Next Article in Journal
A Biomedical Investigation of the Hepatoprotective Effect of Radix salviae miltiorrhizae and Network Pharmacology-Based Prediction of the Active Compounds and Molecular Targets
Next Article in Special Issue
Circulating Cell-Free DNA and Circulating Tumor Cells as Prognostic and Predictive Biomarkers in Advanced Non-Small Cell Lung Cancer Patients Treated with First-Line Chemotherapy
Previous Article in Journal
CmMYB19 Over-Expression Improves Aphid Tolerance in Chrysanthemum by Promoting Lignin Synthesis
Previous Article in Special Issue
Predictive and Prognostic Molecular Biomarkers for Response to Neoadjuvant Chemoradiation in Rectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Polymorphisms Contribute to the Individual Variations of Imatinib Mesylate Plasma Levels and Adverse Reactions in Chinese GIST Patients

1
Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai 201203, China
2
Department of Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
3
Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
4
Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
5
Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2017, 18(3), 603; https://doi.org/10.3390/ijms18030603
Submission received: 31 December 2016 / Revised: 27 February 2017 / Accepted: 4 March 2017 / Published: 13 March 2017

Abstract

:
Imatinib mesylate (IM) has dramatically improved the outcomes of gastrointestinal stromal tumor (GIST) patients. However, the clinical responses of IM may considerably vary among single individuals. This study aimed to investigate the influences of genetic polymorphisms of drug-metabolizing enzyme (CYP3A4), transporters (ABCB1, ABCG2), and nuclear receptor (Pregnane X Receptor (PXR, encoded by NR1I2)) on IM plasma levels and related adverse reactions in Chinese GIST patients. A total of 68 Chinese GIST patients who have received IM 300–600 mg/day were genotyped for six single nucleotide polymorphisms (SNPs) (CYP3A4 rs2242480; ABCB1 rs1045642; ABCG2 rs2231137; NRI12 rs3814055, rs6785049, rs2276706), and the steady-state IM trough plasma concentrations were measured by a validated HPLC method. There were statistically significant variances in the steady-state IM trough plasma concentrations (from 272.22 to 4365.96 ng/mL). Subjects of GG in rs2242480, T allele carriers in rs1045642 and CC in rs3814055 had significantly higher steady-state IM dose-adjusted trough plasma concentrations. Subjects of CC in rs3814055 had significantly higher incidence rate of edema. The genetic polymorphisms of rs2242480, rs1045642, rs3814055 were significantly associated with IM plasma levels, and the genetic variations of rs3814055 were significantly associated with the incidence rate of edema in Chinese GIST patients. The current results may serve as valuable fundamental knowledge for IM therapy in Chinese GIST patients.

Graphical Abstract

1. Introduction

Gastrointestinal stromal tumors (GISTs) are epidemiologically analyzed to be the most common form of the mesenchymal tumor of the gastrointestinal tract, as the worldwide incidence and prevalence are estimated to be approximately 1 to 1.5 per 100,000 per year and 13 per 100,000, respectively [1]. At the present time, the combined application of surgery resection and molecular targeted drugs therapy is the most effective treatment for patients with the middle or high risk GISTs. Imatinib mesylate (IM) is a first-line targeted therapy for inoperable, metastatic, or recurrent KIT-positive GIST and for the adjuvant treatment of patients following resection of primary KIT-positive GIST [2]. After the clinical introduction of IM, the patient outcomes for GIST have been dramatically improved, with an impressive impact on both quality of life and long-term prognosis. However, there are large individual variations in clinical efficacy and adverse reactions of IM [3,4,5]. Ten to fifteen percent of patients who underwent IM treatment after 3–6 months progressed rapidly to widespread metastatic disease [6], about 30% of patients who developed serious adverse reactions had to stop taking medication [7]. Recently, the relationship between IM plasma concentrations and efficacy and toxicity have been described [8,9], minimal plasma concentration thresholds (1100 ng/mL) have been established, under which a substantial increase in treatment failure and drug resistance was observed [10]. Thus, inter-individual variations in IM pharmacokinetics may, therefore, have important clinical consequences. This implicates that timely monitoring of IM plasma concentration is warranted in GIST patients.
It has been found that genetic polymorphisms of main drug-metabolizing enzymes and transporters may significantly influence the inter-individual variations in drug reaction and disposition [11]. IM is orally administrated and was mainly metabolized in the liver by cytochrome P450 3A4 (CYP3A4), and effluxed by ATP Binding Cassette Subfamily B Member 1(ABCB1, P-glycoprotein), ATP Binding Cassette Subfamily G Member 2 (ABCG2, BCRP), etc. The current studies of individualized therapy for IM mainly focused on the genetic polymorphisms of main drug-metabolizing enzymes (CYP3A4) [12] and transporters (ABCB1, ABCG2) [13,14]. However, the variant differences in IM clinical effects and adverse reactions remains controversial, suggesting that there are some other genetic factors neglected. In addition, CYP3A4 rs2242480, the highest frequency single nucleotide polymorphisms (SNPs) in the Chinese population [15] was found to be correlated with the increased activity of CYP3A4, indicating that it is likely to be associated with IM pharmacokinetics.
Recently, nuclear receptors, as domain transcriptional regulators, have been found to play a crucial role in regulating the expression of relevant drug metabolic enzymes and transporters. Pregnane X Receptor (PXR, encoded by NR1I2), a member of the nuclear receptor superfamily, was increasingly found in simultaneously inducing the expression of CYP3A4 [16,17,18,19] and ABCB1 [17,18,20], predicting that the genetic polymorphisms of NRI12 may have an effect on IM pharmacokinetics.
Based on these observations, six SNPs (CYP3A4 rs2242480; ABCB1 rs1045642; ABCG2 rs2231137; NRI12 rs3814055, rs6785049, rs2276706), which have been associated with the expression and/or function of the above drug-metabolizing enzyme and transporters genes and/or proteins in IM pharmacokinetics pathway, and with the minor allele frequency higher than 10% in Asian, were chosen as candidate SNPs, in order to investigate the influence of CYP3A4, ABCB1, ABCG2 and NR1I2 genetic polymorphisms on the steady-state IM dose-adjusted trough plasma concentrations and related adverse reactions in Chinese GIST patients. The results can be served as valuable fundamental knowledge for IM therapy, in order to make the antitumor treatment more successfully, and increase the safety and long-term tolerability of IM in Chinese GIST patients.

2. Results

2.1. Patients Characteristics and IM trough Plasma Concentrations

Sixty-eight Chinese GIST patients were enrolled, including 39 males and 29 females, whose characteristics and biological values are shown in Table 1. There are significant differences in IM trough plasma concentrations, and the mean IM trough plasma concentration of the study population is 1134.30 ng/mL, ranging from 272.22 to 4365.96 ng/mL (Table 2). About 47.06% of patients, whose IM trough plasma concentrations are under the minimal plasma concentration thresholds (1100 ng/mL), this may indicate a high risk of disease progression and treatment failure.

2.2. Genotype Frequencies

Frequencies of the six SNPs’ genotypes in the study population are shown in Table 3. The frequency expected for each genotype was evaluated on the basis of Hardy–Weinberg equilibrium. None of the observed SNPs’ frequencies was significantly different from the expected frequencies (p > 0.05), illustrating that all of the six SNPs’ frequencies in the study are in accordance with Hardy–Weinberg equilibrium. There is almost no linkage disequilibrium among all the six SNPs, and six SNPs in the study were in genetic equilibrium.
Comparisons of mutation frequencies for the six SNPs above, among different ethnic groups, are shown in Table 4. None of the observed SNPs’ frequencies in the study population were significantly different from the Healthy Han Chinese and Asian, but all six SNPs’ mutation frequencies were significantly different from Caucasian and African. These SNPs may contribute to inter-individual variations of IM reaction and disposition among different ethnic groups.

2.3. Effect of Genetic Polymorphisms of Drug-Metabolizing Enzyme CYP3A4 on IM Plasma Levels

For CYP3A4 rs2242480, the steady-state IM dose-adjusted trough plasma concentrations in mutant allele A carriers (GA + AA) (2.27 ± 0.32 ng/mL/mg) were significantly lower than that in wild-types (GG) (4.12 ± 0.40 mg/mL/mg) (p = 0.0171) (Figure 1, Table 5).

2.4. Effect of Genetic Polymorphisms of Transporters ABCB1/ABCG2 on IM Plasma Levels

For ABCB1 rs1045642, the steady-state IM dose-adjusted trough plasma concentrations in mutant allele T carriers (CT + TT) (4.36 ± 0.45 ng/mL/mg) were significantly higher than that in wild-types (CC) (2.56 ± 0.33 mg/mL/mg) (p = 0.0055) (Figure 2a, Table 6).
For ABCG2 rs2231137, there is no significant difference in the mean steady-state IM dose-adjusted trough plasma concentration in observed genotypes (p > 0.05) (Figure 2b, Table 6).

2.5. Effect of Genetic Polymorphisms of Nuclear Receptor PXR on IM Plasma Levels

For NR1I2 rs3814055, the steady-state IM dose-adjusted trough plasma concentrations in mutate allele T carriers (CT + TT) (2.34 ± 0.25 ng/mL/mg) were significantly lower than that in wild-types (CC) (4.26 ± 0.43 mg/mL/mg) (p = 0.0066) (Figure 3a, Table 7).
For NR1I2 rs6785049 and rs2276706, there is no significant difference in the mean steady-state IM dose-adjusted trough plasma concentrations in observed genotypes (p > 0.05). (Figure 3b,c and Table 7).

2.6. Effect of Genetic Polymorphisms on IM Adverse Reactions

Among the 68 Chinese GIST patients, the most frequently observed adverse reactions related to IM include continuous edema, diarrhea, rash and myelosuppression.
For all six of the SNPs, only the mutation of NR1I2 rs3814055 was significantly associated with the incidence rate of continuous edema in the study population. The incidence rate of continuous edema in wild-types (CC) (34.15%) in NR1I2 rs3814055 was significantly higher than that in mutate allele T carriers (CT + TT) (3.70%) (p = 0.0030, Odds ratio (OR) = 13.48, 95% Confidence interval (CI): 1.65–109.98) (Figure 4, Table 8).
For the incidence rates of diarrhea, rash and myelosuppression, there are no significant differences in observed genotypes of all six SNPs (p > 0.05).

3. Discussion

In the past decade, imatinib mesylate, the first molecular-targeted drug with a known mechanism of efficacy, has radically changed the life expectancy of patients with GISTs. However, a large number of researchers have focused on the peculiar proto-oncogene of GISTs, and it can only partially explain the inter-individual variances of clinical response rate. While the germline DNA of patients remains important, as it dictates drug pharmacokinetics that may indirectly determine efficacy and toxicity. Based on these previous observations, we have involved the most relevant pharmacogenetic parameters, such as genetic polymorphisms of drug metabolizing enzymes, transporters and nuclear receptors, in order to investigate their influences on the steady-state IM dose-adjusted trough plasma concentrations and adverse reactions, for further explanation of the inter-individual variances on IM pharmacokinetics.
In our study, there was nearly a 16-fold variance in the steady-state IM trough plasma concentrations (from 272.22 to 4365.96 ng/mL) with IM doses ranging from 300 to 600 mg daily, which is much larger than the previous reports [10,23]. In addition, 47.06% of patients in the study, whose IM trough plasma concentrations had not reached the predefined minimal plasma concentration thresholds (1100 ng/mL), may be at the higher risk of disease progression and treatment failure.
Our study clearly showed the clinical impact of CYP3A4 rs2242480 polymorphisms on IM plasma levels, and the steady-state IM dose-adjusted trough plasma concentrations in mutant allele A carriers (GA + AA) were significantly lower than that in wild-types (GG) (p = 0.0171). However, the influence of rs2242480 polymorphisms on IM plasma levels in Chinese GIST patients has not been investigated previously. rs2242480, characterized by a G to A substitution in intron 10 of CYP3A4, is the most frequent SNP of CYP3A4 in the Chinese population. The frequency of mutant allele A in the study was 17.58%, similar to Healthy Han Chinese and Asian [15,24], but significantly different from Caucasian (7.34%) and African (85.71%). The mutant allele A was reported to be associated with a higher CYP3A4 metabolic activity [25,26,27], thus increasing the clearance of IM, which was mainly metabolized by CYP3A4, leading the lower IM plasma levels. Therefore, the mutant allele A of rs2242480 is a meaningful risk factor for predicting inadequate clinical efficacy of IM, and patients who carry mutant allele A of rs2242480 may be suggested to have a higher dose therapy.
In addition, our study obviously showed the clinical impact of ABCB1 rs1045642 polymorphisms on IM plasma levels, and the steady-state IM dose-adjusted trough plasma concentrations in mutate allele T carriers (CT + TT) were significantly higher than that in wild-types (CC) (p = 0.0066). Interestingly, the same association has been reported in Chronic Myelogenous Leukemia (CML) [28] patients under IM treatment, corroborating the importance of IM pharmacokinetics. The frequency of mutant allele T in the study was 36.76%, similar to Healthy Han Chinese and Asian [21], but significantly different from Caucasian (51.79%) and African (11.06%). The ABCB1 rs1045642 mutant allele T was reported to reduce the transcript levels of ABCB1 mRNA in vivo by as much as two- to four-fold when compared with allele C [29]. Thus, the mutant allele T was associated with a lower production of P-glycoprotein, leading to a lower drug clearance capability [30,31]. This might be responsible for the observation in the study that the mutant allele T carriers (CT + TT) have the higher IM plasma levels. We suggest that the mutant allele T of rs1045642 is a significant risk factor for predicting excessive treatment of IM, and patients who carry the mutant allele T of rs1045642 may be suggested to have a lower dose therapy.
However, the most interesting finding is the influence of NR1I2 rs3814055 polymorphisms on IM plasma levels and adverse reaction. The steady-state IM dose-adjusted trough plasma concentrations in mutate allele T carriers (CT + TT) (2.34 ± 0.25 ng/mL/mg) were significantly lower than that in wild-types (CC), and the incidence rate of edema in mutate allele T carriers (CT + TT) was significantly lower than that in wild-types (CC) (p = 0.0030, OR = 13.48, 95% CI: 1.65–109.98). However, the influence of rs3814055 polymorphisms on IM plasma levels and adverse reactions in Chinese GIST patients has not been reported previously, although a similar association has been reported in the studies on the pharmacokinetics of Tacrolimus in healthy subjects [32]. It is known that PXR is a crucial nuclear receptor to simultaneously regulate the expression of CYP3A4 [16,17,18,19] and ABCB1 [17,18,20]. rs3814055, located in 5′ Untranslated Regions of NRI12, is the highest frequency SNP of NR1I2 in the Chinese population. The frequency of mutant allele T in the study was 23.53%, similar to Healthy Han Chinese and Asian [22], but significantly different from Caucasian (36.58%) and African (30.64%). It is reported that the erythromycin breath test in mutant allele T carriers (CT + TT) was two times stronger than wild-types (CC) by the inducing of rifampicin, indicated that mutant allele T was associated with a higher transcript levels of CYP3A4 mRNA and metabolic activity [33,34], thus increasing the clearance of IM, which was mainly metabolized by CYP3A4, leading to the lower IM plasma levels.
Continuous edema is the most common side effect of IM treatment, with incidence of 37.85% in the study population. Because severe and continuous edema (fluid retention) may result in interruption of IM, careful monitoring of severe edema is especially important in elderly patients (65 and older), patients with preexisting coronary artery disease or renal impairment and patients on higher doses of IM [35,36,37]. However, it is considered to be a concentration-dependent and dose-limiting adverse reaction of IM [38,39,40]—the higher the IM plasma concentrations, the higher the risk of incidence of the adverse reaction. This might be responsible for the observation in this study that the wild-types (CC) in NR1I2 rs3814055 have higher IM plasma levels, leading to a high incidence rate of continuous edema. Therefore, allele C of rs3814055 is a productive risk factor for predicting excessive treatment and high risk of adverse reaction of IM, and patients who carry the allele C of rs3814055 may be suggested to have a lower dose or adjuvant diuretic therapy.

4. Materials and Methods

4.1. Subjects and Study Design

The study was performed according to the declaration of Helsinki and the International Conference on Harmonization-Good Clinical Practice standards. The study protocol was approved by the Ethics Committees of Zhongshan Hospital (ethical approval code: B2015-140R, 20151202) and the Shanghai Cancer Center (ethical approval code: 1604159-6, 20160425), affiliated with Fudan University. Details of the study were explained to all patients and informed consent was obtained. A total of 68 patients (39 males and 29 females) with newly diagnosed GIST were enrolled. All patients were orally administered 300–600 mg/day Imatinib Mesylate (Glivec, Novartis Pharma Stein AG, Basel, Switzerland) for at least one month. All of them were below 80 years of age, and had adequate hepatic and renal functions (Aspartate aminotransferase (AST)/Alanine aminotransferase (ALT) < 2× upper limit of normal (ULN), Total bilirubin in serum (TBIL) < 1.25 × ULN, Serum creatinine (Scr) < 1.5 × ULN). Patients who received medication known to affect IM plasma levels, such as verapamil, ketoconazole, and itraconazole, were excluded.
When IM plasma concentration reached a steady state (IM was regularly taken for 1 month at least), peripheral blood samples (0.5 h before IM dosing) of each GIST patient were drawn for IM trough plasma concentration determination and genotyping analysis, and the characteristics and biological values of the study population were recorded and summarized on the same day.

4.2. IM Trough Plasma Concentration Determination

Within 1 h of peripheral blood sample collection, plasma samples were prepared by centrifuging at 4000× g for 10 min at 4 °C, and a 100 µL aliquot of plasma was prepared by liquid–liquid extraction processing for IM trough plasma concentration determination, following a modified and validated HPLC method as reported earlier [41]. The HPLC system equipped with a Waters 1525 HPLC pump (Waters Corporation, Milford, MA, USA), a 2707 auto-sampler and a 2489 UV-detector linked to the Breeze™ Chromatography Data processing workstation (Waters Corporation, Milford, MA, USA) for recording and storing throughout analysis. Reversed phase HPLC analysis was carried out using a Symmetry C18 column (2.1 mm × 150 mm, 3.5 μm) (Waters Corporation, Milford, MA, USA) maintained at ambient temperature, with a mobile phase of Acetonitrile-20 mM ammonium acetate buffer (pH 6.8) (30:70, v/v), pumped at a flow rate of 0.2 mL/min and UV detection at a wavelength of 265 nm.

4.3. DNA Extraction and Genotyping

Total genomic DNA extraction was carried out using the method described previously [42]. The concentration and purity of extracted total genomic DNA samples were determined by a NANODROP LITE Spectrophotometer (Thermo Fisher Scientific Inc., Waltham, MA, USA). The genetic polymorphisms of CYP3A4 rs2242480, ABCB1 rs1045642, ABCG2 rs2231137 were determined by using polymerase chain reaction (PCR) restriction fragment length polymorphism (RFLP) method, while the genetic polymorphisms of NR1I2 rs3814055, rs6785049, rs2276706 were determined by using PCR and then sequencing. Analyses were performed using an Applied Biosysterms 2707 Thermal Cycler (Applied Biosystems, Foster, CA, USA). Sequencings were carried out on Thermo Fisher Scientific Inc. USA. Genotyping accuracy was confirmed by sequencing for two cases of each genotype. The PCR primers and reaction conditions were listed in the Supplementary Materials section (Tables S1–S3).

4.4. Adverse Reactions Monitoring

All patients were followed up in special clinics for GISTs to monitor IM-induced adverse reactions, which were evaluated according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 (NCI CTCAE v4.0).

4.5. Statistical Analysis

All statistical analyses were performed with SPSS® software, version 21.0 (IBM, Chicago, IL, USA). The results are expressed as mean ± SD, all of the tests were two-sided, and two-sided p < 0.05 was considered as statistically significant.
The Hardy–Weinberg equilibrium test was performed using an appropriate χ2-test. Linkage disequilibrium (LD) based association analysis was measured using the online software SHEsis (Bio-X Life Science Research Center, Shanghai, China) [43]. The statistical differences of IM dose-adjusted trough plasma concentration between genotypes of each SNP were analyzed by Mann-Whitney or Kruskal-Wallis tests. The statistical differences of IM adverse reactions between genotypes of each SNP were analyzed by chi-square or Fisher’s exact tests.

5. Conclusions

In summary, our findings indicate that the genetic polymorphisms of CYP3A4, ABCB1 and NR1I2 may make an important contribution to IM plasma levels and related adverse reactions. Because of the limited sample size of the study, further research should be carried out to verify the associated genetic polymorphisms on IM plasma levels and adverse reactions. The current results may serve as valuable fundamental knowledge for IM therapy in Chinese GIST patients.

Supplementary Materials

Supplementary materials can be found at www.mdpi.com/1422-0067/18/3/603/s1.

Acknowledgments

Our work was supported by the Shanghai Municipal Commission of Health and Family Planning, key development discipline (No. 2015ZB0201), and the Shanghai Municipal Science and Technology Commission (13411950802 and 14411970000).

Author Contributions

Jing Liu, Weimin Cai, and Yuhong Zhou conceived and designed the experiments; Jing Liu and Hanmei Chen performed the experiments; Jing Liu and Zhiyu Chen analyzed the data; Yingyong Hou, Weiqi Lu, Junyi He and Hanxing Tong helped to enroll and monitor the patients; and Jing Liu, Weinmin Cai and Yuhong Zhou wrote the paper.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCB1ATP-binding cassette (ABC) transporters subfamily B member 1
ABCG2ATP-binding cassette (ABC) transporters subfamily G member 2
ALTAlanine aminotransferase
ASTAspartate aminotransferase
CIConfidence interval
CYP3A4Cytochrome P450 3A4
GISTGastrointestinal stromal tumor
HPLCHigh Performance Liquid Chromatography
HWEHardy–Weinberg equilibrium
IMImatinib mesylate
OROdds ratio
PCRPolymerase chain reaction
PXRPregnane X Receptor
RFLPRestriction fragment length polymorphism
ScrSerum creatinine
SNPSingle nucleotide polymorphism
TBILTotal bilirubin in serum
ULNUpper limit of normal

References

  1. Inayat, F.; Saif, M.W. New Drug and Possible New Toxicity—Squamous Cell Carcinoma Following Imatinib in Patients with Gastrointestinal Stromal Tumors. Anticancer Res. 2016, 36, 6201–6204. [Google Scholar] [CrossRef] [PubMed]
  2. Nishida, T.; Blay, J.Y.; Hirota, S.; Kitagawa, Y.; Kang, Y.K. The standard diagnosis, treatment, and follow-up of gastrointestinal stromal tumors based on guidelines. Gastric Cancer 2016, 19, 3–14. [Google Scholar] [CrossRef] [PubMed]
  3. Gao, B.; Yeap, S.; Clements, A.; Balakrishnar, B.; Wong, M.; Gurney, H. Evidence for therapeutic drug monitoring of targeted anticancer therapies. J. Clin. Oncol. 2012, 30, 4017–4025. [Google Scholar] [CrossRef] [PubMed]
  4. Klumpen, H.J.; Samer, C.F.; Mathijssen, R.H.; Schellens, J.H.; Gurney, H. Moving towards dose individualization of tyrosine kinase inhibitors. Cancer Treat. Rev. 2011, 37, 251–260. [Google Scholar] [CrossRef] [PubMed]
  5. Slaviero, K.A.; Clarke, S.J.; Rivory, L.P. Inflammatory response: An unrecognised source of variability in the pharmacokinetics and pharmacodynamics of cancer chemotherapy. Lancet Oncol. 2003, 4, 224–232. [Google Scholar] [CrossRef]
  6. Demetri, G.D.; von Mehren, M.; Blanke, C.D.; van den Abbeele, A.D.; Eisenberg, B.; Roberts, P.J.; Heinrich, M.C.; Tuveson, D.A.; Singer, S.; Janicek, M.; et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N. Engl. J. Med. 2002, 347, 472–480. [Google Scholar] [CrossRef] [PubMed]
  7. Dematteo, R.P.; Ballman, K.V.; Antonescu, C.R.; Maki, R.G.; Pisters, P.W.; Demetri, G.D.; Blackstein, M.E.; Blanke, C.D.; von Mehren, M.; Brennan, M.F.; et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: A randomised, double-blind, placebo-controlled trial. Lancet 2009, 373, 1097–1104. [Google Scholar] [CrossRef]
  8. Delbaldo, C.; Chatelut, E.; Ré, M.; Deroussent, A.; Séronie-Vivien, S.; Jambu, A.; Berthaud, P.; Le Cesne, A.; Blay, J.Y.; Vassal, G. Pharmacokinetic-pharmacodynamic relationships of imatinib and its main metabolite in patients with advanced gastrointestinal stromal tumors. Clin. Cancer Res. 2006, 12, 6073–6078. [Google Scholar] [CrossRef] [PubMed]
  9. Widmer, N.; Decosterd, L.A.; Leyvraz, S.; Duchosal, M.A.; Rosselet, A.; Debiec-Rychter, M.; Csajka, C.; Biollaz, J.; Buclin, T. Relationship of imatinib-free plasma levels and target genotype with efficacy and tolerability. Br. J. Cancer 2008, 98, 1633–1640. [Google Scholar] [CrossRef] [PubMed]
  10. Demetri, G.D.; Wang, Y.; Wehrle, E.; Racine, A.; Nikolova, Z.; Blanke, C.D.; Joensuu, H.; von Mehren, M. Imatinib plasma levels are correlated with clinical benefit in patients with unresectable/metastatic gastrointestinal stromal tumors. J. Clin. Oncol. 2009, 27, 3141–3147. [Google Scholar] [CrossRef] [PubMed]
  11. Evans, W.E.; McLeod, H.L. Pharmacogenomics—Drug disposition, drug targets, and side effects. N. Engl. J. Med. 2003, 348, 538–549. [Google Scholar] [PubMed]
  12. O’Brien, K.M.; Orlow, I.; Antonescu, C.R.; Ballman, K.; McCall, L.; DeMatteo, R.; Engel, L.S. Gastrointestinal stromal tumors, somatic mutations and candidate genetic risk variants. PLoS ONE 2013, 8, e62119. [Google Scholar] [CrossRef] [PubMed]
  13. Koo, D.H.; Ryu, M.H.; Ryoo, B.Y.; Beck, M.Y.; Na, Y.S.; Shin, J.G.; Lee, S.S.; Kim, E.Y.; Kang, Y.K. Association of ABCG2 polymorphism with clinical efficacy of imatinib in patients with gastrointestinal stromal tumor. Cancer Chemother. Pharmacol. 2015, 75, 173–182. [Google Scholar] [CrossRef] [PubMed]
  14. Eechoute, K.; Sparreboom, A.; Burger, H.; Franke, R.M.; Schiavon, G.; Verweij, J.; Loos, W.J.; Wiemer, E.A.; Mathijssen, R.H. Drug transporters and imatinib treatment: Implications for clinical practice. Clin. Cancer Res. 2011, 17, 406–415. [Google Scholar] [CrossRef] [PubMed]
  15. Du, J.; Yu, L.; Wang, L.; Zhang, A.; Shu, A.; Xu, L.; Xu, M.; Shi, Y.; Li, X.; Feng, G.; et al. Differences in CYP3A41G genotype distribution and haplotypes of CYP3A4, CYP3A5 and CYP3A7 in 3 Chinese populations. Clin. Chim. Acta 2007, 383, 172–174. [Google Scholar] [CrossRef] [PubMed]
  16. Xie, W.; Evans, R.M. Orphan nuclear receptors: The exotics of xenobiotics. J. Biol. Chem. 2001, 276, 37739–37742. [Google Scholar] [CrossRef] [PubMed]
  17. Cerveny, L.; Svecova, L.; Anzenbacherova, E.; Vrzal, R.; Staud, F.; Dvorak, Z.; Ulrichova, J.; Anzenbacher, P. Valproic acid induces CYP3A4 and MDR1 gene expression by activation of constitutive androstane receptor and pregnane X receptor pathways. Drug Metab. Dispos. 2007, 35, 1032–1041. [Google Scholar] [CrossRef] [PubMed]
  18. Burk, O.; Arnold, K.A.; Nussler, A.K.; Schaeffeler, E.; Efimova, E.; Avery, B.A.; Avery, M.A.; Fromm, M.F.; Eichelbaum, M. Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. Mol. Pharmacol. 2005, 67, 1954–1965. [Google Scholar] [CrossRef] [PubMed]
  19. Burk, O.; Koch, I.; Raucy, J.; Hustert, E.; Eichelbaum, M.; Brockmöller, J.; Zanger, U.M.; Wojnowski, L. The induction of cytochrome P450 3A5 (CYP3A5) in the human liver and intestine is mediated by the xenobiotic sensors pregnane X receptor (PXR) and constitutively activated receptor (CAR). J. Biol. Chem. 2004, 279, 38379–38385. [Google Scholar] [CrossRef] [PubMed]
  20. Albermann, N.; Schmitz-Winnenthal, F.H.; Z'graggen, K.; Volk, C.; Hoffmann, M.M.; Haefeli, W.E.; Weiss, J. Expression of the drug transporters MDR1/ABCB1, MRP1/ABCC1, MRP2/ABCC2, BCRP/ABCG2, and PXR in peripheral blood mononuclear cells and their relationship with the expression in intestine and liver. Biochem. Pharmacol. 2005, 70, 949–958. [Google Scholar] [CrossRef] [PubMed]
  21. Li, D.; Zhang, G.L.; Lou, Y.Q.; Li, Q.; Wang, X.; Bu, X.Y. Genetic polymorphisms in MDR1 and CYP3A5 and MDR1 haplotype in mainland Chinese Han, Uygur and Kazakh ethnic groups. J. Clin. Pharm. Ther. 2007, 32, 89–95. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, X.D.; Li, J.L.; Su, Q.B.; Deng, X.Y.; Lu, Y.; Chen, J.; Zhang, J.X.; Zhao, L.Z.; Zuo, Z.; Chan, E.; et al. A pharmacogenetic study of pregnane X receptor (NR1I2) in Han Chinese. Curr. Drug Metab. 2007, 8, 778–786. [Google Scholar] [CrossRef] [PubMed]
  23. Yoo, C.; Ryu, M.H.; Kang, B.W.; Yoon, S.K.; Ryoo, B.Y.; Chang, H.M.; Lee, J.L.; Beck, M.Y.; Kim, T.W.; Kang, Y.K. Cross-sectional study of imatinib plasma trough levels in patients with advanced gastrointestinal stromal tumors: Impact of gastrointestinal resection on exposure to imatinib. J. Clin. Oncol. 2010, 28, 1554–1559. [Google Scholar] [CrossRef] [PubMed]
  24. Du, J.; Xing, Q.; Xu, L.; Xu, M.; Shu, A.; Shi, Y.; Yu, L.; Zhang, A.; Wang, L.; Wang, H.; et al. Systematic screening for polymorphisms in the CYP3A4 gene in the Chinese population. Pharmacogenomics 2006, 7, 831–841. [Google Scholar] [CrossRef] [PubMed]
  25. Lane, S.; AI-Zubiedi, S.; Hatch, E.; Matthews, I.; Jorgensen, A.L.; Deloukas, P.; Daly, A.K.; Park, B.K.; Aarons, L.; Ogungbenro, K.; et al. The population pharmacokinetics of R- and S-warfarin: Effect of genetic and clinical factors. Br. J. Clin. Pharmacol. 2012, 73, 66–76. [Google Scholar] [CrossRef] [PubMed]
  26. Du, J.; Zhang, A.; Wang, L.; Xuan, J.; Yu, L.; Chen, R.; Li, X.; Gu, N.; Lin, Z.; Feng, G.; et al. Relationship between response to risperidone, plasma concentrations of risperidone and CYP3A4 polymorphisms in schizophrenia patients. J. Psychopharmacol. 2010, 24, 1115–1120. [Google Scholar] [CrossRef] [PubMed]
  27. Ren, Z.Y.; Xu, X.Q.; Bao, Y.P.; He, J.; Shi, L.; Deng, J.H.; Gao, X.J.; Tang, H.L.; Wang, Y.M.; Lu, L. The impact of genetic variation on sensitivity to opioid analgesics in patients with postoperative pain: A systematic review and meta-analysis. Pain Physician 2015, 18, 131–152. [Google Scholar] [PubMed]
  28. Zheng, Q.; Wu, H.; Yu, Q.; Kim, D.H.; Lipton, J.H.; Angelini, S.; Soverini, S.; Vivona, D.; Takahashi, N.; Cao, J. ABCB1 polymorphisms predict imatinib response in chronic myeloid leukemia patients: A systematic review and meta-analysis. Pharmacogenom. J. 2015, 15, 127–134. [Google Scholar] [CrossRef] [PubMed]
  29. Hoffmeyer, S.; Burk, O.; von Richter, O.; Arnold, H.P.; Brockmöller, J.; Johne, A.; Cascorbi, I.; Gerloff, T.; Roots, I.; Eichelbaum, M.; et al. Functional polymorphisms of the human multidrug-resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 3473–3478. [Google Scholar] [CrossRef] [PubMed]
  30. Breedveld, P.; Pluim, D.; Cipriani, G.; Wielinga, P.; van Tellingen, O.; Schinkel, A.H.; Schellens, J.H. The effect of Bcrp1 (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): Implications for the use of breast cancer resistance protein and P-glycoprotein inhibitors to enable the brain penetration of imatinib in patients. Cancer Res. 2005, 65, 2577–2582. [Google Scholar] [PubMed]
  31. Oostendorp, R.L.; Buckle, T.; Beijnen, J.H.; van Tellingen, O.; Schellens, J.H. The effect of P-gp (Mdr1a/1b), BCRP (Bcrp1) and P-gp/BCRP inhibitors on the in vivo absorption, distribution, metabolism and excretion of imatinib. Investig. New Drugs 2009, 27, 31–40. [Google Scholar] [CrossRef] [PubMed]
  32. Choi, Y.; Jiang, F.; An, H.; Park, H.J.; Choi, J.H.; Lee, H. A pharmacogenomic study on the pharmacokinetics of tacrolimus in healthy subjects using the DMETTM Plus platform. Pharmacogenom. J. 2016. [Google Scholar] [CrossRef]
  33. Zhang, J.; Kuehl, P.; Green, E.D.; Touchman, J.W.; Watkins, P.B.; Daly, A.; Hall, S.D.; Maurel, P.; Relling, M.; Brimer, C.; et al. The human pregnane X receptor: Genomic structure and identification and functional characterization of natural allelic variants. Pharmacogenetics 2001, 11, 555–572. [Google Scholar] [CrossRef] [PubMed]
  34. Lamba, J.; Lamba, V.; Schuetz, E. Genetic variants of PXR (NR1I2) and CAR (NR1I3) and their implications in drug metabolism and pharmacogenetics. Curr. Drug Metab. 2005, 6, 369–383. [Google Scholar] [CrossRef] [PubMed]
  35. Deininger, M.W.; O'Brien, S.G.; Ford, J.M.; Druker, B.J. Practical management of patients with chronic myeloid leukemia receiving imatinib. J. Clin. Oncol. 2003, 21, 1637–1647. [Google Scholar] [CrossRef] [PubMed]
  36. Guilhot, F. Indications for imatinib mesylate therapy and clinical management. Oncologist 2004, 9, 271–281. [Google Scholar] [CrossRef] [PubMed]
  37. Trent, J.C.; Patel, S.S.; Zhang, J.; Araujo, D.M.; Plana, J.C.; Lenihan, D.J.; Fan, D.; Patel, S.R.; Benjamin, R.S.; Khakoo, A.Y. Rare incidence of congestive heart failure in gastrointestinal stromal tumor and other sarcoma patients receiving imatinib mesylate. Cancer 2010, 116, 184–192. [Google Scholar] [CrossRef] [PubMed]
  38. Verweij, J.; Casali, P.G.; Zalcberg, J.; LeCesne, A.; Reichardt, P.; Blay, J.Y.; Issels, R.; van Oosterom, A.; Hogendoorn, P.C.; Van Glabbeke, M.; Bertulli, R.; et al. Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: Randomised trial. Lancet 2004, 364, 1127–1134. [Google Scholar] [CrossRef]
  39. Blanke, C.D.; Rankin, C.; Demetri, G.D.; Ryan, C.W.; von Mehren, M.; Benjamin, R.S.; Raymond, A.K.; Bramwell, V.H.; Baker, L.H.; Maki, R.G.; et al. Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. J. Clin. Oncol. 2008, 26, 626–632. [Google Scholar] [CrossRef] [PubMed]
  40. Thanopoulou, E.; Judson, I. The safety profile of imatinib in CML and GIST: Long-term considerations. Arch. Toxicol. 2012, 86, 1–12. [Google Scholar] [CrossRef] [PubMed]
  41. Oostendorp, R.L.; Beijnen, J.H.; Schellens, J.H.; Tellingen, O.V. Determination of imatinib mesylate and its main metabolite (CGP74588) in human plasma and murine specimens by ion-pairing reversed-phase high-performance liquid chromatography. Biomed. Chromatogr. 2007, 21, 747–754. [Google Scholar] [CrossRef] [PubMed]
  42. Loparev, V.N.; Cartas, M.A.; Monken, C.E.; Velpandi, A.; Srinivasan, A. An efficient and simple method of DNA extraction from whole blood and cell lines to identify infectious agents. J. Virol. Methods 1991, 34, 105–112. [Google Scholar] [CrossRef]
  43. Shi, Y.Y.; He, L. SHEsis, a powerful software platform for analyses of linkage disequilibrium, haplotype construction, and genetic association at polymorphism loci. Cell Res. 2005, 15, 97–98. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The steady-state Imatinib mesylate (IM) dose-adjusted trough plasma concentrations related to CYP3A4 rs2242480 genotypes in 68 Chinese gastrointestinal stromal tumor (GIST) patients. (* p < 0.05).
Figure 1. The steady-state Imatinib mesylate (IM) dose-adjusted trough plasma concentrations related to CYP3A4 rs2242480 genotypes in 68 Chinese gastrointestinal stromal tumor (GIST) patients. (* p < 0.05).
Ijms 18 00603 g001
Figure 2. The steady-state Imatinib mesylate (IM) dose-adjusted trough plasma concentrations related to ABCB1 rs1045642 (a) and ABCG2 rs2231137 (b) genotypes in 68 Chinese gastrointestinal stromal tumor (GIST) patients (** p < 0.01).
Figure 2. The steady-state Imatinib mesylate (IM) dose-adjusted trough plasma concentrations related to ABCB1 rs1045642 (a) and ABCG2 rs2231137 (b) genotypes in 68 Chinese gastrointestinal stromal tumor (GIST) patients (** p < 0.01).
Ijms 18 00603 g002
Figure 3. The steady-state Imatinib mesylate (IM) dose-adjusted trough plasma concentrations related to NR1I2 rs3814055 (a); rs6785049 (b); and rs2276706 (c) genotypes in 68 Chinese gastrointestinal stromal tumor (GIST) patients (** p < 0.01).
Figure 3. The steady-state Imatinib mesylate (IM) dose-adjusted trough plasma concentrations related to NR1I2 rs3814055 (a); rs6785049 (b); and rs2276706 (c) genotypes in 68 Chinese gastrointestinal stromal tumor (GIST) patients (** p < 0.01).
Ijms 18 00603 g003
Figure 4. Comparison of the incidences of edema in 68 Chinese gastrointestinal stromal tumor (GIST) patients based on NR1I2 rs3814055 polymorphisms.
Figure 4. Comparison of the incidences of edema in 68 Chinese gastrointestinal stromal tumor (GIST) patients based on NR1I2 rs3814055 polymorphisms.
Ijms 18 00603 g004
Table 1. Characteristics and biological values of 68 Chinese GIST patients on the IM plasma concentration determination day.
Table 1. Characteristics and biological values of 68 Chinese GIST patients on the IM plasma concentration determination day.
Characters (Unit)Mean ± SD (Range)
Patient characteristics
Number of recipients68
Age (years)56.11 ± 0.50 (29–78)
Gender (male/female)39/29
Height (cm)166.00 ± 7.04 (150.00–182.00)
Weight (kg)61.51 ± 9.47 (45.00–85.00)
BMI22.18 ± 3.25 (15.09–28.65)
Biological values
White blood cell count (×109/L)5.37 ± 1.42 (2.01–12.40)
Red blood cell count (×1012/L)3.70 ± 0.51 (2.42–5.10)
Neutrophil count (×109/L)3.19 ± 1.30 (0.50–11.50)
Hemoglobin (g/L)117.83 ± 12.00 (46.00–152.00)
Platelet (×109/L)187.00 ± 66.57 (29.00–332.00)
Alanine aminotransferase (U/L)30.17 ± 11.28 (5.00–73.00)
Aspartate aminotransferase (U/L)31.67 ± 4.75 (12.80–77.00)
Albumin (g/L)40.60 ± 4.68 (24.00–51.90)
Serum creatinine (μmol/L)77.00 ± 14.13 (37.00–133.00)
Uric acid (μmol/L)227.80 ± 71.55 (96.00–512.00)
Blood urea nitrogen (mmol/L)5.65 ± 1.50 (1.70–202.00)
Abbreviations: GIST, Gastrointestinal stromal tumor; IM, Imatinib mesylate; BMI: Body Mass Index = Weight/(Height/100)2; SD, Standard deviation.
Table 2. The steady-state IM trough plasma concentrations in 68 Chinese GIST patients.
Table 2. The steady-state IM trough plasma concentrations in 68 Chinese GIST patients.
Characters (Unit)Mean ± SD (Range)
Number of total patients68
IM trough plasma concentrations (ng/mL)1134.30 ± 1141.69 (272.22–4365.96)
IM dose-adjusted trough plasma concentrations (ng/mL/mg)3.71 ± 2.53 (0.68–10.91)
Number of patients (<1100 ng/mL)32 (47.06%)
IM trough plasma concentrations (ng/mL)713.86 ± 239.76 (272.22–1095.20)
IM dose-adjusted trough plasma concentrations (ng/mL/mg)1.80 ± 0.61 (0.68–2.74)
Number of patients (>1100 ng/mL)36 (52.94%)
IM trough plasma concentrations (ng/mL)2339.81 ± 840.04 (1118.31–4365.96)
IM dose-adjusted trough plasma concentrations (ng/mL/mg)6.03 ± 2.04 (2.80–10.91)
Abbreviations: IM, Imatinib mesylate; GIST, Gastrointestinal stromal tumor; SD, Standard deviation.
Table 3. Frequencies of six SNPs in 68 Chinese GIST patients.
Table 3. Frequencies of six SNPs in 68 Chinese GIST patients.
SNP_IDGeneGenotypenIdentified Frequency (%)AlleleAllele Frequency (%)HWE
p-Value
rs2242480CYP3A4GG4870.59G82.350.12
GA1623.53A17.65
AA45.88
rs1045642ABCB1CC2942.64C63.240.52
CT2841.18T36.76
TT1116.18
rs2231137ABCG2GG2841.18G63.970.99
GA3145.59A36.03
AA913.23
rs3814055NR1I2CC4160.30C76.470.60
CT2232.35T23.53
TT57.35
rs6785049NR1I2GG2130.88G55.880.99
GA3450.00A44.12
AA1319.12
rs2276706NR1I2GG3957.35G75.000.84
GA2435.30A25.00
AA57.35
Abbreviations: SNPs, single nucleotide polymorphisms; GIST, Gastrointestinal stromal tumor; n, the numbers of patients; HWE, Hardy-Weinberg equilibrium.
Table 4. Comparisons of mutation frequencies for six SNPs among different ethnic groups.
Table 4. Comparisons of mutation frequencies for six SNPs among different ethnic groups.
SNPMutation Frequency (%)
Study PopulationHealthy Han ChineseAsianCaucasianAfrican
nFreq. anFreq. aRef. bnFreq. aRef. bnFreq. aRef. bnFreq. aRef. b
rs22424806817.6897622.10[15]12020.00NCBI2187.34 **NCBI22485.71 c,**NCBI
rs10456426836.7620840.40[21]12043.33NCBI22651.79 **NCBI22611.06 c,**NCBI
rs22311376836.039028.89NCBI12029.55NCBI1201.67 **NCBI1205.00 e,**NCBI
rs38140556823.5328621.80[22]12022.50NCBI22636.58 **NCBI22630.64 dNCBI
rs67850496844.1230037.00[22]17251.67NCBI22662.33 **NCBI2263.56 **NCBI
rs22767066825.0028621.80[22]12022.50NCBI12040.00 **NCBI11627.59 cNCBI
a Frequency; b Reference; c Sub-Saharan African; d African-American; e Yoruba in Ibadan; ** p < 0.01. Abbreviations: SNPs, single nucleotide polymorphisms; n, the numbers of patients.
Table 5. Association of CYP3A4 rs2242480 genotypes with the steady-state IM dose-adjusted concentrations in 68 Chinese GIST patients.
Table 5. Association of CYP3A4 rs2242480 genotypes with the steady-state IM dose-adjusted concentrations in 68 Chinese GIST patients.
SNP_IDGeneGenotypenIM Dose-Adjusted Concentration (ng/mL/mg)
Mean ± SDp
rs2242480CYP3A4GG484.12 ± 0.400.0171 *
GA + AA202.27 ± 0.32
Abbreviations: IM, Imatinib mesylate; GIST, Gastrointestinal stromal tumor; SNP, Single nucleotide polymorphism; n, the numbers of patients; SD, Standard deviation. * p < 0.05.
Table 6. Association of ABCB1/ABCG2 polymorphisms with the steady-state IM dose-adjusted concentrations in 68 Chinese GIST patients.
Table 6. Association of ABCB1/ABCG2 polymorphisms with the steady-state IM dose-adjusted concentrations in 68 Chinese GIST patients.
SNP_IDGeneGenotypenIM Dose-Adjusted Concentration (ng/mL/mg)
Mean ± SDp
rs1045642ABCB1CC292.56 ± 0.330.0055 **
CT + TT394.36 ± 0.45
rs2231137ABCG2GG283.51 ± 0.400.7158
GA + AA403.76 ± 0.50
Abbreviations: IM, Imatinib mesylate; GIST, Gastrointestinal stromal tumor; SNP, Single nucleotide polymorphism; n, the numbers of patients; SD, Standard deviation. ** p < 0.01.
Table 7. Association of NR1I2 polymorphisms with the steady-state IM dose-adjusted concentrations in 68 Chinese GIST patients.
Table 7. Association of NR1I2 polymorphisms with the steady-state IM dose-adjusted concentrations in 68 Chinese GIST patients.
SNP_IDGeneGenotypenIM Dose-Adjusted Concentration (ng/mL/mg)
Mean ± SDp
rs3814055NR1I2CC414.26 ± 0.430.0066 **
CT + TT272.34 ± 0.25
rs6785049NR1I2GG213.17 ± 0.330.2010
GA + AA474.09 ± 0.73
rs2276706NR1I2GG393.55 ± 0.320.9556
GA + AA293.62 ± 1.62
Abbreviations: IM, Imatinib mesylate; GIST, Gastrointestinal stromal tumor; SNP, Single nucleotide polymorphism; n, the numbers of patients; SD, Standard deviation. ** p < 0.01.
Table 8. Comparison of the incidences of edema related to NR1I2 rs3814055 polymorphisms in 68 Chinese GIST patients.
Table 8. Comparison of the incidences of edema related to NR1I2 rs3814055 polymorphisms in 68 Chinese GIST patients.
SNP_IDGeneGenotypenEdema
Yes (%)No (%)pOR95% CI
rs3814055NR1I2CC4114 (34.15)27 (65.85)0.0030 **13.481.65, 109.98
CT + TT271 (3.70)26 (96.30)
Abbreviations: GIST, Gastrointestinal stromal tumor; SNP, Single nucleotide polymorphism; n, the numbers of patients; OR, Odds ratio; CI, Confidence interval. ** p < 0.01.

Share and Cite

MDPI and ACS Style

Liu, J.; Chen, Z.; Chen, H.; Hou, Y.; Lu, W.; He, J.; Tong, H.; Zhou, Y.; Cai, W. Genetic Polymorphisms Contribute to the Individual Variations of Imatinib Mesylate Plasma Levels and Adverse Reactions in Chinese GIST Patients. Int. J. Mol. Sci. 2017, 18, 603. https://doi.org/10.3390/ijms18030603

AMA Style

Liu J, Chen Z, Chen H, Hou Y, Lu W, He J, Tong H, Zhou Y, Cai W. Genetic Polymorphisms Contribute to the Individual Variations of Imatinib Mesylate Plasma Levels and Adverse Reactions in Chinese GIST Patients. International Journal of Molecular Sciences. 2017; 18(3):603. https://doi.org/10.3390/ijms18030603

Chicago/Turabian Style

Liu, Jing, Zhiyu Chen, Hanmei Chen, Yingyong Hou, Weiqi Lu, Junyi He, Hanxing Tong, Yuhong Zhou, and Weimin Cai. 2017. "Genetic Polymorphisms Contribute to the Individual Variations of Imatinib Mesylate Plasma Levels and Adverse Reactions in Chinese GIST Patients" International Journal of Molecular Sciences 18, no. 3: 603. https://doi.org/10.3390/ijms18030603

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop