Open access

Pharmacogenetics and Renal Transplantation

Written By

Chi Yuen Cheung

Submitted: 29 October 2010 Published: 23 August 2011

DOI: 10.5772/18882

From the Edited Volume

Kidney Transplantation - New Perspectives

Edited by Dr.(Md) Magdalena Trzcinska

Chapter metrics overview

2,148 Chapter Downloads

View Full Metrics

1. Introduction

Different patients receiving the same dose of a drug can exhibit a wide range of blood concentrations. This is a main concern regarding the immunosuppressive drugs because of their narrow therapeutic indices. Subtherapeutic blood concentrations are associated with an increased risk of acute rejection while overdosing may increase the risk of over-immunosuppression, with subsequent increased risk of infection and malignancies. Moreover, there are also numerous drug-specific adverse effects. At present, therapeutic drug monitoring (TDM) is used to address the issue of inter-individual variation in pharmacokinetics of immunosuppressive agents. However, TDM cannot influence drug exposure during the first 2-3 days after transplantation. Thus many patients experience a significant delay in achieving target blood concentrations, significantly increasing the risk of acute graft rejection (Clase et al., 2002; Undre et al., 1999). The narrow therapeutic index of these agents prevents use of a strategy based on a higher initial dose for all patients. As a result, there is a clear need for a strategy to allow individualized immunosuppressive drug dosing in the immediate post-transplant period.

After administration, the drug is absorbed and distributed to its site of action, where it interacts with targets such as receptors and enzymes, undergoes metabolism, and is then excreted. Each of these processes might involve clinically significant genetic variations. In the general population, it is estimated that genetics accounts for 20% to 95% of the variability in drug disposition and effects (Kalow et al., 1998). Other non-genetic factors, such as hepatic or renal function, drug interactions, and nature of diseases will also influence the effects of medication. With the introduction of tools for genomic analysis, the DNA variants responsible for the differences in drug-metabolizing capacities were discovered. Subsequently, individuals can be characterized as efficient or poor metabolizers for a particular drug based on their gene polymorphisms encoding protein variants that metabolize the drug. The genetic polymorphisms in drug-metabolizing enzymes together with drug transporters and drug receptors led to the hypothesis that genetic factors may be implicated in the inter-individual variability of the pharmacokinetic or pharmacodynamic characteristics of immunosuppressive drugs, major side effects, and immunologic risks.

By definition, pharmacogenetics is the study of genetic variation that gives rise to differing responses to drugs, whereas pharmacogenomics is the application of genomic technologies to drug discovery (Goldstein et al., 2003; Phillips & Van Bebber, 2005; Stoughton & Friend, 2005). Nowadays, the two terms are often used interchangeably. The promising role of pharmacogenetics and pharmacogenomics illustrates the concept of personalized medicine, in which the characterization of the patients’ genotype may help to identify the right drug and dose for each patient. The wider use of pharmacogenetic testing is also currently viewed as an important tool to improve drug safety and efficacy (Hesselink et al., 2005a; Shastry, 2005).

Advertisement

2. Pharmacokinetics and pharmacogenetics

The relationship between genetic variation and drug response was first observed in the 1950s for drugs metabolized by N-acetyltransferase. Based on the blood concentrations of drugs metabolized by this enzyme, patients were classified into “fast and slow acetylators”. However, the molecular genetic basis for such inherited traits began to be elucidated only in the late 1980s, with the initial cloning and characterization of polymorphic human genes encoding for drug metabolizing enzymes.

Whether pharmacogenetics can be applied successfully in daily clinical practice depends on our understanding of the enzymes which metabolize the drugs. When a drug is administered, it is first absorbed in the intestine, and different proteins in the intestinal wall can determine the amount that finally passes into the blood. The calcineurin inhibitors (CNIs), the mammalian target of rapamycin (mTOR) inhibitors and corticosteroids are all metabolized by the oxidative enzymes in the cytochrome (CYP) 3A family (Dai et al., 2004, 2006; Kamdem et al., 2005) and are substrates for the P-glycoprotein (P-gp) (Miller et al., 1997; Saeki et al., 1993). They work together to form an active barrier to drug absorption, limiting the oral bioavailability of the CNIs and mTOR inhibitors (Zhang & Benet, 2001). Some drugs require binding proteins before their delivery to the targets, such as CNIs that bind to immunophilins as part of their mechanism of action. As a result, only a proportion of an administered drug can reach the target. In order to define the association between the genotypes and the pharmacokinetics of a drug, a group of individuals are given the same dose of a drug, and the blood concentrations will then be measured at different intervals (Dunn et al., 2001; Schiff et al., 2007). These individuals are genotyped for polymorphisms at certain candidate genes, and the association between the genotypes and the pharmacokinetics is subsequently statistically analyzed. The ultimate goal is to find out the gene variants that can help in predicting the pharmacokinetics of a drug, and identifying patients who need a higher dose to reach the desired blood concentration (Evans & McLeod, 2003; Mc Leod & Evans, 2001). The combined analysis of gene variants encoding the different proteins that mediate the drug action may help to determine the final dose necessary to obtain a pharmacological effect (Kruger et al., 2008). This complicated picture shows that a successful pharmacogenetics approach would require the genotyping of several genes in each patient.

Advertisement

3. P-glycoprotein

Many drugs, including some immunosuppressive agents, are pumped out of the endothelial cells by P-gp, encoded by adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) gene (formerly called multidrug resistance-1 [MDR1] gene) (Benet et al., 1999; Lown et al., 1997). One of the main functions of P-gp is to ensure the energy-dependent cellular efflux of substrates. The P-gp expression in the intestinal wall and in the proximal tubular cells of the kidneys suggests that it may have a role in the absorption and excretion of xenobiotics. In the gut, alteration in its expression, function, or both raises the absorption of its substrates (Ambudkar et al., 1999). Individuals with a genotype with high intrinsic P-gp activity have a lower proportion of the drug that reaches the blood stream compared with a genotype with less P-gp activity (Choudhuri & Klaassen, 2006). Various single nucleotide polymorphisms (SNPs) have been identified within the ABCB1 gene over the recent few years (Kim, 2002). An SNP, located in exon 26 (exon 26 3435C>T), was associated with variations in the intestinal expression and function of P-gp. However, this SNP is a silent polymorphism that does not result in any amino acid changes. It was suggested that it may be in linkage disequilibrium with other functional polymorphisms within the ABCB1 gene. A co-segregation of exon 26 3435T with the T allele of the non-synonymous exon 21 SNP (exon 21 2677G>T), resulting in an A893S amino acid change, and also with the T allele of the synonymous exon 12 SNP (exon 12 1236C>T) was reported. This disequilibrium led to haplotype analysis of the ABCB1 gene and identification of the links between the genomic variations represented by each haplotype on ABCB1 function. This approach takes into account the combination of SNPs present in an allele (Kim, 2002) and might be more predictive of changes in response to drugs than SNP-based approaches.

Advertisement

4. CYP3A

CYP3A proteins can be classified into families and subfamilies on the basis of amino acid sequence similarities. Members of the CYP3A subfamily are implicated in the metabolism of structurally diverse endobiotics, drugs, and protoxic or procarcinogenic molecules. Substantial inter-individual differences in CYP3A expression contribute greatly to variations in the oral bioavailability and systemic clearance of CYP3A substrates (Nebert & Russell, 2002). Human CYP3A activities reflect the heterogeneous expression of at least three CYP3A members, CYP3A4, CYP3A5, and CYP3A7, which are adjacent to each other on chromosome band 7q21. CYP3A7 is normally only expressed in fetal liver. CYP3A4 and CYP3A5 have been identified as the major enzymes responsible for the disposition of drugs (Sattler et al., 1992). In enterocytes, CYP3A4 and CYP3A5 are involved in intestinal metabolism, preventing systemic uptake of immunosuppressive agents; while in the liver, they provide a further layer contributing to first-pass metabolism, thus affecting the drug clearance. For example, CYP3A5 variants alter the dose requirement of tacrolimus (Tac). Since CYP3A5 is involved in Tac deactivation, patients with a genotype that encodes for lower enzyme activity would have an increase drug exposure. Thus a lower dose will be required to be within the target blood concentration. The normal (wild-type) sequences of CYP3A4 and CYP3A5 are designated as CYP3A4*1 and CYP3A5*1. The most frequent CYP3A4 SNP linked to different enzymatic activities is -392 A>G in the gene promoter. The -392 G allele (also called CYP3A4*1B) increased CYP3A4 expression in vitro (Rebbeck et al., 1998; Westlind et al., 1999). This SNP is common in individuals of African descent (30%–70%) but rare among whites (1%–10%) (Makeeva et al., 2008; Quaranta et al., 2006). On the other hand, the most important SNP of the CYP3A5 gene leading to the alteration of gene expression and enzymatic activity is the SNP 6986 A>G in intron 3. Analysis revealed that only individuals with at least one CYP3A5*1 allele (A at position 6986) produce high levels of full-length CYP3A5 mRNA and express CYP3A5, which then accounts for at least 50% of the total CYP3A content. Those with the CYP3A5*3 allele (G at position 6986) display sequence variability in intron 3 that creates a cryptic splice site and encodes an aberrantly spliced mRNA with a premature stop codon, leading to the absence of protein expression (Kuehl et al., 2001).

Advertisement

5. Tacrolimus

Among the factors which have been investigated for the possible influence on CNIs pharmacokinetics, polymorphisms in genes coding for CYP3A (3A4 and 3A5) and P-gp received much attention. The impact of CYP3A4, CYP3A5, and MDR1 SNPs on Tac pharmacokinetics has been analyzed extensively in recent years. CYP3A5 expressers, who are carriers of at least one CYP3A5*1 allele, would have higher Tac clearance and lower dose-normalized C0 at different times after renal transplantation compared with CYP3A5*3 homozygotes (Haufroid et al., 2004; Haufroid et al., 2006; Hesselink et al., 2003; Mac, Phee et al., 2005; Mourad et al., 2006; Roy et al., 2006; Tada et al., 2005; Thervet et al., 2003; Tsuchiya et al., 2004; Zhang et al., 2005; Zhao et al., 2005). A study of 118 kidney recipients examined the relationship between CYP3A5*1/*3 and Tac dose-normalized concentrations at 1 week, 1 month, and 3 months post-transplantation. At 1 week, the mean dose-normalized blood concentration was significantly lower in CYP3A5*1 carriers (33 ng/mL per mg/kg/day) compared with CYP3A5*3 homozygotes (102 ng/mL per mg/kg/day). This difference remained significant at 1 month and 3 months post-transplant (Zhang et al., 2005). A temporal change in Tac oral bioavailability has been reported by Kuypers et al. The dose-normalized exposure to Tac increased progressively over a 5-year period in individuals predicted to be CYP3A5 non-expressers but not in CYP3A5 expressers (Kuypers et al., 2007). The frequency of these alleles depends on the population studies: the CYP3A5*1 allele is present in 15 % of the Caucasian, 45% of the African-American (Kuehl et al., 2001), and 25% of the Chinese population (Balram et al., 2003). Since many genetic differences exist between races, it is also important to examine whether the described polymorphisms are related to differences in pharmacokinetic and dosing of Tac in different population. In a study of 103 stable Chinese renal transplant recipients (Cheung et al., 2006), a strong significant genetic effect between CYP3A5*3 polymorphism and both the dose-normalized AUC0-12 and the daily Tac dose has been demonstrated. In fact, the CYP3A5*3 polymorphism may explain 35.3% of the variation in the daily Tac dose observed in the renal transplant recipients. In another study involving Caucasian population (Op den Buijsch et al., 2007), significantly higher dose-normalized C0, dose-normalized AUC0-12 and dose-normalized peak concentrations (Cmax) is demonstrated in carriers of the CYP3A5*3 allele in both early and late post renal transplant recipient groups than in patients homozygous for CYP3A5*1. In their centre, a complete Tac pharmacokinetic profile was usually requested early for patients who failed to achieve the target Tac concentration shortly after transplantation. Since the CYP3A5*1 allele was over-represented in this early phase group in their study, the authors concluded that renal transplant recipients carrying this allele have more difficulties in achieving and maintaining Tac concentrations compared to homozygous carriers of the CYP3A5*3 variant. This might be of importance for the Chinese population in which the CYP3A5*1 allele has a much higher prevalence than in the Caucasian population.

CYP3A5 is closely linked to the CYP3AP1 pseudogene, and the CYP3AP1*1 allele (-44 G) was in strong linkage disequilibrium with the low-expression CYP3A5*3 allele. In fact, the CYP3AP1 genotype resembles the CYP3A5 genotype (Kuehl et al., 2001). A significant association was found between the CYP3AP1 polymorphism and three parameters, namely, drug concentrations during the first week post-transplant, time to reach the target blood concentrations, and the risk of early allograft rejection (Mac , Phee et al., 2002, 2004).

On the other hand, the impact of the CYP3A4 and ABCB1 polymorphisms in Tac pharmacokinetics is not clear. Most studies failed to show any association between CYP3A4 gene polymorphisms and Tac pharmacokinetics although at least one group has reported lower C0 concentrations at 3 and 12 months post-transplantation in carriers of the CYP3A4*1B allele than in the CYP3A4*1 homozygotes (Hesselink et al., 2003). Similarly, there has been conflicting pubished data about the influence of the polymorphisms of the ABCB1 system on the pharmacokinetics of Tac. A number of studies have reported that there seems to be no association between the ABCB1 polymorphisms and Tac dose-normalized C0 (Haufroid et al., 2004; Hebert et al., 2003; Hesselink et al., 2003; Mai et al., 2004; Zhang et al., 2005) or the dose-normalized AUC (Tada et al., 2005; Tsuchiya et al., 2004). However, some studies found a correlation between individual ABCB1 polymorphisms and a higher Tac dose (Mac, Phee et al., 2002; Zheng et al., 2003; Zheng et al., 2004). Carriers of the 2677T or the 3435T MDR1 alleles showed higher dose-normalized Tac C0 concentrations compared with 2677 G-homozygous (GG) and 3435 C-homozygous (CC) patients (Akbas et al., 2006; Anglicheau et al., 2003; Li et al., 2006). Because of differences in design and study populations it might be that a polymorphism, that has a minor influence on the Tac blood concentrations, demonstrates contrasting results among these studies. Moreover, there are several SNPs that may occur together, resulting in different haplotypes. The correlation of these haplotypes with the pharmacokinetics of Tac has not yet been described extensively. In one study, a correlation between the ABCB1 1236C-2677G-3435C haplotype and a higher Tac dose was found (Anglicheau et al., 2003). However, in another study of 63 Caucasian renal transplant recipients with a complete 9-point 12-hour AUC of Tac, 3 SNPs in the ABCB1 system were genotyped. Neither the individual ABCB1 polymorphisms nor the ABCB1 haplotypes were associated with any pharmacokinetic parameter (Op den Buijsch et al., 2007). On the other hand, in a study of Chinese renal transplant recipients (Cheung et al., 2006), individuals carrying the 2677TT or 3435TT genotype has a significantly lower dose-normalized AUC0-12, but no correlation was found between ABCB1 system haplotype and dose-normalized AUC0-12. In multiple regression analysis the 2677TT and 3435TT genotype was not shown to be significant if the CYP3A polymorphism was included. Therefore, the published correlation of SNPs of the ABCB1 system with dose-normalized AUC of Tac might be related to genetic linkage of the ABCB1 system with other polymorphisms, such as the CYP3A system. Individuals with the mutant genotype appear to be over-represented in the CYP3A5 expresser population and under-represented in the CYP3A5 non-expresser population. Thus the interaction between P-gp and CYP3A5 further complicates the analysis of interaction between ABCB1 genotype and Tac pharmacokinetics.

Despite the fact that expression of CYP3A5 results in higher Tac dose requirements and significant delay in achieving target blood concentrations early after transplantation, most of the previous studies failed to identify the association of the genetic polymorphisms with increased incidence of acute rejection (Hesselink et al., 2008; Mac, Phee et al., 2004). However, in a recent prospective study of 62 patients who underwent 10-day scheduled renal graft biopsy, significantly higher overall incidences of early T-cell-mediated rejection of at least Banff grade 1 in severity were detected in CYP3A5 expressers. The severity was also associated with the CYP3A5 genotypes. Moreover, the estimated glomerular filtration rate in CYP3A5 expressers was lower than that of the non-expressers until one month after transplantation (Min et al., 2010). However, further large-scale long-term outcome studies are necessary to confirm the clinical relevance of the findings.

Advertisement

6. Cyclosporine

Healthy volunteers who are CYP3A5*1 carriers had a lower cyclosporine (CsA) AUC when compared with individuals with homozygous for CYP3A5*3 (Min et al., 2004). Moreover, renal transplant recipients who carry CYP3A5*1 were also found to exhibit lower dose-normalized CsA C0 (Haufroid, et al., 2004). However, this association has not been confirmed by most authors (Anglicheau et al., 2004; Hesselink et al., 2003, 2004; Kreutz et al., 2004). This is strange because the clear influence of CYP3A5 genotype on Tac absorption could not be shown similarly in CsA (as both are CNIs). A possible explanation is that the molar dose of CsA administered is approximately 30-fold higher than that for Tac and it blocks a saturable barrier to drug absorption more effectively than for Tac (Higgins et al., 1999). The association between CYP3A4 SNPs and CsA pharmacokinetics is also controversial. The CYP3A4*1B allele has been linked to significantly higher CsA clearance compared with wild-type homozygotes (Hesselink et al., 2004; Min & Ellingrod, 2003). However, this association was not confirmed in other studies (Rivory et al., 2000; von Ahsen et al., 2001). In whites, the 3A4*1B occurs at a low frequency (Coto & Tavira, 2009), and recruitment of the minimum number of carriers to reach statistical significance is difficult.

The association between ABCB1 SNPs and CsA pharmacokinetics is also controversial. In a study involving 106 renal transplant recipients, carriers of the ABCB1 1236 wild-type allele had a lower dose-normalized Cmax and lower increased AUC when compared with the 1236 T allele homozygotes (Anglicheau et al., 2004). In another study with 69 renal transplant recipients, a significantly lower AUC and C2 in carriers of the ABCB1 3435 T allele was shown at day 3 post-transplant but the difference did not remain significant at 1 month (Foote et al., 2007). However, most of the large studies did not find an association between any of the ABCB1 polymorphisms and CsA pharmacokinetics (Haufroid et al., 2004; Kuzuya et al., 2003; Mai et al., 2003). On the other hand, the influence of ABCB1 genotype on pharmacodynamics seems more compelling. The incidence of CsA nephrotoxicity was significantly higher when the donor had the ABCB1 3435TT genotype (Hauser et al., 2005). This is consistent with the hypothesis that local levels of P-gp expression in renal tubular epithelial cells can explain the susceptibility to CNI nephrotoxicity. It has been shown that lower levels of P-gp expression were found in renal biopsies in patients with CNI nephrotoxicity (Joy et al., 2005). CsA nephrotoxicity is also exacerbated by concomitant use of sirolimus, which can be explained by the inhibitory effect of sirolimus on P-gp-mediated efflux and subsequent increased cellular concentration of CsA (Anglicheau et al., 2006). Moreover, another study also found that ABCB1 polymorphisms in donors influence long-term graft outcome. The donor ABCB1 haplotype 1236T/2677T/3435T was significantly associated with increase graft loss, acute rejection episodes and greater decrease in renal function (Woillard et al., 2010).

In addition to CYP and ABCB1 genes, other gene variants can also affect CNIs pharmacokinetics and clinical outcomes. In a study of subpopulation of patients participating in the CAESAR study, 4 gene polymorphisms including ABCB1 G2677T/A, IMPDH2 T3757C, IL-10 C-592A and TNF-alpha G-308A demonstrated a statistically significant association with biopsy-proven acute rejection at 12 months post-transplant (Grinyo et al., 2008). CsA action is mediated by its binding to the cyclophilins (Cyp). In a study involving 290 kidney-transplanted patients, the effect of two CypA polymorphisms on CsA pharmacokinetics and clinical outcomes was analyzed (Moscoso-Solorzano et al., 2008). In vitro studies showed that a promoter SNP (-11 G/C) affected gene expression but was not related to differences in C0 and C2 dose-normalized levels. However, an association between the high expression allele and nephrotoxicity was found but these results need further confirmation.

Advertisement

7. Azathioprine

Most of the pharmacogenetic traits first identified were discovered by phenotypic analysis detecting a bi- or trimodality of an enzymatic activity (Weinshilboum, 2003). Azathioprine, metabolized in part by S-methylation catalyzed by the enzyme thiopurine methyltransferase (TPMT), is an example. Large inter-individual differences were reported in TPMT activity, which was found to be inherited in an autosomal codominant fashion (Weinshilboum et al., 1999). When individuals with low or undetectable TPMT activity received standard doses of azathioprine, they had high concentrations of the active metabolites 6-thioguanine nucleotides and drug-induced myelosuppression. On the other hand, azathioprine efficacy will be reduced in patients with very high levels of TPMT activity which can be attributed to its rapid metabolization (Chocair et al., 1992; Soria-Royer et al., 1993). TPMT activity correlates with both short- and long-term results after renal transplantation (Thervet et al., 2001). Subsequently, these variations in TPMT activity were shown to be attributed to genetic polymorphisms within the TPMT gene. At present, 20 variant alleles (TPMT*2 - *18) have been identified, which are associated with decreased activity when compared with the TPMT*1 wild type allele. TPMT*3A, the most common variant allele responsible for low TPMT activity in whites, encodes a protein with two single nucleotide polymorphisms (SNP), G460A in exon 7 and A719G in exon 10, leading to modifications in the amino acid sequence. The phenotypic test for TPMT activity determination in red blood cells and, subsequently, DNA-based tests, were among the first pharmacogenetic tests to be used in clinical practice.

Advertisement

8. Mycophenolic acid

Mycophenolic acid (MPA) is the active derivative of the prodrug mycophenolate mofetil but MPA itself is also available as enteric coated sodium salt tablet. MPA is metabolized by uridine-glucuronyl-transferase (UGT), primarily UGT1A8 and 1A9, to the inactive metabolite 7-O-glucuronide (MPAG). MPAG is primarily excreted by kidneys, although a proportion is secreted in bile by the drug efflux pump multidrug resistance associated protein 2 (MRP2), now also called ABCC2. Deconjugation by intestinal bacterial flora results in a second peak of absorption at 6 to 8 hours due to enterohepatic recirculation. This second peak accounts for 30-50% of the total AUC. As a result, co-administration with medication that inhibit ABCC2, such as CsA, can result in a significantly reduced MPA exposure (Hesselink et al., 2005b). Several SNPs have been found in the ABCC2 gene (Ito et al., 2001). It has been found that ABCC2 C-24T and C-3972T polymorphisms can protect the renal transplant recipients from a decrease in MPA exposure associated with mild liver dysfunction. Moreover, C-24T SNP was associated with significantly high dose-normalized MPA trough levels at steady state (Naesens et al., 2006).

Polymorphisms have also been identified in both UGT1A9 and 1A8 genes. In vitro studies have shown that polymorphisms in the UGT1A9 gene result in significant alteration of the UGT enzymatic activity. Two polymorphisms in the promoter region of the UGT1A9 gene, namely C-275T>A and C-2152C>T, result in higher MPA glucuronidation rates (Girard et al., 2004). It has been demonstrated that in renal transplant recipients, carriers of either or both polymorphisms had lower MPA AUC and C0 (Johnson et al, 2008; Kuypers et al., 2005; van Schaik et al., 2009). On the other hand, UGT1A8*3 (P277C>Y) polymorphism results in an approximately 30-fold reduction in MPAG formation (Bernard et al., 2006).

MPA inhibits inosine monophosphate dehydrogenase (IMPDH), a key enzyme in the pathway for purine synthesis.. The two isoforms of IMPDH, IMPHD1 and IMPDH2, have similar enzymatic activity. Individuals with low levels of lymphocyte IMPDH activity are more likely to experience toxicity with MPA and those with high levels are more likely to have rejection (Glander et al., 2004). There were different variants of the IMPDH2 gene and the 263L>F variant resulted in a reduction in enzyme activity to 10% of the wild-type (Wang et al., 2007). Several mutants of IMPDH had decreased affinity for MPA (Farazi et al., 1997). However, there are no clinical associations for this SNP. On the other hand, 2 SNPs in the IMPDH1 gene, namely +125G>A and -106G>A, were found to be associated with increase risk of rejection after kidney transplantation but the underlying mechanism remains uncertain (Wang et al., 2008). Despite of availability of different candidate genes, there are still insufficient data to support the use of pharmacogenetic strategy for mycophenolate.

Advertisement

9. Mammalian target of rapamycin inhibitors

There is only limited data concerning the pharmacogenetics of mTOR inhibitors. Usually it takes longer time for sirolimus to achieve desired therapeutic range because of its long half-life (approximately 60 hours). Thus use of pharmacogenetic strategy seems to be an ideal option for sirolimus dosage adjustment. However, the data in literature is controversial. While there were studies showing reduced oral bioavailability of sirolimus in CYP3A5 expressors (Anglicheau et al., 2005; Le Meur et al., 2006), similar association was not found in other studies (Mourad et al., 2005; Renders et al., 2007). Moreover, none of the studies could show the influence of ABCB1 genotype on sirolimus exposure (Anglicheau et al., 2005; Mourad et al., 2005; Renders et al., 2007). As a result, pharmacogenetics is still not suitable for sirolimus dosing.

Advertisement

10. Conclusion and future perspective

Currently TDM is the gold standard for monitoring and titration of immunosuppressive drugs in order to ensure adequate immunosuppression but avoid side effects. However, many patients experience significant delay in achieving therapeutic blood concentrations, resulting in a higher risk of acute rejection. As a result, selection of the best drug with an accurate dose is important. Pharmacogenetics have generated considerable enthusiasm in transplantation medicine in recent years and it is widely believed that “personalized medicine” is the ultimate goal of use of immunosuppressive drugs. Although many genetic factors have been shown to influence pharmacokinetics for the immunosuppressive drugs, only CYP3A5 genotyping may help to guide individual tacrolimus dosing in clinical practice. Moreover, the focus of pharmacogenetic studies has recently shifted from pharmacokinetics to transplantation outcomes, such as renal allograft dysfunction. Although there is substantial evidence that intrarenally expressed ABCB1 is implicated in the pathogenesis of CNI nephrotoxicity, there is no direct evidence in human that the association between ABCB1 genotype and CNI nephrotoxicity is indeed caused by higher intrarenal concentration of CNIs (Hesselink et al., 2010). Further prospective and intervention studies involving genetic profile and transplant outcome are required for recommendation of widespread use of pharmacogenetic testing in routine clinical practice.

References

  1. 1. AkbasS. H.BilgenT.KeserI.TuncerM.YucetinL.TosunO.GultekinM.LuleciG.2006The effect of MDR1 (ABCB1) polymorphism on the pharmacokinetic of tacrolimus in Turkish renal transplant recipients. Transplant Proc, 38512901292
  2. 2. AmbudkarS. V.DeyS.HrycynaC. A.RamachandraM.PastanI.GottesmanM. M.1999Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol, 39361398
  3. 3. AnglicheauD.VerstuyftC. Laurent-PuigP.BecquemontL. SchlageterM.H. CassinatB. BeauneP. LegendreC. ThervetE. (2003). Association of the multidrug resistance-1 gene single-nucleotide polymorphisms with the tacrolimus dose requirements in renal transplant recipients. J Am Soc Nephrol, 71418891896
  4. 4. AnglicheauD.ThervetE.EtienneI.HuraultB.De LignyY.Le MeurG.TouchardM.BuchlerP.Laurent-PuigD.TregouetP.BeauneA.DalyC.LegendreP.Marquet2004CYP3A5 and MDR1 genetic polymorphisms and cyclosporine pharmacokinetics after renal transplantation. Clin Pharmacol Ther, 755422433
  5. 5. AnglicheauD.le CorreD.LechatonS.Laurent-PuigP.KreisH.BeauneP.LegendreC.ThervetE.2005Consequences of genetic polymorphisms for sirolimus requirements after renal transplant in patients on primary sirolimus therapy. Am J Transplant, 53595603
  6. 6. AnglicheauD.PalletN.RabantM.MarquetP.CassinatB.MeriaP.BeauneP.LegendreC.ThervetE.2006Role of P-glycoprotein in cyclosporine cytotoxicity in the cyclosporine-sirolimus interaction. Kidney Int, 70610191025
  7. 7. BalramC.ZhouQ.CheungY. B.LeeE. J.2003CYP3A5*3 and *6 single nucleotide polymorphisms in three distinct Asian populations. Eur J Clin Pharmacol, 592123126
  8. 8. BenetL. Z.IzumiT.ZhangY.SilvermanJ. A.WacherV. J.1999Intestinal MDR transport proteins and P-450 enzymes as barriers to oral drug delivery. J Control Release, 621-22531
  9. 9. BernardO.TojcicJ.JournaultK.PerusseL.GuillemetteC.2006Influence of nonsynonymous polymorphisms of UGT1A8 and UGT2B7 metabolizing enzymes on the formation of phenolic and acyl glucuronides of mycophenolic acid. Drug Metab Dispos, 34915391545
  10. 10. CheungC. Y.OpR. A.denBuijsch. K. M.WongH. W.ChanK. F.ChauC. S.LiK. T.LeungT. H.KwanJ. E.de VriesP. A.WijnenM. P.van Dieijen-VisserO.Bekers2006Influence of different allelic variants of the cytochrome 3A and adenosine triphosphate-binding cassette B1 gene on the tacrolimus pharmacokinetic profile of Chinese renal transplant recipients. Pharmacogenomics, 74563574
  11. 11. ChocairP. R.DuleyJ. A.SimmondsH. A.CameronJ. S.1992The importance of thiopurine methyltransferase activity for the use of azathioprine in transplant recipients. Transplantation, 53510511056
  12. 12. ChoudhuriS.KlaassenC. D.2006Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int J Toxicol, 254231259
  13. 13. ClaseC. M.MahalatiK.KiberdB. A.LawenJ. G.WestK. A.FraserA. D.BelitskyP.2002Adequate early cyclosporin exposure is critical to prevent renal allograft rejection: patients monitored by absorption profiling. Am J Transplant, 28789795
  14. 14. CotoE.TaviraB.2009Pharmacogenetics of calcineurin inhibitors in renal transplantation. Transplantation, 883SS62S67
  15. 15. DaiY.IwanagaK.LinY.S. HebertM.F. DavisC.L. HuangW. KharaschE.D. ThummelK.E. (2004). In vitro metabolism of cyclosporine A by human kidney CYP3A5. Biochem Pharmacol, 96818891902
  16. 16. DaiY.HebertM. F.IsoherranenN.DavisC. L.MarshC.ShenD. D.ThummelK. E.2006Effect of CYP3A5 polymorphism on tacrolimus metabolic clearance in vitro. Drug Metab Dispos, 345836847
  17. 17. DunnC.J. WagstaffA.J. PerryC.M. PloskerG.L. GoaK.L. (2001). Cyclosporin: An updated review of the pharmacokinetic properties, clinical efficacy and tolerability of a microemulsion-based formulation (Neoral)1 in organ transplantation. Drugs, 136119572016
  18. 18. EvansW. E.Mc LeodH. L.2003Pharmacogenomics-Drug disposition, drug targets, and side effects. N Engl J Med, 3486538549
  19. 19. FaraziT.LeichmanJ.HarrisT.CahoonM.HedstromL.1997Isolation and characterization of mycophenolic acid-resistant mutants of inosine-5’-monophosphate dehydrogenase. J Biol Chem, 2722961965
  20. 20. FooteC. J.GreerW.KiberdB.FraserA.LawenJ.NashanB.BelitskyP.2007Polymorphisms of multidrug resistance gene (MDR1) and cyclosporine absorption in de novo renal transplant patients. Transplantation, 831013801384
  21. 21. GirardH.CourtM. H.BernardO.FortierL. C.VilleneuveL.HaoQ.GreenblattD. J.vonL. L.MoltkeL.PerussedC.Guillemette2004Identification of common polymorphisms in the promoter of the UGT1A9 gene: evidence that UGT1A9 protein and activity levels are strongly genetically controlled in the liver. Pharmacogenetics, 148501515
  22. 22. GlanderP.HambachP.BraunK.P. FritscheL.GiessingM. MaiI.EineckeG. WaiserJ. NeumayerH.H. BuddeK. (2004). Pre-transplant inosine monophosphate dehydrogenase activity is associated with clinical outcome after renal transplantation. Am j Transplant, 12420452051
  23. 23. GoldsteinD. B.TateS. K.SisodiyaS. M.2003Pharmacogenetics goes genomic. Nat Rev Genet, 412937947
  24. 24. GrinyoJ.VanrenterghemY.NashanB.VincentiF.EkbergH.LindpaintnerK.RashfordM.Nasmyth-MillerC.VoulgariA.SpleissO.TrumanM.EssiouxL.2008Association of four DNA polymorphisms with acute rejection after kidney transplantation. Transplantation, 219879891
  25. 25. HaufroidV.MouradM.Van KerckhoveV.WawrzyniakJ.De MeyerM.EddourD. C.MalaiseJ.LisonD.SquiffletJ. P.WallemacqP.2004The effect of CYP3A5 and MDR1 (ABCB1) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood levels in stable renal transplant patients. Pharmacogenetics, 143147154
  26. 26. HaufroidV.WallemacqP.Van KerckhoveV.ElensL.De MeyerM.EddourD. C.MalaiseJ.LisonD.MouradM.2006CYP3A5 and ABCB1 polymorphisms and tacrolimus pharmacokinetics in renal transplant candidates: Guidelines from an experimental study. Am J Transplant, 61127062713
  27. 27. HauserI. A.SchaeffelerE.GauerS.ScheuermannE. H.WegnerB.GossmannJ.AckermannH.SeidlC.HocherB.ZangerU. M.GeigerH.EichelbaumM.SchwabM.2005ABCB1 genotype of the donor but not of the recipient is a major risk factor for cyclosporine-related nephrotoxicity after renal transplantation. J Am Soc Nephrol, 16515011511
  28. 28. HebertM. F.DowlingA. L.GierwatowskiC.LinY. S.EdwardsK. L.DavisC. L.MarshC. L.SchuetzE. G.ThummelK. E.2003Association between ABCB1 (multidrug resistance transporter) genotype and post-liver transplantation renal dysfunction in patients receiving calcineurin inhibitors. Pharmacogenetics, 1311661674
  29. 29. HesselinkD. A.van SchaikR. H.van der HeidenI. P.van der WerfM.GregoorP. J.LindemansJ.WeimarW.van GelderT.2003Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Clin Pharmacol Ther, 743245254
  30. 30. HesselinkD. A.van GelderT.van SchaikR. H.BalkA. H.van der HeidenI. P.van DamT.van der WerfM.WeimarW.MathotR. A.2004Population 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, 766545556
  31. 31. HesselinkD. A.van GelderT.van SchaikR. H.2005aThe pharmacogenetics of calcineurin inhibitors: One step closer toward individualized immunosuppression? Pharmacogenomics, 64323337
  32. 32. HesselinkD. A.van HestR. M.MathotR. A.BonthuisF.WeimarW.de BruinR. W.van GelderT.2005bCyclosporine interacts with mycophenolic acid by inhibiting the multidrug resistance-associated protein 2. Am J Transplant, 55987994
  33. 33. HesselinkD. A.van SchaikR. H.van AgterenM.de FijterJ. W.HartmannA.ZeierM.BuddeK.KuypersD. R.PisarskiP.Le MeurY.MamelokR. D.van GelderT.2008CYP3A5 genotype is not associated with a higher risk of acute rejection in tacrolimus-treated renal transplant recipients. Pharmacogenet Genomics, 184339348
  34. 34. HesselinkD. A.BouamarR.van GelderT.2010The pharmacogenetics of calcineurin inhibitor-related nephrotoxicity. Ther Drug Monit, 324387393
  35. 35. HigginsR. M.MorlidgeC.MageeP.Mc Diarmaid-GordonA.LamF. T.KashiH.1999Conversion between cyclosporin and tacrolimus--30-fold dose prediction. Nephrol Dial Transplant, 1461609
  36. 36. ItoS.IeiriI.TanabeM.SuzukiA.HiguchiS.OtsuboK.2001Polymorphism of the ABC transporter genes, MDR1, MRP1 and MRP2/cMOAT, in healthy Japanese subjects. Pharmacogenetics, 112175184
  37. 37. JohnsonL. A.OettingW. S.BasuS.PrausaS.MatasA.JacobsonP. A.2008Pharmacogenetic effect of the UGT polymorphisms on mycophenolate is modified by calcineurin inhibitors. Eur J Clin Pharmacol, 641110471056
  38. 38. JoyM. S.NickeleitV.HoganS. L.ThompsonB. D.FinnW. F.2005Calcineurin inhibitor-induced nephrotoxicity and renal expression of P-glycoprotein. Pharmacotherapy, 256779789
  39. 39. KamdemL. K.StreitF.ZangerU. M.BrockmollerJ.OellerichM.ArmstrongV. W.WojnowskiL.2005Contribution of CYP3A5 to the in vitro hepatic clearance of tacrolimus. Clin Chem, 51813741381
  40. 40. KalowW.TangB. K.EndrenyiL.1998Hypothesis: Comparisons of inter- and intra-individual variations can substitute for twin studies in drug research. Pharmacogenetics, 84283289
  41. 41. KimR. B.2002MDR1 single nucleotide polymorphisms: Multiplicity of haplotypes and functional consequences. Pharmacogenetics, 126425427
  42. 42. KreutzR.ZurcherH.KainS.MartusP.OffermannG.BeigeJ.2004The effect of variable CYP3A5 expression on cyclosporine dosing, blood pressure and long-term graft survival in renal transplant patients. Pharmacogenetics, 1410665671
  43. 43. KrügerB.SchröppelB.MurphyB. T.2008Genetic polymorphisms and the fate of the transplanted organ. Transplant Rev (Orlando) 222131140
  44. 44. KuehlP.ZhangJ.LinY.LambaJ.AssemM.SchuetzJ.WatkinsP. B.DalyA.WrightonS. A.HallS. D.MaurelP.RellingM.BrimerC.YasudaK.VenkataramananR.StromS.ThummelK.BoguskiM. S.SchuetzE.2001Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet, 274383391
  45. 45. KuypersD.R. NaesensM. VermeireS. VanrenterghemY. (2005). The impact of uridine diphosphate-glucuronosyltransferase 1A9 (UGT1A9) gene promoter region single-nucleotide polymorphisms T-275A and C-2152T on early mycophenolic acid dose-interval exposure in de novo renal allograft recipients. Clin Pharmacol Ther478351361
  46. 46. KuypersD. R.de JongeH.NaesensM.LerutE.VerbekeK.VanrenterghemY.2007CYP3A5 and CYP3A4 but not MDR1 single-nucleotide polymorphisms determine long-term tacrolimus disposition and drug-related nephrotoxicity in renal recipients. Clin Pharmacol Ther, 826711725
  47. 47. KuzuyaT.KobayashiT.MoriyamaN.NagasakaT.UchidaK.NakaoA.NabeshimaT.2003Amlodipine, but not MDR1 polymorphisms, alters the pharmacokinetics of cyclosporine A in Japanese kidney transplant recipients. Transplantation, 765865868
  48. 48. Le MeurY.DjebliN.SzelagJ. C.HoizeyG.ToupanoeO.RerolleJ. P.MarquetP.2006CYP3A5*3 influences sirolimus oral clearance in de novo and stable renal transplant recipients. Clin Pharmacol Ther, 8015160
  49. 49. LiD.GuiR.LiJ.HuangZ.NieX.2006Tacrolimus dosing in Chinese renal transplant patients is related to MDR1 gene C3435T polymorphisms. Transplant Proc, 38928502852
  50. 50. LownK. S.MayoR. R.LeichtmanA. B.HsiaoH. L.TurgeonD. K.Schmiedlin-RenP.BrownM. B.GuoW.RossiS. J.BenetL. Z.WatkinsP. B.1997Role of intestinal P-glycoprotein (mdr1) in interpatient variation in the oral bioavailability of cyclosporine. Clin Pharmacol Ther, 623248260
  51. 51. MacI. A.PheeS.FredericksT.TaiP.SyrrisN. D.CarterA.JohnstonL.GoldbergD. W.Holt2002Tacrolimus pharmacogenetics: Polymorphisms associated with expression of cytochrome p4503A5 and P-glycoprotein correlate with dose requirement. Transplantation, 741114861489
  52. 52. MacI. A.PheeS.FredericksT.TaiP.SyrrisN. D.CarterA.JohnstonL.GoldbergD. W.Holt2004The influence of pharmacogenetics on the time to achieve target tacrolimus concentrations after kidney transplantation. Am J Transplant, 46914919
  53. 53. MacI. A.PheeS.FredericksM.MohamedM.MoretonN. D.CarterA.JohnstonL.GoldbergD. W.Holt2005Tacrolimus pharmacogenetics: The CYP3A5*1 allele predicts low dose-normalized tacrolimus blood concentrations in whites and South Asians. Transplantation, 794499502
  54. 54. MaiI.StormerE.GoldammerM.JohneA.KrugerH.BuddeK.RootsI.2003MDR1 haplotypes do not affect the steady-state pharmacokinetics of cyclosporine in renal transplant patients. J Clin Pharmacol, 431011011107
  55. 55. MaiI.PerloffE. S.BauerS.GoldammerM.JohneA.FillerG.BuddeK.RootsI.2004MDR1 haplotypes derived from exons 21 and 26 do not affect the steady-state pharmacokinetics of tacrolimus in renal transplant patients. Br J Clin Pharmacol, 585548553
  56. 56. MakeevaO.StepanovV.PuzyrevV.GoldsteinD. B.GrossmanI.2008Global pharmacogenetics: Genetic substructure of Eurasian populations and its effect on variants of drug-metabolizing enzymes. Pharmacogenomics, 97847868
  57. 57. Mc LeodH. L.EvansW. E.2001Pharmacogenomics: Unlocking the human genome for better drug therapy. Annu Rev Pharmacol Toxicol, 41101121
  58. 58. MillerD. S.FrickerG.DreweJ.1997P-glycoprotein-mediated transport of a fluorescent rapamycin derivative in renal proximal tubule. J Pharmacol Exp Ther, 2821440444
  59. 59. MinD. I.EllingrodV. L.2003Association of the CYP3A4*1B 5 flanking region polymorphism with cyclosporine pharmacokinetics in healthy subjects. Ther Drug Monit, 253305309
  60. 60. MinD. I.EllingrodV. L.MarshS.Mc LeodH.2004CYP3A5 polymorphism and the ethnic differences in cyclosporine pharmacokinetics in healthy subjects 1. Ther Drug Monit, 265524528
  61. 61. MinS. I.KimS. Y.AhnS. H.MinS. K.KimS. H.KimY. S.MoonK. C.OhJ. M.KimS. J.HaJ.2010CYP3A5*1 allele: impacts on early acute rejection and graft function in tacrolimus-based renal transplant recipients. Transplantation, 901213941400
  62. 62. Moscoso-SolorzanoG. T.OrtegaF.RodríguezI.Garcia-CastroM.GomezE.Diaz-CorteC.BaltarJ. M.AlvarezV.OrtizA.CotoE.2008A search for cyclophilin-A gene variants in cyclosporine A-treated renal transplanted patients. Clin Transplant, 226722729
  63. 63. MouradM.MouradG.WallemacqP.GarrigueV.Van BellingenC.Van KerkhoveV.De MeyerM.MalaiseJ.ChaibD.LisonE.D.SquiffletJ. P.HaufroidV.2005Sirolimus and tacrolimus trough concentrations and dose requirements after kidney transplantation in relation to CYP3A5 and MDR1 polymorphisms and steroids. Transplantation, 807977984
  64. 64. MouradM.WallemacqP.De MeyerM.BrandtD.Van KerkhoveV.MalaiseJ.ChaibD.EddourD.LisonV.Haufroid2006The influence of genetic polymorphisms of cytochrome P450 3A5 and ABCB1 on starting dose- and weight-standardized tacrolimus trough concentrations after kidney transplantation in relation to renal function. Clin Chem Lab Med, 441011921198
  65. 65. NaesensM.KuypersD. R.VerbekeK.VanrenterghemY.2006Multidrug resistance protein 2 genetic polymorphisms influence mycophenolic acid exposure in renal allograft recipients. Transplantation, 82810741084
  66. 66. NebertD. W.RussellD. W.2002Clinical importance of the cytochromes P450. Lancet, 360934011551162
  67. 67. OpR. A.denBuijsch. M. H.ChristiaansL. M.StolkJ. E.de VriesC. Y.CheungN. A.UndreJ. P.van HooffM. P.van Dieijen-VisserO.Bekers2007Tacrolimus pharmacokinetics and pharmacogenetics: influence of adenosine triphosphate-binding cassette B1 (ABCB1) and cytochrome (CYP) 3A polymorphisms. Fundam Clin Pharmacol, 214427435
  68. 68. PhillipsK. A.Van BebberS. L.2005Measuring the value of pharmacogenomics. Nat Rev Drug Discov, 46500509
  69. 69. QuarantaS.ChevalierD.AllorgeD.Lo-GuidiceJ. M.Migot-NabiasF.KenaniA.ImbenotteM.BrolyF.LacarelleB.LhermitteM.2006Ethnic differences in the distribution of CYP3A5 gene polymorphisms. Xenobiotica, 361211911200
  70. 70. RebbeckT. R.JaffeJ. M.WalkerA. H.WeinA. J.MalkowiczS. B.1998Modification of clinical presentation of prostate tumors by a novel genetic variant in CYP3A4. J Natl Cancer Inst, 901612251229
  71. 71. RendersL.FrismanM.UferM.MosyaginI.HaenischS.OttU.CaliebeA.DechantM.BraunF.KunzendorfU.CascorbiI.2007CYP3A5 genotype markedly influences the pharmacokinetics of tacrolimus and sirolimus in kidney transplant recipients. Clin Pharmacol Ther, 812228234
  72. 72. RivoryL. P.QinH.ClarkeS. J.ErisJ.DugginG.RayE.TrentR. J.BishopJ. F.2000Frequency of cytochrome P450 3A4 variant genotype in transplant population and lack of association with cyclosporin clearance. Eur J Clin Pharmacol, 565395398
  73. 73. RoyJ. N.BaramaA.PoirierC.VinetB.RogerM.2006Cyp3A4, Cyp3A5, and MDR-1genetic influences on tacrolimus pharmacokinetics in renal transplant recipients. Pharmacogenet Genomics, 169659665
  74. 74. SaekiT.UedaK.TanagawaraY.HoriR.KomanoT.1993Human P-glycoprotein transports cyclosporine A and FK506. J Biol Chem, 268960776080
  75. 75. SattlerM.GuengerichF. P.YunC. H.ChristiansU.SewingK. F.1992Cytochrome P-450 3A enzymes are responsible for biotransformation of FK506 and rapamycin in man and rat. Drug Metab Dispos, 205753761
  76. 76. SchiffJ.ColeE.CantarovichM.2007Therapeutic monitoring of calcineurin inhibitors for the nephrologist. Clin J Am Soc Nephrol, 22374384
  77. 77. ShastryB. S.2005Genetic diversity and new therapeutic concepts. J Hum Genet, 507321328
  78. 78. Soria-RoyerC.LegendreC.MirchevaJ.PremelS.BeauneP.KreisH.1993Thiopurine-methyl-transferase activity to assess azathioprine myelotoxicity in renal transplant recipients. Lancet, 341886015931594
  79. 79. StoughtonR. B.FriendS. H.2005How molecular profiling could revolutionize drug discovery. Nat Rev Drug Discov, 44345350
  80. 80. TadaH.TsuchiyaN.SatohS.KagayaH.LiZ.SatoK.MiuraM.SuzukiT.KatoT.HabuchiT.2005Impact of CYP3A5 and MDR1(ABCB1) C3435T polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Transplant Proc, 37417301732
  81. 81. TsuchiyaN.SatohS.TadaH.LiZ.OhyamaC.SatoK.SuzukiT.HabuchiT.KatoT.2004Influence of CYP3A5 and MDR1 (ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Transplantation, 78811821187
  82. 82. ThervetE.AnglicheauD.ToledanoN.HoullierA. M.NoelL. H.KreisH.BeauneP.LegendreC.2001Long-term results of TMPT activity monitoring in azathioprine-treated renal allograft recipients. J Am Soc Nephrol, 121170176
  83. 83. ThervetE.AnglicheauD.KingB.SchlageterM. H.CassinatB.BeauneP.LegendreC.DalyA. K.2003Impact of cytochrome p450 3A5 genetic polymorphism on tacrolimus doses and concentration-to-dose ratio in renal transplant recipients. Transplantation, 76812331235
  84. 84. UndreN. A.van HooffJ.ChristiaansM.VanrenterghemY.DonckJ.HeemanU.KohnleM.ZankerB.LandW.MoralesJ. M.AndresA.SchaferA.StevensonP.1999Low systemic exposure to tacrolimus correlates with acute rejection. Transplant Proc, 311-2296298
  85. 85. Van SchaikR.H. van AgterenM.de FijterJ.W. HartmannA. SchmidtJ.BuddeK.KuypersD.Le MeurY.van der WerfM.MamelokR. van GelderT. (2009). UGT1A9-275T>A/-2152C>T polymorphisms correlate with low MPA exposure and acute rejection in MMF/tacrolimus-treated kidney transplant patients. Clin Pharmcol Ther, 386319327
  86. 86. vonN.AhsenM.RichterC.GruppB.RingeM.OellerichV. W.Armstrong2001No influence of the MDR-1 C3435T polymorphism 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, 47610481052
  87. 87. WangJ.ZeeviA.WebberS.GirnitaD. M.AddonizioL.SelbyR.HutchinsonI. V.BurckartG. J.2007A novel variant L263F in human inosine 5’-monophosphate dehydrogenase 2 is associated with diminished enzyme activity. Pharmacogenet Genomics, 174283290
  88. 88. WangJ.YangJ. W.ZeeviA.WebberS. A.GirnitaD. M.SelbyR.FuJ.ShahT.PravicaV.HutchinsonI. V.BurckartG. J.2008IMPDH gene polymorphisms and association with acute rejection in renal transplant recipients. Clin Pharmacol Ther, 835711717
  89. 89. WeinshilboumR. M.OtternessD. M.SzumlanskiC. L.1999Methylation pharmacogenetics: Catechol O-methyltransferase, thiopurine methyltransferase, and histamine N-methyltransferase. Annu Rev Pharmacol Toxicol, 391952
  90. 90. WeinshilboumR.2003Inheritance and drug response. N Engl J Med, 3486529537
  91. 91. WestlindA.LöfbergL.TindbergN.AnderssonT. B.Ingelman-SundbergM.1999Interindividual differences in hepatic expression of CYP3A4: Relationship to genetic polymorphism in the 5’-upstream regulatory region. Biochem Biophys Res Commun, 2591201205
  92. 92. WoillardJ. B.RerolleJ. P.PicardN.RousseauA.GuillaudeauA.MunteanuE.EssigM.DrouetM.Le MeurY.MarquetP.2010Donor P-gp polymorphisms strongly influence renal function and graft loss in a cohort of renal transplant recipients on cyclosporine therapy in a long-term follow-up. Clin Pharmacol Ther, 88195100
  93. 93. ZhangX.LiuZ. H.ZhengJ. M.ChenZ. H.TangZ.ChenJ. S.LiL. S.2005Influence of CYP3A5 and MDR1 polymorphisms on tacrolimus concentration in the early stage after renal transplantation. Clin Transplant, 195638643
  94. 94. ZhangY.BenetL. Z.2001The gut as a barrier to drug absorption. Combined role of cytochrome P4503A and P-glycoprotein. Clin Pharmacokinet, 403159168
  95. 95. ZhaoY.SongM.GuanD.BiS.MengJ.LiQ.WangW.2005Genetic polymorphisms of CYP3A5 genes and concentration the cyclosporine and tacrolimus. Transplant Proc, 371178181
  96. 96. ZhengH.WebberS.ZeeviA.SchuetzE.ZhangJ.BowmanP.BoyleG.LawY.MillerS.LambaJ.BurckartG. J.2003Tacrolimus dosing in pediatric heart transplant patients is related to CYP3A5 and MDR1 gene polymorphisms. Am J Transplant, 34477483
  97. 97. ZhengH.ZeeviA.SchuetzE.LambaJ.Mc CurryK.GriffithB. P.WebberS.RistichJ.DauberJ.IaconoA.GrgurichW.ZaldonisD.Mc DadeK.ZhangJ.BurckartG. J.2004Tacrolimus dosing in adult lung transplant patients is related to cytochrome P4503A5 gene polymorphism. J Clin Pharmacol, 442135140

Written By

Chi Yuen Cheung

Submitted: 29 October 2010 Published: 23 August 2011