ArticlePDF Available

Pharmacogenetic determinants associated with sunitinib-induced toxicity and ethnic difference in Korean metastatic renal cell carcinoma patients

Authors:

Abstract and Figures

The aim of this study was to investigate the pharmacogenetic determinants of sunitinib-related toxicity and ethnic difference in metastatic renal cell carcinoma (mRCC) among Korean patients. A pharmacogenetic study was performed in 65 patients with mRCC treated with the standard schedule of sunitinib (50 mg orally once daily for 4 weeks-on/2 weeks-off). Detailed data regarding the toxicity of sunitinib, including thrombocytopenia, neutropenia, anemia, and hand-foot syndrome (HFS), were prospectively collected in a clinical trial program (n = 38) or standard oncology practice (n = 27). Total of 12 genetic polymorphisms in 8 candidate genes (CYP1A1, CYP3A5, ABCB1, ABCG2, PDGFRα, VEGFR2, RET, and FLT3) were analyzed for an association with treatment-related toxicity from sunitinib using Pearson χ (2) test. Common grade 3 or grade 4 treatment-related toxicities were thrombocytopenia (36.9 %, 24/65), neutropenia (18.4 %, 12/65), anemia (7.7 %, 5/65), and HFS (12.3 %, 8/65). Patients carrying an ABCG2 421 AA genotype developed significantly more grade 3 or grade 4 thrombocytopenia, neutropenia, and HFS adjusted for age, sex, and Eastern Cooperative Oncology Group performance status, and body surface area (odds ratio compared with AC/CC genotypes [OR] 9.90, P = 0.04, thrombocytopenia; OR 18.20, P = 0.02, neutropenia; and OR 28.46, P = 0.01, HFS). In addition, total and surface protein ABCG2 protein expression was decreased in ABCG2 421 AA mutant cells compared to wild type. Among 12 genetic polymorphisms, polymorphism in the ABCG2 421C>A gene may be mostly associated with the risk of sunitinib-related toxicity in mRCC patients. Considering the high frequency of 421C>A SNP in Asian, this may be related to differential toxicities among ethnic groups.
Content may be subject to copyright.
1 3
Cancer Chemother Pharmacol (2013) 72:825–835
DOI 10.1007/s00280-013-2258-y
ORIGINAL ARTICLE
Pharmacogenetic determinants associated with sunitinib‑induced
toxicity and ethnic difference in Korean metastatic renal cell
carcinoma patients
Hye Ryun Kim · Hyung Soon Park · Woo Sun Kwon · Ji Hyun Lee ·
Yusuke Tanigawara · Sun Min Lim · Hyo Song Kim ·
Sang Jun Shin · Jung Bae Ahn · Sun Young Rha
Received: 16 March 2013 / Accepted: 1 August 2013 / Published online: 8 September 2013
© Springer-Verlag Berlin Heidelberg 2013
association with treatment-related toxicity from sunitinib
using Pearson χ2 test.
Results Common grade 3 or grade 4 treatment-related
toxicities were thrombocytopenia (36.9 %, 24/65), neu-
tropenia (18.4 %, 12/65), anemia (7.7 %, 5/65), and HFS
(12.3 %, 8/65). Patients carrying an ABCG2 421 AA gen-
otype developed significantly more grade 3 or grade 4
thrombocytopenia, neutropenia, and HFS adjusted for age,
sex, and Eastern Cooperative Oncology Group performance
status, and body surface area (odds ratio compared with
AC/CC genotypes [OR] 9.90, P = 0.04, thrombocytopenia;
OR 18.20, P = 0.02, neutropenia; and OR 28.46, P = 0.01,
HFS). In addition, total and surface protein ABCG2 protein
expression was decreased in ABCG2 421 AA mutant cells
compared to wild type.
Conclusion Among 12 genetic polymorphisms, poly-
morphism in the ABCG2 421C>A gene may be mostly
Abstract
Purpose The aim of this study was to investigate the
pharmacogenetic determinants of sunitinib-related toxic-
ity and ethnic difference in metastatic renal cell carcinoma
(mRCC) among Korean patients.
Methods A pharmacogenetic study was performed in 65
patients with mRCC treated with the standard schedule of
sunitinib (50 mg orally once daily for 4 weeks-on/2 weeks-
off). Detailed data regarding the toxicity of sunitinib,
including thrombocytopenia, neutropenia, anemia, and
hand–foot syndrome (HFS), were prospectively collected
in a clinical trial program (n = 38) or standard oncology
practice (n = 27). Total of 12 genetic polymorphisms in
8 candidate genes (CYP1A1, CYP3A5, ABCB1, ABCG2,
PDGFRα, VEGFR2, RET, and FLT3) were analyzed for an
Electronic supplementary material The online version of this
article (doi:10.1007/s00280-013-2258-y) contains supplementary
material, which is available to authorized users.
H. R. Kim · S. M. Lim · H. S. Kim · S. J. Shin · J. B. Ahn ·
S. Y. Rha
Department of Internal Medicine, College of Medicine, Yonsei
University, Seoul, Korea
H. R. Kim · S. M. Lim · H. S. Kim · S. J. Shin · J. B. Ahn
Division of Medical Oncology, Yonsei Cancer Center,
College of Medicine, Yonsei University, 50 Yonsei-ro, 134
Shinchon-dong, Seodaemun-gu, Seoul 120-752, Korea
H. S. Park · J. H. Lee
Department of Pharmacology, Pharmacogenomic Research
Center for Membrane Transporters, College of Medicine, Yonsei
University, Seoul, Korea
W. S. Kwon
Cancer Metastasis Research Center, College of Medicine, Yonsei
University, Seoul, Korea
W. S. Kwon
Brain Korea 21 Project for Medical Sciences, College
of Medicine, Yonsei University, Seoul, Korea
J. H. Lee
Research Center for Human Natural Defense System, College
of Medicine, Yonsei University, Seoul, Korea
Y. Tanigawara
Department of Clinical Pharmacokinetics
and Pharmacodynamics, School of Medicine, Keio University,
Minato, Japan
S. Y. Rha (*)
Yonsei Cancer Research Institute, College of Medicine, Yonsei
University, Seoul, Korea
e-mail: rha7655@yuhs.ac
826 Cancer Chemother Pharmacol (2013) 72:825–835
1 3
associated with the risk of sunitinib-related toxicity in
mRCC patients. Considering the high frequency of 421C>A
SNP in Asian, this may be related to differential toxicities
among ethnic groups.
Keywords Renal cell carcinoma · Sunitinib · Toxicity ·
Polymorphism
Introduction
Sunitinib is a small-molecule receptor tyrosine kinase
inhibitor (TKI) that has been approved as the first-line
treatment for metastatic renal cell carcinoma (mRCC). The
recommended dose and schedule for sunitinib is 50 mg
each day given orally for 4 consecutive weeks followed
by 2 weeks-off per treatment cycle. Adverse events (AEs)
that frequently occurred in patients treated with sunitinib
were hematological toxicities, including thrombocytopenia,
neutropenia, and anemia, and non-hematological toxicities
such as hand–foot syndrome (HFS) and oral mucositis. It
is noteworthy that sunitinib-induced toxicities are often
associated with treatment discontinuation, interruption, and
dose reduction in approximately 30 % of the patients in
studies conducted with Caucasian population [13].
Intriguingly, recent data reported that there are differ-
ences regarding severity in toxicity even with similar tox-
icity profiles between Asians and non-Asians and sunitinib-
induced AEs were much more common in Asians [46].
Furthermore, 90 % of patients underwent dose modification
related to the AEs in a Japanese study, 58 % of patients did
so in the Asian subpopulation in a global expanded access
program (EAP) study, and 76 % of patients in Korean pop-
ulation [4, 68]. Thus, sunitinib-induced toxicity is a seri-
ous problem that should not be ignored, especially in Asian
patients. A discovery of novel predictive factors regard-
ing sunitinib-induced toxicity will help to optimize drug
treatment in individual patients. So far, a few studies have
described clinicopathological factors such as low body sur-
face area (BSA), old age, female sex, or Asian ethnicity as
determinants associated with severe toxicity [6, 912]. The
pharmacokinetic results showed that the volume of sunitinib
distribution was decreased in females or in patients with low
body weight [13]. However, these reports are insufficient to
explain the phenomenon and individual patients’ diversity.
Although there is an emerging evidence of variability in
sunitinib-induced toxicity according to individual patients
or ethnic groups, the reason for heterogenous tolerability of
sunitinib is not clear. One possible explanation was suggested
in relation to polymorphisms in genes related to the pharma-
cogenetic or pharmacokinetic pathways of sunitinib [1, 14].
However, these studies were mostly conducted in Caucasian
population and Asian data have not been reported so far.
Genes that are related to the pharmacodynamic path-
ways of sunitinib are based on targeted genes including
vascular endothelial growth factor receptors (VEGFRs) 1,
2, and 3, platelet-derived growth factor receptor (PDGFR)
α and β, Fms-like tyrosine kinase 3 receptor (FLT3), and
the receptor encoded by the ret proto-oncogene (RET).
On the other hand, genes in the pharmacokinetic pathway
are the genes of metabolisms such as cytochrome P450
1A1 (CYP1A1) and cytochrome P450 3A5 (CYP3A5), and
the drug efflux genes of ATP binding cassette member G2
(ABCG2) and ATP binding cassette member B1 (ABCB1)
[14]. Especially, ABCG2, also known as breast cancer
resistance protein (BCRP), is an efflux transporter on the
small intestine and has been known to highly interact with
different TKIs such as gefitinib, erlotinib, imatinib, and
nilotinib [1524]. The ABCG2 421C>A non-synonymous
polymorphism is known to induce the changes of protein
expression or activity and be frequent polymorphism in
Korean population [2325].
In this study, we have evaluated whether polymorphisms
of genes regarding pharmacogenetic pathway can be asso-
ciated with sunitinib-induced toxicity and efficacy in
Korean mRCC patients.
Materials and methods
Study population and clinical data collection
We consecutively enrolled 65 histologically confirmed
mRCC patients who were treated with sunitinib and were
available for genetic analysis between March 2006 and
December 2010 at the Yonsei Cancer Center (YCC) in
Seoul, Korea, and who were available for genetic analysis.
Among 65 patients, 38 were treated within as an EAP trial
using sunitinib and the rest 27 patients were treated with
sunitinib as a part of standard oncology practice follow-
ing the similar protocol of the clinical trial. Sunitinib was
administered orally as a single agent at a dosage of 50 mg
daily, consisting of 4 weeks of treatment followed by
2-week rest periods in cycles of 6 weeks until progression
or intolerable toxicity occurred. In our study, we excluded
patients treated for less than one cycle. This study is a ret-
rospective study on blood samples and patients data col-
lected over the past years.
Demographic and clinical data of patients were collected
from the review of electronic medical records. Patient char-
acteristics considered relevant for sunitinib toxicity were
as follows age, sex, BSA, Eastern Cooperative Oncology
Group (ECOG) performance status, histologic type, and
organ function. Risk group stratification was done based
on the Memorial Sloan-Kettering Cancer Center (MSKCC)
risk factor [26]. This study was approved by the Severance
827
Cancer Chemother Pharmacol (2013) 72:825–835
1 3
Hospital Institutional Review Board and all patients signed
written informed consent for the genetic analysis.
Dose reductions of sunitinib were allowed depending on
the type and severity of AEs according to the current guide-
line. Sunitinib toxicity was evaluated using the National
Cancer Institute-Common Toxicity Criteria for Adverse
Effects (NCI-CTCAE) version 3.0. To assess AEs, physical
examination and laboratory assessments were performed at
baseline (before starting sunitinib), after 4 weeks of suni-
tinib therapy, and again after the 2-week rest period (just
before starting the second cycle). We used the AEs after
4 weeks in the first treatment cycle as clinical factor in the
analysis of this study. Among all collected data of AEs, we
focused on thrombocytopenia, neutropenia, anemia, and
HFS, which are more prevalent in Asians than in Caucasian
patients, leading to dose modification or interruption. The
primary outcome measures of this study were thrombocyto-
penia, neutropenia, anemia, and HFS of higher than grade
2 in the first cycle. We grouped the population into patients
with “low” toxicity (grades 0–2) and patients with “high”
toxicity (grades 3–4) for each toxicity type.
We also performed tumor assessments with computed
tomography (CT) or magnetic resonance imaging (MRI)
every 12 weeks until the end of the treatment. Clinical
responses were evaluated using the Response Evaluation
Criteria in Solid Tumor (RECIST version 1.1). PFS was
measured from the first day of sunitinib treatment to tumor
progression or death, whereas OS was measured from the
date of sunitinib treatment until the date of death. Patients
without a known date of death were censored at the time of
the last follow-up.
DNA isolation and analysis of polymorphism
Five milliliter of blood samples was collected in heparinized
tube from each patient and peripheral blood mononuclear
cells (PBMCs) were collected and frozen (80 °C) until the
assay. Genomic DNA (gDNA) was isolated from PBMCs
before sunitinib treatment. PBMCs were isolated from the
blood using Ficoll-Paque (Pharmacia, Uppsala, Sweden)
and gDNA was extracted with the LaboPass™ Blood kit
(Genotein Biotech, Korea) following the manufacturer’s
instructions. The extracted gDNA was then amplified by
PCR using an Eppendorf Master Cycler Gradient (Brink-
mann Instruments, Inc., USA). PCRs were performed.
For the SNaPshot analysis, exo/SAP-purified PCR prod-
ucts were mixed with AmpliTaq DNA polymerase, four flu-
orescently labeled dideoxynucleoside-5-triphosphates, and
the reaction buffer contained in an ABI Prism SNaPshot
multiplex kit (Applied Biosystems, Foster City, CA, USA).
Amplicons were then purified with exo/SAP and analyzed
on an ABI Prism 3700 Automated Sequencer (Applied Bio-
systems, Foster City, CA, USA). To analyze melting curves
with Taqman probe, we genotyped solution-phase hybridi-
zation reactions with 5-nuclease and fluorescence detec-
tion (TaqMan SNP genotyping assay, Applied Biosystems,
Foster City, CA, USA) in a 7300 Real-Time PCR system
(Applera, Norwalk, CT, USA). The PCR contained 20 ng
of genomic DNA, 1 μl of TaqMan Universal Master Mix,
900 nM of each primer, and 200 nM of VIC-labeled and
FAM-labeled probes in 25 μl reactions (Applera, Norwalk,
CT, USA). Amplification conditions were 95 °C for 10 min,
40 cycles of 92 °C for 15 s, and 60 °C for 1 min.
Selection of pharmacogenetic pathway
A total of 12 single-nucleotide polymorphisms (SNPs) in
8 candidate genes, CYP1A1, CYP3A5, ABCG2, ABCB1,
PDGFRα, VEGFR2, RET, and FLT3 were genotyped. Six
polymorphisms in four genes involved in the pharmacoki-
netics and the others involved in the pharmacodynamics of
sunitinib were selected (Table 1). Target polymorphisms
were selected based on the clinical relevance from the
previous reports or the assumption that non-synonymous
amino acid change might lead to the change of protein
functionality.
Assay for total and surface ABCG2 protein expression
To evaluate whether the polymorphism 421C>A might
influence on the expression of ABCG2 transporter, we con-
ducted in vitro experiment using Human Embryonic Kid-
ney (HEK) 293T cell line transfected with ABCG2 wild and
mutant type. First, to compare the total protein expression
level between HEK 293T cell line transfected with ABCG2
wild and mutant type, ABCG2 mutant cells were prepared.
HEK293T cell line was maintained in Dulbecco’s modi-
fied Eagle medium-HG (Invitrogen, Carlsbad, CA) supple-
mented with 10 % fetal bovine serum. Mammalian express-
ible ABCG2 plasmids were constructed using pcDNA 3.1.
Plasmids were transiently transfected into HEK293T cells
using Lipofectamine Plus Reagent (Invitrogen). Second,
total lysates were extracted and Western blotting was con-
ducted with total lysates following routine protocol. Anti-
ABCG2 antibody was purchased from Alexis Corporation
(Lausen, Switzerland). Third, to compare the ABCG2 pro-
tein expression on the surface membrane, we conducted
surface Biotinylation assay. HEK293T cells transfected
with ABCG2 gene wild and mutant type were grown to
80 % confluency in 6-well. The cells were washed with ice-
cold phosphate-buffered saline containing 0.1 Mm CaCl2
and 1 mM MgCl2, and the plasma membrane proteins were
then biotinylated by sulfo-NHS-SS-biotin (Pierce) in phos-
phate-buffered saline for 30 min at 4 °C. After biotinyla-
tion, the cells were washed extensively with 1 % bovine
serum albumin and phosphate-buffered saline to remove
828 Cancer Chemother Pharmacol (2013) 72:825–835
1 3
excess biotin. Then, the cells were lysed, Strepavidin solu-
tion (Streptavidin Agarose Resins, Pierce) was added to the
supernatant, and the mixture was incubated at 4 °C over-
night. Avidin-bound complexes were washed three times
and eluted in SDS sample buffer, resolved by SDS–PAGE,
and immunoblotted with anti-BCRP antibody (Alexis).
Data analysis
Statistical analysis was performed using SPSS 16 (SPSS
Inc., Chicago, IL, USA). Haplotype analysis was performed
using Haploview 3.2 program based on a standard expecta-
tion–maximization algorithm to construct haplotype blocks
[27]. The data were summarized using standard descrip-
tive statistics. For toxicity analysis, we used dichotomous
end points expressed as toxicity higher than grade 2 (yes
or no). All demographic and clinical variables were tested
univariately against the selected primary outcomes using
the χ2 test, Fisher’s exact test, and t tests where appropri-
ate. The statistical evaluation of genotype data and haplo-
types was tested univariately against the selected primary
outcomes using a χ2 test. Genotype frequencies at each
locus were also tested for Hardy–Weinberg equilibrium.
Firstly, association test was conducted with allele or geno-
type level for screening of significant locus. In this study,
twelve loci were evaluated and P value was decided by
Bonferroni’s correction (<0.004). Locus was derived from
screening step, and univariate and multivariate analyses
were performed. All multivariate logistic regression analy-
ses with toxicity were corrected for age, sex, and ECOG
performance status. The Kaplan–Meier method was used to
estimate PFS and OS, and the differences among genetic
polymorphisms were compared using the log-rank test.
All results from univariate and multivariate analyses with
P less than 0.05 were considered significant. All P values
were based on a two-sided hypothesis.
Results
Patients
This study included 51 men and 14 women with a median
age of 59 years (range 36–81). The histology was clear
cell type in 61 patients (93.8 %) and papillary type in
3 patients (4.6 %). Most patients (55/65, 84.6 %) had an
ECOG performance status 0 or 1. Among these 65 patients,
40 (61.5 %) were treatment naïve and 25 (38.5 %) had
received previous medical treatment including 16 patients
treated with cytokine treatment, 6 patients with chemother-
apy, and 3 patients with both immunotherapy and chemo-
therapy. All patients were treated with sunitinib as the
first VEGFR-TKI. Regarding the prognostic stratification,
most patients (90.8 %) were favorable or intermediate risk
group based on the MSKCC risk factor [26]. The detailed
patients’ characteristics are presented in Table 2.
Treatment outcome
All 65 patients received at least 1 week of sunitinib, and
62 (95.3 %) patients received more than 1 cycle of suni-
tinib. With the median 23.8 (range 1.1–57.9) months of
Table 1 Polymorphism genotyped in the pharmacokinetic and pharmacodynamic pathways of sunitinib
Gene rs number Polymorphism Region
CYP1A1 Cytochrome P450 1A1 rs1048943 2,455 A>G Non-synonymous I462V
CYP3A5 Cytochrome P450 3A5 rs776746 219–237 A>G Intron
ABCB1 ATP binding cassette
member B1
rs1128503 1,236 C>T Synonymous G412G
rs2032582 2,677 G>T/A Non-synonymous A893S/T
rs1045642 3,435 C>T Synonymous I1145I
ABCG2 (BCRP) ATP binding
cassette member G2
rs2231142 421 C>A Non-synonymous Q141K
PDGFRαPlatelet-derived
growth factor receptor
rs1800812 537 G>T Promoter
rs35597368 1,580 C>T Non-synonymous P478S
VEGFR2 (KDR) Vascular endothelial
growth factor receptors
rs2305948 889 G>A Non-synonymous V297I
rs1870377 1,416 T>A Non-synonymous H472Q
RET Ret proto-oncogene rs1799939 2,071 G>A Non-synonymous G691S
FLT3 Fms-like tyrosine
kinase 3 receptor
rs1933437 680 C>T Non-synonymous T227M
829
Cancer Chemother Pharmacol (2013) 72:825–835
1 3
follow-up, the median number of treatment cycle was
7 (range 1–38). Main reasons for drug discontinuation
were disease progression (n = 48, 73.8 %) and treat-
ment-related toxicity (n = 3, 4.7 %; 2 patients with both
grades 3–4 hematological and non-hematological tox-
icities; 1 patient with HFS). In terms of dose intensity,
when we calculated the RDI for 62 patients who received
more than one cycles of treatment, the mean and median
RDI for sunitinib were 80.2 % (SD ± 13.1) and 81.1 %
(range 39.0–100). In terms of the best response to suni-
tinib, 4.6 % (3/65) had a complete response (CR), 36.9 %
(24/65) had a partial response (PR), 49.2 % (32/65) had
Table 2 Patient demographics
and clinical characteristics
Risk factors are ECOG PS2,
low hemoglobin, and high
calcium. For patients without
prior cytokine treatment,
additional risk factors were
raised lactate dehydrogenase
and the time of use of interferon
α of <1 year. Patients with
prior cytokine treatment
were classified as favorable,
intermediate, or poor if 0, 1, or
>1 risk factors were present,
respectively. Patients without
prior cytokine treatment
were classified as favorable,
intermediate, or poor if 0, 1–2,
or >2 risk factors were present,
respectively
a All three patients are papillary
type
b ECOG PS = Eastern Coop-
erative Oncology Group perfor-
mance status
c MSKCC = Memorial Sloan-
Kettering Cancer Center
Characteristics Number (%)
Total patient 65 (100)
Sex
Male 51 (78.5)
Female 14 (21.5)
Physiological factor
Age—year (median, range) 59, 36–81
Weight (mean ± SD) 65.3 ± 10.3
BSA (mean ± SD) 1.7 ± 0.1
Histology
Clear 61 (93.8)
Non-clear cell typea3 (4.6)
Unknown 1 (1.5)
ECOG performance statusb
0 18 (27.7)
1 37 (56.9)
2 8 (12.3)
3 1 (1.5)
Previous surgical treatment
Nephrectomy 55 (84.6)
Previous systemic treatment
No 40 (61.5)
Immunotherapy only 16 (24.6)
Chemotherapy only 6 (9.3)
Immunotherapy and chemotherapy 3 (4.6)
Number of previous systemic treatment
0 40 (61.5)
1 22 (33.8)
2 3 (4.7)
Modified MSKCC risk groupc
Favorable 38 (58.5 %)
Intermediate 21(32.3 %)
Poor 6 (9.2 %)
Number of disease site Median 2 (range 1–5)
1 31 (47.6)
2 26 (40.0)
3 8 (12.4)
Common site of metastasis
Lung 45 (69.2)
LN 11 (16.9)
Bone 11 (16.9)
Brain 2 (3.0)
Liver 5 (7.6)
830 Cancer Chemother Pharmacol (2013) 72:825–835
1 3
stable disease (SD), and 9.2 % (6/65) had progressive
disease (PD). The median OS for all patients was not
reached in our study (Supplementary Figure 1A), while
the median PFS for all patients was 13.5 months (Sup-
plementary Figure 1B) with similar PFS in patients with
or without prior treatment (median 14.3 vs. 13.8 months,
respectively, P = NS). In terms of MSKCC risk group,
favorable (n = 38, 58.5 %), intermediated (n = 21,
32.3 %), and poor risk groups (n = 6, 9.2 %) showed the
trends toward shorter PFS (16.3, 7.7, and 3.5 months,
respectively).
Toxicities
Dose reduction was reported in 48 (73.8 %) patients and
treatment interruption in 31 (47.6 %) patients. Sunitinib
was reduced to 37.5 mg/day in 38 patients (58.4 %) and
to further 25 mg/day reductions in 10 (15.4 %) patients.
The frequently developed AEs in all grades during the
first treatment cycle were thrombocytopenia (46.2 %,
30/65), neutropenia (46.2 %, 30/65), anemia (32.3 %,
21/65), and HFS (50.8 %, 33/65). Among them, 36.9 %
(24/65) of grade 3 or grade 4 toxicities developed in
thrombocytopenia, 18.4 % (12/65) in neutropenia, 7.6 %
(5/65) in anemia, and 12.3 % (8/65) in HFS. There was
no sunitinib treatment-related mortality in our study
(Table 3).
Pharmacogenetic determinants for sunitinib-induced
toxicity
To evaluate the association between genetic polymor-
phism and sunitinib-related toxicity, an association
analysis was performed in 12 candidate SNPs (Table 4).
Genotype distributions at all loci were consistent with
Hardy–Weinberg equilibrium (each P > 0.05). Among
the 12 candidate SNPs, the ABCG2 421C>A (Q141K)
polymorphism showed a strong association with suni-
tinib-related toxicity, especially HFS, at the allele fre-
quency level. Regarding HFS toxicities, the “high” tox-
icity (grades 3–4) patient group was more likely to have
the “A” allele than the “low” toxicity (grades 0–2) patient
group (68.75 vs. 25.43 %, P = 0.00003; OR of A vs. C,
6.76, CI 2.16–21.13). The P value for ABCG2 421C>A
remained significant at 0.0003 after employing Bonferro-
ni’s correction for multiple comparisons. However, other
gene polymorphisms showed no difference between the
two groups. In haplotype analysis, haplotypes of ABCB1,
VEGFR2, and PDGFR α was evaluated for association
with toxicity. However, there was no significant associa-
tion between four toxicities and haplotypes (Supplemen-
tary figure 2, Supplementary table 2).
We performed a multivariate logistic regression anal-
ysis for the selected ABCG2 421C>A polymorphism
through univariate analysis to evaluate the association
between sunitinib-related toxicity and this polymorphism.
We analyzed the association between selected end points
of thrombocytopenia, neutropenia, anemia, and HFS and
ABCG2 421C>A polymorphism. The results of the mul-
tivariate logistic regression analysis for the thrombocyto-
penia, neutropenia, anemia, HFS, and any toxicity higher
than grade 2 are summarized in Table 5. To evaluate the
role of ABCG2 421C>A polymorphism in predicting the
sunitinib-induced toxicities, we conducted logistic regres-
sion to compensate for compounding factors (age, sex,
ECOG PS, and BSA). ABCG2 421 AA genotype was asso-
ciated with sunitinib-induced toxicity such as thrombocy-
topenia (OR 9.90, CI 1.16–infinity, P = 0.04), neutrope-
nia (OR 18.20, CI 1.49–222.09, P = 0.02), and HFS (OR
28.46, CI 2.22–364.94, P = 0.01). The presence of the AA
genotype at the ABCG2 421C>A locus was related to a
9.9-fold increase in the risk for thrombocytopenia, 18.2-
fold increase in the risk for neutropenia, and 28.46-fold
increase in the risk of HFS.
Table 3 Distribution of toxicity during first cycle
a Toxicities were evaluated by NCI-CTC version 3.0
b “No” represents grade 0,1, or 2 toxicities
c “Yes” represents grade 3 or 4 toxicities
Toxicity gradeaNo %
Thrombocytopenia 30 46.2
1 1 1.5
2 5 7.7
3 23 35.4
4 1 1.5
Neutropenia 30 46.2
1 8 12.3
2 10 15.4
3 11 16.9
4 1 1.5
Anemia 21 32.3
1 11 16.9
2 5 7.7
3 5 7.7
4 0 0
Any hematologic toxicity >grade 2
Nob38 58.4
Ye sc27 41.6
Hand–foot syndrome 33 50.8
1 15 23.1
2 10 15.4
3 8 12.3
4 0 0
831
Cancer Chemother Pharmacol (2013) 72:825–835
1 3
Table 4 Association of each genetic variation with sunitinib-related toxicity in mRCC patients
Gene name; polymorphic site, allele; amino acid; rs number
Polymorphism Minor Thrombocytopenia Neutropenia Anemia Hand–foot syndrome
Allele Odds ratio 95 % CI P value Odds ratio 95 % CI P value Odds ratio 95 % CI P value Odds ratio 95 % CI P value
CYP1A1
c.2,455 A>G
I462V
G 0.6 0.24–1.49 0.27 0.45 0.12–1.64 0.22 0.61 0.14–2.54 0.44 1.22 0.36–4.13 0.75
CYP3A5
c.219–237 A>G
Intron 3
A 0.42 0.17–1.08 0.07 0.60 0.19–1.92 0.39 1.46 0.35–6.03 0.69 0.43 0.09–2.00 0.36
ABCB1
c.1,236 C>T
G412G
C 1.12 0.54–2.31 0.77 0.56 0.21–1.46 0.23 1.0 0.26–3.73 0.63 0.65 0.21–1.99 0.45
ABCB1
c.2,677 G>T/A
A893S/T
T 1.00 0.48–2.11 0.99 1.12 0.45–2.80 0.81 0.29 0.06–1.45 0.18 1.11 0.37–3.28 0.85
ABCB1
c.3,435 C>T
I1145I
T 0.87 0.41–1.83 0.71 0.89 0.35–2.28 0.82 0.33 0.08–1.25 0.16 1.11 0.38–3.28 0.85
ABCG2
c.421 C>A
Q141K
A 1.74 0.81–3.75 0.15 1.89 0.76–4.75 0.17 1.85 0.38–9.14 0.72 6.76 2.16–21.13 0.00003
PDGFRα
–573G>T
Promoter
T 0.508 0.19–1.38 0.18 0.347 0.076–1.590 0.244 2.04 0.24–16.95 0.69 0.264 0.03–2.10 0.302
PDGFRα
c.1,580 C>T
P478S
C 0.638 0.23–1.77 0.386 0.416 0.090–1.923 0.362 1.80 0.22–15.01 0.49 0.313 0.04–2.51 0.467
VEGFR2
c.889 G>A
V297I
A 0.48 0.15–1.58 0.22 0.25 0.03–1.94 0.19 1.09 0.92–1.16 0.35 2.59 0.73–9.23 0.23
VEGFR2
c.1,416 T>A
H472Q
T 0.87 0.42–1.79 0.70 0.89 0.37–2.20 0.81 1.84 0.45–7.48 0.514 1.33 0.47–3.78 0.59
RET
c.2,071 G>A
G691S
A 0.68 0.22–2.06 0.49 2.88 0.94–8.78 0.09 0.57 0.11–2.94 0.61 2.59 0.73–9.22 0.23
FLT3
c.680 C>T
T227M
C 1.24 0.57–2.71 0.59 1.671 0.658–4.246 0.277 0.56 0.15–2.14 0.46 0.818 0.25–2.72 1
832 Cancer Chemother Pharmacol (2013) 72:825–835
1 3
Pharmacogenetic determinants for sunitinib treatment
outcomes
Among the 12 single-nucleotide polymorphisms in 8 can-
didate genes, only a few polymorphisms from pharmaco-
dynamic genes were associated with OS. Prolonged OS
was found in the univariate analysis of patients with the
A” allele in RET 2251 G/A (GG vs. GA/AA, P = 0.029),
with the “C” allele in PDGFR α 1580T/C (TT vs. TC/CC,
P = 0.037), without the GT in PDGFR α diplotype (GT/
GT vs. others, P = 0.023), and with the GG genotype in
KDR (VEGFR2) (GG vs. GA/AA, P = 0.079) (Supple-
mentary Figure 3). However, these loci were not satisfied
with Bonferroni’s correction cutoff value (<0.004). Also,
there was no association between genetic polymorphism
and PFS in our study.
ABCG2 421 C>A polymorphisms and transporter
expression
To evaluate whether the SNP might influence on the
expression level of the transporter, we conducted in vitro
experiment comparing ABCG2 expression between wild
type and ABCG2 421AA mutant type. Total expression
of ABCG2 protein in whole cell lysate was evaluated by
Western blotting, while ABCG2 protein expression on the
cell surface membrane was done by Biotinylation assay. As
a result, total and surface protein in the mutant transfected
cells was decreased compared with those in the wild-type
ABCG2 cells (Fig. 1). This finding implicates that patients
with Q141K variation of ABCG2 show reduced ABCG2
transporter activity. Decreased transporter expression might
influence in reducing the export into the intestinal lumen
and thus increase the plasma concentration of sunitinib,
which can be associated with increased sunitinib-induced
toxicity risk in patients receiving sunitinib therapy.
Discussion
To the best of our knowledge, this is the study to analyze
the association between sunitinib-induced toxicities and
pharmacogenetic determinants in Asian mRCC patients
treated with sunitinib. The main finding of our study was
that the genetic polymorphism in the ABCG2 421C>A was
associated with the sunitinib-induced toxicities. The risks
of thrombocytopenia, neutropenia, and HFS were signifi-
cantly increased when the AA genotype in ABCG2 421C>A
was present. Consistent with previous reports, our study
showed a higher incidence of thrombocytopenia, neutro-
penia, anemia, and HFS as sunitinib-related toxicities than
those in Caucasian population. Considering the high fre-
quency of ABCG2 421C>A polymorphism in Asian indi-
viduals, this polymorphism might be regard as the determi-
nants for ethnic difference.
According to the International HapMap Project data
(http://www.hapmap.org/), the allelic frequency for this
ABCG2 421C>A differed significantly among ethnic
groups. The minor allele frequency was much lower in
Caucasians than in Asians. The “A” allele of 421C>A
Table 5 Multivariate analysis of ABCG2 421 polymorphism’s
predictive impact on sunitinib-induced toxicities
Bold values represent statistical significance
OR odds ratio, CI confidence interval
a Adjusted for age, sex, ECOG PS, and BSA
ORa95 % CI P value
Thrombocytopenia
ABCG 2 421C/A
CC + CA 1
AA 9.90 1.16–infinity 0.04
Neutropenia
ABCG 2 421C/A
CC + CA 1
AA 18.20 1.49–222.09 0.02
Anemia
ABCG 2 421C/A
CC + CA 1
AA 1.31 0.72–13.21 NS
Hand–foot syndrome
ABCG 2 421C/A
CC + CA 1
AA 28.46 2.22–364.94 0.01
Any hematologic toxicity >grade 2
ABCG 2 421C/A
CC + CA 1
AA 8.24 0.99–infinity 0.05
Fig. 1 ABCG2 protein expression on the cell surface membrane was
done by Biotinylation assay and the total expression of ABCG2 pro-
tein in whole cell lysate was evaluated by Western blotting. Total and
surface protein in the Q141K mutant transfected cells was decreased
compared with those in the wild-type ABCG2 cells. Aldolase was
used as a loading control
833
Cancer Chemother Pharmacol (2013) 72:825–835
1 3
(rs2231142) was found less frequently in Utah residents
with ancestry from northern and western Europe (CEUs)
(11.1 %) than in Asians (Korean, 27.5 %; Japanese, 34.1 %;
Chinese, 29.2 %) [25]. The “A” allele of 421C>A associ-
ated with sunitinib-related toxicity in this study might be
susceptible in other Asian populations, as a case report
from Japan also showed the same results [28]. This ethnic
difference in polymorphism could explain the ethnic differ-
ence in sunitinib toxicity.
Even though RCC is relatively uncommon in the Asian
population compared to the Western population, its inci-
dence is increasing in developed Asian nations such as
Japan or Korea [7]. With increasing experiences with suni-
tinib treatment in Asian mRCC patients, incidences of AEs
are also rising in Asian patients [4, 6, 7]. Recently, Houk
et al. [29] demonstrated that “Asian race” influences the
sunitinib pharmacokinetics by lowering of clearance for
sunitinib and metabolites. These data could explain the
higher toxicities in Asian than those in Caucasian. Suni-
tinib-induced toxicity in Asian population could disturb the
treatment by discontinuation, interruption, and dose reduc-
tion. Although differences in sunitinib-induced toxicity
among ethnicities or individuals are currently well recog-
nized, information regarding the determinants of toxicity is
still scarce.
Recent studies suggested that polymorphisms in specific
genes encoding for metabolizing enzymes, efflux trans-
porters, and targets of sunitinib could be associated with
sunitinib-related toxicities and drug efficacy [1, 14]. In
our study, individual variation of sunitinib-induced toxic-
ity was explained by genetic polymorphisms related to the
pharmacokinetic pathway. However, there were discrepan-
cies among each study because the incidence of genetic
polymorphisms was low and kinds of polymorphisms were
various according to the population.
Our study demonstrated that genetic polymorphism of
ABCG2 421 C>A was associated with sunitinib-induced
toxicity. Differences in sunitinib-induced toxicities accord-
ing to genetic polymorphisms can be explained by under-
standing the function of the ABCG2 transporter. The poten-
tial function of ABCG2 transporters regarding sunitinib
disposition has been explained by various in vitro experi-
ments [30]. These data demonstrated that sunitinib is a
substrate for the ABCG2 transporter and inhibits the efflux
of sunitinib metabolite through ABCG2 transporter via
direct binding [16, 17]. Additionally, polymorphisms in the
ABCG2 gene might have an important impact on ABCG2
protein expression, localization, and function [30]. Our in
vitro data as well as previous data demonstrated that the
non-synonymous SNP of ABCG2 421 C>A, which causes a
glutamine-to-lysine amino acid substitution at position141
(Q141K), has been associated with markedly decreased
levels of ABCG2 protein expression and/or activity [23, 31,
32]. The reduced protein levels and altered ATPase activ-
ity of the AA variant in ABCG2 421 C>A located in api-
cal membrane of small intestinal enterocytes, hepatocytes,
and proximal tubule cells in kidney might affect the oral
absorption and/or elimination of sunitinib [23]. There have
been reports that individuals carrying the ABCG2 421
AA homozygous variant showed higher drug exposure of
ABCG2 substrate such as fluvastatin, pravastatin, simvasta-
tin, and gefitinib [23, 32]. Recently, a Japanese case report
of five mRCC patients demonstrated that the homozygous
AA variant may be associated with severe AEs showing
2.5-fold higher maximum concentration and area under the
concentration of sunitinib [28]. Consistently, our results
showed that sunitinib-induced toxicities were more fre-
quent in the AA genotype of ABCG2 421C>A. Therefore,
based on the previous reports, we could infer that homozy-
gous AA genotype of ABCG2 421C>A reflected in elevated
sunitinib exposure that could explain for more toxicity in
patients with this genotype.
With rapidly evolving therapeutic options for mRCC
[28], predictive biomarkers of mRCC patients treated with
molecular targeted agents are of great clinical benefit.
Although validation of the Motzer’s criteria [26] is impor-
tant, a further implementation of RCC molecular biology
knowledge would be ideal for finding a new predictive
marker in new era of molecular targeted therapies. Serum
VEGF, sVEGFR-2, and sVEGFR-3, which are involved in
the VEGF signaling pathway, might be of value as predic-
tive biomarkers for sunitinib efficacy [3336]. Recent ret-
rospective analysis showed that hypertension after sunitinib
treatment is associated with improved clinical outcomes
[33]. However, these biomarkers or clinicopathological fac-
tors do not reflect the diversity of each individual patient.
Furthermore, clinical factors such as hypertension could
be known after sunitinib treatment and thus it could not be
predictive marker before treatment. Therefore, efforts to
identify genetic markers such as SNPs continue in order to
develop predictive biomarkers of efficacy with respect to
individual diversity before treatment [9, 37].
We additionally analyzed the association between
genetic polymorphism and survival outcome. Genetic pol-
ymorphism in RET 2251 G>A, PDGFR α 1580T>C, and
KDR (VEGFR2) genes was related to OS, but no genetic
polymorphism from our study showed an association with
PFS. One of the explanations may be that clinical responses
might be related to tumor tissue itself, but germ line poly-
morphisms of DNA isolated from PBMC do not reflect
tumor tissue. Therefore, somatic polymorphism using DNA
extracted from tumor tissue would be needed to predict the
treatment outcome. This study is underpowered because
this was retrospectively conducted to evaluate the associa-
tion between genetic polymorphism and toxicity. There-
fore, more studies will be warranted to validate the role of
834 Cancer Chemother Pharmacol (2013) 72:825–835
1 3
ABCG2 polymorphisms in sunitinib toxicity in the future.
In conclusion, the polymorphism in ABCG2 421 C>A
could be a biomarker in association with sunitinib-induced
toxicity in patients with Asian mRCC. An ABCG2 poly-
morphism may partly explain why individual patients show
different toxicities and Asian patients show more severe
toxicities than Caucasian patients. If the predictive role of
ABCG2 polymorphisms is confirmed in future prospec-
tive study, genotyping for ABCG2 could become a clinical
routine practice to select the appropriate dose and help to
achieve the benefits of personalized medicine. In the next
step, prospective pharmacokinetic and safety study may be
needed to confirm the role of ABCG2 in sunitinib toxicity.
An optimal recommended dose for ABCG2 421AA patients
can be also estimated in future pharmacogenetic and phar-
macokinetic safety study.
Acknowledgments This study was supported by a grant from the
Korea Healthcare Technology R&D Project of the Ministry of Health
and Welfare of Korea (A110641) by grant of Health Fellowship Foun-
dation, and by the Public Welfare & Safety research program through
the National Research Foundation of Korea (NRF), funded by the
Ministry of Science, ICT & Future Planning (2010-0020841).
Conflict of interest The authors have no potential conflicts of inter-
est to disclose.
References
1. Garcia-Donas J, Esteban E, Leandro-Garcia LJ, Castellano DE,
del Alba AG, Climent MA, Arranz JA, Gallardo E, Puente J, Bell-
munt J, Mellado B, Martinez E, Moreno F, Font A, Robledo M,
Rodriguez-Antona C (2011) Single nucleotide polymorphism
associations with response and toxic effects in patients with
advanced renal-cell carcinoma treated with first-line sunitinib:
a multicentre, observational, prospective study. Lancet Oncol
12:1143–1150
2. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski
RM, Oudard S, Negrier S, Szczylik C, Pili R, Bjarnason GA,
Garcia-del-Muro X, Sosman JA, Solska E, Wilding G, Thomp-
son JA, Kim ST, Chen I, Huang X, Figlin RA (2009) Overall sur-
vival and updated results for sunitinib compared with interferon
alfa in patients with metastatic renal cell carcinoma. J Clin Oncol
27:3584–3590
3. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski
RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen
I, Bycott PW, Baum CM, Figlin RA (2007) Sunitinib versus
interferon alfa in metastatic renal-cell carcinoma. N Engl J Med
356:115–124
4. Lee SCH, Mainwaring P, Ng C, Chang JWC, Kwong P et al
(2009) An Asian subpopulation analysis of the safety and effi-
cacy of sunitinib in metastatic renal cell carcinoma. Eur J Cancer
Suppl (abstract) 7:428
5. Naito S, Tomita Y, Rha SY, Uemura H, Oya M, Song HZ, Zhong
LH (2010) Wahid MI Kidney cancer working group report. Jpn J
Clin Oncol 40(Suppl 1):i51–i56
6. Kim HS, Hong MH, Kim K, Shin SJ, Ahn JB, Jeung HC, Chung
HC, Koh Y, Lee SH, Bang YJ, Rha SY (2011) Sunitinib for Asian
patients with advanced renal cell carcinoma: a comparable effi-
cacy with different toxicity profiles. Oncology 80:395–405
7. Tomita Y, Shinohara N, Yuasa T, Fujimoto H, Niwakawa M,
Mugiya S, Miki T, Uemura H, Nonomura N, Takahashi M,
Hasegawa Y, Agata N, Houk B, Naito S, Akaza H (2010) Overall
survival and updated results from a phase II study of sunitinib in
Japanese patients with metastatic renal cell carcinoma. Jpn J Clin
Oncol 40:1166–1172
8. Hong MH, Kim HS, Kim C, Ahn JR, Chon HJ, Shin SJ, Ahn
JB, Chung HC, Rha SY (2009) Treatment outcomes of sunitinib
treatment in advanced renal cell carcinoma patients: a single can-
cer center experience in Korea. Cancer Res Treat 41:67–72
9. van der Veldt AA, Boven E, Helgason HH, van Wouwe M, Berk-
hof J, de Gast G, Mallo H, Tillier CN, van den Eertwegh AJ,
Haanen JB (2008) Predictive factors for severe toxicity of suni-
tinib in unselected patients with advanced renal cell cancer. Br J
Cancer 99:259–265
10. Shirao K, Nishida T, Doi T, Komatsu Y, Muro K, Li Y, Ueda E,
Ohtsu A (2010) Phase I/II study of sunitinib malate in Japanese
patients with gastrointestinal stromal tumor after failure of prior
treatment with imatinib mesylate. Invest New Drugs 28:866–875
11. Guo F, Letrent SP, Sharma A (2007) Population pharmacokinet-
ics of a HER2 tyrosine kinase inhibitor CP-724,714 in patients
with advanced malignant HER2 positive solid tumors. Cancer
Chemother Pharmacol 60:799–809
12. Sparreboom A, Wolff AC, Mathijssen RH, Chatelut E, Rowin-
sky EK, Verweij J, Baker SD (2007) Evaluation of alternate size
descriptors for dose calculation of anticancer drugs in the obese. J
Clin Oncol 25:4707–4713
13. Houk BE, Bello CL, Kang D, Amantea M (2009) A population
pharmacokinetic meta-analysis of sunitinib malate (SU11248)
and its primary metabolite (SU12662) in healthy volunteers and
oncology patients. Clin Cancer Res 15:2497–2506
14. van Erp NP, Eechoute K, van der Veldt AA, Haanen JB, Reyners
AK, Mathijssen RH, Boven E, van der Straaten T, Baak-Pablo
RF, Wessels JA, Guchelaar HJ, Gelderblom H (2009) Pharmaco-
genetic pathway analysis for determination of sunitinib-induced
toxicity. J Clin Oncol 27:4406–4412
15. Ozvegy-Laczka C, Varady G, Koblos G, Ujhelly O, Cervenak
J, Schuetz JD, Sorrentino BP, Koomen GJ, Varadi A, Nemet K,
Sarkadi B (2005) Function-dependent conformational changes
of the ABCG2 multidrug transporter modify its interaction
with a monoclonal antibody on the cell surface. J Biol Chem
280:4219–4227
16. Ozvegy-Laczka C, Cserepes J, Elkind NB, Sarkadi B (2005)
Tyrosine kinase inhibitor resistance in cancer: role of ABC multi-
drug transporters. Drug Resist Updat 8:15–26
17. Ozvegy-Laczka C, Hegedus T, Varady G, Ujhelly O, Schuetz
JD, Varadi A, Keri G, Orfi L, Nemet K, Sarkadi B (2004) High-
affinity interaction of tyrosine kinase inhibitors with the ABCG2
multidrug transporter. Mol Pharmacol 65:1485–1495
18. Burger H, van Tol H, Brok M, Wiemer EA, de Bruijn EA,
Guetens G, de Boeck G, Sparreboom A, Verweij J, Nooter K
(2005) 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 4:747–752
19. Yang CH, Huang CJ, Yang CS, Chu YC, Cheng AL, Whang-Peng
J, Yang PC (2005) Gefitinib reverses chemotherapy resistance in
gefitinib-insensitive multidrug resistant cancer cells expressing
ATP-binding cassette family protein. Cancer Res 65:6943–6949
20. Leggas M, Panetta JC, Zhuang Y, Schuetz JD, Johnston B, Bai F,
Sorrentino B, Zhou S, Houghton PJ, Stewart CF (2006) Gefitinib
modulates the function of multiple ATP-binding cassette trans-
porters in vivo. Cancer Res 66:4802–4807
21. Brendel C, Scharenberg C, Dohse M, Robey RW, Bates SE,
Shukla S, Ambudkar SV, Wang Y, Wennemuth G, Burchert A,
Boudriot U, Neubauer A (2007) Imatinib mesylate and nilotinib
835
Cancer Chemother Pharmacol (2013) 72:825–835
1 3
(AMN107) exhibit high-affinity interaction with ABCG2 on
primitive hematopoietic stem cells. Leukemia 21:1267–1275
22. Lemos C, Jansen G, Peters GJ (2008) Drug transporters: recent
advances concerning BCRP and tyrosine kinase inhibitors. Br J
Cancer 98:857–862
23. Keskitalo JE, Pasanen MK, Neuvonen PJ, Niemi M (2009) Dif-
ferent effects of the ABCG2 c.421C>A SNP on the pharmacoki-
netics of fluvastatin, pravastatin and simvastatin. Pharmacog-
enomics 10:1617–1624
24. Lemos C, Giovannetti E, Zucali PA, Assaraf YG, Scheffer GL,
van der Straaten T, D’Incecco A, Falcone A, Guchelaar HJ,
Danesi R, Santoro A, Giaccone G, Tibaldi C, Peters GJ (2011)
Impact of ABCG2 polymorphisms on the clinical outcome and
toxicity of gefitinib in non-small-cell lung cancer patients. Phar-
macogenomics 12:159–170
25. Kim KA, Joo HJ, Park JY (2010) ABCG2 polymorphisms,
34G>A and 421C>A in a Korean population: analysis and a com-
prehensive comparison with other populations. J Clin Pharm Ther
35:705–712
26. Motzer RJ, Bacik J, Schwartz LH, Reuter V, Russo P, Marion S,
Mazumdar M (2004) Prognostic factors for survival in previously
treated patients with metastatic renal cell carcinoma. J Clin Oncol
22:454–463
27. Barrett JC, Fry B, Maller J, Daly MJ (2005) Haploview: analy-
sis and visualization of LD and haplotype maps. Bioinformatics
21:263–265
28. Mizuno T, Terada T, Kamba T, Fukudo M, Katsura T, Nakamura
E, Ogawa O, Inui K (2010) ABCG2 421C>A polymorphism and
high exposure of sunitinib in a patient with renal cell carcinoma.
Ann Oncol 21:1382–1383
29. Houk BE, Bello CL, Kang D, Amantea M (2009) A population
pharmacokinetic meta-analysis of sunitinib malate (SU11248)
and its primary metabolite (SU12662) in healthy volunteers and
oncology patients. Clin Cancer Res 15:2497–2506
30. Shukla S, Robey RW, Bates SE, Ambudkar SV (2009) 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 37:359–365
31. Imai Y, Nakane M, Kage K, Tsukahara S, Ishikawa E, Tsuruo T,
Miki Y, Sugimoto Y (2002) C421A polymorphism in the human
breast cancer resistance protein gene is associated with low
expression of Q141K protein and low-level drug resistance. Mol
Cancer Ther 1:611–616
32. Lemos C, Giovannetti E, Zucali PA, Assaraf YG, Scheffer GL,
van der Straaten T, D’Incecco A, Falcone A, Guchelaar HJ,
Danesi R, Santoro A, Giaccone G, Tibaldi C, Peters GJ (2011)
Impact of ABCG2 polymorphisms on the clinical outcome and
toxicity of gefitinib in non-small-cell lung cancer patients. Phar-
macogenomics 12:159–170
33. Deprimo SE, Bello CL, Smeraglia J, Baum CM, Spinella D,
Rini BI, Michaelson MD, Motzer RJ (2007) Circulating protein
biomarkers of pharmacodynamic activity of sunitinib in patients
with metastatic renal cell carcinoma: modulation of VEGF and
VEGF-related proteins. J Transl Med 5:32
34. Bear HD, Anderson S, Smith RE, Geyer CE Jr, Mamounas
EP, Fisher B, Brown AM, Robidoux A, Margolese R, Kahlen-
berg MS, Paik S, Soran A, Wickerham DL, Wolmark N (2006)
Sequential preoperative or postoperative docetaxel added to pre-
operative doxorubicin plus cyclophosphamide for operable breast
cancer: national Surgical Adjuvant Breast and Bowel Project Pro-
tocol B-27. J Clin Oncol 24:2019–2027
35. Motzer RJ, Bukowski RM, Figlin RA, Hutson TE, Michaelson
MD, Kim ST, Baum CM, Kattan MW (2008) Prognostic nom-
ogram for sunitinib in patients with metastatic renal cell carci-
noma. Cancer 113:1552–1558
36. Porta C, Paglino C, De Amici M, Quaglini S, Sacchi L, Imarisio
I, Canipari C (2010) Predictive value of baseline serum vascu-
lar endothelial growth factor and neutrophil gelatinase-associated
lipocalin in advanced kidney cancer patients receiving sunitinib.
Kidney Int 77:809–815
37. van der Veldt AA, Eechoute K, Gelderblom H, Gietema J, Guche-
laar HJ, van Erp NP, van den Eertwegh AJ, Haanen JB, Mathi-
jssen RH, Wessels JA (2011) Genetic polymorphisms associated
with a prolonged progression-free survival in patients with met-
astatic renal cell cancer treated with sunitinib. Clin Cancer Res
17:620–629
... Typically, a higher frequency of ABCG2 efflux transporter rs2231142 polymorphism was reported in Asian populations [25]. In vitro, the ABCG2 421C>A mutant corresponding to this polymorphism leads to decreased protein expression [26]. In HMEC normal endothelial cells, exposure to subtoxic concentrations of sunitinib increases ABCG2 pump expression, thus favoring sunitinib efflux from the cells [27]. ...
... In HMEC normal endothelial cells, exposure to subtoxic concentrations of sunitinib increases ABCG2 pump expression, thus favoring sunitinib efflux from the cells [27]. Among Korean patients treated with sunitinib, ABCG2 rs2231142 polymorphism was associated with more frequent toxicities [26]. Our meta-analysis comparing 4319 Asian patients with 31,914 other patients enables the high-level recommendation to prefer sunitinib or cabozantinib for Asian patients as these two drugs were associated with a lower prevalence of severe hypertension and hand-foot syndrome. ...
Article
Full-text available
Targeting of angiogenesis has become a major therapeutic approach for the treatment of various advanced cancers. There are many unresolved questions on the toxicity of anti-angiogenic tyrosine kinase inhibitors (TKIs). We performed a meta-analysis to assess the toxicity prevalence of the different anti-angiogenic TKIs among cancer patients and in subpopulations of interest including patients with renal cell carcinoma. We searched the MEDLINE and Cochrane Library databases to November 2023. Clinical trials were eligible if they set out to report the grade ≥3 toxicities related to one of the seven currently approved anti-angiogenic TKIs as monotherapies. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method was applied with PROSPERO (CRD42023411946). The 421 eligible studies included a total of 56,895 cancer patients treated with anti-angiogenic TKI monotherapy. Twenty-four different cancer types were identified, mainly renal cell carcinoma (41.9% of the patients). The anti-angiogenic TKI was sorafenib (34.5% of the patients), sunitinib (30.5%), regorafenib (10.7%), pazopanib (9.4%), cabozantinib (7.7%), axitinib (4.3%), and lenvatinib (2.9%). The pooled prevalence of grade 3 and 4 toxicities was 56.1% (95% confidence interval 53.5–58.6), with marked between-study heterogeneity (I2 = 96.8%). Toxicity profiles varied considerably depending on the type of TKI, the cancer type, and the specific patient characteristics. In particular, Asian patients and elderly people had higher prevalences of severe toxicities, with pazopanib being the best-tolerated drug. For patients treated with sunitinib, particularly those with metastatic RCC, there was no significant difference in terms of toxicity according to the regimen schedule. This meta-analysis highlights the toxicity profiles of anti-angiogenic TKI monotherapies, and thus enables high-level recommendations for the choice of anti-angiogenic TKIs on the basis of the patient’s age, ethnicity, comorbidities, and comedications, for personalized treatment.
... More examples are represented by the neurological effect of FOLFOX therapy that could be predicted by a pharmacogenomic panel performed before therapy, as well as the lethal effect of the DPYD risk genetic variant. In addition, therapy based on cytarabine and its related drug Gemcitabine is affected by several polymorphisms found in the Cytidine Deaminase (CDA) gene [106][107][108] . ...
... The accessibility of more than 20 approved antiretroviral drugs and the opportunity for individual genotype profiling allows the development of regimens that minimize potential DDIs and improve compliance with ART during AC therapy 97,[107][108][109] . Anthracyclines, antimetabolite agents, antitumor antibiotics, and platinum agents undergo non-CYP450 routes of elimination and are unlikely to be altered by ART. ...
Article
Full-text available
The introduction of highly active antiretroviral therapy (ART) has deeply modified the outcome of HIV patients by improving their overall survival and ameliorating their quality of life (QoL). The prolongation of these patients' survival has led to an increased risk of highly diffused non-infectious diseases, e.g., cardiovascular diseases, endocrine disease, neurological diseases, and cancer. The management of antiretroviral therapy and anticancer agents (AC) can be challenging, due to the possible drug-drug interactions (DDI) between AC and ART. For this reason, a multidisciplinary approach is always preferred as demonstrated by the GICAT (Italian Cooperation Group on AIDS and Tumors). This review aims to analyze the current scientific data regarding the possible effects of ART on the management of HIV-positive cancer patients and to evaluate the possible DDIs that must be taken into consideration when co-administrating ART and AC. A collaboration between all the involved professional figures, particularly infectious disease specialists and oncologists, represents the key to the correct managing of these patients in order to guarantee the best oncological outcome possible.
... About 8% of patients have the TKD subtype, which has a controversial but more favorable prognosis [102]. In previous studies, the rs1933437 variant has been reported to be associated with toxicity in treatment with sunitinib [105]. Midostaurin inhibits FLT3 receptor signal transduction and induces cell cycle arrest and apoptosis in leukemic cells expressing both ITD and TKD subtypes. ...
Article
Full-text available
Most malignant hematological diseases are generally a consequence of acquired mutations or rearrangements in cell replication processes. Acute myeloid leukemia (AML) is a clinically and molecularly heterogeneous disease that results from acquired genetic and epigenetic alterations in hematopoietic progenitor cells. Despite the advances made in understanding the pathogenesis of this disease, the overall survival of patients remains very low due to the high relapse rate. Pharmacogenetics and massive sequencing studies have allowed the identification of new recurrent mutations with significant prognostic impact in AML; furthermore, it seems likely that whole genome sequencing will soon become a standard diagnostic test, which will allow the molecular diagnosis of patients. Therefore, it is necessary to develop molecular targets that open new therapeutic perspectives and allow individualized treatment of patients with this aggressive disease. Chronic myeloid leukemia (CML) is the first neoplastic disease for which a characteristic genetic alteration was described. It has, by definition, a genetic marker, the BCR::ABL1 rearrangement, as a consequence of the t9;22(q34;q11) translocation. Its study is essential for the diagnosis of this entity and also for monitoring the response to treatment. Drugs known as tyrosine kinase inhibitors (TKIs) that target the BCR::ABL1 protein (oral targeted therapy) are the conventional treatment of CML, representing a change of paradigm in the management of oncohematological patients.
Article
Full-text available
The clinical application of Pharmacogenomics (PGx) has improved patient safety. However, comprehensive PGx testing has not been widely adopted in clinical practice, and significant opportunities exist to further optimize PGx in cancer care. This systematic review and meta‐analysis aim to evaluate the safety outcomes of reported PGx‐guided strategies (Analysis 1) and identify well‐studied emerging pharmacogenomic variants that predict severe toxicity and symptom burden (Analysis 2) in patients with cancer. We searched MEDLINE, EMBASE, CENTRAL, clinicaltrials.gov , and International Clinical Trials Registry Platform from inception to January 2023 for clinical trials or comparative studies evaluating PGx strategies or unconfirmed pharmacogenomic variants. The primary outcomes were severe adverse events (SAE; ≥ grade 3) or symptom burden with pain and vomiting as defined by trial protocols and assessed by trial investigators. We calculated pooled overall relative risk (RR) and 95% confidence interval (95%CI) using random effects models. PROSPERO, registration number CRD42023421277. Of 6811 records screened, six studies were included for Analysis 1, 55 studies for Analysis 2. Meta‐analysis 1 (five trials, 1892 participants) showed a lower absolute incidence of SAEs with PGx‐guided strategies compared to usual therapy, 16.1% versus 34.0% (RR = 0.72, 95%CI 0.57–0.91, p = 0.006, I ² = 34%). Meta‐analyses 2 identified nine medicine(class)‐variant pairs of interest across the TYMS , ABCB1 , UGT1A1 , HLA‐DRB1 , and OPRM1 genes. Application of PGx significantly reduced rates of SAEs in patients with cancer. Emergent medicine‐variant pairs herald further research into the expansion and optimization of PGx to improve systemic anti‐cancer and supportive care medicine safety and efficacy.
Article
During the past decade, many high-alert medications have been developed and used in clinical practice. Particularly, in the pharmacotherapy of high-alert medications with large individual differences, more attention is needed. To achieve appropriate and individualized pharmacotherapy, there are many issues to be addressed from a clinical pharmacology perspective, such as enhanced monitoring and prior risk identification. This paper is focusing on the therapeutic drug monitoring of molecularly targeted anticancer drugs, and the provision of real-world evidence based on the clinical implementation of pharmacogenetic testing.
Article
Full-text available
Background Pharmacogenomics (PGx), forming the basis of precision medicine, has revolutionized traditional medical practice. Currently, drug responses such as drug efficacy, drug dosage, and drug adverse reactions can be anticipated based on the genetic makeup of the patients. The pharmacogenomic data of Pakistani populations are limited. This study investigates the frequencies of pharmacogenetic variants and their clinical relevance among ethnic groups in Pakistan. Methods The Pharmacogenomics Knowledge Base (PharmGKB) database was used to extract pharmacogenetic variants that are involved in medical conditions with high (1A + 1B) to moderate (2A + 2B) clinical evidence. Subsequently, the allele frequencies of these variants were searched among multiethnic groups of Pakistan (Balochi, Brahui, Burusho, Hazara, Kalash, Pashtun, Punjabi, and Sindhi) using the 1000 Genomes Project (1KGP) and ALlele FREquency Database (ALFRED). Furthermore, the published Pharmacogenomics literature on the Pakistani population was reviewed in PubMed and Google Scholar. Results Our search retrieved (n = 29) pharmacogenetic genes and their (n = 44) variants with high to moderate evidence of clinical association. These pharmacogenetic variants correspond to drug-metabolizing enzymes (n = 22), drug-metabolizing transporters (n = 8), and PGx gene regulators, etc. (n = 14). We found 5 pharmacogenetic variants present at >50% among 8 ethnic groups of Pakistan. These pharmacogenetic variants include CYP2B6 (rs2279345, C; 70%-86%), CYP3A5 (rs776746, C; 64%-88%), FLT3 (rs1933437, T; 54%-74%), CETP (rs1532624, A; 50%-70%), and DPP6 (rs6977820, C; 61%-86%) genes that are involved in drug response for acquired immune deficiency syndrome, transplantation, cancer, heart disease, and mental health therapy, respectively. Conclusions This study highlights the frequency of important clinical pharmacogenetic variants (1A, 1B, 2A, and 2B) among multi-ethnic Pakistani populations. The high prevalence (>50%) of single nucleotide pharmacogenetic variants may contribute to the drug response/diseases outcome. These PGx data could be used as pharmacogenetic markers in the selection of appropriate therapeutic regimens for specific ethnic groups of Pakistan.
Chapter
The ABCG2 half‐transporter plays an important in normal physiology predominantly in protective roles at the maternal‐fetal barrier and the blood–brain barrier and affects oral bioavailability of substrates. High expression in some cancers suggests it may also play a role in multidrug resistance in cancer. Polymorphic variants have been shown to impair trafficking of the protein to the cell surface, with the Q141K variant being associated with a higher incidence of gout. Loss of ABCG2 expression forms the genetic bases of the Jr(a−) blood type. This chapter will focus on more recent ABCG2 studies with particular focus on genetic variants and atomic structures. These more recent findings may lead to a better understanding of the function of ABCG2, potentially leading to the development of better inhibitors or to the development of compounds that are not substrates for the transporter.
Chapter
Interindividual variation in membrane transporter expression and function can lead to changes in pharmacokinetics, pharmacodynamics and drug toxicity. Genetic polymorphisms in membrane transporters are one source of variability in their function. Extensive catalogs of common genetic variation in multiple world populations are publicly available and increased sequencing efforts are driving the identification of rare variation across the human genome. Computational predictions or experimental validation of the consequence of genetic variation in membrane transporters are limited, making it difficult to link with clinical drug response. This chapter focuses on the membrane transporters most studied during drug development and describes functionally significant genetic variation and clinically relevant changes in drug disposition, response or toxicity associated with common genetic variants of these transporters. The contribution of pharmacogenetic research to our understanding of membrane transporter substrate selectivity and their role in pharmacokinetics and drug response is also discussed.
Article
Full-text available
In a phase II, open-label, multicentre Japanese study, sunitinib demonstrated antitumour activity and acceptable tolerability in metastatic renal cell carcinoma patients. Final survival analyses and updated results are reported. Fifty-one Japanese patients with a clear-cell component of metastatic renal cell carcinoma (25 treatment-naïve; 26 cytokine-refractory) received sunitinib 50 mg orally, once daily (Schedule 4/2). Overall and progression-free survivals were estimated by the Kaplan-Meier method. Objective response rate (per Response Evaluation Criteria in Solid Tumours) and safety were assessed with an updated follow-up. First-line and pretreated patients received a median 6.0 and 9.5 treatment cycles, respectively. Investigator-assessed, end-of-study objective response rate was 52.0, 53.8 and 52.9% in first-line, pretreated and overall intent-to-treat populations, respectively. The median progression-free survival was 12.2 and 10.6 months in first-line and pretreated patients, respectively. Fourteen patients per group died (56 and 54%), and the median overall survival was 33.1 and 32.5 months, respectively. The most common treatment-related Grade 3 or 4 adverse events and laboratory abnormalities were fatigue (24%), hand-foot syndrome (18%), decreased platelet count (55%), decreased neutrophil count (53%) and increased lipase (49%). No Grade 5 treatment-related adverse events occurred. Forty patients (78%) required dose reduction, and 13 (25%) discontinued, due to treatment-related adverse events. With the median overall survival benefit exceeding 2.5 years, and acceptable tolerability, in first-line and pretreated Japanese metastatic renal cell carcinoma patients with Eastern Cooperative Oncology Group performance status 0/1, sunitinib showed a favourable risk/benefit profile, similar to Western studies. However, there was a trend towards greater efficacy and more haematological adverse events in Japanese patients.
Article
Background: To conduct a population pharmacokinetic (PopPK) analysis for CP-724714, a novel HER2 tyrosine kinase inhibitor under development for the treatment of advanced HER2 positive cancers.Methods: Concentration data (n=560) from 30 cancer patients receiving daily oral CP-724714 in 21-day cycles in an open label First-in-Human dose-escalation study were used. Cohorts of 250 mg QD, 250 mg BID, 250 mg TID and 400 mg BID were evaluated. Serum levels of CP-724714 were obtained after single dose and at steady state. Mixed effect analysis (FOCE) in NONMEM and bootstrapping were performed. The effect of covariates was assessed. Diagnostic plots, decrease of objective function value (10.83) and predictive check were used as model selection criteria.Results: A 2-compartment 1st-order absorption PK model with interpatient variability on CL/F and Vc/F, and BSA as covariate on CL/F and Vc/F was developed. Results revealed linear PK across the dose range evaluated. CL/F and Vc/F increased with increasing BSA. Interpatient variability of CL/F and Vc/F was reduced by 30% with BSA as a covariate, respectively. Age, gender, ethnicity, and LFTs did not influence CP-724714 disposition.Conclusions: The final PopPK model adequately described CP-724714 PK profiles. BSA was identified as a covariate that reduced PK variability and improved model predictability. Future studies will further assess the importance of BSA as a PK covariate. The proposed PopPK model could be used to simulate CP-724714 Phase 2 trials.
Article
Sunitinib is a tyrosine kinase inhibitor with proven efficacy in renal-cell carcinoma, but some patients do not respond or need dose reductions due to toxicity. Because there are no validated molecular predictors of response or toxicity to sunitinib, we aimed to identify genetic markers predictive of outcome and toxic effects. In our observational, prospective study we enrolled previously untreated adults (≥ 18 years) with clear-cell renal-cell carcinoma at 15 institutions in the Spanish Oncology Genitourinary Group in Spain. Patients received sunitinib according to local practice guidelines. We assessed RECIST response, progression-free survival (PFS), overall survival, and toxicity of sunitinib with 16 key polymorphisms in nine genes: VEGFR2 (rs2305948 and rs1870377), VEGFR3 (rs307826, rs448012, and rs307821), PDGFR-α (rs35597368), VEGF-A (rs2010963, rs699947, and rs1570360), IL8 (rs1126647), CYP3A4 (rs2740574), CYP3A5 (rs776746), ABCB1 (rs1045642, rs1128503, and rs2032582), and ABCB2 (rs2231142). We assessed associations with efficacy and toxicity by use of univariable and multivariable analyses (with clinical factors associated with outcomes as covariates). We adjusted for multiplicity using the Bonferroni method; p values of less than 0·0031 before adjustment were deemed to still be significant after adjustment. We enrolled 101 patients between Oct 10, 2007, and Dec 13, 2010. 95 of these patients were included in toxicity analyses and 89 in the efficacy analyses. Two VEGFR3 missense polymorphisms were associated with reduced PFS with sunitinib on multivariable analysis: rs307826 (hazard ratio [HR] per allele 3·57, 1·75-7·30; p(unadjusted)=0·00049, p(adjusted)=0·0079) and rs307821 (3·31, 1·64-6·68; p(unadjusted)=0·00085, p(adjusted)=0·014). The CYP3A5*1 (rs776746) high metabolising allele was associated in a multivariable analysis with an increased risk of dose reductions due to toxicity (HR per allele 3·75, 1·67-8·41; p(unadjusted)=0·0014, p(adjusted)=0·022). No other SNPs were associated with sunitinib response or toxicity. Polymorphisms in VEGFR3 and CYP3A5*1 might be able to define a subset of patients with renal-cell carcinoma with decreased sunitinib response and tolerability. If confirmed, these results should promote interventional studies testing alternative therapeutic approaches for patients with such variants. Pfizer.
Article
We aimed to describe the efficacy and safety of sunitinib in unselected Korean advanced renal cell carcinoma (RCC) patients. From November 2005 to August 2008, 132 histologically confirmed advanced RCC patients (100 in the global expanded access program) were enrolled. Response and toxicity were assessed regularly according to the protocol. Within this population, 82.6% had clear cell RCC, and 28.8% were treatment naïve. Patients received a median of 5 cycles of sunitinib (range 1-30), and the mean relative dose intensity was 82.0 ± 14.20 (SD). The progression-free survival (PFS) and overall survival rates were 8.2 and 23.1 months, respectively. For the 130 evaluable patients, the objective response rate was 34.1% (n = 45); 44.7% (n = 59) exhibited stable disease. Reasons for discontinuation were disease progression (75.0%) and toxicity (7.6%). The most frequent adverse events were thrombocytopenia (75.0%), neutropenia (70.5%), and anemia (69.7%). Low body surface area (OR = 4.2, 95% CI 1.2-13.8, p = 0.02) and previously treated status (OR = 3.1, 95% CI 1.3-7.4, p = 0.01) were highly predictive of grade 3-4 toxicities. Based on these findings, a nomogram predicting the probability of 12-month PFS was constructed, giving a concordance index of 0.675. Despite the different toxicity profiles, maintaining adequate dose modifications and a careful follow-up enables comparable treatment outcomes for unselected Korean advanced RCC patients.
Article
The current study investigates whether or not functional polymorphisms in the ATP-binding cassette transporter gene ABCG2 might affect gefitinib activity and/or toxicity in non-small-cell lung cancer (NSCLC) patients. Towards this end, ABCG2 polymorphisms and expression were assessed in DNA and tumors from 94 NSCLC patients treated with gefitinib, whereas their associations with toxicity/response and time-to-progression/overall survival were evaluated using Pearson-χ(2) and log-rank-test, respectively. Patients carrying an ABCG2 -15622T/T genotype or harboring at least one TT copy in the ABCG2 (1143C/T, -15622C/T) haplotype developed significantly more grade 2/3 diarrhea (p < 0.01). No associations were found between polymorphisms and outcome. Consistently, ABCG2 protein levels in tumors were not significantly different between patients harboring different ABCG2 variants. The ABCG2 -15622C/T polymorphism and ABCG2 (1143C/T, -15622C/T) haplotype resulted in a gefitinib-dependent, moderate-to-severe diarrhea suggesting that these pharmacogenetic markers should be considered to optimize NSCLC treatment.
Article
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. 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. 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). 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.
Article
ABCG2, also known as Breast Cancer Resistance Peptide (BCRP) or mitoxantrone-resistant protein, is the second member of the G-family of ABC transporters. The frequencies of ABCG2 34G>A and 421C>A polymorphisms in a Korean population were assessed using a newly developed multiplex pyrosequencing method, and compared with the corresponding frequencies seen in other ethnic groups. We designed a multiplex pyrosequencing method to simultaneously detect ABCG2 421C>A and 34G>A polymorphisms and analysed the allele frequencies of these polymorphisms in 250 Korean subjects. The results showed 100% concordance between single and multiplex pyrosequencing methods. We also validated the polymorphisms identified by pyrosequencing with a direct sequencing method using randomly selected samples. The allele frequencies of ABCG2 421C>A and 34G>A in the population tested were 0·298 and 0·190 respectively. The allele frequency of the 421C>A polymorphism is comparable to other Asian populations, including Japanese and Chinese. However, both frequencies are different from those of Caucasians and Africans. The multiplex pyrosequencing method used to detect two ABCG2 polymorphisms concurrently is a rapid and reliable genotyping method for the detection of important ABCG2 genetic polymorphisms. The ABCG2 34G>A and 421C>A polymorphisms are frequently found in the Korean population. The frequencies are similar to those seen in other Asian populations including Japanese and Chinese, but very different to those of Caucasian and African-American populations.
Article
Unlabelled: Kidney cancer accounts for approximately 2% of all cancers worldwide, with renal cell carcinoma being the most common form and this report is focused on renal cell carcinoma. Globally, the incidence and mortality rates are increasing by 2-3% per decade. Kidney cancer is less common in Asia compared with the West. Cigarette smoking, obesity, acquired cystic kidney disease and inherited susceptibility are known risk factors for kidney cancer. The National Comprehensive Cancer Network Guidelines recommend surgical excision as first line of treatment for Stage I, II or III kidney cancer patients and Stage IV patients with resectable tumours. Immunotherapy has a 20-year history in treatment of metastatic kidney cancer. High-dose interleukin-2 (IL-2) is administered in some countries, whereas low-dose IL-2 and interferon-alpha (IFN-α) are popular in Japan. Molecular-targeted drugs, including sunitinib, bevacizumab and sorafenib, are being used for previously untreated and refractory patients. Asian and non-Asian populations have shown large differences in the incidences of adverse events with sorafenib and sunitinib. Consensus statement: Kidney cancer is relatively uncommon in Asia compared with the West, but its incidence is increasing in more developed Asian nations. Guidelines from the National Comprehensive Cancer Network , etc., for treating metastatic renal cell carcinoma are based on Phase III clinical trials conducted primarily in Western patients. Targeted therapies are now becoming primary recommendations, but efficacy/toxicity data from Asian patients are lacking. Some drugs cause adverse effects in Asians because their recommended dosages are optimal for Caucasians but may be too high for Asians. Further research is necessary to develop optimal treatment strategies for Asians.