An Integrated Clinical and Genetic Prediction Model for Tacrolimus Levels in Pediatric Solid Organ Transplant Recipients

Sandar Min, Tanya Papaz, A Nicole Lambert, Upton Allen, Patricia Birk, Tom Blydt-Hansen, Bethany J Foster, Hartmut Grasemann, Lorraine Hamiwka, Catherine Litalien, Vicky Ng, Noureddine Berka, Patricia Campbell, Claude Daniel, Chee Loong Saw, Kathryn Tinckam, Simon Urschel, Sara L Van Driest, Rulan Parekh, Seema Mital, Sandar Min, Tanya Papaz, A Nicole Lambert, Upton Allen, Patricia Birk, Tom Blydt-Hansen, Bethany J Foster, Hartmut Grasemann, Lorraine Hamiwka, Catherine Litalien, Vicky Ng, Noureddine Berka, Patricia Campbell, Claude Daniel, Chee Loong Saw, Kathryn Tinckam, Simon Urschel, Sara L Van Driest, Rulan Parekh, Seema Mital

Abstract

Background: There are challenges in achieving and maintaining therapeutic tacrolimus levels after solid organ transplantation (SOT). The purpose of this genome-wide association study was to generate an integrated clinical and genetic prediction model for tacrolimus levels in pediatric SOT.

Methods: In a multicenter prospective observational cohort study (2015-2018), children <18 years old at their first SOT receiving tacrolimus as maintenance immunosuppression were included (455 as discovery cohort; 322 as validation cohort). Genotyping was performed using a genome-wide single nucleotide polymorphism (SNP) array and analyzed for association with tacrolimus trough levels during 1-y follow-up.

Results: Genome-wide association study adjusted for clinical factors identified 25 SNPs associated with tacrolimus levels; 8 were significant at a genome-wide level (P < 1.025 × 10-7). Nineteen SNPs were replicated in the validation cohort. After removing SNPs in strong linkage disequilibrium, 14 SNPs remained independently associated with tacrolimus levels. Both traditional and machine learning approaches selected organ type, age at transplant, rs776746, rs12333983, and rs12957142 SNPs as the top predictor variables for dose-adjusted 36- to 48-h posttacrolimus initiation (T1) levels. There was a significant interaction between age and organ type with rs776476*1 SNP (P < 0.05). The combined clinical and genetic model had lower prediction error and explained 30% of the variation in dose-adjusted T1 levels compared with 18% by the clinical and 12% by the genetic only model.

Conclusions: Our study highlights the importance of incorporating age, organ type, and genotype in predicting tacrolimus levels and lays the groundwork for developing an individualized age and organ-specific genotype-guided tacrolimus dosing algorithm.

Conflict of interest statement

The authors declare no conflicts of interest.

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

Graphical abstract
Graphical abstract
FIGURE 1.
FIGURE 1.
A, Box plot of tacrolimus levels by time after transplant (n = 455 recipients; 2375 tacrolimus levels). The x-axis shows tacrolimus levels at 6 posttransplant time points (36–48 h post-Tac initiation (n = 398), 7 d (n = 410), 14 d (n = 409), 30 d (n = 403), 3 mo (n = 398), and 12 mo posttransplant (n = 357) in kidney (purple), heart (red), liver (blue), and lung (green) transplant recipients. The boxes represent medians and interquartile ranges, the whiskers represent minimum and maximum values, and the dots represent outliers. B, Forest plot of variables associated with tacrolimus trough levels during 1-y follow-up posttransplant using linear-mixed effects model. Tacrolimus levels were higher in liver, heart, and lung recipients compared with kidney recipients. Tacrolimus levels were higher with increasing age, concomitant CYP3A4 inhibitor use, and higher tacrolimus dosage. Dots represent parameter estimate for tacrolimus levels, and bars represent 95% confidence intervals. *P < 0.05; **P < 0.01; ***P < 0.001. †Posttacrolimus initiation. PC1, first principal component; PC2, second principal component; TAC, tacrolimus.
FIGURE 2.
FIGURE 2.
GWAS SNPs associated with tacrolimus trough levels. A, Manhattan plot showing SNPs associated with tacrolimus levels on GWAS analysis with majority of the significantly associated SNPs residing on chromosome 7. The red line represents genome-wide significance at P < 1.025 × 10−7, and the blue line represents P < 1 × 10−5. B, Q-Q plot of the observed vs expected P for SNP association with tacrolimus levels. C, Forest plot of SNPs independently associated with tacrolimus levels during 1-y follow-up posttransplant. Colored dots represent parameter estimate for tacrolimus levels, and bars represent 95% confidence intervals. GWAS, genome-wide association study; SNP, single nucleotide polymorphism.
FIGURE 3.
FIGURE 3.
Interaction of age and organ type with association of rs776746*1 SNP with T1 levels. A, Interaction plot of age and rs776746*1 SNP (adjusted for organ type). Blue dots represent log-transformed dose-adjusted T1 levels in CYP3A5 nonexpressors, red dots represent log-transformed dose-adjusted T1 levels in CYP3A5 expressors, and bars represent 95% CI in 3 age groups. The difference in log-transformed dose-adjusted T1 levels between CYP3A5 nonexpressors and expressors was higher in infants (n = 238; effect size 0.73; P < 0.001) and adolescents (n = 219; effect size 0.67; P < 0.001) and lower in children (n = 234; effect size 0.34; P < 0.01) (P < 0.05 between children and other age groups). B, Interaction plot of organ type and rs776746*1 SNP (adjusted for age). Blue dots represent log-transformed dose-adjusted T1 levels in CYP3A5 nonexpressors, red dots represent log-transformed dose-adjusted T1 levels in CYP3A5 expressors, and bars represent 95% CI in 3 different organ types. The difference in log-transformed dose-adjusted T1 levels between CYP3A5 nonexpressors and CYP3A5 expressors was higher in heart (effect size 0.92; P < 0.001) than in kidney (effect size 0.42; P < 0.001) or liver recipients (effect size 0.46; P < 0.001) (P < 0.01 between heart and other organ recipients). ***P < 0.001, and **P < 0.01. SNP, single nucleotide polymorphism.
FIGURE 4.
FIGURE 4.
Model performance for dose-adjusted T1 level prediction. A, Coefficient path of lasso model with a red vertical line indicating selected λ = 0.23, which has smallest out of sample MSE. The y-axis indicates standardized coefficient. Each colored line represents the independent variable and its coefficient. The variable with the largest standardized coefficient has highest impact on dose-adjusted T1 levels. As variables enter the model from left to right, the variable which enters the model first (ie, diverges from the 0 line) is the most important predictor and the variable which enters last is the least important variable. The x-axis represents the tuning parameter (λ) of lasso model. B, Standardized coefficient plot of the lasso model showing the relationship between the variables and dose-adjusted T1 levels with the variables above the zero line being positively related while those below the line being negatively associated with dose-adjusted T1 levels. C, Plot of observed vs dose-adjusted T1 levels predicted by clinical only model (blue), SNP only model (red), and combined clinical and SNP model (green) in the validation cohort. The dotted line represents a 45-degree perfect fitted line. (D) Plot of observed vs dose-adjusted T1 levels predicted by the combined clinical and SNP model in heart (orange), liver (gray), and kidney (magenta) recipients. The dotted line represents a 45-degree perfect fitted line. MSE, mean squared error; SNP, single nucleotide polymorphism.

References

    1. Kearns GL, Abdel-Rahman SM, Alander SW, et al. . Developmental pharmacology–drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349:1157–1167.
    1. Christians U, Jacobsen W, Benet LZ, et al. . Mechanisms of clinically relevant drug interactions associated with tacrolimus. Clin Pharmacokinet. 2002;41:813–851.
    1. Sikma MA, van Maarseveen EM, van de Graaf EA, et al. . Pharmacokinetics and toxicity of tacrolimus early after heart and Lung transplantation. Am J Transplant. 2015;15:2301–2313.
    1. Gueta I, Markovits N, Yarden-Bilavsky H, et al. . High tacrolimus trough level variability is associated with rejections after heart transplant. Am J Transplant. 2018;18:2571–2578.
    1. Davis S, Gralla J, Klem P, et al. . Tacrolimus intrapatient variability, time in therapeutic range, and risk of De Novo Donor-specific antibodies. Transplantation. 2020;104:881–887.
    1. Elens L, Capron A, van Schaik RH, et al. . Impact of CYP3A4*22 allele on tacrolimus pharmacokinetics in early period after renal transplantation: toward updated genotype-based dosage guidelines. Ther Drug Monit. 2013;35:608–616.
    1. Oetting WS, Schladt DP, Guan W, et al. ; DeKAF Investigators. Genomewide association study of tacrolimus concentrations in African American kidney transplant recipients identifies multiple CYP3A5 alleles. Am J Transplant. 2016;16:574–582.
    1. Birdwell KA, Decker B, Barbarino JM, et al. . Clinical pharmacogenetics implementation consortium (CPIC) guidelines for CYP3A5 genotype and tacrolimus dosing. Clin Pharmacol Ther. 2015;98:19–24.
    1. Gijsen V, Mital S, van Schaik RH, et al. . Age and CYP3A5 genotype affect tacrolimus dosing requirements after transplant in pediatric heart recipients. J Heart Lung Transplant. 2011;30:1352–1359.
    1. Min S, Papaz T, Lafreniere-Roula M, et al. . A randomized clinical trial of age and genotype-guided tacrolimus dosing after pediatric solid organ transplantation. Pediatr Transplant. 2018;22:e13285.
    1. Rojas L, Neumann I, Herrero MJ, et al. . Effect of CYP3A5*3 on kidney transplant recipients treated with tacrolimus: a systematic review and meta-analysis of observational studies. Pharmacogenomics J. 2015;15:38–48.
    1. Haufroid V, Wallemacq P, VanKerckhove V, et al. . CYP3A5 and ABCB1 polymorphisms and tacrolimus pharmacokinetics in renal transplant candidates: guidelines from an experimental study. Am J Transplant. 2006;6:2706–2713.
    1. Papaz T, Allen U, Blydt-Hansen T, et al. . Pediatric outcomes in transplant: PersOnaliSing immunosuppression to ImproVe efficacy (POSITIVE study): the collaboration and design of a National transplant precision medicine program. Transplant Direct. 2018;4:e410.
    1. Birdwell KA, Grady B, Choi L, et al. . The use of a DNA biobank linked to electronic medical records to characterize pharmacogenomic predictors of tacrolimus dose requirement in kidney transplant recipients. Pharmacogenet Genomics. 2012;22:32–42.
    1. Keating BJ, Van Setten J, Jacobson P, et al. . Design and implementation of the international genetics and translational research in transplantation network. Transplantation. 2015;99:2401–2412.
    1. Howie B, Marchini J, Stephens M. Genotype imputation with thousands of genomes. G3 (Bethesda). 2011;1:457–470.
    1. Bonferroni C. Teoria Statistica Delle Classi e Calcolo Delle Probabilità. Vol 8. Pubblicazioni del R Istituto Superiore di Scienze Economiche e Commerciali di Firenze; 1936.
    1. Kelly PJ, Stallard N, Whittaker JC. Statistical design and analysis of pharmacogenetic trials. Stat Med. 2005;24:1495–1508.
    1. DeSarbo WS, Hwang H, Jedidi K. Redundancy Analysis. Wiley StatsRef: Statistics Reference Online. 2016:1–18.
    1. Churpek MM, Yuen TC, Winslow C, et al. . Multicenter comparison of machine learning methods and conventional regression for predicting clinical deterioration on the wards. Crit Care Med. 2016;44:368–374.
    1. Montini G, Ujka F, Varagnolo C, et al. . The pharmacokinetics and immunosuppressive response of tacrolimus in paediatric renal transplant recipients. Pediatr Nephrol. 2006;21:719–724.
    1. Wallemacq P, Armstrong VW, Brunet M, et al. . Opportunities to optimize tacrolimus therapy in solid organ transplantation: report of the European consensus conference. Ther Drug Monit. 2009;31:139–152.
    1. Oetting WS, Wu B, Schladt DP, et al. . 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. 2018;18:501–505.
    1. Sohn M, Kim MG, Han N, et al. . Whole exome sequencing for the identification of CYP3A7 variants associated with tacrolimus concentrations in kidney transplant patients. Sci Rep. 2018;8:18064.
    1. Liu J, Ouyang Y, Chen D, et al. . Donor and recipient P450 gene polymorphisms influence individual pharmacological effects of tacrolimus in Chinese liver transplantation patients. Int Immunopharmacol. 2018;57:18–24.
    1. Liu MZ, He HY, Zhang YL, et al. . IL-3 and CTLA4 gene polymorphisms may influence the tacrolimus dose requirement in Chinese kidney transplant recipients. Acta Pharmacol Sin. 2017;38:415–423.
    1. Wang P, Mao Y, Razo J, et al. . Using genetic and clinical factors to predict tacrolimus dose in renal transplant recipients. Pharmacogenomics. 2010;11:1389–1402.
    1. Oetting WS, Wu B, Schladt DP, et al. . Attempted validation of 44 reported SNPs associated with tacrolimus troughs in a cohort of kidney allograft recipients. Pharmacogenomics. 2018;19:175–184.
    1. Lamba V, Panetta JC, Strom S, et al. . Genetic predictors of interindividual variability in hepatic CYP3A4 expression. J Pharmacol Exp Ther. 2010;332:1088–1099.
    1. Elens L, van Schaik RH, Panin N, et al. . 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.
    1. Vafadari R, Bouamar R, Hesselink DA, et al. . Genetic polymorphisms in ABCB1 influence the pharmacodynamics of tacrolimus. Ther Drug Monit. 2013;35:459–465.
    1. Staatz CE, Goodman LK, Tett SE. Effect of CYP3A and ABCB1 single nucleotide polymorphisms on the pharmacokinetics and pharmacodynamics of calcineurin inhibitors: part I. Clin Pharmacokinet. 2010;49:141–175.
    1. Merali ZG, Witiw CD, Badhiwala JH, et al. . Using a machine learning approach to predict outcome after surgery for degenerative cervical myelopathy. PLoS One. 2019;14:e0215133.
    1. Lacroix D, Sonnier M, Moncion A, et al. . Expression of CYP3A in the human liver–evidence that the shift between CYP3A7 and CYP3A4 occurs immediately after birth. Eur J Biochem. 1997;247:625–634.
    1. Stevens JC, Hines RN, Gu C, et al. . Developmental expression of the major human hepatic CYP3A enzymes. J Pharmacol Exp Ther. 2003;307:573–582.
    1. Murry DJ, Crom WR, Reddick WE, et al. . Liver volume as a determinant of drug clearance in children and adolescents. Drug Metab Dispos. 1995;23:1110–1116.
    1. Strougo A, Yassen A, Monnereau C, et al. . Predicting the “First dose in children” of CYP3A-metabolized drugs: evaluation of scaling approaches and insights into the CYP3A7-CYP3A4 switch at young ages. J Clin Pharmacol. 2014;54:1006–1015.
    1. Liddle C, Goodwin BJ, George J, et al. . Separate and interactive regulation of cytochrome P450 3A4 by triiodothyronine, dexamethasone, and growth hormone in cultured hepatocytes. J Clin Endocrinol Metab. 1998;83:2411–2416.
    1. Thervet E, Loriot MA, Barbier S, et al. . Optimization of initial tacrolimus dose using pharmacogenetic testing. Clin Pharmacol Ther. 2010;87:721–726.
    1. Shuker N, Bouamar R, van Schaik RH, et al. . 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.
    1. Passey C, Birnbaum AK, Brundage RC, et al. . Dosing equation for tacrolimus using genetic variants and clinical factors. Br J Clin Pharmacol. 2011;72:948–957.
    1. Boughton O, Borgulya G, Cecconi M, et al. . 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.
    1. Elens L, Hesselink DA, van Schaik RH, et al. . The CYP3A4*22 allele affects the predictive value of a pharmacogenetic algorithm predicting tacrolimus predose concentrations. Br J Clin Pharmacol. 2013;75:1545–1547.
    1. Levy AR, Sobolev B, James D, et al. . The costs of change: direct medical costs of solid organ transplantation in British Columbia, Canada, 1995-2003. Value Health. 2009;12:282–292.
    1. Brown NW, Gonde CE, Adams JE, et al. . Low hematocrit and serum albumin concentrations underlie the overestimation of tacrolimus concentrations by microparticle enzyme immunoassay versus liquid chromatography-tandem mass spectrometry. Clin Chem. 2005;51:586–592.
    1. Fukudo M, Yano I, Masuda S, et al. . Population pharmacokinetic and pharmacogenomic analysis of tacrolimus in pediatric living-donor liver transplant recipients. Clin Pharmacol Ther. 2006;80:331–345.

Source: PubMed

3
구독하다