Expansion Phase 1 Study of Pegargiminase Plus Pemetrexed and Cisplatin in Patients With Argininosuccinate Synthetase 1-Deficient Mesothelioma: Safety, Efficacy, and Resistance Mechanisms

Peter W Szlosarek, Melissa M Phillips, Iuliia Pavlyk, Jeremy Steele, Jonathan Shamash, James Spicer, Sanjeev Kumar, Simon Pacey, Xiaoxing Feng, Amanda Johnston, John Bomalaski, Graeme Moir, Kelvin Lau, Stephen Ellis, Michael Sheaff, Peter W Szlosarek, Melissa M Phillips, Iuliia Pavlyk, Jeremy Steele, Jonathan Shamash, James Spicer, Sanjeev Kumar, Simon Pacey, Xiaoxing Feng, Amanda Johnston, John Bomalaski, Graeme Moir, Kelvin Lau, Stephen Ellis, Michael Sheaff

Abstract

Introduction: Pegargiminase (ADI-PEG 20; ADI) degrades arginine and potentiates pemetrexed (Pem) cytotoxicity in argininosuccinate synthetase 1 (ASS1)-deficient malignant pleural mesothelioma (MPM). We conducted a phase 1 dose-expansion study at the recommended phase 2 dose of ADI-PEG 20 with Pem and cisplatin (ADIPemCis), to further evaluate arginine-lowering therapy in ASS1-deficient MPM and explore the mechanisms of resistance.

Methods: A total of 32 patients with ASS1-deficient MPM (11 epithelioid; 10 biphasic;11 sarcomatoid) who were chemonaive received weekly intramuscular pegargiminase (36 mg/m2) with Pem (500 mg/m2) and cisplatin (75 mg/m2) intravenously, every 3 weeks (six cycles maximum). Maintenance pegargiminase was permitted until disease progression or withdrawal. Safety, pharmacodynamics, immunogenicity, and efficacy were determined. Biopsies were performed in progressing patients to explore the mechanisms of resistance to pegargiminase.

Results: The treatment was well tolerated. Most adverse events were of grade 1/2, whereas four nonhematologic grade 3/4 adverse events related to pegargiminase were reversible. Plasma arginine decreased whereas citrulline increased; this was maintained by 18 weeks of ADIPemCis therapy. The disease control rate in 31 assessed patients was 93.5% (n = 29 of 31; 95% confidence interval [CI]: 78.6%-99.2%), with a partial response rate of 35.5% (n = 11 of 31; 95% CI: 19.2%-54.6%). The median progression-free and overall survivals were 5.6 (95% CI: 4.0-6.0) and 10.1 (95% CI: 6.1-11.1) months, respectively. Progression biopsies on pegargiminase revealed a statistically significant influx of macrophages (n = 6; p = 0.0255) and patchy tumoral ASS1 reexpression (n = 2 of 6). In addition, we observed increased tumoral programmed death-ligand 1-an ADI-PEG 20 inducible gene-and the formation of CD3-positive T lymphocyte aggregates on disease progression (n = 2 of 5).

Conclusions: The dose expansion of ADIPemCis confirmed the high clinical activity and good tolerability in ASS1-deficient poor-prognosis mesothelioma, underpinning an ongoing phase 3 study (ClinicalTrials.govNCT02709512). Notably, resistance to pegargiminase correlated with marked macrophage recruitment and-along with the tumor immune microenvironment-warrants further study to optimize arginine deprivation for the treatment of mesothelioma.

Keywords: ADIPemCis; ASS1; Arginine; Macrophages; Mesothelioma.

© 2020 The Authors.

Figures

Figure 1
Figure 1
CONSORT diagram. AE, adverse event; ASS1, argininosuccinate synthetase 1; MPM, malignant pleural mesothelioma; SARC, sarcomatoid.
Figure 2
Figure 2
Pharmacodynamics and response. (A) Pharmacodynamics of arginine and citrulline in patients treated with ADIPemCis. Serum arginine and citrulline are revealed by week of treatment (mean ± SEM). (B) Serum levels of anti–ADI-PEG 20 antibodies in all patients by week of ADIPemCis (mean ± SEM). (C) Waterfall plot of response by modified RECIST to ADIPemCis. (D) Spider plots revealing response duration to ADIPemCis. Ab, antibody; ADI, arginine deiminase; ADIPemCis, ADI-PEG 20 with pemetrexed and cisplatin; B, biphasic; E, epithelioid; PEG, PEGylated; RECIST, Response Evaluation Criteria in Solid Tumors; S, sarcomatoid.
Figure 3
Figure 3
Survival outcomes. (A) PFS by MPM histologic subtype. (B) Kaplan-Meier survival estimates by MPM histologic subtype. CI, confidence interval; MPM, malignant pleural mesothelioma; OS, overall survival; PFS, progression-free survival.
Figure 4
Figure 4
Baseline and progression biopsies analyzed for ASS1 and CD68. (A) Tumoral ASS1 reexpression at progression noted in two patients (×200 magnification; epithelioid and sarcomatoid). (B) Increase in CD68pos macrophages at disease progression in ASS1–deficient tumoral regions (n = 6; p = 0.0255; paired t test); two epithelioid, one sarcomatoid, and three biphasic tumors (N.B. one epithelioid tumor was reclassified as biphasic on operative rebiopsy). (C) Representative serial sections of epithelioid, biphasic, and sarcomatoid MPM at baseline and progression stained for ASS1 and CD68, revealing the increase in ASS1posCD68pos macrophages at progression (×200 magnification). ASS1, argininosuccinate synthetase 1; MPM, malignant pleural mesothelioma; N.B., nota bene; pos, positive.
Figure 5
Figure 5
Baseline and progression biopsies analyzed for PD-L1 and CD3 modulation of PD-L1 expression and CD3pos lymphocytes in two patients at progression (×200 magnification; epithelioid and biphasic). PD-L1 increased from 10% to 30% (in epithelioid disease) and 0% to 5% (in biphasic disease) with clustering of CD3pos T cells in both patients at progression. PD-L1, programmed death-ligand 1; pos, positive.

References

    1. Klebe S., Brownlee N.A., Mahar A. Sarcomatoid mesothelioma: a clinical-pathologic correlation of 326 cases. Mod Pathol. 2010;23:470–479.
    1. Vigneswaran W.T., Kircheva D.Y., Ananthanarayanan V. Amount of epithelioid differentiation is a predictor of survival in malignant pleural mesothelioma. Ann Thorac Surg. 2017;103:962–966.
    1. Vogelzang N.J., Rusthoven J.J., Symanowski J. Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol. 2003;21:2636–2644.
    1. Szlosarek P.W., Klabatsa A., Pallaska A. In vivo loss of expression of argininosuccinate synthetase in malignant pleural mesothelioma is a biomarker for susceptibility to arginine depletion. Clin Cancer Res. 2006;12:7126–7131.
    1. Allen M.D., Luong P., Hudson C. Prognostic and therapeutic impact of argininosuccinate synthetase 1 control in bladder cancer as monitored longitudinally by PET imaging. Cancer Res. 2014;74:896–907.
    1. Rabinovich S., Adler L., Yizhak K. Diversion of aspartate in ASS1-deficient tumors fosters de novo pyrimidine synthesis. Nature. 2015;527:379–383.
    1. Keshet R., Szlosarek P., Carracedo A., Erez A. Rewiring urea cycle metabolism in cancer to support anabolism. Nat Rev Cancer. 2018;18:634–645.
    1. Ensor C.M., Holtsberg F.W., Bomalaski J.S., Clark M.A. PEGylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 2002;62:5443–5450.
    1. Cheng P.N., Lam T.L., Lam W.M. PEGylated recombinant human arginase (rhArg-peg5,000mw) inhibits the in vitro and in vivo proliferation of human hepatocellular carcinoma through arginine depletion. Cancer Res. 2007;67:309–317.
    1. Phillips M.M., Sheaff M.T., Szlosarek P.W. Targeting arginine-dependent cancers with arginine-degrading enzymes: opportunities and challenges. Cancer Res Treat. 2013;45:251–262.
    1. Szlosarek P.W., Steele J.P., Nolan L. Arginine deprivation with PEGylated arginine deiminase in patients with argininosuccinate synthetase 1-deficient malignant pleural mesothelioma: a randomized clinical trial. JAMA Oncol. 2017;3:58–66.
    1. Beddowes E., Spicer J., Chan P.Y. 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;35:1778–1785.
    1. Locke M., Ghazaly E., Freitas M.O. Inhibition of the polyamine synthesis pathway is synthetically lethal with loss of argininosuccinate synthase 1. Cell Rep. 2016;16:1604–1613.
    1. Tsai W.B., Aiba I., Lee S.Y., Feun L., Savaraj N., Kuo M.T. Resistance to arginine deiminase treatment in melanoma cells is associated with induced argininosuccinate synthetase expression involving c-Myc/HIF-1alpha/Sp4. Mol Cancer Ther. 2009;8:3223–3233.
    1. Feun L.G., Marini A., Walker G. Negative argininosuccinate synthetase expression in melanoma tumors may predict clinical benefit from arginine-depleting therapy with PEGylated arginine deiminase. Br J Cancer. 2012;106:1481–1485.
    1. Battisti S., Valente D., Albonici L., Bei R., Modesti A., Palumbo C. Nutritional stress and arginine auxotrophy confer high sensitivity to chloroquine toxicity in mesothelioma cells. Am J Respir Cell Mol Biol. 2012;46:498–506.
    1. Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
    1. Balkwill F.R., Mantovani A. Cancer-related inflammation: common themes and therapeutic opportunities. Semin Cancer Biol. 2012;22:33–40.
    1. Burt B.M., Rodig S.J., Tilleman T.R., Elbardissi A.W., Bueno R., Sugarbaker D.J. Circulating and tumor-infiltrating myeloid cells predict survival in human pleural mesothelioma. Cancer. 2011;117:5234–5244.
    1. Miselis N.R., Wu Z.J., Van Rooijen N., Kane A.B. Targeting tumor-associated macrophages in an orthotopic murine model of diffuse malignant mesothelioma. Mol Cancer Ther. 2008;7:788–799.
    1. Peddi P.F., Peddi S., Santos E.S., Morgensztern D. Central nervous system toxicities of chemotherapeutic agents. Expert Rev Anticancer Ther. 2014;14:857–863.
    1. Bueno R., Stawiski E.W., Goldstein L.D. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nat Genet. 2016;48:407–416.
    1. Scagliotti G.V., Gaafar R., Nowak A.K. Nintedanib in combination with pemetrexed and cisplatin for chemotherapy-naive patients with advanced malignant pleural mesothelioma (LUME-Meso): a double-blind, randomised, placebo-controlled phase 3 trial. Lancet Respir Med. 2019;7:569–580.
    1. Zalcman G., Mazieres J., Margery J. Bevacizumab for newly diagnosed pleural mesothelioma in the Mesothelioma Avastin Cisplatin Pemetrexed Study (MAPS): a randomised, controlled, open-label, phase 3 trial [published correction appears in Lancet. 2016;387:e24] Lancet. 2016;387:1405–1414.
    1. Forest F., Patoir A., Dal Col P. Nuclear grading, BAP1, mesothelin and PD-L1 expression in malignant pleural mesothelioma: prognostic implications. Pathology. 2018;50:635–641.
    1. Chou A., Toon C.W., Clarkson A., Sheen A., Sioson L., Gill A.J. The epithelioid BAP1-negative and p16-positive phenotype predicts prolonged survival in pleural mesothelioma. Histopathology. 2018;72:509–515.
    1. Tsao A.S., Miao J., Wistuba I.I. Phase II trial of cediranib in combination with cisplatin and pemetrexed in chemotherapy-naïve patients with unresectable malignant pleural mesothelioma (SWOG S0905) J Clin Oncol. 2019;37:2537–2547.
    1. Nussler A.K., Billiar T.R., Liu Z.Z., Morris S.M., Jr. Coinduction of nitric oxide synthase and argininosuccinate synthetase in a murine macrophage cell line. Implications for regulation of nitric oxide production. J Biol Chem. 1994;269:1257–1261.
    1. Phillips M., Szyszko T., Hall P. Abstract B33: expansion study of PEGylated arginine deiminase (ADI-PEG 20), pemetrexed, and cisplatin in patients with ASS1-deficient non-squamous non-small cell lung cancer (TRAP) Clin Cancer Res. 2018;24(suppl)
    1. Phillips M., Szlosarek P.W. In: Tumour-Associated Macrophages. Lawrence T., Hagemann T., editors. Springer; New York, NY: 2012. Arginine metabolism and tumour associated macrophages; pp. 77–90.
    1. Jayasingam S.D., Citartan M., Thang T.H., Mat Zin A.A., Ang K.C., Ch’ng E.S. Evaluating the polarization of tumor-associated macrophages into M1 and M2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice. Front Oncol. 2020;9:1512.
    1. Klionsky D.J., Abdelmohsen K., Abe A. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition) [published correction appears in Autophagy. 2016;12:443. Selliez, Iban [corrected to Seiliez, Iban]] Autophagy. 2016;12:1–222.
    1. Hall P.E., Lewis R., Syed N. 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;25:2708–2716.
    1. Kim R.H., Coates J.M., Bowles T.L. Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res. 2009;69:700–708.
    1. Delage B., Luong P., Maharaj L. Promoter methylation of argininosuccinate synthetase-1 sensitises lymphomas to arginine deiminase treatment, autophagy and caspase-dependent apoptosis. Cell Death Dis. 2012;3:e342.
    1. Brin E., Wu K., Lu H.T., 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;8:58948–58963.
    1. Chang K.Y., Chiang N.J., Yen C.J. A phase Ib study of ADI-PEG 20 plus pembrolizumab in advanced solid cancers. J Clin Oncol. 2018;36(supple15) 2556–2556.
    1. Alley E.W., Lopez J., Santoro A. Clinical safety and activity of pembrolizumab in patients with malignant pleural mesothelioma (KEYNOTE-028): preliminary results from a non-randomised, open-label, phase 1b trial. Lancet Oncol. 2017;18:623–630.
    1. Quispel-Janssen J., van der Noort V., de Vries J.F. Programmed death 1 blockade with nivolumab in patients with recurrent malignant pleural mesothelioma. J Thorac Oncol. 2018;13:1569–1576.
    1. Hassan R., Thomas A., Nemunaitis J.J. Efficacy and safety of avelumab treatment in patients with advanced unresectable mesothelioma: phase 1b results from the JAVELIN solid tumor trial. JAMA Oncol. 2019;5:351–357.
    1. Okada M., Kijima T., Aoe K. Clinical efficacy and safety of nivolumab: results of a multicenter, open-label, single-arm, Japanese phase II study in malignant pleural mesothelioma (MERIT) Clin Cancer Res. 2019;25:5485–5492.
    1. Awad M.M., Jones R.E., Liu H. Cytotoxic T cells in PD-L1-positive malignant pleural mesotheliomas are counterbalanced by distinct immunosuppressive factors. Cancer Immunol Res. 2016;4:1038–1048.
    1. Rodriguez P.C., Ochoa A.C., Al-Khami A.A. Arginine metabolism in myeloid cells shapes innate and adaptive immunity. Front Immunol. 2017;8:93.
    1. Murray P.J. Amino acid auxotrophy as a system of immunological control nodes. Nat Immunol. 2016;17:132–139.
    1. Lemos H., Huang L., Prendergast G.C., Mellor A.L. Immune control by amino acid catabolism during tumorigenesis and therapy. Nat Rev Cancer. 2019;19:162–175.
    1. Lee J.S., Adler L., Karathia H. Urea cycle dysregulation generates clinically relevant genomic and biochemical signatures. Cell. 2018;174:1559–1570.
    1. Ries C.H., Cannarile M.A., Hoves S. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25:846–859.
    1. Sharma B., Nawandar D.M., Nannuru K.C., Varney M.L., Singh R.K. Targeting CXCR2 enhances chemotherapeutic response, inhibits mammary tumor growth, angiogenesis, and lung metastasis. Mol Cancer Ther. 2013;12:799–808.
    1. Matlung H.L., Szilagyi K., Barclay N.A., van den Berg T.K. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol Rev. 2017;276:145–164.
    1. Schürch C.M., Forster S., Brühl F., Yang S.H., Felley-Bosco E., Hewer E. The “don’t eat me” signal CD47 is a novel diagnostic biomarker and potential therapeutic target for diffuse malignant mesothelioma. Oncoimmunology. 2017;7
    1. Gordon S.R., Maute R.L., Dulken B.W. PD-1 expression by tumor-associated macrophages inhibits phagocytosis and tumor immunity. Nature. 2017;545:495–499.
    1. Vitale I., Manic G., Coussens L.M., Kroemer G., Galluzzi L. Macrophages and metabolism in the tumor microenvironment. Cell Metab. 2019;30:36–50.

Source: PubMed

3
Sottoscrivi