Phase 1, pharmacogenomic, dose-expansion study of pegargiminase plus pemetrexed and cisplatin in patients with ASS1-deficient non-squamous non-small cell lung cancer

Peter W Szlosarek, Akhila G Wimalasingham, Melissa M Phillips, Peter E Hall, Pui Ying Chan, John Conibear, Louise Lim, Sukaina Rashid, Jeremy Steele, Paula Wells, Chiung-Fang Shiu, Chih-Ling Kuo, Xiaoxing Feng, Amanda Johnston, John Bomalaski, Stephen Ellis, Marianne Grantham, Michael Sheaff, Peter W Szlosarek, Akhila G Wimalasingham, Melissa M Phillips, Peter E Hall, Pui Ying Chan, John Conibear, Louise Lim, Sukaina Rashid, Jeremy Steele, Paula Wells, Chiung-Fang Shiu, Chih-Ling Kuo, Xiaoxing Feng, Amanda Johnston, John Bomalaski, Stephen Ellis, Marianne Grantham, Michael Sheaff

Abstract

Introduction: We evaluated the arginine-depleting enzyme pegargiminase (ADI-PEG20; ADI) with pemetrexed (Pem) and cisplatin (Cis) (ADIPemCis) in ASS1-deficient non-squamous non-small cell lung cancer (NSCLC) via a phase 1 dose-expansion trial with exploratory biomarker analysis.

Methods: Sixty-seven chemonaïve patients with advanced non-squamous NSCLC were screened, enrolling 21 ASS1-deficient subjects from March 2015 to July 2017 onto weekly pegargiminase (36 mg/m2 ) with Pem (500 mg/m2 ) and Cis (75 mg/m2 ), every 3 weeks (four cycles maximum), with maintenance Pem or pegargiminase. Safety, pharmacodynamics, immunogenicity, and efficacy were determined; molecular biomarkers were annotated by next-generation sequencing and PD-L1 immunohistochemistry.

Results: ADIPemCis was well-tolerated. Plasma arginine and citrulline were differentially modulated; pegargiminase antibodies plateaued by week 10. The disease control rate was 85.7% (n = 18/21; 95% CI 63.7%-97%), with a partial response rate of 47.6% (n = 10/21; 95% CI 25.7%-70.2%). The median progression-free and overall survivals were 4.2 (95% CI 2.9-4.8) and 7.2 (95% CI 5.1-18.4) months, respectively. Two PD-L1-expressing (≥1%) patients are alive following subsequent pembrolizumab immunotherapy (9.5%). Tumoral ASS1 deficiency enriched for p53 (64.7%) mutations, and numerically worse median overall survival as compared to ASS1-proficient disease (10.2 months; n = 29). There was no apparent increase in KRAS mutations (35.3%) and PD-L1 (<1%) expression (55.6%). Re-expression of tumoral ASS1 was detected in one patient at progression (n = 1/3).

Conclusions: ADIPemCis was safe and highly active in patients with ASS1-deficient non-squamous NSCLC, however, survival was poor overall. ASS1 loss was co-associated with p53 mutations. Therapies incorporating pegargiminase merit further evaluation in ASS1-deficient and treatment-refractory NSCLC.

Trial registration: ClinicalTrials.gov NCT02029690.

Keywords: ADIPemCis; ASS1; KRAS; PD-L1; arginine; arginine deiminase; non-squamous NSCLC; p53.

Conflict of interest statement

PW Szlosarek received support from the Higher Education Funding Council for England (HEFCE) and research grant support from Polaris Group. Ms Shiu, Ms Kuo, Dr Bomalaski, Dr Johnson, and Dr Feng are paid employees of Polaris Pharmaceuticals. The remaining authors did not report any relevant conflict of interest.

© 2021 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
CONSORT diagram
FIGURE 2
FIGURE 2
Pharmacodynamics and response. (A) Pharmacodynamics of arginine and citrulline in patients treated with ADIPemCis. Serum [arginine] and [citrulline] are shown by week of treatment (means ± SEM). (B) Serum levels of anti‐ADI‐PEG 20 antibodies in all patients by week of ADIPemCis (Mean ± SEM); Ab, Antibody. (C) Waterfall plot of response by RECIST 1.1. to ADIPemCis. (D) Spider plots showing response duration to ADIPemCis
FIGURE 3
FIGURE 3
Survival outcomes for ASS1‐deficient patients (A) Progression‐free survival. (B) Kaplan–Meier survival estimates
FIGURE 4
FIGURE 4
Survival outcomes for ASS1‐proficient patients Kaplan–Meier survival estimates for ASS1‐proficient patients screened as part of this study (n = 29) and treated with standard of care cisplatin and pemetrexed chemotherapy and compared to ASS1‐deficient patients receiving ADIPemCis (n = 21)

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(6):394‐424.
    1. Inamura K. Lung cancer: understanding its molecular pathology and the 2015 WHO classification. Front Oncol. 2017;7:193.
    1. Arbour KC, Riely GJ. Systemic therapy for locally advanced and metastatic non‐small cell lung cancer: a review. JAMA. 2019;322(8):764‐774.
    1. Husson A, Brasse‐Lagnel C, Fairand A, Renouf S, Lavoinne A. Argininosuccinate synthetase from the urea cycle to the citrulline‐NO cycle. Eur J Biochem. 2003;270(9):1887‐1899.
    1. Kim SH, Roszik J, Grimm EA, Ekmekcioglu S. Impact of l‐arginine metabolism on immune response and anticancer immunotherapy. Front Oncol. 2018;8:67.
    1. Delage B, Fennell DA, Nicholson L, et al. Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. Int J Cancer. 2010;126(12):2762‐2772.
    1. Keshet R, Szlosarek P, Carracedo A, Erez A. Rewiring urea cycle metabolism in cancer to support anabolism. Nat Rev Cancer. 2018;18(10):634‐645.
    1. Allen MD, Luong P, Hudson C, et al. Prognostic and therapeutic impact of argininosuccinate synthetase 1 control in bladder cancer as monitored longitudinally by PET imaging. Cancer Res. 2014;74(3):896‐907.
    1. Phillips MM, Sheaff MT, Szlosarek PW. Targeting arginine‐dependent cancers with arginine‐degrading enzymes: opportunities and challenges. Cancer Res Treat. 2013;45(4):251‐262.
    1. Szlosarek PW, Steele JP, Nolan L, et al. Arginine deprivation with pegylated arginine deiminase in patients with argininosuccinate synthetase 1‐deficient malignant pleural mesothelioma: a randomized clinical trial. JAMA Oncol. 2017;3(1):58‐66.
    1. Huang HY, Wu WR, Wang YH, et al. ASS1 as a novel tumor suppressor gene in myxofibrosarcomas: aberrant loss via epigenetic DNA methylation confers aggressive phenotypes, negative prognostic impact, and therapeutic relevance. Clin Cancer Res. 2013;19(11):2861‐2872.
    1. Qiu F, Chen Y‐R, Liu X, et al. Arginine starvation impairs mitochondrial respiratory function in ASS1‐deficient breast cancer cells. Sci Signal. 2014;7(319):ra31.
    1. Kobayashi E, Masuda M, Nakayama R, et al. Reduced argininosuccinate synthetase is a predictive biomarker for the development of pulmonary metastasis in patients with osteosarcoma. Mol Cancer Ther. 2010;9(3):535‐544.
    1. Nicholson LJ, Smith PR, Hiller L, et al. Epigenetic silencing of argininosuccinate synthetase confers resistance to platinum‐induced cell death but collateral sensitivity to arginine auxotrophy in ovarian cancer. Int J Cancer. 2009;125(6):1454‐1463.
    1. Chang JT, Lee YM, Huang RS. The impact of the Cancer Genome Atlas on lung cancer. Transl Res. 2015;166(6):568‐585. 10.1016/j.trsl.2015.08.001
    1. Rabinovich S, Adler L, Yizhak K, et al. Diversion of aspartate in ASS1‐deficient tumours fosters de novo pyrimidine synthesis. Nature. 2015;527(7578):379‐383.
    1. Szlosarek PW, Phillips MM, Pavlyk I, et al. Expansion phase 1 study of pegargiminase plus pemetrexed and cisplatin in patients with argininosuccinate synthetase 1‐deficient mesothelioma: safety, efficacy, and resistance mechanisms. JTO Clin Res Rep. 2020;1(4):1‐11. 10.1016/j.jtocrr.2020.100093
    1. Hall PE, Lewis R, Syed N, et al. A phase I study of pegylated arginine deiminase (pegargiminase), cisplatin, and pemetrexed in argininosuccinate synthetase 1‐deficient recurrent high‐grade glioma. Clin Cancer Res. 2019;25(9):2708‐2716.
    1. Beddowes E, Spicer J, Chan PY, et al. Phase 1 dose‐escalation study of pegylated arginine deiminase, cisplatin, and pemetrexed in patients with argininosuccinate synthetase 1‐deficient thoracic cancers. J Clin Oncol. 2017;35(16):1778‐1785.
    1. Scagliotti GV, Parikh P, von Pawel J, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy‐naive patients with advanced‐stage non‐small‐cell lung cancer. J Clin Oncol. 2008;26(21):3543‐3551.
    1. Gandhi L, Rodríguez‐Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non‐small‐cell lung cancer. N Engl J Med. 2018;378(22):2078‐2092.
    1. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first‐line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378(24):2288‐2301.
    1. Reck M, Rodríguez‐Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD‐L1‐positive non‐small‐cell lung cancer. N Engl J Med. 2016;375(19):1823‐1833.
    1. Herbst RS, Giaccone G, de Marinis F, et al. Atezolizumab for first‐line treatment of PD‐L1‐selected patients with NSCLC. N Engl J Med. 2020;383(14):1328‐1339.
    1. Van Allen EM, Golay HG, Liu Y, et al. Long‐term benefit of PD‐L1 blockade in lung cancer associated with JAK3 activation. Cancer Immunol Res. 2015;3(8):855‐863.
    1. Choi M, Kadara H, Zhang J, et al. Mutation profiles in early‐stage lung squamous cell carcinoma with clinical follow‐up and correlation with markers of immune function. Ann Oncol. 2017;28(1):83‐89.
    1. Aggarwal C, Rodriguez Abreu D, Felip E, et al. Prevalence of PD‐L1 expression in patients with non‐small cell lung cancer screened for enrollment in KEYNOTE‐001, ‐010, and ‐024. Ann Oncol. 2016;27:359‐78.
    1. Dietel M, Savelov N, Salanova R, et al. Real‐world prevalence of programmed death ligand 1 expression in locally advanced or metastatic non‐small‐cell lung cancer: the global, multicenter EXPRESS study. Lung Cancer. 2019;134:174‐179.
    1. Gelatti ACZ, Cordeiro de Lima VC, Freitas H, et al. Real‐world prevalence of PD‐L1 expression among tumor samples from patients with non‐small‐cell lung cancer. Clin Lung Cancer. 2020;21(6):e511‐e515.
    1. Brin E, Wu K, Lu HT, He Y, Dai Z, He W. PEGylated arginine deiminase can modulate tumor immune microenvironment by affecting immune checkpoint expression, decreasing regulatory T cell accumulation and inducing tumor T cell infiltration. Oncotarget. 2017;8(35):58948‐58963.
    1. Duan Q, Zhang H, Zheng J, Zhang L. Turning cold into hot: firing up the tumor microenvironment. Trends Cancer. 2020;6(7):605‐618.
    1. Lee JS, Adler L, Karathia H, et al. Urea cycle dysregulation generates clinically relevant genomic and biochemical signatures. Cell. 2018;174(6):1559‐70 e22.
    1. Chang KY, Chiang NJ, Wu SY, et al. Phase 1b study of pegylated arginine deiminase (ADI‐PEG 20) plus pembrolizumab in advanced solid cancers. Oncoimmunology. 2021;10(1). 10.1080/2162402X.2021.1943253
    1. Gibbons DL, Byers LA, Kurie JM. Smoking, p53 mutation, and lung cancer. Mol Cancer Res. 2014;12(1):3‐13.
    1. Miyamoto T, Lo PHY, Saichi N, et al. Argininosuccinate synthase 1 is an intrinsic Akt repressor transactivated by p53. Sci Adv. 2017;3(5):e1603204.
    1. Berkers CR, Maddocks OD, Cheung EC, Mor I, Vousden KH. Metabolic regulation by p53 family members. Cell Metab. 2013;18(5):617‐633.
    1. Robles AI, Linke SP, Harris CC. The p53 network in lung carcinogenesis. Oncogene. 2002;21(45):6898‐6907.
    1. Donehower LA, Soussi T, Korkut A, et al. Integrated analysis of TP53 gene and pathway alterations in the cancer genome atlas. Cell Rep. 2019;28(5):1370‐1384.e5.
    1. Skoulidis F, Heymach JV. Co‐occurring genomic alterations in non‐small‐cell lung cancer biology and therapy. Nat Rev Cancer. 2019;19(9):495‐509.
    1. Aggarwal C, Davis CW, Mick R, et al. Influence of TP53 mutation on survival in patients with advanced EGFR‐mutant non‐small‐cell lung cancer. JCO Precis Oncol. 2018;2018.
    1. Prudner BC, Rathore R, Robinson AM, et al. Arginine starvation and docetaxel induce c‐Myc‐driven hENT1 surface expression to overcome gemcitabine resistance in ASS1‐negative tumors. Clin Cancer Res. 2019;25(16):5122‐5134.
    1. Bueno R, Stawiski EW, Goldstein LD, et al. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nat Genet. 2016;48(4):407‐416.
    1. Markowitz P, Patel M, Groisberg R, et al. Genomic characterization of malignant pleural mesothelioma and associated clinical outcomes. Cancer Treat Res Commun. 2020;25:100232.
    1. Consortium APG . AACR Project GENIE: Powering Precision Medicine through an International Consortium. Cancer Discov. 2017;7(8):818‐831.
    1. Cancer Genome Atlas Research N . Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543‐550.
    1. Shepherd FA, Lacas B, Le Teuff G, et al. Pooled analysis of the prognostic and predictive effects of TP53 comutation status combined with KRAS or EGFR mutation in early‐stage resected non‐small‐cell lung cancer in four trials of adjuvant chemotherapy. J Clin Oncol. 2017;35(18):2018‐2027.
    1. Ruiz‐Cordero R, Ma J, Khanna A, et al. Simplified molecular classification of lung adenocarcinomas based on EGFR, KRAS, and TP53 mutations. BMC Cancer. 2020;20(1):83.
    1. Skoulidis F, Goldberg ME, Greenawalt DM, et al. STK11/LKB1 mutations and PD‐1 inhibitor resistance in KRAS‐mutant lung adenocarcinoma. Cancer Discov. 2018;8(7):822‐835.
    1. Galan‐Cobo A, Sitthideatphaiboon P, Qu X, et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS‐mutant Lung adenocarcinoma. Cancer Res. 2019;79(13):3251‐3267.
    1. Lohinai Z, Klikovits T, Moldvay J, et al. KRAS‐mutation incidence and prognostic value are metastatic site‐specific in lung adenocarcinoma: poor prognosis in patients with KRAS mutation and bone metastasis. Sci Rep. 2017;7:39721.
    1. Ghimessy A, Radeczky P, Laszlo V, et al. Current therapy of KRAS‐mutant lung cancer. Cancer Metastasis Rev. 2020;39(4):1159‐1177.
    1. Pavlyk I, Foster J, Dexter K, et al. Pegylated arginine deiminase sensitizes ASS1‐negative and KRAS mutant non‐small cell lung cancer to PD‐1 blockade immunotherapy. Cancer Res. 2020;80:nr 2217. 10.1158/1538-7445.AM2020-2217
    1. Tomlinson BK, Thomson JA, Bomalaski JS, et al. Phase I trial of arginine deprivation therapy with ADI‐PEG 20 plus docetaxel in patients with advanced malignant solid tumors. Clin Cancer Res. 2015;21(11):2480‐2486.
    1. Poillet‐Perez L, Xie X, Zhan LE, et al. Autophagy maintains tumour growth through circulating arginine. Nature. 2018;563(7732):569‐573.

Source: PubMed

3
구독하다