A Phase II Study of Arginine Deiminase (ADI-PEG20) in Relapsed/Refractory or Poor-Risk Acute Myeloid Leukemia Patients

Hui-Jen Tsai, Shih Sheng Jiang, Wen-Chun Hung, Gautam Borthakur, Sheng-Fung Lin, Naveen Pemmaraju, Elias Jabbour, John S Bomalaski, Ya-Ping Chen, Hui-Hua Hsiao, Ming-Chung Wang, Ching-Yuan Kuo, Hung Chang, Su-Peng Yeh, Jorge Cortes, Li-Tzong Chen, Tsai-Yun Chen, Hui-Jen Tsai, Shih Sheng Jiang, Wen-Chun Hung, Gautam Borthakur, Sheng-Fung Lin, Naveen Pemmaraju, Elias Jabbour, John S Bomalaski, Ya-Ping Chen, Hui-Hua Hsiao, Ming-Chung Wang, Ching-Yuan Kuo, Hung Chang, Su-Peng Yeh, Jorge Cortes, Li-Tzong Chen, Tsai-Yun Chen

Abstract

Exogenous arginine is required for growth in some argininosuccinate synthetase (ASS)-deficient cancers. Arginine deiminase (ADI) inhibits growth in various ASS-deficient cancers by depleting arginine. The efficacy of pegylated ADI (ADI-PEG20) in relapsed/refractory/poor-risk acute myeloid leukemia (AML) was evaluated in 43 patients in a prospective, phase II trial (NCT01910012 (10/07/2013), https://ichgcp.net/clinical-trials-registry/NCT01910012?term = ADI-PEG20&rank = 12 ). Despite almost all pre-treatment tumor samples showing ASS deficiency, the best response among 21 evaluable patients was complete response (CR) in 2 (9.5%) and stable disease in 7 (33.3%), yielding a disease control rate (DCR) of 42.9%. The response durations of the two patients with CR were 7.5 and 8.8 months. DCR was correlated with a median of 8 weeks of arginine depletion to ≤10 μM. Using whole transcriptome sequencing, we compared gene expression profiling of pre- and post-treatment bone marrow samples of the two responders and three non-responders. The expression levels of some markers for AML subtypes and c-MYC regulated genes were considered potential predictors of response to ADI-PEG20. These results suggest that ASS deficiency is a prerequisite but not a sufficient condition for response to ADI-PEG20 monotherapy in AML. Predictive biomarkers and mechanistic explorations will be critical for identifying appropriate patients for future AML trials of ADI-PEG20.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Progression-free survival and overall survival of all intention-to-treat patients.
Figure 2
Figure 2
The mean circulating arginine and citrulline levels and the titers of anti-ADI-PEG20 antibodies in the intention-to treat patients during the ADI-PEG20 treatment.
Figure 3
Figure 3
The complete blood count (CBC) and bone marrow (BM) profiles of Case 1 and Case 2 during ADI-PEG20 treatment. (A) The dynamic CBC and BM changes in Case 1 after ADI-PEG20 treatment and the timing of packed red blood cell and platelet transfusion. (B) The dynamic CBC and BM changes in Case 2 after ADI-PEG20 treatment.
Figure 4
Figure 4
mRNA expression of associated genes in responders and non-responders before and after ADI-PEG20 treatment. (A) The relative mRNA expression levels of RHAG, ANK1, and NPM1 in the bone marrow mononuclear cells (BMMCs) of Case 1 and Case 2 at the indicated time points by QR-PCR. BMMC1B is the BMMCs of Case 1 before ADI-PEG20 treatment. BMMC1A is the BMMCs of Case 1 after ADI-PEG20 and in complete remission status. BMMC1R is the BMMCs of Case 1 collected at the time of relapse after ADI-PEG20 treatment. BMMC2B is the BMMCs of Case 2 before ADI-PEG20 treatment. BMMC2A is the BMMCs of Case 2 after ADI-PEG20 and in complete remission. The samples of BMMC1B and BMMC1R were compared with BMMC1A for analysis. The sample of BMMC2B was compared with BMMC1A for analysis. (B) The mRNA expression of c-MYC in AML cases responsive and non-responsive to ADI-PEG20. (left) Fold changes of mRNA expression of c-MYC in BMMC1B, and 1 R relative to BMMC1A and in BMMC 2B relative to BMMC2A. Case 1 and Case 2 were responders to ADI-PEG20. (right) Fold changes of mRNA expression of c-MYC in BMMC4A, 9 A, 10 A, 11 A, and 14 A relative to BMMC4B, 9B, 10B, 11B, and 14B, respectively. Cases 4, 9, 10, 11, and 14 were non-responders to ADI-PEG20. (Cases 4, 10, and 11were PD; Cases 9 and 14 were SD) “A” indicates after ADI-PEG20, “B” indicates before ADI-PEG20.

References

    1. Döhner H, et al. Acute myeloid leukemia. N Engl J Med. 2015;373:1136–1152. doi: 10.1056/NEJMra1406184.
    1. Health Promotion Administration, Ministry of Health and Welfare, Taiwan Health Promotion Administration Annual Report. Available from: . Date of access: 10/05/2017 (2016).
    1. Hengeveld M, et al. Intensive consolidation therapy compared with standard consolidation and maintenance therapy for adults with acute myeloid leukaemia aged between 46 and 60 years: final results of the randomized phase III study (AML 8B) of the European Organization for Research and Treatment of Cancer (EORTC) and the Gruppo Italiano Malattie Ematologiche Malignedell ‘Adulto (GIMEMA) Leukemia Cooperative Groups. Ann Hematol. 2012;91:825–835. doi: 10.1007/s00277-012-1436-z.
    1. Jehn U, et al. Non-infusional vs intravenous consolidation chemotherapy in elderly patients with acute myeloid leukemia: final results of the EORTC-GIMEMA AML-13 randomized phase III trial. Leukemia. 2006;20:1723–1730. doi: 10.1038/sj.leu.2404356.
    1. Patel JP, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366:1079–1089. doi: 10.1056/NEJMoa1112304.
    1. Knapper S, et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood. 2006;108:3262–3270. doi: 10.1182/blood-2006-04-015560.
    1. Jain N, et al. Phase II study of the oral MEK inhibitor Selumetinib in advanced acute myeloid leukemia: a University of Chicago phase II consortium trial. Clin Cancer Res. 2014;20:490–498. doi: 10.1158/1078-0432.CCR-13-1311.
    1. Garcia-Manero G, et al. Phase I dose escalation trial of ilorasertib, a dual Aurora/VEGF receptor kinase inhibitor, in patients with hematologic malignancies. Invest New Drugs. 2015;33:870–880. doi: 10.1007/s10637-015-0242-6.
    1. Phillips MM, et al. Targeting arginine-dependent cancers with arginine-degrading enzymes: opportunities and challenges. Cancer Res Treat. 2013;45:251–262. doi: 10.4143/crt.2013.45.4.251.
    1. Schimke RT, et al. The generation of energy by the arginine dihydrolase pathway in Mycoplasma hominis 07. J Biol Chem. 1966;241:2228–2236.
    1. Tytell AA, et al. Growth response of stable and primary cell cultures to L-ornithine, L-citrulline, and L-arginine. Exp Cell Res. 1960;20:84–91. doi: 10.1016/0014-4827(60)90225-1.
    1. Delage B, et al. Promoter methylation of argininosuccinate synthetase-1 sensitises lymphomas to arginine deiminase treatment, autophagy and caspase-dependent apoptosis. Cell Death Dis. 2012;3:e342. doi: 10.1038/cddis.2012.83.
    1. Ensor CM, et al. 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. Kim RH, et al. Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res. 2009;69:700–708. doi: 10.1158/0008-5472.CAN-08-3157.
    1. Glazer ES, et al. Phase II study of pegylated arginine deiminase for nonresectable and metastatic hepatocellular carcinoma. J Clin Oncol. 2010;28:2220–2226. doi: 10.1200/JCO.2009.26.7765.
    1. Feun LG, et al. Negative argininosuccinate synthetase expression in melanoma tumours may predict clinical benefit from arginine-depleting therapy with pegylated arginine deiminase. Br J Cancer. 2012;106:1481–1485. doi: 10.1038/bjc.2012.106.
    1. Yang TS, et al. A randomised phase II study of pegylated arginine deiminase (ADI-PEG 20) in Asian advanced hepatocellular carcinoma patients. Br J Cancer. 2010;103:954–960. doi: 10.1038/sj.bjc.6605856.
    1. Szlosarek P. et al. Pegylated arginine deiminase (ADI-PEG20) as a potential novel therapy for argininosuccinatesynthetase-deficient acute myeloid leukemia. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research-Innovation and Collaboration: The Path to Progress; 2011 Apr 2-6; Orlando, FL Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl), Abstract nr 4067. doi:10.1158/1538-7445.AM2011-4067.
    1. Miraki-Moud F, et al. Arginine deprivation using pegylated arginine deiminase has activity against primary acute myeloid leukemia cells in vivo. Blood. 2015;125:4060–4068. doi: 10.1182/blood-2014-10-608133.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Jaatinen T, et al. Global expression profile of human cord blood-derived CD133+ cells. Stem Cell. 2006;24:631–641. doi: 10.1634/stemcells.2005-0185.
    1. Schuringa JJ, et al. Constitutive activation of STAT5A promotes human hematopoietic stem cell self-renewal and erythroid differentiation. J Exp Med. 2004;200:623–635. doi: 10.1084/jem.20041024.
    1. Olsson AY, et al. Role of E2F3 expression in modulating cellular proliferation rate in human bladder and prostate cancer cells. Oncogene. 2007;26:1028–1037. doi: 10.1038/sj.onc.1209854.
    1. Adzhubei IA, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7:248–249. doi: 10.1038/nmeth0410-248.
    1. Kumar P, et al. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073–1081. doi: 10.1038/nprot.2009.86.
    1. Corominas M, et al. Hypersensitivity reactions to biological drugs. J Investig Allergol Clin Immunol. 2014;24:212–225.
    1. Szlosarek PW, et al. Metabolic response to pegylated arginine deiminase in mesothelioma with promoter methylation of argininosuccinatesynthetase. J Clin Oncol. 2013;31:e111–113. doi: 10.1200/JCO.2012.42.1784.
    1. Vadlamudi S, et al. Studies on neutralization of L-asparaginase activity in vitro and in vivo. Cancer. 1971;27:1321–1327. doi: 10.1002/1097-0142(197106)27:6<1321::AID-CNCR2820270607>;2-Y.
    1. Plunkett W. Arginine addiction in AML. Blood. 2015;125:3971–3972. doi: 10.1182/blood-2015-05-643411.
    1. Patil MD, et al. Arginine dependence of tumor cells: targeting a chink in cancer’s armor. Oncogene. 2016;35:4957–4972. doi: 10.1038/onc.2016.37.
    1. Cheson BD, et al. International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia.Revised recommendations of the International Working Group for Diagnosis, Standardizationof Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol. 2003;21:4642–4649. doi: 10.1200/JCO.2003.04.036.
    1. Sekeres MA, et al. A phase 2 study of lenalidomidemonotherapy in patients with deletion 5q acute myeloid leukemia: Southwest Oncology Group Study S0605. Blood. 2011;118:523–528. doi: 10.1182/blood-2011-02-337303.
    1. Kim D, et al. TopHat2:accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36. doi: 10.1186/gb-2013-14-4-r36.
    1. Langmead B, et al. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923.

Source: PubMed

3
Předplatit