Skip to main content

REVIEW article

Front. Pharmacol., 31 July 2020
Sec. Pharmacogenetics and Pharmacogenomics
This article is part of the Research Topic Recent Developments in Pharmacogenetics and Pharmacogenomics View all 11 articles

The Clinical Impact of the C0/D Ratio and the CYP3A5 Genotype on Outcome in Tacrolimus Treated Kidney Transplant Recipients

Teun van Gelder*Teun van Gelder1*Soufian Meziyerh,Soufian Meziyerh2,3Jesse J. SwenJesse J. Swen1Aiko P. J. de Vries,Aiko P. J. de Vries2,3Dirk Jan A. R. MoesDirk Jan A. R. Moes1
  • 1Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, Netherlands
  • 2Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, Netherlands
  • 3Leiden Transplant Center, Leiden University Medical Center, Leiden, Netherlands

Tacrolimus is metabolized by CYP3A4 and CYP3A5 enzymes. Patients expressing CYP3A5 (in Caucasian patients about 15% of the population but more frequent in African Americans and Asians) have a dose requirement that is around 50% higher than non-expressers to reach the target concentration. CYP3A5 expressers can be considered fast metabolizers. The trough concentration/dose (C0/D) ratio of tacrolimus has recently been proposed as a prognostic marker for poor outcome after kidney transplantation. Patients with a low C0/D ratio (also referred to as fast metabolizers) seem to have more tacrolimus-related nephrotoxicity, more BK-viremia, and a lower graft survival. At first sight, the expression of CYP3A5 and a low C0/D ratio seem to be overlapping factors, both pointing towards patients in whom a higher tacrolimus dose is needed to reach the tacrolimus target concentration. However, there are important differences, and these differences may explain why the impact of the C0/D ratio on long term outcome is stronger than for CYP3A5 genotype status. Patients with a low C0/D ratio require a high tacrolimus dose and are exposed to high tacrolimus peak concentrations. The higher peak exposure to tacrolimus (and/or its metabolites) may explain the higher incidence of nephrotoxicity, BK-viremia and graft loss. A potential confounder is the concurrent maintenance treatment of corticosteroids, as steroids are sometimes continued in patients at high immunological risk. Steroids induce the metabolism of tacrolimus via pregnane X receptor mediated increased CYP3A4 expression, resulting in lower tacrolimus C0/D ratio in high risk patients. Also non-adherence may result in lower C0/D ratio which is also associated with poor outcome. The C0/D ratio of tacrolimus does seem to identify a group of patients with increased risk of poor outcome after kidney transplantation. Our recommendation is to monitor tacrolimus peak concentrations in these patients, and if these are high then target slightly lower pre-dose concentrations. Another possibility would be to switch to a prolonged release formulation or to dose the drug more frequently, in smaller doses, to avoid high peak concentrations.

Introduction

Tacrolimus is the first choice calcineurin inhibitor (CNI) in kidney transplant patients. Maintaining the tacrolimus exposure within the therapeutic window is considered to be essential to prevent the development of cellular and antibody-mediated rejections and to minimize drug-related toxicity.

The pharmacokinetics (PK) of tacrolimus is characterized by poor and highly variable oral bioavailability (mean 25%, range 5–90%) (Shuker et al., 2015). Also drug–drug interactions, epigenetic changes in the expression of metabolizing enzymes and patient adherence contribute to a large inter-patient and intra-patient variability in tacrolimus exposure (Vanhove et al., 2016). An important part of the inter-patient variability is explained by the presence of a single nucleotide polymorphism (SNP) in the gene encoding for the cytochrome P450 (CYP) 3A5 enzyme (6986A>G). Patients expressing CYP3A5 (those carrying the A nucleotide, defined as the *1 allele) have a dose requirement that is around 50% higher than non-expressers (those homozygous for the G nucleotide, defined as the *3 allele) (Hesselink et al., 2014). Also the CYP3A4 gene carries a SNP (SNP in intron 6 (rs35599367C>T)) that is significantly associated with the tacrolimus dose requirement, but to a lesser degree than the CYP3A5 gene polymorphism (Elens et al., 2011). Genome wide association studies have shown that there are no other common single genetic variants aside from the CYP3A gene that significantly influences the tacrolimus PK (Oetting et al., 2018; Oetting et al., 2019).

Intra-patient variability in tacrolimus exposure is easily identified by repetitive measurement of drug concentrations in patients on maintenance treatment. Ten years ago Borra et al. demonstrated that patients with high intra-patient variability more often reached a composite endpoint consisting of graft loss, biopsy-proven chronic allograft nephropathy, and ‘doubling in plasma creatinine concentration in the period between t = 12 months post-transplantation and last follow-up’ (Borra et al., 2010). A high intra-patient variability in tacrolimus exposure is now recognized as a predictor of poor clinical outcome (van Gelder, 2014; Mendoza Rojas et al., 2019). Rodrigo et al. found that a higher intra-patient variability was independently related to development of donor-specific antibodies and graft-loss (Rodrigo et al., 2016). Identification of patients with a high intra-patient variability is therefore important as it is a modifiable risk factor, and interventions may improve long-term outcomes (Neuberger et al., 2017).

Concentration/Dose (C0/D) Ratio and Outcome

More recently the concentration/dose (C0/D) ratio of tacrolimus has also been proposed as a prognostic marker for poor outcome. The C0/D ratio can be calculated by dividing the tacrolimus pre-dose concentration (C0) by the corresponding daily tacrolimus dose (D).

Formula: C0/D ratio=tacrolimus trough concentration (ng/mL)/daily dose (mg)

Thölking et al. were the first to hypothesize that the metabolization rate of tacrolimus, expressed as the C0/D ratio, would be a prognostic factor of clinical outcome (Thölking et al., 2014). The mean of the C0/D ratios calculated at months 1, 3, and 6 after renal transplantation was used to categorize patients as fast, intermediate, and slow metabolizers. The incidence of T cell-mediated rejection or antibody-mediated rejection was not related to the tacrolimus metabolizer status, but in the group of fast metabolizers more often CNI nephrotoxicity (p = 0.015) and BK-virus associated nephropathy (p = 0.024) were observed. They concluded that the tacrolimus C0/D ratio is a simple and inexpensive tool to identify patients at risk for the development of CNI nephrotoxicity or BK nephropathy. Moreover, a C0/D ratio < 1.05 was also associated with a higher mortality in a 24 months follow-up. In a five-year follow-up study from the same group the patient survival was noticeably reduced in fast metabolizers as compared to intermediate/slow metabolizers (89.9 vs. 95.3%, log-rank p = 0.036), and in a Cox regression analysis fast metabolizer status was an independent predictor of both graft and patient survival (Schütte-Nütgen et al., 2019). The suggested intervention could be to switch fast Tac metabolizers from tacrolimus based therapy to treatment with a mammalian target of rapamycin inhibitor (mTORi) or cyclosporine, but there is no evidence that this intervention does improve long term outcome. Although the authors do acknowledge that tacrolimus metabolism is also related to the CYP3A5 genotype, they claim that the prognostic value of the C0/D ratio is stronger than that of the genotype.

In a subsequent study, patients were categorized into three metabolizer groups based on the same cut-off values. Patients with a tacrolimus C0/D ratio < 1.05 ng/ml/mg were characterized as fast metabolizers, patients with a C0/D ratio of 1.05–1.54 ng/ml/mg as intermediate metabolizers, and those with a C0/D ratio ≥ 1.55 ng/ml/mg were defined as slow metabolizers (Thölking et al., 2016). Also in this study a fast tacrolimus metabolism was associated with increased risk of BK viremia. The potential explanation for the effect of the metabolizer status on outcome is that in patients with a faster metabolism the drug dose required to reach the target tacrolimus trough concentration is higher. As a result, the tacrolimus peak levels in the first hours after oral administration are higher. Evidence for this hypothesis was obtained in an additional study in 56 renal transplant recipients, in whom the tacrolimus concentrations 2 h after drug intake (C2) in patients with a low C0/D ratio (high metabolizers) were increased compared to the other patients (20.2 ± 10.3 ng/ml vs. 9.8 ± 4.2 ng/ml, respectively; p = 0.004) (Thölking et al., 2019). In daily practice most centers only monitor pre-dose tacrolimus concentrations, and the higher peak levels often go unnoticed.

A Polish group recently also reported impaired outcome data in patients with a low C0/D ratio from a large group of 571 renal transplant patients (Kwiatkowska et al., 2019). In these patients the C0/D ratio was calculated at their most recent out-patient appointment (mean time after transplantation = 84 months), and this ratio was then correlated with the change in renal function from transplantation to last follow-up. Also in this study a higher metabolization status was associated with a significantly greater drop in the eGFR. In a smaller study, also from Poland, the highly significant relationship between C0/D ratio at 6 months and kidney function 2 years after transplantation was confirmed in a linear regression model (p = 0.007) (Nowicka et al., 2019).

In 2020 Jouve et al. published a retrospective study on more than 1,000 kidney transplant patients treated with tacrolimus and with more than 1 year follow-up (Jouve et al., 2020). This study was called the TOMATO study, which stands for TacrOlimus MetAbolization in kidney TransplantatiOn. In a multivariate analysis the C0/D ratio at month 3 and month 6 proved to be independent of early predictors of death-censored kidney graft survival. The authors stressed the importance of mechanistic studies to understand how the C0/D ratio causes its effect.

Taber et al. studied the impact of tacrolimus pharmacokinetics in African Americans (AAs) (Taber et al., 2015). In AAs with sub-therapeutic tacrolimus concentrations, the incidence of acute cellular rejection and of antibody-mediated rejection was increased. But also patients who did achieve therapeutic tacrolimus concentrations were at an increased risk of developing interstitial fibrosis and tubular atrophy (IF/TA), reflecting nephrotoxicity. Most likely, in these AA patients therapeutic tacrolimus concentrations are reached at the cost of high drug doses, and the high peak concentrations inherent to these dosages result in more nephrotoxicity.

A faster metabolization of tacrolimus will result in higher concentrations of tacrolimus metabolites. These metabolites may accumulate in the blood and or renal tubular cells and cause nephrotoxicity. Compared to nonexpressers, the CYP3A5 expressers have a 2.0- to 2.7-fold higher metabolite/parent AUC ratio for these metabolites (Zheng et al., 2012). As several immunoassays suffer from cross-reactivity of these metabolites (without reporting a concentration of the individual metabolites), the mean of the tacrolimus concentrations measured with these immunoassays is higher than if measured with mass spectrometry based assays (Akamine et al., 2018). A survey in 2015 showed that for TDM of tacrolimus 53% of the laboratories used LC–MS/MS and 47% immunoassays (n = 72) (Christians et al., 2015). Accumulation of tacrolimus metabolites in the blood may go unnoticed if TDM is done exclusively on the basis of LC–MS/MS.

CYP3A5 Genotype and C0/D Ratio

As already mentioned, there are multiple studies that show that renal transplant patients who express the CYP3A5 enzyme need a higher dose to reach the target tacrolimus concentration compared to non-expressers (Hesselink et al., 2003; Thervet et al., 2003; Haufroid et al., 2004). Thus, CYP3A5 expressers in general have a low C0/D ratio. In a Caucasian population the prevalence of patients expressing CYP3A5 is low (10%) (van Schaik et al., 2002). In contrast, in patients from African descent about 40–50% express CYP3A5, and in Asian patients the prevalence of CYP3A5 expressers is even higher (50–70%) (Andrews et al., 2016).

In most centers the starting dose of tacrolimus is based on body weight. Standard tacrolimus dosing is 0.2 mg/kg bodyweight, divided in two doses. The guideline of the Clinical Pharmacogenetics Implementation Consortium (CPIC) recommends to increase the starting dose in CYP3A5 expressers 1.5–2 fold, but not to exceed 0.3 mg/kg/day (Birdwell et al., 2015). The higher starting dose is meant to avoid early underexposure to tacrolimus in the first days after transplantation.

CYP3A5 and Outcome

In 1994 an analysis was published on differences in outcome of black and white patients after kidney transplantation based on a large dataset from the United Network for Organ Sharing (UNOS). An impaired graft survival in black patients was found. The difference in outcome was attributed to poor socioeconomic factors and reduced access to health care in African Americans and to poor HLA-matching (Koyama et al., 1994). In this study CYP3A5 genotype data were not available. A French study demonstrated that ethnic origin did not affect outcome after renal transplantation in France, and it was suggested that the poor results of renal transplantation in patients of African origin in the US could be improved with universal immunosuppressive drug coverage (Pallet et al., 2005). Also in this French study CYP3A5 genotype data were not available.

In a meta-analysis Rojas found that the CYP3A5 expresser genotype might be associated with a higher risk of acute rejection (OR 1.32, 95% CI 1.02–1.71) and a trend towards more chronic nephrotoxicity (OR 1.81, 95% CI 0.89–3.68) (Rojas et al., 2015). However, a large-scale genome-wide association study did not identify strong donor or recipient genetic predictors of allograft survival or renal function outside the HLA region (Hernandez-Fuentes et al., 2018; Stapleton et al., 2019). Also a candidate gene association study of allograft loss in renal transplant recipients CYP3A5 was not related to outcome (Woillard et al., 2018).

In a recently published meta-analysis, the effect-size of CYP3A5 polymorphism on the risk of rejection was estimated in Asian and European kidney transplant populations (Khan et al., 2020). In European populations no significant association was found with rejection episodes between expressers and nonexpressers (OR: 1.12; p = 0.47). However, in Asian patients a higher risk of rejection was found after follow-up of 3 years post-transplantation (OR: 1.68; p < 0.05), respectively. An explanation for this difference, other than a higher prevalence of CYP3A5 expressers in Asian populations could not be given. Data on longer term follow-up are scarce, and this higher risk estimate would need confirmation in larger datasets.

C0/D Ratio as Prognostic Factor for Outcome

In daily practice tacrolimus pre-dose concentrations are being monitored, and routinely monitoring tacrolimus peak concentrations or AUC is unusual since it is more cumbersome both for patients and clinicians. Whether or not the patient needs a high tacrolimus dose to reach the target concentration is not taken into account. However, patients who need a high tacrolimus dose may be exposed to substantially higher tacrolimus peak concentrations and tacrolimus-AUC compared to patients who reach the same pre-dose concentrations with lower dosages. A study of 46 African Americans, of whom 35 were CYP3A5-expresser, showed that the tacrolimus pre-dose concentrations were similar in CYP3A5-expressers and non-expressers (6.26 and 6.24 ng/ml respectively) (Trofe-Clark et al., 2018). In the expressers group a much higher tacrolimus dose was required to reach these pre-dose concentrations (10.1 vs 6.3 mg/day), and as a result the Cmax was also higher in the expressers (25.5 ng/ml and 19.5 ng/ml respectively; p = 0.04). The higher peaks and higher AUC may lead to chronic nephrotoxicity and ultimately contribute to graft loss. In a study on chronic irreversible drug-induced nephrotoxicity Kuypers et al. also found that especially patients with a high early tacrolimus dose requirement developed this form of toxicity (Kuypers et al., 2010). In their study this was predominantly but not exclusively encountered in CYP3A5 expressers.

At present tacrolimus is being dosed based on trough concentrations in the vast majority of centers. There is limited experience with dosing tacrolimus based on AUC. In Leiden the tacrolimus target AUC0–12h for maintenance treatment (>6 months post-transplantation) is 80 ug*h/L. Based on a pharmacokinetic model the upper threshold of this target relates to a peak concentration of 22 ng/ml (Scholten et al., 2005). In case of high peak concentrations and high AUC the tacrolimus dose can be reduced. If peaks are high but AUC is within the target, then dose could be divided into smaller portions (from twice daily to three times daily), but this will negatively affect adherence. An alternative option would be to change from a twice daily immediate release formulation to a once daily prolonged release formulation. Besides avoiding high peaks this will also improve adherence. Another option would be to switch to mTOR-inhibitors as suggested by Thölking et al. (Schütte-Nütgen et al., 2019). There are no studies available that show that either of these options is beneficial.

At first sight the expression of CYP3A5 and a low C0/D ratio seem to be overlapping factors, both pointing towards patients in whom a higher tacrolimus dose is needed to reach the tacrolimus target concentration. However, there are important differences, and these differences may explain why the impact of the C0/D ration on long term outcome is stronger than for CYP3A5 genotype status.

First of all, on average the CYP3A5 genotype does lead to a higher dose requirement, but there is considerable overlap between expressers and non-expressers (Thervet et al., 2003). Some of the CYP3A5 expressers do not have a fast metabolizer phenotype, and they reach target concentrations with conventional tacrolimus doses. In these patients there is not a high peak concentration after drug intake. In contrast, patients with a low C0/D ratio all have a fast metabolizer phenotype and invariably need high doses of tacrolimus. As a result, the impact of the C0/D ratio may be stronger than the CYP3A5 genotype.

Another factor that may impact on the prognostic significance of the C0/D ratio is the use of corticosteroids in patients at increased risk of rejection. For patients at low-immunological risk the avoidance, or early withdrawal of steroids, is well tolerated, but in patients with higher risk this may lead to an increased incidence of acute cellular rejections or late antibody medicated rejections (Pascual, 2011). Therefore, continued exposure to steroids may be linked to a higher immunological risk. As steroids are known to induce CYP3A enzymes the tacrolimus metabolism is faster in patients treated with steroids, and C0/D ratio is lower (Hesselink et al., 2003; van Duijnhoven et al., 2003). Steroid use may thus be a confounder by indication, affecting the C0/D ratio. However, in the recently published TOMATO-study the C0/D ratio was independently associated with death-censored kidney-graft survival, even when corrected for continued corticosteroid use (Jouve et al., 2020).

An important limitation is that the number of studies on this topic is still limited and that several of these studies have been published by one center (Thölking et al.). In order to be more certain on the clinical relevance of the C0/D ratio it would be good if more well documented analyses in larger data sets, including correction for potential confounding factors such as ethnicity, steroid use, and intra-patient variability, would be performed. Furthermore, interventions such as dose reductions and switching to prolonged release formulations should ideally be tested in prospective controlled studies. Novel developments in therapeutic drug monitoring, including home-based dried blood spot (DBS) sampling, offer the potential to facilitate repetitive large scale AUC assessment as an alternative to the conventional TDM sampling at the (out-patient) clinic. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) assays have been developed to quantify tacrolimus in DBS samples (Veenhof et al., 2017; Zwart et al., 2018), and at home sampling has been shown to be feasible in the kidney transplant population. Adherence to immunosuppressive therapy has been shown to decrease over time (Massey et al., 2015). Furthermore, non-adherence is a strong predictor for poor outcome and is related to development of donor specific antibodies and a higher risk of rejection (Takemoto et al., 2007). Patients who do not take (part of) their daily drug dose seemingly have a fast metabolism and a low C0/D ratio. In these patients the poor outcome is not due to the faster metabolism, or due to high peak concentrations but to non-adherence.

Conclusion

The C0/D ratio of tacrolimus does seem to identify a group of patients with increased risk of poor outcome after kidney transplantation. Our recommendation is to check tacrolimus peak concentrations in these patients, and if these are high then target for slightly lower pre-dose concentrations. Assessment of limited sampling AUC can assist in finetuning the tacrolimus dose.

Author Contributions

TG and DM developed the idea for this manuscript. The first draft version was written by TG. All authors contributed to the article and approved the submitted version.

Conflict of Interest

In the last 3 years TG has received lecture fees and study grants from Chiesi and Astellas, in addition to consulting fees from Roche Diagnostics, Vitaeris, Astellas, Aurinia Pharma and Novartis. In all cases money has been transferred to hospital accounts, and none has been paid to his personal bank accounts. TG does not have employment or stock ownership at any of these companies, and neither does he have patents or patent applications.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Akamine, Y., Kagaya, H., Ohkubo, T., Satoh, S., Miura, M. (2018). A comparison of the effects of CYP3A5 polymorphism on tacrolimus blood concentrations measured by 4 immunoassay methods in renal transplant patients. J. Clin. Pharm. Ther. 43 (2), 181–188. doi: 10.1111/jcpt.12624

PubMed Abstract | CrossRef Full Text | Google Scholar

Andrews, L. M., De Winter, B. C., Van Gelder, T., Hesselink, D. A. (2016). Consideration of the ethnic prevalence of genotypes in the clinical use of tacrolimus. Pharmacogenomics 17 (16), 1737–1740. doi: 10.2217/pgs-2016-0136

PubMed Abstract | CrossRef Full Text | Google Scholar

Birdwell, K. A., Decker, B., Barbarino, J. M., Peterson, J. F., Stein, C. M., Sadee, W., et al. (2015). Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP3A5 Genotype and Tacrolimus Dosing. Clin. Pharmacol. Ther. 98 (1), 19–24. doi: 10.1002/cpt.113

PubMed Abstract | CrossRef Full Text | Google Scholar

Borra, L. C., Roodnat, J. I., Kal, J. A., Mathot, R. A., Weimar, W., van Gelder, T. (2010). High within-patient variability in the clearance of tacrolimus is a risk factor for poor long-term outcome after kidney transplantation. Nephrol. Dial Transpl. 25 (8), 2757–2763. doi: 10.1093/ndt/gfq096

CrossRef Full Text | Google Scholar

Christians, U., Vinks, A. A., Langman, L. J., Clarke, W., Wallemacq, P., van Gelder, T., et al. (2015). Impact of Laboratory Practices on Interlaboratory Variability in Therapeutic Drug Monitoring of Immunosuppressive Drugs. Ther. Drug Monit. 37 (6), 718–724. doi: 10.1097/FTD.0000000000000205

PubMed Abstract | CrossRef Full Text | Google Scholar

Elens, L., Bouamar, R., Hesselink, D. A., Haufroid, V., van der Heiden, I. P., van Gelder, T., et al. (2011). A new functional CYP3A4 intron 6 polymorphism significantly affects tacrolimus pharmacokinetics in kidney transplant recipients. Clin. Chem. 57 (11), 1574–1583. doi: 10.1373/clinchem.2011.165613

PubMed Abstract | CrossRef Full Text | Google Scholar

Haufroid, V., Mourad, M., Van Kerckhove, V., Wawrzyniak, J., De Meyer, M., Eddour, D. C., et al. (2004). The effect of CYP3A5 and MDR1 (ABCB1) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood levels in stable renal transplant patients. Pharmacogenetics 14 (3), 147–154. doi: 10.1097/00008571-200403000-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

Hernandez-Fuentes, M. P., Franklin, C., Rebollo-Mesa, I., Mollon, J., Delaney, F., Perucha, E., et al. (2018). United Kingdom and Ireland Renal Transplant Consortium (UKIRTC) and the Wellcome Trust Case Control Consortium (WTCCC)-3. Long- and short-term outcomes in renal allografts with deceased donors: A large recipient and donor genome-wide association study. Am. J. Transplant. 18 (6), 1370–1379. doi: 10.1111/ajt.14594

PubMed Abstract | CrossRef Full Text | Google Scholar

Hesselink, D. A., van Schaik, R. H., van der Heiden, I. P., van der Werf, M., Smak Gregoor, P. J., Lindemans, J., et al. (2003). Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Clin. Pharmacol. Ther. 74 (3), 245–254. doi: 10.1016/S0009-9236(03)00168-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Hesselink, D. A., Ngyuen, H., Wabbijn, M., Gregoor, P. J., Steyerberg, E. W., van Riemsdijk, I. C., et al. (2003). Tacrolimus dose requirement in renal transplant recipients is significantly higher when used in combination with corticosteroids. Br. J. Clin. Pharmacol. 56 (3), 327–330. doi: 10.1046/j.0306-5251.2003.01882.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hesselink, D. A., Bouamar, R., Elens, L., van Schaik, R. H., van Gelder, T. (2014). The role of pharmacogenetics in the disposition of and response to tacrolimus in solid organ transplantation. Clin. Pharmacokinet. 53 (2), 123–139. doi: 10.1007/s40262-013-0120-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Jouve, T., Fonrose, X., Noble, J., Janbon, B., Fiard, G., Malvezzi, P., et al. (2020). The TOMATO Study (Tacrolimus Metabolization in Kidney Transplantation): Impact of the Concentration-Dose Ratio on Death-censored Graft Survival. Transplantation 104 (6), 1263–1271. doi: 10.1097/TP.0000000000002920

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, A. R., Raza, A., Firasat, S., Abid, A. (2020). CYP3A5 gene polymorphisms and their impact on dosage and trough concentration of tacrolimus among kidney transplant patients: a systematic review and meta-analysis. Pharmacogenomics J. 20 (4), 553–562. doi: 10.1038/s41397-019-0144-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Koyama, H., Cecka, J. M., Terasaki, P. I. (1994). Kidney transplants in black recipients. HLA matching and other factors affecting long-term graft survival. Transplantation 57 (7), 1064–1068. doi: 10.1097/00007890-199404150-00014

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuypers, D. R., Naesens, M., de Jonge, H., Lerut, E., Verbeke, K., Vanrenterghem, Y. (2010). Tacrolimus dose requirements and CYP3A5 genotype and the development of calcineurin inhibitor-associated nephrotoxicity in renal allograft recipients. Ther. Drug Monit. 32 (4), 394–404. doi: 10.1097/FTD.0b013e3181e06818

PubMed Abstract | CrossRef Full Text | Google Scholar

Kwiatkowska, E., Kwiatkowski, S., Wahler, F., Gryczman, M., Domańki, L., Marchelk-Myśliwiec, M., et al. (2019). C/D Ratio in Long-Term Renal Function. Transplant. Proc. 51 (10), 3265–3270. doi: 10.1016/j.transproceed.2019.08.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Massey, E. K., Tielen, M., Laging, M., Timman, R., Beck, D. K., Khemai, R., et al. (2015). Discrepancies between beliefs and behavior: a prospective study into immunosuppressive medication adherence after kidney transplantation. Transplantation 99 (2), 375–380. doi: 10.1097/TP.0000000000000608

PubMed Abstract | CrossRef Full Text | Google Scholar

Mendoza Rojas, A., Hesselink, D. A., van Besouw, N. M., Baan, C. C., van Gelder, T. (2019). Impact of low tacrolimus exposure and high tacrolimus intra-patient variability on the development of de novo anti-HLA donor-specific antibodies in kidney transplant recipients. Expert Rev. Clin. Immunol. 15 (12), 1323–1331. doi: 10.1080/1744666X.2020.1693263

PubMed Abstract | CrossRef Full Text | Google Scholar

Neuberger, J. M., Bechstein, W. O., Kuypers, D. R., Burra, P., Citterio, F., De Geest, S., et al. (2017). Practical Recommendations for Long-term Management of Modifiable Risks in Kidney and Liver Transplant Recipients: A Guidance Report and Clinical Checklist by the Consensus on Managing Modifiable Risk in Transplantation (COMMIT) Group. Transplantation 101 (4S Suppl 2), S1–S56. doi: 10.1097/TP.0000000000001651

CrossRef Full Text | Google Scholar

Nowicka, M., Górska, M., Nowicka, Z., Edyko, K., Edyko, P., Wiślicki, S., et al. (2019). Tacrolimus: Influence of the Posttransplant Concentration/Dose Ratio on Kidney Graft Function in a Two-Year Follow-Up. Kidney Blood Press Res. 44 (5), 1075–1088. doi: 10.1159/000502290

PubMed Abstract | CrossRef Full Text | Google Scholar

Oetting, W. S., Wu, B., Schladt, D. P., Guan, W., Remmel, R. P., Mannon, R. B., et al. (2018). Genome-wide association study identifies the common variants in CYP3A4 and CYP3A5 responsible for variation in tacrolimus trough concentration in Caucasian kidney transplant recipients. Pharmacogenomics J. 18 (3), 501–505. doi: 10.1038/tpj.2017.49

PubMed Abstract | CrossRef Full Text | Google Scholar

Oetting, W. S., Wu, B., Schladt, D. P., Guan, W., van Setten, J., Keating, B. J. (2019). et al; DeKAF Genomics and GEN-03 Investigators. Genetic Variants Associated With Immunosuppressant Pharmacokinetics and Adverse Effects in the DeKAF Genomics Genome-wide Association Studies. Transplantation 103 (6), 1131–1139. doi: 10.1097/TP.0000000000002625

PubMed Abstract | CrossRef Full Text | Google Scholar

Pallet, N., Thervet, E., Alberti, C., Emal-Aglaé, V., Bedrossian, J., Martinez, F., et al. (2005). Kidney transplant in black recipients: are African Europeans different from African Americans? Am. J. Transplant. 5 (11), 2682–2687. doi: 10.1111/j.1600-6143.2005.01057.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascual, J. (2011). Steroid avoidance or withdrawal in kidney transplantation. Curr. Opin. Organ Transpl. 16 (6), 600–605. doi: 10.1097/MOT.0b013e32834c23fa

CrossRef Full Text | Google Scholar

Rodrigo, E., Segundo, D. S., Fernández-Fresnedo, G., López-Hoyos, M., Benito, A., Ruiz, J. C., et al. (2016). Within-Patient Variability in Tacrolimus Blood Levels Predicts Kidney Graft Loss and Donor-Specific Antibody Development. Transplantation 100 (11), 2479–2485. doi: 10.1097/TP.0000000000001040

PubMed Abstract | CrossRef Full Text | Google Scholar

Rojas, L., Neumann, I., Herrero, M. J., Bosó, V., Reig, J., Poveda, J. L., et al. (2015). Effect of CYP3A5*3 on kidney transplant recipients treated with tacrolimus: a systematic review and meta-analysis of observational studies. Pharmacogenomics J. 15 (1), 38–48. doi: 10.1038/tpj.2014.38

PubMed Abstract | CrossRef Full Text | Google Scholar

Schütte-Nütgen, K., Thölking, G., Steinke, J., Pavenstädt, H., Schmidt, R., Suwelack, B., et al. (2019). Fast Tac Metabolizers at Risk ⁻ It is Time for a C/D Ratio Calculation. J. Clin. Med. 8 (5), E587. doi: 10.3390/jcm8050587

PubMed Abstract | CrossRef Full Text | Google Scholar

Scholten, E. M., Cremers, S. C., Schoemaker, R. C., Rowshani, A. T., van Kan, E. J., den Hartigh, J., et al. (2005). AUC-guided dosing of tacrolimus prevents progressive systemic overexposure in renal transplant recipients. Kidney Int. 67 (6), 2440–2447. doi: 10.1111/j.1523-1755.2005.00352.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Shuker, N., van Gelder, T., Hesselink, D. A. (2015). Intra-patient variability in tacrolimus exposure: causes, consequences for clinical management. Transplant. Rev. (Orlando). 29 (2), 78–84. doi: 10.1016/j.trre.2015.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Stapleton, C. P., Heinzel, A., Guan, W., van der Most, P. J., van Setten, J., Lord, G. M., et al. (2019). n PJ; UK Ireland Renal Transplant Consortium; DeKAF Genomics and GEN03 Studies; International Genetics and Translational Research in Transplantation Network. The impact of donor and recipient common clinical and genetic variation on estimated glomerular filtration rate in a European renal transplant population. Am. J. Transpl. 19 (8), 2262–2273. doi: 10.1111/ajt.15326

CrossRef Full Text | Google Scholar

Taber, D. J., Gebregziabher, M. G., Srinivas, T. R., Chavin, K. D., Baliga, P. K., Egede, L. E. (2015). African-American race modifies the influence of tacrolimus concentrations on acute rejection and toxicity in kidney transplant recipients. Pharmacotherapy 35 (6), 569–577. doi: 10.1002/phar.1591

PubMed Abstract | CrossRef Full Text | Google Scholar

Takemoto, S. K., Pinsky, B. W., Schnitzler, M. A., Lentine, K. L., Willoughby, L. M., Burroughs, T. E., et al. (2007). A retrospective analysis of immunosuppression compliance, dose reduction and discontinuation in kidney transplant recipients. Am. J. Transpl. 7 (12), 2704–2711. doi: 10.1111/j.1600-6143.2007.01966.x

CrossRef Full Text | Google Scholar

Thölking, G., Fortmann, C., Koch, R., Gerth, H. U., Pabst, D., Pavenstädt, H., et al. (2014). The tacrolimus metabolism rate influences renal function after kidney transplantation. PloS One 9 (10), e111128. doi: 10.1371/journal.pone.0111128

PubMed Abstract | CrossRef Full Text | Google Scholar

Thölking, G., Schmidt, C., Koch, R., Schuette-Nuetgen, K., Pabst, D., Wolters, H., et al. (2016). Influence of tacrolimus metabolism rate on BKV infection after kidney transplantation. Sci. Rep. 6, 32273. doi: 10.1038/srep32273

PubMed Abstract | CrossRef Full Text | Google Scholar

Thölking, G., Schütte-Nütgen, K., Schmitz, J., Rovas, A., Dahmen, M., Bautz, J., et al. (2019). A Low Tacrolimus Concentration/Dose Ratio Increases the Risk for the Development of Acute Calcineurin Inhibitor-Induced Nephrotoxicity. J. Clin. Med. 8 (10), E1586. doi: 10.3390/jcm8101586

PubMed Abstract | CrossRef Full Text | Google Scholar

Thervet, E., Anglicheau, D., King, B., Schlageter, M. H., Cassinat, B., Beaune, P., et al. (2003). Impact of cytochrome p450 3A5 genetic polymorphism on tacrolimus doses and concentration-to-dose ratio in renal transplant recipients. Transplantation 76 (8), 1233–1235. doi: 10.1097/01.TP.0000090753.99170.89

PubMed Abstract | CrossRef Full Text | Google Scholar

Trofe-Clark, J., Brennan, D. C., West-Thielke, P., Milone, M. C., Lim, M. A., Neubauer, R., et al. (2018). Results of ASERTAA, a Randomized Prospective Crossover Pharmacogenetic Study of Immediate-Release Versus Extended-Release Tacrolimus in African American Kidney Transplant Recipients. Am. J. Kidney Dis. 71 (3), 315–326. doi: 10.1053/j.ajkd.2017.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

van Duijnhoven, E. M., Boots, J. M., Christiaans, M. H., Stolk, L. M., Undre, N. A., van Hooff, J. P. (2003). Increase in tacrolimus trough levels after steroid withdrawal. Transpl. Int. 16 (10), 721–725. doi: 10.1111/j.1432-2277.2003.tb00230.x

PubMed Abstract | CrossRef Full Text | Google Scholar

van Gelder, T. (2014). Within-patient variability in immunosuppressive drug exposure as a predictor for poor outcome after transplantation. Kidney Int. 85 (6), 1267–1268. doi: 10.1038/ki.2013.484

PubMed Abstract | CrossRef Full Text | Google Scholar

van Schaik, R. H., van der Heiden, I. P., van den Anker, J. N., Lindemans, J. (2002). CYP3A5 variant allele frequencies in Dutch Caucasians. Clin. Chem. 48 (10), 1668–1671. doi: 10.1093/clinchem/48.10.1668

PubMed Abstract | CrossRef Full Text | Google Scholar

Vanhove, T., Annaert, P., Kuypers, D. R. (2016). Clinical determinants of calcineurin inhibitor disposition: a mechanistic review. Drug Metab. Rev. 48, 88–112. doi: 10.3109/03602532.2016.1151037

PubMed Abstract | CrossRef Full Text | Google Scholar

Veenhof, H., Koster, R. A., Alffenaar, J. C., Berger, S. P., Bakker, S. J. L., Touw, D. J. (2017). Clinical Validation of Simultaneous Analysis of Tacrolimus, Cyclosporine A, and Creatinine in Dried Blood Spots in Kidney Transplant Patients. Transplantation 101 (7), 1727–1733. doi: 10.1097/TP.0000000000001591

PubMed Abstract | CrossRef Full Text | Google Scholar

Woillard, J. B., Gatault, P., Picard, N., Arnion, H., Anglicheau, D., Marquet, P., et al. (2018). and recipient candidate gene association study of allograft loss in renal transplant recipients receiving a tacrolimus-based regimen. Am. J. Transpl. Dec18 (12), 2905–2913. doi: 10.1111/ajt.14894

CrossRef Full Text | Google Scholar

Zheng, S., Tasnif, Y., Hebert, M. F., Davis, C. L., Shitara, Y., Calamia, J. C., et al. (2012). Measurement and compartmental modeling of the effect of CYP3A5 gene variation on systemic and intrarenal tacrolimus disposition. Clin. Pharmacol. Ther. 92 (6), 737–745. doi: 10.1038/clpt.2012.175

PubMed Abstract | CrossRef Full Text | Google Scholar

Zwart, T. C., Gokoel, S. R. M., van der Boog, P. J. M., de Fijter, J. W., Kweekel, D. M., Swen, J. J., et al. (2018). Therapeutic drug monitoring of tacrolimus and mycophenolic acid in outpatient renal transplant recipients using a volumetric dried blood spot sampling device. Br. J. Clin. Pharmacol. 84 (12), 2889–2902. doi: 10.1111/bcp.13755

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: tacrolimus, transplantation, kidney, CYP3A5, pharmacogenetics

Citation: van Gelder T, Meziyerh S, Swen JJ, de Vries APJ and Moes DJAR (2020) The Clinical Impact of the C0/D Ratio and the CYP3A5 Genotype on Outcome in Tacrolimus Treated Kidney Transplant Recipients. Front. Pharmacol. 11:1142. doi: 10.3389/fphar.2020.01142

Received: 10 February 2020; Accepted: 13 July 2020;
Published: 31 July 2020.

Edited by:

José A. G. Agúndez, University of Extremadura, Spain

Reviewed by:

Raman Venkataramanan, University of Pittsburgh, United States
Bryce Alan Kiberd, Dalhousie University, Canada

Copyright © 2020 van Gelder, Meziyerh, Swen, de Vries and Moes. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Teun van Gelder, T.van_Gelder1@lumc.nl

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.