Imatinib mesylate is approved for the treatment of chronic myeloid leukemia (CML) and advanced gastrointestinal stromal tumors (GIST). Unfortunately, in the course of treatment, disease progression occurs in the majority of patients with GIST. Lowered plasma trough levels of imatinib over time potentially cause disease progression, a phenomenon known as “acquired pharmacokinetic drug resistance.” This outcome may be the result of an altered expression pattern or activity of drug transporters. To date, the role of both efflux transporters (ATP-binding cassette transporters, such as ABCB1 and ABCG2) and uptake transporters [solute carriers such as organic cation transporter 1 (OCT1) and organic anion transporting polypeptide 1A2 (OATP1A2)] in imatinib pharmacokinetics and pharmacodynamics has been studied. In vitro experiments show a significant role of ABCB1 and ABCG2 in cellular uptake and retention of imatinib, although pharmacokinetic and pharmacogenetic data are still scarce and contradictory. ABCB1 and ABCC1 expression was shown in GIST, whereas ABCB1, ABCG2, and OCT1 were found in mononuclear cells in CML patients. Several studies have reported a clinical relevance of tumor expression or activity of OCT1 in CML patients. Further (clinical) studies are required to quantify drug transporter expression over time in organs involved in imatinib metabolism, as well as in tumor tissue. In addition, more pharmacogenetic studies will be needed to validate associations. Clin Cancer Res; 17(3); 406–15. ©2010 AACR.

Translational Relevance

Since the start of this millennium, imatinib has become known as a very potent targeted agent for the treatment of chronic myeloid leukemia (CML) and gastrointestinal stromal tumors (GIST). However, in patients treated with imatinib, disease progression ultimately occurs because of resistance mechanisms. To overcome drug resistance, it is essential to know more about the mechanisms behind this phenomenon. In this review, the role of influx and efflux transporters is discussed in depth to give clinical insight into pharmacokinetic and pharmacodynamic pathways that are possibly influenced by drug transporter expression or activity. Finally, a better understanding of these mechanisms should lead to strategies to prolong the efficacy of imatinib therapy in both of these diseases.

Imatinib mesylate (Gleevec, Novartis International AG) is the first approved rationally designed inhibitor of specific protein tyrosine kinases. The drug inhibits ABL and the BCR-ABL fusion protein [expressed in Philadelphia chromosome–positive chronic myeloid leukemia (CML)], c-KIT [expressed in gastrointestinal stromal tumors (GIST)], and the platelet-derived growth factor receptor (PDGF-R; i.e., expressed in some sarcomas; refs. 1–4). Imatinib has become the standard treatment for patients with chronic myeloid leukemia (5, 6) and GIST (7–10).

Although response rates in imatinib-treated patients are high, ranging between 70 to 90% of patients with GIST as well as CML (9, 11, 12), nonresponse or disease progression after a certain period of time may occur. Genetic mutations or gene amplification of the drug targets are known mechanisms for this observed (acquired) drug resistance (12–15). Accumulating data, however, indicate a contributing role of pharmacokinetics in imatinib efficacy, as well as for the initial therapeutic response, and for the time to progression. Drug uptake and efflux transporters are likely to be involved in imatinib absorption, distribution, and excretion, thereby influencing pharmacokinetics. Imatinib is almost completely absorbed (>97%; ref. 16) and is, then, extensively metabolized in the liver with CGP74588 as its most active metabolite, predominantly formed by cytochrome P450 isoform 3A4 and 3A5 (CYP3A4, CYP3A5) as shown in Fig. 1 (17). This metabolite is equipotent to its parental compound in vitro. Other cytochrome P450 isoforms (CYP1A2, CYP2D6, CYP2C8, CYP2C9, and CYP2C19) also play a (minor) role in imatinib metabolism (17, 18). At clinically relevant concentrations, imatinib is bound to plasma proteins, mainly albumin and α1-acid glycoprotein (16, 19). Both imatinib and its active metabolite are excreted in feces and, to a lesser extent, in urine.

Figure 1.

Scheme for the involvement of drug transporters in imatinib pharmacokinetics. Carriers and enzymes involved in imatinib absorption, metabolism, and excretion, respectively, in their anatomical, tissue, and/or cellular localization.

Figure 1.

Scheme for the involvement of drug transporters in imatinib pharmacokinetics. Carriers and enzymes involved in imatinib absorption, metabolism, and excretion, respectively, in their anatomical, tissue, and/or cellular localization.

Close modal

Initial drug resistance could be correlated with drug exposure. For instance, imatinib trough levels, the lowest drug concentration, right before administration of a new dose, in nonresponding patients are significantly lower (see Table 1) than in responding patients (20–22). Moreover, imatinib pharmacokinetics may also contribute to acquired drug resistance. This finding was shown in a small-population pharmacokinetics study in patients with GIST (23). After long-term treatment (>1 year), imatinib clearance increased by ≈33% and systemic exposure decreased by ≈42%, compared with the start of treatment, possibly suggesting involvement of pharmacokinetics in the development of resistance. This change in pharmacokinetics is likely also related to the reduced side effects over time (14) and may also explain why dose escalation of imatinib at first progression results in clinical benefit in subsets of patients with advanced CML and GIST (24–26).

Table 1.

Imatinib trough levels (Cmin) in imatinib-treated chronic myeloid leukemia and gastrointestinal stromal cell tumor patients with or without clinical response

Tumor typeDose range (mg)No. of patientsNo. of respondersMean or median Cmin in responders (ng/mL)Median free Cmin in responders (ng/mL)No. of nonrespondersMean or median Cmin in nonresponders (ng/mL)Median free Cmin in nonresponders (ng/mL)P-valueReference
CML 400–800 351 297 1,009 (±544)a,b n.a.c 54 812 (± 409)a n.a.c 0.01 20 
CML 50–800 254 218 1,057 (±585)a,b n.a.c 36 835 (± 524)a n.a.c 0.033 30 
CML 50–800 254 166 1,107 (±594)a,d n.a.c 88 873 (± 528)a n.a.c 0.002 30 
CML 400–600 68 56 1,123 (±617)a,b n.a.c 12 694 (± 556)a n.a.c 0.03 21 
CML 400–600 68 34 1,452 (±649)a,d n.a.c 34 869 (± 427)a n.a.c 0.001 21 
CML 400 40 20 2,340 (±520)a,e n.a.c 20 690 (± 150)a n.a.c 0.002 29 
GIST 400–800 73 57 1,446 (414–3,336)f,g n.a.c 16 1,155 (545–4,182)f,h n.a.c 0.25 28 
GISTi Not reported 33 14 n.a.c 25.7 (13.7–27)j 19 n.a.c 10.1 (6.1–17.4)j,k 0.013 22 
Tumor typeDose range (mg)No. of patientsNo. of respondersMean or median Cmin in responders (ng/mL)Median free Cmin in responders (ng/mL)No. of nonrespondersMean or median Cmin in nonresponders (ng/mL)Median free Cmin in nonresponders (ng/mL)P-valueReference
CML 400–800 351 297 1,009 (±544)a,b n.a.c 54 812 (± 409)a n.a.c 0.01 20 
CML 50–800 254 218 1,057 (±585)a,b n.a.c 36 835 (± 524)a n.a.c 0.033 30 
CML 50–800 254 166 1,107 (±594)a,d n.a.c 88 873 (± 528)a n.a.c 0.002 30 
CML 400–600 68 56 1,123 (±617)a,b n.a.c 12 694 (± 556)a n.a.c 0.03 21 
CML 400–600 68 34 1,452 (±649)a,d n.a.c 34 869 (± 427)a n.a.c 0.001 21 
CML 400 40 20 2,340 (±520)a,e n.a.c 20 690 (± 150)a n.a.c 0.002 29 
GIST 400–800 73 57 1,446 (414–3,336)f,g n.a.c 16 1,155 (545–4,182)f,h n.a.c 0.25 28 
GISTi Not reported 33 14 n.a.c 25.7 (13.7–27)j 19 n.a.c 10.1 (6.1–17.4)j,k 0.013 22 

Abbreviations: CML, chronic myeloid leukemia; GIST, gastrointestinal stromal tumors.

aMean Cmin (±SD).

bResponse is defined as complete cytogenetic response.

cNot available.

dResponse is defined as major molecular response.

eResponse is defined as complete hematologic response at 3 months or major cytogenetic response at 6 months.

fMedian Cmin (and range).

gResponse is defined by the response evaluation criteria in solid tumors (RECIST) as stable disease, partial response or complete response.

hNonresponse is defined as disease progression by RECIST or not assessable.

iPatients with exon 9 mutated or wt KIT GIST.

jMedian free Cmin (and range) deduced from imatinib and AGP levels.

kNonresponse is defined by RECIST as disease progression.

Despite a broad interpatient variability in imatinib plasma exposure (27), more recent data suggest the clinical significance of keeping imatinib plasma concentrations within a therapeutic range. That is, in GIST and CML, a lower response rate and/or shorter time to progression may occur when imatinib plasma levels drop below approximately 1,000 ng/mL (21, 28–30). As reported in the study with GIST patients, imatinib steady state trough levels were higher than mentioned in literature (31). Therefore, an association with clinical benefit may be biased by the overestimation of trough levels. Nonetheless, steady state plasma concentrations above 1,000 ng/mL are often easily reached with a daily dose of 400 mg imatinib (31–33). Yet, in a subset of patients, this plasma concentration is not reached with this standard dose. Understanding the causes for this variability may be of clinical importance.

One of the patient factors that is likely to be relevant for the observed differences in imatinib pharmacokinetics is the possible interpatient variability in drug transporter expression and activity. In this review, we give a detailed overview of the potential clinical relevance of recently characterized drug transporters for imatinib therapy.

Efflux transporters

Imatinib is a substrate of ATP-binding cassette (ABC) transporters such as the ABC subfamily B member 1 (ABCB1; formerly known as P-glycoprotein or MDR1; refs. 32–38) and subfamily G member 2 [ABCG2; formerly known as breast cancer resistance protein (BCRP); refs. 37, 39, 40], which are involved in its excretion process. These drug transporters use the hydrolysis of ATP and subsequent phosphorylation of the transporter as an energy source, enabling active transport of substrates across various biomembranes (41, 42). Studies have reported imatinib as an inhibitor of ABC transporters (35, 43, 44), but there is growing consensus that ABC transporter inhibition by imatinib is dose dependent with inhibition only occurring at higher imatinib concentrations (Table 2; refs. 35, 37, 38). ABCB1 and ABCG2 are expressed in a variety of tissues, including liver (at the sinusoidal basolateral membrane, as well as the apical bile canalicular membrane of hepatocytes; refs. 1, 37, 45, 46), intestine (at the apical membrane; see Fig. 1; refs. 45–47), kidney, placenta (46, 48), and the blood-brain barrier (41, 49). The role of these efflux transporters in acquired drug resistance has been investigated more intensively than the role of uptake transporters because of the evident physiologic role of efflux transporters as a defense mechanism against penetration of xenobiotics.

Table 2.

Interaction between drug transporters and imatinib pharmacokinetics and pharmacodynamics

Drug transporterStudy designEffects on imatinib PKEffects on imatinib PDTransporter interactionEffect on imatinib IURReference
ABCB1 In vitro    IUR↓ 34–35,39 
 In vitro  Resistance ↑   35 
 In vivo Systemic clearance ↑    33,57 
 In vitro   Inducer  32,40 
 In vitro   Inhibitor  32,35,37 
 In vitro   Substrate  37–38 
ABCG2 In vitro    IUR = 44 
 In vitro    IUR↓ 39 
 In vitro  No resistance   44 
 In vivo (mice) Systemic clearance ↑    33,57 
 In vivo (mice) Plasma concentration =    60 
 In vivo (mice) Liver concentration =    60 
 In vitro   Inhibitor  37,43–44 
 In vitro   Inducer  40 
 In vivo (mice)   No inducer  60 
 In vitro   Substrate  37–39 
OCT1 In vitro    IUR = 51 
 In vitro    IUR↑ 50,62 
 In vitro  Resistance ↓   50 
 Clinical (CML)  Resistance ↓   63,65–66 
 In vitro   Substrate  50,62 
OATP1A2 In vitro   Substrate  51 
OATP1B3 In vitro   Substrate  51 
OCTN2 In vitro   Substrate  51 
Drug transporterStudy designEffects on imatinib PKEffects on imatinib PDTransporter interactionEffect on imatinib IURReference
ABCB1 In vitro    IUR↓ 34–35,39 
 In vitro  Resistance ↑   35 
 In vivo Systemic clearance ↑    33,57 
 In vitro   Inducer  32,40 
 In vitro   Inhibitor  32,35,37 
 In vitro   Substrate  37–38 
ABCG2 In vitro    IUR = 44 
 In vitro    IUR↓ 39 
 In vitro  No resistance   44 
 In vivo (mice) Systemic clearance ↑    33,57 
 In vivo (mice) Plasma concentration =    60 
 In vivo (mice) Liver concentration =    60 
 In vitro   Inhibitor  37,43–44 
 In vitro   Inducer  40 
 In vivo (mice)   No inducer  60 
 In vitro   Substrate  37–39 
OCT1 In vitro    IUR = 51 
 In vitro    IUR↑ 50,62 
 In vitro  Resistance ↓   50 
 Clinical (CML)  Resistance ↓   63,65–66 
 In vitro   Substrate  50,62 
OATP1A2 In vitro   Substrate  51 
OATP1B3 In vitro   Substrate  51 
OCTN2 In vitro   Substrate  51 

Abbreviations: PD, pharmacodynamics; PK, pharmacokinetics; IUR, intracellular uptake and retention.

Uptake transporters

Meanwhile, more than one fourth of the present-day anticancer drugs are oral formulations, stressing the possible relevance of intestinal absorption through uptake transporters expressed on the apical membrane of enterocytes. These solute carriers (SLC) use electrochemical gradients of ions to transport substrates across a membrane. Mainly the role of organic cation transporter 1 (OCT1 or the SLC22A1 gene product) and to a lesser extent organic anion transporting polypeptide 1A2 (OATP1A2, the SLCO1A2 gene product) in imatinib uptake has been described (50, 51). Furthermore, imatinib proved to be a good substrate for the solute carriers OATP1B3 (SLCO1B3 gene product) and OCTN2 (SLC22A5 gene product), both expressed on the basolateral membrane of hepatocytes (Fig. 1; ref. 51).

Absorption

This knowledge raises the question whether altered pharmacokinetics can be (in part) the result of (over) expression of drug transporters. At duodenal pH 5 to 6, imatinib is mainly charged (52), implying active intestinal transport; this renders intestinally located solute carriers such as OATP1A2 and OCTN2 as good candidates for intestinal imatinib uptake (53, 54). However, to date, little is known about the influence of these uptake transporters on imatinib pharmacokinetics. Another good candidate for systemic imatinib uptake is ABCC4, expressed on the basolateral membrane of hepatocytes (51). This efflux transporter could pump imatinib from the liver to the systemic circulation.

As mentioned above, imatinib is absorbed very efficiently, which is somewhat surprising, considering the high affinity of imatinib for ABC transporters, expressed on the canalicular membrane of hepatocytes and on enterocytes. A possible explanation for this apparent contradiction could be local substrate inhibition of efflux transporters by imatinib, bearing in mind its dose-dependent interaction. Furthermore, absolute bioavailability could also be influenced by the balance between efflux and influx transport over the intestinal barrier, favoring active imatinib uptake.

Tissue distribution

Liver distribution

Imatinib is actively cleared from the blood into the liver, where it is metabolized extensively. Possible candidates for this active transport are OATP1B3, OCTN2, and OCT1, predominantly located at the basolateral membrane of hepatocytes (Fig. 1; refs. 16, 51, 55, 56). However, in vivo or clinical data, supporting the role of solute carriers in imatinib clearance, are not available.

Brain distribution

Systemic treatment of brain tumors (primary as well as metastases) is limited because of low penetration of drugs into the brain tissue. Distribution to the brain is primarily prevented by the blood-brain barrier formed by the endothelial cells of brain capillaries. These endothelial cells also express ABCB1 and ABCG2 (41, 49), which actively prevent xenobiotics from diffusing into the brain. This finding was illustrated in Bcrp and Mdr1a/1b (rodent analogs of ABCG2 and ABCB1, respectively) knockout mouse models, showing that imatinib brain penetration significantly increased in knockout mice compared with wild-type mice, with a greater difference in Mdr1a/1b knockout (57). Another study with a rodent model showed that combined Bcrp and Mdr1a/1b knockout proved to significantly increase brain penetration compared with individual Bcrp or Mdr1a/1b knockouts (33). These data suggest that inhibition of efflux transporters at the blood-brain barrier may provide more tools in the treatment of brain metastases in imatinib-treated patients. A few obstacles remain, however. For instance, Gardner and colleagues showed that inhibition of ABCB1 and ABCG2 in mice resulted in a proportional increase in systemic exposure to imatinib in plasma and brain, leaving the brain-to-plasma concentration ratio unaltered (58). This finding suggests that reduced systemic elimination of imatinib leads to the observed increase in imatinib exposure to the brain as a result of higher imatinib concentrations at the blood-brain barrier, rather than a modification of the barrier itself. Furthermore, it is still unclear if efflux inhibitors will increase imatinib levels in tumor cells located in the central nervous system, because these inhibitors may be merely increasing brain uptake of substrates but not necessarily uptake into brain tumors (59).

Excretion

Biliary secretion

Up until now, in vivo experiments on the importance of drug transporters for imatinib excretion have shown only minor effects. Systemic clearance of imatinib in Mdr1a/1b and Bcrp1 knockout mice was 1.3-fold and 1.6-fold lower than wild-type mice (57). A combined Mdr1a/1b/Bcrp1 knockout showed a 1.8-fold reduction in imatinib plasma clearance compared with wild-type mice when imatinib was administered intravenously (33). Interestingly, no differences in pharmacokinetic parameters were found between Mdr1a/1b/Bcrp knockouts and wild-type mice after oral administration of imatinib. Whether ABCB1 and ABCG2 contribute to imatinib clearance in humans to a similar degree and, more importantly, whether there is a possible upregulation of these efflux transporters in excretory organs during imatinib treatment is unknown. These murine data, however, suggest a minor contribution of efflux transporters to imatinib clearance, compared with the hepatic metabolism. Indeed, an extensive first-pass metabolism of imatinib could contribute more substantially to systemic clearance than Mdr1a/1b and Bcrp1 efflux.

Furthermore, protein expression of Abcb1 and Bcrp1 in mice did not differ after long-term treatment with orally administered imatinib (60). After daily administration for 4 consecutive weeks, no upregulation of Abcb1 and Bcrp1 in mouse liver and intestinal tissues was found. Also, no significant change in plasma and liver concentrations of imatinib was seen. Theoretically, however, the length of treatment required to induce upregulation of these drug transporters in mice might be longer or the activity of both efflux transporters over the course of time may change without a quantitative change in expression.

Renal excretion

Although renal excretion accounts for less than 10% of imatinib excretion (31, 61), increased plasma exposure and decreased clearance in imatinib-treated cancer patients with impaired renal function, were seen (61). This finding may be due to increased levels of circulating uremic toxins. One such toxin inhibits OATP1B3 function in a rodent model (55), supporting the possibility that uremic toxins can directly reduce hepatic uptake of imatinib by OATP1B3. Further elucidation of this mechanism is needed.

Role of OCT1 in chronic myeloid leukemia blasts

There is substantial evidence that tumor OCT1 expression or activity determines therapeutic outcome in imatinib-treated CML patients. Thomas and colleagues were the first to show that inhibition of OCT1 in peripheral blood leukocytes from 6 CML patients, decreased intracellular imatinib uptake (62). This finding was confirmed in another study, showing that imatinib uptake in a CML cell line significantly correlated with OCT1 mRNA expression (63). Furthermore, White and colleagues showed that in vitro sensitivity for imatinib strongly correlated with the intracellular uptake and retention of imatinib in mononuclear cells of untreated CML patients (50). When prazosin, an OCT1 inhibitor, was added to these cells, the concentration needed to inhibit molecular drug targets was significantly increased. Furthermore, it was also shown that only the activity of OCT1 in mature CML blasts is associated with therapeutic outcome and not the OCT1 activity in immature CD34+ cells (64). This could imply that effective tumoral uptake of imatinib by OCT1 may be decisive for therapeutic response in CML patients. On the other hand, Hu and colleagues showed that intracellular uptake of imatinib in (nonleukemic) cells overexpressing OCT1 was only minimally higher than in control cells (51). In a gene expression analysis, they showed that SLC22A1 is significantly interrelated with ABCB1, ABCG2, and SLCO1A2. Alternatively, SLC22A1 gene expression could, therefore, be a marker for expression and subsequent activity of other transporters involved in imatinib transport.

SLC22A1 gene expression did prove to be a good predictor of clinical outcome in imatinib-treated CML patients (63, 65). Pretreatment OCT1 expression levels in 32 CML patients were 8 times higher in responders than in nonresponding patients (65). This result was confirmed in 70 CML patients, in which high baseline OCT1 RNA expression levels correlated with better cytogenetic response at 6 months and prolonged progression-free and overall survival (63). White and colleagues showed that chronic phase CML patients with low OCT1 activity showed clinical benefit from imatinib dose escalation, but they reported no correlation between clinical efficacy and OCT1 mRNA levels (66).

ABC transporter expression in gastrointestinal stromal tumor and chronic myeloid leukemia

ABCB1 and ABCC1 are expressed in approximately three quarters of GISTs, which is 2- to 3-fold more than the expression in leiomyosarcomas (67–69). On the other hand, Western blot analysis of 21 GIST specimens showed no expression of ABCG2 (68). So, in contrast to the possible influence of ABCG2 on the intestinal uptake of imatinib (33, 40, 57), there seems to be no role for ABCG2 on a tumoral level in GIST patients. Little can be said of the impact of ABCB1 and ABCC1 expression in stromal tumor cells on imatinib therapy in these GIST patients because only a very limited number of patients in these studies were treated with imatinib.

Although preclinical data (34–36) show that cellular (over) expression of ABCB1 leads to a reduced intracellular accumulation of imatinib, it is not clear whether long-term treatment with imatinib induces overexpression of this transporter in tumor cells. Mahon and colleagues examined various cell lines and found no upregulation of the expression of the ABCB1 gene by imatinib in time (70). In contrast, bone marrow mononuclear cells in CML patients resistant to imatinib showed an overexpression of ABCB1 and ABCG2 (although not statistically significant; ref. 65). In addition, a gene expression analysis in CML patients showed that expression of ABCC3 in CML blasts was unique to patients with disease recurrence (71). More studies with larger populations are needed to elucidate the possible tumoral upregulation of efflux transporters during imatinib therapy and its pharmacodynamic effects.

Pharmacokinetic impact of genetic variation in ABC transporters

To date, pharmacogenetic association studies were predominantly done for ABCB1 and ABCG2 (Table 3). Associations between 2 single nucleotide polymorphisms, known to reduce the activity of ABCB1 and ABCG2, respectively, and steady state imatinib pharmacokinetics in 82 patients with mainly GIST, have been investigated (72). Sixteen patients had a heterozygous variant (421 C > A) genotype for ABCG2 and 20 patients expressed a homozygous variant for ABCB1 3435 C > T. No significant differences in imatinib pharmacokinetics were seen compared with the homozygous wild-type patients. On the other hand, Takahashi and colleagues recently showed that imatinib trough levels were significantly higher in CML patients carrying an ABCG2 421A allele (in homozygous as well as heterozygous variant genotypes; ref. 73).

Table 3.

Studied single-nucleotide polymorphisms in patients involved in imatinib pharmacokinetics and pharmacodynamics

Transporter genePolymorphismEffects on imatinib PKEffects on imatinib PDNo. of patientsReference
ABCB1 3435 T No  82 72 
 3435 T Cmin = a Response = b 67 73 
 TTT haplotypec Cmina  90 77 
 TTT haplotypec Clearance ↑  22 74 
 3435 T  Overall survival ↓d 229 78 
 3435 T  Resistance ↑e 52 79 
 1236 T  Resistance ↑e 52 79 
 1236 T  Response ↑b 90 77 
 1236 T Cmin = a Response = b 67 73 
 2677 T/A Cmin = a Response = b 67 73 
 2677 G  Response ↓b 90 77 
 CGC haplotypec  Response ↓b 90 77 
ABCG2 421 A No  82 72 
 421 A Cmina Response = b 67 73 
 34 A  Response ↓f 229 78 
ABCC2 −24 T Cmin = a Response = b 67 73 
SLC22A1 181 T SS imatinib plasma level =  74 51 
 1393 A SS imatinib plasma level =  74 51 
 181 T  Response = b 32 80 
 480 G  Response ↓g 229 78 
 480 G Cmin = a Response = b 67 73 
 1022 T Cmin = a Response = b 67 73 
 1222 G Cmin = a Response ↑b 67 73 
 156 C Cmin = a Response = b 67 73 
SLCO1B3 334 G Cmin = a Response = b 67 73 
Transporter genePolymorphismEffects on imatinib PKEffects on imatinib PDNo. of patientsReference
ABCB1 3435 T No  82 72 
 3435 T Cmin = a Response = b 67 73 
 TTT haplotypec Cmina  90 77 
 TTT haplotypec Clearance ↑  22 74 
 3435 T  Overall survival ↓d 229 78 
 3435 T  Resistance ↑e 52 79 
 1236 T  Resistance ↑e 52 79 
 1236 T  Response ↑b 90 77 
 1236 T Cmin = a Response = b 67 73 
 2677 T/A Cmin = a Response = b 67 73 
 2677 G  Response ↓b 90 77 
 CGC haplotypec  Response ↓b 90 77 
ABCG2 421 A No  82 72 
 421 A Cmina Response = b 67 73 
 34 A  Response ↓f 229 78 
ABCC2 −24 T Cmin = a Response = b 67 73 
SLC22A1 181 T SS imatinib plasma level =  74 51 
 1393 A SS imatinib plasma level =  74 51 
 181 T  Response = b 32 80 
 480 G  Response ↓g 229 78 
 480 G Cmin = a Response = b 67 73 
 1022 T Cmin = a Response = b 67 73 
 1222 G Cmin = a Response ↑b 67 73 
 156 C Cmin = a Response = b 67 73 
SLCO1B3 334 G Cmin = a Response = b 67 73 

Abbreviations: PK, pharmacokinetics; PD, pharmacodynamics; SS, steady state.

aImatinib plasma trough level.

bMajor molecular response.

cDescription haplotype: 1236C > T, 2677G > T/A, 3435C > T.

dDefined as the period from initiation of imatinib therapy until the date of death from any cause or the date of last follow-up.

eNonresponse defined as absence of cytogenetic response.

fMajor or complete cytogenetic response.

gHigher rate of loss of cytogenetic or molecular response.

Gurney and colleagues found that patients with a TTT haplotype in ABCB1 1236C > T, 2677G > T/A, and 3435 C > T loci had significant higher estimated imatinib clearances (74). This finding is contradictory to reports showing lower mRNA and protein levels when a homozygous T allele for ABCB1 3435C > T was present (75), and to the findings of others who observed a decreased hepatic 99mTC-MIBI elimination rate, a phenotypic marker for ABCB1-mediated drug clearance, in patients with the TTT haplotype (76). In addition, in CML patients, a TTT-haplotype was associated with higher imatinib trough levels (77).

Clinical impact of genetic variation in ABC transporters

Up until now, pharmacogenetic association studies assessing clinical efficacy were exclusively done in CML patients. A poor response was observed in CML patients who were homozygous for the G allele in ABCG2 34G > A (78). As for ABCB1, a 1236T allele was associated with better response, whereas a 2677G allele or a CGC haplotype for the 1236, 2,677, and 3,435 loci were associated with worse response in CML patients (77). However, Kim and colleagues observed a reduced overall survival in CML patients carrying a TT genotype for 3435C > T locus, when analyzed univariately (78). This finding was confirmed by another group, who observed more resistance in CML patients carrying T alleles at positions 1236 and 3,435 (79). All in all, data on the role of pharmacogenetics in response and survival in CML patients receiving imatinib therapy are scarce and poorly validated or reproduced.

Pharmacokinetic and clinical impact of genetic variations in solute carriers

To date, studies assessing the possible role of genetic polymorphisms in solute carriers in imatinib therapy are limited to OCT1 and OATP1B3. Allelic variants of the SLC22A1 gene (encoding for the OCT1 protein) with known reduced function showed no effect on steady state imatinib plasma levels (181C > T and 1393G > A), in a group of GIST and CML patients compared with the reference genotype (51). Furthermore, Zach and colleagues found no significant differences in response in patients heterozygous for the T allele in the SLC22A1 181C > T polymorphism (80). Also, no correlation between SLCO1B3 334T > G polymorphism and imatinib exposure or clinical response was seen in CML patients (73). On the other hand, CML patients carrying a homozygous GG genotype for the SLC22A1 480C > G polymorphism showed a lower response rate (78). Furthermore, a higher response rate was seen in CML patients carrying a GG genotype for the 1222A > G locus (73). Unfortunately, imatinib trough levels did not significantly differ for these patients, carrying a 1222GG genotype, as compared with the reference allele.

Although in vitro studies show that imatinib exposure leads to an upregulation of ABCB1 and ABCG2 in human colon carcinoma cells (40), currently no data are available on the expression of these drug transporters in human intestinal cells under imatinib therapy. Future studies should assess the possible correlation between their expression pattern in excretory organs over time with imatinib pharmacokinetics and clinical outcome. In order to study the role of these transporters in the observed decline in imatinib clearance, a series of intestinal biopsies at different time points are needed.

Current studies also show that the majority of GIST expresses both ABCB1 and ABCC1, but its clinical importance is not yet elucidated. Although tumoral expression of OCT1 in CML patients has already been correlated with therapeutic outcome, more data are needed on the expression pattern of drug transporters in tumor cells in both CML and GIST patients and their possible pharmacodynamic impact. At least a quantification of these transporters over time should be made in GIST biopsies or mononuclear cells in CML patients in order to assess the possibility of an altered expression pattern of drug transporters as a mechanistic explanation for an altered sensitivity to the drug.

Finally, pharmacogenetic association data will need to be validated or reproduced. At this point, no clear guidelines exist on the design of pharmacogenetic studies. Preventing selection bias, adequate power analysis, clear endpoints, correction for genetic and nongenetic covariates, and other factors are often poorly implemented. Therefore, more pharmacogenetic studies assessing the association with imatinib pharmacokinetics and/or pharmacodynamics are desired, and study design will need to be uniform. In the present context of rapidly emerging promising compounds, the latter is of utmost importance if we want to personalize dosing and treatment sequences of rationally designed molecules to an individual patient's needs.

No potential conflicts of interest were disclosed.

1.
Buchdunger
E
,
Zimmermann
J
,
Mett
H
,
Meyer
T
,
Müller
M
,
Druker
BJ
, et al
Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative
.
Cancer Res
1996
;
56
:
100
4
.
2.
Lydon
NB
,
Druker
BJ
. 
Lessons learned from the development of imatinib
.
Leuk Res
2004
;
28
[Suppl 1]
:
S29
38
.
3.
Verweij
J
,
Judson
I
,
van Oosterom
A
. 
STI571: a magic bullet?
Eur J Cancer
2001
;
37
:
1816
9
.
4.
de Jong
FA
,
Verweij
J
. 
Role of imatinib mesylate (Gleevec/Glivec) in gastrointestinal stromal tumors
.
Expert Rev Anticancer Ther
2003
;
3
:
757
66
.
5.
O'Brien
SG
,
Guilhot
F
,
Larson
RA
,
Gathmann
I
,
Baccarani
M
,
Cervantes
F
, et al
Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia
.
N Engl J Med
2003
;
348
:
994
1004
.
6.
Druker
BJ
,
Guilhot
F
,
O'Brien
SG
,
Gathmann
I
,
Kantarjian
H
,
Gattermann
N
, et al
Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia
.
N Engl J Med
2006
;
355
:
2408
17
.
7.
van Oosterom
AT
,
Judson
I
,
Verweij
J
,
Stroobants
S
,
Donato di Paola
E
,
Dimitrijevic
S
, et al
Safety and efficacy of imatinib (STI571) in metastatic gastrointestinal stromal tumours: a phase I study
.
Lancet
2001
;
358
:
1421
3
.
8.
Demetri
GD
,
von Mehren
M
,
Blanke
CD
,
Van Den Abbeele
AD
,
Eisenberg
B
,
Roberts
PJ
, et al
Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors
.
N Engl J Med
2002
;
347
:
472
80
.
9.
Verweij
J
,
Casali
PG
,
Zalcberg
J
,
LeCesne
A
,
Reichardt
P
,
Blay
JY
, et al
Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial
.
Lancet
2004
;
364
:
1127
34
.
10.
Joensuu
H
,
Fletcher
C
,
Dimitrijevic
S
,
Silberman
S
,
Roberts
P
,
Demetri
G
. 
Management of malignant gastrointestinal stromal tumours
.
Lancet Oncol
2002
;
3
:
655
64
.
11.
Blanke
CD
,
Rankin
C
,
Demetri
GD
,
Ryan
CW
,
von Mehren
M
,
Benjamin
RS
, 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
32
.
12.
Druker
BJ
. 
Translation of the Philadelphia chromosome into therapy for CML
.
Blood
2008
;
112
:
4808
17
.
13.
Heinrich
MC
,
Corless
CL
,
Blanke
CD
,
Demetri
GD
,
Joensuu
H
,
Roberts
PJ
, et al
Molecular correlates of imatinib resistance in gastrointestinal stromal tumors
.
J Clin Oncol
2006
;
24
:
4764
74
.
14.
Sleijfer
S
,
Wiemer
E
,
Seynaeve
C
,
Verweij
J
. 
Improved insight into resistance mechanisms to imatinib in gastrointestinal stromal tumors: a basis for novel approaches and individualization of treatment
.
Oncologist
2007
;
12
:
719
26
.
15.
Liegl
B
,
Kepten
I
,
Le
C
,
Zhu
M
,
Demetri
GD
,
Heinrich
MC
, et al
Heterogeneity of kinase inhibitor resistance mechanisms in GIST
.
J Pathol
2008
;
216
:
64
74
.
16.
Peng
B
,
Dutreix
C
,
Mehring
G
,
Hayes
MJ
,
Ben-Am
M
,
Seiberling
M
, et al
Absolute bioavailability of imatinib (Glivec) orally versus intravenous infusion
.
J Clin Pharmacol
2004
;
44
:
158
62
.
17.
Gschwind
HP
,
Pfaar
U
,
Waldmeier
F
,
Zollinger
M
,
Sayer
C
,
Zbinden
P
, et al
Metabolism and disposition of imatinib mesylate in healthy volunteers
.
Drug Metab Dispos
2005
;
33
:
1503
12
.
18.
Nebot
N
,
Crettol
S
,
D'Esposito
F
,
Tattam
B
,
Hibbs
DE
,
Murray
M
. 
Participation of CYP2C8 and CYP3A4 in the N-demethylation of imatinib in human hepatic microsomes
.
Br J Pharmacol
2010
;
161
:
1059
69
.
19.
Gambacorti-Passerini
C
,
Zucchetti
M
,
Russo
D
,
Frapolli
R
,
Verga
M
,
Bungaro
S
, et al
Alpha1 acid glycoprotein binds to imatinib (STI571) and substantially alters its pharmacokinetics in chronic myeloid leukemia patients
.
Clin Cancer Res
2003
;
9
:
625
32
.
20.
Larson
RA
,
Druker
BJ
,
Guilhot
F
,
O'Brien
SG
,
Riviere
GJ
,
Krahnke
T
, et al
Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study
.
Blood
2008
;
111
:
4022
8
.
21.
Picard
S
,
Titier
K
,
Etienne
G
,
Teilhet
E
,
Ducint
D
,
Bernard
MA
, et al
Trough imatinib plasma levels are associated with both cytogenetic and molecular responses to standard-dose imatinib in chronic myeloid leukemia
.
Blood
2007
;
109
:
3496
9
.
22.
Widmer
N
,
Decosterd
LA
,
Csajka
C
,
Montemurro
M
,
Haouala
A
,
Leyvraz
S
, et al
Imatinib plasma levels: correlation with clinical benefit in GIST patients
.
Br J Cancer
2010
;
102
:
1198
9
.
23.
Judson
I
,
Ma
P
,
Peng
B
,
Verweij
J
,
Racine
A
,
di Paola
ED
, et al
Imatinib pharmacokinetics in patients with gastrointestinal stromal tumour: a retrospective population pharmacokinetic study over time. EORTC Soft Tissue and Bone Sarcoma Group
.
Cancer Chemother Pharmacol
2005
;
55
:
379
86
.
24.
Kantarjian
HM
,
Larson
RA
,
Guilhot
F
,
O'Brien
SG
,
Mone
M
,
Rudoltz
M
, et al
Efficacy of imatinib dose escalation in patients with chronic myeloid leukemia in chronic phase
.
Cancer
2009
;
115
:
551
60
.
25.
Zalcberg
JR
,
Verweij
J
,
Casali
PG
,
Le Cesne
A
,
Reichardt
P
,
Blay
JY
, et al
Outcome of patients with advanced gastro-intestinal stromal tumours crossing over to a daily imatinib dose of 800 mg after progression on 400 mg
.
Eur J Cancer
2005
;
41
:
1751
7
.
26.
Park
I
,
Ryu
MH
,
Sym
SJ
,
Lee
SS
,
Jang
G
,
Kim
TW
, et al
Dose escalation of imatinib after failure of standard dose in Korean patients with metastatic or unresectable gastrointestinal stromal tumor
.
Jpn J Clin Oncol
2009
;
39
:
105
10
.
27.
Nikolova
Z
,
Peng
B
,
Hubert
M
,
Sieberling
M
,
Keller
U
,
Ho
YY
, et al
Bioequivalence, safety, and tolerability of imatinib tablets compared with capsules
.
Cancer Chemother Pharmacol
2004
;
53
:
433
8
.
28.
Demetri
GD
,
Wang
Y
,
Wehrle
E
,
Racine
A
,
Nikolova
Z
,
Blanke
CD
, et al
Imatinib plasma levels are correlated with clinical benefit in patients with unresectable/metastatic gastrointestinal stromal tumors
.
J Clin Oncol
2009
;
27
:
3141
7
.
29.
Singh
N
,
Kumar
L
,
Meena
R
,
Velpandian
T
. 
Drug monitoring of imatinib levels in patients undergoing therapy for chronic myeloid leukaemia: comparing plasma levels of responders and non-responders
.
Eur J Clin Pharmacol
2009
;
65
:
545
9
.
30.
Takahashi
N
,
Wakita
H
,
Miura
M
,
Scott
SA
,
Nishii
K
,
Masuko
M
, et al
Correlation between imatinib pharmacokinetics and clinical response in Japanese patients with chronic-phase chronic myeloid leukemia
.
Clin Pharmacol Ther
2010
;
88
:
809
13
.
31.
Peng
B
,
Lloyd
P
,
Schran
H
. 
Clinical pharmacokinetics of imatinib
.
Clin Pharmacokinet
2005
;
44
:
879
94
.
32.
Hegedus
T
,
Orfi
L
,
Seprodi
A
,
Varadi
A
,
Sarkadi
B
,
Keri
G
. 
Interaction of tyrosine kinase inhibitors with the human multidrug transporter proteins, MDR1 and MRP1
.
Biochim Biophys Acta
2002
;
1587
:
318
25
.
33.
Oostendorp
RL
,
Buckle
T
,
Beijnen
JH
,
van Tellingen
O
,
Schellens
JH
. 
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
.
Invest New Drugs
2009
;
27
:
31
40
.
34.
Widmer
N
,
Colombo
S
,
Buclin
T
,
Decosterd
LA
. 
Functional consequence of MDR1 expression on imatinib intracellular concentrations
.
Blood
2003
;
102
:
1142
.
35.
Hamada
A
,
Miyano
H
,
Watanabe
H
,
Saito
H
. 
Interaction of imatinib mesilate with human P-glycoprotein
.
J Pharmacol Exp Ther
2003
;
307
:
824
8
.
36.
Zhang
Y
,
Bachmeier
C
,
Miller
DW
. 
In vitro and in vivo models for assessing drug efflux transporter activity
.
Adv Drug Deliv Rev
2003
;
55
:
31
51
.
37.
Dohse
M
,
Scharenberg
C
,
Shukla
S
,
Robey
RW
,
Volkmann
T
,
Deeken
JF
, et al
Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib and dasatinib
.
Drug Metab Dispos
2010
;
38
:
1371
80
.
38.
Shukla
S
,
Sauna
ZE
,
Ambudkar
SV
. 
Evidence for the interaction of imatinib at the transport-substrate site(s) of the multidrug-resistance-linked ABC drug transporters ABCB1 (P-glycoprotein) and ABCG2
.
Leukemia
2008
;
22
:
445
7
.
39.
Burger
H
,
van Tol
H
,
Boersma
AW
,
Brok
M
,
Wiemer
EA
,
Stoter
G
, et al
Imatinib mesylate (STI571) is a substrate for the breast cancer resistance protein (BCRP)/ABCG2 drug pump
.
Blood
2004
;
104
:
2940
2
.
40.
Burger
H
,
van Tol
H
,
Brok
M
,
Wiemer
EA
,
de Bruijn
EA
,
Guetens
G
, et al
Chronic imatinib mesylate exposure leads to reduced intracellular drug accumulation by induction of the ABCG2 (BCRP) and ABCB1 (MDR1) drug transport pumps
.
Cancer Biol Ther
2005
;
4
:
747
52
.
41.
Schinkel
AH
,
Smit
JJ
,
van Tellingen
O
,
Beijnen
JH
,
Wagenaar
E
,
van Deemter
L
, et al
Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs
.
Cell
1994
;
77
:
491
502
.
42.
Giacomini
KM
,
Sugiyama
Y
. 
Membrane transporters and drug response
. 
Goodman & Gilman's The pharmacological basis of therapeutics
. 11th ed.
McGraw-Hill
; 
2005
.
p. 41
70
.
43.
Ozvegy-Laczka
C
,
Hegedus
T
,
Varady
G
,
Ujhelly
O
,
Schuetz
JD
,
Viradi
A
, et al
High-affinity interaction of tyrosine kinase inhibitors with the ABCG2 multidrug transporter
.
Mol Pharmacol
2004
;
65
:
1485
95
.
44.
Houghton
PJ
,
Germain
GS
,
Harwood
FC
,
Schuetz
JD
,
Stewart
CF
,
Buchdunger
E
, et al
Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro
.
Cancer Res
2004
;
64
:
2333
7
.
45.
Thiebaut
F
,
Tsuruo
T
,
Hamada
H
,
Gottesman
MM
,
Pastan
I
,
Willingham
MC
. 
Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues
.
Proc Natl Acad Sci U S A
1987
;
84
:
7735
8
.
46.
Maliepaard
M
,
Scheffer
GL
,
Faneyte
IF
,
van Gastelen
MA
,
Pijnenborg
AC
,
Schinkel
AH
, et al
Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues
.
Cancer Res
2001
;
61
:
3458
64
.
47.
Behrens
I
,
Kamm
W
,
Dantzig
AH
,
Kissel
T
. 
Variation of peptide transporter (PepT1 and HPT1) expression in Caco-2 cells as a function of cell origin
.
J Pharm Sci
2004
;
93
:
1743
54
.
48.
Nagashige
M
,
Ushigome
F
,
Koyabu
N
,
Hirata
K
,
Kawabuchi
M
,
Hirakawa
T
, et al
Basal membrane localization of MRP1 in human placental trophoblast
.
Placenta
2003
;
24
:
951
8
.
49.
Cooray
HC
,
Blackmore
CG
,
Maskell
L
,
Barrand
MA
. 
Localisation of breast cancer resistance protein in microvessel endothelium of human brain
.
Neuroreport
2002
;
13
:
2059
63
.
50.
White
DL
,
Saunders
VA
,
Dang
P
,
Engler
J
,
Zannettino
AC
,
Cambareri
AC
, et al
OCT-1-mediated influx is a key determinant of the intracellular uptake of imatinib but not nilotinib (AMN107): reduced OCT-1 activity is the cause of low in vitro sensitivity to imatinib
.
Blood
2006
;
108
:
697
704
.
51.
Hu
S
,
Franke
RM
,
Filipski
KK
,
Hu
C
,
Orwick
SJ
,
de Bruijn
EA
, et al
Interaction of imatinib with human organic ion carriers
.
Clin Cancer Res
2008
;
14
:
3141
8
.
52.
Szakacs
Z
,
Beni
S
,
Varga
Z
,
Orfi
L
,
Keri
G
,
Noszal
B
. 
Acid-base profiling of imatinib (gleevec) and its fragments
.
J Med Chem
2005
;
48
:
249
55
.
53.
Lee
W
,
Glaeser
H
,
Smith
LH
,
Roberts
RL
,
Moeckel
GW
,
Gervasini
G
, et al
Polymorphisms in human organic anion-transporting polypeptide 1A2 (OATP1A2): implications for altered drug disposition and central nervous system drug entry
.
J Biol Chem
2005
;
280
:
9610
7
.
54.
Koepsell
H
,
Lips
K
,
Volk
C
. 
Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications
.
Pharm Res
2007
;
24
:
1227
51
.
55.
Franke
RM
,
Sparreboom
A
. 
Inhibition of imatinib transport by uremic toxins during renal failure
.
J Clin Oncol
2008
;
26
:
4226
7
, author reply 7–8.
56.
Zhang
L
,
Dresser
MJ
,
Gray
AT
,
Yost
SC
,
Terashita
S
,
Giacomini
KM
. 
Cloning and functional expression of a human liver organic cation transporter
.
Mol Pharmacol
1997
;
51
:
913
21
.
57.
Breedveld
P
,
Pluim
D
,
Cipriani
G
,
Wielinga
P
,
van Tellingen
O
,
Schinkel
AH
, et al
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
82
.
58.
Gardner
ER
,
Smith
NF
,
Figg
WD
,
Sparreboom
A
. 
Influence of the dual ABCB1 and ABCG2 inhibitor tariquidar on the disposition of oral imatinib in mice
.
J Exp Clin Cancer Res
2009
;
28
:
99
.
59.
Shen
J
,
Carcaboso
AM
,
Hubbard
KE
,
Tagen
M
,
Wynn
HG
,
Panetta
JC
, et al
Compartment-specific roles of ATP-binding cassette transporters define differential topotecan distribution in brain parenchyma and cerebrospinal fluid
.
Cancer Res
2009
;
69
:
5885
92
.
60.
Gardner
ER
,
Sparreboom
A
,
Verweij
J
,
Figg
WD
. 
Lack of ABC transporter autoinduction in mice following long-term exposure to imatinib
.
Cancer Biol Ther
2008
;
7
:
412
5
.
61.
Gibbons
J
,
Egorin
MJ
,
Ramanathan
RK
,
Fu
P
,
Mulkerin
DL
,
Shibata
S
, et al
Phase I and pharmacokinetic study of imatinib mesylate in patients with advanced malignancies and varying degrees of renal dysfunction: a study by the National Cancer Institute Organ Dysfunction Working Group
.
J Clin Oncol
2008
;
26
:
570
6
.
62.
Thomas
J
,
Wang
L
,
Clark
RE
,
Pirmohamed
M
. 
Active transport of imatinib into and out of cells: implications for drug resistance
.
Blood
2004
;
104
:
3739
45
.
63.
Wang
L
,
Giannoudis
A
,
Lane
S
,
Williamson
P
,
Pirmohamed
M
,
Clark
RE
. 
Expression of the uptake drug transporter hOCT1 is an important clinical determinant of the response to imatinib in chronic myeloid leukemia
.
Clin Pharmacol Ther
2008
;
83
:
258
64
.
64.
Engler
JR
,
Frede
A
,
Saunders
V
,
Zannettino
A
,
White
DL
,
Hughes
TP
. 
The poor response to imatinib observed in CML patients with low OCT-1 activity is not attributable to lower uptake of imatinib into their CD34+ cells
.
Blood
2010
;
116
:
2776
8
.
65.
Crossman
LC
,
Druker
BJ
,
Deininger
MW
,
Pirmohamed
M
,
Wang
L
,
Clark
RE
. 
hOCT 1 and resistance to imatinib
.
Blood
2005
;
106
:
1133
4
,
author reply 4.
66.
White
DL
,
Saunders
VA
,
Dang
P
,
Engler
J
,
Venables
A
,
Zrim
S
, et al
Most CML patients who have a suboptimal response to imatinib have low OCT-1 activity: higher doses of imatinib may overcome the negative impact of low OCT-1 activity
.
Blood
2007
;
110
:
4064
72
.
67.
Plaat
BE
,
Hollema
H
,
Molenaar
WM
,
Torn
Broers GH
,
Pijpe
J
,
Mastik
MF
, et al
Soft tissue leiomyosarcomas and malignant gastrointestinal stromal tumors: differences in clinical outcome and expression of multidrug resistance proteins
.
J Clin Oncol
2000
;
18
:
3211
20
.
68.
Theou
N
,
Gil
S
,
Devocelle
A
,
Julié
C
,
Lavergne-Slove
A
,
Beauchet
A
, et al
Multidrug resistance proteins in gastrointestinal stromal tumors: site-dependent expression and initial response to imatinib
.
Clin Cancer Res
2005
;
11
:
7593
8
.
69.
Perez-Gutierrez
S
,
Gonzalez-Campora
R
,
Amerigo-Navarro
J
,
Beato-Moreno
A
,
Sánchez-León
M
,
Pareja
Megía JM
, et al
Expression of P-glycoprotein and metallothionein in gastrointestinal stromal tumor and leiomyosarcomas. Clinical implications
.
Pathol Oncol Res
2007
;
13
:
203
8
.
70.
Mahon
FX
,
Deininger
MW
,
Schultheis
B
,
Chabrol
J
,
Reiffers
J
,
Goldman
JM
, et al
Selection and characterization of BCR-ABL positive cell lines with differential sensitivity to the tyrosine kinase inhibitor STI571: diverse mechanisms of resistance
.
Blood
2000
;
96
:
1070
9
.
71.
Radich
JP
,
Dai
H
,
Mao
M
,
Oehler
V
,
Schelter
J
,
Druker
B
, et al
Gene expression changes associated with progression and response in chronic myeloid leukemia
.
Proc Natl Acad Sci U S A
2006
;
103
:
2794
9
.
72.
Gardner
ER
,
Burger
H
,
van Schaik
RH
,
van Oosterom
AT
,
de Bruijn
EA
,
Guetens
G
, et al
Association of enzyme and transporter genotypes with the pharmacokinetics of imatinib
.
Clin Pharmacol Ther
2006
;
80
:
192
201
.
73.
Takahashi
N
,
Miura
M
,
Scott
SA
,
Kagaya
H
,
Kameoka
Y
,
Tagawa
H
, et al
Influence of CYP3A5 and drug transporter polymorphisms on imatinib trough concentration and clinical response among patients with chronic phase chronic myeloid leukemia
.
J Hum Genet
2010
;
55
:
731
7
.
74.
Gurney
H
,
Wong
M
,
Balleine
RL
,
Rivory
LP
,
McLachlan
AJ
,
Hoskins
JM
, et al
Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype
.
Clin Pharmacol Ther
2007
;
82
:
33
40
.
75.
Wang
D
,
Johnson
AD
,
Papp
AC
,
Kroetz
DL
,
Sadee
W
. 
Multidrug resistance polypeptide 1 (MDR1, ABCB1) variant 3435C>T affects mRNA stability
.
Pharmacogenet Genomics
2005
;
15
:
693
704
.
76.
Wong
M
,
Evans
S
,
Rivory
LP
,
Hoskins
JM
,
Mann
GJ
,
Farlow
D
, et al
Hepatic technetium Tc 99m-labeled sestamibi elimination rate and ABCB1 (MDR1) genotype as indicators of ABCB1 (P-glycoprotein) activity in patients with cancer
.
Clin Pharmacol Ther
2005
;
77
:
33
42
.
77.
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 imatinib in chronic myeloid leukemia
.
Blood
2008
;
112
:
2024
7
.
78.
Kim
DH
,
Sriharsha
L
,
Xu
W
,
Kamel-Reid
S
,
Liu
X
,
Siminovitch
K
, et al
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
2009
;
15
:
4750
8
.
79.
Ni
LN
,
Li
JY
,
Miao
KR
,
Qiao
C
,
Zhang
SJ
,
Qiu
HR
, et al
Multidrug resistance gene (MDR1) polymorphisms correlate with imatinib response in chronic myeloid leukemia
.
Med Oncol
2010
.
Epub 2010 Mar 4
.
80.
Zach
O
,
Krieger
O
,
Foedermayr
M
,
Zellhofer
B
,
Lutz
D
. 
OCT1 (SLC22A1) R61C polymorphism and response to imatinib treatment in chronic myeloid leukemia patients
.
Leuk Lymphoma
2008
;
49
:
2222
3
.