Skip to main content

Effects of ABCG2 C421A and ABCG2 G34A genetic polymorphisms on clinical outcome and response to imatinib mesylate, in Iranian chronic myeloid leukemia patients

Abstract

Background

Chronic myeloid leukemia (CML) is a multifactorial clonal myeloid neoplasm that mainly arises from the Philadelphia chromosome. Even though imatinib mesylate (IM) is considered the gold standard for first-line treatment, a number of CML patients have shown IM resistance that can be influenced by many factors, including pharmacogenetic variability. The present study examined whether two common single nucleotide polymorphisms (SNPs) of ABCG2 (G34A and C421A) contribute to IM resistance and/or good responses.

Material and methods

A total of 72 CML patients were genotyped with high-resolution melting (HRM) and restriction fragment length polymorphism-polymerase chain reaction (RFLP-PCR). We also determined the cytogenetic and hematological response, as evaluable factors for measuring response to imatinib.

Results

In the current study, we explored the relationship between the different variants of ABCG2 G34A and C421A and clinical response to imatinib among CML patients. There were no statistically significant differences between genotypes of C421A and G34A and allele frequencies among the resistant and responder groups, with response to IM (P > 0.05). Also, we found no statistically significant association between genotypes and cytogenetic and hematological responses.

Conclusion

This is the first study to investigate the association between genotypes of the G34A and C421A SNPs and the outcome of IM treatment in Iranian population. As a whole, genotyping of these SNPs is unhelpful in predicting IM response in CML patients.

Background

Chronic myeloid leukemia (CML) is a multifactorial clonal myeloid neoplasm originating from malignant hematopoietic stem cells [1,2,3] with an approximate incidence of one to two per 100,000 adults worldwide [4]. The reciprocal translocation (t (9; 22) (q34; q11)) resulting in the Philadelphia (Ph) chromosome formation is pivotal to the pathogenesis of CML [8]. The ensuing oncoprotein named breakpoint cluster region- abelson (BCR-ABL) is an active tyrosine kinase triggering many downstream signaling pathways such as RAS, RAF, JUN kinase, MYC, and STAT, consequently increasing proliferation, apoptosis prevention, mutation accumulation, and genomic instability [2, 4, 5].

CML treatment prior to the 2000s was confined to nonspecific drugs such as busulfan, hydroxyurea, and interferon-alpha (IFN-α) [6]. Alternative therapies, including allogeneic stem cell transplantation (allo-SCT), are also available but come with health risks and mortality [4]. Imatinib mesylate (IM), used to treat a variety of cancers and is a selective tyrosine kinase inhibitor (TKI), is known to be the gold standard of first-line treatment for Philadelphia chromosome-positive CML [7, 8]. The death rate in CML patients has been dropped since the introduction of IM in 2000 [9]. Inactive (closed) form of BCR-ABL is bound by IM, which prevents ATP binding [10]. Following this interaction, the subsequent phosphorylation and activation of downstream pathways are inhibited, resulting in apoptosis promotion of leukemic cells, and inhibiting leukemogenesis [11,12,13]. In line with the results of recent studies, IM significantly improved CML patients' clinical outcomes and prognoses. However, despite its remarkable efficacy, cancer treatment is hampered in some cases by primary or acquired resistance to IM and severe side effects [11, 14,15,16,17,18,19]. Increasing the dose of IM from 400 to 800 mg/day or switching to second-generation tyrosine kinase inhibitors such as dasatinib or nilotinib is recommended when there is no adequate response to IM or treatment fails [20].

IM resistance can be attributed to BCR-ABL-dependent and BCR-ABL-independent mechanisms [21, 22]. Mutations in BCR-ABL's tyrosine kinase domain, which is the most common cause of resistance, and overexpression of the BCR-ABL protein are examples of BCR-ABL-dependent mechanisms [15, 22,23,24,25,26]. BCR-ABL-independent causes can include activation of downstream oncogenic pathways, alterations of apoptosis-related genes, and IM pharmacokinetics (IM blood and/or intracellular levels), which are mostly still unknown [27,28,29,30,31].

CML patients' drug response and IM pharmacokinetics are influenced by efflux and influx transporters, such as ABCB1, ABCG2, and OCT1 (SLC22A1) [32,33,34,35,36,37,38]. There has been evidence that genetic polymorphisms in such genes lead to transporter function alteration and affect IM response [21, 39]. Previous studies have analyzed the association between ABCG2 (ATP-binding cassette subfamily G member 2) polymorphisms and pharmacokinetic inter-individual variations, along with the efficacy and toxicity of drugs [40,41,42]. To date, more than 80 ABCG2 single nucleotide polymorphisms (SNPs) have been detected in various populations via direct DNA sequencing [43, 44]. The two common ABCG2 variants are G34A (V12M, rs2231137) and C421A (Q141K, rs2231142), which are located on exon 2 and 5, respectively [21, 41, 45]. In this study, we investigated the relationship between these two SNPs and the IM response/resistance among Iranian CML patients.

Material and methods

Study subjects

The study involved 72 Ph-positive CML patients (37 females and 35 males) receiving 400 mg IM (Gleevec, STI571 (signal transduction inhibitor 571), and CGP57148B) daily for at least three months. Before participation in this study, these patients with chronic phase CML were diagnosed by the Department of Hematology, Omid Hospital (Isfahan, Iran). Written informed consent was obtained from all patients, and peripheral blood samples were collected in EDTA-containing tubes. Participants taking IM metabolism-interfering drugs, such as Phenobarbital and Phenytoin, were excluded. At the time of diagnosis, Philadelphia chromosome and BCR-ABL fusion mRNA presence were confirmed, respectively, by cytogenetic analysis and RT-PCR technique. Table 1 summarizes the demographic and clinical characteristics of patients. This study was approved by the Yazd University of Medical Sciences local Ethics Committee (IR.SSU.MEDICINE.REC.1398.091).

Table 1 Demographic and clinical characteristics of study subjects

Clinical assessment

The guidance for evaluation of clinical response and ordinary follow-up of patients were chased as given in “European Leukemia Net: guideline for managing CML patients”. Bone marrow morphology and cytogenetic studies were utilized for early diagnosis of patients and definition of the phase of the disease at the time of clinical demonstration. The primary treatment prescribed was 400 mg of IM [46].

The response to the drug was evaluated at regular intervals. For this reason, hematological (WBC count < 10 × 109/L; basophils < 5%; an absence of myelocytes, promyelocytes, or myeloblasts in peripheral blood; platelet counts < 450 × 109/L) and molecular (BCR-ABL1/ABL1 ≤ 0.1%) response every 3 months, and the cytogenetic (0% Ph + meta phases) response at 6 months was assessed [47, 48].

An indication of resistance can be determined by a lack of complete hematological response at 3 months, complete cytogenetic response (0% Ph-positive metaphases) at intervals of 12 months and (MMR) major molecular response (BCR-ABLIS ≤ 0.1%) at 18 months [47].

In this study, CML patients were followed up at an interval of 6 months. During this period, hematological, cytogenetic, and molecular responses were evaluated.

DNA extraction and SNP genotyping

Genomic DNA was extracted from peripheral blood lymphocytes using the PrimePrep Genomic DNA isolation kit (Genet Bio, Korea) based on the manufacturer’s protocol. Qualitative and quantitative assessments of the isolated DNA were examined utilizing electrophoresis on 2% agarose gel and Thermo Scientific NanoDrop 2000 Spectrophotometer, respectively.

The G34A polymorphism was genotyped utilizing the allelic discrimination assay by High-Resolution Melting (HRM) technique (Rotor Gene-6000, Corbett Research, Sydney, Australia). PCR was performed in a final volume of 20 μL PCR mixture using HOT FIREPol® EvaGreen® HRM Mix (Solis BioDyne, Estonia). The HRM conditions were as follows: one cycle of 95 °C for 12 min to activate HOT FIREPOL DNA polymerase; 40 cycles of 95 °C for 15 s, 58 °C for 20 s, 72 °C for 20 s; and a HRM step from 76 to 86 °C rising at 0.2 °C with 2-s hold time after each step. The results were analyzed by Q 5plex HRM software V.2.3.4. Having found no feasible 80–100-bp primers, PCR–RFLP-based genotyping using the BseMI enzyme was designed to genotype the C421A polymorphism. After amplification DNA fragments by PCR, they were cut by 2 units of restriction enzyme BseMI for 16 h at 55 °C. Then, DNA fragments created after digestion run on a 3% agarose gel. Electrophoresis of these fragments could distinguish the genotypes based on fragments length. Primer sequences were designed using GeneRunner software. Primer sequences were as follows: Forward 5′AGGATGATGTTGTGATGGGC3′, Reverse 5′TGACCCTGTTAATCCGTTCG3′ for C421A and Forward 5′ GTTGTGCCTGTCTTCCCAT 3′, Reverse 5′ TCGACAAGGTAGAAAGCCAC 3′ for G34A. We randomly sequenced 10% of the samples using the ABI 3730XL automatic sequencer (Applied Biosystems, USA) to confirm results.

Statistical analysis

Data analysis was performed using SPSS software package version 26 (SPSS Inc., Chicago, IL, USA). The Chi-square test and independent samples t-test were used to assess the differences between demographics and response/resistance to IM. Then, the binary logistic model was exploited to calculate odds ratios (ORs) and 95% confidence intervals (CIs). We also performed Wilcoxon analysis and paired T-test to find out whether genotypes were associated with hematological responses. A P value lower than 0.05 was considered statistically significant.

Results

Table 1 lists the demographic and clinical characteristics of study participants. There was no statistically significant difference between the response to treatment and age (P = 0.179) or sex (P = 0.851). Furthermore, neither C421A (P = 0.146, P = 0.170) nor G34A (P = 0.235, P = 0.398) genotype and allele frequencies provided any statistically significant association with response to IM (Tables 2, 3).

Table 2 C421 and G34A genotype frequencies in the responder and resistant groups
Table 3 C421 and G34A allele frequencies in the responder and resistant groups

A hematologic index (Hb, WBC, and Plt count) analysis was also performed to find out how well the drug altered responder and resistant patients' hematologic profiles. Comparing the primary and secondary responses before and after drug intake, no significant relationship between drug use and hematological index was revealed, according to the data provided in Table 4. Moreover, genotypes did not significantly affect hematologic outcomes (WBC, Plt, and Hb count) (Table 5) and cytogenetic response (Table 6).

Table 4 The comparison of hematological response between patients with different genotypes
Table 5 The comparison of hematological response between responder and resistant groups
Table 6 The comparison of cytogenetic response between different genotype groups

Discussion

Two types of ABCG2 variants, C421A and G34A, have been extensively studied in order to predict the IM response among CML patients. However, the results were noticeably contrasting. We compared the distribution of ABCG2 C421A and ABCG2 G34A polymorphism genotypes among responding and resistant CML patients treated with IM. According to our results, these SNPs did not show significantly different distributions between the two groups. Between the genotypes C421A and G34A of ABCG2 and the IM response, no statistically significant association could be established [47]. Also, ABCG2 C421A was not effective in reaching an optimal response to IM treatment [49, 50]. In comparison with ABCG2 C421A polymorphism, ABCG2 G34A polymorphism has a contradictory effect on IM response [51]. In study carried out in 2016, ABCG2 is not involved in forecasting optimal molecular responses after IM consumption [52]. In the present study, no information on patients' molecular responses was available.

Furthermore, we found that the frequencies of these alleles were insignificantly different between these two groups and were not found to be risk factors for resistance. Our results are in disagreement with some studies that the ABCG2 polymorphism is associated with response to IM [2, 21, 53].

The association of each SNP genotype of ABCG2 G34A and C421A with hematological responses (WBC, Hb, and Plt counts) was also examined. IM is effective in the hematological response since BCR-ABL protein is effective in proliferation of blood cells (myeloid), and BCR-ABL is IM's target, although according to Table 4, hematological indices did not vary significantly between the groups with different genotypes and drug therapy with IM did not significantly influence primary or secondary hematological response. In this regard, similarly results from study of CML patients in western of Iran showed no statistically significant correlation between ABCG2 C421A and hematological response (Hb, WBC, and Plt counts) [2]. The disconnection needs to be confirmed by further investigation.

Several studies have shown that the association between SNPs genotypes and cytogenetic response is more important for evaluating drug response than the relationship between SNPs genotypes and hematological response. According to our findings, no statistically significant relationship was found between G34A and C421A variants and cytogenetic response in both groups. Similarly, no significant correlation for ABCG2 C421A has been reported [21, 54]. Also, no statistically significant association has been observed between cytogenetic response and SNP genotype of ABCG2 C421A and G34A, which is consistent with some previously published research [47].

Conclusion

To sum up, our study reveals that ABCG2/C421A and ABCG2/G34A polymorphisms are insignificantly associated with the optimal response rate to IM and cytogenetic/hematologic response and cannot be used as a predictive marker for optimal response/primary failure in CML patients receiving IM. Nevertheless, our study is the first that investigates the association between hematologic index among responder and resistant groups, as well as the association between these indexes and drug consumption. Our study had some restrictions such as not checking the molecular response test. In addition, the statistical population of the present study for assessing the relationship between response to treatment and combined SNPs was small. Therefore, additional studies with larger and different populations for evaluating the relationship between ABCB1, SLC22A1, CYP3A4, and CYP3A5 polymorphisms and response/resistance to IM are suggested.

Availability of data and materials

The data that support the findings of this study are available on request from the corresponding author.

Abbreviations

CML:

Chronic myeloid leukemia

IM:

Imatinib mesylate

Ph:

Philadelphia

SNP:

Single nucleotide polymorphisms

TKI:

Tyrosine kinase inhibitor

References

  1. Chorzalska A, Ahsan N, Rao RSP, Roder K, Yu X, Morgan J et al (2018) Overexpression of Tpl2 is linked to imatinib resistance and activation of MEK-ERK and NF-κB pathways in a model of chronic myeloid leukemia. Mol Oncol 12(5):630–647

    Article  CAS  Google Scholar 

  2. Salimizand H, Amini S, Abdi M, Ghaderi B, Azadi NA (2016) Concurrent effects of ABCB1 C3435T, ABCG2 C421A, and XRCC1 Arg194Trp genetic polymorphisms with risk of cancer, clinical output, and response to treatment with imatinib mesylate in patients with chronic myeloid leukemia. Tumour Biol 37(1):791–798

    Article  CAS  Google Scholar 

  3. Louati N, Turki F, Mnif H, Frikha R (2022) MDR1 gene polymorphisms and imatinib response in chronic myeloid leukemia: a meta-analysis. J Oncol Pharm Pract 28(1):39–48

    Article  CAS  Google Scholar 

  4. Jabbour E, Kantarjian H (2020) Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am J Hematol 95(6):691–709

    Article  CAS  Google Scholar 

  5. Gromicho M, Dinis J, Magalhães M, Fernandes AR, Tavares P, Laires A et al (2011) Development of imatinib and dasatinib resistance: dynamics of expression of drug transporters ABCB1, ABCC1, ABCG2, MVP, and SLC22A1. Leuk Lymphoma 52(10):1980–1990

    Article  CAS  Google Scholar 

  6. Jabbour E, Kantarjian H (2022) Chronic myeloid leukemia: 2022 update on diagnosis, therapy, and monitoring. Am J Hematol 97:1236–1256

    Article  CAS  Google Scholar 

  7. Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP et al (2017) Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med 376(10):917–927

    Article  CAS  Google Scholar 

  8. Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N et al (2006) Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med 355(23):2408–2417

    Article  CAS  Google Scholar 

  9. American Cancer Society (2015) Cancer facts & figures 2015. American Cancer Society, New York

    Google Scholar 

  10. Tanis KQ, Veach D, Duewel HS, Bornmann WG, Koleske AJ (2003) Two distinct phosphorylation pathways have additive effects on Abl family kinase activation. Mol Cell Biol 23(11):3884–3896

    Article  CAS  Google Scholar 

  11. Mughal TI, Radich JP, Deininger MW, Apperley JF, Hughes TP, Harrison CJ et al (2016) Chronic myeloid leukemia: reminiscences and dreams. Haematologica 101(5):541

    Article  CAS  Google Scholar 

  12. Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, Kuriyan J (2000) Structural mechanism for STI-571 inhibition of abelson tyrosine kinase. Science 289(5486):1938–1942

    Article  CAS  Google Scholar 

  13. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S et al (1996) Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med 2(5):561–566

    Article  CAS  Google Scholar 

  14. Gotta V, Bouchet S, Widmer N, Schuld P, Decosterd LA, Buclin T et al (2014) Large-scale imatinib dose–concentration–effect study in CML patients under routine care conditions. Leuk Res 38(7):764–772

    Article  CAS  Google Scholar 

  15. Quintás-Cardama A, Kantarjian HM, Cortes JE (2009) Mechanisms of primary and secondary resistance to imatinib in chronic myeloid leukemia. Cancer Control 16(2):122–131

    Article  Google Scholar 

  16. Zhang WW, Cortes JE, Yao H, Zhang L, Reddy NG, Jabbour E et al (2009) Predictors of primary imatinib resistance in chronic myelogenous leukemia are distinct from those in secondary imatinib resistance. J Clin Oncol 27(22):3642

    Article  CAS  Google Scholar 

  17. Karabay AZ, Koc A, Ozkan T, Hekmatshoar Y, Altinok Gunes B, Sunguroglu A et al (2018) Expression analysis of Akirin-2, NFκB-p65 and β-catenin proteins in imatinib resistance of chronic myeloid leukemia. Hematology 23(10):765–770

    Article  CAS  Google Scholar 

  18. Housman G, Byler S, Heerboth S, Lapinska K, Longacre M, Snyder N et al (2014) Drug resistance in cancer: an overview. Cancers 6(3):1769–1792

    Article  Google Scholar 

  19. Rabian F, Lengline E, Rea D (2019) Towards a personalized treatment of patients with chronic myeloid leukemia. Curr Hematol Malig Rep 14(6):492–500

    Article  Google Scholar 

  20. Lardo M, Castro M, Moiraghi B, Rojas F, Borda N, Rey JA et al (2015) MDR1/ABCB1 gene polymorphisms in patients with chronic myeloid leukemia. Blood Res 50(3):154–159

    Article  CAS  Google Scholar 

  21. Au A, Aziz Baba A, Goh AS, Wahid Fadilah SA, Teh A, Rosline H et al (2014) Association of genotypes and haplotypes of multi-drug transporter genes ABCB1 and ABCG2 with clinical response to imatinib mesylate in chronic myeloid leukemia patients. Biomed Pharmacother 68(3):343–349

    Article  CAS  Google Scholar 

  22. Jabbour E, Kantarjian H, Jones D, Talpaz M, Bekele N, O’Brien S et al (2006) Frequency and clinical significance of BCR-ABL mutations in patients with chronic myeloid leukemia treated with imatinib mesylate. Leukemia 20(10):1767–1773

    Article  CAS  Google Scholar 

  23. Azevedo AP, Reichert A, Afonso C, Alberca MD, Tavares P, Lima F (2017) BCR-ABL V280G mutation, potential role in Imatinib resistance: first case report. Clin Med Insights Oncol 11:1179554917702870

    Article  Google Scholar 

  24. Kaplan JB, Platanias LC (2017) Another tyrosine kinase inhibitor-resistance mutation within the BCR-ABL kinase domain: chasing our tails? Leuk Lymphoma 58(7):1526–1527

    Article  CAS  Google Scholar 

  25. Kantarjian HM, Talpaz M, Giles F, O’Brien S, Cortes J (2006) New insights into the pathophysiology of chronic myeloid leukemia and imatinib resistance. Ann Intern Med 145(12):913–923

    Article  Google Scholar 

  26. La Rosée P, Hochhaus A (2010) Molecular pathogenesis of tyrosine kinase resistance in chronic myeloid leukemia. Curr Opin Hematol 17(2):91–96

    Article  Google Scholar 

  27. Elias MH, Azlan H, Sulong S, Baba AA, Ankathil R (2018) Aberrant DNA methylation at HOXA4 and HOXA5 genes are associated with resistance to imatinib mesylate among chronic myeloid leukemia patients. Cancer Rep 1(2):e1111

    Article  Google Scholar 

  28. Diamond JM, Melo JV (2011) Mechanisms of resistance to BCR–ABL kinase inhibitors. Leuk Lymphoma 52(sup1):12–22

    Article  CAS  Google Scholar 

  29. Kim DHD, Sriharsha L, Xu W, Kamel-Reid S, Liu X, Siminovitch K et al (2009) Clinical relevance of a pharmacogenetic approach using multiple candidate genes to predict response and resistance to imatinib therapy in chronic myeloid leukemia. Clin Cancer Res 15(14):4750–4758

    Article  CAS  Google Scholar 

  30. Donato NJ, Wu JY, Stapley J, Lin H, Arlinghaus R, Aggarwal B et al (2004) Imatinib mesylate resistance through BCR-ABL independence in chronic myelogenous leukemia. Can Res 64(2):672–677

    Article  CAS  Google Scholar 

  31. Dai Y, Rahmani M, Corey SJ, Dent P, Grant S (2004) A Bcr/Abl-independent, Lyn-dependent form of imatinib mesylate (STI-571) resistance is associated with altered expression of Bcl-2. J Biol Chem 279(33):34227–34239

    Article  CAS  Google Scholar 

  32. Fletcher JI, Haber M, Henderson MJ, Norris MD (2010) ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer 10(2):147–156

    Article  CAS  Google Scholar 

  33. Allen JD, Schinkel AH (2002) Multidrug resistance and pharmacological protection mediated by the breast cancer resistance protein (BCRP/ABCG2) 1 supported in part by Dutch Cancer Society Grants NKI 97-1433 and NKI 97-1434 (to AHS). 1. Mol Cancer Ther 1(6):427–434

    CAS  Google Scholar 

  34. Gottesman MM, Fojo T, Bates SE (2002) Multidrug resistance in cancer: role of ATP–dependent transporters. Nat Rev Cancer 2(1):48–58

    Article  CAS  Google Scholar 

  35. Litman T, Druley T, Stein W, Bates S (2001) From MDR to MXR: new understanding of multidrug resistance systems, their properties and clinical significance. Cell Mol Life Sci CMLS 58(7):931–959

    Article  CAS  Google Scholar 

  36. Miao ZH, Tang T, Zhang YX, Zhang JS, Ding J (2003) Cytotoxicity, apoptosis induction and downregulation of MDR-1 expression by the anti-topoisomerase II agent, salvicine, in multidrug-resistant tumor cells. Int J Cancer 106(1):108–115

    Article  CAS  Google Scholar 

  37. Yagüe E, Higgins C, Raguz S (2004) Complete reversal of multidrug resistance by stable expression of small interfering RNAs targeting MDR1. Gene Ther 11(14):1170–1174

    Article  Google Scholar 

  38. Munteanu E, Verdier M, Grandjean-Forestier F, Stenger C, Jayat-Vignoles C, Huet S et al (2006) Mitochondrial localization and activity of P-glycoprotein in doxorubicin-resistant K562 cells. Biochem Pharmacol 71(8):1162–1174

    Article  CAS  Google Scholar 

  39. de Lima LT, Vivona D, Bueno CT, Hirata RD, Hirata MH, Luchessi AD et al (2014) Reduced ABCG2 and increased SLC22A1 mRNA expression are associated with imatinib response in chronic myeloid leukemia. Med Oncol (Northwood, Lond, Engl) 31(3):851

    Article  Google Scholar 

  40. Woodward OM, Köttgen A, Coresh J, Boerwinkle E, Guggino WB, Köttgen M (2009) Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci 106(25):10338–10342

    Article  CAS  Google Scholar 

  41. Keskitalo J, Zolk O, Fromm MF, Kurkinen K, Neuvonen PJ, Niemi M (2009) ABCG2 polymorphism markedly affects the pharmacokinetics of atorvastatin and rosuvastatin. Clin Pharmacol Ther 86(2):197–203

    Article  CAS  Google Scholar 

  42. Sissung TM, Baum CE, Kirkland CT, Gao R, Gardner ER, Figg WD (2010) Pharmacogenetics of membrane transporters: an update on current approaches. Mol Biotechnol 44(2):152–167

    Article  CAS  Google Scholar 

  43. El Mesallamy HO, Rashed WM, Hamdy NM, Hamdy N (2014) High-dose methotrexate in Egyptian pediatric acute lymphoblastic leukemia: the impact of ABCG2 C421A genetic polymorphism on plasma levels, what is next? J Cancer Res Clin Oncol 140(8):1359–1365

    Article  Google Scholar 

  44. Sparreboom A, Nooter K (2000) Does P-glycoprotein play a role in anticancer drug pharmacokinetics? Drug Resist Updates 3(6):357–363

    Article  CAS  Google Scholar 

  45. Bruhn O, Cascorbi I (2014) Polymorphisms of the drug transporters ABCB1, ABCG2, ABCC2 and ABCC3 and their impact on drug bioavailability and clinical relevance. Expert Opin Drug Metab Toxicol 10(10):1337–1354

    Article  CAS  Google Scholar 

  46. Baccarani M, Cortes J, Pane F, Niederwieser D, Saglio G, Apperley J et al (2009) Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. J Clin Oncol 27(35):6041

    Article  CAS  Google Scholar 

  47. Nair D, Dhangar S, Shanmukhaiah C, Vundinti BR (2017) Association of genetic polymorphisms of the ABCG2, ABCB1, SLCO1B3 genes and the response to Imatinib in chronic myeloid leukemia patients with chronic phase. Meta Gene 11:14–19

    Article  Google Scholar 

  48. Gardner E (2008) Factors affecting pharmacokinetic variability of imatinib mesylate (Gleevec, STI-571)

  49. Nambu T, Hamada A, Nakashima R, Yuki M, Kawaguchi T, Mitsuya H et al (2011) Association of SLCO1B3 polymorphism with intracellular accumulation of imatinib in leukocytes in patients with chronic myeloid leukemia. Biol Pharm Bull 34(1):114–119

    Article  CAS  Google Scholar 

  50. Belohlavkova P, Vrbacky F, Voglova J, Racil Z, Zackova D, Hrochova K et al (2018) The significance of enzyme and transporter polymorphisms for imatinib plasma levels and achieving an optimal response in chronic myeloid leukemia patients. Arch Med Sci 14:1416

    Article  CAS  Google Scholar 

  51. Niebudek K, Balcerczak E, Mirowski M, Pietrzak J, Zawadzka I, Żebrowska-Nawrocka M (2019) The contribution of ABCG2 G34A and C421A polymorphisms to multiple myeloma susceptibility. Onco Targets Ther 12:1655

    Article  CAS  Google Scholar 

  52. da Cunha VF, Mauricio Scheiner MA, Moellman-Coelho A, Mencalha AL, Renault IZ, Rumjanek VM et al (2016) Low ABCB1 and high OCT1 levels play a favorable role in the molecular response to imatinib in CML patients in the community clinical practice. Leuk Res 51:3–10

    Article  Google Scholar 

  53. Omran MM, Abdelfattah R, Moussa HS, Alieldin N, Shouman SA (2020) Association of the trough, peak/trough ratio of imatinib, pyridine-N-oxide imatinib and ABCG2 SNPs 34 G>A and SLCO1B3 334 T>G with imatinib response in Egyptian chronic myeloid leukemia patients. Front Oncol 10:1348

    Article  Google Scholar 

  54. Seong S, Lim M, Sohn S, Moon J, Oh S-J, Kim BS et al (2013) Influence of enzyme and transporter polymorphisms on trough imatinib concentration and clinical response in chronic myeloid leukemia patients. Ann Oncol 24(3):756–760

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to thank all patients and health staff (of Genome Medical Genetics Center and Seyed Al-Shohada Hospital in Isfahan, Iran) who participated in this study. Also, we thank DR. Mohammad Forat Yazdi and DR. Fatemeh Shishe Bor who contributed to sample collection. In addition, we thank DR. Mohammad Kazemi who involved in performing this project.

Funding

For this research, the author(s) did not receive any funding.

Author information

Authors and Affiliations

Authors

Contributions

NN contributed significantly to the preparation of this study by collected samples from Sayed Al-Shohada Hospital, designing the protocol of work, contributing to practical part of the study, and writing and editing the manuscript. VM, ZK, and MK contributed to collect samples from Sayed Al-Shohada Hospital and provide patients’ data. Also, ZK contributed to practical part of the study, especially PCR–RFLP technique. EZ, EA, and FZ edited the manuscript, and analyzed and interpreted the patient data. SK was primarily responsible for the writing of the manuscript. All authors have read and approved the manuscript.

Corresponding authors

Correspondence to Seyed Mehdi Kalantar or Mansoor Salehi.

Ethics declarations

Ethics approval and consent to participate

This research was approved by the Yazd University of Medical Sciences Local Ethics Committee, and informed written consent was taken from every participant in the study; the committee’s reference number is IR.SSU.MEDICINE.REC.1398.091.

Consent for publication

Consent to publish from the patient had been taken.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nouri, N., Mehrzad, V., Khalaj, Z. et al. Effects of ABCG2 C421A and ABCG2 G34A genetic polymorphisms on clinical outcome and response to imatinib mesylate, in Iranian chronic myeloid leukemia patients. Egypt J Med Hum Genet 24, 1 (2023). https://doi.org/10.1186/s43042-022-00379-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43042-022-00379-6

Keywords