GSTM1 and GSTT1 double null genotypes determining cell fate and proliferation as potential risk factors of relapse in children with hematological malignancies after hematopoietic stem cell transplantation

Simona Jurkovic Mlakar, Satyanarayana Chakradhara Rao Uppugunduri, Tiago Nava, Vid Mlakar, Hadrien Golay, Shannon Robin, Nicolas Waespe, Mohamed Aziz Rezgui, Yves Chalandon, Jaap Jan Boelens, Robert G M Bredius, Jean-Hugues Dalle, Christina Peters, Selim Corbacioglu, Henrique Bittencourt, Maja Krajinovic, Marc Ansari, paediatric diseases working party of the European society for blood and marrow transplantation, Simona Jurkovic Mlakar, Satyanarayana Chakradhara Rao Uppugunduri, Tiago Nava, Vid Mlakar, Hadrien Golay, Shannon Robin, Nicolas Waespe, Mohamed Aziz Rezgui, Yves Chalandon, Jaap Jan Boelens, Robert G M Bredius, Jean-Hugues Dalle, Christina Peters, Selim Corbacioglu, Henrique Bittencourt, Maja Krajinovic, Marc Ansari, paediatric diseases working party of the European society for blood and marrow transplantation

Abstract

Purpose: This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models.

Methods: GSTM1- and GSTT1-null alleles were genotyped using germline DNA from whole blood prior to a conditioning BU-based regimen. Association of GSTM1- and GSTT1-null variants with relapse incidence was analyzed using multivariable competing risk analysis. BU-induced cell death studies were conducted in GSTs- null and non-null LCLs and CRISPR-Cas9 gene-edited THP1 leukemia cell lines.

Results: Carrying GSTM1/GSTT1 double null genotype was found to be an independent risk factor for post-HSCT relapse in 86 children (adjusted HR: 6.52 [95% Cl, 2.76-15.42; p = 1.9 × 10-5]). BU-induced cell death preferentially in THP1GSTM1(non-null) and LCLsGSTM1(non-null) as shown by decreased viability, increased necrosis and levels of the oxidized form of glutathione compared to null cells, while GSTT1 non-null cells showed increased baseline proliferation.

Conclusion: The clinical association suggests that GSTM1/GSTT1 double null genotype could serve as genetic stratification biomarker for the high risk of post-HSCT relapse. Functional studies have indicated that GSTM1 status modulates BU-induced cell death. On the other hand, GSTT1 is proposed to be involved in baseline cell proliferation.

Trial registration: ClinicalTrials.gov NCT01257854.

Keywords: Acute leukemia; Busulfan resistance; Hematological malignancies; Hematopoietic stem cell transplantation; Null genotypes of glutathione S-transferases; Post-transplant relapse.

Conflict of interest statement

The authors declare that they have no competing interests related to the submitted work.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Cumulative incidence plots of relapse in univariable competing risk analyses stratified by GST-null variants. Results are plotted for (A) GSTT1(−/−) group versus groups GSTT1( ±) and GSTT1(+ / +) together presented as GSTT1( +); (B) GSTM1(−/−) group versus groups GSTM1( ±) and GSTM1(+ / +) together presented as GSTM1( +); (C) combined GSTM1(−/−)/GSTT1(−/−) versus other genotype combinations [GSTM1(−/−)/GSTT1( +), GSTM1( +)/GSTT1(−/−) and GSTM1( +)/GSTT1( +)]; (D) combined GSTM1(−/−)/GSTT1(−/−) versus grouped other genotype combinations [GSTM1( +)/GSTT1( +)]. p values for the difference in cumulative incidence of relapse were calculated with Gray’s test with death as a competing event. HR, hazard ratio; GSTM1-null and GSTT1-null are presented as GSTM1(−/−) and GSTT1(−/−), respectively. GSTM1 non-null and GSTT1 non-null genotypes are presented as GSTM1( +) and GSTT1( +), respectively
Fig. 2
Fig. 2
IC50-values for BU in GSTM1-null and GSTT1-null LCLs, THP1GSTM1(−/−) and THP1GSTT1(−/−) cells. IC50 values for BU were stratified according to GSTM1- (A) and GSTT1-null variants (C) in LCLs and CRISPR–Cas9 gene-edited THP1GSTM1(−/−) (B) and THP1GSTT1(−/−) (D) cell models. Concentration–response titration points were fitted to a Hill equation for BU. The 50% inhibitory concentrations of BU (BU-IC50) were determined by dose–response curve fitting using Prism 5.02 software (GraphPad SoftwareInc., CA. USA). The coefficient of determination (R2) of each plate was used to assess experimental reproducibility and was set to be above 0.95. Independent experiments were repeated at least three times. Non-parametric unpaired t test was used in LCLs (A, C). Pairwise comparisons by t test between GST(−/−) variants in THP1-CRISPR–Cas9 models (B., D.) were used. In THP1-CRISPR–Cas9 cell models (B., D.), dots represented are specific clones with identified GST(−/−) variants (+ / + vs. /) based on Sanger DNA-sequencing. p values below 0.05 were considered statistically significant. ns, not significant. GSTM1-null and GSTT1-null are presented as GSTM1(−/−) and GSTT1(−/−), respectively. GSTM1 non-null and GSTT1 non-null genotypes are presented as GSTM1( +) and GSTT1( +), respectively
Fig. 3
Fig. 3
Baseline cell growth against GSTM1-null and GSTT1-null genotypes in LCLs and THP1GSTT1(−/−) cells. Baseline cell growth was assessed against GSTM1-null (A) and GSTT1-null (B) genotypes in LCLs at 48 h end-point analysis and the (C) RealTime Cell Viability assay–Kinetics plot was performed for 72 h stratified by GSTT1-null genotype at baseline (proliferation profile) in THP1GSTT1(−/−) cell models. (A, B) On the y-axis, the basal cell growth rate (r) was calculated using the following formula, appropriate for the usual exponential kinetics of cell growth (N; the number of cells) after the defined time (t; h): Nt = No.2tr. (C) Measurement of baseline reducing the potential of viable cells according to GSTT1(−/−) variant in CRISPR–Cas9 gene-edited cell models was performed. The unpaired t test between GST genotypes in LCLs (A., B.) and Pairwise comparisons by t test between GSTT1 genotypes in THP1-CRISPR–Cas9 models were used. p values below 0.05 were considered statistically significant. GSTM1-null and GSTT1-null are presented as GSTM1(−/−) and GSTT1(−/−), respectively. GSTM1 non-null and GSTT1 non-null genotypes are presented as GSTM1( +) and GSTT1( +), respectively
Fig. 4
Fig. 4
Number of necrotic and apoptotic cells in LCLs and caspase 3/7 activity in LCLs, THP1GSTM1(−/−) and THP1GSTT1(−/−) cells. Flow cytometric analysis (FACS) by using Annexin V/PI assay was used to assess primary necrosis (A), late apoptosis (B), live cells (C) and early apoptosis (D) in LCLs stratified according to GSTM1-null variants; and caspase 3/7 activity (E) in LCLs and THP-CRISPR–Cas9 models stratified according to GST-null variants at 250, 500 and 1000 μM BU 48 h post-treatment. Statistical analysis was performed by two-way ANOVA considering 250, 500 and 1000 μM BU concentrations (genotype and treatment factors); t tests between GST(−/−) variants in each condition separately were used; no statistically significant differences were observed between GSTM1 + and GSTM1(−/−) LCLs and THP-CRISPR–Cas9 models in either 1% DMSO or medium only; p values below 0.05 were considered statistically significant. GSTM1-null and GSTT1-null are presented as GSTM1(−/−) and GSTT1(−/−), respectively. GSTM1 non-null and GSTT1 non-null genotypes are presented as GSTM1( +) and GSTT1( +), respectively
Fig. 5
Fig. 5
Glutathione levels in LCLs stratified according to GSTM1-null variant, THP1GSTM1(−/−) and THP1GSTT1(−/−) cells. [GSSG/GSHT] ratio (A) and GSHT (B) in LCLs were calculated stratified according to GSTM1-null variant; and [GSSG/GSHT] ratios in CRISPR–Cas9 edited THP1GSTM1(−/−) (C) and THP1GSTT1(−/−) (D) cell models after the treatment with 500 μM BU. Statistical analysis was performed by the two-way ANOVA considering 250 or 500 μM BU concentration (genotype factor); t tests between GST(−/−) variants in each condition separately were used; no statistically significant differences were observed between GST(−/−) variants in LCLs and THP-CRISPR–Cas9 models in either 1% DMSO or medium; p values below 0.05 were considered statistically significant. GSTM1-null and GSTT1-null are presented as GSTM1(−/−) and GSTT1(−/−), respectively. GSTM1 non-null and GSTT1 non-null genotypes are presented as GSTM1( +) and GSTT1( +), respectively

References

    1. Ansari M, et al. GSTA1 diplotypes affect busulfan clearance and toxicity in children undergoing allogeneic hematopoietic stem cell transplantation: a multicenter study. Oncotarget. 2017;8:90852–90867.
    1. Balta G, Yuksek N, Ozyurek E, Ertem U, Hicsonmez G, Altay C, Gurgey A. Characterization of MTHFR, GSTM1, GSTT1, GSTP1, and CYP1A1 genotypes in childhood acute leukemia. Am J Hematol. 2003;73:154–160.
    1. Barragan E, Collado M, Cervera J, Martin G, Bolufer P, Roman J, Sanz MA. The GST deletions and NQO1*2 polymorphism confers interindividual variability of response to treatment in patients with acute myeloid leukemia. Leuk Res. 2007;31:947–953.
    1. Barrett AJ, Battiwalla M. Relapse after allogeneic stem cell transplantation. Expert Rev Hematol. 2010;3:429–441.
    1. Bartelink IH, et al. Association of busulfan exposure with survival and toxicity after haemopoietic cell transplantation in children and young adults: a multicentre, retrospective cohort analysis. Lancet Haematol. 2016;3:e526–e536.
    1. Board PG, Menon D. Glutathione transferases, regulators of cellular metabolism and physiology. Biochim Biophys Acta. 2013;1830:3267–3288.
    1. Borst L, Buchard A, Rosthoj S, Wesolowska A, Wehner PS, Wesenberg F, Dalhoff K, Schmiegelow K. Gene dose effects of GSTM1, GSTT1 and GSTP1 polymorphisms on outcome in childhood acute lymphoblastic leukemia. J Pediatr Hematol Oncol. 2012;34:38–42.
    1. Bremer S, Floisand Y, Brinch L, Gedde-Dahl T, Bergan S. Glutathione transferase gene variants influence busulfan pharmacokinetics and outcome after myeloablative conditioning. Ther Drug Monit. 2015;37:493–500.
    1. Chen CL, Liu Q, Pui CH, Rivera GK, Sandlund JT, Ribeiro R, Evans WE, Relling MV. Higher frequency of glutathione S-transferase deletions in black children with acute lymphoblastic leukemia. Blood. 1997;89:1701–1707.
    1. Ciurea SO, Andersson BS. Busulfan in hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2009;15:523–536.
    1. Czerwinski M, Gibbs JP, Slattery JT. Busulfan conjugation by glutathione S-transferases alpha, mu, and pi. Drug Metab Dispos. 1996;24:1015–1019.
    1. Czerwinski M, Kiem HP, Slattery JT. Human CD34+ cells do not express glutathione S-transferases alpha. Gene Ther. 1997;4:268–270.
    1. DeLeve LD, Wang X. Role of oxidative stress and glutathione in busulfan toxicity in cultured murine hepatocytes. Pharmacology. 2000;60:143–154.
    1. Elhasid R, Krivoy N, Rowe JM, Sprecher E, Adler L, Elkin H, Efrati E. Influence of glutathione S-transferase A1, P1, M1, T1 polymorphisms on oral busulfan pharmacokinetics in children with congenital hemoglobinopathies undergoing hematopoietic stem cell transplantation. Pediatr Blood Cancer. 2010;55:1172–1179.
    1. El-Serafi I, Terelius Y, Abedi-Valugerdi M, Naughton S, Saghafian M, Moshfegh A, Mattsson J, Potacova Z, Hassan M. Flavin-containing monooxygenase 3 (FMO3) role in busulphan metabolic pathway. PLoS ONE. 2017;12:e0187294.
    1. Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.
    1. Franca R, et al. Glutathione S-transferase homozygous deletions and relapse in childhood acute lymphoblastic leukemia: a novel study design in a large Italian AIEOP cohort. Pharmacogenomics. 2012;13:1905–1916.
    1. Gaziev J, et al. Novel pharmacokinetic behavior of intravenous busulfan in children with thalassemia undergoing hematopoietic stem cell transplantation: a prospective evaluation of pharmacokinetic and pharmacodynamic profile with therapeutic drug monitoring. Blood. 2010;115:4597–4604.
    1. Goekkurt E, Stoehlmacher J, Stueber C, Wolschke C, Eiermann T, Iacobelli S, Zander AR, Ehninger G, Kroger N. Pharmacogenetic analysis of liver toxicity after busulfan/cyclophosphamide-based allogeneic hematopoietic stem cell transplantation. Anticancer Res. 2007;27:4377–4380.
    1. Gray RJ. A class of K-sample tests for comparing the cumulative incidence of a competing risk. Ann Stat. 1988;16:1141–1154.
    1. Hall AG, Autzen P, Cattan AR, Malcolm AJ, Cole M, Kernahan J, Reid MM. Expression of mu class glutathione S-transferase correlates with event-free survival in childhood acute lymphoblastic leukemia. Cancer Res. 1994;54:5251–5254.
    1. Hamilton BK, Copelan EA. Concise review: the role of hematopoietic stem cell transplantation in the treatment of acute myeloid leukemia. Stem Cells. 2012;30:1581–1586.
    1. Hao C, Ma X, Wang L, Zhang W, Hu J, Huang J, Yang W. Predicting the presence and mechanism of busulfan drug-drug interactions in hematopoietic stem cell transplantation using pharmacokinetic interaction network-based molecular structure similarity and network pharmacology. Eur J Clin Pharmacol. 2020;77(4):595–605.
    1. Hoban PR, Robson CN, Davies SM, Hall AG, Cattan AR, Hickson ID, Harris AL. Reduced topoisomerase II and elevated alpha class glutathione S-transferase expression in a multidrug resistant CHO cell line highly cross-resistant to mitomycin C. Biochem Pharmacol. 1992;43:685–693.
    1. Horowitz MM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990;75:555–562.
    1. Kim SD, et al. Influence of GST gene polymorphisms on the clearance of intravenous busulfan in adult patients undergoing hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2011;17:1222–1230.
    1. Lee CJ, et al. Late effects after ablative allogeneic stem cell transplantation for adolescent and young adult acute myeloid leukemia. Blood Adv. 2020;4:983–992.
    1. Leonardi DB, Abbate M, Riccheri MC, Nunez M, Alfonso G, Gueron G, De Siervi A, Vazquez E, Cotignola J. Improving risk stratification of patients with childhood acute lymphoblastic leukemia: Glutathione-S-Transferases polymorphisms are associated with increased risk of relapse. Oncotarget. 2017;8:110–117.
    1. Li M, Zheng M, Chen H, Yu H. Effects of GST variants on the risk odds of hematological malignancy: a meta-analysis. J Cell Biochem. 2018;120(5):8570–8580.
    1. Lin DX, Tang YM, Peng Q, Lu SX, Ambrosone CB, Kadlubar FF. Susceptibility to esophageal cancer and genetic polymorphisms in glutathione S-transferases T1, P1, and M1 and cytochrome P450 2E1. Cancer Epidemiol Biomarkers Prev. 1998;7:1013–1018.
    1. Marin F, Garcia N, Munoz X, Capella G, Gonzalez CA, Agudo A, Sala N. Simultaneous genotyping of GSTT1 and GSTM1 null polymorphisms by melting curve analysis in presence of SYBR Green I. J Mol Diagn. 2010;12:300–304.
    1. McCune JS, Holmberg LA. Busulfan in hematopoietic stem cell transplant setting. Expert Opin Drug Metab Toxicol. 2009;5:957–969.
    1. McCune JS, Gooley T, Gibbs JP, Sanders JE, Petersdorf EW, Appelbaum FR, Anasetti C, Risler L, Sultan D, Slattery JT. Busulfan concentration and graft rejection in pediatric patients undergoing hematopoietic stem cell transplantation. Bone Marrow Transplant. 2002;30:167–173.
    1. Myers AL, Kawedia JD, Champlin RE, Kramer MA, Nieto Y, Ghose R, Andersson BS. Clarifying busulfan metabolism and drug interactions to support new therapeutic drug monitoring strategies: a comprehensive review. Expert Opin Drug Metab Toxicol. 2017;13:901–923.
    1. Peters C, et al. Total body irradiation or chemotherapy conditioning in childhood all: a multinational, randomized, noninferiority phase III study. J Clin Oncol. 2021;39:295–307.
    1. Philippe M, Goutelle S, Guitton J, Fonrose X, Bergeron C, Girard P, Bertrand Y, Bleyzac N. Should busulfan therapeutic range be narrowed in pediatrics? Experience from a large cohort of hematopoietic stem cell transplant children. Bone Marrow Transplant. 2016;51:72–78.
    1. Rocha JC, et al. Pharmacogenetics of outcome in children with acute lymphoblastic leukemia. Blood. 2005;105:4752–4758.
    1. Sachet M, Liang YY, Oehler R. The immune response to secondary necrotic cells. Apoptosis. 2017;22:1189–1204.
    1. Saitou M, Ishida T. Distributions of the GSTM1 and GSTT1 null genotypes worldwide are characterized by latitudinal clines. Asian Pac J Cancer Prev. 2015;16:355–361.
    1. Shah NN, Borowitz MJ, Steinberg SM, Robey NC, Gamper CJ, Symons HJ, Loeb DM, Wayne AS, Chen AR. Factors predictive of relapse of acute leukemia in children after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2014;20:1033–1039.
    1. Smith MT, Evans CG, Doane-Setzer P, Castro VM, Tahir MK, Mannervik B. Denitrosation of 1,3-bis(2-chloroethyl)-1-nitrosourea by class mu glutathione transferases and its role in cellular resistance in rat brain tumor cells. Cancer Res. 1989;49:2621–2625.
    1. Srivastava A, Poonkuzhali B, Shaji RV, George B, Mathews V, Chandy M, Krishnamoorthy R. Glutathione S-transferase M1 polymorphism: a risk factor for hepatic venoocclusive disease in bone marrow transplantation. Blood. 2004;104:1574–1577.
    1. Stanulla M, Schrappe M, Brechlin AM, Zimmermann M, Welte K. Polymorphisms within glutathione S-transferase genes (GSTM1, GSTT1, GSTP1) and risk of relapse in childhood B-cell precursor acute lymphoblastic leukemia: a case-control study. Blood. 2000;95:1222–1228.
    1. Takanashi M, Morimoto A, Yagi T, Kuriyama K, Kano G, Imamura T, Hibi S, Todo S, Imashuku S. Impact of glutathione S-transferase gene deletion on early relapse in childhood B-precursor acute lymphoblastic leukemia. Haematologica. 2003;88:1238–1244.
    1. Teachey DT, Hunger SP. Predicting relapse risk in childhood acute lymphoblastic leukaemia. Br J Haematol. 2013;162:606–620.
    1. Terakura S, et al. Analysis of glutathione S-transferase and cytochrome P450 gene polymorphism in recipients of dose-adjusted busulfan-cyclophosphamide conditioning. Int J Hematol. 2020;111:84–92.
    1. Tew KD, Townsend DM. Glutathione-s-transferases as determinants of cell survival and death. Antioxid Redox Signal. 2012;17:1728–1737.
    1. Udensi UK, Tchounwou PB. Dual effect of oxidative stress on leukemia cancer induction and treatment. J Exp Clin Cancer Res. 2014;33:106.
    1. Uppugunduri CRS, et al. The association of combined GSTM1 and CYP2C9 genotype status with the occurrence of hemorrhagic cystitis in pediatric patients receiving myeloablative conditioning regimen prior to allogeneic hematopoietic stem cell transplantation. Front Pharmacol. 2017;8:451.
    1. Weiss JR, Baer MR, Ambrosone CB, Blanco JG, Hutson A, Ford LA, Moysich KB. Concordance of pharmacogenetic polymorphisms in tumor and germ line DNA in adult patients with acute myeloid leukemia. Cancer Epidemiol Biomarkers Prev. 2007;16:1038–1041.
    1. Woo MH, et al. Glutathione S-transferase genotypes in children who develop treatment-related acute myeloid malignancies. Leukemia. 2000;14:232–237.
    1. Xiao Z, Yang L, Xu Z, Zhang Y, Liu L, Nie L, Li L, Wang J, Hao Y. Glutathione S-transferases (GSTT1 and GSTM1) genes polymorphisms and the treatment response and prognosis in Chinese patients with de novo acute myeloid leukemia. Leuk Res. 2008;32:1288–1291.
    1. Xiao Q, Deng D, Li H, Ye F, Huang L, Zhang B, Ye B, Mo Z, Yang X, Liu Z. GSTT1 and GSTM1 polymorphisms predict treatment outcome for acute myeloid leukemia: a systematic review and meta-analysis. Ann Hematol. 2014;93:1381–1390.
    1. Yeshurun M, et al. The impact of the graft-versus-leukemia effect on survival in acute lymphoblastic leukemia. Blood Adv. 2019;3:670–680.
    1. Zareifar S, Monabati A, Saeed A, Fakhraee F, Cohan N. The association of glutathione S-transferase gene mutations (including GSTT1 and GSTM1) with the prognostic factors and relapse in acute lymphoblastic leukemia. Pediatr Hematol Oncol. 2013;30:568–573.
    1. Zhang HY, Zhang J, Wu T, Bai H. Polymorphism of glutathione S-transferases and genetic sensitivity of childhood acute lymphoblastic leukemia: a meta-analysis. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2017;25:16–23.
    1. Zmorzynski S, Swiderska-Kolacz G, Koczkodaj D, Filip AA. Significance of polymorphisms and expression of enzyme-encoding genes related to glutathione in hematopoietic cancers and solid tumors. Biomed Res Int. 2015;2015:853573.

Source: PubMed

3
订阅