Skip to main content
Bentham Open Access logoLink to Bentham Open Access
. 2018 May;19(6):513–522. doi: 10.2174/1389200219666180129151948

Pharmacokinetics, Pharmacodynamics and Pharmacogenetics of Tacrolimus in Kidney Transplantation

Meng Yu 1, Mouze Liu 2, Wei Zhang 2, Yingzi Ming 1,*
PMCID: PMC6182932  PMID: 29380698

Abstract

Background:

Tacrolimus (Tac, or FK506), a calcineurin inhibitor (CNI), is the first-line immu-nosuppressant which consists of the footstone as immunosuppressive regimens in kidney transplantation. However, the drug toxicity and the significant differences of pharmacokinetics (PK) and pharmacodynam-ics (PD) among individuals are hidden troubles for clinical application. Recently, emerging evidences of Tac pharmacogenetics (PG) regarding drug absorption, metabolism, disposition, excretion and response are discovered for better understanding of this drug.

Method:

We reviewed the published articles regarding the Tac PG and its effects on PK and PD in kidney transplantation. In addition, we summarized information on polygenic algorithms.

Results:

The polymorphism of genes encoding metabolic enzymes and transporters related to Tac were largely investigated, but the results were inconsistent. In addition to CYP3A4, CYP3A5 and P-gp (also known as ABCB1), single nucleotide polymorphisms (SNPs) might also affect the PK and PD parameters of Tac.

Conclusion:

The correlation between Tac PK, PD and PG is very complex. Although many factors need to be verified, it is envisaged that thorough understanding of PG may assist clinicians to predict the optimal starting dosage, help adjust the maintenance regimen, as well as identify high risk patients for adverse ef-fects or drug inefficacy

Keywords: Pharmacokinetics, pharmacodynamics, pharmacogenetics, Tacrolimus, dosing algorithms, solid organ transplantation

1. INTRODUCTION

Tacrolimus (Tac, or FK506), a calcineurin inhibitor (CNI), is a 23-membered macrolide lactone isolated from Streptomyces tsukubaensis in 1987 for the first time [1]. In 1994, the US Food and Drug Administration firstly approved Tac for liver transplantation. Due to its excellent efficacy, Tac has been extended as a first-line regimen for kidney, heart, lung, intestinal and bone marrow transplantation.

A patient's initial Tac dose is conventionally determined based on body weight and adjusted according to Tac blood concentration. Oral Tac is absorbed in the gastrointestinal tract, and is metabolized by liver enzymes, mainly the cytochrome P450 (CYP450) system, which is easily interacted with many substances. Unfortunately, the therapeutic index of Tac is narrow, and the pharmacokinetics (PK) and pharmacodynamics (PD) features vary dramatically among individuals. As a result, it is often difficult to reach or maintain the target Tac blood concentration, and the patients could be at the risk of either graft rejection or toxicity. Genetic factors including CYP3A5*3, CYP3A4*1B, CYP3A4*22, ABCB1 and POR*28 have been reported frequently for their influence on Tac dose requirement, which reveals the importance of pharmacogenetics (PG) of Tac.

Herein, we summarize the latest research progress of Tac PG, and discuss its effect on PK and PD in kidney transplantation. With thorough understanding of Tac PG, it may assist clinicians to achieve individualized Tac treatment in future.

2. PHARMACOKINETICS

The average bioavailability of Tac is merely 25%, and it varies dramatically among individuals, ranging from 5 to 90% [2, 3]. About 99% of Tac binds to erythrocytes after entering the systemic circulation, but only the dissociated portion can enter the lymphatic system and play its major immunosuppressive effect [4]. The first pass effect of absorption in small intestine and liver, and the pumping action of the small intestine contribute to the poor bioavailability. The synergistic effects of CYP3A and P-glycoprotein (P-gp, or ABCB1) can significantly influence the absorption of Tac in the small intestine [4]. P-gp can also inhibit the entry of Tac into organs or septal structures, including blood-brain barrier, testis, placenta, heart and kidney. In P-gp knockout transgenic mice, the Tac concentration in brain cells was significantly increased [5]. This indicates the importance of P-gp in the variability of Tac distribution.

Tac is primarily metabolized by the CYP3A enzyme system, which includes CYP3A5, CYP3A4, CYP3A7 and CYP3A43, and is expressed in small intestine, liver and kidney [6]. Liver is the main site for Tac metabolism, while considerable pre-systemic biotransformation occurs in small intestine [7]. In kidney, the main CYP3A isoform expressed is CYP3A5, which may play an important role in local Tac metabolism [8, 9].

Compared with CYP3A5, the catalytic efficiency of CYP3A4 was relatively low [10]. CYP3A7 has little influence on the metabolism of Tac, while the role of CYP3A43 is unclear [11, 12].

The total body clearance (TBC) of Tac is relatively low, around 0.06 L/(h⋅kg)-1, while the half-life is long and variable, ranging from 4 to 41 h (about 12 h on average) [2, 3, 13]. Approximately 95% of Tac metabolites are excreted by bile, and urinary excretion is only about 2% [7]. Only 0.5% of the original drug is excreted through urine and feces.

The metabolites of Tac may also be substrates for potential drug transporters. For instance, metabolites formed by Tac in mucosa may return to the intestinal lumen via P-gp transport [14]. In kidney, P-gp expressed on brush border of proximal tubular epithelial cells and more distally on the renal tubule may contribute to renal elimination; by contrast, P-gp on the canalicular surface of hepatocytes controls excretion into bile [15].

Given all the evidences above, it suggests that the expression and/or function of CYP3A5, CYP3A4 and P-gp are closely related to the PK of Tac. Therefore, genetic polymorphism of these proteins or factors that have an impact on them can affect the PK of Tac and lead to inter-individual differences. The relationship between PK and PG is reviewed in Table 1 [6, 8, 11-13, 16-28].

Table 1.

PK and PG.

Genes rsID Alleles Pharmacokinetic Parameters References
CYP3A5*3 rs776746 *3/*3 ↓ Da,b ↓ CL/F ↑C0/D ↑ Cmax/D ↑AUC12/D ↑C0 [12, 13, 27]
CYP3A5*6 rs10264272 *6/*1 *6/*6 ↑C0/D [18]
CYP3A5*7 rs41303343 *7/*1 *7/*7 ↑C0/D [18]
CYP3A4*1Bc rs2740574 *1B/1 *1B/*1B ↑D ↓C0/D [12]
CYP3A4*18c rs28371759 *18/*1 *18/*18 ↑CL/F ↓C0/D ↓C2/D [12, 16, 28]
CYP3A4*22 rs35599367 *22/*1 *22/*22 ↑C0/D ↓D [6, 8, 19]
ABCB1 3435C>T rs1045642 T-T ↑D ↓D ↑C0/D [12, 16, 28]
ABCB1 2677G>T/A rs2032582 G-T T-T ↑C0/D ↑D ↓D [11, 12, 16]
ABCB1 1236C>T rs1128503 C-T T-T ↑C0/D [11, 12, 16]
Haploid ABCB1 3435C>T rs1045642 T-T-T ↓D ↑C0/D (vs. C-G-C) [11, 12, 16]
ABCB1 2677G>T/A rs2032582
ABCB1 1236C>T rs1128503
PXR 8055C>T rs2276707 C-T T-T ↑AUC12/D [26]
POR*28 rs1057868 *28/*28 *28/*1 ↓C0d ↓AUC24d ↓Cmaxd ↑Dd [17, 20-25]

AUCx: Area under the concentration time curve(0~x h); CL/F: Apparent clearance rate; C0: Tac trough concentration; Cmax: Maximum blood concentration; D: Dose requirement.

a: An example how to read the table: compared with other genetic variants (*3/*1 or *1/*1), type *3/*3 may decrease the Tac dose requirement; b: in order to make the table legible, only the common parameters mentioned frequently in literature are listed; c: there is Linkage disequilibrium with CYP3A5*3; d: only make sense when the recipient is CYP3A5*1 allele carrier.

3. PHARMACODYNAMICS

Tac is an effective immunosuppressive drug to prevent rejection. Meanwhile, it also has considerable toxicity and a narrow therapeutic window. In clinic, even though the concentration of some patients is within the therapeutic window, they might also suffer from toxicity or rejection. On the contrary, certain patients do not reject the allografts with a low Tac exposure. The variability between individuals may lie on the genetics polymorphism. The PG-based differences in Tac PD about adverse effects are summarized in Table 2 [11, 24, 25, 27, 29-42].

Table 2.

PD (adverse effects) and PG.

Genes rsID Genotype Tac-induced Adverse Events Risk
Acute
Rejection
Nephrotoxicity DGF PTDM Hypertension Neurotoxicity
CYP3A5*3 rs776746 *3/*3 a↑[31, 34]
[30]
↑[58] ↓[35, 36] ↓[70]
ABCB1 3435C>T rs1045642 T-T ↑[50] (D&R)
↓[37] (D)
↓[38]
ABCB1 2677G>T/A rs2032582 G-(T/A) or (T/A)-(T/A) ↑[38]
↓[11]
ABCB1 1236C>T rs1128503 C-C ↑[11]
Haploid ABCB1 3435C>T rs1045642 T-G-C ↑[11]
ABCB1 2677G>T/A rs2032582
ABCB1 1236C>T rs1128503
PXR 8055C>T rs2276707 T-T ↑[57] (D)
POR*28 rs1057868 *28/*28 or *28/*1 ↑[17]
TGF-β1 29T>C rs1800470 C-T ↑[39-41]
TGF-β1 74G>C rs1800471 G-C ↑[39, 40]
PPARA rs4253728 G>A rs4253728 A-A or A-G ↑[17]
CYP2C8*3 rs11572080 *3/*3 or *3/*1 ↑[42] ↑[59]

a: an example to read the table: compared with other genetic variants (*3/*1 or *1/*1), type *3/*3 may increase the risk of Tac-induced nephrotoxicity, investigated in reference 25 and 133.

When there are ↑ and ↓ on a same line, it means there are conflicting results demonstrated by different groups. (D&R): means the genotype of both the donor and recipient is effective in influencing the risk; (D): means only the donor genotype may contribute to the influence. Not all research results are listed, only those seems to be reliable are demonstrated here, for details, readers can refer to the paragraph or the references.

3.1. Acute Rejection (AR)

The genetics basis associated with acute rejection has been extensively studied [43]. In a cohort study carried out by Oetting WS et al., 969 kidney transplant recipients were recruited and 23 genetic variants which were associated with AR according to previous reports, were analyzed. Interestingly, only one single-nucleotide polymorphism (SNP) within the coagulation factor V gene (rs6025, Leiden mutation), was proved to be significantly associated with AR [44].

Moreover, studies also showed that SNPs of CYP3A and ABCB1 were not the risk factors of rejection [16, 45].

3.2. Nephrotoxicity

CNI-induced nephrotoxicity is likely related to intra-renal concentrations of CNIs, which may not be properly reflected by whole-blood CNI concentrations [29, 46-48]. As mentioned above, the main CYP3A isoform expressed in the kidney is CYP3A5, which contributes to local Tac metabolism and limit the Tac local accumulation [8, 9]. However, contradictory results have been reported on their relationship [30, 31]. Possible reasons for these discrepancies include differences in ethnicity, sample size and the definition of nephrotoxicity. In addition, there is a hypothesis that, it is not Tac itself but its metabolites that are responsible for its nephrotoxicity.

ABCB1 expressed in renal tubules may limit the local accumulation of Tac and its metabolites in kidney through facilitating them into urine [15]. Thus, a lower ABCB1 expression in kidney may be associated with increased risk of chronic kidney damage caused by Tac. However the results of the reported literatures remains inconsistent [32, 49, 50]. One possible explanation is that what really works is the function of ABCB1 other than its expression. Besides, CYP3A5 may have different interplay with ABCB1 in vascular and tubule-interstitial compartments of kidney [30].

In addition to ABCB1 and CYP3A, CYP2C8 is also related to CNI-nephrotoxicity. CYP2C8 is a member of the P450 superfamily which plays a role in the metabolic process of arachidonic acid (AA) to epoxyeicosatrienoic acids (EETs) in kidney [51-53]. EETs are biologically active to help the kidneys to counter the vasoconstrictive effects of CNIs, which indicates the beneficial role of CYP2C8 in CNI-nephrotoxicity.

3.3. DGF

Delayed graft function (DGF) is a frequent complication after kidney transplantation which reduces long-term allograft survival. Although the definition may differ, a widely-recognized definition is the need for dialysis within the first week after transplantation [54, 55]. Ischemia/reperfusion injury is the main reason for DGF [54, 56]. Hauser et al. reported that PXR significantly increased the risk of DGF [57]. Another study found that DGF was associated with CYP3A5 [58]. Besides, association between DGF and CYP2C8 has also been reported [59].

3.4. Post Transplanted Diabetes Mellitus (PTDM)

New-onset diabetes is another frequent complication after transplantation due to the immunosuppressive drugs, such as glucocorticoids and mammalian target of rapamycin (mTOR) inhibitors. Tac can also cause glycometabolism disorder, and is more diabetogenic than cyclosporine [33]. The mechanisms involve impairment of insulin secretion and insulin gene expression as well as directly toxicity to Langerhans in islets [60]. Recent studies revealed that the genetic background was one of the risk factors for PTDM. The PPARA rs4253728 A>G and POR*28 variant alleles could increase the risk of developing PTDM [60]. Polymorphisms in the genes of vitamin D receptor, promoter region of the IL-6, transcription factor 7-like 2 (rs7903146) and zinc transporter-8 (SLC30A8; rs13266634) have also been reported [17, 61-65].

3.5. Hypertension

Hypertension caused by Tac is due to activating the renal sodium chloride co-transporter, which disturbs the“with-no-lysine”(WNK) kinase network [66].

CYP3A5 and ABCB1 have been reported to play a role in regulating the metabolism of 6β-hydrocortisol and the transport of aldosterone, which influences the metabolism of water and sodium in kidney [67]. The genetic polymorphisms of ABCB1 and CYP3A5 may be related to hypertension, but it needs further research [68].

3.6. Neurotoxicity

Neurotoxic effects of Tac include tremor, headache, insomnia, and peripheral neuropathy [69]. Although the exact pathophysiology of Tac-induced neurotoxicity is unclear, how Tac penetrates into the central nervous system (CNS) is the critical step for neurotoxicity. Yanagimachi et al. Reported that the CYP3A5*1 allele could increase the risk of neurotoxicity [70]. They also suggested that it might not be Tac itself but its metabolites that caused neurotoxicity. ABCB1, which is expressed in the blood brain barrier, also takes part in Tac transport in CNS. In mice, dysfunction of ABCB1 leaded to accumulation of Tac in the CNS [5, 71]. However, whether ABCB1 genotype has an impact on Tac-induced neurotoxicity in clinic is still unclear.

So far, the genetics of Tac PD has been less well-investigated than that of Tac PK. Tissue concentrations of CNIs have hitherto received little attention due to technical difficulties, although only the intra-renal Tac can play a role. Meanwhile, more attention should be paid to unbound blood Tac concentration, which may have a closer relationship to Tac toxicity [72]. The current evidences of the relationship between PG and PD are summarized in Table 2. With better understanding of the relation between gene polymorphisms and Tac PD, we might be able to assess the efficacy of Tac through the detection of gene polymorphisms in future.

4. Pharmacogenetics

Definitely, the correlation between Tac PK, PD and PG is very complex. Published investigations mainly focused on the polymorphism of genes encoding metabolic enzymes and transporters. In addition to CYP3A4, CYP3A5 and P-gp (also known as ABCB1), there are more SNPs that may affect the PK and PD parameters of Tac as mentioned above. The following is a brief summary.

4.1. CYP3A5

Polymorphisms in the CYP3A5 gene explain 40-50% of the variability in Tac dose requirement [73, 74]. The hottest SNP studied most in CYP3A5 is CYP3A5*3, which is an A to G transition at position 6986 within intron 3 (rs776746) [67]. This mutation leads to alternative splicing, and truncation of the protein, which decreases the function of CYP3A5 enzyme [75]. As a result, CYP3A5 expressers (CYP3A5*1/*1 or CYP3A5*1/*3 genotype) have significantly lower dose-adjusted C0 compared to CYP3A5 non-expressers (CYP3A5*3/*3 genotype), and the requirement of Tac dose is CYP3A5*1/*1>*1/*3>*3/*3 [67]. In Chinese, the frequency of CYP3A5*3 allele is as high as 77.8%, which may be an explanation for the lower dosage required for Chinese people [76]. Meanwhile, as for the postoperative adverse drug reactions including abnormal liver function and renal toxicity, there is a conflicting result that type *1/*3 and *3/*3 are significantly higher than type *1/*1 [30, 77].

Following standard bodyweight-based dosing, the exposure of Tac for CYP3A5 expressers could be insufficient at the early stage post transplantation, so that the risk for AR might increase. MacPhee et al. demonstrated that even under the therapeutic drug monitoring (TDM), CYP3A5 expressers did achieve the target Tac concentration with a delay [78]. However, no evidence supports the hypothesis that this delay would increase rejection. It was reported that CYP3A5 expressers only had an earlier onset of AR compared with non-expressers (median time, 7 days versus 13 days), but the rate of biopsy-proven acute rejection had no significant difference [77]. This is in consistent with some other investigations [11, 79-86].

Meanwhile, the CYP3A5 genotype-based Tac initial dose has been verified extensively. Several randomized-controlled clinical trials (RCT) suggested that CYP3A5 genotypes could be helpful in predicting the initial dose of Tac [87-89]. However, it is still unclear whether this strategy was superior in terms of efficacy when compared with conventional TDM [90]. Nevertheless, recipients who received a CYP3A5 genotype-based Tac dose needed significantly less time and fewer dose adaptations to reach target, which, in our view would be helpful in some potential ways for the recipients.

Other CYP3A5 SNPs include CYP3A5*6 (rs10264272) and CYP3A5*7 (rs41303343). CYP3A5*6 encodes a G to A transition at position 14690, causing a splice variant mRNA and deletion of exon 7, resulting in nonfunctional CYP3A5 protein [18, 75]. CYP3A5*7 denotes a single base insertion at codon 346, causing a frameshift and resulting in a truncated mRNA and nonfunctional CYP3A5 [91].

4.2. CYP3A4

As for CYP3A4 gene, two SNPs in relation to Tac PK have been investigated extensively: CYP3A4*1B SNP (rs2740574) and CYP3A4*22 SNP (rs35599367). The CYP3A4*1B SNP involves an A to G transition at position -392 in the promoter region of CYP3A4, and is associated with an increase of CYP3A4 activity [92]. It showed that the C0/D ratio of Tac in patients with the *1B mutation was reduced by 35% compared with that of wild-type homozygotes [93]. However, there is a linkage disequilibrium(LD) between CYP3A4*1B and rs776746 of the CYP3A5 gene. It is possible that the effect of CYP3A4*1B on Tac PK and PD is caused by rs776746, which has been shown in several published studies [12, 94]. Therefore, the exact effect of CYP3A4*1B alone on Tac is still unclear. The CYP3A4*22 SNP (rs35599367) contains a transition of C to T in intron 6 and is associated with reduced CYP3A4 mRNA expression and CYP3A4 enzyme activity in vitro [95]. In clinic observation of kidney transplantation, the CYP3A4*22 required less Tac dose to achieve the target exposure. What's more, it was not influenced by the CYP3A5 genotype [6]. Similar result was observed in pediatric heart transplant recipients [96]. To reach similar target concentrations, the requirement of Tac dose was 30% less in CYP3A4*22 carriers than that in CYP3A4*1/*1 carriers.

CYP3A4 * 18 (rs28371759) may also have an impact on Tac PK. This SNP is located in intron 10, with a transition of T to C at position 878. This mutation may increase the activity of CYP3A4 enzyme, and thereby increase the Tac clearance rate and plasma drug concentration [97].

Recently a new and rare CYP3A4 variant was found, which is now designated as CYP3A4*26 [98]. This variant is a c.802C>T transition and results in a premature stop codon at position 268 in exon 9 (R268*) [98]. The truncated CYP3A4 protein is non-functional. Werk et al. [99] first identified this mutation when they observed an unusually low Tac dose requirement in a kidney transplant recipient. This patient had very high Tac exposure following standard Tac dosing and only reached the therapeutic window once the Tac dose was reduced to 0.5 mg thrice weekly. This patient was a CYP3A5*3 homozygote and was also homozygous for CYP3A4*26, and therefore experienced complete failure of CYP3A enzyme activity.

When combining CYP3A4 and CYP3A5 genotypes, Elens et al. [19] were able to predict Tac dose requirements better compared with the CYP3A4 or CYP3A5 genotype alone. Based on these observations, it has been proposed to prescribe different Tac doses for ultra-rapid (CYP3A5 expressers and CYP3A4 *1/*1), intermediate (CYP3A5 non-expressers and CYP3A4*1/*1) and poor (CYP3A5 non-expressers and CYP3A4*22 carriers) CYP3A metabolizers, respectively [100].

4.3. ABCB1 Gene

P-gp, also known as ABCB1 or MDR1 is a glycoprotein encoded by human ABCB1 gene. As discussed above, it serves as drug transporter of Tac, and plays an important role in Tac PK. Recently, P-gp has been found to contain more than 50 SNPs. Among them, the ABCB1 3435C>T (rs1045642), 1236C>T (rs1128503) and 2677G>T/A (rs2032582; Ala893Ser/Thr) SNPs have drawn the most attention after intensive investigation [101-103].

The ABCB1 3435C>T (rs1045642) might be the hottest locus among all the ABCB1 gene SNPs. Reportedly, the frequency of this mutation in Orientals is 37-49% [104]. The variation of rs1045642 locus might reduce the expression and function of P-gp in the duodenum, and thus potentially affect the bioavailability of Tac [16].

Another retrospective study recruited 81 recipients and found that after one month of renal transplantation, the daily Tac dose and the concentration/dose ratio were highly associated with 2677G>T/A SNP [105]. In detail, wild-type patients required 40% higher Tac dose compared with homozygous carriers of 2677G>T/A SNP (P≤0.05), while the concentration/dose ratio was 36% lower in the wild-type patients (P≤0.02). The haplotype analysis further confirmed the results and suggested that 3435C>T and 2677G>T/A SNPs were associated with daily Tac dose requirements.

In addition, the study of these three SNP haploid (1236C>T, 2677G>T/A and 3435C>T, which are in linkage disequilibrium) found that C-G-C (haplotype 1) and T-T/A-T (haplotype2) accounted for 45.4% and 36.2% of the haplotypes, respectively; individuals with haplotype 1 required significantly higher daily doses of Tac than those with haplotype 2 [105].

Because lymphocytes also express ABCB1 on the membrane, the activity of ABCB1 may affect the intracellular accumulation of Tac where the drug exerts its biologic effect as well. Vafadari et al. found that patients with the ABCB1 3435CC genotype needed more Tac for inhibition of IL-2 production in T-cells compared with 3435TT genotype patients [106]. Capron et al. found that patients with the ABCB1 3435T or the 2677T/A allele had 1.3-fold higher Tac concentrations within circulating lymphocytes compared with wild-type homozygotes [107]. These studies provide evidences that ABCB1 3435C>T and 2677G>T/A affect Tac distribution into lymphocytes with the variant alleles which are associated with an increased pharmacodynamic effect of Tac. Therefore, ABCB1 SNPs may also play a role in Tac-induced nephrotoxicity, as tissue concentrations of Tac are believed to be more related to its renal side effects.

Although there are many studies on the association of ABCB1 gene polymorphism with Tac PK or PD, the results remain inconsistent. To further confirm the association, large-scale genotype-phenotype correlation trials are encouraged.

4.4. POR

POR is essential for CYP-mediated drug oxidation as an electron donor [108]. POR*28 (rs1057868; A503V) is a coding variant in POR gene, which is believed to be effective in increasing the activity of POR and thus leads to the increasing activities of CYP3A4 and CYP3A5 [17, 28]. An investigation demonstrated that the POR*28 cloud lead to an increase of CYP3A5-mediated Tac metabolism, but in CYP3A5 non-expressers, there was no increase of CYP3A4-mediated Tac metabolism, which indicated a possibility of interaction between POR, CYP3A5 and Tac [109].

In addition, it has also been reported frequently that POR*28 carriers have lower adjusted Tac C0 and Tac C0/Tac dose in heart or kidney transplantation, no matter for the adult or the pediatric [21-24]. Although the strength of this association seems weak and has limited clinical impact on Tac dose requirements (15%-20%), POR*28 may explain a part of Tac variability, and POR*28 carriers may experience faster Tac metabolism [21, 23, 25].

4.5. PXR

As a nuclear transcription, the human pregnane X receptor (PXR), which is encoded by NR1I2, regulates the expression of CYP3A and ABCB1. Polymorphisms of NR1I2 have been reported, but the results regarding their association with Tac dose requirement are conflicting [26, 73, 100].

The six-base pair deletion mutation (rs3842689, -/GAGAAG) in the NR1I2 promoter region was first discovered by Uno et al. [110, 111], which occurs in a potential liver nuclear factor (HNF-1) binding site. Research indicated that the PXR Six-base deletion mutation could reduce the expression of PXR mRNA, and thus, significantly reduce the expression of CYP3A4 and ABCB1 [112]. However, Z.P. Wang et al. reported that the PXR rs3842689 native homozygous WW genotype was only a risk factor for gastrointestinal reactions, and the other genotypes seemed to play a minor role in Tac-induced adverse reactions [113].

4.6. PPAR-α

The expression and activity of CYP3A is also related to the nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR-α). Two sequence variants in the PPAR-α gene (PPARA), PPARA c.209-1003G>A and c.208+3819A>G, can reduce the PPAR-α expression and contribute to the intra- and inter-individual variability of CYP3A [114]. In one study with 229 kidney transplant recipients, the Tac C0/D ratio was significantly higher in patients with one or more PPARA variant alleles [94]. Patients who were homozygous for PPARA-α c.209-1003G>A showed significantly higher Tac exposure, which was consistent with the reduced CYP3A4 protein and activity observed in vitro [114]. At present, PPARA c.208+3819A>G appears to have the strongest influence on Tac PK, though it still needs confirmation.

As always the case, there are opposite findings, which show that the PPARA c.209-1003G>A has no effect on Tac C0/D [115, 116]. These paradoxical results need further investigation.

4.7. Other SNPs

The multidrug resistance-associated protein 2 (MRP2), which is encoded by the ABCC2, may also be associated with Tac metabolism [117]. Reportedly, ABCC2 c.3972C>T (rs3740066) SNP significantly increased the dose-normalized concentration of Tac, whereas ABCC2 c.-24C>T (rs717620) SNP had no influence on it [117]. Genvigir et al. also showed that the ABCC2 c.3972C>T polymorphism affected Tac C/D in Brazilian kidney transplant recipients [118]. However, Renders et al. showed that there was no association between the ABCC2 c.3972C>T polymorphism and the Tac concentration in German patients [82].

The CYP2C8 enzyme, which is highly expressed in the liver, can also be found in extrahepatic tissues like kidney [119]. Reported by Suarez-Kurtz et al. [120], The CYP2C8*3 was associated with higher Tac C0/D, but only in CYP3A5 none-expressers. Furthermore, CYP2C8*3 and CYP2J2 c.-76G>T SNPs were reported to influence the renal function of the patients and the occurrence of adverse events during treatment with Tac and mycophenolate sodium [118].

Genetic polymorphisms in IL-18 (e.g., rs5744247) and IL-10 (e.g., -819 C/T and -592 C/A) can also affect Tac dose requirements [81, 94]. However, the exact mechanism by which they affect Tac dose requirements is unknown [121, 122]. Recently, our group also demonstrated that IL-3 rs181781 and CTLA4 rs4553808 genetic polymorphisms probably influence the Tac dose requirements in Chinese kidney transplant recipients [123].

5. CLINICAL APPLICATION

The metabolism of Tac can be influenced by many factors, including ethnicity, age, gender, concomitant medication, hepatic and renal dysfunction, and genetic factors such as CYP3A5, CYP3A4 and ABCB1 SNPs [12, 16, 124]. Among them, CYP3A5 genotype is associated with a remarkable impact on Tac PK, while the effects of other genetic polymorphisms are limited or even contradictory [125-127]. A number of algorithms containing clinical and/or PG factors have been constructed to predict the requirement of Tac dose; meanwhile, retrospective and prospective trials have been conducted to verify these algorithms [87, 125, 128-134].

The first dosing algorithm was created by Passey et al. in 2011. The CYP3A5 genotype was the only genetic factor included in the algorithm [130], while this dosing algorithm was not able to predict estimated Tac clearance accurately in another study in the UK in 2013 [135]. Another dosing algorithm reported by Passey et al. was improved by incorporating the CYP3A4*22 allele [136]. There are also other dosing algorithms reported by different groups demonstrating their successful performance [87, 125]. In general, PG factors guided Tac dosing enabled more renal recipients to achieve target Tac trough (C0) levels. What’s more, it took less time for these patients to reach the target concentration with fewer dose modifications. These successes in clinic revealed the possibility to improve clinical outcomes of Tac therapy by taking PG factors into account.

In our previous study, we collected the clinical data of 1045 renal transplant patients from multiple transplant centers in the past 7 years, developed machine-learning models to predict Tac stable dose(TSD) in renal transplant recipients [124]. To our knowledge, this is the first study using machine-learning models to predict TSD. Comparing conventional dosing algorithms performed by multiple linear regression with the eight new techniques of machine learning, we demonstrated that the technique of regression tree was the best to predict TSD. With the highest ideal rate, the algorithm performed by this technique might provide a more accurate approach which could help achieve personalized medicine in clinic. One concern for our study is that only the data from the Chinese was analyzed; studies in other ethnic groups may come to different results.

Algorithms that are widely accepted in clinic should be validated in populations of different ethnicity. Genetic variations of drug-metabolizing enzymes show remarkable differences between different peoples. For example, the allelic frequency of the CYP3A5*3 allele, which is common among Caucasian patients (90-93%), is less frequently seen in Asian (60-73%) or African (32%) descent [137, 138]. When it comes to a particular group, the situation may be much more complicated. In a study of six different Chinese ethnic groups, Lai et al. [128] reported the frequencies of CYP3A5*3 variant alleles differed dramatically in Uygur Chinese (88.1%), Kazakh Chinese (84.5%), Tibetan Chinese (80.3%), Han (67.3%), Bai Chinese (70.2%) and Wa Chinese (56.3%). More details about the allelic frequencies of the most common SNPs in CYP3A5, CYP3A4, ABCB1 and POR*28 in various ethnic groups were summarized by J.T. Tang et al. [139].

Although it is still a question whether genotype testing would improve clinical outcome, it is definitely true that this technique is effective on Tac pharmacokinetic parameters. At present, the TDM is still the indispensable part in current management of Tac, which obviously cannot be replaced by polygenic algorithms. However, more and more evidences show the association between the genotypes and Tac PK parameters in recent years, which suggests the potential for greater predictive value of polygenic algorithms. In fact, many centers have already established or applied some dosing algorithms, which are primarily based on CYP3A5 genotype. The Clinical Pharmacogenetics Implementation Consortium (CPIC) has also published Tac dosing guidelines based on CYP3A5 genotype expression [140, 141]. Unfortunately, it is still questionable whether these techniques have been widely used in standard practice to guide Tac dosing. So far, the evidence has been limited for us to provide an appropriate genotype testing strategy for each ethnic group, and the Tac dosing guidelines based on CYP3A5 genotype published by CPIC seem to be the only one which is relatively authoritative. But we do hope that more and more clinicians could try to use the genotype-guided dosing followed by TDM and pay more attention on Tac adverse effects simultaneously. If so, we may get more valuable evidence to achieve the optimal individual therapy.

CONCLUSION AND FUTURE PROSPECTS

In order to better understand the individual differences of Tac, tremendous efforts have been made. The genetics polymorphisms, including SNPs of CYP3A4, CYP3A5, ABCB1 etc., play an important role in the variability of Tac. Although it is not sure whether genotype testing of these alleles would improve clinical outcome of kidney transplantation, this technique is definitely effective in depicting the PK parameters of Tac. Algorithms based on multiple genotypes have a better performance in predicting the required dose, which helps recipients achieve target Tac concentration faster with fewer dose adjustments. It is inspiring that the transplant community devotes such great efforts to the PG research. We believe that, by combining genetics with demographic, clinical, epigenetic and environmental information, predictive algorithms may be developed to achieve more reliable dosing, thereby avoiding patients exposing to ineffective or overly toxic regimens.

Furthermore, if the intra-cellular or tissue drug quantification can be carried out in daily clinical practice, and novel techniques such as mass spectrometry can be applied, we may soon clarify the significance of Tac metabolites and tissue Tac concentrations.

To achieve Tac tailored treatment, another obstacle in the development of PG in China is the cost of genetic testing which has not been covered by insurance. Economic condition has limited the routine clinical practice of genotyping. All in all, we still have a lot of difficulties to overcome. But considering that Tac is likely to be around for the next decade, all the efforts are worthy.

Consent for Publication

Not applicable.

Acknowledgements

Declared none.

Conflict of Interest

The authors declare no conflict of interest, financial or otherwise.

REFERENCES

  • 1.Pritchard D.I. Sourcing a chemical succession for cyclosporin from parasites and human pathogens. Drug Discov. Today. 2005;10:688–691. doi: 10.1016/S1359-6446(05)03395-7. [DOI] [PubMed] [Google Scholar]
  • 2.Staatz C.E., Tett S.E. Clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplantation. Clin. Pharmacokinet. 2004;43:623–653. doi: 10.2165/00003088-200443100-00001. [DOI] [PubMed] [Google Scholar]
  • 3.Venkataramanan R., Swaminathan A., Prasad T., Jain A., Zuckerman S., Warty V., McMichael J., Lever J., Burckart G., Starzl T. Clinical pharmacokinetics of tacrolimus. Clin. Pharmacokinet. 1995;29:404–430. doi: 10.2165/00003088-199529060-00003. [DOI] [PubMed] [Google Scholar]
  • 4.Christians U., Strom T., Zhang Y.L., Steudel W., Schmitz V., Trump S., Haschke M. Active drug transport of immunosuppressants: new insights for pharmacokinetics and pharmacodynamics. Ther. Drug Monit. 2006;28:39–44. doi: 10.1097/01.ftd.0000183385.27394.e7. [DOI] [PubMed] [Google Scholar]
  • 5.Yokogawa K., Takahashi M., Tamai I., Konishi H., Nomura M., Moritani S., Miyamoto K., Tsuji A. P-glycoprotein-dependent disposition kinetics of tacrolimus: studies in mdr1a knockout mice. Pharm. Res. 1999;16:1213–1218. doi: 10.1023/a:1018993312773. [DOI] [PubMed] [Google Scholar]
  • 6.Elens L., Bouamar R., Hesselink D.A., Haufroid V., van der Heiden I.P., van Gelder T., van Schaik R.H. A new functional CYP3A4 intron 6 polymorphisms significantly affects tacrolimus pharmacokinetics in kidney transplant recipients. Clin. Chem. 2011;57:1574–1583. doi: 10.1373/clinchem.2011.165613. [DOI] [PubMed] [Google Scholar]
  • 7.Möller A., Iwasaki K., Kawamura A., Teramura Y., Shiraga T., Hata T., Schäfer A., Undre N.A. The disposition of 14C-labeled tacrolimus after intravenous and oral administration in healthy human subjects. Drug Metab. Dispos. 1999;27:633–636. [PubMed] [Google Scholar]
  • 8.Murray G.I., McFadyen M.C., Mitchell R.T., Cheung Y.L., Kerr A.C., Melvin W.T. Cytochrome P450 CYP3A in human renal cell cancer. Br. J. Cancer. 1999;79:1836–1842. doi: 10.1038/sj.bjc.6690292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Koch I., Weil R., Wolbold R., Brockmöller J., Hustert E., Burk O., Nuessler A., Neuhaus P., Eichelbaum M., Zanger U., Wojnowski L. Interindividual variability and tissue-specificity in the expression of cytochrome P450 3A mRNA. Drug Metab. Dispos. 2002;30:1108–1114. doi: 10.1124/dmd.30.10.1108. [DOI] [PubMed] [Google Scholar]
  • 10.Dai Y., Hebert M.F., Isoherranen N., Davis C.L., Marsh C., Shen D.D., Thummel K.E. Effect of CYP3A5 polymorphism on tacrolimus metabolic clearance in vitro. Drug Metab. Dispos. 2006;34:836–847. doi: 10.1124/dmd.105.008680. [DOI] [PubMed] [Google Scholar]
  • 11.Gervasini G., Garcia M., Macias R.M., Cubero J.J., Caravaca F., Benitez J. Impact of genetic polymorphisms on tacrolimus pharmacokinetics and the clinical outcome of renal transplantation. Transpl. Int. 2012;25:471–480. doi: 10.1111/j.1432-2277.2012.01446.x. [DOI] [PubMed] [Google Scholar]
  • 12.Staatz C.E., Goodman L.K., Tett S.E. Effect of CYP3A and ABCB1 single nucleotide polymorphisms on the pharmacokinetics and pharmacodynamics of calcineurin inhibitors: Part I. Clin. Pharmacokinet. 2010;49:141–175. doi: 10.2165/11317350-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 13.van Maarseveen E.M., Rogers C.C., Trofe-Clark J., van Zuilen A.D., Mudrikova T. Drug-drug interactions between antiretroviral and immunosuppressive agents in HIV-infected patients after solid organ transplantation: A review. AIDS Patient Care STDS. 2012;26:568–581. doi: 10.1089/apc.2012.0169. [DOI] [PubMed] [Google Scholar]
  • 14.Cummins C.L., Jacobsen W., Christians U., Benet L.Z. CYP3A4-transfected Caco-2 cells as a tool for understanding biochemical absorption barriers: studies with sirolimus and midazolam. J. Pharmacol. Exp. Ther. 2004;308:143–155. doi: 10.1124/jpet.103.058065. [DOI] [PubMed] [Google Scholar]
  • 15.Thiebaut F., Tsuruo T., Hamada H., Gottesman M.M., Pastan I., Willingham M.C. Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues. Proc. Natl. Acad. Sci. USA. 1987;84:7735–7738. doi: 10.1073/pnas.84.21.7735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Grinyó J., Vanrenterghem Y., Nashan B., Vincenti F., Ekberg H., Lindpaintner K., Rashford M., Nasmyth-Miller C., Voulgari A., Spleiss O., Truman M., Essioux L. Association of four DNA polymorphisms with acute rejection after kidney transplantation. Transpl. Int. 2008;21:879–891. doi: 10.1111/j.1432-2277.2008.00679.x. [DOI] [PubMed] [Google Scholar]
  • 17.Min S.I., Kim S.Y., Ahn S.H., Min S.K., Kim S.H., Kim Y.S., Moon K.C., Oh J.M., Kim S.J., Ha J. CYP3A5 *1 allele: impacts on early acute rejection and graft function in tacrolimus-based renal transplant recipients. Transplantation. 2010;90:1394–1400. doi: 10.1097/TP.0b013e3181fa93a4. [DOI] [PubMed] [Google Scholar]
  • 18.Staatz C.E., Goodman L.K., Tett S.E. Effect of CYP3A and ABCB1 single nucleotide polymorphisms on the pharmacokinetics and pharmacodynamics of calcineurin inhibitors: Part II. Clin. Pharmacokinet. 2010;49:207–221. doi: 10.2165/11317550-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 19.Shuker N., Bouamar R., Weimar W., van Schaik R.H., van Gelder T., Hesselink D.A. ATP-binding cassette transporters as pharmacogenetic biomarkers for kidney transplantation. Clin. Chim. Acta. 2012;413:1326–1337. doi: 10.1016/j.cca.2011.09.040. [DOI] [PubMed] [Google Scholar]
  • 20.Naesens M., Kuypers D.R., Sarwal M. Calcineurin inhibitor nephrotoxicity. Clin. J. Am. Soc. Nephrol. 2009;4:481–508. doi: 10.2215/CJN.04800908. [DOI] [PubMed] [Google Scholar]
  • 21.van Gelder T., Balk A.H., Zietse R., Hesse C., Mochtar B., Weimar W. Renal insufficiency after heart transplantation: a case-control study. Nephrol. Dial. Transplant. 1998;13:2322–2326. doi: 10.1093/ndt/13.9.2322. [DOI] [PubMed] [Google Scholar]
  • 22.Noll B.D., Coller J.K., Somogyi A.A., Morris R.G., Russ G.R., Hesselink D.A., Van Gelder T., Sallustio B.C. Measurement of cyclosporine A in rat tissues and human kidney transplant biopsies-a method suitable for small (<1 mg) samples. Ther. Drug Monit. 2011;33:688–693. doi: 10.1097/FTD.0b013e318236315d. [DOI] [PubMed] [Google Scholar]
  • 23.Noll B.D., Coller J.K., Somogyi A.A., Morris R.G., Russ G.R., Hesselink D.A., Van Gelder T., Sallustio B.C. Validation of an LC-MS/MS method to measure tacrolimus in rat kidney and liver tissue and its application to human kidney biopsies. Ther. Drug Monit. 2013;35:617–623. doi: 10.1097/FTD.0b013e31828e8162. [DOI] [PubMed] [Google Scholar]
  • 24.Kuypers D.R., Naesens M., de Jonge H., Lerut E., Verbeke K., Vanrenterghem Y. Tacrolimus dose requirements and CYP3A5 genotype and the development of calcineurin inhibitor-associated nephrotoxicity in renal allograft recipients. Ther. Drug Monit. 2010;32:394–404. doi: 10.1097/FTD.0b013e3181e06818. [DOI] [PubMed] [Google Scholar]
  • 25.Chen J.S., Li L.S., Cheng D.R., Ji S.M., Sun Q.Q., Cheng Z., Wen J.Q., Sha G.Z., Liu Z.H. Effect of CYP3A5 genotype on renal allograft recipients treated with tacrolimus. Transplant. Proc. 2009;41:1557–1561. doi: 10.1016/j.transproceed.2009.01.097. [DOI] [PubMed] [Google Scholar]
  • 26.Metalidis C., Lerut E., Naesens M., Kuypers D.R. Expression of CYP3A5 and P-glycoprotein in renal allografts with histological signs of calcineurin inhibitor nephrotoxicity. Transplantation. 2011;91:1098–1102. doi: 10.1097/TP.0b013e3182177502. [DOI] [PubMed] [Google Scholar]
  • 27.Naesens M., Lerut E., de Jonge H., Van Damme B., Vanrenterghem Y., Kuypers D.R. Donor age and renal P-glycoprotein expression associate with chronic histological damage in renal allografts. J. Am. Soc. Nephrol. 2009;20:2468–2480. doi: 10.1681/ASN.2009020192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Joy M.S., Nickeleit V., Hogan S.L., Thompson B.D., Finn W.F. Calcineurin inhibitor-induced nephrotoxicity and renal expression of P-glycoprotein. Pharmacotherapy. 2005;25:779–789. doi: 10.1592/phco.2005.25.6.779. [DOI] [PubMed] [Google Scholar]
  • 29.Imig J.D. Eicosanoid regulation of the renal vasculature. Am. J. Physiol. Renal Physiol. 2000;279:F965–F981. doi: 10.1152/ajprenal.2000.279.6.F965. [DOI] [PubMed] [Google Scholar]
  • 30.Node K., Huo Y., Ruan X., Yang B., Spiecker M., Ley K., Zeldin D.C., Liao J.K. Anti-inflammatory properties of cytochrome P450 epoxygenase-derived eicosanoids. Science. 1999;285:1276–1279. doi: 10.1126/science.285.5431.1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Roman R.J. P-450 metabolites of arachidonic acid in the control of cardiovascular function. Physiol. Rev. 2002;82:131–185. doi: 10.1152/physrev.00021.2001. [DOI] [PubMed] [Google Scholar]
  • 32.Perico N., Cattaneo D., Sayegh M.H., Remuzzi G. Delayed graft function in kidney transplantation. Lancet. 2004;364:1814–1827. doi: 10.1016/S0140-6736(04)17406-0. [DOI] [PubMed] [Google Scholar]
  • 33.Yarlagadda S.G., Coca S.G., Garg A.X., Doshi M., Poggio E., Marcus R.J., Parikh C.R. Marked variation in the definition and diagnosis of delayed graft function: a systematic review. Nephrol. Dial. Transplant. 2008;23:2995–3003. doi: 10.1093/ndt/gfn158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ghods A.J., Savaj S., Abbasi M., Heidari H., Rokhsatyazdi H. The incidence and risk factors of delayed graft function in 689 consecutive living unrelated donor renal transplantation. Transplant. Proc. 2007;39:846–847. doi: 10.1016/j.transproceed.2007.04.011. [DOI] [PubMed] [Google Scholar]
  • 35.Hauser I.A., Kruck S., Gauer S., Nies A.T., Winter S., Bedke J., Geiger H., Hoefeld H., Kleemann J., Asbe-Vollkopf A., Engel J., Burk O., Schwab M., Schaeffeler E. Human pregnane X receptor genotype of the donor but not of the recipient is a risk factor for delayed graft function after renal transplantation. Clin. Pharmacol. Ther. 2012;91:905–916. doi: 10.1038/clpt.2011.346. [DOI] [PubMed] [Google Scholar]
  • 36.Kuypers D.R., de Jonge H., Naesens M., Vanrenterghem Y. A prospective, open-label, observational clinical cohort study of the association between delayed renal allograft function, tacrolimus exposure, and CYP3A5 genotype in adult recipients. Clin. Ther. 2010;32:2012–2023. doi: 10.1016/j.clinthera.2010.11.010. [DOI] [PubMed] [Google Scholar]
  • 37.Gervasini G., Garcia M., Macias R.M., Benitez J., Caravaca F., Cubero J.J. CYP2C8*3 polymorphism and donor age are associated with allograft dysfunction in kidney transplant recipients treated with calcineurin inhibitors. J. Clin. Pharmacol. 2013;53:427–434. doi: 10.1002/jcph.15. [DOI] [PubMed] [Google Scholar]
  • 38.Webster A.C., Woodroffe R.C., Taylor R.S., Chapman J.R., Craig J.C. Tacrolimus versus ciclosporin as primary immunosuppression for kidney transplant recipients: Meta-analysis and meta-regression of randomised trial data. BMJ. 2005;331:810. doi: 10.1136/bmj.38569.471007.AE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yates C.J., Fourlanos S., Hjelmesaeth J., Colman P.G., Cohney S.J. New-onset diabetes after kidney transplantation-changes and challenges. Am. J. Transplant. 2012;12:820–828. doi: 10.1111/j.1600-6143.2011.03855.x. [DOI] [PubMed] [Google Scholar]
  • 40.Elens L., Sombogaard F., Hesselink D.A., Van Schaik R.H., Van Gelder T. Single-nucleotide polymorphisms in P450 oxidoreductase and peroxisome proliferator-activated receptor-α are associated with the development of new-onset diabetes after transplantation in kidney transplant recipients treated with tacrolimus. Pharmacogenet. Genomics. 2013;23:649–657. doi: 10.1097/FPC.0000000000000001. [DOI] [PubMed] [Google Scholar]
  • 41.Numakura K., Satoh S., Tsuchiya N., Horikawa Y., Inoue T., Kakinuma H., Matsuura S., Saito M., Tada H., Suzuki T., Habuchi T. Clinical and genetic risk factors for posttransplant diabetes mellitus in adult renal transplant recipients treated with tacrolimus. Transplantation. 2005;80:1419–1424. doi: 10.1097/01.tp.0000181142.82649.e3. [DOI] [PubMed] [Google Scholar]
  • 42.Bamoulid J., Courivaud C., Deschamps M., Mercier P., Ferrand C., Penfornis A., Tiberghien P., Chalopin J.M., Saas P., Ducloux D. IL-6 promoter polymorphism -174 is associated with new-onset diabetes after transplantation. J. Am. Soc. Nephrol. 2006;17:2333–2340. doi: 10.1681/ASN.2006010066. [DOI] [PubMed] [Google Scholar]
  • 43.Kang E.S., Kim M.S., Kim Y.S., Hur K.Y., Han S.J., Nam C.M., Ahn C.W., Cha B.S., Kim S.I., Lee H.C. A variant of the transcription factor 7-like 2 (TCF7L2) gene and the risk of posttransplantation diabetes mellitus in renal allograft recipients. Diabetes Care. 2008;31:63–68. doi: 10.2337/dc07-1005. [DOI] [PubMed] [Google Scholar]
  • 44.Kang E.S., Kim M.S., Kim Y.S., Kim C.H., Han S.J., Chun S.W., Hur K.Y., Nam C.M., Ahn C.W., Cha B.S., Kim S.I., Lee H.C. A polymorphism in the zinc transporter gene SLC30A8 confers resistance against posttransplantation diabetes mellitus in renal allograft recipients. Diabetes. 2008;57:1043–1047. doi: 10.2337/db07-0761. [DOI] [PubMed] [Google Scholar]
  • 45.Ghisdal L., Baron C., Le M.Y., Lionet A., Halimi J.M., Rerolle J.P., Glowacki F., Lebranchu Y., Drouet M., Noël C., El H.H., Cochaux P., Wissing K.M., Abramowicz D., Abramowicz M. TCF7L2 polymorphism associates with new-onset diabetes after transplantation. J. Am. Soc. Nephrol. 2009;20:2459–2467. doi: 10.1681/ASN.2008121314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hoorn E.J., Walsh S.B., McCormick J.A., Fürstenberg A., Yang C.L., Roeschel T., Paliege A., Howie A.J., Conley J., Bachmann S., Unwin R.J., Ellison D.H. The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Nat. Med. 2011;17:1304–1309. doi: 10.1038/nm.2497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hesselink D.A., Bouamar R., Elens L., Van Schaik R.H., Van Gelder T. The role of pharmacogenetics in the disposition of and response to tacrolimus in solid organ transplantation. Clin. Pharmacokinet. 2014;53:123–139. doi: 10.1007/s40262-013-0120-3. [DOI] [PubMed] [Google Scholar]
  • 48.Bochud M., Bovet P., Burnier M., Eap C.B. CYP3A5 and ABCB1 genes and hypertension. Pharmacogenomics. 2009;10:477–487. doi: 10.2217/14622416.10.3.477. [DOI] [PubMed] [Google Scholar]
  • 49.Wijdicks E.F. Neurotoxicity of immunosuppressive drugs. Liver Transpl. 2001;7:937–942. doi: 10.1053/jlts.2001.27475. [DOI] [PubMed] [Google Scholar]
  • 50.Yanagimachi M., Naruto T., Tanoshima R., Kato H., Yokosuka T., Kajiwara R., Fujii H., Tanaka F., Goto H., Yagihashi T., Kosaki K., Yokota S. Influence of CYP3A5 and ABCB1 gene polymorphisms on calcineurin inhibitor-related neurotoxicity after hematopoietic stem cell transplantation. Clin. Transplant. 2010;24:855–861. doi: 10.1111/j.1399-0012.2009.01181.x. [DOI] [PubMed] [Google Scholar]
  • 51.Cordon-Cardo C., O’Brien J.P., Casals D., Rittman-Grauer L., Biedler J.L., Melamed M.R., Bertino J.R. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc. Natl. Acad. Sci. USA. 1989;86:695–698. doi: 10.1073/pnas.86.2.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zahir H., McCaughan G., Gleeson M., Nand R.A., McLachlan A.J. Changes in tacrolimus distribution in blood and plasma protein binding following liver transplantation. Ther. Drug Monit. 2004;26:506–515. doi: 10.1097/00007691-200410000-00008. [DOI] [PubMed] [Google Scholar]
  • 53.Press R.R., Ploeger B.A., Den Hartigh J., Van der Straaten T., Van Pelt J., Danhof M., De Fijter J.W., Guchelaar H.J. Explaining variability in tacrolimus pharmacokinetics to optimize early exposure in adult kidney transplant recipients. Ther. Drug Monit. 2009;31:187–197. doi: 10.1097/FTD.0b013e31819c3d6d. [DOI] [PubMed] [Google Scholar]
  • 54.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) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood levels in stable renal transplant patients. Pharmacogenetics. 2004;14:147–154. doi: 10.1097/00008571-200403000-00002. [DOI] [PubMed] [Google Scholar]
  • 55.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 polymorphic CYP3A5 expression. Nat. Genet. 2001;27:383–391. doi: 10.1038/86882. [DOI] [PubMed] [Google Scholar]
  • 56.Hu Y.F., He J., Chen G.L., Wang D., Liu Z.Q., Zhang C., Duan L.F., Zhou H.H. CYP3A5*3 and CYP3A4*18 single nucleotide polymorphisms in a Chinese population. Clin. Chim. Acta. 2005;353:187–192. doi: 10.1016/j.cccn.2004.11.005. [DOI] [PubMed] [Google Scholar]
  • 57.Wei-lin W., Jing J., Shu-sen Z., Li-hua W., Ting-bo L., Song-feng Y., Sheng Y. Tacrolimus dose requirement in relation to donor and recipient ABCB1 and CYP3A5 gene polymorphisms in Chinese liver transplant patients. Liver Transpl. 2006;12:775–780. doi: 10.1002/lt.20709. [DOI] [PubMed] [Google Scholar]
  • 58.MacPhee I.A., Fredericks S., Tai T., Syrris P., Carter N.D., Johnston A., Goldberg L., Holt D.W. The influence of pharmacogenetics on the time to achieve target tacrolimus concentrations after kidney transplantation. Am. J. Transplant. 2004;4:914–919. doi: 10.1111/j.1600-6143.2004.00435.x. [DOI] [PubMed] [Google Scholar]
  • 59.Hesselink D.A., Van Schaik R.H., Van Agteren M., De Fijter J.W., Hartmann A., Zeier M., Budde K., Kuypers D.R., Pisarski P., Le M.Y., Mamelok R.D., Van Gelder T. CYP3A5 genotype is not associated with a higher risk of acute rejection in tacrolimus-treated renal transplant recipients. Pharmacogenet. Genomics. 2008;18:339–348. doi: 10.1097/FPC.0b013e3282f75f88. [DOI] [PubMed] [Google Scholar]
  • 60.Zhang X., Liu Z.H., Zheng J.M., Chen Z.H., Tang Z., Chen J.S., Li L.S. Influence of CYP3A5 and MDR1 polymorphisms on tacrolimus concentration in the early stage after renal transplantation. Clin. Transplant. 2005;19:638–643. doi: 10.1111/j.1399-0012.2005.00370.x. [DOI] [PubMed] [Google Scholar]
  • 61.Roy J.N., Barama A., Poirier C., Vinet B., Roger M. Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients. Pharmacogenet. Genomics. 2006;16:659–665. doi: 10.1097/01.fpc.0000220571.20961.dd. [DOI] [PubMed] [Google Scholar]
  • 62.Renders L., Frisman M., Ufer M., Mosyagin I., Haenisch S., Ott U., Caliebe A., Dechant M., Braun F., Kunzendorf U., Cascorbi I. CYP3A5 genotype markedly influences the pharmacokinetics of tacrolimus and sirolimus in kidney transplant recipients. Clin. Pharmacol. Ther. 2007;81:228–234. doi: 10.1038/sj.clpt.6100039. [DOI] [PubMed] [Google Scholar]
  • 63.Rong G., Jing L., Deng-Qing L., Hong-Shan Z., Shai-Hong Z., Xin-Min N. Influence of CYP3A5 and MDR1(ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in Chinese renal transplant recipients. Transplant. Proc. 2010;42:3455–3458. doi: 10.1016/j.transproceed.2010.08.063. [DOI] [PubMed] [Google Scholar]
  • 64.Glowacki F., Lionet A., Buob D., Labalette M., Allorge D., Provôt F., Hazzan M., Noël C., Broly F., Cauffiez C. CYP3A5 and ABCB1 polymorphisms in donor and recipient: impact on Tacrolimus dose requirements and clinical outcome after renal transplantation. Nephrol. Dial. Transplant. 2011;26:3046–3050. doi: 10.1093/ndt/gfr253. [DOI] [PubMed] [Google Scholar]
  • 65.Ferraresso M., Tirelli A., Ghio L., Grillo P., Martina V., Torresani E., Edefonti A. Influence of the CYP3A5 genotype on tacrolimus pharmacokinetics and pharmacodynamics in young kidney transplant recipients. Pediatr. Transplant. 2007;11:296–300. doi: 10.1111/j.1399-3046.2006.00662.x. [DOI] [PubMed] [Google Scholar]
  • 66.Zhao W., Elie V., Roussey G., Brochard K., Niaudet P., Leroy V., Loirat C., Cochat P., Cloarec S., André J.L., Garaix F., Bensman A., Fakhoury M., Jacqz-Aigrain E. Population pharmacokinetics and pharmacogenetics of tacrolimus in de novo pediatric kidney transplant recipients. Clin. Pharmacol. Ther. 2009;86:609–618. doi: 10.1038/clpt.2009.210. [DOI] [PubMed] [Google Scholar]
  • 67.Thervet E., Loriot M.A., Barbier S., Buchler M., Ficheux M., Choukroun G., Toupance O., Touchard G., Alberti C., Le P.P., Moulin B., Le M.Y., Heng A.E., Subra J.F., Beaune P., Legendre C. Optimization of initial tacrolimus dose using pharmacogenetic testing. Clin. Pharmacol. Ther. 2010;87:721–726. doi: 10.1038/clpt.2010.17. [DOI] [PubMed] [Google Scholar]
  • 68.Pallet N., Etienne I., Buchler M., Bailly E., De Ligny B.H., Choukroun G., Colosio C., Thierry A., Vigneau C., Moulin B., Le M.Y., Heng A.E., Legendre C., Beaune P., Loriot M.A., Thervet E. Long-term clinical impact of adaptation of initial tacrolimus dosing to CYP3A5 genotype. Am. J. Transplant. 2016;16:2670–2675. doi: 10.1111/ajt.13788. [DOI] [PubMed] [Google Scholar]
  • 69.Shuker N., Bouamar R., Van Schaik R.H., Clahsen-van G.M.C., Damman J., Baan C.C., Van de Wetering J., Rowshani A.T., Weimar W., Van Gelder T., Hesselink D.A. A randomized controlled trial comparing the efficacy of Cyp3a5 genotype-based with body-weight-based tacrolimus dosing after living donor kidney transplantation. Am. J. Transplant. 2016;16:2085–2096. doi: 10.1111/ajt.13691. [DOI] [PubMed] [Google Scholar]
  • 70.Kuypers D.R. Pharmacogenetic vs. concentration-controlled optimization of tacrolimus dosing in renal allograft recipients. Clin. Pharmacol. Ther. 2010;88:595–596. doi: 10.1038/clpt.2010.129. [DOI] [PubMed] [Google Scholar]
  • 71.Santoro A., Felipe C.R., Tedesco-Silva H., Medina-Pestana J.O., Struchiner C.J., Ojopi E.B., Suarez-Kurtz G. Pharmacogenetics of calcineurin inhibitors in Brazilian renal transplant patients. Pharmacogenomics. 2011;12:1293–1303. doi: 10.2217/pgs.11.70. [DOI] [PubMed] [Google Scholar]
  • 72.Hustert E., Haberl M., Burk O., Wolbold R., He Y.Q., Klein K., Nuessler A.C., Neuhaus P., Klattig J., Eiselt R., Koch I., Zibat A., Brockmöller J., Halpert J.R., Zanger U.M., Wojnowski L. The genetic determinants of the CYP3A5 polymorphism. Pharmacogenetics. 2001;11:773–779. doi: 10.1097/00008571-200112000-00005. [DOI] [PubMed] [Google Scholar]
  • 73.Knops N., Levtchenko E., Van den Heuvel B., Kuypers D. From gut to kidney: Transporting and metabolizing calcineurin-inhibitors in solid organ transplantation. Int. J. Pharm. 2013;452:14–35. doi: 10.1016/j.ijpharm.2013.05.033. [DOI] [PubMed] [Google Scholar]
  • 74.Hesselink D.A., Van Schaik R.H., Van der Heiden I.P., Van der Werf M., Gregoor P.J., Lindemans J., Weimar W., Van Gelder T. Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Clin. Pharmacol. Ther. 2003;74:245–254. doi: 10.1016/S0009-9236(03)00168-1. [DOI] [PubMed] [Google Scholar]
  • 75.Bandur S., Petrasek J., Hribova P., Novotna E., Brabcova I., Viklicky O. Haplotypic structure of ABCB1/MDR1 gene modifies the risk of the acute allograft rejection in renal transplant recipients. Transplantation. 2008;86:1206–1213. doi: 10.1097/TP.0b013e318187c4d1. [DOI] [PubMed] [Google Scholar]
  • 76.Wang D., Guo Y., Wrighton S.A., Cooke G.E., Sadee W. Intronic polymorphism in CYP3A4 affects hepatic expression and response to statin drugs. Pharmacogenomics J. 2011;11:274–286. doi: 10.1038/tpj.2010.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gijsen V.M., Van Schaik R.H., Elens L., Soldin O.P., Soldin S.J., Koren G., De Wildt S.N. CYP3A4*22 and CYP3A combined genotypes both correlate with tacrolimus disposition in pediatric heart transplant recipients. Pharmacogenomics. 2013;14:1027–1036. doi: 10.2217/pgs.13.80. [DOI] [PubMed] [Google Scholar]
  • 78.Chiu K.W., Hu T.H., Nakano T., Chen K.D., Lai C.Y., Hsu L.W., Tseng H.P., Chiu H.C., Cheng Y.F., Goto S., Chen C.L. Biological interactions of CYP2C19 genotypes with CYP3A4*18, CYP3A5*3, and MDR1-3435 in living donor liver transplantation recipients. Transplant. Res. 2013;2:6. doi: 10.1186/2047-1440-2-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Werk A.N., Cascorbi I. Functional gene variants of CYP3A4. Clin. Pharmacol. Ther. 2014;96:340–348. doi: 10.1038/clpt.2014.129. [DOI] [PubMed] [Google Scholar]
  • 80.Werk A.N., Lefeldt S., Bruckmueller H., Hemmrich-Stanisak G., Franke A., Roos M., Küchle C., Steubl D., Schmaderer C., Bräsen J.H., Heemann U., Cascorbi I., Renders L. Identification and characterization of a defective CYP3A4 genotype in a kidney transplant patient with severely diminished tacrolimus clearance. Clin. Pharmacol. Ther. 2014;95:416–422. doi: 10.1038/clpt.2013.210. [DOI] [PubMed] [Google Scholar]
  • 81.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 requirements and trough blood levels in stable renal transplant patients. Pharmacogenomics. 2011;12:1383–1396. doi: 10.2217/pgs.11.90. [DOI] [PubMed] [Google Scholar]
  • 82.Elens L., Bouamar R., Shuker N., Hesselink D.A., van Gelder T., van Schaik R.H. Clinical implementation of pharmacogenetics in kidney transplantation: calcineurin inhibitors in the starting blocks. Br. J. Clin. Pharmacol. 2014;77:715–728. doi: 10.1111/bcp.12253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hoffmeyer S., Burk O. von, R. O.; Arnold, H. P.; Brockmöller, J.; Johne, A.; Cascorbi, I.; Gerloff, T.; Roots, I.; Eichelbaum, M.; Brinkmann, U. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc. Natl. Acad. Sci. USA. 2000;97:3473–3478. doi: 10.1073/pnas.050585397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang D., Johnson A.D., Papp A.C., Kroetz D.L., Sadée W. Multidrug resistance polypeptide 1 (MDR1, ABCB1) variant 3435C>T affects mRNA stability. Pharmacogenet. Genomics. 2005;15:693–704. [PubMed] [Google Scholar]
  • 85.Kimchi-Sarfaty C., Oh J.M., Kim I.W., Sauna Z.E., Calcagno A.M., Ambudkar S.V., Gottesman M.M.A. “silent” polymorphism in the MDR1 gene changes substrate specificity. Science. 2007;315:525–528. doi: 10.1126/science.1135308. [DOI] [PubMed] [Google Scholar]
  • 86.Chen B., Fang J., Zhang W., Jin Z., Yu Z., Cai W. Detection of C1236T, G2677T/A, and C3435T polymorphism of MDR1 by amplification refractory mutation system PCR. J. Clin. Lab. Anal. 2009;23:110–116. doi: 10.1002/jcla.20299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Anglicheau D., Verstuyft C., Laurent-Puig P., Becquemont L., Schlageter M.H., Cassinat B., Beaune P., Legendre C., Thervet E. Association of the multidrug resistance-1 gene single-nucleotide polymorphisms with the tacrolimus dose requirements in renal transplant recipients. J. Am. Soc. Nephrol. 2003;14:1889–1896. doi: 10.1097/01.asn.0000073901.94759.36. [DOI] [PubMed] [Google Scholar]
  • 88.Vafadari R., Bouamar R., Hesselink D.A., Kraaijeveld R., van Schaik R.H., Weimar W., Baan C.C., van Gelder T. Genetic polymorphisms in ABCB1 influence the pharmacodynamics of tacrolimus. Ther. Drug Monit. 2013;35:459–465. doi: 10.1097/FTD.0b013e31828c1581. [DOI] [PubMed] [Google Scholar]
  • 89.Capron A., Mourad M., De Meyer M., De Pauw L., Eddour D.C., Latinne D., Elens L., Haufroid V., Wallemacq P. CYP3A5 and ABCB1 polymorphisms influence tacrolimus concentrations in peripheral blood mononuclear cells after renal transplantation. Pharmacogenomics. 2010;11:703–714. doi: 10.2217/pgs.10.43. [DOI] [PubMed] [Google Scholar]
  • 90.Hart S.N., Zhong X.B. P450 oxidoreductase: genetic polymorphisms and implications for drug metabolism and toxicity. Expert Opin. Drug Metab. Toxicol. 2008;4:439–452. doi: 10.1517/17425255.4.4.439. [DOI] [PubMed] [Google Scholar]
  • 91.Elens L., Nieuweboer A.J., Clarke S.J., Charles K.A., de Graan A.J., Haufroid V., van Gelder T., Mathijssen R.H., van Schaik R.H. Impact of POR*28 on the clinical pharmacokinetics of CYP3A phenotyping probes midazolam and erythromycin. Pharmacogenet. Genomics. 2013;23:148–155. doi: 10.1097/FPC.0b013e32835dc113. [DOI] [PubMed] [Google Scholar]
  • 92.Zhang J.J., Zhang H., Ding X.L., Ma S., Miao L.Y. Effect of the P450 oxidoreductase 28 polymorphism on the pharmacokinetics of tacrolimus in Chinese healthy male volunteers. Eur. J. Clin. Pharmacol. 2013;69:807–812. doi: 10.1007/s00228-012-1432-1. [DOI] [PubMed] [Google Scholar]
  • 93.Lesche D., Sigurdardottir V., Setoud R., Oberhänsli M., Carrel T., Fiedler G.M., Largiadèr C.R., Mohacsi P., Sistonen J. CYP3A5*3 and POR*28 genetic variants influence the required dose of tacrolimus in heart transplant recipients. Ther. Drug Monit. 2014;36:710–715. doi: 10.1097/FTD.0000000000000080. [DOI] [PubMed] [Google Scholar]
  • 94.de Jonge H., Metalidis C., Naesens M., Lambrechts D., Kuypers D.R. The P450 oxidoreductase *28 SNP is associated with low initial tacrolimus exposure and increased dose requirements in CYP3A5-expressing renal recipients. Pharmacogenomics. 2011;12:1281–1291. doi: 10.2217/pgs.11.77. [DOI] [PubMed] [Google Scholar]
  • 95.Elens L., Hesselink D.A., Bouamar R., Budde K., de Fijter J.W., De Meyer M., Mourad M., Kuypers D.R., Haufroid V., Van Gelder T., Van Schaik R.H. Impact of POR*28 on the pharmacokinetics of tacrolimus and cyclosporine A in renal transplant patients. Ther. Drug Monit. 2014;36:71–79. doi: 10.1097/FTD.0b013e31829da6dd. [DOI] [PubMed] [Google Scholar]
  • 96.Gijsen V.M., van Schaik R.H., Soldin O.P., Soldin S.J., Nulman I., Koren G., De Wildt S.N. P450 oxidoreductase *28 (POR*28) and tacrolimus disposition in pediatric kidney transplant recipients-a pilot study. Ther. Drug Monit. 2014;36:152–158. doi: 10.1097/FTD.0b013e3182a3f282. [DOI] [PubMed] [Google Scholar]
  • 97.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. Pharmacol. 2014;70:685–693. doi: 10.1007/s00228-014-1656-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Benkali K., Prémaud A., Picard N., Rérolle J.P., Toupance O., Hoizey G., Turcant A., Villemain F., Le M.Y., Marquet P., Rousseau A. Tacrolimus population pharmacokinetic-pharmacogenetic analysis and Bayesian estimation in renal transplant recipients. Clin. Pharmacokinet. 2009;48:805–816. doi: 10.2165/11318080-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 99.Barraclough K.A., Isbel N.M., Lee K.J., Bergmann T.K., Johnson D.W., McWhinney B.C., Ungerer J.P., Campbell S.B., Leary D.R., Bialasiewicz S., Rockett R.J., Staatz C.E. NR1I2 polymorphisms are related to tacrolimus dose-adjusted exposure and BK viremia in adult kidney transplantation. Transplantation. 2012;94:1025–1032. doi: 10.1097/TP.0b013e31826c3985. [DOI] [PubMed] [Google Scholar]
  • 100.Uno Y., Sakamoto Y., Yoshida K., Hasegawa T., Hasegawa Y., Koshino T., Inoue I. Characterization of six base pair deletion in the putative HNF1-binding site of human PXR promoter. J. Hum. Genet. 2003;48:594–597. doi: 10.1007/s10038-003-0076-5. [DOI] [PubMed] [Google Scholar]
  • 101.Liu Y., Ji W., Yin Y., Fan L., Zhang J., Yun H., Wang N., Li Q., Wei Z., Ouyang D., Zhou H.H. The effects of splicing variant of PXR PAR-2 on CYP3A4 and MDR1 mRNA expressions. Clin. Chim. Acta. 2009;403:142–144. doi: 10.1016/j.cca.2009.02.006. [DOI] [PubMed] [Google Scholar]
  • 102.Wang Z.P., Zhao M., Qu Q.S., Miao S.Z. Effect of pregnane X receptor polymorphisms on tacrolimus blood concentrations and the resulting adverse reactions in kidney transplantation recipients. Genet. Mol. Res. 2016;15(3):15038464. doi: 10.4238/gmr.15038464. [DOI] [PubMed] [Google Scholar]
  • 103.Klein K., Thomas M., Winter S., Nussler A.K., Niemi M., Schwab M., Zanger U.M. PPARA: a novel genetic determinant of CYP3A4 in vitro and in vivo. Clin. Pharmacol. Ther. 2012;91:1044–1052. doi: 10.1038/clpt.2011.336. [DOI] [PubMed] [Google Scholar]
  • 104.Kurzawski M., Malinowski D., Dziewanowski K., Droździk M. Impact of PPARA and POR polymorphisms on tacrolimus pharmacokinetics and new-onset diabetes in kidney transplant recipients. Pharmacogenet. Genomics. 2014;24:397–400. doi: 10.1097/FPC.0000000000000067. [DOI] [PubMed] [Google Scholar]
  • 105.Bruckmueller H., Werk A.N., Renders L., Feldkamp T., Tepel M., Borst C., Caliebe A., Kunzendorf U., Cascorbi I. Which genetic determinants should be considered for tacrolimus dose optimization in kidney transplantation? A combined analysis of genes affecting the CYP3A locus. Ther. Drug Monit. 2015;37:288–295. doi: 10.1097/FTD.0000000000000142. [DOI] [PubMed] [Google Scholar]
  • 106.Ogasawara K., Chitnis S.D., Gohh R.Y., Christians U., Akhlaghi F. Multidrug resistance-associated protein 2 (MRP2/ABCC2) haplotypes significantly affect the pharmacokinetics of tacrolimus in kidney transplant recipients. Clin. Pharmacokinet. 2013;52:751–762. doi: 10.1007/s40262-013-0069-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.FDV G.; Nishikawa, A. M.; Felipe, C. R.; Tedesco-Silva, H.; Oliveira, N.; ABC, S.; Medina-Pestana, J. O.; Doi, S. Q.; Hirata, M. H.; RDC, H. Influence of ABCC2, CYP2C8, and CYP2J2 polymorphisms on tacrolimus and mycophenolate sodium-based treatment in brazilian kidney transplant recipients. Pharmacotherapy. 2017;37(5):535–545. doi: 10.1002/phar.1928. [DOI] [PubMed] [Google Scholar]
  • 108.Aquilante C.L., Niemi M., Gong L., Altman R.B., Klein T.E. PharmGKB summary: very important pharmacogene information for cytochrome P450, family 2, subfamily C, polypeptide 8. Pharmacogenet. Genomics. 2013;23:721–728. doi: 10.1097/FPC.0b013e3283653b27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Suarez-Kurtz G., Pena S.D., Struchiner C.J., Hutz M.H. Pharmacogenomic diversity among brazilians: influence of ancestry, self-reported color, and geographical origin. Front. Pharmacol. 2012;3:191. doi: 10.3389/fphar.2012.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fan J., Zhang X., Ren L., Chen D., Wu S., Guo F., Qin S., Wang Z., Lin Z., Xing T., Sun X., Peng Z. Donor IL-18 rs5744247 polymorphism as a new biomarker of tacrolimus elimination in Chinese liver transplant patients during the early post-transplantation period: results from two cohort studies. Pharmacogenomics. 2015;16:239–250. doi: 10.2217/pgs.14.166. [DOI] [PubMed] [Google Scholar]
  • 111.Zhang X., Wang Z., Fan J., Liu G., Peng Z. Impact of interleukin-10 gene polymorphisms on tacrolimus dosing requirements in Chinese liver transplant patients during the early posttransplantation period. Eur. J. Clin. Pharmacol. 2011;67:803–813. doi: 10.1007/s00228-011-0993-8. [DOI] [PubMed] [Google Scholar]
  • 112.Liu M.Z., He H.Y., Zhang Y.L., Hu Y.F., He F.Z., Luo J.Q., Luo Z.Y., Chen X.P., Liu Z.Q., Zhou H.H., Shao M.J., Ming Y.Z., Xin H.W., Zhang W. IL-3 and CTLA4 gene polymorphisms may influence the tacrolimus dose requirement in Chinese kidney transplant recipients. Acta Pharmacol. Sin. 2017;38:415–423. doi: 10.1038/aps.2016.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tang J., Liu R., Zhang Y.L., Liu M.Z., Hu Y.F., Shao M.J., Zhu L.J., Xin H.W., Feng G.W., Shang W.J., Meng X.G., Zhang L.R., Ming Y.Z., Zhang W. Application of machine-learning models to predict tacrolimus stable dose in renal transplant recipients. Sci. Rep. 2017;7:42192. doi: 10.1038/srep42192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.de Jonge H., de Loor H., Verbeke K., Vanrenterghem Y., Kuypers D.R. In vivo CYP3A4 activity, CYP3A5 genotype, and hematocrit predict tacrolimus dose requirements and clearance in renal transplant patients. Clin. Pharmacol. Ther. 2012;92:366–375. doi: 10.1038/clpt.2012.109. [DOI] [PubMed] [Google Scholar]
  • 115.Jacobson P.A., Oetting W.S., Brearley A.M., Leduc R., Guan W., Schladt D., Matas A.J., Lamba V., Julian B.A., Mannon R.B., Israni A. Novel polymorphisms associated with tacrolimus trough concentrations: Results from a multicenter kidney transplant consortium. Transplantation. 2011;91:300–308. doi: 10.1097/TP.0b013e318200e991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.van Gelder T., Hesselink D.A. Dosing tacrolimus based on CYP3A5 genotype: Will it improve clinical outcome. Clin. Pharmacol. Ther. 2010;87:640–641. doi: 10.1038/clpt.2010.42. [DOI] [PubMed] [Google Scholar]
  • 117.Li J.L., Wang X.D., Chen S.Y., Liu L.S., Fu Q., Chen X., Teng L.C., Wang C.X., Huang M. Effects of diltiazem on pharmacokinetics of tacrolimus in relation to CYP3A5 genotype status in renal recipients: from retrospective to prospective. Pharmacogenomics J. 2011;11:300–306. doi: 10.1038/tpj.2010.42. [DOI] [PubMed] [Google Scholar]
  • 118.Passey C., Birnbaum A.K., Brundage R.C., Schladt D.P., Oetting W.S., Leduc R.E., Israni A.K., Guan W., Matas A.J., Jacobson P.A. Validation of tacrolimus equation to predict troughs using genetic and clinical factors. Pharmacogenomics. 2012;13:1141–1147. doi: 10.2217/pgs.12.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Passey C., Birnbaum A.K., Brundage R.C., Oetting W.S., Israni A.K., Jacobson P.A. Dosing equation for tacrolimus using genetic variants and clinical factors. Br. J. Clin. Pharmacol. 2011;72:948–957. doi: 10.1111/j.1365-2125.2011.04039.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Provenzani A., Notarbartolo M., Labbozzetta M., Poma P., Vizzini G., Salis P., Caccamo C., Bertani T., Palazzo U., Polidori P., Gridelli B., D’Alessandro N. Influence of CYP3A5 and ABCB1 gene polymorphisms and other factors on tacrolimus dosing in Caucasian liver and kidney transplant patients. Int. J. Mol. Med. 2011;28:1093–1102. doi: 10.3892/ijmm.2011.794. [DOI] [PubMed] [Google Scholar]
  • 121.Li L., Li C.J., Zheng L., Zhang Y.J., Jiang H.X., Si-Tu B., Li Z.H. Tacrolimus dosing in Chinese renal transplant recipients: a population-based pharmacogenetics study. Eur. J. Clin. Pharmacol. 2011;67:787–795. doi: 10.1007/s00228-011-1010-y. [DOI] [PubMed] [Google Scholar]
  • 122.Kim I.W., Moon Y.J., Ji E., Kim K.I., Han N., Kim S.J., Shin W.G., Ha J., Yoon J.H., Lee H.S., Oh J.M. Clinical and genetic factors affecting tacrolimus trough levels and drug-related outcomes in Korean kidney transplant recipients. Eur. J. Clin. Pharmacol. 2012;68:657–669. doi: 10.1007/s00228-011-1182-5. [DOI] [PubMed] [Google Scholar]
  • 123.Wang P., Mao Y., Razo J., Zhou X., Wong S.T., Patel S., Elliott E., Shea E., Wu A.H., Gaber A.O. Using genetic and clinical factors to predict tacrolimus dose in renal transplant recipients. Pharmacogenomics. 2010;11:1389–1402. doi: 10.2217/pgs.10.105. [DOI] [PubMed] [Google Scholar]
  • 124.Boughton O., Borgulya G., Cecconi M., Fredericks S., Moreton-Clack M., MacPhee I.A. A published pharmacogenetic algorithm was poorly predictive of tacrolimus clearance in an independent cohort of renal transplant recipients. Br. J. Clin. Pharmacol. 2013;76:425–431. doi: 10.1111/bcp.12076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Elens L., Hesselink D.A., Van Schaik R.H., Van Gelder T. The CYP3A4*22 allele affects the predictive value of a pharmacogenetic algorithm predicting tacrolimus predose concentrations. Br. J. Clin. Pharmacol. 2013;75:1545–1547. doi: 10.1111/bcp.12038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Xie H.G., Wood A.J., Kim R.B., Stein C.M., Wilkinson G.R. Genetic variability in CYP3A5 and its possible consequences. Pharmacogenomics. 2004;5:243–272. doi: 10.1517/phgs.5.3.243.29833. [DOI] [PubMed] [Google Scholar]
  • 127.Liu Y.T., Hao H.P., Liu C.X., Wang G.J., Xie H.G. Drugs as CYP3A probes, inducers, and inhibitors. Drug Metab. Rev. 2007;39:699–721. doi: 10.1080/03602530701690374. [DOI] [PubMed] [Google Scholar]
  • 128.Lai Y., Zhang J., Wang Y.X., Wang X.D., Li J.L., Wang Y.H., Zeng Y.J., Huang M. CYP3A5*3 and MDR-1 C3435T single nucleotide polymorphisms in six Chinese ethnic groups. Pharmazie. 2011;66:136–140. [PubMed] [Google Scholar]
  • 129.Tang J.T., Andrews L.M., Van Gelder T., Shi Y.Y., van Schaik R.H., Wang L.L., Hesselink D.A. Pharmacogenetic aspects of the use of tacrolimus in renal transplantation: Recent developments and ethnic considerations. Expert Opin. Drug Metab. Toxicol. 2016;12:555–565. doi: 10.1517/17425255.2016.1170808. [DOI] [PubMed] [Google Scholar]
  • 130.Birdwell K.A., Decker B., Barbarino J.M., Peterson J.F., Stein C.M., Sadee W., Wang D., Vinks A.A., He Y., Swen J.J., Leeder J.S., van Schaik R., Thummel K.E., Klein T.E., Caudle K.E., MacPhee I.A. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP3A5 Genotype and Tacrolimus Dosing. Clin. Pharmacol. Ther. 2015;98:19–24. doi: 10.1002/cpt.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Díaz-Molina B., Tavira B., Lambert J.L., Bernardo M.J., Alvarez V., Coto E. Effect of CYP3A5, CYP3A4, and ABCB1 genotypes as determinants of tacrolimus dose and clinical outcomes after heart transplantation. Transplant. Proc. 2012;44:2635–2638. doi: 10.1016/j.transproceed.2012.09.062. [DOI] [PubMed] [Google Scholar]
  • 132.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 subjects: a population pharmacokinetic analysis. J. Clin. Pharm. Ther. 2011;36:614–624. doi: 10.1111/j.1365-2710.2010.01206.x. [DOI] [PubMed] [Google Scholar]
  • 133.Quteineh L., Verstuyft C., Furlan V., Durrbach A., Letierce A., Ferlicot S., Taburet A.M., Charpentier B., Becquemont L. Influence of CYP3A5 genetic polymorphism on tacrolimus daily dose requirements and acute rejection in renal graft recipients. Basic Clin. Pharmacol. Toxicol. 2008;103:546–552. doi: 10.1111/j.1742-7843.2008.00327.x. [DOI] [PubMed] [Google Scholar]
  • 134.Torio A., Auyanet I., Montes-Ares O., Guerra R.M., Fernandez E.J., Perez M.A., Ramirez A., Checa M.D. Effect of CYP3A51/3 polymorphism on blood pressure in renal transplant recipients. Transplant. Proc. 2012;44:2596–2598. doi: 10.1016/j.transproceed.2012.09.047. [DOI] [PubMed] [Google Scholar]
  • 135.Ferraresso M., Turolo S., Ghio L., Tirelli A.S., Belingheri M., Villa R., Groppali E., Edefonti A. Association between CYP3A5 polymorphisms and blood pressure in kidney transplant recipients receiving calcineurin inhibitors. Clin. Exp. Hypertens. 2011;33:359–365. doi: 10.3109/10641963.2011.561896. [DOI] [PubMed] [Google Scholar]
  • 136.Moore J., McKnight A.J., Döhler B., Simmonds M.J., Courtney A.E., Brand O.J., Briggs D., Ball S., Cockwell P., Patterson C.C., Maxwell A.P., Gough S.C., Opelz G., Borrows R. Donor ABCB1 variant associates with increased risk for kidney allograft failure. J. Am. Soc. Nephrol. 2012;23:1891–1899. doi: 10.1681/ASN.2012030260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Yamauchi A., Ieiri I., Kataoka Y., Tanabe M., Nishizaki T., Oishi R., Higuchi S., Otsubo K., Sugimachi K. Neurotoxicity induced by tacrolimus after liver transplantation: relation to genetic polymorphisms of the ABCB1 (MDR1) gene. Transplantation. 2002;74:571–572. doi: 10.1097/00007890-200208270-00024. [DOI] [PubMed] [Google Scholar]
  • 138.van de Wetering J., Weimar C.H., Balk A.H., Roodnat J.I., Holweg C.T., Baan C.C., van Domburg R.T., Weimar W. The impact of transforming growth factor-beta1 gene polymorphism on end-stage renal failure after heart transplantation. Transplantation. 2006;82:1744–1748. doi: 10.1097/01.tp.0000250360.78553.5e. [DOI] [PubMed] [Google Scholar]
  • 139.Lácha J., Hubácek J.A., Viklický O., Málek I., Hutchinson I., Vítko S. TGF-beta1 gene polymorphism is a risk factor for renal dysfunction in heart transplant recipients. Transplant. Proc. 2001;33:1567–1569. doi: 10.1016/s0041-1345(00)02596-3. [DOI] [PubMed] [Google Scholar]
  • 140.Baan C.C., Balk A.H., Holweg C.T., van Riemsdijk I.C., Maat L.P., Vantrimpont P.J., Niesters H.G., Weimar W. Renal failure after clinical heart transplantation is associated with the TGF-beta 1 codon 10 gene polymorphism. J. Heart Lung Transplant. 2000;19:866–872. doi: 10.1016/s1053-2498(00)00155-8. [DOI] [PubMed] [Google Scholar]
  • 141.Smith H.E., Jones J.P., Kalhorn T.F., Farin F.M., Stapleton P.L., Davis C.L., Perkins J.D., Blough D.K., Hebert M.F., Thummel K.E., Totah R.A. Role of cytochrome P450 2C8 and 2J2 genotypes in calcineurin inhibitor-induced chronic kidney disease. Pharmacogenet. Genomics. 2008;18:943–953. doi: 10.1097/FPC.0b013e32830e1e16. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Current Drug Metabolism are provided here courtesy of Bentham Science Publishers

RESOURCES