• Aucun résultat trouvé

Effect of interaction of glutathione S-transferases (T1 and M1) on the hematologic and cytogenetic responses in chronic myeloid leukemia patients treated with imatinib

N/A
N/A
Protected

Academic year: 2021

Partager "Effect of interaction of glutathione S-transferases (T1 and M1) on the hematologic and cytogenetic responses in chronic myeloid leukemia patients treated with imatinib"

Copied!
6
0
0

Texte intégral

(1)

O R I G I N A L P A P E R

Effect of interaction of glutathione S-transferases (T1 and M1) on the hematologic and cytogenetic responses in chronic myeloid leukemia patients treated with imatinib

Y. KassogueM. QuachouhH. Dehbi A. Quessar S. BenchekrounS. Nadifi

Received: 13 March 2014 / Accepted: 22 May 2014 ÓSpringer Science+Business Media New York 2014

Abstract The glutathioneS-transferases (GSTs) are phase II xenobiotic metabolizing enzymes known to be involved in the detoxification of carcinogens and anticancer drugs.

Individual genetic variation linked to inherited polymor- phisms ofGSTT1andGSTM1leading to a complete loss of enzyme activity could expose subjects to develop cancer or to induce drug resistance. Indeed, despite the impressive results obtained with the imatinib, some patients with chronic myeloid leukemia (CML) fail to achieve the expected results or develop resistance. The present study aimed to examine the impact ofGSTT1andGSTM1poly- morphisms on the response to imatinib in patients with CML.

Multiplex polymerase chain reaction was used to detect the genotypes ofGSTT1andGSTM1in 60 CML patients. We found that side effects were more frequent in patients car- ryingGSTT1null when compared toGSTT1present carriers (31 vs. 16.6 %; v2=6.2; p=0.013). The loss of

hematologic response was statistically greater in patients carrying the combined genotype GSTT1 present/GSTM1 present (26.3 %) when compared to GSTT1 null/GSTM1 present (12.8 %), GSTT1present/GSTM1 null (8.3 %) and GSTT1 null/GSTM1 null (0 %), (v2=18.85; p\0.001).

The complete cytogenetic response was higher in patients harboring the GSTT1 null/GSTM1 null (75 %) compared withGSTT1null/GSTM1present (55.6 %),GSTT1present/

GSTM1 null (50 %) and GSTT1 present/GSTM1 present (47.8). On the other hand, the frequency of none cytogenetic responders was more common in patients carryingGSTT1 present/GSTM1present (34.8 %) when compared to other genotype combinations (v2=20.99;p =0.05). Moreover, theGSTT1present/GSTM1present appeared to be associated with a final dose of 600 or 800 mg of imatinib, but not sig- nificantly. Based on these findings, we find that the interac- tion between GSTT1 and GSTM1 seems to influence treatment outcome in patients with CML. Therefore, further investigations are required to confirm these results, for better genotype–phenotype correlation.

Keywords GSTT1GSTM1 Chronic myeloid leukemiaImatinibDrug response

Introduction

Chronic Myeloid leukemia (CML), characterized by a malignant proliferation of cells from the granulocytic lineage, is a consequence of the reciprocal translocation t (9; 22) (q34; q11) or the BCR-ABL fusion gene at the molecular level. CML represents approximately 15-20 % of leukemia in adults [1, 2]. It is noteworthy that the intro- duction of imatinib mesylate (Gleevec), a tyrosine kinase inhibitor targeting the BCR-ABL fusion in CML treatment, Y. KassogueH. DehbiS. Nadifi (&)

Genetics and Molecular Pathology Laboratory, Medical School of Casablanca, University Hassan II, 19 Rue Tarik Ibnou Ziad, BP. 9154, Casablanca, Morocco

e-mail: nadifi@labgenmed.com Y. Kassogue

e-mail: kassoy2@yahoo.fr H. Dehbi

e-mail: hinddehbi@gmail.com

M. QuachouhA. QuessarS. Benchekroun

Department of Onco-Hematology, Ibn Rochd University Hospital, Casablanca, Morocco

e-mail: qachouhmeryem@gmail.com A. Quessar

e-mail: quessar.a@gmail.com S. Benchekroun

e-mail: medredaz@gmail.com DOI 10.1007/s12032-014-0047-z

(2)

has improved the quality of life of patients, reducing the response time, toxicity, morbidity and mortality associated with the disease [3,4]. However, despite these commend- able results achieved with imatinib, some patients fail to reach adequate hematologic, cytogenetic and molecular responses. It has been reported that mutations or amplifi- cation of BCR-ABL kinase domain [5–7] or variation in the bioavailability of imatinib linked to inherited genetic dif- ferences of the enzymes involved in imatinib metabolism could explain interindividual differences in the treatment outcome [8,9]. Indeed, it is well established that the highly polymorphic cytochrome P450 enzymes particularly CYP3A4 plays a non-negligible role in the metabolism of imatinib with other isoenzymes as CYP1A2, CYP2D6, CYP2C9 and CYP2C19 [10]. Furthermore, other studies have shown a correlation between polymorphisms in the multidrug resistance gene (MDR1) and imatinib response [11, 12]. It was also reported that polymorphisms in the super family of glutathione S-transferases (GSTs) gene were associated with treatment outcome in patients receiving imatinib [13–15]. GSTs are phase II xenobiotic metabolizing enzymes, which participate in the transfor- mation and activation of a wide range of substrates including carcinogens, pesticides, anticancer drugs and harmful compounds from the environment by conjugating them to soluble glutathione [16]. GSTs are composed of microsomal and cytosolic or soluble GSTs; Cytosolic GST includes eight classes divided intoa(GSTA),l(GSTM),h (GSTT),p(GSTP),f(GSTZ),r(GSTS),j(GSTK) andx (GSTO) [17].GSTM1andGSTT1 belong to the class mu and theta and are located on chromosomes 1p13.3 and 22q11.2, respectively [18,19]. The detoxifying activity of GSTs enzymes protects cells from the adverse effects of xenobiotics, but can reduce the effectiveness of drugs in cancer cells, resulting in toxicity or drug resistance [20,21].

Deletions ofGSTM1andGSTT1gene leading to a complete absence of enzyme activity have been found to be associ- ated with a better outcome in the treatment of some cancers [22,23], but other studies showed no relationship [24,25].

Considering the few data on the pharmacogenetic aspects of CML in relation to the polymorphisms in GSTs genes, we carried out the present study with the aim to investigate the impact of the genotypes of GSTT1 and GSTM1 on the response to imatinib in CML patients.

Materials and Methods

Patients

In the present study, 60 CML patients with a median age of 40.5 years range (20-74 years) at the time of diagnosis have participated. The patients were monitored at the

department of Onco-Hematology of the Ibn Rochd Uni- versity Hospital in Casablanca, Morocco, from 2009 to 2013. All patients were in chronic phase and were con- firmed with the presence of the reciprocal translocation t (9; 22) (q34; q11). All patients started with 400 mg of imatinib per day; this dose was escalated to 600 or 800 mg per day for patients who did not reach complete hemato- logic response (CHR) at 3 months, major cytogenetic response (MCyR) at 6 months, or complete cytogenetic response (CCyR) at 12 months. Clinical and biological data were obtained from the data file of each patient.

Hematologic and cytogenetic responses have been descri- bed by Deininger et al. [26] and Kantarjian et al. [27]. The local Ethics Committee approved the study, and all par- ticipants agreed and signed the informed consent. Four milliliters of peripheral venous blood were collected in an EDTA tube from each patient and stored at minus 20°C until DNA extraction. The salting-out method was used to extract DNA from white blood cells [28] at the laboratory of genetic and molecular diseases.

Identification ofGSTM1andGSTT1polymorphisms A multiplex polymerase chain reaction (PCR), in which BCL2gene as an internal control was used to identify the genotypes of GSTM1andGSTT1. The PCR mixture con- sisted of 100 ng of genomic DNA, 1X of 5X GoTaq Flexi Buffer (Promega), 1.5 mM of MgCl2, 0.2 mM of each dNTP, 10 pM of each primer and 0.5 U of GoTaq poly- merase (Promega) completed to 25ll with molecular grade water. The forward and reverse primers forGSTT1, GSTM1 and BCL2 were previously described [29]. PCR amplification was carried out with an initial denaturation at 94°C for 5 min and then followed by 35 cycles at 94°C for 1 min, 61°C for 1 min, 72°C for 1 min and a last extension at 72°C for 7 min. DNA fragments were ana- lyzed through a 2 % agarose gel stained with 0.5lg/mL ethidium bromide. Expected product’s sizes were 480, 219 and 154 bp for GSTT1, GSTM1 and BCL2, respectively.

The presence ofBCL2 withoutGSTT1or GSTM1 or both shows their deletion.

Statistical analysis

The effect of GSTM1 and GSTT1 genotypes on hemato- logic, cytogenetic responses, as well as the toxicity related to imatinib was estimated by chi-square test (v2) or Fisher’s exact test. The same test was used to measure the effects of the interaction between GSTT1 andGSTM1on the response to imatinib. A value of p\0.05 was con- sidered significant. The statistical package SPSS version 16 (SPSS Inc., Chicago, IL, USA) was used for the analysis.

(3)

Results

In the present study, we examined the impact ofGSTT1and GSTM1deletions on the response to imatinib in 60 CML patients. The CHR at 3 months showed no particular trend between GSTT1 present and GSTT1 null genotypes (85.1 vs. 92.3 %;v2=0.46; p=0.5). TheGSTM1present and GSTM1 null genotype showed similar trends in terms of hematologic response (84.8 vs. 88.9 %; v2=0.21;

p=0.5) (Table1).

The effect of interaction of different genotype combi- nations betweenGSTT1 andGSTM1showed that the loss of hematologic response (HRL) in patients who have already reached CHR at 3 months was significantly more common in patients carrying the GSTT1 present/GSTM1 present (26.3 %) compared with GSTT1 null/GSTM1 present (12.8 %), GSTT1 present/GSTM1 null carriers (8.3 %) and GSTT1 null/GSTM1 null (0 %) (v2=18.85;

p\0.001) (Fig.1).

However, patients carrying the GSTT1 null genotype were more likely to develop side effects related to imatinib compared with patients harboring the GSTT1 present (31 vs. 16.6 %;v2=6.2;p=0.013) (Fig.2). In the contrast, the distribution of side effects were similar between GSTM1null andGSTM1 present carriers (18.8 vs. 20 %;

v2=0.06; p=0.8) (Fig.3).

As shown in Table1, the rate of CCyR was comparable betweenGSTT1 present andGSTT1null genotype carriers (48.9 vs. 53.8 %). A similar trend was observed between

GSTM1 present and GSTM1 null genotype carriers (48.5 vs. 55.6 %). However, the frequency of none cytogenetic responders (NCyR) was higher in patients carrying the GSTT1 present genotype compared with GSTT1 null car- riers (23.5 vs. 7.7 %); similar results were observed in patients carrying GSTM1 present when compared to GSTM1null carriers (27.2 vs. 11.1 %).

The effect of interaction between the two genes showed that patients carrying the double deletion GSTT1 null/

GSTM1 null were found to be more cytogenetically sen- sitive to imatinib (75 %) followed byGSTT1null/GSTM1 present carriers (55.6 %), GSTT1 present/GSTM1 null (50 %) and GSTT1present/GSTM1present (47.8). On the Table 1 Demographic characteristics and hemato-cytogenetic responses of patients to imatinib according toGSTT1andGSTM1genotypes

Parameters GSTT1 GSTM1

Present N (%) Null N (%) Present N (%) Null N (%)

Sex

Female 30 (85.7) 5 (14.3) 19 (54.3) 16 (45.7)

Male 17 (68) 8 (32) 14 (56) 11 (44)

v2=2.69; df=1;p=0.093 v2=0.02; df=1;p=0.55 Hematologic responsea

Yes 40 (85.1) 12 (92.1) 28 (84.8) 24 (88.9)

No 7 (14.9) 1 (7.7) 5 (15.2) 3 (11.1)

v2=0.46; df=1;p=0.5 v2=0.21; df=1;p=0.5

Cytogenetic responseb

Complete 23 (48.9) 7 (53.8) 16 (48.5) 15 (55.6)

Partial 8 (17) 2 (15.4) 4 (12.2) 5 (18.5)

Minor 4 (8.5) 0 (0) 1 (3) 3 (11.1)

Minimal 1 (2.1) 3 (23.1) 3 (9.1) 1 (3.7)

None 11 (23.5) 1 (7.7) 9 (27.2) 3 (11.1)

v2=9.13; df=4;p=0.058 v2=4.65; df=4;p=0.32

a Hematologic response at 3 months,bcytogenetic response at 18 months

Fig. 1 Distribution of the loss of hematologic response (HRL) in patients, based on different genotype combinations. (Gray means absence of HRL;blackmeans the presence of HRL)

(4)

other hand, the frequency of NCyR to imatinib was higher in patients carrying the GSTT1 present/GSTM1 present genotype (34.8 %) (Table2).

As shown in Fig.4, approximately half of patients car- rying both functional genotypes of GSTT1/GSTM1 have continued with a final dose of 600 mg of imatinib. In addition, the increase of imatinib dose to 600 mg was twice as high among patients with both functional genotypes of GSTT1 and GSTM1 compared with other genotype com- binations. Surprisingly, the dose of 800 mg was observed only in patients carrying at least theGSTT1present in the combination.

Discussion

The GSTs enzymes play an important role in the detoxi- fication, which protects cells from the toxic effects of xenobiotic. Indeed, it is well known that the polymor- phisms in GST enzymes are often associated with a decrease or complete absence of enzymatic activity [30].

Such variability in gene expression linked to inherited genetic differences might influence individual susceptibil- ity to diseases and the response to drugs. It was also reported that overexpression of GSTs enzymes could cause chemoresistance due to the rapid elimination of cancer drugs, resulting in an inadequate therapeutic response [31].

In this study, no significant difference was observed when examining hematologic response for GSTT1 and GSTM1. However, the interaction of GSTT1 and GSTM1 have demonstrated that HRL was more frequent in patients carrying the GSTT1 present/GSTM1 present genotype, while the double deletion GSTT1 null/GSTM1 null appeared to protect patients from HRL, but not signifi- cantly. Indeed, this finding may be explained by possible

rapid elimination of imatinib in patients harboringGSTT1 present/GSTM1present genotype, but also by the low fre- quency of the double deletion in our patients. Stanulla et al.

[22] have reported that theGSTT1orGSTM1deletion was Fig. 2 Distribution of side effects related to imatinib in patients

harboring GSTT1 null or GSTT1 present genotypes. (Gray means absence of HRL;blackmeans the presence of HRL)

Fig. 3 Distribution of side effects related to imatinib in patients harboring GSTM1 null or GSTM1present genotypes. (Gray means absence of side effects;blackmeans the presence of side effects)

Table 2 Effect of the interaction of different genotype combinations between GSTT1 and GSTM1 on the cytogenetic response at 18 months

Response Nulla/ nullb

Nulla/ presentb

Presenta/ nullb

Presenta/ presentb

N (%) N (%) N (%) N (%)

Complete 3 (75) 5 (55.6) 12 (50) 11 (47.8)

Partial 1(25) 0 (0) 5 (20.8) 3 (13)

Minor 0 (0) 0 (0) 3 (12.5) 1 (4.4)

Minimal 0 (0) 3 (33.3) 1 (4.2) 0 (0)

None 0 (0) 1 (11.1) 3 (12.5) 8 (34.8)

v2=20.99; df=12;p=0.050

a GSTT1,bGSTM1

Fig. 4 Variation of the final daily dose of imatinib based on different genotype combinations of genotypes. (Gray means 400 mg; black means 600 mg;Redmeans 800 mg)

(5)

associated with a twofold reduction, in the relapse of acute lymphoblastic leukemia after remission.

The frequency ofGSTT1null genotype was statistically higher in patients who developed side effects when com- pared to patients carrying the GSTT1 present genotype (Fig.2). Mossallam et al. [32] have reported that toxic death appeared to be associated withGSTT1null carriers in patients with acute myeloid leukemia. In contrast, the GSTM1 genotypes showed no particular trend (Fig.1).

Therefore, we can hypothesize that the identification of GSTT1 genotype can predict side effects in patients receiving imatinib.

We found that the rate of CCyR was greater in patients harboring GSTT1 null genotype compared with GSTT1 present carriers, as well as between GSTM1 null and GSTM1present genotypes, but not significantly. This rel- ative difference in the therapeutic response might be explained by the fact that patients carrying the functional genotype of GSTT1 or GSTM1 seem to have a shorter plasma half-life in the metabolism of imatinib compared with subjects with the deleted gene. Moreover, this finding is supported by the relatively high frequency of NCyR in subjects carrying the functional genotypes of GSTT1 or GSTM1(Table1).

The interaction between GSTT1 and GSTM1 revealed that patients bearing the double deletion of GSTT1 and GSTM1genes are more likely to achieve CCyR, unlike to patients carrying both genes at the functional state. It is noteworthy that HRL was also greater in this group of patients. Therefore, we can suggest that a short plasma half-life of imatinib might be associated with the presence of functional genotypes ofGSTT1andGSTM1(Table 2). It was also reported that overexpression of GSTs might expose patients to chemoresistance [21].

Considering the final daily dose of imatinib adminis- tered, we observed that patients bearing the combined GSTT1 present/GSTM1 present genotype were more can- didates at a final dose of 600 mg. Furthermore, theGSTT1 present/GSTM1present genotype appears to be associated with a final dose of 600 or 800 mg. Therefore, we can speculate that patients harboring the combined GSTT1 present/GSTM1present genotype might eliminate imatinib faster than other combinations, which could explain the observed resistance.

Conclusion

We looked the impact of GSTT1 and GSTM1 polymor- phisms of glutathione S-transferase on CML patients receiving imatinib. It appears from the present study that patients carrying the GSTT1 present/GSTM1 present genotype are more prone to HRL. We noticed that the side

effects were more common in patients with GSTT1 null genotype. Furthermore, the frequency of CCyR was higher in patients with GSTT1 null/GSTM1null genotype. How- ever, the rate of NCyR was higher in patients harboring GSTT1 present/GSTM1 present genotype. Therefore, polymorphisms of GSTT1 and GSTM1may influence the clinical and cytogenetic outcome of patients treated with imatinib. Considering the sample size, additional studies are required to confirm our findings.

Acknowledgments The authors thank the Hassan II Academy of Science and Technology for the financial support.

Conflict of interest None.

References

1. Deininger MWN, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood. 2000;96:3343–56.

2. Schiffer CA. BCR-ABL tyrosine kinase inhibitors for chronic myelogenous leukemia. N Engl J Med. 2007;357:258–65.http://

www.ncbi.nlm.nih.gov/pubmed/17634461.

3. Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gat- termann N, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–17.

http://www.ncbi.nlm.nih.gov/pubmed/17151364.

4. Gathmann I, Baccarani M, Cervantes F, Cornelissen JJ, Fischer T, et al. Imatinib compared with interferon and low-dose cyt- arabine for newly-diagnosed chronic-phase chronicmyeloid leu- kemia. N Engl J Med. 2003;348:994–1004.

5. Branford S, Rudzki Z, Walsh S, Parkinson I, Grigg A, Szer J, et al. Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P- loop) are associated with a poor prognosis. Blood. 2003;102:

276–83.http://www.ncbi.nlm.nih.gov/pubmed/12623848.

6. Werner B, Lutz D, Bru¨mmendorf TH, Traulsen A, Balabanov S.

Dynamics of resistance development to imatinib under increasing selection pressure: a combination of mathematical models and in vitro data. PLoS ONE. 2011;6:1–8.

7. Bose P, Park H, Al-Khafaji J, Grant S. Strategies to circumvent the T315I gatekeeper mutation in the Bcr-Abl tyrosine kinase.

Leuk Res Rep. 2013;2:18–20.

8. Vivona D, Bueno CT, Lima LT, Hirata RDC, Hirata MH, Lu- chessi AD, et al. ABCB1 haplotype is associated with major molecular response in chronic myeloid leukemia patients treated with standard-dose of imatinib. Blood cells Mol Dis.

2012;48:132–6.http://www.ncbi.nlm.nih.gov/pubmed/22134106.

9. Kassogue Y, Quachouh M, Dehbi H, Quessar A, Benchekroun S, Nadifi S. Functional polymorphism of CYP2B6 G15631T is associated with hematologic and cytogenetic response in chronic myeloid leukemia patients treated with imatinib. Med Oncol.

(Northwood, London, England). 2014;31:782–6. http://www.

ncbi.nlm.nih.gov/pubmed/24293093.

10. Villuendas R, Steegmann JL, Polla´n M, Tracey L, Granda a, Ferna´ndez-Ruiz E, et al. Identification of genes involved in imatinib resistance in CML: a gene-expression profiling approach. Leukemia. 2006;20:1047–54.http://www.ncbi.nlm.nih.

gov/pubmed/16598311.

11. Dulucq S, Bouchet S, Turcq B, Lippert E, Etienne G, Reiffers J, et al. Multidrug resistance gene (MDR1) polymorphisms are associated with major molecular responses to standard-dose

(6)

imatinib in chronic myeloid leukemia. Blood. 2008;112:2024–7.

http://www.ncbi.nlm.nih.gov/pubmed/18524988.

12. Vivona D, Bueno CT, Lima LT, Hirata RDC, Hirata MH, Lu- chessi AD, et al. ABCB1 haplotype is associated with major molecular response in chronic myeloid leukemia patients treated with standard-dose of imatinib. Blood Cells Mol Dis.

2012;48:132–6.http://www.ncbi.nlm.nih.gov/pubmed/22134106.

13. Sailaja K, Surekha D, Rao DN, Rao DR, Vishnupriya S. Asso- ciation of the GSTP1 gene (Ile105Val) polymorphism with chronic myeloid leukemia. Asian Pac J Cancer Prev. APJCP [Internet]. 2010;11:461–4.http://www.ncbi.nlm.nih.gov/pubmed/

20843134.

14. Koh Y, Kim D-Y, Park S-H, Jung S-H, Park E, Kim H-J, et al.

GSTT1 copy number gain is a poor predictive marker for esca- lated-dose imatinib treatment in chronic myeloid leukemia:

genetic predictive marker found using array comparative geno- mic hybridization. Cancer Genet Cytogenet. 2010;203:215–21.

http://www.ncbi.nlm.nih.gov/pubmed/21156236.

15. Elhoseiny S, El-wakil M, Fawzy M, Rahman AA. GSTP1 (Ile105Val) gene polymorphism: risk and treatment response in chronic myeloid leukemia. J Cancer Ther. 2014;5:1–10.

16. Boyer TD. The glutathione S-transferases: an update. Hepatology.

1989;9:486–96.http://www.ncbi.nlm.nih.gov/pubmed/2646197.

17. Mannervik B, Awasthi YC, Board PG, Hayes JD, Di llo C, Ketterer B, et al. Nomenclature for human glutathione transfer- ases. Biochem J. 1992;282:305–6.

18. Pearson WR, Vorachek WR, Xu SJ, Berger R, Hart I, Vannais D, et al. Identification of class-mu glutathione transferase genes GSTM1 GSTM5 on human chromosome 1p13. Am J Hum Genet.

1993;53:220–33.

19. Webb G, Vaska V, Coggan M, Board P. Chromosomal locali- zation of the gene for the human theta class glutathione trans- ferase (GSTT1). Genomics. 1996;33:121–3. http://www.ncbi.

nlm.nih.gov/pubmed/8617495.

20. Ruzza P, Calderan A. Glutathione transferase (GST)-activated prodrugs. Pharmaceutics. 2013;5:220–31.

21. O’Brien ML, Tew KD. Glutathione and related enzymes in multi- drug resistance. Eur J Cancer (Oxford, England: 1990). 1996;32A:

967–78.http://www.ncbi.nlm.nih.gov/pubmed/10500793.

22. Stanulla M, Schrappe M, Brechlin AM, Zimmermann M, Welte K, Mu A. 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;95:1222–8.

23. Ambrosone CB, Sweeney C, Coles BF, Thompson PA, Mcclure GY, Korourian S, et al. Polymorphisms in glutathione S-trans- ferases (GSTM1 and GSTT1) and survival after treatment for breast cancer after treatment for breast cancer 1. Cancer Res.

2001;61:7130–5.

24. Chen CL, Liu Q, Pui CH, Rivera GK, Sandlund JT, Ribeiro R, et al. Higher frequency of glutathione S-transferase deletions in black children with acute lymphoblastic leukemia. Blood.

1997;89:1701–7.http://www.ncbi.nlm.nih.gov/pubmed/9057653.

25. Davies S, Bhatia S, Ross J, Kiffmeyer W, Gaynon P, Radloff G, et al. Glutathione S-transferase P1 genotypes, genetic suscepti- bility and outcome of therapy in thai childhood acute lympho- blastic leukemia. Blood [Internet]. 2002;100:67–71.http://www.

ncbi.nlm.nih.gov/pubmed/17696749.

26. Deininger MW, O’Brien SG, Ford JM, Druker BJ. Practical management of patients with chronic myeloid leukemia receiving imatinib. J Clin Oncol. 2003;21:1637–47.http://www.ncbi.nlm.

nih.gov/pubmed/12668652.

27. Kantarjian HM, O’Brien S, Cortes J, Giles FJ, Rios MB, Shan J, et al. Imatinib mesylate therapy improves survival in patients with newly diagnosed Philadelphia chromosome-positive chronic myelogenous leukemia in the chronic phase: comparison with historic data. Cancer. 2003;98:2636–42. http://www.ncbi.nlm.

nih.gov/pubmed/14669283.

28. Miller S, Dykes D, Polesky H. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res.

1988;16:1215.

29. Voso MT, D’Alo’ F, Putzulu R, Mele L, Scardocci A, Chiusolo P, et al. Negative prognostic value of glutathione S-transferase (GSTM1 and GSTT1) deletions in adult acute myeloid leukemia.

Blood. 2002;100:2703–7. http://www.ncbi.nlm.nih.gov/pubmed/

12351375.

30. Smith CM, Kelsey KT, Wiencke JK, Leyden K, Levin S, Christiani DC. Inherited glutathione-S-transferase deficiency is a risk factor for pulmonary asbestosis. Cancer Epidemiol Biomarkers Prev.

1994;3:471–7.http://www.ncbi.nlm.nih.gov/pubmed/8000297.

31. McIlwain CC, Townsend DM, Tew KD. Glutathione S-transferase polymorphisms: cancer incidence and therapy. Oncogene. 2006;25:

1639–48. rhttp://www.ncbi.nlm.nih.gov/pubmed/16550164.

32. Mossallam GI, Abdel Hamid TM, Samra M a. Glutathione S-transferase GSTM1 and GSTT1 polymorphisms in adult acute myeloid leukemia; its impact on toxicity and response to che- motherapy. J Egypt Natl Cancer Inst. 2006;18:264–73. http://

www.ncbi.nlm.nih.gov/pubmed/17671537.

Références

Documents relatifs

The level of blast CD33 expression positively impacts the effect of gemtuzumab ozogamicin in patients with acute myeloid leukemia... Andrulis M, Lehners N, Capper

In the present study, a comprehensive analysis of 14 SNPs in genes involved in the NER pathway was performed in 92 early chronic-phase CML patients treated

Les moyens plus limités avec lesquels les patrons de petites entreprises familiales doivent composer dans la gestion de leur établissement aident à comprendre qu’ils soient

IM therapy drives the leukemic population into the "immune window", allowing the patient's autologous immune cells to expand and eventually mount an efficient recognition

A fauna de répteis descrita até ao momento no Jurássico Superior da Bacia Lusitânica está composta por mais de três dezenas de táxons que se distribuem entre

This chapter will present the process that is recommended to follow in order to build a socially sustainable supply chain using SSCM tools. Before presenting the process

This retrospective study, carried out by the French Innovative Leukemia Organization, included 188 tri12 patients with comprehensive information on immunoglobulin heavy chain

Fabien Zoulim, Thierry Poynard, Françoise Degos, Alain Slama, Abdelkhader Al Hasnaoui, Patrick Blin, Florence Mercier, Paul Deny, Paul Landais, Parviz. Parvaz,