ArticlePDF Available

Association Between CYP3A4 and CYP3A5 Genotypes and Cyclosporine's Blood Level and Doses Among Jordanian Kidney Transplanted Patients

Authors:

Abstract and Figures

BACKGROUND Cyclosporine is used as an immunosuppressive agent in kidney transplantation. It has a narrow therapeutic window. Cyclosporine is predominantly metabolized by CYP3A4 and CYP3A5. The most common single nucleotide polymorphisms (SNPs) affecting cyclosporine metabolism (CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3) were investigated among Jordanian kidney transplanted patients to find out the genotypes and allele frequencies of these SNPs. Additionally, this study investigated whether genotypes of CYP3A4 and CYP3A5 affects C2 blood levels, dosing of cyclosporine and the prevalence of acute rejection. METHODS Blood samples of 109 adult patients taking cyclosporine as their primary immunosuppressant for kidney transplantation were collected from Prince Hamzah Hospital, Amman, Jordan. Patients’ first C2 blood levels and their first two given doses were collected. Patients were genotyped for the four SNPs using Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) assay method. RESULTS Allele frequencies among Jordanian patients for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3 were 0.037, 0.399, 0.037 and 0.271, respectively. There was a significant association between CYP3A4*22 and mean difference of the second and first given doses (P=0.034). There was a big difference between CYP3A4*22 and the mean of the first C2 blood levels (P=0.063). CONCLUSION There was a strong association between CYP3A4*22 and the mean difference of the second and first given doses. There was a trend of significant difference between the mean of the first C2 blood levels among heterozygous CYP3A4*22 patients. Pharmacogenomics may hold promise in assisting the prediction of the best cyclosporine dose and C2 blood level among Jordanian kidney transplanted patients.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.net
682 Current Drug Metabolism, 2019, 20, 682-694
RESEARCH ARTICLE
! 1389-2002/19 $58.00+.00 © 2019 Bentham Science Publishers!
Association Between CYP3A4 and CYP3A5 Genotypes and Cyclosporine's Blood
Levels and Doses among Jordanian Kidney Transplanted Patients
Sahar El-Shair1,*, Mohammad Al Shhab1, Khaled Zayed2, Moaath Alsmady1 and Malek Zihlif1
1Department of Pharmacology, The University of Jordan, Amman, Jordan 2Department of Nephrology and Kidney Transplantation,
Prince Hamzah Hospital, Amman, Jordan
A R T I C L E H I S T O R Y
Received: June 06, 2019
Revised: July 16, 2019
Accepted: July 25, 2019
DOI:
10.2174/1389200220666190806141825
Abstract: Background: Cyclosporine is used as an immunosuppressive agent in kidney transplantation. It has a
narrow therapeutic window. Cyclosporine is predominantly metabolized by CYP3A4 and CYP3A5. The most com-
mon Single Nucleotide Polymorphisms (SNPs) affecting cyclosporine metabolism (CYP3A4*1B, CYP3A4*1G,
CYP3A4*22 and CYP3A5*3) were investigated among Jordanian kidney transplanted patients to find out the geno-
types and allele frequencies of these SNPs. Additionally, this study investigated whether genotypes of CYP3A4 and
CYP3A5 affect C2 blood levels, dosing of cyclosporine and the prevalence of acute rejection.
Methods: Blood samples of 109 adult patients taking cyclosporine as their primary immunosuppressant for kidney
transplantation were collected from the Prince Hamzah Hospital, Amman, Jordan. Patients’ first C2 blood levels and
their first two given doses were collected. Patients were genotyped for the four SNPs using Polymerase Chain Reac-
tion-restriction Fragment Length Polymorphism (PCR-RFLP) assay method.
Results: Allele frequencies among Jordanian patients for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3
were 0.037, 0.399, 0.037 and 0.271, respectively. There was a significant association between CYP3A4*22 and mean
difference in the second and first given doses (P=0.034). There was a big difference between CYP3A4*22 and the
mean of the first C2 blood levels (P=0.063).
Conclusion: There was a strong association between CYP3A4*22 and the mean difference between the second and
first given doses. There was a trend of significant difference between the mean of the first C2 blood levels among
heterozygous CYP3A4*22 patients. Pharmacogenomics may hold promise in assisting the prediction of the best
cyclosporine dose and C2 blood level among Jordanian kidney transplant patients.
Keywords: Cyclosporine, kidney transplantation, CYP3A4*1B, CYP3A4*1G, CYP3A4*22, CYP3A5*3, Jordanian.
1. INTRODUCTION
Kidney transplantation is the optimal modality of Renal Re-
placement Therapy (RRT) for patients with end-stage renal disease
[1]. Much of the success of kidney transplantation seen nowadays is
due to the use of immunosuppressive medications [2, 3]. Immuno-
suppressive agents hav e a narrow therapeutic window and are asso-
ciated with great inter- and intra-individual response variations [4-
6]. Subtherapeutic blood concentrations are associated with an in-
creased risk of acute rejection. Whereas, overdosing is associated
with an increased risk of adverse events [7].
Cyclosporine (CsA) is one of the cornerstone immunosuppres-
sive agents used in kidney transplantation along with steroids and
antiproliferative drugs [3]. Many factors affect CsA metabolism
such as diet, age, drugs, genetics and others [8, 9]. Pharmacokinet-
ics (PK) and Pharmacodynamics (PD) are difficult to predict in all
transplanted patients [1]. The best clinical practice involves the use
of standard dosing based on body weight and Therapeutic Drug
Monitoring (TDM) by measuring cyclosporine blood levels [1, 9].
However, TDM cannot reflect drug concentration at its site of ac-
tion nor guarantee optimal organ suppression [1]. Some patients
may even develop acute rejection or drug-related adverse events
despite having the drug within the therapeutic window [10, 11].
Genetics was estimated to account for 20-95% of variability in drug
disposition and effect [5, 12-14]. Hence, the emergence of Pharma-
cogenomics (PG) science aims to aid and guide, along with TDM,
future prescription of immunosuppressants [1].
*Address correspondence to this author at the Department of Pharmacology,
University of Jordan, Amman, Jordan. Tel: +962795321521;
E-mail: pharm-d.sahar.elshair@hotmail.com
CsA is extensively metabolized by the isoenzyme CYP3A4 as it
is responsible for about 80% of CsA metabolism [15]. There are
more than 30 Single Nucleotide Polymorphism (SNPs) identified in
the CYP3A4 gene [14, 16]. The most frequent allele known to af-
fect cyclosporine is CYP3A4*1B. Other alleles may include
CYP3A4*22, CYP3A4*1G, CYP3A4*3, CYP3A4 rs 4646437C˃T
and 2 pseudogenes [17-19]. Table 1 [20-22] and Fig. (1) summarize
major CYP3A4 alleles.
CYP3A4*1B is an A- to G- mutation in the 5’ regulatory region
at 290 locus of CYP3A4 (A290G, rs2740574) [23, 24]. It was re-
ported with controversial assumptions regarding its mRNA tran-
scription and enzymatic activity [17, 25-27]. Some support the as-
sumption of an impaired or decreased CYP3A4 enzymatic activity
[23, 28, 29]. Others support the opposite assumption of increased
enzymatic activity and increased rate of drug metabolism as men-
tioned by Barbarino et al. (2013) and Crettol et al. (2008) [17, 18].
Whereas, others presume its in-vitro and in-vivo studies that do not
show any association between genotypes and phenotypes at all [26].
CYP3A4*1G is a highly frequent allele in intron 10 of the
CYP3A4 gene (rs2242480) involving a -G to -A substitution at posi-
tion 82266 [30]. This SNP is also associated with controversial as-
sumptions where some authors presume it is associated with reduced
enzymatic activity [31, 32]. Therefore, patients carrying the mutant
allele have higher CsA levels and consequently require lower doses.
While, others presume its association with a higher enzymatic activity
[25, 30, 33] and the resulting lower CsA levels which thus require
higher doses than patients possessing the wild allele.
CYP3A4*22 is a newly identified SNP in intron 6 found on
CYP3A4 gene that results from a substitution of C to T allele (522-
Association Between CYP3A4 and CYP3A5 Genotypes Current Drug Metabolism, 2019, Vol. 20, No. 8 683
191C>T; rs35599367) [34]. Some reported its association with
decreased CYP3A4 mRNA hepatic levels [18, 35, 36]. Thus, the T-
variant allele was correlated with a decreased CYP3A4 enzymatic
activity [34] when compared to both CT and CC genotypes.
CYP3A5 is considered the second most important CYP3A
isoenzyme in the human liver and intestine [9, 14]. CYP3A5 has a
similar protein sequence to CYP3A4 and similar substrate specific-
ity [1]. It may represent up to 50% of the total hepatic CYP3A con-
tent in people expressing it [17, 37]. Though CsA is oxidized by
CYP3A4, some of its metabolites are formed by CYP3A5, therefore
having a significant impact on CsA PK [36, 38]. There are at least 7
variant alleles of CYP3A5 identified to date. Fig. (2) summarizes
the major CYP3A5 alleles.
The expression of CYP3A5 is affected by a splicing mutation
(A6986G, rs776746) within intron 3 [5, 16, 37] o f the mentioned
gene. The replacement of nucleotide A (CYP3A5*1) by nucleotide G
(CYP3A5*3) at locus 6986 creates an mRNA splice defect with a
premature stop codon resulting in an inactive truncated protein [5, 16,
36]. Thus, CYP3A5*3 variant is a loss-of-function allele [16, 37].
The aim of this study was to examine the genotypes and allele
frequencies of CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and
CYP3A5*3 polymorphisms among Jordanian kidney transplanted
patients. Additionally, this study investigated any association be-
tween genotypes of CYP3A4 and CYP3A5 and all of the C2 blood
levels, dosing of CsA and the prevalence of acute rejection.
2. METHODS AND MATERIALS
A total of 160 kidney transplant patients who attended Neph-
rology and Kidney Transplant Clinics at Prince Hamzah Hospital,
Jordan, were interviewed after obtaining their consent. The study’s
informed consent and case report forms were both approved by the
Institutional Review Board of Prince Hamzah Hospital.
Patients were assigned and identified with a random number.
Around 3-5 ml venous whole blood samples were drawn and col-
lected in Ethylene Diamine Tetra-acetic Acid (EDTA) tubes identi-
fied with a random number for each patient. Tubes were stored at
4°C until analysis. The genomic DNA of each patient was exam-
ined for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3
polymorphisms. A total of 51 patients were excluded from the
study.
2.1. Inclusion Criteria
Jordanian adult (18 years old and above) patients, including
both female and male population, who underwent kidney transplant
during 2009-2017 and who were prescribed cyclosporine as their
primary immunosuppressant agent.
Table 1. Major CYP3A4 alleles [20-22].
Allele
Nucleotide Change
Protein Effect
CYP3A4*1B (rs2740574)
-392A<G
Promoter
CYP3A4*1G (rs2242480)
20230G>A
Intronic
CYP3A4*2 (rs55785340)
15713T>C
Ser222Pro
CYP3A4*3 (rs4986910)
23171T>C
Met445Thr
CYP3A4*8 (rs72552799)
13908G>A
Arg130Gln
CYP3A4*12 (rs12721629)
21896C>T
Leu373Phe
CYP3A4*13 (rs4986909)
22026C>T
Pro416Leu
CYP3A4*15A (rs4986907)
14269G>A
Arg162Gln
CYP3A4*17 (rs4987161)
15615T>C
Phe189Ser
CYP3A4*18 (rs28371759)
20070T>C
Leu293Pro
CYP3A4*22 (rs35599367)
15389C>T
Intronic
Fig. (1). Different CYP3A4 alleles [21].
684 Current Drug Metabolism, 2019, Vol. 20, No. 8 El-Shair et al.
2.2. Exclusion Criteria
Any patient younger than 18 years, any patient prescribed tac-
rolimus as their primary immunosuppressive drug and any patient
prescribed cyclosporine for reasons other than kidney transplant.
2.3. Genome DNA Extraction
DNA was extracted from lymphocytes of peripheral whole
blood using DNA extraction kits (Wizard Genomic DNA Purifica-
tion Kit, Promega, USA) according to the manufacturer’s protocol.
2.4. Genotyping
In order to study the different genotypes o f the patients, a spe-
cific sequence containing the SNP of the gene of interest was am-
plified by Polymerase Chain Reaction (PCR), then through Restric-
tion Fragment Length Polymorphism-Polymerase Chain Reaction
(RFLPs-PCR) process. Specific restriction enzymes were used to
digest normal or mutant alleles at specific sites.
All primers were designed by Primer 3’- according to the Rs-
number of the SNP as shown in Table 2. PCR was performed using
PCR-Thermocycler (Bio-Rad, USA) as described in Table 3. The
resultant amplified sequences were confirmed on agarose gel elec-
trophoresis and visualized using Red Safe. The PCR products were
digested by specific restriction enzyme and RFLP mixture was
prepared as shown in Table 4. Digested products were mixed with
5µl loading dye and confirmed on agarose gel electrophoresis.
Genotypes and phenotypes were recorded according to the size of
the resulted fragments as described in Table 4.
2.5. Statistical Analysis
2.5.1. Prediction of Allele and Genotype Frequencies
Allele and genotype frequencies of 109 patients were estimated.
In addition, the Hardy-Weinberg Equilibrium (HWE) was used.
One for Chi-square value and the other for P-value of the Chi-
Square test. If the P value of the chi-square test is < 0.05, it is then
not consistent with HWE.
2.5.2. Statistical Tests
Normally distributed continuous variables such as age, weight,
C2 levels and doses given were all analyzed using ANOVA test.
Pearson Chi-square test was used to test discrete variables of inde-
pendence to determine the association between all different SNPs
(CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3) and
gender, the prevalence of adverse events and prevalence of acute
rejection. A P-value of 0.05 or less was considered significant. All
the statistical analyses were performed using Statistical Package for
Social Sciences (SPSS Inc., Chicago, Illinois) version 23.0.
2.5.3. Comparison of Allele Frequencies Between Jordanian
Population and other Ethnic Groups
In order to compare allele frequencies between the Jordanian
population and other ethnic populations, the statistical z-test was
Fig. (2). Different CYP3A5 alleles [27].
Arrow heads indicate mutated locations that result in amino acid changes, premature stop codon and alternative splicing.
CYP: Cytochrome P450; INS: Insertion; ORF: Open reading frame; SV: Splicing variant; UTR: Untranslated regions.
Table 2. Primers sequences and expected product size for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3 PCR amplifica-
tion.
SNP
Primers
Sequence (5’ 3’)
Rs Number
PCR Product S ize
CYP3A4*1B - F
ATCTGTGTGAGGAGTTTGGT
CYP3A4*1B
CYP3A4*1B - R
GTGAGGCTGTTGGATTGTTT
2740574
470
CYP3A4*1G - F
TCAGAGCCTTCCTACATAGAG
CYP3A4*1G
CYP3A4*1G - R
GTCTTTCCTCTCCTTTCAGC
2242480
315
CYP3A4*22 - F
ACTGTAGTTGTCTGATAGTGGG
CYP3A4*22
CYP3A4*22 - R
CGCTGAGTGGAGAAAGATGT
35599367
290
CYP3A5*3 - F
AAGTCCTCAGAATCCACAGCG
CYP3A5*3
CYP3A5*3 R
CACACAGCAAGAGTCTCACAC
776746
416
*Primer sequences are designed by Primer 3’- according to rs-number and tested usin g NCBI website.
Association Between CYP3A4 and CYP3A5 Genotypes Current Drug Metabolism, 2019, Vol. 20, No. 8 685
used at 5% level of significance. If z-value is less than 1.96,
then the two proportions of allele frequencies are significantly
similar. On the other hand, if z-value is greater than 1.96, then
allele frequencies are significantly different between the two
groups.
3. RESULTS
3.1. Study Population Characteristics
This study included 109 kidney transplant recipients who vis-
ited the Nephrology and Kidney Transplant Clinics at Prince
Hamzah Hospital periodically and who underwent kidney transplant
from 2006 until early 2017. Patients who were taking cyclosporine
(CsA) drug as their primary immunosuppressant along with predni-
solone and mycophenolic acid as maintenance anti-rejection ther-
apy were included. Their data including C2 blood levels, first two
given doses, the prevalence of drug-related adverse events and
prevalence of acute rejection were reviewed.
Out of the 109 kidney transplant patients, 65% were male and
35% were female between the ages of 18-73 years. The demo-
graphic data included in this study (Table 5) are gender, age,
weight, year of transplant, donor type, Drug-drug Interactions
(DDI) found and the most common reported adverse events. The
most commonly reported drug-related adverse events among our
patient groups were gingival hyperplasia and hirsutism as shown in
Table 5.
All amplified sequences of all alleles tested were confirmed on
1.5% agarose gel electrophoresis and visualized using Red Safe.
The PCR-RFLP analysis of all SNPs was performed on all the 109
subjects. The PCR product was digested by MboII, RsaI, DraIII and
SspI enzymes for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and
CYP3A5*3, respectively. They were visualized using 3.5 % aga-
rose gel after being stained by Ethidium Bromide. Summary of
gene and allele frequencies with their consistency with HWE is
shown in Table 6.
3.2. C2 Levels and Polymorphisms
The first C2 level was taken for each patient. The association
between the four SNPs and the mean for the first C2 levels are
shown in Table 7. The four polymorphisms showed no significant
association with the mean of the first C2 levels.
Table 3. PCR conditions used and agarose gel confirmations for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3 genotypes.
PCR Conditions
SNP
Initial
Denaturation
Denaturation
Annealing
Extension
Number of
Cycles
Final
Extension
Confirmed on
Agarose Gel (%)
CYP3A4*1B
54 for 30 seconds
36 cycle
1.5
CYP3A4*1G
52 for 30 seconds
36 cycle
1.5
CYP3A4*22
51 for 30 seconds
36 cycle
2.5
CYP3A5*3
95ºC for 5
minutes
95ºC for 30
seconds
55.4 for 30 seconds
72ºC for 1
minute
39 cycle
72ºC for 7 min
1.5
Table 4. Genotypes and phenotypes of digested fragments for CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3.
SNP
Restriction
Enzyme
Alleles
Fragment Length
Genotypes
Phenotypes
References
AA
69, 104, 297 bp
Homozygous wild
Extensive metabolizers
AG
69, 104, 297, 470 bp
Heterozygous
Intermediate metabolizers
CYP3A4*1B
MboII
GG
470 bp
Homozygous mutant
Poor metabolizers
[23, 28]
GG
192, 123 bp
Homozygous wild
Extensive metabolizers
GA
315, 192, 123 bp
Heterozygous
Intermediate metabolizers
CYP3A4*1G
RsaI
AA
315 bp
Homozygous mutant
Poor metabolizers
[31, 32]
CC
290 bp
Homozygous wild
Extensive metabolizers
CT
97, 193, 290 bp
Heterozygous
Intermediate metabolizers
CYP3A4*22
DraIII
TT
97, 193 bp
Homozygous mutant
Poor metabolizers
[18, 35]
AA
190 + 226 bp
Homozygous wild
Extensive metabolizers
AG
190 + 226 + 416 bp
Heterozygous
Intermediate metabolizers
CYP3A5*3
SspI
GG
416 bp
Homozygous mutant
Non-functioning
[16, 39]
BP: Base pair.
686 Current Drug Metabolism, 2019, Vol. 20, No. 8 El-Shair et al.
Table 5. Demographic data of our study group.
Demographic Data
Variables
No.
%
Mean
Males
71
65
Gender
Females
38
35
-
Total
109
100
-
18-25
19
17
26-35
31
29
36-45
26
24
46-55
21
19
56-65
11
10
Age
66-75 or more
1
1
38.56
Total
109
100
-
35
0
0
36-45
10
9
46-55
15
14
56-65
19
17
66-75
25
23
76-85
17
16
86-95
17
16
96-105
6
6
Weight
106
0
0
69.72
Total
109
100
-
2010
2
2
2011
8
7
2012
10
9
2013
25
23
2014
24
22
2015
25
23
Year of Transplantation
2016
15
14
-
Total
109
100
-
LR
101
93
Donor Type
LNR
8
7
-
Total
109
100
-
No DDI
80
73
With CYP-450 Inhibitors
27
25
DDI
With CYP-450 Inducers
2
2
-
Total
109
100
-
Table (5) contd….
Association Between CYP3A4 and CYP3A5 Genotypes Current Drug Metabolism, 2019, Vol. 20, No. 8 687
Adverse Events
Reported AE
No AE
Gingival
Hyperplasia
Hirsutism
Increased
KFT
Increased LFT
Rash
Total
AA
83
(82%)
11
(11%)
5
(5%)
0
1
(1%)
1
(1%)
101
AG
5
(83%)
0
0
1
(17%)
0
0
6
CYP3A4*1B
GG
0
0
0
0
1
(100%)
0
1
Total
88
11
5
1
2
1
108
GG
29
(85%)
4
(12%)
0
0
0
1
(3%)
34
GA
48
(77%)
7
(11%)
4
(6%)
1
(2%)
2
(3%)
0
62
CYP3A4*1G
AA
11
(92%)
0
1
(8%)
0
0
0
12
Total
88
11
5
1
2
1
108
CYP3A4*22
CC
82
(82%)
10
(10%)
5
(5%)
1
(1%)
1
(1%)
1
(1%)
100
CT
6
(75%)
1
(12.5%)
0
0
1
(12.5%)
0
8
TT
0
0
0
0
0
0
0
Total
88
11
5
1
2
1
108
CYP3A5*3
AA
63
(83%)
7
(9%)
3
(4%)
1
(1.32%)
1
(1.32%)
1
(1.32%)
76
AG
3
(60%)
1
(20%)
1
(20%)
0
0
0
5
GG
22
(81%)
3
(11%)
1
(4%)
0
1
(4%)
0
27
Total
88
11
5
1
2
1
108
CYP-450: Cytochrome P-450.
DDI: Drug-Drug Interaction.
AE: Adverse Events.
Table 6. Gene and allele frequencies of CYP3A4*1B, CYP3A4*1G, CYP3A4*22 and CYP3A5*3 polymorphisms among Jordanian
patients and their consistency with Hardy-Weinberg Equilibriu m.
HWE
SNP
Number of Patients
Frequency
Chi Sq Value
P-Value of Chi-Sq
CYP3A4*1B
AA (wild)
102/109
0.94
AG
6/109
0.06
Gene Frequency
GG (mutant)
1/109
0.01
A allele
210/218
0.963
Allele Frequency
G allele
8/218
0.037
5.344
0.021
Table (6) contd….
688 Current Drug Metabolism, 2019, Vol. 20, No. 8 El-Shair et al.
HWE
SNP
Number of Patients
Frequency
Chi Sq Value
P-Value of Chi-Sq
CYP3A4*1G
GG (wild)
34/109
0.31
GA
63/109
0.58
Gene Frequency
AA (mutant)
12/109
0.11
G allele
131/218
0.601
Allele Frequency
A allele
87/218
0.399
4.583
0.032
CYP3A4*22
CC (wild)
101/109
0.93
CT
8/109
0.07
Gene Frequency
TT (mutant)
0/109
0
C allele
210/218
0.963
Allele Frequency
T allele
8/218
0.037
0.158
0.691
CYP3A5*3
AA (wild)
77/109
0.71
AG
5/109
0.05
Gene Frequency
GG (mutant)
27/109
0.25
A allele
159/218
0.729
Allele Frequency
G allele
59/218
0.271
85.142
0.000
Table 7. Associations between 1st C2 level and SNPs among Jordanian patients taking cyclosporine.
1st Level
N
Mean
SD
St.Err
F
Sig
AA
102
(93%)
1253.89
579.49
57.38
AG
6
(6%)
1274.83
631.45
257.79
CYP3A4*1B
GG
1
(1%)
1600.00
-
-
Total
109
1258.22
577.59
55.32
0.178
0.837
GG
34
(31%)
1281.35
653.58
112.09
GA
63
(58%)
1274.98
551.85
69.53
CYP3A4*1G
AA
12
(11%)
1104.67
498.51
143.91
Total
109
1258.22
577.59
55.32
0.473
0.624
CC
101
(93%)
1287.10
573.21
57.04
CT
8
(7%)
893.63
537.90
190.18
CYP3A4*22
TT
0
0
0
0
Total
109
1258.22
577.59
55.32
3.520
0.063
AA
77
(71%)
1268.58
578.56
65.93
AG
5
(4%)
1342.20
366.65
163.97
CYP3A5*3
GG
27
(25%)
1213.11
618.76
119.08
Total
109
1258.22
577.59
55.32
0.145
0.865
SD: Standard Deviation.
St.Err: Standard Error.
Association Between CYP3A4 and CYP3A5 Genotypes Current Drug Metabolism, 2019, Vol. 20, No. 8 689
Table 8. Associations between differences between 2nd and 1st given doses and SNPs among Jordanian patients taking cyclosporine.
2nd Given Dose
N
Mean
SD
St.Err
F
Sig
AA
102
(93%)
0.98
33.78
3.34
AG
6
(6%)
4.17
33.23
13.57
CYP3A4*1B
GG
1
(1%)
-25.00
-
-
Total
109
0.92
33.537
3.21
0.323
0.725
GG
34
(31%)
1.84
30.793
5.28
GA
63
(58%)
-1.59
35.89
4.52
CYP3A4*1G
AA
12
(11%)
11.46
27.93
8.06
Total
109
0.92
33.54
3.21
0.778
0.462
CC
101
(93%)
-0.99
32.44
3.23
CT
8
(7%)
25.00
40.09
14.17
CYP3A4*22
TT
0
0
0
0
Total
109
0.92
33.54
3.21
4.600
0.034
AA
77
(71%)
0.65
36.10
4.11
AG
5
(4%)
-15.00
13.69
6.12
CYP3A5*3
GG
27
(25%)
4.63
27.77
5.34
Total
109
0.92
33.54
3.21
0.727
0.486
SD: Standard Deviation
St.Err: Standard Error
3.3. Differences between Given Dose and Polymorphisms
The difference between the second and first given doses was
calculated for each patient. The association between the four SNPs
and the mean difference between the second and first given doses
are shown in Table 8. A statistically significant association was
only observed between CYP3A4*22 and the mean difference be-
tween the second and first given doses with a P -value of 0.034. The
rest of the polymorphisms showed no associations.
3.4. Prevalence of Acute Rejection and Polymorphisms
Prevalence of acute rejection was recorded for each patient. The
association between the prev alence of acute rejection and the four
SNPs is shown in Table 9. No significant association was observed
with any of the four SNPs and prevalence of acute rejection in the
study group.
3.5. Comparison of Allele Frequencies between Jordanian
Population and other Ethnic Groups
The allele frequencies among Jordanian population compared to
other ethnic groups are shown in Table 10.
4. DISCUSSION
CsA is extensively metabolized in the liver and small intestine
by both CYP3A4 and CYP3A5. Both the cytochromes are consid-
ered major reflectors of overall CYP3A activity [9, 50]. The most
common SNPs affecting cyclosporine’s PK include CYP3A4*1B,
CYP3A4*1G, CYP3A4*22 and CYP3A5*3.
The first SNP affecting CsA, CYP3A4*1B, is highly variable
among different racial populations. As Jordan population is consid-
ered, a mixed Caucasian population, this study found that the minor
allele frequency of CYP3A4*1B (G allele) is 0.037, which varies
from previous studies among Jordanians (P=0.107) as reported by
Yousef et al. (2016) [40], but it was consistent with another study
among Caucasian populations (P=0.000) [44]. Cattaneo et al.
(2004) also reported it to be around 4% among Caucasians and
66.7% among the black population [29]. Moreover, van Schaik
(2008) reported this SNP to be more common among African-
Americans when compared to Caucasians. As the case among Cau-
casians, this varian t was also considered rare among Asians [24].
The results of this study did not show any association between
CYP3A4*1B and any CsA PK parameters. Such results are similar
to the results observed by Rivory et al. (2001) [23] and Ashavaid et
al. (2010) [52].
Regarding CYP3A4*1G, which has not been studied among
Jordanian population, the minor allele frequency was 0.399. This
significantly differs from the frequencies among Caucasians
(P=0.000) [46] and Chinese (P=0.025) [47] populations. Regarding
this SNP and its effect on CsA PK parameters, there was no asso-
ciation with any significant effect on PK parameters. Such results
are in agreement with a study conducted at the early stage
690 Current Drug Metabolism, 2019, Vol. 20, No. 8 El-Shair et al.
Table 9. Associations between the prevalence of acute rejection and SNPs among Jordanian patients taking cyclosporine.
Incidence of Acute Rejection
No
Yes
Total
Pearson Chi Square
AA
74
(73%)
27
(27%)
101
AG
2
(33%)
4
(67%)
6
CYP3A4*1B
GG
1
(100%)
0
1
Total
77
31
108
0.090
GG
24
(73%)
9
(27%)
33
GA
42
(67%)
21
(33%)
63
CYP3A4*1G
AA
11
(92%)
1
(8%)
12
Total
77
31
108
0.210
CC
72
(72%)
28
(28%)
100
CT
5
(63%)
3
(37%)
8
CYP3A4*22
TT
0
0
0
Total
77
31
108
0.415
AA
57
(75%)
19
(25%)
76
AG
3
(60%)
2
(40%)
5
CYP3A5*3
GG
17
(63%)
10
(37%)
27
Total
77
31
108
0.419
Table 10. Comparisons of allele frequencies between Jordanian transplantation population and other ethnic transplantation groups
for the four polymorphisms.
Ethnic
References
Sample Size
Allele Frequency
Z-Score
P-Value
CYP3A4*1B
A Allele
G Allele
Jordanians
Present Study
109
0.963
0.037
---
---
Jordanians
[40]
137
0.912
0.088
1.61
0.107
Turkish
[41]
186
0.986
0.014
1.29
0.197
Spanish
[42]
177
0.96
0.04
0.13
0.897
British
[41]
200
0.935
0.065
1.03
0.303
Caucasians
[43]
135
0.93
0.07
1.12
0.263
Caucasians
[44]
241
0.689
0.311
5.68
0
Asians
[45]
151
0.42
0.58
9.04
0
African Americans
[45]
151
0.42
0.58
9.04
0
Table (10) contd….
Association Between CYP3A4 and CYP3A5 Genotypes Current Drug Metabolism, 2019, Vol. 20, No. 8 691
Ethnic
References
Sample Size
Allele Frequency
Z-Score
P-Value
CYP3A4*1B
A Allele
G Allele
CYP3A4*1G
G Allele
A Allele
Jordanians
Present Study
109
0.601
0.399
---
---
European Americans
[46]
48
0.917
0.083
3.97
0
Chinese
[5]
126
0.738
0.262
2.24
0.025
Chinese
[47]
91
0.75
0.25
2.23
0.026
African Americans
[46]
48
0.781
0.219
2.19
0.029
CYP3A4*22
C Allele
T Allele
Jordanians
Present Study
109
0.963
0.037
---
---
Caucasians
[36]
177
0.97
0.03
0.32
0.749
Caucasians
[44]
241
0.71
0.29
5.37
0
CYP3A5*3
A Allele
G Allele
Jordanians
Present Study
109
0.729
0.271
---
---
Jordanians
[40]
137
0.120
0.880
9.73
0.000
Caucasians
[44]
241
0.963
0.037
6.48
0.000
Caucasians
[48]
95
0.906
0.094
3.22
0.001
Koreans
[46]
92
0.745
0.255
0.26
0.795
Koreans
[49]
194
27.6
0.724
7.63
0.0000
African Americans
[46]
48
0.802
0.198
0.97
0.332
post-transplantation on 126 renal transplant patients who revealed
that the wild CYP3A4*1 allele and mutant CYP3A4*1G carriers do
not differ significantly in cyclosporine’s PK [5]. In addition, Zhang
et al. (2013) did not observe any significant influence of this SNP
on cyclosporine C0/Dose among 101 Chinese renal transplant pa-
tients [53].
Findings in this study differ from the study by Hu et al. (2007)
on 26 Chinese healthy subjects who showed a significantly higher
clearance rate in patients having the mutant allele, 50% reduction in
C2 concentration, a lower mean maximum plasma concentration
(Cmax) and a lower mean AUC0-4 [54]. Similarly, Qiu et al.
(2008) also observed in their study that homozygous wild patients
had a 40% higher dose-adjusted trough levels (C0/dose) on days
16-30 post-transplantation when compared to homozygous
CYP3A4*1G mutant patients and C2-post-dose-adjusted level
(C2/dose) for homozygous wild CYP3A4*1 was 35% higher on
days 16-30 and 19% higher on days 8-15 when compared to homo-
zygous mutant patients [55].
The second SNP that was also not investigated in Jordan before
is the CYP3A4*22. Our results revealed the minor allele frequency
to be 0.037, which is significantly different from the frequency
reported by Lunde et al. (2014) (P=0.749) among Caucasians [36].
Interestingly, in this study, there was a trend of significant dif-
ference between the mean of the first C2 blood levels among homo-
zygous wild patients and C2 blood levels among heterozygous
CYP3A4*22 patients (P= 0.063). Elens et al. (2011) also reported a
significant association between CYP3A4*22 and CsA trough levels
C0 (P=0.033) [56].
Additionally, a significant association was found between
CYP3A4*22 polymorphism and the mean difference between the
second and the first given dose (P=0.034). In order to eliminate the
confounder’s effect of the patient’s body weight, the difference
between the second and first doses was calculated.
Limited studies are available regarding the effect of CYP3A4*22
on either CsA’s PK parameters or the difference between the given
doses. Many studies concentrated on the effect of genotypes on actual
given doses, which was thought to be highly influenced by body
weight, were excluded from this study. This was shown by von Ah-
sen et al. (2001), who failed to show any significant association be-
tween CYP3A4*1B genotypes and given doses when a study was
conducted on 124 renal transplant patients [23].
In the case of CYP3A5*3, the minor allele (G allele) frequency
in our study, was found to be 0.271. It had a P-value of 0.000 when
compared to previous studies among Jordanians [40], and a P-value
of 0.001 when compared to Caucasians in a different study with an
allele frequency of 0.094 [48]. This variation in minor allele fre-
quency has also been observed among Koreans (P=0.795 vs. P=0,
respectively) [46, 49] which could have been attributed to different
patient sample selection criteria among different studies of different
ethnic groups.
The results of this study did not show any significant correla-
tion of this allele on CsA’s PK parameters which differed from
Haufroid et al. (2004), who observed a 1.6-fold higher dose-
adjusted trough levels in CYP3A5 non-expressers among 50 stable
kidney transplant patients when compared to CYP3A5 expressers
[57]. Similarly, Tang et al. (2010) observed an increased dose-
adjusted C0 concentration for CYP3A5 non-expressers among 1821
renal transplant patients [16].
Regarding genotypes and the prevalence of acute rejection, the
results of this study did not show any significant association be-
692 Current Drug Metabolism, 2019, Vol. 20, No. 8 El-Shair et al.
tween the four genotypes and acute rejection prevalence. These
observations differ from Crettol et a l. (2008) who observed a
significant association between CYP3A4*1B and the prevalence of
acute rejection [17]. However, Tang et al. (2010), Zhu et al. (2011)
and Qiu et al. (2011) all failed to reveal any significant effect of
CYP3A5*3 genotypes on the prevalence of acute rejection [16, 47,
58].
Lastly, this study has some limitations such as a relatively small
sample and the different cyclosporine sampling methods when
compared to different institutions.
CONCLUSION
There was a strong association between CYP3A4*22 and mean
difference between the second and first given doses (P=0.034).
There was a trend of significant difference between the mean of the
first C2 blood levels among heterozygous CYP3A4*22 patients
(P=0.063). Pharmacogenomics may hold promise in assisting the
prediction of the best cyclosporine dose and C2 blood level among
Jordanian kidney transplant patients.
LIST OF ABBREVIATIONS
AE = Adverse Events
AUC0-4 = Area Under the Curve from 0 to 4 Hours
Bp = Base Pair
C0 = Cyclosporine trough Concentration at Zero Hour
C2 = 2-hours post-dose Cyclosporine Blood Concentra-
tion
Cmax = Maximum Plasma Concentration
CsA = Cyclosporine
CYP-450 = Cytochrome P-450
DDI = Drug-Drug Interaction
EDTA = Ethylene Diamine Tetra-acetic Acid
PCR = Polymerase Chain Reaction
PD = Pharmacodynamics
PG = Pharmacogenomics
PK = Pharmacokinetics
RFLPs-PCR = Restriction Fragment Length Polymorphism-
polymerase Chain Reaction
RRT = Renal Replacement Therapy
S.D. = Standard Deviation
SNP = Single Nucleotide Polymorphism
SPSS = Statistical Package for Social Science
St. Err = Standard Error
TDM = Therapeutic Drug Monitoring
ETHICS APPROVAL AND CONSENT TO PARTICIPATE
Ethical approval of IRB at Prince Hamzah Hospital was granted
on 17/Dec/2018 (Ref. No.: (3309/32).
HUMAN AND ANIMAL RIGHTS
The reported experiments were in accordance with the ethical
standards of the committee responsible for human experimentation
(institutional and national), and with the Helsinki Declaration of
1975, as revised in 2013 (http://ethics.iit.edu/ecodes/node/3931).
CONSENT FOR PUBLICATION
Approved by the Institutional Review Board of Prince Hamzah
Hos-pital, Jordan.
AVAILABILITY OF DATA AND MATERIALS
Not applicable.
FUNDING
This work was supported by the Pharmacology Dept. at the
University of Jordan and Deanship of Scientific and Academic
Research at the University of Jordan.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or other-
wise.
ACKNOWLEDGEMENTS
We thank Dr. Rula Saeed (Jordan Hospital). We also thank Dr.
Elias Turk, Nurse Diyaa Fahjan, Nurse Mohammad Khdour, Nurse
Mahmoud Nababteh, staff members of Nephrology and Kidney
Transplantation Clinic and all staff members of Prince Hamzah
Hospital.
REFERENCES
[1] Ware, N. The role of genetics in drug dosing. Pediatr. Nephrol.,
2012, 27(9), 1489-1498.
[http://dx.doi.org/10.1007/s00467-012-2105-0] [PMID: 22358188]
[2] Ingelfinger, J.R.; Schwartz, R.S. Immunosuppression--the promise
of specificity. N. Engl. J. Med., 2005, 353(8), 836-839.
[http://dx.doi.org/10.1056/NEJMe058166] [PMID: 16120865]
[3] McKay, D.; Steinberg, S. Kidney transplantation: A guide to the
care of kidney transplant recipients; Springer, 2010.
[http://dx.doi.org/10.1007/978-1-4419-1690-7]
[4] Picard, N.; Marquet, P. The influence of pharmacogenetics and
cofactors on clinical outcomes in kidney transplantation. Expert
Opin. Drug Metab. Toxicol., 2011, 7(6), 731-743.
[http://dx.doi.org/10.1517/17425255.2011.570260] [PMID:
21434840]
[5] Meng, X.G.; Guo, C.X.; Feng, G.Q.; Zhao, Y.C.; Zhou, B.T.; Han,
J.L.; Chen, X.; Shi, Y.; Shi, H.Y.; Yin, J.Y.; Peng, X.D.; Pei, Q.;
Zhang, W.; Wang, G.; He, M.; Liu, M.; Yang, J.K.; Zhou, H.H. As-
sociation of CYP3A polymorphisms with the pharmacokinetics of
cyclosporine A in early post-renal transplant recipients in China.
Acta Pharmacol. Sin., 2012, 33(12), 1563-1570.
[http://dx.doi.org/10.1038/aps.2012.136] [PMID: 23085740]
[6] Rosso Felipe, C.; de Sandes, T.V.; Sampaio, E.L.; Park, S.I.; Silva,
H.T., Jr; Medina Pestana, J.O. Clinical impact of polymorphisms of
transport proteins and enzymes involved in the metabolism of im-
munosuppressive drugs. Transplant. Proc., 2009, 41(5), 1441-
1455.
[http://dx.doi.org/10.1016/j.transproceed.2009.03.024] [PMID:
19545654]
[7] Cheung, C.Y. Pharmacogenetics and renal transplantation; IN-
TECH Open Access Publisher, 2011.
[8] Canafax, D.M. Minimizing cyclosporine concentration variability
to optimize transplant outcome. Clin. Transplant., 1995, 9(1), 1-13.
[PMID: 7742576]
[9] Eng, H.S.; Mohamed, Z.; Calne, R.; Lang, C.C.; Mohd, M.A.; Seet,
W.T.; Tan, S.Y. The influence of CYP3A gene polymorphisms on
cyclosporine dose requirement in renal allograft recipients. Kidney
Int., 2006, 69(10), 1858-1864.
[http://dx.doi.org/10.1038/sj.ki.5000325] [PMID: 16612333]
[10] Cattaneo, D.; Perico, N.; Remuzzi, G. From pharmacokinetics to
pharmacogenomics: A new approach to tailor immunosuppressive
therapy. Am. J. Transplant, 2004, 4(3), 299-310.
[http://dx.doi.org/10.1111/j.1600-6143.2004.00312.x]
[11] Bäckman, L.; Levy, M.F.; Klintmalm, G. Whole-blood and plasma
levels of FK 506 after liver transplantation: Results from the US
Multicenter Trial. FK506 Multicenter Study Group. Transplant.
Proc., 1995, 27(1), 1124-1124.
[PMID: 7533361]
[12] Zaza, G.; Granata, S.; Sallustio, F.; Grandaliano, G.; Schena, F.P.
Pharmacogenomics: A new paradigm to personalize treatments in
nephrology patients. Clin. Exp. Immunol., 2010, 159(3), 268-280.
[http://dx.doi.org/10.1111/j.1365-2249.2009.04065.x] [PMID:
19968662]
Association Between CYP3A4 and CYP3A5 Genotypes Current Drug Metabolism, 2019, Vol. 20, No. 8 693
[13] Kalow, W.; Tang, B.K.; Endrenyi, L. Hypothesis: Comparisons of
inter- and intra-individual variations can substitute for twin studies
in drug research. Pharmacogenetics, 1998, 8(4), 283-289.
[http://dx.doi.org/10.1097/00008571-199808000-00001] [PMID:
9731714]
[14] Lamba, J.K.; Lin, Y.S.; Thummel, K.; Daly, A.; Watkins, P.B.;
Strom, S.; Zhang, J.; Schuetz, E.G. Common allelic variants of cy-
tochrome P4503A4 and their prevalence in different populations.
Pharmacogenetics, 2002, 12(2), 121-132.
[http://dx.doi.org/10.1097/00008571-200203000-00006] [PMID:
11875366]
[15] Combalbert, J.; Fabre, I.; Fabre, G.; Dalet, I.; Derancourt, J.; Cano,
J.P.; Maurel, P. Metabolism of cyclosporin A. IV. Purification and
identification of the rifampicin-inducible human liver cytochrome
P-450 (cyclosporin A oxidase) as a product of P450IIIA gene sub-
family. Drug Metab. Dispos., 1989, 17(2), 197-207.
[PMID: 2565211]
[16] Tang, H.L.; Ma, L.L.; Xie, H.G.; Zhang, T.; Hu, Y.F. Effects of the
CYP3A5*3 variant on cyclosporine exposure and acute rejection
rate in renal transplant patients: a meta-analysis. Pharmacogenet.
Genomics, 2010, 20(9), 525-531.
[http://dx.doi.org/10.1097/FPC.0b013e32833ccd56] [PMID:
20588203]
[17] Crettol, S.; Venetz, J-P.; Fontana, M.; Aubert, J-D.; Pascual, M.;
Eap, C.B. CYP3A7, CYP3A5, CYP3A4, and ABCB1 genetic po-
lymorphisms, cyclosporine concentration, and dose requirement in
transplant recipients. Ther. Dru g Monit., 2008, 30(6), 689-699.
[http://dx.doi.org/10.1097/FTD.0b013e31818a2a60] [PMID:
18978522]
[18] Barbarino, J.M.; Staatz, C.E.; Venkataramanan, R.; Klein, T.E.;
Altman, R.B. PharmGKB summary: Cyclosporine and tacrolimus
pathways. Pharmacogenet. Genomics, 2013, 23(10), 563-585.
[http://dx.doi.org/10.1097/FPC.0b013e328364db84] [PMID:
23922006]
[19] Schirmer, M.; Rosenberger, A.; Klein, K.; Kulle, B.; Toliat, M.R.;
Nürnberg, P.; Zanger, U.M.; Wojnowski, L. Sex-dependent genetic
markers of CYP3A4 expression and activity in human liver micro-
somes. Pharmacogenomics, 2007, 8(5), 443-453.
[http://dx.doi.org/10.2217/14622416.8.5.443] [PMID: 17465708]
[20] Fohner, A.; Muzquiz, L.I.; Austin, M.A.; Gaedigk, A.; Gordon, A.;
Thornton, T.; Rieder, M.J.; Pershouse, M.A.; Putnam, E.A.;
Howlett, K.; Beatty, P.; Thummel, K.E.; Woodahl, E.L. Pharmaco-
genetics in american indian populations: Analysis of cyp2d6,
cyp3a4, cyp3a5, and cyp2c9 in the confederated salish and koo-
tenai tribes. Pharmacogenet. Genomics, 2013, 23(8), 403-414.
[http://dx.doi.org/10.1097/FPC.0b013e3283629ce9] [PMID:
23778323]
[21] Werk, A.N.; Cascorbi, I. Functional gene variants of CYP3A4.
Clin. Pharmacol. Ther., 2014, 96(3), 340-348.
[http://dx.doi.org/10.1038/clpt.2014.129] [PMID: 24926778]
[22] Skagen, K. J. Interindividual variability in the cytochrome p450
3a4 drug metabolizing enzyme: Effect of the cyp3a4* 1g genetic
variant. PhD Thesis, The University of Montana: Missoula, August
2014.
[23] Von Ahsen, N.; Richter, M.; Grupp, C.; Ringe, B.; Oellerich, M.;
Armstrong, V.W. No influence of the MDR-1 C3435T polymor-
phism or a CYP3A4 promoter polymorphism (CYP3A4-V allele)
on dose-adjusted cyclosporin A trough concentrations or rejection
incidence in stable renal transplant recipients. Clin. Chem., 2001,
47(6), 1048-1052.
[PMID: 11375290]
[24] Van Schaik, R.H.; de Wildt, S.N.; Van Iperen, N.M.; Uitterlinden,
A.G.; Van Den Anker, J.N.; Lindemans, J. CYP3A4-V polymor-
phism detection by PCR-restriction fragment length polymorphism
analysis and its allelic frequency among 199 Dutch Caucasians.
Clin. Chem., 2000, 46(11), 1834-1836.
[PMID: 11067821]
[25] Wang, D.; Guo, Y.; Wrighton, S.A.; Cooke, G.E.; Sadee, W. In-
tronic polymorphism in CYP3A4 affects hepatic expression and re-
sponse to statin drugs. Pharmacogenomics J., 2011, 11(4), 274-
286.
[http://dx.doi.org/10.1038/tpj.2010.28] [PMID: 20386561]
[26] Westlind, A.; Löfberg, L.; Tindberg, N.; Andersson, T.B.; Ingel-
man-Sundberg, M. Interindividual differences in hepatic expression
of CYP3A4: Relationship to genetic polymorphism in the 5-
upstream regulatory region. Biochem. Biophys. Res. Commun.,
1999, 259(1), 201-205.
[http://dx.doi.org/10.1006/bbrc.1999.0752] [PMID: 10334940]
[27] Lee, S.-J.; Goldstein, J.A. Functionally defective or altered cyp3a4
and cyp3a5 single nucleotide polymorphisms and their detection
with genotyping tests. Pharmacogenomics, 2005, 6(4), 357-371.
[28] Albekairy, A.; Alkatheri, A.; Fujita, S.; Hemming, A.; Howard, R.;
Reed, A.; Karlix, J. Cytochrome p450 3a4* 1b as pharmacoge-
nomic predictor of tacrolimus pharmacokinetics and clinical out-
come in the liver transplant recipients. Saudi J. Gastroenterol.,
2013, 19(2), 89.
[http://dx.doi.org/10.4103/1319-3767.108484] [PMID: 23481136]
[29] Cattaneo, D.; Perico, N.; Remuzzi, G. From pharmacokinetics to
pharmacogenomics: A new approach to tailor immunosuppressive
therapy. Am. J. Transplant., 2004, 4(3), 299-310.
[http://dx.doi.org/10.1111/j.1600-6143.2004.00312.x] [PMID:
14961981]
[30] Shi, X.J.; Geng, F.; Jiao, Z.; Cui, X.Y.; Qiu, X.Y.; Zhong, M.K.
Association of ABCB1, CYP3A4*18B and CYP3A5*3 genotypes
with the pharmacokinetics of tacrolimus in healthy Chinese sub-
jects: A population pharmacokinetic analysis. J. Clin. Pharm.
Ther., 2011, 36(5), 614-624.
[http://dx.doi.org/10.1111/j.1365-2710.2010.01206.x] [PMID:
21916909]
[31] He, B.X.; Shi, L.; Qiu, J.; Tao, L.; Li, R.; Yang, L.; Zhao, S.J. A
functional polymorphism in the CYP3A4 gene is associated with
increased risk of coronary heart disease in the Chinese Han popula-
tion. Basic Clin. Pharmacol. Toxicol., 2011, 108(3), 208-213.
[http://dx.doi.org/10.1111/j.1742-7843.2010.00657.x] [PMID:
21199372]
[32] Shu, W.Y.; Li, J.L.; Wang, X.D.; Huang, M. Pharmacogenomics
and personalized medicine: A review focused on their application
in the Chinese population. Acta Pharmacol. Sin., 2015, 36(5), 535-
543.
[http://dx.doi.org/10.1038/aps.2015.10] [PMID: 25891088]
[33] Zeng, Y.; He, Y.J.; He, F.Y.; Fan, L.; Zhou, H.H. Effect of bifen-
date on the pharmacokinetics of cyclosporine in relation to the
CYP3A4*18B genotype in healthy subjects. Acta Pharmacol. Sin.,
2009, 30(4), 478-484.
[http://dx.doi.org/10.1038/aps.2009.27] [PMID: 19343062]
[34] Wang, D.; Sadee, W. The making of a cyp3a biomarker panel for
guiding drug therapy J. Pers. Med, 2012, 2, 175-191.
[35] Elens, L.; Bouamar, R.; Hesselink, D.A .; Haufroid, V.; van Gelder,
T.; van Schaik, R.H. The new CYP3A4 intron 6 C>T polymor-
phism (CYP3A4*22) is associated with an increased risk of de-
layed graft function and worse renal function in cyclosporine-
treated kidney transplant patients. Pharmacogenet. Genomics,
2012, 22(5), 373-380.
[http://dx.doi.org/10.1097/FPC.0b013e328351f3c1] [PMID:
22388796]
[36] Lunde, I.; Bremer, S.; Midtvedt, K.; Mohebi, B.; Dahl, M.; Bergan,
S.; Åsberg, A.; Christensen, H. The influence of CYP3A, PPARA,
and POR genetic variants on the pharmacokinetics of tacrolimus
and cyclosporine in renal transplant recipients. Eur. J. Clin. Phar-
macol., 2014, 70(6), 685-693.
[http://dx.doi.org/10.1007/s00228-014-1656-3] [PMID: 24658827]
[37] Kuehl, P.; Zhang, J.; Lin, Y.; Lamba, J.; Assem, M.; Schuetz, J.;
Watkins, P.B.; Daly, A.; Wrighton, S.A.; Hall, S.D.; Maurel, P.;
Relling, M.; Brimer, C.; Yasuda, K.; Venkataramanan, R.; Strom,
S.; Thummel, K.; Boguski, M.S.; Schuetz, E. Sequence diversity in
CYP3A promoters and characterization of the genetic basis of po-
lymorphic CYP3A5 expression. Nat. Genet., 2001, 27(4), 383-391.
[http://dx.doi.org/10.1038/86882] [PMID: 11279519]
[38] Dai, Y.; Iwanaga, K.; Lin, Y.S.; Hebert, M.F.; Davis, C.L.; Huang,
W.; Kharasch, E.D.; Thummel, K.E. In vitro metabolism of cy-
closporine A by human kidney CYP3A5. Biochem. Pharmacol.,
2004, 68(9), 1889-1902.
[http://dx.doi.org/10.1016/j.bcp.2004.07.012] [PMID: 15450954]
[39] Birdwell, K. Role of pharmacogenomics in dialysis and transplan-
tation. Curr. Opin. Nephrol. Hypertens., 2014, 23(6), 570-577.
[http://dx.doi.org/10.1097/MNH.0000000000000065] [PMID:
25162201]
[40] Yousef, A-M.; Qosa, H.; Bulatova, N.; Abuhaliema, A.; Almad-
houn, H.; Khayyat, G.; Olemat, M. Effects of genetic polymor-
phism in cyp3a4 and cyp3a5 genes on tacrolimus dose among kid-
694 Current Drug Metabolism, 2019, Vol. 20, No. 8 El-Shair et al.
ney transplant recipients. Iran. J. Kidney Dis., 2016, 10(3), 156-
163.
[PMID: 27225724]
[41] Saytolu, M.; Yildiz, I.; Hatirnaz, Ö.; Özbek, U. Common cyto-
chrome p4503a (cyp3a4 and cyp3a5) and thiopurine s-methyl trans-
ferase (tpmt) polymorphisms in turkish population. Turk. J. Med.
Sci., 2006, 36(1), 11-15.
[42] Gervasini, G.; Vizcaino, S.; Gasiba, C.; Carrillo, J.A.; Benitez, J.
Differences in CYP3A5*3 genotype distribution and combinations
with other polymorphisms between Spaniards and other Caucasian
populations. Ther. Drug Monit., 2005, 27(6), 819-821.
[http://dx.doi.org/10.1097/01.ftd.0000186914.32038.a0] [PMID:
16306861]
[43] Umamaheswaran, G.; Krishna Kumar, D.; Adithan, C. Distribution
of genetic polymorphisms of genes encoding drug metabolizing en-
zymes & drug transporters-a review with indian perspective. Indian
J Med Res., 2014, 139(1), 27-65.
[PMID: 24604039]
[44] Kurzawski, M.; Dąbrowska, J.; Dziewanowski, K.; Domański, L.;
Perużyńska, M.; Droździk, M. CYP3A5 and CYP3A4, but not
ABCB1 polymorphisms affect tacrolimus dose-adjusted trough
concentrations in kidney transplant recipients. Pharmacogenomics,
2014, 15(2), 179-188.
[http://dx.doi.org/10.2217/pgs.13.199] [PMID: 24444408]
[45] Hesselink, D.A.; Van Gelder, T.; Van Schaik, R.H.; Balk, A.H.;
Van Der Heiden, I.P.; Van Dam, T.; Van Der Werf, M.; Weimar,
W.; Mathot, R.A. Population pharmacokinetics of cyclosporine in
kidney and heart transplant recipients and the influence of ethnicity
and genetic polymorphisms in the MDR-1, CYP3A4, and CYP3A5
genes. Clin. Pharmacol. Ther., 2004, 76(6), 545-556.
[http://dx.doi.org/10.1016/j.clpt.2004.08.022] [PMID: 15592326]
[46] Lee, J.S.; Cheong, H.S.; Kim, L.H.; Kim, J.O.; Seo, D.W.; Kim,
Y.H.; Chung, M.W.; Han, S.Y.; Shin, H.D. Screening of genetic
polymorphisms of cyp3a4 and cyp3a5 genes. Korean J. Physiol.
Pharmacol., 2013, 17(6), 479-484.
[http://dx.doi.org/10.4196/kjpp.2013.17.6.479] [PMID: 24381495]
[47] Qiu, F.; He, X-J.; Sun, Y-X.; Li-Ling, J.; Zhao, L-M. Influence of
ABCB1, CYP3A4*18B and CYP3A5*3 polymorphisms on cy-
closporine A pharmacokinetics in bone marrow transplant recipi-
ents. Pharmacol. Rep., 2011, 63(3), 815-825.
[http://dx.doi.org/10.1016/S1734-1140(11)70594-1] [PMID:
21857093]
[48] Garsa, A.A.; McLeod, H.L.; Marsh, S. CYP3A4 and CYP3A5
genotyping by pyrosequencing. BMC Med. Genet., 2005, 6(1), 19.
[http://dx.doi.org/10.1186/1471-2350-6-19] [PMID: 15882469]
[49] Park, S.Y.; Kang, Y.S.; Jeong, M.S.; Yoon, H.K.; Han, K.O. Fre-
quencies of CYP3A5 genotypes and haplotypes in a Korean popu-
lation. J. Clin. Pharm. Ther., 2008, 33(1), 61-65.
[http://dx.doi.org/10.1111/j.1365-2710.2008.00879.x] [PMID:
18211618]
[50] Renwick, A.G.; Robertson, D.R.; Macklin, B.; Challenor, V.;
Waller, D.G.; George, C.F. The pharmacokinetics of oral nifedip-
ine-a population study. Br. J. Clin. Pharmacol., 1988, 25(6), 701-
708.
[http://dx.doi.org/10.1111/j.1365-2125.1988.tb05256.x] [PMID:
3203042]
[51] Rivory, L.P.; Qin, H.; Clarke, S.J.; Eris, J.; Duggin, G.; Ray, E.;
Trent, R.J.; Bishop, J.F. Frequency of cytochrome P450 3A4 vari-
ant genotype in transplant population and lack of association with
cyclosporin clearance. Eur. J. Clin. Pharmacol., 2000, 56(5), 395-
398.
[http://dx.doi.org/10.1007/s002280000166] [PMID: 11009048]
[52] Ashavaid, T.; Raje, H.; Shalia, K.; Shah, B. Effect of gene poly-
morphisms on the levels of calcineurin inhibitors in Indian renal
transplant recipients. Indian J. Nephrol., 2010, 20(3), 1 46-151.
[http://dx.doi.org/10.4103/0971-4065.70846] [PMID: 21072155]
[53] Zhang, Y.; Li, J.L.; Fu, Q.; Wang, X.D.; Liu, L.S.; Wang, C.X.;
Xie, W.; Chen, Z.J.; Shu, W.Y.; Huang, M. Associations of
ABCB1, NFKB1, CYP3A, and NR1I2 polymorphisms with cy-
closporine trough concentrations in Chinese renal transplant recipi-
ents. Acta Pharmacol. Sin., 2013, 34(4), 555-560.
[http://dx.doi.org/10.1038/aps.2012.200] [PMID: 23503472]
[54] Hu, Y-F.; Tu, J-H.; Tan, Z-R.; Liu, Z-Q.; Zhou, G.; He, J.; Wang,
D.; Zhou, H-H. Association of CYP3A4*18B polymorphisms with
the pharmacokinetics of cyclosporine in healthy subjects. Xenobi-
otica, 2007, 37(3), 315-327.
[http://dx.doi.org/10.1080/00498250601149206] [PMID:
17624028]
[55] Qiu, X.Y.; Jiao, Z.; Zhang, M.; Zhong, L.J.; Liang, H.Q.; Ma, C.L.;
Zhang, L.; Zhong, M.K. Association of MDR1, CYP3A4*18B, and
CYP3A5*3 polymorphisms with cyclosporine pharmacokinetics in
Chinese renal transplant recipients. Eur. J. Clin . Pharmacol., 2008,
64(11), 1069-1084.
[http://dx.doi.org/10.1007/s00228-008-0520-8] [PMID: 18636247]
[56] Elens, L.; van Schaik, R.H.; Panin, N.; de Meyer, M.; Wallemacq,
P.; Lison, D.; Mourad, M.; Haufroid, V. Effect of a new functional
CYP3A4 polymorphism on calcineurin inhibitors’ dose require-
ments and trough blood levels in stable renal transplant patients.
Pharmacogenomics, 2011, 12(10), 1383-1396.
[http://dx.doi.org/10.2217/pgs.11.90] [PMID: 21902502]
[57] Haufroid, V.; Mourad, M.; Van Kerckhove, V.; Wawrzyniak, J.;
De Meyer, M.; Eddour, D.C.; Malaise, J.; Lison, D.; Squifflet, J-P.;
Wallemacq, P. The effect of CYP3A5 and MDR1 (ABCB1) poly-
morphisms on cyclosporine and tacrolimus dose requirements and
trough blood levels in stable renal transplant patients. Pharmaco-
genetics, 2004, 14(3), 147-154.
[http://dx.doi.org/10.1097/00008571-200403000-00002] [PMID:
15167702]
[58] Zhu, H.J.; Yuan, S.H.; Fang, Y.; Sun, X.Z.; Kong, H.; Ge, W.H.
The effect of CYP3A5 polymorphism on dose-adjusted cy-
closporine concentration in renal transplant recipients: A meta-
analysis. Pharmacogenomics J., 2011, 11(3), 23 7-246.
[http://dx.doi.org/10.1038/tpj.2010.26] [PMID: 20368718]
... It is metabolized by cytochrome P-450, encoded by the CYP genes cluster. It is well known that polymorphisms of the intracellular metabolizer enzyme CYP and the trans-membrane transport protein ABCB1 may influence enzymatic intracellular activity, modifying drugs metabolism [11][12][13][14][15][16][17][18][19]. Patients with the A allele on CYP3A5*3 need to double the dose of tacrolimus in order to reach therapeutic blood concentration [20]. ...
... The role of changes in drug metabolism, induced by polymorphisms of a number of genes, has been repeatedly underlined in the last two decades [11][12][13][14][15][16][17][18][19]. ...
Article
Full-text available
Background: We evaluated the role of CYP3A5, ABCB1 and SXR gene polymorphisms in the occurrence of acute kidney rejection in a cohort of pediatric renal transplant recipients. Methods: Forty-nine patients were genotyped for CYP3A5, ABCB1 and SXR polymorphisms and evaluated with tacrolimus through levels in a retrospective monocenter study. Results: Patients with the A allele of CYP3A5 treated with tacrolimus had a higher risk of acute rejection than those without the A allele, while patients carrying the homozygous GG variant for SXR A7635GG did not show any episode of acute rejection. Conclusion: Genetic analysis of polymorphisms implicated in drug metabolism and tacrolimus trough levels may help to forecast the risk of acute rejection and individualize drug dosage in children undergoing renal transplantation.
... Research (Apellániz-Ruiz et al. 2015) indicates that the CYP3A4*20 allele is specific to the Spanish population, present in 1.2% of individuals (up to 3.8% in specific regions). Although no studies have reported the impact of CYP3A4 genetic polymorphism on the pharmacokinetics or pharmacodynamics of ziprasidone, multiple studies on the impact of other CYP3A4 substrates have been published (Bins et al. 2019;El-Shair et al. 2019;Hannachi et al. 2020;Olagunju et al. 2014). Quetiapine, a commonly used antipsychotic in psychiatry, is extensively metabolized in the liver primarily by CYP3A4 (Aichhorn et al. 2006). ...
Article
Full-text available
Ziprasidone is widely used in the treatment of psychiatric disorders. Despite its prevalence, there is a notable lack of population pharmacokinetics (PPK) studies on ziprasidone in serum, both domestically and internationally. This study aimed to comprehensively investigate the various factors influencing the PPK characteristics of Ziprasidone, thereby providing a scientific basis for personalized treatment strategies in clinical settings. This is a retrospective study. A non-linear mixed-effects modeling method was used for data analysis, with the ziprasidone PPK model established using the Phoenix NLME 8.1 software. Model evaluation employed goodness-of-fit plots, visual predictive checks, and Bootstrap methods to ensure reliability and accuracy. To further validate the model’s applicability, data from an additional 30 patients meeting the same inclusion criteria but not included in the final model were collected for external validation. Simulations were performed to explore the personalized dosage regimens. This retrospective analysis collected 547 drug concentration data points from 185 psychiatric disorder patients, along with related medical records. The data included detailed demographic information (such as age, gender, weight), dosing regimens, laboratory test results, and concomitant medication details. In the final model, Ka was fixed at 0.5 h⁻¹ based on literature, and the population typical values for ziprasidone clearance (CL) and volume of distribution (V) were 18.74 L/h and 110.24 L, respectively. Co-administration of lorazepam and valproic acid significantly influenced the clearance of ziprasidone. Moreover, the model evaluation indicated good stability and predictive accuracy. A simple to use dosage regimen table was derived based on the results of simulations. This study successfully established and validated a PPK model for ziprasidone in Chinese patients with psychiatric disorders. The model provides a scientific reference for individualized dosing of ziprasidone and holds the potential to optimize treatment strategies, thereby enhancing therapeutic efficacy and safety.
... Other CYP3A4 polymorphisms play a role in the pharmacodynamics of CsA: a Jordanian study found a strong direct correlation between the CYP3A4*22 polymorphism and a higher C0/dose [21]. ...
Article
Full-text available
Kidney transplantation is the preferred therapeutic option for end-stage kidney disease, but, despite major therapeutic advancements, allograft rejection continues to endanger graft survival. Every patient is unique due to his or her clinical history, drug metabolism, genetic background, and epigenetics. For this reason, examples of “personalized medicine” and “precision medicine” have steadily increased in recent decades. The final target of precision medicine is to maximize drug efficacy and minimize toxicity for each individual patient. Immunosuppressive drugs, in the setting of kidney transplantation, require a precise dosage to avoid either adverse events (overdosage) or a lack of efficacy (underdosage). In this review, we will explore the knowledge regarding the pharmacogenomics of the main immunosuppressive medications currently utilized in kidney transplantation. We will focus on clinically relevant pharmacogenomic data, that is, the polymorphisms of the genes that metabolize immunosuppressive drugs.
... The CYP3A4 gene is located on chromosome 7q21.3-q22.1 consisting of 13 exons (Keshava et al., 2004). The most important single nucleotide polymorphism (SNP) within the CYP3A4 family is CYP3A4*1B (rs2740574) (Alessandrini et al., 2013), an A to G transition at nucleotide 392 in the promoter sequence of the gene (El-Shair et al., 2019). This SNP is associated with poor metabolism of artemether and lumefantrine (Staehli Hodel et al., 2013) (Piedade and Gil, 2011). ...
Article
Full-text available
Background : Therapeutic efficacy of artemether-lumefantrine is highly dependent on adequate systemic exposure to the partner drug lumefantrine particularly day 7 lumefantrine plasma concentration. There has been contradicting findings on the role of the cut-off values in predicting treatment outcomes among malaria patients in malaria endemic regions. This study assesses the day 3 and 7 lumefantrine plasma concentrations including related determinant factors and their influence on treatment outcomes among treated Tanzanian children and adults in uncontrolled conditions (real life condition). Methods : Data was nested from an efficacy study employing the WHO protocol, 2015 for monitoring antimalarial drug efficacy. Lumefantrine plasma concentration was measured by high performance liquid chromatography with ultraviolet (HPLC-UV). Results: Lumefantrine plasma concentrations below 175ng/ml and 200ng/ml on day 3 and 7 did not affect adequate clinical and parasitological response (ACPR) and recurrence of infection (p=0.428 and 0.239 respectively). Age and baseline parasitemia were not associated to day 3 median lumefantrine plasma concentrations (p =0.08 and 0.31 respectively) and day 7 lumefantrine plasma concentrations (p= 0.07 and 0.41respectively). However, the day 3 and day 7 lumefantrine plasma concentrations were significantly higher in males compared to females (p=0.03 and 0.042 respectively). Conclusion : Lumefantrine plasma concentrations below cut off points (175ng/ml and 200ng/ml) on day 3 and 7 did not influence treatment outcomes.
... Several studies have suggested the conflicting roles of CYP3A4*1G in CYP3A4 activity. CYP3A4*1G polymorphism enhances the disposition of CYP3A4 substrates, including atorvastatin (Gao et al., 2008), cyclosporin A (El-Shair et al., 2019), and tacrolimus (Tamashiro et al., 2017;Tang et al., 2019). In contrast, CYP3A4*1G polymorphism is reported to decrease the disposition of fentanyl (Yuan et al., 2015). ...
Article
Full-text available
Teneligliptin, a dipeptidyl peptidase-4 inhibitor, is used to treat type 2 diabetes mellitus. FMO3 and CYP3A4 metabolize teneligliptin into teneligliptin sulfoxide. This study examined the effects of FMO3 (rs909530, rs1800822, rs2266780, and rs2266782) and CYP3A4 (rs2242480) polymorphisms on teneligliptin pharmacokinetics at a steady state among 23 healthy participants administered 20 mg teneligliptin daily for 6 days. Subjects with FMO3 rs909530, rs2266780, and rs2266782 polymorphisms exhibited a significant gene dosage-dependent increase in maximum steady-state plasma drug concentration (Cmax,ss) and area under the drug concentration vs time curve (AUC) (p<0.05). However, the Cmax values significantly decreased but the AUC values did not significantly vary in subjects with CYP3A4 polymorphism (rs2242480). These results suggest that FMO3 and CYP3A4 polymorphisms affect teneligliptin pharmacokinetics in humans. The findings of this study provide a scientific basis for the inter-individual variation in teneligliptin disposition.
Article
Full-text available
Background Sub-Saharan Africa (SSA) population is genetically diverse and heterogenous thus variability in drug response among individuals is predicted to be high. Cytochrome P450 (CYP450) polymorphisms is a major source of variability in drug response. This systematic review presents the influence of CYP450 single nucleotide polymorphisms (SNPs), particularly CYP3A4*1B, CYP2B6*6 and CYP3A5*3 on antimalarial drug plasma concentrations, efficacy and safety in SSA populations. Methods Searching for relevant studies was done through Google Scholar, Cochrane Central Register of controlled trials (CENTRAL), PubMed, Medline, LILACS, and EMBASE online data bases. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines were used. Two independent reviewers extracted data from the studies. Results Thirteen studies reporting the influence of CYP450 SNPs on plasma concentrations, efficacy and safety were included in the final data synthesis. CYP3A4*1B, CYP3A5*5, CYP2B6*6 and CYP2C8*2 did not affect antimalarial drug plasma concentration significantly. There was no difference in treatment outcomes between malaria patients with variant alleles and those with wild type alleles. Conclusion This review reports lack of influence of CYP3A4*1B, CYP3A5*3, CYP2C8*3 and CYP2B6*6 SNPs on PK profiles, efficacy and safety in SSA among P. falciparum malaria patients.
Article
Full-text available
Cyclosporine‐A (Cyc‐A) was initially prescribed as systemic therapy for patients receiving solid organ transplants or in patients with graft versus host disease (GVHD). Topical Cyc‐A is an ideal form of Cyclosporine in the treatment of mucocutaneous disorders as it causes fewer systemic side effects and has more stable results than steroids; however, poor absorption through the skin makes the development of new formulations necessary to improve skin permeability. To evaluate the efficacy and safety of topical Cyc‐A in different dermatological conditions. A thorough systematic review was performed on PubMed/Medline, Embase, Scopus, and Web of Science databases as well as Google Scholar, and relevant studies from 2000 until January 3rd, 2022, were selected. The study was conducted according to the Preferred Reporting Items for Systematic reviews and Meta‐Analysis (PRISMA). Topical Cyc‐A was observed to be an effective medication in the treatment of oral lichen planus, psoriasis, burning mouth syndrome, Pyoderma Gangrenosum, and Zoon's balanitis. Adverse side effects such as dysphagia, burning sensation, lips swealing, and gastrointestinal upset were reported following Cyc‐A mouthwash use, whereas mild erythema, dryness, and fissuring of the skin were observed following the Cyc‐A lipogel application. Topical Cyc‐A was found to be a good alternative to traditional treatment regimens for immune‐mediated mucocutaneous conditions. Cyc‐A can be considered as a safe and efficient option in cases of long‐term treatment as it does not have the same adverse effects of long‐term steroids. This article is protected by copyright. All rights reserved.
Article
Full-text available
Cytochrome P450 3A4 (CYP3A4) is the most important drug metabolizing enzyme in the liver, responsible for the oxidative metabolism of ∼50% of clinically prescribed drugs. Therefore, genetic variation in CYP3A4 could potentially affect the pharmacokinetics, toxicity and clinical outcome of drug treatment. Thus far, pharmacogenetics for CYP3A4 has not received much attention. However, the recent discovery of the intron 6 single-nucleotide polymorphism (SNP) rs35599367C > T, encoding the CYP3A4∗22 allele, led to several studies into the pharmacogenetic effect of CYP3A4∗22 on different drugs. This allele has a relatively minor allele frequency of 3-5% and an effect on CYP3A4 enzymatic activity. Thus far, no review summarizing the data published on several drugs is available yet. This article therefore addresses the current knowledge on CYP3A4∗22. This information may help in deciding if, and for which drugs, CYP3A4∗22 genotype-based dosing could be helpful in improving drug therapy. CYP3A4∗22 was shown to significantly influence the pharmacokinetics of several drugs, with currently being most thoroughly investigated tacrolimus, cyclosporine, and statins. Additional studies, focusing on toxicity and clinical outcome, are warranted to demonstrate clinical utility of CYP3A4∗22 genotype-based dosing.
Article
Objectives: CYP3A5 and ABCB1 are highly implicated in the pharmacokinetics and pharmacodynamics of immunosuppressive agents, such as calcineurin inhibitors and mammalian target of rapamycin inhibitors. The polymorphisms of their coding genes play important roles in the interindividual and intraindividual differences of bioavailability of these drugs. In this study, our objective was to investigate, in a Lebanese population,the frequency of ABCB1C3435T (rs1045642) and CYP3A5*3 (rs776746) polymorphisms and to compare the results to preexisting data from other populations. Materials and methods: We determined the frequencies of the allelic variants of interest for 1824 Lebanese participants, and we compared these results with those from other major ethnic groups. Results: The allelic frequencies were 91.4% (C) and 8.6% (T) for CYP3A5*3 and 50.8% (T) and 49.2% (C) for ABCB1 C3435T. Our results were significantly different from most other world populations, except the European population. Conclusions: The frequencies of gene variants of interest in our Lebanese population were similar to those found in European populations. Most of our study population were CYP3A5*3 carriers, and more than half may have a lower P-glycoprotein efflux pump. These characteristics might render Lebanese transplant recipients more prone to the development of drug toxicity and in need of lower drug doses.
Article
Full-text available
Tacrolimus (Tac) and cyclosporine (CsA) are mainly metabolized by CYP3A4 and CYP3A5. Several studies have demonstrated an association between the CYP3A5 genotype and Tac dose requirements. Recently, CYP3A4, PPARA, and POR gene variants have been shown to influence CYP3A metabolism. The present study investigated potential associations between CYP3A5*3, CYP3A4*22, PPARA c.209-1003G>A and c.208 + 3819A>G, and POR*28 alleles and dose-adjusted concentrations (C/D) of Tac and CsA in 177 renal transplant patients early post-transplant. All patients (n = 177) were genotyped for CYP3A4*22, CYP3A5*3, POR*28, PPARA c.209-1003G>A, and PPARA c.208 + 3819A>G using real-time polymerase chain reaction (PCR) and melting curve analysis with allele-specific hybridization probes or PCR restriction fragment length polymorphisms (RFLP) methods. Drug concentrations and administered doses were retrospectively collected from patient charts at Oslo University Hospital, Rikshospitalet, Norway. One steady-state concentration was collected for each patient. We confirmed a significant impact of the CYP3A5*3 allele on Tac exposure. Patients with POR*28 and PPARA variant alleles demonstrated 15 % lower (P = 0.04) and 19 % higher (P = 0.01) Tac C0/D respectively. CsA C2/D was 53 % higher among CYP3A4*22 carriers (P = 0.03). The results support the use of pre-transplant CYP3A5 genotyping to improve initial dosing of Tac, and suggest that Tac dosing may be further individualized by additional POR and PPARA genotyping. Furthermore, initial CsA dosing may be improved by pre-transplant CYP3A4*22 determination.
Article
Full-text available
Phase I and II drug metabolizing enzymes (DME) and drug transporters are involved in the absorption, distribution, metabolism as well as elimination of many therapeutic agents, toxins and various pollutants. Presence of genetic polymorphisms in genes encoding these proteins has been associated with marked inter-individual variability in their activity that could result in variation in drug response, toxicity as well as in disease predisposition. The emergent field pharmacogenetics and pharmacogenomics (PGx) is a promising discipline, as it predicts disease risk, selection of proper medication with regard to response and toxicity, and appropriate drug dosage guidance based on an individual's genetic make-up. Consequently, genetic variations are essential to understand the ethnic differences in disease occurrence, development, prognosis, therapeutic response and toxicity. For that reason, it is necessary to establish the normative frequency of these genes in a particular population before unraveling the genotype-phenotype associations. Although a fair amount of allele frequency data are available in Indian populations, the existing pharmacogenetic data have not been compiled into a database. This review was intended to compile the normative frequency distribution of the variants of genes encoding DMEs (CYP450s, TPMT, GSTs, COMT, SULT1A1, NAT2 and UGTs) and transporter proteins (MDR1, OCT1 and SLCO1B1) with Indian perspective.
Article
Full-text available
CYP3A ranks among the most abundant cytochrome P450 enzymes in the liver, playing a dominant role in metabolic elimination of clinically used drugs. A main member in CYP3A family, CYP3A4 expression and activity vary considerably among individuals, attributable to genetic and non-genetic factors, affecting drug dosage and efficacy. However, the extent of genetic influence has remained unclear. This review assesses current knowledge on the genetic factors influencing CYP3A4 activity. Coding region CYP3A4 polymorphisms are rare and account for only a small portion of inter-person variability in CYP3A metabolism. Except for the promoter allele CYP3A4*1B with ambiguous effect on expression, common CYP3A4 regulatory polymorphisms were thought to be lacking. Recent studies have identified a relatively common regulatory polymorphism, designated CYP3A4*22 with robust effects on hepatic CYP3A4 expression. Combining CYP3A4*22 with CYP3A5 alleles *1, *3 and *7 has promise as a biomarker predicting overall CYP3A activity. Also contributing to variable expression, the role of polymorphisms in transcription factors and microRNAs is discussed.
Article
Full-text available
Background: Tacrolimus (TAC), acting as a calcineurin inhibitor, is an immunosuppressant widely used after kidney transplantation. TAC requires blood concentration monitoring due to large interindividual variability in its pharmacokinetics and a narrow therapeutic index. Since genetic factors are considered responsible for a part of the observed pharmacokinetic variability, hereby SNPs within the CYP3A4, CYP3A5 and ABCB1 genes in kidney transplant patients of Polish Caucasian origin were investigated. Patients & methods: A total of 241 patients treated with TAC through the first year after kidney transplantation were genotyped for the presence of common SNPs: rs776746:A>G (CYP3A5*3), rs35599367:C>T (CYP3A4*22), rs2740574:A>G (CYP3A4*1B) and rs1045642:C>T (ABCB1 3435C>T) using TaqMan(®) assays. Results: CYP3A5 expressers received significantly higher weight-adjusted TAC doses, and were characterized by markedly lower C0 and dose adjusted C0 values in the course of treatment. CYP3A4*1B was significantly associated with TAC pharmacokinetics in univariate analysis. Impact of the CYP3A4*22 allele was significant only at particular time points, that is, 3 months after transplantation, with marginal significance 6 months after transplantation. The ABCB1 genotype did not influence TAC pharmacokinetics. Multivariate analysis of all the studied loci demonstrated that only the CYP3A5*1 (starting from month 1) and CYP3A4*22 alleles (at 3 and 6 months) were independent predictors of TAC dose-adjusted C0. Conclusion: Our results confirm the impact of the CYP3A4*22 allele on TAC pharmacokinetics, as a second significant genetic factor (in addition to the CYP3A5*1 allele) influencing TAC dose-adjusted blood concentrations in kidney transplant recipients.
Book
Kidney Transplantation: A Guide to the Care of Kidney Transplant Recipients is an easy to read, up to date, clinical resource written by experts in the field of kidney transplantation. The book explains how donors and recipients are selected for transplantation, how the surgical procedure is performed, and how the experts recognize and treat rejection. This guide to the care of the kidney transplant recipient aims to provide practical guidelines for management of the post-transplant recipient and is targeted to community nephrologists and general internists who care for the patient with a kidney transplant. Dianne B. McKay, MD, is Associate Professor at The Scripps Research Institute, La Jolla, California. Steven M. Steinberg, MD, is affiliated with the Sharp Memorial Hospital, San Diego, California.
Article
Introduction: This study aimed to evaluate the effects of single nucleotide polymorphisms CYP3A4*1B and CYP3A5*3 on tacrolimus dose requirement among kidney transplant recipients. Materials and methods: Blood levels of tacrolimus were measured using microparticle enzyme immunoassay. Genotyping analysis utilized specific polymerase chain reaction-restriction fragment length polymorphism methods for 137 kidney transplant recipients. Results: The median tacrolimus dose was significantly lower in the CYP3A4*1/*1 carriers (0.06 mg/kg/d; range, 0.007 mg/kg/d to 0.17 mg/kg/d) as compared to the CYP3A4*1B/*1B carriers (0.1 mg/kg/d; range, 0.03 mg/kg/d to 0.22 mg/kg/d; P = .001). Patients with at least 1 CYP3A5*1 wild-type allele required higher median doses of tacrolimus (median, 0.08 mg/kg/d; range, 0.03 mg/kg/d to 0.22 mg/kg/d) as compared to the CYP3A5*3 carriers (median, 0.05 mg/kg/d; range, 0.007 mg/kg/d to 0.17 mg/kg/d; P = .002). Conclusions: This study showed that tacrolimus dose requirement is lower in Jordanian kidney transplant recipients compared to other populations. Moreover, we found a correlation between genetic variations in CYP3A4 and CYP3A5 enzymes and tacrolimus blood levels among our kidney transplant recipients.
Article
Polymorphisms in the genes encoding cytochrome p450 (CYP) and thiopurine S-methyl transferase (TPMT) enzymes catalyze the metabolic reactions of several drugs. These polymorphisms might be responsible for adverse drug reactions. Turkish population data for these genes still needs to be elucidated. We aimed to detect the allele frequencies of thiopurine S-methyl transferase (TPMT), cytochrome p4503A4*1B (CYP3A4*1B) and cytochrome p4503A4*3 (CYP3A5*3) gene variants in the Turkish population. We examined the TPMT (*1, *2. *3A, *3C), CYP3A4*1B and CYP3A5*3 variant allele frequencies in a group of healthy Turkish Caucasian blood donors by using PCR-RFLP, allele-specific PCR and direct sequencing techniques. The frequencies of four allelelic variants of TPMT gene, are *Z (238G>C)(2.0%), *3A (460G>A and 719A>G)(1.0%), *3B (460G>A)(0.0%) and *3C (719A>G) (1.4%). We observed CYP3A4*1B allele frequency in 1.4% and CYP3A5*3 allele frequency in 7.5% of our population. This study provides the first analysis of TPMT, CYP3A4*1B and CYP3A5*3 mutant allele frequencies in the Turkish population.
Article
The field of pharmacogenomics was initiated in the 1950s and began to thrive after the completion of the human genome project 10 years ago. Thus far, more than 100 drug labels and clinical guidelines referring to pharmacogenomic biomarkers have been published, and several key pharmacogenomic markers for either drug safety or efficacy have been identified and subsequently adopted in clinical practice as pre-treatment genetic tests. However, a tremendous variation of genetic backgrounds exists between different ethnic groups. The application of pharmacogenomics in the Chinese population is still a long way off, since the published guidelines issued by the organizations such as US Food and Drug Administration require further confirmation in the Chinese population. This review highlights important pharmacogenomic discoveries in the Chinese population and compares the Chinese population with other nations regarding the pharmacogenomics of five most commonly used drugs, ie, tacrolimus, cyclosporine A, warfarin, cyclophosphamide and azathioprine.
Article
Purpose of review: Pharmacogenomics is the study of differences in drug response on the basis of individual genetic background. With rapidly advancing genomic technologies and decreased costs of genotyping, the field of pharmacogenomics continues to develop. Application to patients with kidney disease provides growing opportunities for improving drug therapy. Recent findings: Pharmacogenomics studies are lacking in patients with chronic kidney disease and dialysis, but are abundant in the kidney transplant field. A potentially clinically actionable genetic variant exists in the CYP3A5 gene, with the initial tacrolimus dose selection being optimized based on CYP3A5 genotype. Although many pharmacogenomics studies have focused on transplant immunosuppression pharmacokinetics, an expanding literature on pharmacodynamic outcomes, such as calcineurin inhibitor toxicity and new onset diabetes, is providing new information on patients at risk. Summary: Appropriately powered pharmacogenomics studies with well-defined phenotypes are needed to validate existing studies and unearth new findings in patients with kidney disease, especially the chronic kidney disease and dialysis population.
Article
Cytochrome P450 3A (CYP3A4) is involved in the metabolism of more drugs in clinical use than any other foreign compound metabolizing enzyme in humans. Recently, there is increasing evidence that variants in the CYP3A4 gene have functional significance and - in rare cases - lead to loss of activity implying tremendous consequences for the patients. This review article highlights the functional consequences all CYP3A4 variants, recognized by the Human Cytochrome P450 (CYP) Allele Nomenclature Database.Clinical Pharmacology & Therapeutics (2014); Accepted article preview online 13 June 2014; doi:10.1038/clpt.2014.129.