063.
    1. Koizumi W, Narahara H, Hara T, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): A phase III trial. Lancet Oncol. 2008;9:215–221. doi: 10.1016/S1470-2045(08)70035-4.
    1. Yamada Y, Higuchi K, Nishikawa K, et al. Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naïve patients with advanced gastric cancer. Ann. Oncol. 2015;26:141–148. doi: 10.1093/annonc/mdu472.
    1. Lee KW, Chung IJ, Ryu MH, et al. Multicenter phase III trial of S-1 and cisplatin versus S-1 and oxaliplatin combination chemotherapy for first-line treatment of advanced gastric cancer (SOPP trial) Gastric Cancer. 2021;24:156–167. doi: 10.1007/s10120-020-01101-4.
    1. U.S. Department of Health and Human Services. National Institutes of Health, National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) Version 4.03. (2010). .
    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. Yamaguchi K, Satoh T, Muro K, et al. Phase 1b study of andecaliximab (GS-5745, ADX) as monotherapy and in combination with nivolumab (nivo) in Japanese subjects with gastric or GEJ adenocarcinoma. J. Clin. Oncol. 2019;37:137–137. doi: 10.1200/JCO.2019.37.4_suppl.137.
    1. Yoshikawa AK, Yamaguchi K, Muro K, et al. Safety and tolerability of andecaliximab as monotherapy and in combination with an anti-PD-1 antibody in Japanese patients with gastric or gastroesophageal junction adenocarcinoma: A phase 1b study. J. Immunother. Cancer. 2022;10:e003518. doi: 10.1136/jitc-2021-003518.
    1. JGC Association Japanese gastric cancer treatment guidelines 2018 (5th edition) Gastric Cancer. 2021;24:1–21. doi: 10.1007/s10120-020-01042-y.
    1. Boku N, Yamamoto S, Fukuda H, et al. Fluorouracil versus combination of irinotecan plus cisplatin versus S-1 in metastatic gastric cancer: a randomised phase 3 study. Lancet Oncol. 2009;10:1063–1069. doi: 10.1016/S1470-2045(09)70259-1.
    1. Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N. Engl. J. Med. 2008;358:36–46. doi: 10.1056/NEJMoa073149.
    1. Shah MA, Bodoky G, Starodub A, et al. Phase III study to evaluate efficacy and safety of andecaliximab with mFOLFOX6 as first-line treatment in patients with advanced gastric or GEJ adenocarcinoma (GAMMA-1) J. Clin. Oncol. 2021;39:990–1000. doi: 10.1200/JCO.20.02755.
    1. Catenacci DVT, Kang Y-K, Saeed A, et al. FIGHT: A randomized, double-blind, placebo-controlled, phase II study of bemarituzumab (bema) combined with modified FOLFOX6 in 1L FGFR2b+ advanced gastric/gastroesophageal junction adenocarcinoma (GC) J. Clin. Oncol. 2021;39:4010–4010. doi: 10.1200/JCO.2021.39.15_suppl.4010.
    1. Ooki A, Yamaguchi K. The dawn of precision medicine in diffuse-type gastric cancer. Ther. Adv. Med. Oncol. 2022;14:17588359221083049. doi: 10.1177/17588359221083049.
    1. Zhao R, Liu XQ, Wu XP, et al. Vascular endothelial growth factor (VEGF) enhances gastric carcinoma invasiveness via integrin alpha(v)beta6. Cancer Lett. 2010;287:150–156. doi: 10.1016/j.canlet.2009.06.006.
    1. Shao L, Zhang B, Wang L, Wu L, Kan Q, Fan K. MMP-9-cleaved osteopontin isoform mediates tumor immune escape by inducing expansion of myeloid-derived suppressor cells. Biochem. Biophys. Res. Commun. 2017;493:1478–1484. doi: 10.1016/j.bbrc.2017.10.009.
    1. Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625–637. doi: 10.1016/S0092-8674(02)00754-7.
    1. Denney H, Clench MR, Woodroofe MN. Cleavage of chemokines CCL2 and CXCL10 by matrix metalloproteinases-2 and -9: Implications for chemotaxis. Biochem. Biophys. Res. Commun. 2009;382:341–347. doi: 10.1016/j.bbrc.2009.02.164.
    1. Shah MA, Cunningham D, Metges JP, et al. Randomized, open-label, phase 2 study of andecaliximab plus nivolumab versus nivolumab alone in advanced gastric cancer identifies biomarkers associated with survival. J. Immunother. Cancer. 2021;9:1–10.
    1. Kang YK, Chen LT, Ryu MH, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): A randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022;23:234–247. doi: 10.1016/S1470-2045(21)00692-6.
    1. Chen SZ, Yao HQ, Zhu SZ, Li QY, Guo GH, Yu J. Expression levels of matrix metalloproteinase-9 in human gastric carcinoma. Oncol. Lett. 2015;9:915–919. doi: 10.3892/ol.2014.2768.
    1. Redondo-Muñoz J, Ugarte-Berzal E, García-Marco JA, et al. Alpha4beta1 integrin and 190-kDa CD44v constitute a cell surface docking complex for gelatinase B/MMP-9 in chronic leukemic but not in normal B cells. Blood. 2008;112:169–178. doi: 10.1182/blood-2007-08-109249.
    1. Takaishi S, Okumura T, Tu S, et al. Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells. 2009;27:1006–1020. doi: 10.1002/stem.30.
    1. Ishimoto T, Nagano O, Yae T, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19:387–400. doi: 10.1016/j.ccr.2011.01.038.
    1. Yosef G, Hayun H, Papo N. Simultaneous targeting of CD44 and MMP9 catalytic and hemopexin domains as a therapeutic strategy. Biochem. J. 2021;478:1139–1157. doi: 10.1042/BCJ20200628.
    1. Alford VM, Kamath A, Ren X, et al. Targeting the hemopexin-like domain of latent matrix metalloproteinase-9 (proMMP-9) with a small molecule inhibitor prevents the formation of focal adhesion junctions. ACS Chem. Biol. 2017;12:2788–2803. doi: 10.1021/acschembio.7b00758.

Source: PubMed

3
Předplatit