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

Chakradhara Rao S Uppugunduri, Flavia Storelli, Vid Mlakar, Patricia Huezo-Diaz Curtis, Aziz Rezgui, Yves Théorêt, Denis Marino, Fabienne Doffey-Lazeyras, Yves Chalandon, Peter Bader, Youssef Daali, Henrique Bittencourt, Maja Krajinovic, Marc Ansari, Chakradhara Rao S Uppugunduri, Flavia Storelli, Vid Mlakar, Patricia Huezo-Diaz Curtis, Aziz Rezgui, Yves Théorêt, Denis Marino, Fabienne Doffey-Lazeyras, Yves Chalandon, Peter Bader, Youssef Daali, Henrique Bittencourt, Maja Krajinovic, Marc Ansari

Abstract

Hemorrhagic cystitis (HC) is one of the complications of busulfan-cyclophosphamide (BU-CY) conditioning regimen during allogeneic hematopoietic stem cell transplantation (HSCT) in children. Identifying children at high risk of developing HC in a HSCT setting could facilitate the evaluation and implementation of effective prophylactic measures. In this retrospective analysis genotyping of selected candidate gene variants was performed in 72 children and plasma Sulfolane (Su, water soluble metabolite of BU) levels were measured in 39 children following treatment with BU-CY regimen. The cytotoxic effects of Su and acrolein (Ac, water soluble metabolite of CY) were tested on human urothelial cells (HUCs). The effect of Su was also tested on cytochrome P 450 (CYP) function in HepaRG hepatic cells. Cumulative incidences of HC before day 30 post HSCT were estimated using Kaplan-Meier curves and log-rank test was used to compare the difference between groups in a univariate analysis. Multivariate Cox regression was used to estimate hazard ratios with 95% confidence intervals (CIs). Multivariate analysis included co-variables that were significantly associated with HC in a univariate analysis. Cumulative incidence of HC was 15.3%. In the univariate analysis, HC incidence was significantly (p < 0.05) higher in children older than 10 years (28.6 vs. 6.8%) or in children with higher Su levels (>40 vs. <11%) or in carriers of both functional GSTM1 and CYP2C9 (33.3 vs. 6.3%) compared to the other group. In a multivariate analysis, combined GSTM1 and CYP2C9 genotype status was associated with HC occurrence with a hazards ratio of 4.8 (95% CI: 1.3-18.4; p = 0.02). Ac was found to be toxic to HUC cells at lower concentrations (33 μM), Su was not toxic to HUC cells at concentrations below 1 mM and did not affect CYP function in HepaRG cells. Our observations suggest that pre-emptive genotyping of CYP2C9 and GSTM1 may aid in selection of more effective prophylaxis to reduce HC development in pediatric patients undergoing allogeneic HSCT. Article summary: (1) Children carrying functional alleles in GSTM1 and CYP2C9 are at high risk for developing hemorrhagic cystitis following treatment with busulfan and cyclophosphamide based conditioning regimen. (2) Identification of children at high risk for developing hemorrhagic cystitis in an allogeneic HSCT setting will enable us to evaluate and implement optimal strategies for its prevention. Trial registration: This study is a part of the trail "clinicaltrials.gov identifier: NCT01257854."

Keywords: CYPs; HepaRG; acrolein; busulfan; conjugation; cyclophosphamide; induction; urothelial cells.

Figures

FIGURE 1
FIGURE 1
Hypothetical model for Hemorrhagic Cystitis occurrence in children receiving BU-CY regimen prior to allogeneic HSCT. Busulfan (BU) and cyclophosphamide (CY) metabolism predominantly occurs in the liver generating the water soluble metabolites sulfolane (Su) and acrolein (Ac), respectively. During the metabolic process BU utilizes glutathione (GSH) and activity of GSTs such as GST alpha 1 (GSTA1), GST mu1 (GSTM1) plays an important role in conjugation of BU with GSH. GSH is also essential for scavenging the CY active metabolites including Ac also catalyzed by GSTs. When CY is administered after BU, the water soluble metabolite of BU, i.e., Su could influence the activity of cytochrome P 450 enzymes (CYPs) and thus might affect the activation of CY, or on the other hand higher Su levels also represents the utilization of GSH and formation of γ-glutamyl-dehydroalanyl-glycine (EdAG) affecting the availability of reduced form of GSH. Su might also affect the function of GSTs and other enzymes in BU, CY metabolism and proteins such as Mitochondrial uncoupling proteins (UCP) encoded by SLC25A27 gene affecting cell survival. Su possibly affect the function of ADH (alcohol dehydrogenase) enzyme resulting in increased levels of hydroxy CY metabolite, resulting increased Ac formation. BU could irreversibly alkylate the glutaredoxins (Grx) and Thioredoxins (Trx) increasing the oxidative stress, thus increasing toxicity of incoming CY metabolites such as 4 hydroxy cyclophosphamide (4OH-CY); Ac, chloro acetaldehyde (CA), and Phosphoramide mustard (PM). Altered activity of CYPs due to less functional alleles in the encoding genes might have a predictable function of Su formation and CY activation. Circulating CY metabolites (4OH CY, PM) including Ac inhibit cytokine function by alkylation and also cause immunosuppression, further increasing the chances of viremia and viruria (BK and/or JC virus). Ac conjugated with GSH in the liver gets delivered to the bladder and releases free Ac which then could alkylates cysteine residues and causing toxicity. Altogether increased rate of metabolite delivery to the urinary bladder together with viral load and lack of protection due to increased oxidative stress, endothelial damage to the blood vessels, urothelial damage and enhanced viral replication results in bladder inflammation and hematuria and the condition hemorrhagic cystitis.
FIGURE 2
FIGURE 2
Sufolane levels and occurrence of Hemorrhagic cystitis. (A,B) Su levels between patients with and without HC. (C) ROC curve for Su levels as a predictive marker for HC (n = 39). A cut off of 49.43 and 36.28 ng/mL/mg/kg for HC, before dose 7 and after dose 9, respectively [before dose 7; Area = 0.75 (95% CI: 0.59–0.93; p = 0.02); after dose 9; Area = 0.74 (95% CI: 0.57–0.91), p = 0.03]. (D,E) Cumulative incidences HC. Patients with HC/total number of patients in each group. (D) Before dose 7 (E) after dose 9.
FIGURE 3
FIGURE 3
Incidence of Hemorrhagic cystitis. (A) in relation to the CYP2C9 genotype status. Group I represents patients without dysfunctional alleles, and II represents carriers of at least one dysfunctional allele. (B) In relation to GSTM1 genotype, group I represents nonNull carriers; and group II represents Null allele carriers. (C) In relation to combined CYP2C9 and GSTM1 genotypes group I represents carriers of normal functional CYP2C9 and GSTM1 genotypes and group II represents carriers of at least one dysfunctional allele in either CYP2C9 or GSTM1 or both. Number of patients with HC/total number of patients in each group and p-values are mentioned on the plots. Multivariate Cox-regression p-value and hazards ratio with 95% confidence intervals are mentioned for 3C.

References

    1. Al-Rawithi S., El-Yazigi A., Ernst P., Al-Fiar F., Nicholls P. J. (1998). Urinary excretion and pharmacokinetics of acrolein and its parent drug cyclophosphamide in bone marrow transplant patients. Bone Marrow Transplant. 22 485–490. 10.1038/sj.bmt.1701355
    1. Ansari M., Huezo-Diaz P., Uppugunduri C. R. S., Nava T., Rezgui M. A., Mlakar V., et al. (2017). Functional GSTA1 haplotypes affect clearance and toxicity of busulfan when administered in 16 doses to pediatric patients undergoing hematopoietic stem cell transplantation: a multicenter prospective study on behalf of the pediatric disease working party of the European society for blood and marrow transplantation (EBMT). Blood 128 665.
    1. Ansari M., Rezgui M. A., Theoret Y., Uppugunduri C. R., Mezziani S., Vachon M. F., et al. (2013). Glutathione S-transferase gene variations influence BU pharmacokinetics and outcome of hematopoietic SCT in pediatric patients. Bone Marrow Transplant. 48 939–946. 10.1038/bmt.2012.265
    1. Ansari M., Theoret Y., Rezgui M. A., Peters C., Mezziani S., Desjean C., et al. (2014). Association between busulfan exposure and outcome in children receiving intravenous busulfan before hematopoietic stem cell transplantation. Ther. Drug Monit. 36 93–99. 10.1097/FTD.0b013e3182a04fc7
    1. Antherieu S., Chesne C., Li R., Camus S., Lahoz A., Picazo L., et al. (2010). Stable expression, activity, and inducibility of cytochromes P450 in differentiated HepaRG cells. Drug Metab. Dispos. 38 516–525. 10.1124/dmd.109.030197
    1. Balasubramanian P., Desire S., Panetta J. C., Lakshmi K. M., Mathews V., George B., et al. (2012). Population pharmacokinetics of cyclophosphamide in patients with thalassemia major undergoing HSCT. Bone Marrow Transplant. 47 1178–1185. 10.1038/bmt.2011.254
    1. Ben-Barouch S., Cohen O., Vidal L., Avivi I., Ram R. (2016). Busulfan fludarabine vs busulfan cyclophosphamide as a preparative regimen before allogeneic hematopoietic cell transplantation: systematic review and meta-analysis. Bone Marrow Transplant. 51 232–240. 10.1038/bmt.2015.238
    1. Berhane K., Widersten M., Engstrom A., Kozarich J. W., Mannervik B. (1994). Detoxication of base propenals and other alpha, beta-unsaturated aldehyde products of radical reactions and lipid peroxidation by human glutathione transferases. Proc. Natl. Acad. Sci. U.S.A. 91 1480–1484. 10.1073/pnas.91.4.1480
    1. Cai J., Bhatnagar A., Pierce W. M., Jr. (2009). Protein modification by acrolein: formation and stability of cysteine adducts. Chem. Res. Toxicol. 22 708–716. 10.1021/tx800465m
    1. Cantoni N., Gerull S., Heim D., Halter J., Bucher C., Buser A., et al. (2011). Order of application and liver toxicity in patients given BU and CY containing conditioning regimens for allogeneic hematopoietic SCT. Bone Marrow Transplant. 46 344–349. 10.1038/bmt.2010.137
    1. Cesaro S., Facchin C., Tridello G., Messina C., Calore E., Biasolo M. A., et al. (2008). A prospective study of BK-virus-associated haemorrhagic cystitis in paediatric patients undergoing allogeneic haematopoietic stem cell transplantation. Bone Marrow Transplant. 41 363–370. 10.1038/sj.bmt.1705909
    1. Conklin D. J., Haberzettl P., Lesgards J. F., Prough R. A., Srivastava S., Bhatnagar A. (2009). Increased sensitivity of glutathione S-transferase P-null mice to cyclophosphamide-induced urinary bladder toxicity. J. Pharmacol. Exp. Ther. 331 456–469. 10.1124/jpet.109.156513
    1. Gargiulo G., Orlando L., Alberani F., Crabu G., Di Maio A., Duranti L., et al. (2014). Haemorrhagic cystitis in haematopoietic stem cell transplantation (HSCT): a prospective observational study of incidence and management in HSCT centres within the GITMO network (Gruppo Italiano Trapianto Midollo Osseo). Ecancermedicalscience 8 420 10.3332/ecancer.2014.420
    1. Grafstrom R. C., Dypbukt J. M., Willey J. C., Sundqvist K., Edman C., Atzori L., et al. (1988). Pathobiological effects of acrolein in cultured human bronchial epithelial cells. Cancer Res. 48 1717–1721.
    1. Hashmi M., Vamvakas S., Anders M. W. (1992). Bioactivation mechanism of S-(3-oxopropyl)-N-acetyl-L-cysteine, the mercapturic acid of acrolein. Chem. Res. Toxicol. 5 360–365. 10.1021/tx00027a007
    1. Hayden R. T., Gu Z., Liu W., Lovins R., Kasow K., Woodard P., et al. (2015). Risk factors for hemorrhagic cystitis in pediatric allogeneic hematopoietic stem cell transplant recipients. Transpl. Infect. Dis. 17 234–241. 10.1111/tid.12364
    1. Hingorani S. (2009). “Kidney and bladder complications of hematopoietic cell transplantation,” in Thomas Hematopoietic Cell Transplantation, 4th Edn, eds Appelbaum F. R., Forman S. J., Negin R. S., Blume K. G. (Chichester: Wiley-Blackwell Publishing; ), 1473–1486.
    1. Hirsch H. H., Steiger J. (2003). Polyomavirus BK. Lancet Infect. Dis. 3 611–623. 10.1016/S1473-3099(03)00770-9
    1. Jin L., Szeto K. Y., Zhang L., Du W., Sun H. (2004). Inhibition of alcohol dehydrogenase by bismuth. J. Inorg. Biochem. 98 1331–1337. 10.1016/j.jinorgbio.2004.03.016
    1. Kachel D. L., Martin W. J., II (1994). Cyclophosphamide-induced lung toxicity: mechanism of endothelial cell injury. J. Pharmacol. Exp. Ther. 268 42–46.
    1. Kehrer J. P., Biswal S. S. (2000). The molecular effects of acrolein. Toxicol. Sci. 57 6–15. 10.1093/toxsci/57.1.6
    1. Leung A. Y., Yuen K. Y., Kwong Y. L. (2005). Polyoma BK virus and haemorrhagic cystitis in haematopoietic stem cell transplantation: a changing paradigm. Bone Marrow Transplant. 36 929–937. 10.1038/sj.bmt.1705139
    1. Lunde L. E., Dasaraju S., Cao Q., Cohn C. S., Reding M., Bejanyan N., et al. (2015). Hemorrhagic cystitis after allogeneic hematopoietic cell transplantation: risk factors, graft source and survival. Bone Marrow Transplant. 50 1432–1437. 10.1038/bmt.2015.162
    1. Patel J. M., Block E. R. (1993). Acrolein-induced injury to cultured pulmonary artery endothelial cells. Toxicol. Appl. Pharmacol. 122 46–53. 10.1006/taap.1993.1170
    1. Qin W. S., Deng Y. H., Cui F. C. (2016). Sulforaphane protects against acrolein-induced oxidative stress and inflammatory responses: modulation of Nrf-2 and COX-2 expression. Arch. Med. Sci. 12 871–880. 10.5114/aoms.2016.59919
    1. Ramu K., Fraiser L. H., Mamiya B., Ahmed T., Kehrer J. P. (1995). Acrolein mercapturates: synthesis, characterization, and assessment of their role in the bladder toxicity of cyclophosphamide. Chem. Res. Toxicol. 8 515–524. 10.1021/tx00046a005
    1. Ren S., Kalhorn T. F., Slattery J. T. (1999). Inhibition of human aldehyde dehydrogenase 1 by the 4-hydroxycyclophosphamide degradation product acrolein. Drug Metab. Dispos. 27 133–137.
    1. Rezvani A. R., McCune J. S., Storer B. E., Batchelder A., Kida A., Deeg H. J., et al. (2013). Cyclophosphamide followed by intravenous targeted busulfan for allogeneic hematopoietic cell transplantation: pharmacokinetics and clinical outcomes. Biol. Blood Marrow Transplant. 19 1033–1039. 10.1016/j.bbmt.2013.04.005
    1. Ruggeri A., Roth-Guepin G., Battipaglia G., Mamez A. C., Malard F., Gomez A., et al. (2015). Incidence and risk factors for hemorrhagic cystitis in unmanipulated haploidentical transplant recipients. Transpl. Infect. Dis. 17 822–830. 10.1111/tid.12455
    1. Russell S. J., Vowels M. R., Vale T. (1994). Haemorrhagic cystitis in paediatric bone marrow transplant patients: an association with infective agents, GVHD and prior cyclophosphamide. Bone Marrow Transplant. 13 533–539.
    1. Scian M., Atkins W. M. (2015). The busulfan metabolite EdAG irreversibly glutathionylates glutaredoxins. Arch. Biochem. Biophys. 583 96–104. 10.1016/j.abb.2015.08.005
    1. Seber A., Shu X. O., Defor T., Sencer S., Ramsay N. (1999). Risk factors for severe hemorrhagic cystitis following BMT. Bone Marrow Transplant. 23 35–40. 10.1038/sj.bmt.1701523
    1. Silva Lde P., Patah P. A., Saliba R. M., Szewczyk N. A., Gilman L., Neumann J., et al. (2010). Hemorrhagic cystitis after allogeneic hematopoietic stem cell transplants is the complex result of BK virus infection, preparative regimen intensity and donor type. Haematologica 95 1183–1190. 10.3324/haematol.2009.016758
    1. Takamoto S., Sakura N., Namera A., Yashiki M. (2004). Monitoring of urinary acrolein concentration in patients receiving cyclophosphamide and ifosphamide. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 806 59–63. 10.1016/j.jchromb.2004.02.008
    1. Timm R., Kaiser R., Lotsch J., Heider U., Sezer O., Weisz K., et al. (2005). Association of cyclophosphamide pharmacokinetics to polymorphic cytochrome P450 2C19. Pharmacogenomics J. 5 365–373. 10.1038/sj.tpj.6500330
    1. Tsuboi K., Kishi K., Ohmachi K., Yasuda Y., Shimizu T., Inoue H., et al. (2003). Multivariate analysis of risk factors for hemorrhagic cystitis after hematopoietic stem cell transplantation. Bone Marrow Transplant. 32 903–907. 10.1038/sj.bmt.1704240
    1. Uhlen M., Fagerberg L., Hallstrom B. M., Lindskog C., Oksvold P., Mardinoglu A., et al. (2015). Proteomics. Tissue-based map of the human proteome. Science 347:1260419 10.1126/science.1260419
    1. Uppugunduri C. R., Rezgui M. A., Diaz P. H., Tyagi A. K., Rousseau J., Daali Y., et al. (2014). The association of cytochrome P450 genetic polymorphisms with sulfolane formation and the efficacy of a busulfan-based conditioning regimen in pediatric patients undergoing hematopoietic stem cell transplantation. Pharmacogenomics J. 14 263–271. 10.1038/tpj.2013.38
    1. Versace F., Uppugunduri C. R., Krajinovic M., Theoret Y., Gumy-Pause F., Mangin P., et al. (2012). A novel method for quantification of sulfolane (a metabolite of busulfan) in plasma by gas chromatography-tandem mass spectrometry. Anal. Bioanal. Chem. 404 1831–1838. 10.1007/s00216-012-6330-y
    1. Xie H., Griskevicius L., Stahle L., Hassan Z., Yasar U., Rane A., et al. (2006). Pharmacogenetics of cyclophosphamide in patients with hematological malignancies. Eur. J. Pharm. Sci. 27 54–61. 10.1016/j.ejps.2005.08.008

Source: PubMed

3
订阅