Purpose: The objective of this study was to identify genetic polymorphisms related to the pharmacokinetics and pharmacodynamics of sunitinib that are associated with a prolonged progression-free survival (PFS) and/or overall survival (OS) in patients with clear-cell metastatic renal cell cancer (mRCC) treated with sunitinib.

Experimental design: A retrospective multicenter pharmacogenetic association study was performed in 136 clear-cell mRCC patients treated with sunitinib. A total of 30 polymorphisms in 11 candidate genes, together with clinical characteristics were tested univariately for association with PFS as primary and OS as secondary outcome. Candidate variables with P < 0.1 were analyzed in a multivariate Cox regression model.

Results: Multivariate analysis showed that PFS was significantly improved when an A-allele was present in CYP3A5 6986A/G [hazard ratio (HR), 0.27; P = 0.032], a CAT copy was absent in the NR1I3 haplotype (5719C/T, 7738A/C, 7837T/G; HR, 1.76; P = 0.017) and a TCG copy was present in the ABCB1 haplotype (3435C/T, 1236C/T, 2677G/T; HR, 0.52; P = 0.033). Carriers with a favorable genetic profile (n = 95) had an improved PFS and OS as compared with noncarriers (median PFS and OS: 13.1 versus 7.5 months and 19.9 versus 12.3 months). Next to the genetic variants, the Memorial Sloan-Kettering Cancer Center prognostic criteria were associated with PFS and OS (HR, 1.99 and 2.27; P < 0.001).

Conclusions: This exploratory study shows that genetic polymorphisms in three genes involved in sunitinib pharmacokinetics are associated with PFS in mRCC patients treated with this drug. These findings advocate prospective validation and further elucidation of these genetic determinants in relation to sunitinib exposure and efficacy. Clin Cancer Res; 17(3); 620–9. ©2010 AACR.

Translational Relevance

Currently, sunitinib is the most widely prescribed drug for the treatment of metastatic renal cell cancer. Unfortunately, only a part of treated patients will benefit from sunitinib therapy, despite the implementation of clinical prognostic criteria in the choice of therapy. As multiple systemic treatment modalities arise, a further refinement is needed to identify renal cell cancer patients who predispose to benefit from sunitinib treatment and patients who do not. One of the possible options to study the differential response to sunitinib treatment is to identify genetic polymorphisms related to the pharmacokinetics and pharmacodynamics of this drug. In the future, genetic variants may be added to the current prognostic criteria, enabling physicians to predict benefit from sunitinib in individual patients.

For decades, the treatment options of metastatic renal cell cancer (mRCC) have been limited and systemic treatment primarily consisted of immunotherapy with cytokines. Increasing knowledge of the underlying biology of renal cell cancer (RCC), in particular the clear-cell subtype, has expanded the treatment options for patients with mRCC (1). RCC is characterized by an inactivated von Hippel–Lindau (VHL) tumor suppressor gene. Inactivated VHL leads to elevated protein levels of hypoxia-induced factor-α which upregulates VEGF and platelet-derived growth factor (PDGF) genes and proteins. The development of targeted therapy against signaling of these proteins has significantly improved the perspectives of patients with mRCC.

Currently, sunitinib is the most widely prescribed drug for the treatment of mRCC and has been registered as first-line and second-line therapy. Sunitinib is an oral tyrosine kinase inhibitor (TKI) which targets several receptors including VEGF receptors-1,-2,-3, PDGF receptors (PDGFR)-α and –β, c-KIT, and FLT-3. In a randomized controlled trial, sunitinib significantly prolonged the progression-free survival (PFS) and overall survival (OS) as compared with interferon-α (2, 3). Although sunitinib can achieve partial response rates of up to 40% (3–5), approximately 35% of mRCC patients do not benefit from sunitinib treatment (4, 5). Because sunitinib treatment may also result in unnecessary toxicities (6, 7), pretreatment markers to identify mRCC patients with a favorable outcome to sunitinib treatment are warranted.

Sunitinib efficacy may be dependent on its exposure which is regulated by efflux pumps and metabolizing enzymes. After oral administration, the systemic exposure of sunitinib is initially determined by its absorption in the gastrointestinal tract (Fig. 1). This process may be regulated by active drug transport over the intestinal wall, as sunitinib may be a substrate for polyspecific efflux transporters, expressed on enterocytes (8, 9). The efflux transporters ABCB1 (ATP binding cassette member B1, formerly known as P-glycoprotein or MDR1) and ABCG2 [ATP binding cassette member G2, formerly known as breast cancer-resistance protein (BCRP) or mitoxantrone resistant protein (MXR)] are expressed in the intestine and liver, and are involved in the oral absorption and biliary secretion of several anticancer drugs (10). Therefore, expression levels and functionality of these drug transporters may have important consequences for the efficacy of sunitinib.

Figure 1.

Schematic diagram of the pharmacokinetic and pharmacodynamic pathways that may be involved in the efficacy of sunitinib in mRCC. *, NR1I2 and NR1I3 regulate CYP3A4 expression.

Figure 1.

Schematic diagram of the pharmacokinetic and pharmacodynamic pathways that may be involved in the efficacy of sunitinib in mRCC. *, NR1I2 and NR1I3 regulate CYP3A4 expression.

Close modal

The cytochrome P450 (CYP) 3A (CYP3A) family is the predominant drug metabolizing enzyme and CYP3A4 is thought to be the key enzyme for the biotransformation of sunitinib (11). CYP3A4 is predominantly found in the liver and its expression is regulated by the ligand-activated nuclear receptors NR1I2 [pregnane X receptor (PXR)] and NR1I3 [constitutive androstane receptor (CAR)] (12). In addition, other enzymes of the cytochrome P450 family (CYP3A5, CYP1A1, and CYP1A2) may metabolize sunitinib, as these enzymes are known to be involved in the metabolism of other TKIs (13).

Besides pharmacokinetic factors, pharmacodynamic factors may determine the efficacy of sunitinib. In RCC, sunitinib is thought to exert its major therapeutic effect by inhibition of the VEGFR on tumor-associated endothelium, leading to reduced tumor angiogenesis (14). In addition, inhibition of the PDGFR might increase the antiangiogenic effects of sunitinib by targeting pericytes, which are able to protect endothelial cells from apoptosis (15). As the main targets for sunitinib are thought to be located in the microenvironment of tumor cells, the efficacy of sunitinib treatment may be related to the genetics of the surrounding microenvironment (16). Particularly, genetic variation in VEGFR-2 may affect sunitinib activity, because VEGFR-2 is expressed in the normal endothelium (17) and the tumor vasculature may develop from preexisting vessels of the host (18).

Single nucleotide polymorphisms (SNP) in genes encoding for efflux transporters, metabolizing enzymes, and drug targets may affect the efficacy of sunitinib in mRCC, as SNPs in specific genes have previously been associated with sunitinib-induced toxicities in patients with mRCC and gastrointestinal stromal tumors (19, 20). Therefore, SNPs may be useful markers for personalized treatment planning and may be candidate markers for selecting mRCC patients for sunitinib treatment. The objective of the current study was to identify SNPs involved in the pharmacokinetics and pharmacodynamics of sunitinib that are associated with a prolonged PFS and/or OS in mRCC patients.

Study population

In our previous study, 219 sunitinib-treated patients with various malignancies were included to investigate the association between SNPs and sunitinib-induced toxicities (19). In the present study, a subset of patients with histologically proven clear-cell RCC was selected for the analyses. A total of 136 consecutive mRCC patients who initiated sunitinib treatment between December 2005 and May 2008 were included. Sunitinib was administered orally at a dose of 50 mg daily, consisting of 4 weeks of treatment followed by a 2-week rest-period in cycles of 6 weeks. Dose reductions of sunitinib were allowed depending on the type and severity of adverse events according to the current guidelines (21). The study was approved by the medical ethics review board.

Study design

Demographic and clinical data of patients were reported on case record forms designed for data collection for this study. Patient characteristics considered relevant for PFS and OS analysis were age, gender, Eastern Cooperative Oncology Group (ECOG) performance status, prior systemic therapy, prior radiotherapy, the number of metastatic sites, and the risk factors according to Memorial Sloan-Kettering Cancer Center (MSKCC) prognostic criteria, which is based on 5 risk factors including low Karnofsky performance status (< 80%), high lactate dehydrogenase (LDH; >1.5 times the upper limit of normal), low hemoglobin level, high corrected serum calcium (> 10 mg/dL), and time from initial diagnosis to treatment < 1 year (22). Residual blood or serum samples taken for routine patient care were stored at −20°C at the local hospital laboratory. Of each patient one whole blood or serum sample was collected from the participating hospitals. All samples were anonymized by a third party, according to the instructions stated in the Codes for Proper Use and Proper Conduct in the Self-Regulatory Codes of Conduct (www.federa.org).

Genetic polymorphisms

Nineteen polymorphisms in 7 genes involved in the pharmacokinetics and 11 polymorphisms in 4 genes involved in the pharmacodynamics of sunitinib were selected (Supplementary Table S1). Selection criteria for the polymorphisms were a minor allelic frequency > 0.2 in Caucasians and an assumed clinical relevance based on previously reported associations or the assumption that nonsynonymous amino acid change leads to changed protein functionality.

The 11 candidate genes were selected on their potential relation with the pharmacokinetics and pharmacodynamics of sunitinib (Fig. 1). First, the candidate genotypes were selected by literature review. If there was no available data in the literature review, we referred to the SNPs from the dbSNP database (http://www.ncbi.nlm.nih.gov/sites/entrez). ABCB1, ABCG2, NR1I2, NR1I3, CYP3A5, CYP1A1, and CYP1A2 were selected for the pharmacokinetic pathways, whereas VEGFR-2, VEGFR-3, PDGFR-α, and FLT-3 were selected for the pharmacodynamic pathways. The most common functional SNPs in human ABCB1 are the synonymous 3435C > T and 1236C > T changes and the nonsynonymous 2677G > T change. As functional studies have shown that the haplotype of these three SNPs is a silent mutation and alters the function of the efflux transporter including its substrate specificity (23), the haplotype of ABCB1, instead of the three individual SNPs, was included in the analysis. Although VEGFR-1 is a target of sunitinib, and CYP3A4 is an important enzyme for metabolism of sunitinib (11), no polymorphisms of VEGFR-1 and CYP3A4 were analyzed, as no functional polymorphisms met the criteria for SNP selection.

Methods for genotyping assay validation and haplotype estimation have been described previously (19). Briefly, germline DNA was isolated from 1 mL of serum or EDTA-blood with the Magnapure LC (Roche Diagnostics). Polymorphic sites in genomic DNA were analyzed with TaqMan assays (Applied Biosystems).

Statistical design and data analysis

For PFS and OS, data collection was closed on August 31, 2009. The primary outcome measures of this study, PFS, was defined as the time between the first day of sunitinib and the date of progressive disease (PD) according to Response Evaluation Criteria in Solid Tumors (24), clear clinical evidence of PD or death due to PD within 12 weeks after the last response evaluation. If a patient had not progressed, PFS was censored at the time of the last follow-up. If the PD date was unknown or a patient died due to PD later than 12 weeks after the last response evaluation, PFS was censored at the last adequate tumor assessment. OS was the secondary outcome and was defined as the time between the first day of sunitinib treatment and the date of death or the date at which patients were last known to be alive.

All patient characteristics were tested univariately against the primary outcome using Kaplan–Meier and Cox-regression analysis, depending on the tested variables. The polymorphisms and haplotypes were tested univariately against PFS and OS using the Kaplan–Meier method. For this initial analysis, the general model was used. Given the explorative nature of this study, variables with a P ≤ 0.1 were selected as candidate variables for multivariate Cox-regression analyses. Data were fitted to the most appropriate model (multiplicative, dominant or recessive) and tested in the multivariate Cox regression survival analysis with PFS and OS as depending variables. Additional patient characteristics were introduced in the multivariate analyses based on univariately tested results if P < 0.1. Hazard ratios (HR) were generated considering patients with the most common clinical factor or genotype as the reference group. Missing data were kept as missing except for factors in the MSKCC score and the ECOG performance status. Patients with missing performance status (n = 2), LDH (n = 2), hemoglobin (n = 1), and baseline calcium values (n = 2) were assumed to be part of the worse prognosis scores. Accordingly, MSKCC scores were increased with one risk factor in 5 patients and with two risk factors in one patient. As a result 3 patients were categorized into the intermediate risk group, whereas 3 other patients were categorized into the poor risk group. Patients with missing ECOG performance statuses (n = 2) were scored as ECOG = 1. To test these assumptions, the multivariate analyses were performed with and without the replacement of the patients with missing factors in the MSKCC score. Similar results were generated, indicating that the replacement was legitimate. All statistical analyses were performed using SPSS 16.0 software. A 20% improvement (HR, 0 = 0.44) in PFS at ∼1 year in patients with sunitinib was judged to be clinically meaningful by the investigators designing the study. Forty-six events with disease progression were estimated to be needed to detect such an improvement using a two-sided, unstratified log-rank test with an overall significance level of 0.05 and power of 0.80. All results from the multivariate analyses with P < 0.05 were considered significant. Because this is an explorative study, no correction for multiple testing was made.

Study population

The main patient characteristics are presented in Table 1. Thirty-one (22.8%) patients had one metastatic site, 47 (34.6%) patients had two metastatic sites, and 58 (42.6%) patients had at least 3 metastatic sites. According to the MSKCC prognostic criteria most patients (59.6%) were categorized into the intermediate risk group, whereas 24.3% and 16.2% of the patients were categorized into the favorable and poor risk group, respectively.

Table 1.

Patient characteristics

CharacteristicNo.%
Median age, y 60  
 Range 25–84  
Sex   
 Male 83 61.0 
 Female 53 39.0 
Ethnicity   
 Caucasian 130 95.6 
 Black 2.2 
 Asian 0.7 
 Latin–American 0.7 
 Arab 0.7 
ECOG performance status   
 0 66 48.5 
 1 54 39.7 
 2 10 7.4 
 3 2.9 
 Missing 1.5 
Previous medical treatments   
 Yes 56 41.2 
 No 80 58.8 
Previous treatment regimen   
 Cytokine-based therapy 45 33.1 
 Antiangiogenic therapy 2.9 
 Both 5.1 
Previous nephrectomy   
 Yes 105 77.2 
 No 31 22.8 
Previous radiation therapy   
 Yes 34 25.0 
 No 102 75.0 
No. of metastatic sites   
 1 31 22.8 
 2 47 34.6 
 ≥3 58 42.6 
Sites of metastases   
 Lung 100 73.5 
 Liver 33 24.3 
 Bone 46 33.8 
 Lymph nodes 61 44.9 
 Brain 3.7 
MSKCC risk factorsa   
 0 (favorable) 33 24.3 
 1–2 (intermediate) 81 59.6 
 ≥3 (poor) 22 16.2 
CharacteristicNo.%
Median age, y 60  
 Range 25–84  
Sex   
 Male 83 61.0 
 Female 53 39.0 
Ethnicity   
 Caucasian 130 95.6 
 Black 2.2 
 Asian 0.7 
 Latin–American 0.7 
 Arab 0.7 
ECOG performance status   
 0 66 48.5 
 1 54 39.7 
 2 10 7.4 
 3 2.9 
 Missing 1.5 
Previous medical treatments   
 Yes 56 41.2 
 No 80 58.8 
Previous treatment regimen   
 Cytokine-based therapy 45 33.1 
 Antiangiogenic therapy 2.9 
 Both 5.1 
Previous nephrectomy   
 Yes 105 77.2 
 No 31 22.8 
Previous radiation therapy   
 Yes 34 25.0 
 No 102 75.0 
No. of metastatic sites   
 1 31 22.8 
 2 47 34.6 
 ≥3 58 42.6 
Sites of metastases   
 Lung 100 73.5 
 Liver 33 24.3 
 Bone 46 33.8 
 Lymph nodes 61 44.9 
 Brain 3.7 
MSKCC risk factorsa   
 0 (favorable) 33 24.3 
 1–2 (intermediate) 81 59.6 
 ≥3 (poor) 22 16.2 

aRisk groups according to Memorial Sloan-Kettering Cancer Center (MSKCC) prognostic criteria [based on the 5 risk factors: low Karnofsky performance status (<80%), high LDH (>1.5 times the upper limit of normal), low serum hemoglobin, high corrected serum calcium (>10 mg/dL), and time from initial diagnosis to treatment <1 year] (22).

At the time of the analysis, 47 (34.6%) patients were alive and 92 (67.6%) patients had disease progression. Overall, the median PFS time was 10.0 months (range, 7.6–12.4 months) and the median OS time was 16.3 months (range, 13.5–19.2 months). Of the clinical characteristics, the MSKCC risk factors had the largest contribution to PFS and OS (P = 0.001 and P < 0.001, respectively; Tables 2 and 3). In addition, the number of metastatic sites, age, and the ECOG performance status were prognostic for PFS (P = 0.019, 0.047, and 0.049, respectively), whereas only the ECOG performance status and the number of metastatic sites were also prognostic for OS (P = 0.004 and 0.058, respectively).

Table 2.

Univariate and multivariate analyses of progression-free survival in mRCC patients treated with sunitinib

FactorsNo.Univariate analysesaMultivariate analyses
Median PFS (mo)95% CIPHRb95% CIP
Clinical factors        
MSKCC risk factorsc    0.001 1.988 1.394–2.837 <0.001d 
 0 33 24.3 11.3–37.2     
 1–2 81 8.7 6.3–11.2     
 ≥3 22 7.0 0.0–14.4     
No. of metastatic sites    0.019 1.400 1.042–1.880 0.025d 
 1 31 19.4 7.8–31.0     
 2 47 11.0 5.1–16.8     
 ≥3 58 8.1 7.6–8.6     
Age (HR = 1.024 per year increase)   1.000–1.048 0.047 1.031 per year increase 1.003–1.060 0.029 
Genetic factors pharmacokinetic pathway        
CYP3A5 6986A/G    0.017    
 GG versus 117 9.3 6.9–11.8   0.032 
 AG + AA 11 Not reached –  0.266 0.079–0.892  
NR1I3 haplotypee    0.021   0.017 
 Other–other versus 75 13.3 7.9–18.8    
 CAT-other + CAT-CAT 60 8.0 7.5–8.6  1.758 1.108–2.790  
NR1I2 8055C/T    0.025   0.656 
 CC + CT versus 119 10.8 8.0–13.6    
 TT 17 6.7 3.6–9.9  1.638 0.187–14.380  
NR1I2 -25385C/T    0.032   0.795 
 CC + CT versus 118 10.8 8.0–13.6    
 TT 18 6.7 2.8–10.7  0.755 0.091–6.273  
ABCB1 haplotypef    0.072   0.033 
 Other–other versus 100 8.4 7.0–9.7    
 TCG–other 29 15.2 6.1–24.3  0.522 0.287–0.950  
ABCG2 34G/A    0.077   0.497 
 GG versus 124 9.0 6.9–11.2    
 AG 12 19.4 0.0–40.8  0.713 0.269–1.891  
FactorsNo.Univariate analysesaMultivariate analyses
Median PFS (mo)95% CIPHRb95% CIP
Clinical factors        
MSKCC risk factorsc    0.001 1.988 1.394–2.837 <0.001d 
 0 33 24.3 11.3–37.2     
 1–2 81 8.7 6.3–11.2     
 ≥3 22 7.0 0.0–14.4     
No. of metastatic sites    0.019 1.400 1.042–1.880 0.025d 
 1 31 19.4 7.8–31.0     
 2 47 11.0 5.1–16.8     
 ≥3 58 8.1 7.6–8.6     
Age (HR = 1.024 per year increase)   1.000–1.048 0.047 1.031 per year increase 1.003–1.060 0.029 
Genetic factors pharmacokinetic pathway        
CYP3A5 6986A/G    0.017    
 GG versus 117 9.3 6.9–11.8   0.032 
 AG + AA 11 Not reached –  0.266 0.079–0.892  
NR1I3 haplotypee    0.021   0.017 
 Other–other versus 75 13.3 7.9–18.8    
 CAT-other + CAT-CAT 60 8.0 7.5–8.6  1.758 1.108–2.790  
NR1I2 8055C/T    0.025   0.656 
 CC + CT versus 119 10.8 8.0–13.6    
 TT 17 6.7 3.6–9.9  1.638 0.187–14.380  
NR1I2 -25385C/T    0.032   0.795 
 CC + CT versus 118 10.8 8.0–13.6    
 TT 18 6.7 2.8–10.7  0.755 0.091–6.273  
ABCB1 haplotypef    0.072   0.033 
 Other–other versus 100 8.4 7.0–9.7    
 TCG–other 29 15.2 6.1–24.3  0.522 0.287–0.950  
ABCG2 34G/A    0.077   0.497 
 GG versus 124 9.0 6.9–11.2    
 AG 12 19.4 0.0–40.8  0.713 0.269–1.891  

aOnly factors with P < 0.1 level are presented; factors with P < 0.1 in the univariate analyses were selected for multivariate analyses.

bHR < 1.0 indicates that the factor associates with improved PFS, HR > 1.0 associates with worse PFS.

cRisk groups according to Memorial Sloan-Kettering Cancer Center (MSKCC) prognostic criteria [based on the 5 risk factors: low Karnofsky performance status (<80%), high LDH (>1.5 times the upper limit of normal), low serum hemoglobin, high corrected serum calcium (>10 mg/dL), and time from initial diagnosis to treatment <1 year] (22).

dMultiplicative model, HR per increase in MSKCC class or number of metastatic sites class.

Description haplotypes: e = NR1I3 5719C/T, 7738A/C, and 7837T/G; f = ABCB1 3435C/T, 1236C/T, and 2677G/T.

Table 3.

Univariate and multivariate analyses of overall survival in mRCC patients treated with sunitinib

Univariate analysesaMultivariate analyses
FactorsNo.Median OS (mo)95% CIPHRb95% CIP
Clinical factors        
MSKCC risk factorsd    <0.001   <0.001d 
 0 33 Not reached  2.273 1.595–3.238  
 1—2 81 14.8 11.8–17.7     
 ≥3 22 10.9 7.2–14.7     
No. of metastatic sites    0.058   0.097d 
 1 31 28.8 15.3–42.2  1.273 0.957–1.693  
 2 47 15.6 13.5–17.7     
 ≥3 58 13.2 9.5–16.9     
Genetic factors pharmacokinetic pathway        
NR1I2 -25385C/T    0.017   0.178 
 CC + CT versus 118 17.1 12.9–21.2    
 TT 18 10.2 7.4–13.1  1.490 0.834–2.660  
ABCB1 haplotypee    0.097   0.078 
 Other–other versus 100 15.4 12.4–18.3    
 TCG–other 29 23.9 9.0–38.7  0.593 0.332–1.061  
ABCG2 34G/A    0.072   0.069 
 GG versus 124 15.4 12.5–18.3    
 AG 12 39.9 16.7–63.1  0.416 0.162–1.070  
Genetic factors pharmacodynamic pathway        
PDGFR-α haplotypef    0.002   0.108 
 GCGT–GCGT vs. 78 24.2 17.2–31.2    
 GCG–other + other–other 56 14.8 11.6–17.9  1.458 0.920–2.310  
VEGFR-2 1718T/A    0.022   0.016 
 AA + AT versus 125 16.3 12.4–20.2    
 TT 9.4 7.2–11.7  2.907 1.224–6.903  
Univariate analysesaMultivariate analyses
FactorsNo.Median OS (mo)95% CIPHRb95% CIP
Clinical factors        
MSKCC risk factorsd    <0.001   <0.001d 
 0 33 Not reached  2.273 1.595–3.238  
 1—2 81 14.8 11.8–17.7     
 ≥3 22 10.9 7.2–14.7     
No. of metastatic sites    0.058   0.097d 
 1 31 28.8 15.3–42.2  1.273 0.957–1.693  
 2 47 15.6 13.5–17.7     
 ≥3 58 13.2 9.5–16.9     
Genetic factors pharmacokinetic pathway        
NR1I2 -25385C/T    0.017   0.178 
 CC + CT versus 118 17.1 12.9–21.2    
 TT 18 10.2 7.4–13.1  1.490 0.834–2.660  
ABCB1 haplotypee    0.097   0.078 
 Other–other versus 100 15.4 12.4–18.3    
 TCG–other 29 23.9 9.0–38.7  0.593 0.332–1.061  
ABCG2 34G/A    0.072   0.069 
 GG versus 124 15.4 12.5–18.3    
 AG 12 39.9 16.7–63.1  0.416 0.162–1.070  
Genetic factors pharmacodynamic pathway        
PDGFR-α haplotypef    0.002   0.108 
 GCGT–GCGT vs. 78 24.2 17.2–31.2    
 GCG–other + other–other 56 14.8 11.6–17.9  1.458 0.920–2.310  
VEGFR-2 1718T/A    0.022   0.016 
 AA + AT versus 125 16.3 12.4–20.2    
 TT 9.4 7.2–11.7  2.907 1.224–6.903  

aOnly factors with P < 0.1 level are presented; factors with P < 0.1 in the univariate analyses were selected for multivariate analyses.

bHR < 1.0 indicates that the factor associates with improved OS, HR > 1.0 associates with worse OS.

cRisk groups according to Memorial Sloan-Kettering Cancer Center (MSKCC) prognostic criteria [based on the 5 risk factors: low Karnofsky performance status (<80%), high LDH (>1.5 times the upper limit of normal), low serum hemoglobin, high corrected serum calcium (>10 mg/dL), and time from initial diagnosis to treatment <1 year] (22).

dMultiplicative model, HR per increase in MSKCC class or number of disease sites class.

Description haplotypes: e = ABCB1 3435C/T, 1236C/T and 2677G/T; f = PDGFR-α 1580T/C -1171C/G -735G/A -573G/T.

Baseline characteristics entered into the multivariate Cox models included the MSKCC risk factors, the number of metastatic sites, and age for PFS analyses, and the MSKCC risk factors and the number of metastatic sites for OS analyses. The ECOG performance status was excluded from the multivariate analyses due to colinearity with the MSKCC prognostic criteria (22).

Pharmacogenetic factors for sunitinib and progression-free survival

Among the 30 studied polymorphisms, only polymorphisms related to the pharmacokinetics of sunitinib were predictive of PFS (Table 2). A prolonged PFS was found in the univariate analysis of patients with presence of the A-allele in CYP3A5 6986A/G (P = 0.017), absence of a CAT copy in the NR1I3 haplotype (P = 0.021), presence of the C-allele in NR1I2 8055C/T (P = 0.025), presence of the C-allele in NR1I2 -25385C/T (P = 0.032), presence of a TCG copy in the ABCB1 haplotype (P = 0.072) and presence of the A-allele in ABCG2 34G/A (P = 0.077). Together with the MSKCC risk factors, the numbers of metastatic sites and age, these polymorphisms were entered into the multivariate Cox model.

Multivariate analysis confirmed the following factors as significant (<0.05) predictors of improved PFS: the MSKCC risk factors (HR: 1.988; 95% CI, 1.394–2.837), the number of metastatic sites (HR: 1.400; 95% CI, 1.042–1.880), age (HR: 1.031 per year increase; 95% CI, 1.003–1.060), presence of the A-allele in CYP3A5 6986A/G (HR: 0.266; 95% CI, 0.079–0.892), absence of a CAT copy in the NR1I3 haplotype (HR: 1.758; 95% CI, 1.108–2.790), and the presence of a TCG copy in the ABCB1 haplotype (HR: 0.522; 95% CI, 0.287–0.950).

Pharmacogenetic factors for sunitinib and overall survival

In univariate analysis, polymorphisms related to the pharmacokinetics and pharmacodynamics of sunitinib were predictive of OS (Table 3). Of the pharmacokinetic polymorphisms, presence of the C-allele in NR1I2-25385C/T (P = 0.017), presence of a TCG copy in the ABCB1 haplotype (P = 0.097) and presence of the A-allele in ABCG2 34G/A (P = 0.072) were associated with a prolonged OS. In addition, univariate analyses identified two pharmacodynamic polymorphisms including two GCGT copies in the PDGFR-α haplotype and presence of the A-allele in VEGFR-2 1718T/A as factors for a prolonged OS (P = 0.002 and 0.022, respectively).

Multivariate analysis confirmed the MSKCC risk factors (HR: 2.273; 95% CI, 1.595–3.238) and the presence of the A-allele in VEGFR-2 1718T/A (HR: 2.907; 95% CI, 1.224–6.903) as significant (< 0.05) predictors of a prolonged OS. In multivariate analysis, there was a trend toward an improved OS for patients with a TCG copy in the ABCB1 haplotype (HR: 0.593; 95% CI, 0.332–1.061; P = 0.078) or presence of the A-allele in ABCG2 34G/A (HR: 0.416; 95% CI, 0.162–1.070; P = 0.069).

Favorable genetic profiles and outcome

Polymorphisms that were associated with an improved PFS were combined in a predictive model. Patients were categorized as carriers of the favorable genetic profiles when they had at least an A-allele in CYP3A5, a TCG copy in the ABCB1 haplotype or a missing CAT copy in the NR1I3 haplotype. Carriers with a favorable genetic profile (n = 95) had an improved PFS and OS as compared with noncarriers (median PFS: 13.1 versus 7.5 months, P = 0.001 and median OS: 19.9 versus 12.3 months, P = 0.009). Multivariate analysis including the clinical factors showed consistent predictive value of the model for PFS and showed a trend for OS (HR: 0.541; 95% CI, 0.340–0.860, P = 0.009 and HR: 0.667; 95% CI, 0.420–1.058, P = 0.085, respectively; Fig. 2).

Figure 2.

Favorable () and nonfavorable genetic profile () in mRCC patients treated with sunitinib for progression-free (A) and overall survival (B) using multivariate Cox regression analysis. Patients were categorized as carriers of the favorable genetic profiles when they had at least an A-allele in CYP3A5, a TCG copy in the ABCB1 haplotype or a missing CAT copy in the NR1I3 haplotype.

Figure 2.

Favorable () and nonfavorable genetic profile () in mRCC patients treated with sunitinib for progression-free (A) and overall survival (B) using multivariate Cox regression analysis. Patients were categorized as carriers of the favorable genetic profiles when they had at least an A-allele in CYP3A5, a TCG copy in the ABCB1 haplotype or a missing CAT copy in the NR1I3 haplotype.

Close modal

In mRCC patients treated with sunitinib, the MSKCC risk groups and the number of metastatic sites are clinical factors that are usually associated with PFS and OS (2,3,7). However, these clinical factors are prognostic criteria that are associated with the extent of the disease and do not necessarily predict antitumor efficacy of a specific drug. As an increasing number of drugs is currently available for the treatment of mRCC (25), tools are needed to identify patients who predispose to benefit from sunitinib treatment and to select individual mRCC patients for treatment with this drug. The efficacy of sunitinib may be influenced by multiple genes encoding for enzymes, efflux transporters and targets related to the pharmacokinetics and pharmacodynamics of sunitinib. Therefore, we analyzed whether SNPs in the pharmacokinetic and pharmacodynamic pathways of sunitinib were predictive of PFS and OS in patients with clear-cell mRCC. Our study showed that next to 3 clinical characteristics (MSKCC prognostic criteria, number of metastatic sites, and age), 3 genetic variants in the CYP3A5, NR1I3, and ABCB1 genes were predictive factors for PFS. In addition, a role of an A-allele in VEGFR-2 1718T/A for a prolonged OS as a secondary outcome was found.

Clinical benefit from sunitinib treatment may depend on systemic exposure to sunitinib. Sunitinib is metabolized primarily to the active N-de-ethylated metabolite SU12662, which reaches similar plasma concentrations and has equipotent biochemical activity as the parent compound (26). Thereafter, SU12662 undergoes a second N-de-ethylation step, which occurs at a slower rate, to the inactive metabolite SU14335. Recently, a meta-analysis of pharmacokinetic data in 443 patients treated with sunitinib, showed that higher plasma levels of sunitinib and its active metabolite SU12662 were associated with a prolonged time–to–tumor progression and OS (27). Currently, it is not clear which underlying factors account for the observed interindividual differences in plasma levels of sunitinib and its active metabolite SU12662. Interindividual differences in sunitinib exposure may be the result of variations in sunitinib absorption, metabolism, distribution, and excretion through metabolizing enzymes and transporter proteins. Concerning the pharmacokinetics of sunitinib, the present study identified variants in three genes (CYP3A5, NR1I3, and ABCB1) as predictive factors for PFS in mRCC patients treated with sunitinib. Although these polymorphisms were not predictive of OS, there was a trend toward a prolonged OS for patients with a TCG copy in the ABCB1 haplotype. Additional treatment after discontinuation of sunitinib treatment may explain the discrepancy between the results of the PFS and OS analyses, as 26% of patients were subsequently treated with at least one other agent, including sorafenib (22%), temsirolimus (2%), and everolimus (4%).

CYP3A4 is the major key enzyme for the biotransformation of sunitinib (11). However, no polymorphisms of CYP3A4 were analyzed in the present study, as there are no functional polymorphisms in CYP3A4 that meet our described criteria for SNP selection yet. The CYP3A5 enzyme is another important enzyme for the metabolism of several TKIs including erlotinib, gefitinib, and imatinib (13). Similarly, the CYP3A5 enzyme may metabolize sunitinib and was therefore included in the analysis, though the sunitinib-metabolizing capacity of CYP3A5 has to be confirmed. In the current study, presence of the A-allele in CYP3A5, an SNP which leads to the CYP3A5 expressor phenotype (28, 29), was a predictive factor for a prolonged PFS. As the CYP3A5 expressor phenotype may lead to increased metabolism of sunitinib, these findings suggest that the prolonged PFS in patients with presence of the A-allele in CYP3A5 may be caused by increased levels of the active metabolite SU12662, which has a longer half-life than the parent compound (80–100 hours versus 40–60 hours; ref. 26). Furthermore, polymorphisms in other genes (NR1I2 and NR1I3) that regulate the expression of CYP3A4 (12) were identified as predictive factors for outcome in sunitinib-treated mRCC patients.

The efflux transporters ABCB1 and ABCG2 play an important role in drug absorption, excretion, cellular accumulation, and resistance (10). Consequently, polymorphisms in ABCB1 and ABCG2 may affect drug absorption and excretion of sunitinib. In the present study, the found associations between polymorphisms in the ABCB1 haplotype (a TCG copy) and the ABCG2 gene (presence of the A-allele), and improved outcome suggest that these polymorphisms may lead to reduced efflux transport of sunitinib into the gastrointestinal lumen and bile, resulting in increased systemic exposure of sunitinib. In vitro studies have reported conflicting data on the affinity of sunitinib for ABCB1 and ABCG2 (8, 9). Hu and colleagues (8) found a moderate affinity of sunitinib for ABCB1 and a negligible transport of sunitinib in cells overexpressing ABCG2, whereas another study reported higher sunitinib affinity for ABCG2 compared with ABCB1 (9). Furthermore, sunitinib reversed ABCG2-mediated multidrug resistance by inhibiting the drug efflux function of ABCG2 (30). This inhibitory capacity of sunitinib on ABCG2 appeared to be sensitive for the ABCG2 1291T > C genotype (31). In addition, another SNP in ABCG2 (421C > A) was associated with increased sunitinib exposure (32).

The ABC transporters may contribute to multidrug resistance in tumors by actively extruding drugs from cancer cells. In RCC, an increase in ABCB1 expression (33, 34) and activity (35) has been reported, suggesting a contribution of ABCB1 to the resistance of RCC to some anticancer drugs. Although polymorphisms in ABCB1 and ABCG2 may be associated with the development of RCC (36, 37), it is currently not known whether polymorphisms in ABCB1 and ABCG2 are associated with the expression and function of these transporters at the somatic level in renal cancer cells (38). Nevertheless, the role of efflux transporters in tumor cells may be limited for acquired resistance to sunitinib, which may develop after an initial response to sunitinib, as acquired resistance to sunitinib may be more related to physiological changes in the microenvironment of tumors, allowing reestablishment of angiogenesis during sunitinib treatment (16).

Clinical efficacy of treatment with TKIs may also be related to specific mutations in drug targets, as was shown for imatinib and gefitinib (39–40). Currently, it is not known which targets in RCC predict response to sunitinib or whether the somatic polymorphisms of targets in RCC correlate with genetic polymorphisms obtained from germline cells. Of the studied pharmacodynamic polymorphisms of sunitinib, only a polymorphism of VEGFR-2 1718T/A was associated with a decreased OS in multivariate analysis, whereas the presence of two GCGT copies in the PDGFR-α haplotype was associated with a prolonged OS in univariate analysis. However, no significant association between these polymorphisms and PFS was found. These findings may suggest that polymorphisms in VEGFR-2 and PDGFR-α may be associated with the nature of the disease and may therefore be prognostic instead of predictive. However, prospective validation in an independent mRCC cohort that is not treated with sunitinib is necessary to determine whether the associated polymorphisms of the present study are predictive markers of sunitinib activity or prognostic markers of mRCC disease.

In our previous study, several polymorphisms in genes involved in the pharmacokinetic and pharmacodynamic pathways of sunitinib were associated with sunitinib-induced toxicity (19). Polymorphisms of NR1I3 (absence of a CAG copy in the haplotype), ABCB1 (presence of a TTT copy), and VEGFR-2 (T allele in 1191 C/T) were significantly related with an increased risk for leucocytopenia, hand-foot syndrome, and any toxicity > grade 2, respectively (19). In the present study however, other genotypes or haplotypes in NR1I3, ABCB1, and VEGFR-2 were associated with clinical outcome. It is currently not clear how the different genotypes and haplotypes between these studies are related, but it is conceivable that severity and prevalence of some sunitinib-induced toxicities may basically depend on inhibition of a specific molecular pathway rather than variation in exposure, whereas other sunitinib-induced toxicities may mainly depend on exposure to the drug. In addition, the discrepancy between these two studies may be the result of different study populations, as patients with different malignancies were included in our previous study. Recently, Houk and colleagues (27) have shown a relationship between sunitinib exposure and the probability of grade ≥ 1 fatigue, the absolute neutrophil count, and the changes in diastolic blood pressure. However, these sunitinib-induced toxicities cannot be extrapolated to our previous toxicity study (19), as different toxicities and grades of toxicity were analyzed in both studies.

A potential limitation of the study is the retrospective design. As a result, pharmacokinetic data were not available to correlate polymorphisms of CYP3A5, NR1I3, and ABCB1 with plasma levels of sunitinib and its active metabolite SU12662. If future studies reveal a relation between sunitinib exposure and presence of an A-allele in CYP3A5 6986A/G, absence of a CAT copy in the NR1I3 haplotype or presence of a TCG copy in the ABCB1 haplotype, the sunitinib starting dose may be adjusted to dose-escalation of sunitinib > 50 mg/daily for patients without these genotypes and haplotypes. The nonbeneficial genetic profile may be used to select patients who may be eligible for alternative dosing schedules with intensive monitoring of plasma levels of sunitinib and its active metabolite SU12662. Before this genetic profile can be implemented, prospective validation in an independent patient population is necessary.

In conclusion, pharmacokinetic but not pharmacodynamic polymorphisms were independent predictive factors for PFS in patients with clear-cell mRCC who were treated with sunitinib. Patients with an A-allele in CYP3A5 6986A/G, absence of a CAT copy in the NR1I3 haplotype, or presence of a TCG copy in the ABCB1 haplotype had a prolonged PFS. These polymorphisms may be valuable factors to identify patients with reduced exposure to sunitinib in order to improve treatment strategies in these patients. The findings of this study advocate more pharmacokinetic studies in patients treated with sunitinib to further elucidate the role of these genetic determinants in sunitinib exposure and efficacy.

No potential conflicts of interest were disclosed.

We thank L. Vroling, R.R. de Haas, C. Tillier, T. van der Straaten, H. Mallo, L. Wever, J. Ouwerkerk, M.A.G. den Hollander, P. de Bruijn, and A. Nanninga for their help with sample and data collection, J. Swen for help with Figure 1, and E. Boven, A.K.L. Reyners, T. Korse, and J. Bonfrer for their cooperation.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Motzer
RJ
,
Bukowski
RM
. 
Targeted therapy for metastatic renal cell carcinoma
.
J Clin Oncol
2006
;
24
:
5601
8
.
2.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
,
Michaelson
MD
,
Bukowski
RM
,
Rixe
O
, et al
Sunitinib versus interferon alfa in metastatic renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
115
24
.
3.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
,
Michaelson
MD
,
Bukowski
RM
,
Oudard
S
, et al
Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma
.
J Clin Oncol
2009
;
27
:
3584
90
.
4.
Motzer
RJ
,
Michaelson
MD
,
Redman
BG
,
Hudes
GR
,
Wilding
G
,
Figlin
RA
, et al
Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma
.
J Clin Oncol
2006
;
24
:
16
24
.
5.
Motzer
RJ
,
Rini
BI
,
Bukowski
RM
,
Curti
BD
,
George
DJ
,
Hudes
GR
, et al
Sunitinib in patients with metastatic renal cell carcinoma
.
JAMA
2006
;
295
:
2516
24
.
6.
Faivre
S
,
Delbaldo
C
,
Vera
K
,
Robert
C
,
Lozahic
S
,
Lassau
N
, et al
Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer
.
J Clin Oncol
2006
;
24
:
25
35
.
7.
Van der Veldt
AA
,
Boven
E
,
Helgason
HH
,
Van Wouwe
M
,
Berkhof
J
,
De Gast
G
, et al
Predictive factors for severe toxicity of sunitinib in unselected patients with advanced renal cell cancer
.
Br J Cancer
2008
;
99
:
259
65
.
8.
Hu
S
,
Chen
Z
,
Franke
R
,
Orwick
S
,
Zhao
M
,
Rudek
MA
, et al
Interaction of the multikinase inhibitors sorafenib and sunitinib with solute carriers and ATP-binding cassette transporters
.
Clin Cancer Res
2009
;
15
:
6062
9
.
9.
Shukla
S
,
Robey
RW
,
Bates
SE
,
Ambudkar
SV
. 
Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2
.
Drug Metab Dispos
2009
;
37
:
359
65
.
10.
Dietrich
CG
,
Geier
A
,
Oude Elferink
RPJ
. 
ABC of oral bioavailability: transporters as gatekeepers in the gut
.
Gut
2003
;
52
:
1788
95
.
11.
Rock
EP
,
Goodman
V
,
Jiang
JX
,
Mahjoob
K
,
Verbois
SL
,
Morse
D
, et al
Food and drug administration drug approval summary: sunitinib malate for the treatment of gastrointestinal stromal tumor and advanced renal cell carcinoma
.
Oncologist
2007
;
12
:
107
13
.
12.
Tirona
RG
,
Lee
W
,
Leake
BF
,
Lan
LB
,
Cline
CB
,
Lamba
V
, et al
The orphan nuclear receptor HNF4[alpha] determines PXR- and CAR-mediated xenobiotic induction of CYP3A4
.
Nat Med
2003
;
9
:
220
4
.
13.
Van Erp
NP
,
Gelderblom
H
,
Guchelaar
HJ
. 
Clinical pharmacokinetics of tyrosine kinase inhibitors
.
Cancer Treat Rev
2009
;
35
:
692
706
.
14.
Brugarolas
J
. 
Renal-cell carcinoma–molecular pathways and therapies
.
N Engl J Med
2007
;
356
:
185
7
.
15.
Bergers
G
,
Song
S
,
Meyer-Morse
N
,
Bergsland
E
,
Hanahan
D
. 
Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors
.
J Clin Invest
2003
;
111
:
1287
95
.
16.
Rini
BI
,
Atkins
MB
. 
Resistance to targeted therapy in renal-cell carcinoma
.
Lancet Oncol
2009
;
10
:
992
1000
.
17.
Ferrara
N
,
Gerber
HP
,
LeCouter
J
. 
The biology of VEGF and its receptors
.
Nat Med
2003
;
9
:
669
76
.
18.
Carmeliet
P
,
Jain
RK
. 
Angiogenesis in cancer and other diseases
.
Nature
2000
;
407
:
249
57
.
19.
Van Erp
NP
,
Eechoute
K
,
Van der Veldt
AA
,
Haanen
JB
,
Reyners
AK
,
Mathijssen
RH
, et al
Pharmacogenetic pathway analysis for determination of sunitinib-induced toxicity
.
J Clin Oncol
2009
;
27
:
4406
12
.
20.
Van Erp
NP
,
Mathijssen
RHJ
,
Van der Veldt
AA
,
Haanen
JB
,
Reyners
AK
,
Eechoute
K
, et al
Myelosuppression by sunitinib is flt-3 genotype dependent
.
Br J Cancer
2010
;
103
:
757
8
.
21.
Gore
ME
,
Szczylik
C
,
Porta
C
,
Bracarda
S
,
Bjarnason
GA
,
Oudard
S
, et al
Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial
.
Lancet Oncol
2009
;
10
:
757
63
.
22.
Motzer
RJ
,
Bacik
J
,
Murphy
BA
,
Russo
P
,
Mazumdar
M
. 
Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma
.
J Clin Oncol
2002
;
20
:
289
96
.
23.
Kimchi-Sarfaty
C
,
Oh
JM
,
Kim
IW
,
Sauna
ZE
,
Calcagno
AM
,
Ambudkar
SV
, et al
A “silent” polymorphism in the MDR1 gene changes substrate specificity
.
Science
2007
;
315
:
525
8
.
24.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
25.
Rini
BI
. 
Metastatic renal cell carcinoma: many treatment options, one patient
.
J Clin Oncol
2009
;
27
:
3225
34
.
26.
Adams
VR
,
Leggas
M
. 
Sunitinib malate for the treatment of metastatic renal cell carcinoma and gastrointestinal stromal tumors
.
Clin Ther
2007
;
29
:
1338
53
.
27.
Houk
BE
,
Bello
CL
,
Poland
B
,
Rosen
LS
,
Demetri
GD
,
Motzer
RJ
. 
Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis
.
Cancer Chemother Pharmacol
2010
;
66
:
357
71
28.
Lee
SJ
,
Usmani
KA
,
Chanas
B
,
Ghanayem
B
,
Xi
T
,
Hodgson
E
, et al
Genetic findings and functional studies of human CYP3A5 single nucleotide polymorphisms in different ethnic groups
.
Pharmacogenetics
2003
;
13
:
461
72
.
29.
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
.
30.
Dai
CL
,
Liang
YJ
,
Wang
YS
,
Tiwari
AK
,
Yan
YY
,
Wang
F
, et al
Sensitization of ABCG2-overexpressing cells to conventional chemotherapeutic agent by sunitinib was associated with inhibiting the function of ABCG2
.
Cancer Lett
2009
;
279
:
74
83
.
31.
Kawahara
H
,
Noguchi
K
,
Katayama
K
,
Mitsuhashi
J
,
Sugimoto
Y
. 
Pharmacological interaction with sunitinib is abolished by a germ-line mutation (1291T>C) of BCRP/ABCG2 gene
.
Cancer Sci
2010
;
101
:
1493
500
.
32.
Mizuno
T
,
Terada
T
,
Kamba
T
,
Fukudo
M
,
Katsura
T
,
Nakamura
E
, et al
ABCG2 421C>A polymorphism and high exposure of sunitinib in a patient with renal cell carcinoma
.
Ann Oncol
2010
;
21
:
1382
3
.
33.
Szakacs
G
,
Annereau
JP
,
Lababidi
S
,
Shankavaram
U
,
Arciello
A
,
Bussey
KJ
, et al
Predicting drug sensitivity and resistance: profiling ABC transporter genes in cancer cells
.
Cancer Cell
2004
;
6
:
129
37
.
34.
Walsh
N
,
Larkin
A
,
Kennedy
S
,
Connolly
L
,
Ballot
J
,
Ooi
W
, et al
Expression of multidrug resistance markers ABCB1 (MDR-1/P-gp) and ABCC1 (MRP-1) in renal cell carcinoma
.
BMC Urol
2009
;
9
:
6
.
35.
Soto-Vega
E
,
Arroyo
C
,
Richaud-Patin
Y
,
Garcia-Carrasco
M
,
Vazquez-Lavista
LG
,
Llorente
L
. 
P-glycoprotein activity in renal clear cell carcinoma
.
Urol Oncol
2009
;
27
:
363
6
.
36.
Korenaga
Y
,
Naito
K
,
Okayama
N
,
Hirata
H
,
Suehiro
Y
,
Hamanaka
Y
, et al
Association of the BCRP C421A polymorphism with nonpapillary renal cell carcinoma
.
Int J Cancer
2005
;
117
:
431
4
.
37.
Siegsmund
M
,
Brinkmann
U
,
Schaffeler
E
,
Weirich
G
,
Schwab
M
,
Eichelbaum
M
, et al
Association of the P-glycoprotein transporter MDR1C3435T polymorphism with the susceptibility to renal epithelial tumors
.
J Am Soc Nephrol
2002
;
13
:
1847
54
.
38.
Haenisch
S
,
Zimmermann
U
,
Dazert
E
,
Wruck
CJ
,
Dazert
P
,
Siegmund
S
, et al
Influence of polymorphisms of ABCB1 and ABCC2 on mRNA and protein expression in normal and cancerous kidney cortex
.
Pharmacogenomics J
2006
;
7
:
56
65
.
39.
Heinrich
MC
,
Corless
CL
,
Demetri
GD
,
Blanke
CD
,
Von Mehren
M
,
Joensuu
H
, et al
Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor
.
J Clin Oncol
2003
;
21
:
4342
9
.
40.
Paez
JG
,
Janne
PA
,
Lee
JC
,
Tracy
S
,
Greulich
H
,
Gabriel
S
, et al
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy
.
Science
2004
;
304
:
1497
500
.

Supplementary data