1
|
Naguib H, Katz-Greenberg G, Harris M, Gommer J, Yang LZ, Erkanli A, Byrns J. Evaluation of Tacrolimus Concentrations and Clinical Outcomes Between Extended and Immediate Release Formulations in Kidney Transplant. J Pharm Pract 2024; 37:1283-1290. [PMID: 38683344 DOI: 10.1177/08971900241248862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/01/2024]
Abstract
Objectives: Tacrolimus remains the mainstay of immunosuppression in kidney transplantation. Understanding the relationship between therapeutic tacrolimus levels and outcomes of acute rejection, patient/graft survival, and tolerability are important. The relationship between time to therapeutic tacrolimus levels and outcomes has not been well established, specifically with the use of extended release tacrolimus formulation (LCP-Tac). This study investigated time to therapeutic tacrolimus levels of 2 tacrolimus formulations, LCP-Tac and immediate release tacrolimus (IR-Tac), as a predictor of clinical outcomes. Methods: This was a single-center, retrospective, cohort study of kidney transplant recipients at Duke Hospital between 2013-2021. The primary objective evaluated the difference in time to therapeutic tacrolimus levels with LCP-Tac vs IR-Tac regimens. Secondary endpoints included time within therapeutic range during the first 3 months post-transplant, incidence of biopsy-proven rejection, development of de novo donor specific antibodies, and patient and allograft survival at 12 months post-transplant. Results: 128 patients were included (63 in LCP-Tac group and 65 in IR-Tac group). The time to therapeutic tacrolimus level was similar between formulations (7.2 days with LCP-Tac compared to 6.7 days with IR-Tac, P = .63). The time within therapeutic range during the first 3 months post-transplant, via modified Rosendaal, was similar with LCP-Tac and IR-Tac (56.1% vs 64.8%, respectively). Rates of biopsy-proven acute rejection at 12 months were similar (7/63 (11.1%) compared to 4/65 (6.2%)). There was no difference in patient/graft survival between groups. Conclusions: The time to therapeutic tacrolimus levels did not differ based on tacrolimus formulation and was not correlated with clinical outcomes.
Collapse
Affiliation(s)
- Hannah Naguib
- Department of Pharmacy, Indiana University Health Adult Academic Health Center, Indianapolis, IN, USA
| | | | - Matt Harris
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| | - Jennifer Gommer
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| | - Lexie Z Yang
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Alaattin Erkanli
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Jennifer Byrns
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| |
Collapse
|
2
|
Saqr A, Al-Kofahi M, Mohamed M, Dorr C, Remmel RP, Onyeaghala G, Oetting WS, Guan W, Mannon RB, Matas AJ, Israni A, Jacobson PA. Steroid-tacrolimus drug-drug interaction and the effect of CYP3A genotypes. Br J Clin Pharmacol 2024; 90:2837-2848. [PMID: 38994750 DOI: 10.1111/bcp.16172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/11/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
AIMS Tacrolimus, metabolized by CYP3A4 and CYP3A5 enzymes, is susceptible to drug-drug interactions (DDI). Steroids induce CYP3A genes to increase tacrolimus clearance, but the effect is variable. We hypothesized that the extent of the steroid-tacrolimus DDI differs by CYP3A4/5 genotypes. METHODS Kidney transplant recipients (n = 2462) were classified by the number of loss of function alleles (LOF) (CYP3A5*3, *6 and *7 and CYP3A4*22) and steroid use at each tacrolimus trough in the first 6 months post-transplant. A population pharmacokinetic analysis was performed by nonlinear mixed-effect modelling (NONMEM) and stepwise covariate modelling to define significant covariates affecting tacrolimus clearance. A stochastic simulation was performed and translated into a Shiny application with the mrgsolve and Shiny packages in R. RESULTS Steroids were associated with modestly higher (3%-11.8%) tacrolimus clearance. Patients with 0-LOF alleles receiving steroids showed the greatest increase (11.8%) in clearance compared to no steroids, whereas those with 2-LOFs had a negligible increase (2.6%) in the presence of steroids. Steroid use increased tacrolimus clearance by 5% and 10.3% in patients with 1-LOF and 3/4-LOFs, respectively. CONCLUSIONS Steroids increase the clearance of tacrolimus but vary slightly by CYP3A genotype. This is important in individuals of African ancestry who are more likely to carry no LOF alleles, may more commonly receive steroid treatment, and will need higher tacrolimus doses.
Collapse
Affiliation(s)
- Abdelrahman Saqr
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Mahmoud Al-Kofahi
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
- Gilead Sciences, Inc., Foster City, California, USA
| | - Moataz Mohamed
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Casey Dorr
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rory P Remmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guillaume Onyeaghala
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - William S Oetting
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Roslyn B Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska, Omaha, Nebraska, USA
| | - Arthur J Matas
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ajay Israni
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Epidemiology & Community Health, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Pamala A Jacobson
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Marquet P, Anglicheau D, Humeau A, Adrouche S, Saada L, Bisiaux J, Guillemin S, Lardy-Cléaud A, Rostaing L. Tacrolimus Dose Requirement in De Novo Adult Kidney Transplant Patients Treated With Adoport ® Can Be Anticipated. Transpl Int 2024; 37:13495. [PMID: 39469664 PMCID: PMC11513580 DOI: 10.3389/ti.2024.13495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/04/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024]
Abstract
All the factors potentially influencing tacrolimus dose requirement and combinations thereof have never been thoroughly investigated, precluding accurate prediction of tacrolimus starting dose. This prospective, non-interventional, multicenter study in de novo adult kidney transplant recipients over the first year after transplantation aimed to investigate the factors influencing tacrolimus dose-standardized trough blood concentration (C0/D) over the first week post-transplant (D4-D7, primary objective), D8-M3 and M3-M12 (secondary objectives). Statistical analysis employed mixed linear models with repeated measures. Eighteen sites enrolled 440 patients and followed them up for 9.5 ± 4.1 months. Age at baseline (p = 0.0144), end-stage renal disease (p = 0.0092), CYP3A phenotype (p < 0.0001), dyslipidemia at baseline (p = 0.0031), hematocrit (p = 0.0026), total bilirubin (p = 0.0261) and plasma creatinine (p = 0.0484) independently increased with log(C0/D) over D4-D7, explaining together 72.3% of the interindividual variability, and representing a robust model to estimate tacrolimus initial dose. Donor age and CYP3A phenotype were also influential over D8-M3 and M3-12, in addition to recipient age. Corticosteroids, diabetes at baseline, and ASAT yielded inconstant results between D8-M3 and M3-M12. We found no ethnicity effect when CYP3A phenotype was accounted for, and no food effect. Intra-individual variability over M3-M12 was moderate, and significantly lower in patients with chronic hepatic disorder (p = 0.0196) or cancer (p = 0.0132).
Collapse
Affiliation(s)
- Pierre Marquet
- Department of Pharmacology, Toxicology and Pharmacovigilance, Centre Hospitalier Universitaire de Limoges, Limoges, France
- Pharmacology and Transplantation, UMR1248 Inserm Université de Limoges, Limoges, France
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Université Paris Cité, Paris, France
| | - Antoine Humeau
- Pharmacology and Transplantation, UMR1248 Inserm Université de Limoges, Limoges, France
| | | | - Lakhdar Saada
- Medical Department, SANDOZ S.A.S, Levallois-Perret, France
| | | | | | | | - Lionel Rostaing
- Department of Nephrology, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| |
Collapse
|
4
|
Yang CW, Velez JCQ, Cohen DL. Immediate-Release versus Extended-Release Tacrolimus: Comparing Blood Pressure Control in Kidney Transplant Recipients - A Retrospective Cohort Study. Nephron Clin Pract 2024; 149:57-65. [PMID: 39307127 PMCID: PMC11850199 DOI: 10.1159/000541334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/07/2024] [Accepted: 08/07/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Hypertension (HTN) is a common side effect of tacrolimus (Tac), the first-line antirejection medication for kidney transplant recipients. The impact of immediate-release tacrolimus (Tac IR) dosed twice daily versus extended-release tacrolimus (Tac ER) dosed once daily on long-term blood pressure control in kidney transplant recipients remains understudied. This study aims to compare the use of Tac IR versus Tac ER in kidney transplant recipients and evaluate the effects of the different formulations on systolic blood pressure (SBP), diastolic blood pressure (DBP), and HTN crisis. METHODS This retrospective cohort study at a single institution collected baseline characteristics, time-varying exposure to Tac IR versus Tac ER, SBP, DBP, HTN crisis, and confounders at each posttransplant visit. A marginal structural linear mixed-effects model was employed to analyze the longitudinal blood pressure control in kidney transplant recipients receiving Tac IR and Tac ER. RESULTS The final analysis included 654 patients, with mean ages of 52.0 years for Tac IR and 50.3 years for Tac ER. Males constituted 56.7% in Tac IR and 55.0% in Tac ER. Notably, the black population had 2.44 times higher odds of receiving Tac ER after adjusting for the rest of the baseline characteristics. No difference was found between longitudinal SBP (p = 0.386, 95% CI: -1.00, 2.57) or DBP (p = 0.797, 95% CI: -1.38, 1.06). CONCLUSION Our study indicates that posttransplant patients taking Tac ER exhibit no difference in chronic SBP and DBP controls compared to Tac IR.
Collapse
Affiliation(s)
- Chien-Wen Yang
- Department of Nephrology, Ochsner Medical Center, New Orleans, LA, United States
- Ochsner Clinical School, The University of Queensland, Brisban, Australia
| | - Juan Carlos Q Velez
- Department of Nephrology, Ochsner Medical Center, New Orleans, LA, United States
- Ochsner Clinical School, The University of Queensland, Brisban, Australia
| | - Debbie L. Cohen
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
5
|
Chamzas A, Tellez E, SyBing A, Gobburu JVS, Gopalakrishnan M. Optimizing tacrolimus dosing in Hispanic renal transplant patients: insights from real-world data. Front Pharmacol 2024; 15:1443988. [PMID: 39364052 PMCID: PMC11446860 DOI: 10.3389/fphar.2024.1443988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Aim Tacrolimus, an immunosuppressant used to prevent organ rejection in renal transplant patients, exhibits high inter-patient variability, necessitating therapeutic drug monitoring. Early post-transplant tacrolimus exposure in Hispanics is understudied. Although genotypic information is linked to pharmacokinetic differences, its clinical application remains limited. This study aimed to use a real-world data-driven, pharmacokinetic model-based approach for tacrolimus in Hispanics to determine a suitable initial dose and design an optimal dose titration strategy by simulations to achieve plasma trough concentration target levels of 10-12 ng/mL at the earliest. Methods Sparse concentration-time data of tacrolimus were obtained from electronic medical records for self-identified Hispanic subjects following renal transplant. Rich pharmacokinetic literature data was leveraged to estimate structural pharmacokinetic model parameters, which were then fixed in the current analysis. Only apparent clearance was estimated with the sparse tacrolimus data and potential covariates were identified. Simulations of various starting doses and different dose titration strategies were then evaluated. Results The analysis included 121 renal transplant patients with 2,215 trough tacrolimus concentrations. A two-compartment transit absorption model with allometrically scaled body weight and time-varying hematocrit on apparent clearance adequately described the data. The estimated apparent clearance was 13.7 L/h for a typical patient weighing 70 kg and at 30% hematocrit, demonstrating a 40% decrease in clearance compared to other patient populations. Model based simulations indicated the best initial dose for the Hispanic population is 0.1 mg/kg/day. The proposed titration strategy, with three dose adjustments based on trough levels of tacrolimus, increased the proportion of patients within the target range (10-12 ng/mL) more than 2.5-fold and decreased the proportion of patients outside the therapeutic window by 50% after the first week of treatment. Conclusion Hispanic renal transplant population showed an estimated 40% decrease of apparent clearance in the typical patient compared to other populations with similar characteristics. The proposed dose adjustment attained the target range rapidly and safely. This study advocates for tailored tacrolimus dosing regimens based on population pharmacokinetics to optimize therapy in Hispanic renal transplant recipients.
Collapse
Affiliation(s)
- Athanasios Chamzas
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | | | - Andrew SyBing
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Jogarao V. S. Gobburu
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Mathangi Gopalakrishnan
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, MD, United States
| |
Collapse
|
6
|
Mohamed ME, Guo B, Wu B, Schladt DP, Muthusamy A, Guan W, Abrahante JE, Onyeaghala G, Saqr A, Pankratz N, Agarwal G, Mannon RB, Matas AJ, Oetting WS, Remmel RP, Israni AK, Jacobson PA, Dorr CR. Extreme phenotype sampling and next generation sequencing to identify genetic variants associated with tacrolimus in African American kidney transplant recipients. THE PHARMACOGENOMICS JOURNAL 2024; 24:29. [PMID: 39179559 DOI: 10.1038/s41397-024-00349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/08/2024] [Revised: 07/19/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
African American (AA) kidney transplant recipients (KTRs) have poor outcomes, which may in-part be due to tacrolimus (TAC) sub-optimal immunosuppression. We previously determined the common genetic regulators of TAC pharmacokinetics in AAs which were CYP3A5 *3, *6, and *7. To identify low-frequency variants that impact TAC pharmacokinetics, we used extreme phenotype sampling and compared individuals with extreme high (n = 58) and low (n = 60) TAC troughs (N = 515 AA KTRs). Targeted next generation sequencing was conducted in these two groups. Median TAC troughs in the high group were 7.7 ng/ml compared with 6.3 ng/ml in the low group, despite lower daily doses of 5 versus 12 mg, respectively. Of 34,542 identified variants across 99 genes, 1406 variants were suggestively associated with TAC troughs in univariate models (p-value < 0.05), however none were significant after multiple testing correction. We suggest future studies investigate additional sources of TAC pharmacokinetic variability such as drug-drug-gene interactions and pharmacomicrobiome.
Collapse
Affiliation(s)
- Moataz E Mohamed
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Bin Guo
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Baolin Wu
- Department of Epidemiology and Biostatistics, University of California Irvine, Irvine, CA, USA
| | - David P Schladt
- Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| | | | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Juan E Abrahante
- Research Informatics, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Guillaume Onyeaghala
- Hennepin Healthcare Research Institute, Minneapolis, MN, USA
- Nephrology Division, Hennepin Healthcare, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Abdelrahman Saqr
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Gaurav Agarwal
- Division of Nephrology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Roslyn B Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Arthur J Matas
- Division of Transplantation, Department of Surgery, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - William S Oetting
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Rory P Remmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Ajay K Israni
- Hennepin Healthcare Research Institute, Minneapolis, MN, USA
- Nephrology Division, Hennepin Healthcare, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamala A Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Casey R Dorr
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA.
- Hennepin Healthcare Research Institute, Minneapolis, MN, USA.
- Nephrology Division, Hennepin Healthcare, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Clinical and Translational Sciences Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
7
|
Obayemi JE, Callans L, Nair N, Gao H, Gandla D, Loza BL, Gao S, Mohebnasab M, Trofe-Clark J, Jacobson P, Keating B. Assessing the Utility of a Genotype-Guided Tacrolimus Equation in African American Kidney Transplant Recipients: A Single Institution Retrospective Study. J Clin Pharmacol 2024; 64:944-952. [PMID: 38766706 DOI: 10.1002/jcph.2461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 05/22/2024]
Abstract
Tacrolimus metabolism is heavily influenced by the CYP3A5 genotype, which varies widely among African Americans (AA). We aimed to assess the performance of a published genotype-informed tacrolimus dosing model in an independent set of adult AA kidney transplant (KTx) recipients. CYP3A5 genotypes were obtained for all AA KTx recipients (n = 232) from 2010 to 2019 who met inclusion criteria at a single transplant center in Philadelphia, Pennsylvania, USA. Medical record data were used to calculate predicted tacrolimus clearance using the published AA KTx dosing equation and two modified iterations. Observed and model-predicted trough levels were compared at 3 days, 3 months, and 6 months post-transplant. The mean prediction error at day 3 post-transplant was 3.05 ng/mL, indicating that the model tended to overpredict the tacrolimus trough. This bias improved over time to 1.36 and 0.78 ng/mL at 3 and 6 months post-transplant, respectively. Mean absolute prediction error-a marker of model precision-improved with time to 2.33 ng/mL at 6 months. Limiting genotype data in the model decreased bias and improved precision. The bias and precision of the published model improved over time and were comparable to studies in previous cohorts. The overprediction observed by the published model may represent overfitting to the initial cohort, possibly limiting generalizability.
Collapse
Affiliation(s)
- Joy E Obayemi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lauren Callans
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Nikhil Nair
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Hui Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Divya Gandla
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Bao-Li Loza
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Maedeh Mohebnasab
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Trofe-Clark
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pamala Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Brendan Keating
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, New York University, New York, NY, USA
| |
Collapse
|
8
|
Mohamed M, Guo B, Wu B, Schladt D, Muthusamy A, Guan W, Abrahante J, Onyeaghala G, Saqr A, Pankratz N, Agarwal G, Mannon R, Matas A, Oetting W, Remmel R, Israni A, Jacobson P, Dorr C. Extreme Phenotype Sampling and Next Generation Sequencing to Identify Genetic Variants Associated with Tacrolimus in African American Kidney Transplant Recipients. RESEARCH SQUARE 2024:rs.3.rs-4050136. [PMID: 38558983 PMCID: PMC10980152 DOI: 10.21203/rs.3.rs-4050136/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 04/04/2024]
Abstract
African American (AA) kidney transplant recipients (KTRs) have poor outcomes, which may in-part be due to tacrolimus (TAC) sub-optimal immunosuppression. We previously determined the common genetic regulators of TAC pharmacokinetics in AAs which were CYP3A5 *3, *6, and *7. To identify low-frequency variants that impact TAC pharmacokinetics, we used extreme phenotype sampling and compared individuals with extreme high (n=58) and low (n=60) TAC troughs (N=515 AA KTRs). Targeted next generation sequencing was conducted in these two groups. Median TAC troughs in the high group were 7.7 ng/ml compared with 6.3 ng/ml in the low group, despite lower daily doses of 5 versus 12mg, respectively. Of 34,542 identified variants across 99 genes, 1,406 variants were suggestively associated with TAC troughs in univariate models (p-value <0.05), however none were significant after multiple testing correction. We suggest future studies investigate additional sources of TAC pharmacokinetic variability such as drug-drug-gene interactions and pharmacomicrobiome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Pamala Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota
| | | |
Collapse
|
9
|
Masimirembwa C, Ramsay M, Botha J, Ellis E, Etheredge H, Hurrell T, Kanji CR, Kapungu NN, Maher H, Mthembu B, Naidoo J, Scholefield J, Rambarran S, van der Schyff F, Smyth N, Strobele B, Thelingwani RS, Loveland J, Fabian J. The African Liver Tissue Biorepository Consortium: Capacitating Population-Appropriate Drug Metabolism, Pharmacokinetics, and Pharmacogenetics Research in Drug Discovery and Development. Drug Metab Dispos 2023; 51:1551-1560. [PMID: 37751997 DOI: 10.1124/dmd.123.001400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/24/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Pharmaceutical companies subject all new molecular entities to a series of in vitro metabolic characterizations that guide the selection and/or design of compounds predicted to have favorable pharmacokinetic properties in humans. Current drug metabolism research is based on liver tissue predominantly obtained from people of European origin, with limited access to tissue from people of African origin. Given the interindividual and interpopulation genomic variability in genes encoding drug-metabolizing enzymes, efficacy and safety of some drugs are poorly predicted for African populations. To address this gap, we have established the first comprehensive liver tissue biorepository inclusive of people of African origin. The African Liver Tissue Biorepository Consortium currently includes three institutions in South Africa and one in Zimbabwe, with plans to expand to other African countries. The program has collected 67 liver samples as of July 2023. DNA from the donors was genotyped for 120 variants in 46 pharmacogenes and revealed variants that are uniquely found in African populations, including the low-activity, African-specific CYP2C9*5 and *8 variants relevant to the metabolism of diclofenac. Larger liver tissue samples were used to isolate primary human hepatocytes. Viability of the hepatocytes and microsomal fractions was demonstrated by the activity of selected cytochrome P450s. This resource will be used to ensure the safety and efficacy of existing and new drugs in African populations. This will be done by characterizing compounds for properties such as drug clearance, metabolite and enzyme identification, and drug-drug and drug-gene interactions. SIGNIFICANCE STATEMENT: Standard optimization of the drug metabolism of new molecular entities in the pharmaceutical industry uses subcellular fractions such as microsomes and isolated primary hepatocytes, being done mainly with tissue from donors of European origin. Pharmacogenetics research has shown that variants in genes coding for drug-metabolizing enzymes have interindividual and interpopulation differences. We established an African liver tissue biorepository that will be useful in ensuring drug discovery and development research takes into account drug responses in people of African origin.
Collapse
Affiliation(s)
- Collen Masimirembwa
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Michele Ramsay
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Jean Botha
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Ewa Ellis
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Harriet Etheredge
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Tracey Hurrell
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Comfort Ropafadzo Kanji
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Nyasha Nicole Kapungu
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Heather Maher
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Busisiwe Mthembu
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Jerolen Naidoo
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Janine Scholefield
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Sharan Rambarran
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Francisca van der Schyff
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Natalie Smyth
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Bernd Strobele
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Roslyn Stella Thelingwani
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - Jerome Loveland
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| | - June Fabian
- African institute of biomedical Science and Technology (AiBST), Harare, Zimbabwe (C.M., C.R.K., N.N.K., R.S.T.); Sydney Brenner Institute of Molecular Bioscience (SBIMB), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (C.M., M.R., B.M., N.S.); Wits Donald Gordon Medical Centre (WDGMC), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (H.E., H.M., S.R., B.S., F.V.S., J.L., J.F.); Karolinska Institute, Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Karolinska University Hospital Huddinge, Sweden (E.E.); Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria, South Africa (T.H., J.N., J.S.); and Transplant Services, Intermountain Medical Center, Salt Lake City, Utah (J.B.)
| |
Collapse
|
10
|
Newman J, Patel N, Patel S, Sprague T, Bartlett F, Rao N, Andrade E, Rohan V, DuBay D, Casey MJ, Taber D. Impact of obesity on the conversion of immediate-release tacrolimus to extended-release tacrolimus in kidney transplant recipients. Clin Transplant 2023; 37:e15149. [PMID: 37788162 DOI: 10.1111/ctr.15149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023]
Abstract
Outcomes analyzing conversion from IR-tacrolimus (IR) to LCP-tacrolimus (LCP) in obesity are limited. This was a retrospective longitudinal cohort study of patients converted from IR to LCP from June 2019 to October 2020. Primary outcomes were conversion ratios for weight-based dose at a steady-state therapeutic level and identification of appropriate dosing weight. Other outcomes included tacrolimus coefficient of variation (CV), time in therapeutic range (TITR), adverse events, infections, donor specific antibodies (DSAs), and acute rejection. A total of 292 patients were included; 156 and 136 patients with a BMI < 30 and BMI ≥ 30 kg/m2 , respectively. Baseline characteristics were similar, except for pancreas transplant, diabetes, and HLA mismatch. IR to LCP conversion ratio ranged from .73 to .79. Mean LCP dose was similar (.08 vs. .07 mg/kg/day for BMI < 30 and BMI ≥ 30 kg/m2 , respectively); there was a significant difference in IR and LCP mg/kg dosing at steady state with TBW (.11 mg/kg vs.09 mg/kg and .08 mg/kg vs. .06 mg/kg, respectively). The most appropriate dosing weight was adjusted body weight (AdjBW), consistent across IR and LCP steady-state doses, and might yield more accurate steady-state dosing requirements. In multivariable modeling, BMI was a significant predictor of steady state mg/kg dosing at therapeutic goal for total body weight (TBW), but not ideal body weight (IBW) or AdjBW.
Collapse
Affiliation(s)
- Jessica Newman
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Neha Patel
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shikha Patel
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Taylor Sprague
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Felicia Bartlett
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Nikhil Rao
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Erika Andrade
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Vinayak Rohan
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Derek DuBay
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Michael J Casey
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David Taber
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Pharmacy, Ralph H Johnson VAMC, Charleston, South Carolina, USA
| |
Collapse
|
11
|
Schagen MR, Volarevic H, Francke MI, Sassen SDT, Reinders MEJ, Hesselink DA, de Winter BCM. Individualized dosing algorithms for tacrolimus in kidney transplant recipients: current status and unmet needs. Expert Opin Drug Metab Toxicol 2023; 19:429-445. [PMID: 37642358 DOI: 10.1080/17425255.2023.2250251] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/24/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Tacrolimus is a potent immunosuppressive drug with many side effects including nephrotoxicity and post-transplant diabetes mellitus. To limit its toxicity, therapeutic drug monitoring (TDM) is performed. However, tacrolimus' pharmacokinetics are highly variable within and between individuals, which complicates their clinical management. Despite TDM, many kidney transplant recipients will experience under- or overexposure to tacrolimus. Therefore, dosing algorithms have been developed to limit the time a patient is exposed to off-target concentrations. AREAS COVERED Tacrolimus starting dose algorithms and models for follow-up doses developed and/or tested since 2015, encompassing both adult and pediatric populations. Literature was searched in different databases, i.e. Embase, PubMed, Web of Science, Cochrane Register, and Google Scholar, from inception to February 2023. EXPERT OPINION Many algorithms have been developed, but few have been prospectively evaluated. These performed better than bodyweight-based starting doses, regarding the time a patient is exposed to off-target tacrolimus concentrations. No benefit in reduced tacrolimus toxicity has yet been observed. Most algorithms were developed from small datasets, contained only a few tacrolimus concentrations per person, and were not externally validated. Moreover, other matrices should be considered which might better correlate with tacrolimus toxicity than the whole-blood concentration, e.g. unbound plasma or intra-lymphocytic tacrolimus concentrations.
Collapse
Affiliation(s)
- Maaike R Schagen
- Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Erasmus MC, Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
| | - Helena Volarevic
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marith I Francke
- Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Sebastiaan D T Sassen
- Erasmus MC, Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marlies E J Reinders
- Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda C M de Winter
- Erasmus MC, Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Impact of CYP3A5 genotype on de-novo LCP tacrolimus dosing and monitoring in kidney transplantation. Pharmacogenet Genomics 2023; 33:59-65. [PMID: 36877088 DOI: 10.1097/fpc.0000000000000494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/07/2023]
Abstract
OBJECTIVES LCP tac has a recommended starting dose of 0.14 mg/kg/day in kidney transplant. The goal of this study was to assess the influence of CYP3A5 on perioperative LCP tac dosing and monitoring. METHODS This was a prospective observational cohort study of adult kidney recipients receiving de-novo LCP tac. CYP3A5 genotype was measured and 90-day pharmacokinetic and clinical were assessed. Patients were classified as CYP3A5 expressors (*1 homozygous or heterozygous) or nonexpressors (LOF *3/*6/*7 allele). RESULTS In this study, 120 were screened, 90 were contacted and 52 provided consent; 50 had genotype results, and 22 patients expressed CYP3A5*1. African Americans (AA) comprised 37.5% of nonexpressors versus 81.8% of expressors (P = 0.001). Initial LCP tac dose was similar between CYP3A5 groups (0.145 vs. 0.137 mg/kg/day; P = 0.161), whereas steady state dose was higher in expressors (0.150 vs. 0.117 mg/kg/day; P = 0.026). CYP3A5*1 expressors had significantly more tac trough concentrations of less than 6 ng/ml and significantly fewer tac trough concentrations of more than 14 ng/ml. Providers were significantly more likely to under-adjust LCP tac by 10 and 20% in CYP3A5 expressors versus nonexpressors (P < 0.03). In sequential modeling, CYP3A5 genotype status explained the LCP tac dosing requirements significantly more than AA race. CONCLUSION CYP3A5*1 expressors require higher doses of LCP tac to achieve therapeutic concentrations and are at higher risk of subtherapeutic trough concentrations, persisting for 30-day posttransplant. LCP tac dose changes in CYP3A5 expressors are more likely to be under-adjusted by providers.
Collapse
|
13
|
Haverals L, Roosens L, Wouters K, Marquet P, Monchaud C, Massart A, Abramowicz D, Hellemans R. Does the Tacrolimus Trough Level Adequately Predict Drug Exposure in Patients Requiring a High Tacrolimus Dose? Transplant Direct 2023; 9:e1439. [PMID: 37009168 PMCID: PMC10065838 DOI: 10.1097/txd.0000000000001439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/20/2022] [Revised: 11/08/2022] [Accepted: 12/01/2022] [Indexed: 04/04/2023] Open
Abstract
Tacrolimus (Tac) has a narrow therapeutic range. Dosing is generally targeted at Tac trough levels (C 0), notwithstanding conflicting reports on the correlation between Tac C 0 and systemic exposure measured by the area-under-the-concentration-over-time curve (AUC). The Tac dose required to meet the target C 0 varies highly among patients. We hypothesized that patients requiring a relatively high Tac dose for a certain C 0 may show a higher AUC. Methods We retrospectively analyzed data from 53 patients in which a 24-h Tac AUC24 estimation was performed at our center. Patients were divided into those taking a low (≤0.15 mg/kg) or high (>0.15 mg/kg) once-daily Tac dose. Multiple linear regression models were used to investigate if the association between C 0 and AUC24 changes according to dose level. Results Despite the large difference in mean Tac dose between the low- and high-dose group (7 versus 17 mg/d), C 0 levels were similar. However, the mean AUC24 was substantially higher in the high-dose group (320 ± 96 h·μg/L versus 255 ± 81 h·μg/L, P < 0.001). This difference remained significant after adjusting for age and race. For a same C 0, every 0.01 mg/kg increase in Tac dose resulted in an AUC24 increase of 3.59 h·μg/L. Conclusions This study challenges the general belief that C 0 levels are sufficiently reliable to estimate systemic drug exposure. We demonstrated that patients requiring a relatively high Tac dose to attain therapeutic C 0 levels have higher drug exposure and could therefore potentially be overdosed.
Collapse
Affiliation(s)
- Lien Haverals
- Department of Nephrology, Antwerp University Hospital, Edegem, Belgium
| | - Laurence Roosens
- Department of Clinical and Biological Sciences, Antwerp University Hospital, Edegem, Belgium
| | - Kristien Wouters
- Department of Statistics, Antwerp University Hospital, Edegem, Belgium
| | - Pierre Marquet
- Department of Pharmacology and Transplantation, University of Limoges, CHU Limoges, Limoges, France
| | - Caroline Monchaud
- Department of Pharmacology and Transplantation, University of Limoges, CHU Limoges, Limoges, France
| | - Annick Massart
- Department of Nephrology, Antwerp University Hospital, Edegem, Belgium
| | - Daniel Abramowicz
- Department of Nephrology, Antwerp University Hospital, Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Edegem, Belgium
| | - Rachel Hellemans
- Department of Nephrology, Antwerp University Hospital, Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Edegem, Belgium
| |
Collapse
|
14
|
Markianos K, Dong F, Gorman B, Shi Y, Dochtermann D, Saxena U, Devineni P, Moser J, Muralidhar S, Ramoni R, Tsao P, Pyarajan S, Przygodzki R. Pharmacogenetic allele variant frequencies: An analysis of the VA's Million Veteran Program (MVP) as a representation of the diversity in US population. PLoS One 2023; 18:e0274339. [PMID: 36827430 PMCID: PMC9956596 DOI: 10.1371/journal.pone.0274339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2022] [Accepted: 02/03/2023] [Indexed: 02/26/2023] Open
Abstract
We present allele frequencies of pharmacogenomics relevant variants across multiple ancestry in a sample representative of the US population. We analyzed 658,582 individuals with genotype data and extracted pharmacogenomics relevant single nucleotide variant (SNV) alleles, human leukocyte antigens (HLA) 4-digit alleles and an important copy number variant (CNV), the full deletion/duplication of CYP2D6. We compiled distinct allele frequency tables for European, African American, Hispanic, and Asian ancestry individuals. In addition, we compiled allele frequencies based on local ancestry reconstruction in the African-American (2-way deconvolution) and Hispanic (3-way deconvolution) cohorts.
Collapse
Affiliation(s)
- Kyriacos Markianos
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Frederic Dong
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Bryan Gorman
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Yunling Shi
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Daniel Dochtermann
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Uma Saxena
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Poornima Devineni
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Jennifer Moser
- The office of Research and Development, US Department of Veterans Affairs, Washington, DC, United States of America
| | - Sumitra Muralidhar
- The office of Research and Development, US Department of Veterans Affairs, Washington, DC, United States of America
| | - Rachel Ramoni
- The office of Research and Development, US Department of Veterans Affairs, Washington, DC, United States of America
| | - Philip Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston HealthCare System, Boston, MA, United States of America
| | - Ronald Przygodzki
- The office of Research and Development, US Department of Veterans Affairs, Washington, DC, United States of America
| | | |
Collapse
|
15
|
Impact of CYP3A5 Status on the Clinical and Financial Outcomes Among African American Kidney Transplant Recipients. Transplant Direct 2022; 8:e1379. [PMID: 36204191 PMCID: PMC9529042 DOI: 10.1097/txd.0000000000001379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/06/2022] Open
Abstract
Pharmacogenetic profiling of transplant recipients demonstrates that the marked variation in the metabolism of immunosuppressive medications, particularly tacrolimus, is related to genetic variants. Patients of African ancestry are less likely to carry loss-of-function (LoF) variants in the CYP3A5 gene and therefore retain a rapid metabolism phenotype and higher clearance of tacrolimus. Patients with this rapid metabolism typically require higher dosing to achieve therapeutic trough concentrations. This study aims to further characterize the impact of CYP3A5 genotype on clinical outcomes and financial expenditure.
Collapse
|
16
|
Genetic polymorphisms of pharmacogenomic VIP variants in the Lahu population from Yunnan Province. Gene 2022; 844:146825. [PMID: 35995116 DOI: 10.1016/j.gene.2022.146825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/10/2022] [Revised: 06/29/2022] [Accepted: 08/16/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Pharmacogenomics has been widely used to study the very important pharmacogenetic (VIP) variants among populations, but information on pharmacogenomics in the Lahu population is limited. The purpose of this study was to determine the differences in the distribution of VIP variants between the Lahu and the other 26 populations. METHODS We genotyped 55 VIP variants of 27 genes in the Lahu population from the PharmGKB database. χ2 test was used to compare the genotype and allele frequencies between the Lahu and the other 26 populations from the 1000 Genomes Project. RESULTS The genotype and allele frequencies of single nucleotide polymorphisms (SNPs) on rs20417 (PTGS2), rs776746 (CYP3A5), rs2115819 (ALOX5), and rs3093105 (CYP4F2) were considerably different in the Lahu population compared with those in the other 26 populations. Besides, based on the PharmGKB database, we identified several VIP variants that may alter the drug metabolism of aspirin (PTGS2), tacrolimus (CYP3A5), montelukast (ALOX5), and vitamin E (CYP4F2). CONCLUSION The results show that there are significant differences in the genotype frequency distribution between the Lahu and the other 26 populations. Our study supplements the pharmacogenomics information of the Lahu population and provides a theoretical basis for individualized medicine in Lahu.
Collapse
|
17
|
Liu XL, Guan YP, Wang Y, Huang K, Jiang FL, Wang J, Yu QH, Qiu KF, Huang M, Wu JY, Zhou DH, Zhong GP, Yu XX. Population Pharmacokinetics and Initial Dosage Optimization of Tacrolimus in Pediatric Hematopoietic Stem Cell Transplant Patients. Front Pharmacol 2022; 13:891648. [PMID: 35873585 PMCID: PMC9298550 DOI: 10.3389/fphar.2022.891648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/08/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There is a substantial lack of tacrolimus pharmacokinetic information in pediatric hematopoietic stem cell transplant (HSCT) patients. This study aimed to develop population pharmacokinetics (PopPK) of tacrolimus in pediatric HSCT patients and to devise model-guided dosage regimens. Methods: A retrospective analysis was performed on 86 pediatric HSCT patients who received tacrolimus intravenously or orally. A total of 578 tacrolimus trough concentrations (C0) were available for pharmacokinetic analysis using a non-linear mixed-effects modeling method. Demographic and clinical data were included and assessed as covariates via the stepwise method. Bayesian estimators were used to devise pediatric dosage regimens that targeted C0 of 5-15 ng mL-1. Results: A one-compartment model with first-order absorption adequately described the tacrolimus pharmacokinetics. Clearance (CL), volume of distribution (V), and typical bioavailability (F) in this study were estimated to be 2.42 L h-1 (10.84%), 79.6 L (16.51%), and 19% (13.01%), respectively. Body weight, hematocrit, post-transplantation days, and caspofungin and azoles concomitant therapy were considered significant covariates for tacrolimus CL. Hematocrit had a significant impact on the V of tacrolimus. In the subgroup cohort of children (n = 24) with CYP3A5 genotype, the clearance was 1.38-fold higher in CYP3A5 expressers than in non-expressers. Simulation indicated that the initial dosage optimation of tacrolimus for intravenous and oral administration was recommended as 0.025 and 0.1 mg kg-1 d-1 (q12h), respectively. Conclusion: A PopPK model for tacrolimus in pediatric HSCT patients was developed, showing good predictive performance. Model-devised dosage regimens with trough tacrolimus concentrations provide a practical strategy for achieving the therapeutic range.
Collapse
Affiliation(s)
- Xiao-Lin Liu
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yan-Ping Guan
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ying Wang
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Huang
- Department of Paediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fu-Lin Jiang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian Wang
- Department of Paediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi-Hong Yu
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Kai-Feng Qiu
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Huang
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun-Yan Wu
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dun-Hua Zhou
- Department of Paediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guo-Ping Zhong
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Xia Yu
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Zdesenko G, Mduluza T, Mutapi F. Pharmacogenetics of Praziquantel Metabolism: Evaluating the Cytochrome P450 Genes of Zimbabwean Patients During a Schistosomiasis Treatment. Front Genet 2022; 13:914372. [PMID: 35754834 PMCID: PMC9213834 DOI: 10.3389/fgene.2022.914372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/06/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Schistosomiasis is a parasitic disease infecting over 236 million people annually, with the majority affected residing on the African continent. Control of this disease is reliant on the drug praziquantel (PZQ), with treatment success dependent on an individual reaching PZQ concentrations lethal to schistosomes. Despite the complete reliance on PZQ to treat schistosomiasis in Africa, the characterization of the pharmacogenetics associated with PZQ metabolism in African populations has been sparse. We aimed to characterize genetic variation in the drug-metabolising cytochrome P450 enzymes (CYPs) and determine the association between each variant and the efficacy of PZQ treatment in Zimbabwean patients exposed to Schistosoma haematobium infection. Genomic DNA from blood samples of 114 case-control Zimbabweans infected with schistosomes were sequenced using the CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5 genes as targets. Bioinformatic tools were used to identify and predict functional effects of detected single nucleotide polymorphisms (SNPs). A random forest (RF) model was then used to assess SNPs most predictive of PZQ efficacy, with a misclassification rate of 29%. SNPs were detected across all six genes, with 70 SNPs identified and multiple functional changes to the CYP enzymes predicted. Only four SNPs were significantly associated with PZQ efficacy using χ2 tests, with rs951840747 (OR: 3.61, p = 0.01) in the CYP1A2 gene having the highest odds of an individual possessing this SNP clearing infection, and rs6976017 (OR: 2.19, p = 0.045) of CYP3A5 determined to be the most predictive of PZQ efficacy via the RF. Only the rs28371702 (CC) genotype (OR: 2.36, p = 0.024) of CYP2D6 was significantly associated with an unsuccessful PZQ treatment. This study adds to the genomic characterization of the diverse populations in Africa and identifies variants relevant to other pharmacogenetic studies crucial for the development and usage of drugs in these populations.
Collapse
Affiliation(s)
- Grace Zdesenko
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom.,Ashworth Laboratories, NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Edinburgh, United Kingdom
| | - Takafira Mduluza
- Ashworth Laboratories, NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Edinburgh, United Kingdom.,Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| | - Francisca Mutapi
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom.,Ashworth Laboratories, NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
19
|
Zhang SF, Tang BH, An-Hua W, Du Y, Guan ZW, Li Y. Effect of drug combination on tacrolimus target dose in renal transplant patients with different CYP3A5 genotypes. Xenobiotica 2022; 52:312-321. [PMID: 35395919 DOI: 10.1080/00498254.2022.2064252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/18/2022]
Abstract
Various factors, including genetic polymorphisms, drug-drug interactions, and patient characteristics influence the blood concentrations of tacrolimus in renal transplant patients. In the present study, we established a population pharmacokinetic model to explore the effect of combined use of Wuzhi capsules/echinocandins and the patients' biochemical parameters such as hematocrit on blood concentrations and target doses of tacrolimus in renal transplant patients with different CYP3A5 genotypes. The aim of the study was to propose an individualized tacrolimus administration regimen for early renal transplant recipients.In this retrospective cohort study, we included 240 renal transplant recipients within 21 days of surgery (174 males and 66 females, mean age 39.4 years), who received tacrolimus alone (n = 54), in combination with Wuzhi capsules (99) or caspofungin (57) or micafungin (30). We collected demographic characteristics, clinical indicators, CYP3A5 genotypes, and 1950 steady-state trough concentrations of tacrolimus and included them in population pharmacokinetic model. An additional 110 renal transplant recipients and 625 steady-state trough concentrations of tacrolimus were included for external validation of the model. The population pharmacokinetic model was established and Monte Carlo was used to simulate probabilities for achieving the target concentration for individual tacrolimus administration.A two-compartment model of first-order absorption and elimination was developed to describe the population pharmacokinetics of tacrolimus. CYP3A5 genotypes and co-administration of Wuzhi capsules, as well as time after renal transplantation and hematocrit, were important factors affecting the clearance of tacrolimus. We found no obvious change in trend in the scatter plot of tacrolimus clearance rate vs. hematocrit. The Monte Carlo simulation indicated the following recommended doses of tacrolimus alone: 0.14 mg·kg-1·d-1 for genotype CYP3A5*1*1, 0.12 mg·kg-1·d-1 for CYP3A5*1*3, and 0.10 mg·kg-1·d-1 for CYP3A5*3*3. For patients receiving the combination with Wuzhi capsules, the recommended doses of tacrolimus were 0.10 mg·kg-1·d-1 for CYP3A5*1*1, 0.08 mg·kg-1·d-1 for CYP3A5*1*3, and 0.06 mg·kg-1·d-1 for CYP3A5*3*3 genotypes. Caspofungin or micafungin had no effect on the clearance of tacrolimus in renal transplant recipients.The population pharmacokinetics of tacrolimus in renal transplant patients was evaluated and the individual administration regimen of tacrolimus was simulated. For early kidney transplant recipients receiving tacrolimus treatment, not only body weight, but also CYP3A5 genotypes and drugs used in combination should be considered when determining the target dose of tacrolimus.
Collapse
Affiliation(s)
- Shu-Fang Zhang
- School of Pharmacy, Shandong First Medical University, Tai'an, China.,Department of Pharmacy, Tai'an City Central Hospital, Tai'an, China
| | - Bo-Hao Tang
- School of Pharmaceutical Science, Shandong University, Ji'nan, China
| | - Wei An-Hua
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Du
- School of Pharmacy, Shandong First Medical University, Tai'an, China
| | - Zi-Wan Guan
- School of Pharmaceutical Science, Shandong University, Ji'nan, China
| | - Yan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, China
| |
Collapse
|
20
|
Al-Kofahi M, Oetting WS, Schladt DP, Remmel RP, Guan W, Wu B, Dorr CR, Mannon RB, Matas AJ, Israni AK, Jacobson PA. Precision Dosing for Tacrolimus Using Genotypes and Clinical Factors in Kidney Transplant Recipients of European Ancestry. J Clin Pharmacol 2021; 61:1035-1044. [PMID: 33512723 PMCID: PMC11240873 DOI: 10.1002/jcph.1823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2020] [Accepted: 01/26/2021] [Indexed: 12/14/2022]
Abstract
Genetic variation in the CYP3A4 and CYP3A5 (CYP3A4/5) genes, which encode the key enzymes in tacrolimus metabolism, is associated with tacrolimus clearance and dose requirements. Tacrolimus has a narrow therapeutic index with high intra- and intersubject variability, in part because of genetic variation. High tacrolimus clearance and low trough concentration are associated with a greater risk for rejection, whereas high troughs are associated with calcineurin-induced toxicity. The objective of this study was to develop a model of tacrolimus clearance with a dosing equation accounting for genotypes and clinical factors in adult kidney transplant recipients of European ancestry that could preemptively guide dosing. Recipients receiving immediate-release tacrolimus for maintenance immunosuppression from 2 multicenter studies were included. Participants in the GEN03 study were used for tacrolimus model development (n = 608 recipients) and was validated by prediction performance in the DeKAF Genomics study (n = 1361 recipients). Nonlinear mixed-effects modeling was used to develop the apparent oral tacrolimus clearance (CL/F) model. CYP3A4/5 genotypes and clinical covariates were tested for their influence on CL/F. The predictive performance of the model was determined by assessing the bias (median prediction error [ME] and median percentage error [MPE]) and the precision (root median squared error [RMSE]) of the model. CYP3A5*3, CYP3A4*22, corticosteroids, calcium channel blocker and antiviral drug use, age, and diabetes significantly contributed to the interindividual variability of oral tacrolimus apparent clearance. The bias (ME, MPE) and precision (RMSE) of the final model was good, 0.49 ng/mL, 6.5%, and 3.09 ng/mL, respectively. Prospective testing of this equation is warranted.
Collapse
Affiliation(s)
- Mahmoud Al-Kofahi
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - William S Oetting
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - David P Schladt
- Hennepin Health Research Institute, Minneapolis, Minnesota, USA
| | - Rory P Remmel
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weihua Guan
- Department of Biostatistics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Baolin Wu
- Department of Biostatistics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Casey R Dorr
- Hennepin Health Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, Hennepin Healthcare, University of Minnesota, Minneapolis, Minnesota, USA
| | - Roslyn B Mannon
- Division of Nephrology, University of Nebraska, Omaha, Nebraska, USA
| | - Arthur J Matas
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ajay K Israni
- Hennepin Health Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, Hennepin Healthcare, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Epidemiology & Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pamala A Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
21
|
Bishop JR, Huang RS, Brown JT, Mroz P, Johnson SG, Allen JD, Bielinski SJ, England J, Farley JF, Gregornik D, Giri J, Kroger C, Long SE, Luczak T, McGonagle EJ, Ma S, Matey ET, Mandic PK, Moyer AM, Nicholson WT, Petry N, Pawloski PA, Schlichte A, Schondelmeyer SW, Seifert RD, Speedie MK, Stenehjem D, Straka RJ, Wachtl J, Waring SC, Ness BV, Zierhut HA, Aliferis C, Wolf SM, McCarty CA, Jacobson PA. Pharmacogenomics education, research and clinical implementation in the state of Minnesota. Pharmacogenomics 2021; 22:681-691. [PMID: 34137665 DOI: 10.2217/pgs-2021-0058] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
Several healthcare organizations across Minnesota have developed formal pharmacogenomic (PGx) clinical programs to increase drug safety and effectiveness. Healthcare professional and student education is strong and there are multiple opportunities in the state for learners to gain workforce skills and develop advanced competency in PGx. Implementation planning is occurring at several organizations and others have incorporated structured utilization of PGx into routine workflows. Laboratory-based and translational PGx research in Minnesota has driven important discoveries in several therapeutic areas. This article reviews the state of PGx activities in Minnesota including educational programs, research, national consortia involvement, technology, clinical implementation and utilization and reimbursement, and outlines the challenges and opportunities in equitable implementation of these advances.
Collapse
Affiliation(s)
- Jeffrey R Bishop
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.,Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - R Stephanie Huang
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Jacob T Brown
- Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota College of Pharmacy, Duluth, MN 55812, USA
| | - Pawel Mroz
- Department of Laboratory Medicine & Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Steven G Johnson
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Josiah D Allen
- University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.,Medigenics Consulting LLC, Minneapolis, MN 55407, USA
| | - Suzette J Bielinski
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Joel F Farley
- Department of Pharmaceutical Care & Health Systems, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - David Gregornik
- Pharmacogenomics Program, Children's Minnesota, Minneapolis, MN 55407, USA
| | - Jyothsna Giri
- Mayo Clinic Center for Individualized Medicine, Mayo Clinic College of Medicine & Science, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Susie E Long
- MHealth Fairview. Acute Care Pharmacy Services, Minneapolis, MN 55455, USA
| | - Tiana Luczak
- Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota College of Pharmacy, Duluth, MN 55812, USA.,Essentia Health, Duluth, MN 55805, USA
| | - Erin J McGonagle
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Sisi Ma
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Eric T Matey
- Department of Pharmacy, Mayo Clinic College of Medicine & Science, Mayo Clinic, Rochester, MN 55905, USA
| | - Pinar K Mandic
- Department of Finance, University of Minnesota Carlson School of Management, Minneapolis, MN 55455, USA
| | - Ann M Moyer
- Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine & Science, Mayo Clinic, Rochester, MN 55905, USA
| | - Wayne T Nicholson
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic College of Medicine & Science, Mayo Clinic, Rochester, MN 55905, USA
| | - Natasha Petry
- Sanford Health Imagenetics, Sioux Falls, SD 57105, USA.,Department of Pharmacy Practice, North Dakota State University College of Health Professions, Fargo, ND 58108, USA
| | | | | | - Stephen W Schondelmeyer
- Department of Pharmaceutical Care & Health Systems, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Randall D Seifert
- Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota College of Pharmacy, Duluth, MN 55812, USA
| | - Marilyn K Speedie
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - David Stenehjem
- Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota College of Pharmacy, Duluth, MN 55812, USA
| | - Robert J Straka
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Jason Wachtl
- Geritom Medical, Inc, Bloomington, MN 55438, USA
| | | | - Brian Van Ness
- Department of Genetics, Cell Biology & Development, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Heather A Zierhut
- Department of Genetics, Cell Biology & Development, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Constantin Aliferis
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Susan M Wolf
- Law School, Medical School, Consortium on Law & Values in Health, Environment & the Life Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Catherine A McCarty
- Department of Family Medicine & Biobehavioral Health, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Pamala A Jacobson
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| |
Collapse
|
22
|
Abstract
Clinical outcomes after organ transplantation have greatly improved in the past 2 decades with the discovery and development of immunosuppressive drugs such as calcineurin inhibitors, antiproliferative agents, and mammalian target of rapamycin inhibitors. However, individualized dosage regimens have not yet been fully established for these drugs except for therapeutic drug monitoring-based dosage modification because of extensive interindividual variations in immunosuppressive drug pharmacokinetics. The variations in immunosuppressive drug pharmacokinetics are attributed to interindividual variations in the functional activity of cytochrome P450 enzymes, UDP-glucuronosyltransferases, and ATP-binding cassette subfamily B member 1 (known as P-glycoprotein or multidrug resistance 1) in the liver and small intestine. Some genetic variations have been found to be involved to at least some degree in pharmacokinetic variations in post-transplant immunosuppressive therapy. It is well known that the frequencies and effect size of minor alleles vary greatly between different races. Thus, ethnic considerations might provide useful information for optimizing individualized immunosuppressive therapy after organ transplantation. Here, we review ethnic factors affecting the pharmacokinetics of immunosuppressive drugs requiring therapeutic drug monitoring, including tacrolimus, cyclosporine, mycophenolate mofetil, sirolimus, and everolimus.
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Despite advances in medical and device-based therapies for advanced heart failure as well as public policy, disparities by race/ethnicity persist in heart failure clinical outcomes. The purpose of this review is to describe disparities in outcomes by race--ethnicity in patients after receipt of heart transplantation and left ventricular assist device (LVAD), and the current understanding of factors contributing to these disparities. RECENT FINDINGS The proportion of black and Latinx patients receiving advanced heart failure therapies continues to rise, and they have worse hemodynamic profiles at the time of referral for heart transplantation and LVAD. Black patients have lower rates of survival after heart transplantation, in part because of higher rates of cellular and humoral rejection that may be mediated through unique gene pathways, and increased risk for allosensitization and de-novo donor-specific antibodies. Factors that have previously been cited as reasons for worse outcomes in race--ethnic minorities, including psychosocial risk and lower SES, may not be as strongly correlated with outcomes after LVAD. SUMMARY Black and Latinx patients are sicker at the time of referral for advanced heart failure therapies. Despite higher psychosocial risk factors among race--ethnic minorities, outcomes after LVAD appear to be similar to white patients. Black patients continue to have lower posttransplant survival, because of a complex interplay of immunologic susceptibility, clinical and socioeconomic factors. No single factor accounts for the disparities in clinical outcomes for race--ethnic minorities, and thus consideration of these components together is critical in management of these patients.
Collapse
|
24
|
Faravardeh A, Akkina S, Villicana R, Guerra G, Moten MA, Meier-Kriesche U, Stevens DR, Patel SJ, Bunnapradist S. Efficacy and Safety of Once-Daily LCP-Tacrolimus Versus Twice-Daily Immediate-Release Tacrolimus in Adult Hispanic Stable Kidney Transplant Recipients: Sub-Group Analysis from a Phase 3 Trial. Ann Transplant 2021; 26:e929535. [PMID: 33859155 PMCID: PMC8056872 DOI: 10.12659/aot.929535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The pharmacokinetics and metabolism of tacrolimus, an immunosuppressant commonly used to prevent transplant rejection, can differ in specific subpopulations. This analysis examined treatment outcomes and safety of immediate-release tacrolimus (IR-Tac) and LCP-tacrolimus (LCPT) in stable Hispanic kidney transplant recipients. MATERIAL AND METHODS This was a post hoc analysis of clinical trial data from Hispanic adult stable kidney transplant recipients randomized to remain on IR-Tac or convert from IR-Tac to a reduced dose of LCPT (NCT00817206). Composite treatment failure was evaluated at 12 months. Estimated glomerular filtration rate and tacrolimus trough concentrations were evaluated over 12 months. RESULTS Fifty-five stable (LCPT n=26, IR-Tac n=29) kidney transplant recipients who self-identified as Hispanic or Latino were included in this analysis. Composite treatment failure occurred in 1 patient (4%) who converted to LCPT and 1 (3%) who remained on IR-Tac. The estimated glomerular filtration rate was stable over time and similar in the 2 treatment groups (P=0.08). Tacrolimus trough levels for both groups were similar over time in the 2 treatment groups (P=0.98). Treatment-emergent adverse events were similar in patients who converted to LCPT and in those who remained on IR-Tac. CONCLUSIONS Efficacy and safety were similar in Hispanic kidney transplant recipients who converted from IR-Tac to LCPT and in those remaining on IR-Tac.
Collapse
Affiliation(s)
| | - Sanjeev Akkina
- Loyola Outpatient Center, Loyola University Medical Center, Maywood, IL, USA
| | - Rafael Villicana
- Loma Linda University Transplant Institute, Loma Linda University Health, Loma Linda, CA, USA
| | - Giselle Guerra
- Miami Transplant Institute, University of Miami Health System, Miami, FL, USA
| | - Misbah A Moten
- Department of Medical Affairs, Veloxis Pharmaceuticals, Cary, NC, USA
| | | | - Daniel R Stevens
- Department of Medical Affairs, Veloxis Pharmaceuticals, Cary, NC, USA
| | - Samir J Patel
- Department of Medical Affairs, Veloxis Pharmaceuticals, Cary, NC, USA
| | | |
Collapse
|
25
|
Abstract
The efficacy, safety, and tolerability of drugs are dependent on numerous factors that influence their disposition. A dose that is efficacious and safe for one individual may result in sub-therapeutic or toxic blood concentrations in others. A significant source of this variability in drug response is drug metabolism, where differences in presystemic and systemic biotransformation efficiency result in variable degrees of systemic exposure (e.g., AUC, Cmax, and/or Cmin) following administration of a fixed dose.Interindividual differences in drug biotransformation have been studied extensively. It is recognized that both intrinsic factors (e.g., genetics, age, sex, and disease states) and extrinsic factors (e.g., diet , chemical exposures from the environment, and the microbiome) play a significant role. For drug-metabolizing enzymes, genetic variation can result in the complete absence or enhanced expression of a functional enzyme. In addition, upregulation and downregulation of gene expression, in response to an altered cellular environment, can achieve the same range of metabolic function (phenotype), but often in a less predictable and time-dependent manner. Understanding the mechanistic basis for variability in drug disposition and response is essential if we are to move beyond the era of empirical, trial-and-error dose selection and into an age of personalized medicine that will improve outcomes in maintaining health and treating disease.
Collapse
Affiliation(s)
- Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
| | - Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Brice D Thompson
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Christi W Cho
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Tambur AR, Campbell P, Chong AS, Feng S, Ford ML, Gebel H, Gill RG, Kelsoe G, Kosmoliaptsis V, Mannon RB, Mengel M, Reed EF, Valenzuela NM, Wiebe C, Dijke IE, Sullivan HC, Nickerson P. Sensitization in transplantation: Assessment of risk (STAR) 2019 Working Group Meeting Report. Am J Transplant 2020; 20:2652-2668. [PMID: 32342639 PMCID: PMC7586936 DOI: 10.1111/ajt.15937] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/12/2019] [Revised: 04/01/2020] [Accepted: 04/20/2020] [Indexed: 01/25/2023]
Abstract
The purpose of the STAR 2019 Working Group was to build on findings from the initial STAR report to further clarify the expectations, limitations, perceptions, and utility of alloimmune assays that are currently in use or in development for risk assessment in the setting of organ transplantation. The goal was to determine the precision and clinical feasibility/utility of such assays in evaluating both memory and primary alloimmune risks. The process included a critical review of biologically driven, state-of-the-art, clinical diagnostics literature by experts in the field and an open public forum in a face-to-face meeting to promote broader engagement of the American Society of Transplantation and American Society of Histocompatibility and Immunogenetics membership. This report summarizes the literature review and the workshop discussions. Specifically, it highlights (1) available assays to evaluate the attributes of HLA antibodies and their utility both as clinical diagnostics and as research tools to evaluate the effector mechanisms driving rejection; (2) potential assays to assess the presence of alloimmune T and B cell memory; and (3) progress in the development of HLA molecular mismatch computational scores as a potential prognostic biomarker for primary alloimmunity and its application in research trial design.
Collapse
Affiliation(s)
- Anat R. Tambur
- Department of SurgeryComprehensive Transplant CenterNorthwestern UniversityChicagoIllinoisUSA
| | - Patricia Campbell
- Department of Laboratory Medicine & PathologyUniversity of AlbertaEdmontonCanada
| | - Anita S. Chong
- Section of TransplantationDepartment of SurgeryThe University of ChicagoChicagoIllinoisUSA
| | - Sandy Feng
- Department of SurgeryUCSF Medical CenterSan FranciscoCaliforniaUSA
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant CenterEmory UniversityAtlantaGeorgiaUSA
| | - Howard Gebel
- Department of PathologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Ronald G. Gill
- Department of ImmunologyUniversity of ColoradoDenverColoradoUSA
| | - Garnett Kelsoe
- Department of ImmunologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | | | - Roslyn B. Mannon
- Department of MedicineDivision of NephrologyUniversity of Alabama School of MedicineBirminghamAlabamaUSA
| | - Michael Mengel
- Department of Laboratory Medicine & PathologyUniversity of AlbertaEdmontonCanada
| | - Elaine F. Reed
- Department of Pathology and Laboratory MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Nicole M. Valenzuela
- Department of Pathology and Laboratory MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chris Wiebe
- Department of MedicineUniversity of ManitobaWinnipegManitobaCanada
| | - I. Esme Dijke
- Department of Laboratory Medicine & PathologyUniversity of AlbertaEdmontonCanada
| | - Harold C. Sullivan
- Department of PathologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Peter Nickerson
- Department of MedicineUniversity of ManitobaWinnipegManitobaCanada
| |
Collapse
|
27
|
Chen L, Li C, Huo N, Mishuk AU, Hansen RA, Harris I, Kiptanui Z, Qian J. Oral generic tacrolimus initiation and substitution in the Medicaid population: a new user cohort study. Curr Med Res Opin 2020; 36:1533-1540. [PMID: 32644886 DOI: 10.1080/03007995.2020.1793750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Although existing studies have compared clinical efficacy and pharmacokinetics of generic with brand tacrolimus, little is known about generic tacrolimus utilization patterns or factors predicting generic substitution. This study examines associations between patient-level factors and generic tacrolimus initiation or substitution among new users of oral generic or brand-name tacrolimus. METHODS This new user retrospective cohort study identified 374 beneficiaries who initiated generic or brand tacrolimus treatment, using 100% Medicaid administrative claims data from 11 states in 2011-2012. Outcomes were generic tacrolimus initiation and substitution within 12 months of treatment initiation. Multivariable logistic regression and Cox proportional hazards models were used to identify factors associated with generic tacrolimus initiation and substitution. RESULTS Two-thirds of oral tacrolimus new users initiated generic tacrolimus. Patients who were older age and black were significantly more likely to initiate generic tacrolimus (adjusted odds ratio [AOR] = 1.02, 95% confidence interval [CI] = 1.001-1.03; AOR = 2.54, 95% CI = 1.40-4.62; respectively). Patients with more concomitant prescriptions, more comorbidities, or higher initial daily dosage had significantly lower likelihoods of initiating generic tacrolimus (AOR = 0.93, 95% CI = 0.87-0.99; AOR = 0.87, 95% CI = 0.77-0.99; AOR = 0.96, 95% CI = 0.93-0.993). Among brand tacrolimus new users, 23.6% substituted with generics within 12 months, and an addition of prior hospitalization or unit of initial daily dosage increment was associated with 35% (subdistribution hazard ratio [SHR] = 1.35, 95% CI = 1.02-1.76) or 2% (SHR = 1.02, 95% CI = 1.00-1.04) increase in likelihood of generic substitution, respectively. CONCLUSIONS Understanding associations between patient-level factors with generic tacrolimus initiation and substitution could help practitioners and policymakers monitor treatment effect and facilitate generic tacrolimus utilization.
Collapse
Affiliation(s)
- Li Chen
- Department of Medicine, Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN, USA
| | - Chao Li
- Department of Health Outcomes Research and Policy, Auburn University, Auburn, AL, USA
| | - Nan Huo
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Ahmed Ullah Mishuk
- Department of Health Outcomes Research and Policy, Auburn University, Auburn, AL, USA
| | - Richard A Hansen
- Department of Health Outcomes Research and Policy, Auburn University, Auburn, AL, USA
| | | | | | - Jingjing Qian
- Department of Health Outcomes Research and Policy, Auburn University, Auburn, AL, USA
| |
Collapse
|
28
|
Brazeau DA, Attwood K, Meaney CJ, Wilding GE, Consiglio JD, Chang SS, Gundroo A, Venuto RC, Cooper L, Tornatore KM. Beyond Single Nucleotide Polymorphisms: CYP3A5∗3∗6∗7 Composite and ABCB1 Haplotype Associations to Tacrolimus Pharmacokinetics in Black and White Renal Transplant Recipients. Front Genet 2020; 11:889. [PMID: 32849848 PMCID: PMC7433713 DOI: 10.3389/fgene.2020.00889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/19/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A5 (CYP3A5) isoenzymes and membrane transport by P-glycoprotein. Interpatient pharmacokinetic variability has been associated with genotypic variants for both CYP3A5 or ABCB1. Tacrolimus pharmacokinetics was investigated in 65 stable Black and Caucasian post-renal transplant patients by assessing the effects of multiple alleles in both CYP3A5 and ABCB1. A metabolic composite based upon the CYP3A5 polymorphisms: ∗3(rs776746), ∗6(10264272), and ∗7(41303343), each independently responsible for loss of protein expression was used to classify patients as extensive, intermediate and poor metabolizers. In addition, the role of ABCB1 on tacrolimus pharmacokinetics was assessed using haplotype analysis encompassing the single nucleotide polymorphisms: 1236C > T (rs1128503), 2677G > T/A(rs2032582), and 3435C > T(rs1045642). Finally, a combined analysis using both CYP3A5 and ABCB1 polymorphisms was developed to assess their inter-related influence on tacrolimus pharmacokinetics. Extensive metabolizers identified as homozygous wild type at all three CYP3A5 loci were found in 7 Blacks and required twice the tacrolimus dose (5.6 ± 1.6 mg) compared to Poor metabolizers [2.5 ± 1.1 mg (P < 0.001)]; who were primarily Whites. These extensive metabolizers had 2-fold faster clearance (P < 0.001) with 50% lower AUC∗ (P < 0.001) than Poor metabolizers. No differences in C12 h were found due to therapeutic drug monitoring. The majority of blacks (81%) were classified as either Extensive or Intermediate Metabolizers requiring higher tacrolimus doses to accommodate the more rapid clearance. Blacks who were homozygous for one or more loss of function SNPS were associated with lower tacrolimus doses and slower clearance. These values are comparable to Whites, 82% of who were in the Poor metabolic composite group. The ABCB1 haplotype analysis detected significant associations of the wildtype 1236T-2677T-3435T haplotype to tacrolimus dose (P = 0.03), CL (P = 0.023), CL/LBW (P = 0.022), and AUC∗ (P = 0.078). Finally, analysis combining CYP3A5 and ABCB1 genotypes indicated that the presence of the ABCB1 3435 T allele significantly reduced tacrolimus clearance for all three CPY3A5 metabolic composite groups. Genotypic associations of tacrolimus pharmacokinetics can be improved by using the novel composite CYP3A5∗3∗4∗5 and ABCB1 haplotypes. Consideration of multiple alleles using CYP3A5 metabolic composites and drug transporter ABCB1 haplotypes provides a more comprehensive appraisal of genetic factors contributing to interpatient variability in tacrolimus pharmacokinetics among Whites and Blacks.
Collapse
Affiliation(s)
- Daniel A. Brazeau
- Department of Pharmacy Practice, Administration and Research, School of Pharmacy, Marshall University, Huntington, WV, United States
| | - Kristopher Attwood
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Calvin J. Meaney
- Immunosuppressive Pharmacology Research Program, Translational Pharmacology Research Core, NYS Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, United States
- School of Pharmacy and Pharmaceutical Sciences, Buffalo, NY, United States
| | - Gregory E. Wilding
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Joseph D. Consiglio
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - Shirley S. Chang
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Erie County Medical Center, Buffalo, NY, United States
| | - Aijaz Gundroo
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Erie County Medical Center, Buffalo, NY, United States
| | - Rocco C. Venuto
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Erie County Medical Center, Buffalo, NY, United States
| | - Louise Cooper
- Immunosuppressive Pharmacology Research Program, Translational Pharmacology Research Core, NYS Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, United States
- School of Pharmacy and Pharmaceutical Sciences, Buffalo, NY, United States
| | - Kathleen M. Tornatore
- Immunosuppressive Pharmacology Research Program, Translational Pharmacology Research Core, NYS Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, United States
- School of Pharmacy and Pharmaceutical Sciences, Buffalo, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW The current tools to proactively guide and individualize immunosuppression in solid organ transplantation are limited. Despite continued improvements in posttransplant outcomes, the adverse effects of over-immunosuppression or under-immunosuppression are common. The present review is intended to highlight recent advances in individualized immunosuppression. RECENT FINDINGS There has been a great focus on genomic information to predict drug dose requirements, specifically on single nucleotide polymorphisms of CYP3A5 and ABCB1. Furthermore, biomarker studies have developed ways to better predict clinical outcomes, such as graft rejection. SUMMARY The integration of advanced computing tools, such as artificial neural networks and machine learning, with genome sequencing has led to intriguing findings on individual or group-specific dosing requirements. Rapid computing allows for processing of data and discovering otherwise undetected clinical patterns. Genetic polymorphisms of CYP3A5 and ABCB1 have yielded results to suggest varying dose requirements correlated with race and sex. Newly proposed biomarkers offer precise and noninvasive ways to monitor patient's status. Cell-free DNA quantitation is increasingly explored as an indicator of allograft injury and rejection, which can help avoid unneeded biopsies and more frequently monitor graft function.
Collapse
Affiliation(s)
- Shengyi Fu
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
30
|
Impacts of High Intra- and Inter-Individual Variability in Tacrolimus Pharmacokinetics and Fast Tacrolimus Metabolism on Outcomes of Solid Organ Transplant Recipients. J Clin Med 2020; 9:jcm9072193. [PMID: 32664531 PMCID: PMC7408675 DOI: 10.3390/jcm9072193] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022] Open
Abstract
Tacrolimus is a first-line calcineurin inhibitor (CNI) and an integral part of the immunosuppressive strategy in solid organ transplantation. Being a dose-critical drug, tacrolimus has a narrow therapeutic index that necessitates periodic monitoring to maintain the drug’s efficacy and reduce the consequences of overexposure. Tacrolimus is characterized by substantial intra- and inter-individual pharmacokinetic variability. At steady state, the tacrolimus blood concentration to daily dose ratio (C/D ratio) has been described as a surrogate for the estimation of the individual metabolism rate, where a low C/D ratio reflects a higher rate of metabolism. Fast tacrolimus metabolism (low C/D ratio) is associated with the risk of poor outcomes after transplantation, including reduced allograft function and survival, higher allograft rejection, CNI nephrotoxicity, a faster decline in kidney function, reduced death-censored graft survival (DCGS), post-transplant lymphoproliferative disorders, dyslipidemia, hypertension, and cardiovascular events. In this article, we discuss the potential role of the C/D ratio in a noninvasive monitoring strategy for identifying patients at risk for potential adverse events post-transplant.
Collapse
|
31
|
Genetic Variants Associated With Immunosuppressant Pharmacokinetics and Adverse Effects in the DeKAF Genomics Genome-wide Association Studies. Transplantation 2020; 103:1131-1139. [PMID: 30801552 PMCID: PMC6597284 DOI: 10.1097/tp.0000000000002625] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The immunosuppressants tacrolimus and mycophenolate are important components to the success of organ transplantation, but are also associated with adverse effects, such as nephrotoxicity, anemia, leukopenia, and new-onset diabetes after transplantation. In this report, we attempted to identify genetic variants which are associated with these adverse outcomes. METHODS We performed a genome-wide association study, using a genotyping array tailored specifically for transplantation outcomes containing 722 147 single nucleotide polymorphisms, and 2 cohorts of kidney allograft recipients-a discovery cohort and a confirmation cohort-to identify and then confirm genetic variants associated with immunosuppressant pharmacokinetics and adverse outcomes. RESULTS Several genetic variants were found to be associated with tacrolimus trough concentrations. We did not confirm variants associated with the other phenotypes tested although several suggestive variants were identified. CONCLUSIONS These results show that adverse effects associated with tacrolimus and mycophenolate are complex, and recipient risk is not determined by a few genetic variants with large effects with but most likely are due to many variants, each with small effect sizes, and clinical factors.
Collapse
|
32
|
Vangala C, Niu J, Montez-Rath ME, Yan J, Navaneethan SD, Naik AD, Winkelmayer WC. Hip Fracture Risk among Hemodialysis-Dependent Patients Prescribed Opioids and Gabapentinoids. J Am Soc Nephrol 2020; 31:1325-1334. [PMID: 32371535 DOI: 10.1681/asn.2019090904] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2019] [Accepted: 03/11/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Despite opioids' known association with hip fracture risk in the general population, they are commonly prescribed to patients with ESKD. Whether use of opioids or gabapentinoids (also used to treat pain in patients with ESKD) contributes to hip fracture risk in patients with ESKD on hemodialysis remains unknown. METHODS In a case-control study nested within the US Renal Data System, we identified all hip fracture events recorded among patients dependent on hemodialysis from January 2009 through September 2015. Eligible cases were risk-set matched on index date with ten eligible controls. We required >1 year of Medicare Parts A and B coverage and >3 years of part D coverage to study cumulative longer-term exposure. To examine new, short-term exposure, we selected individuals with >18 months of Part D coverage and no prior opioid or gabapentinoid use between 18 and 7 months before index. We used conditional logistic regression to estimate unadjusted and multivariable-adjusted odds ratios (ORs) and 95% confidence intervals (95% CI). RESULTS For the longer-term analyses, we identified 4912 first-time hip fracture cases and 49,120 controls. Opioid use was associated with increased hip fracture risk (adjusted OR, 1.39; 95% CI, 1.26 to 1.53). Subgroups of low, moderate, and high use yielded adjusted ORs of 1.33 (95% CI, 1.20 to 1.47), 1.53 (95% CI, 1.36 to 1.72), and 1.66 (95% CI, 1.45 to 1.90), respectively. The association with hip fractures was also elevated with new, short-term use (adjusted OR, 1.38; 95% CI, 1.25 to 1.52). There were no associations between gabapentinoid use and hip fracture. CONCLUSIONS Among patients dependent on hemodialysis in the United States, both short-term and longer-term use of opioid analgesics were associated with hip fracture events.
Collapse
Affiliation(s)
- Chandan Vangala
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas .,Clinical Effectiveness and Population Health, Houston Center for Innovations in Quality, Effectiveness, and Safety, Houston, Texas.,Section of Nephrology & Solid Organ Transplant, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Jingbo Niu
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas.,Methodology & Analytics Core, Houston Center for Innovations in Quality, Effectiveness, and Safety, Houston, Texas
| | - Maria E Montez-Rath
- Division of Nephrology, Stanford University School of Medicine, Palo Alto, California
| | - Jingyin Yan
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Sankar D Navaneethan
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas.,Section of Nephrology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Aanand D Naik
- Education & Training Core, Houston Center for Innovations in Quality, Effectiveness, and Safety, Houston, Texas
| | - Wolfgang C Winkelmayer
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
33
|
Radouani F, Zass L, Hamdi Y, Rocha JD, Sallam R, Abdelhak S, Ahmed S, Azzouzi M, Benamri I, Benkahla A, Bouhaouala-Zahar B, Chaouch M, Jmel H, Kefi R, Ksouri A, Kumuthini J, Masilela P, Masimirembwa C, Othman H, Panji S, Romdhane L, Samtal C, Sibira R, Ghedira K, Fadlelmola F, Kassim SK, Mulder N. A review of clinical pharmacogenetics Studies in African populations. Per Med 2020; 17:155-170. [PMID: 32125935 PMCID: PMC8093600 DOI: 10.2217/pme-2019-0110] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022]
Abstract
Effective interventions and treatments for complex diseases have been implemented globally, however, coverage in Africa has been comparatively lower due to lack of capacity, clinical applicability and knowledge on the genetic contribution to disease and treatment. Currently, there is a scarcity of genetic data on African populations, which have enormous genetic diversity. Pharmacogenomics studies have the potential to revolutionise treatment of diseases, therefore, African populations are likely to benefit from these approaches to identify likely responders, reduce adverse side effects and optimise drug dosing. This review discusses clinical pharmacogenetics studies conducted in African populations, focusing on studies that examined drug response in complex diseases relevant to healthcare. Several pharmacogenetics associations have emerged from African studies, as have gaps in knowledge.
Collapse
Affiliation(s)
- Fouzia Radouani
- Research Department, Chlamydiae & Mycoplasmas Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco
| | - Lyndon Zass
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, CIDRI Africa Wellcome Trust Centre, University of Cape Town, South Africa
| | - Yosr Hamdi
- Laboratory of Biomedical Genomics & Oncogenetics, Institut Pasteur de Tunis, Université Tunis El Manar, 13, Place Pasteur BP 74, 1002 Tunis, Belvédère, Tunisie
| | - Jorge da Rocha
- Sydney Brenner Institute for Molecular Bioscience, University of The Witwatersrand, Johannesburg, South Africa
| | - Reem Sallam
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbaseya, Cairo 11381, Egypt
| | - Sonia Abdelhak
- Laboratory of Biomedical Genomics & Oncogenetics, Institut Pasteur de Tunis, Université Tunis El Manar, 13, Place Pasteur BP 74, 1002 Tunis, Belvédère, Tunisie
| | - Samah Ahmed
- Centre for Bioinformatics & Systems Biology, Faculty of Science, University of Khartoum, 321 Khartoum, Sudan.,Faculty of Clinical & Industrial Pharmacy, National University, Khartoum, Sudan
| | - Maryame Azzouzi
- Research Department, Chlamydiae & Mycoplasmas Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco
| | - Ichrak Benamri
- Research Department, Chlamydiae & Mycoplasmas Laboratory, Institut Pasteur du Maroc, Casablanca 20360, Morocco.,Systems & Data Engineering Team, National School of Applied Sciences of Tangier, Morocco
| | - Alia Benkahla
- Laboratory of Bioinformatics, Biomathematics & Biostatistics LR 16 IPT 09, Institute Pasteur de Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms & Therapeutic Molecules, Pasteur Institute of Tunis, 13 Place Pasteur, BP74, Tunis Belvedere- University of Tunis El Manar, Tunisia
| | - Melek Chaouch
- Laboratory of Bioinformatics, Biomathematics & Biostatistics LR 16 IPT 09, Institute Pasteur de Tunis, Tunisia
| | - Haifa Jmel
- Laboratory of Biomedical Genomics & Oncogenetics, Institut Pasteur de Tunis, Université Tunis El Manar, 13, Place Pasteur BP 74, 1002 Tunis, Belvédère, Tunisie
| | - Rym Kefi
- Laboratory of Biomedical Genomics & Oncogenetics, Institut Pasteur de Tunis, Université Tunis El Manar, 13, Place Pasteur BP 74, 1002 Tunis, Belvédère, Tunisie
| | - Ayoub Ksouri
- Laboratory of Bioinformatics, Biomathematics & Biostatistics LR 16 IPT 09, Institute Pasteur de Tunis, Tunisia.,Laboratory of Venoms & Therapeutic Molecules, Pasteur Institute of Tunis, 13 Place Pasteur, BP74, Tunis Belvedere- University of Tunis El Manar, Tunisia
| | - Judit Kumuthini
- H3ABioNet, Bioinformatics Department, Centre for Proteomic & Genomic Research, Cape Town, South Africa
| | - Phumlani Masilela
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, CIDRI Africa Wellcome Trust Centre, University of Cape Town, South Africa
| | - Collen Masimirembwa
- Sydney Brenner Institute for Molecular Bioscience, University of The Witwatersrand, Johannesburg, South Africa.,DMPK Department, African Institute of Biomedical Science & Technology, Harare, Zimbabwe
| | - Houcemeddine Othman
- Sydney Brenner Institute for Molecular Bioscience, University of The Witwatersrand, Johannesburg, South Africa
| | - Sumir Panji
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, CIDRI Africa Wellcome Trust Centre, University of Cape Town, South Africa
| | - Lilia Romdhane
- Laboratory of Biomedical Genomics & Oncogenetics, Institut Pasteur de Tunis, Université Tunis El Manar, 13, Place Pasteur BP 74, 1002 Tunis, Belvédère, Tunisie.,Département des Sciences de la Vie, Faculté des Sciences de Bizerte, Université Carthage, 7021 Jarzouna, BP 21, Tunisie
| | - Chaimae Samtal
- Biotechnology Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohammed Ben Abdellah University, Fez 30000, Morocco.,Department of Biology, University of Mohammed Premier, Oujda, Morocco.,Department of Biology Faculty of Sciences, University of Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Rania Sibira
- Centre for Bioinformatics & Systems Biology, Faculty of Science, University of Khartoum, 321 Khartoum, Sudan.,Department of Neurosurgery, National Center For Neurological Sciences, Khartoum, Sudan
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics & Biostatistics LR 16 IPT 09, Institute Pasteur de Tunis, Tunisia
| | - Faisal Fadlelmola
- Centre for Bioinformatics & Systems Biology, Faculty of Science, University of Khartoum, 321 Khartoum, Sudan
| | - Samar Kamal Kassim
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Ain Shams University, Abbaseya, Cairo 11381, Egypt
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, CIDRI Africa Wellcome Trust Centre, University of Cape Town, South Africa
| |
Collapse
|
34
|
Ahsan T, Urmi NJ, Sajib AA. Heterogeneity in the distribution of 159 drug-response related SNPs in world populations and their genetic relatedness. PLoS One 2020; 15:e0228000. [PMID: 31971968 PMCID: PMC6977754 DOI: 10.1371/journal.pone.0228000] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/16/2019] [Accepted: 01/03/2020] [Indexed: 12/25/2022] Open
Abstract
Interethnic variability in drug response arises from genetic differences associated with drug metabolism, action and transport. These genetic variations can affect drug efficacy as well as cause adverse drug reactions (ADRs). We retrieved drug-response related single nucleotide polymorphism (SNP) associated data from databases and analyzed to elucidate population specific distribution of 159 drug-response related SNPs in twenty six populations belonging to five super-populations (African, Admixed Americans, East Asian, European and South Asian). Significant interpopulation differences exist in the minor (variant) allele frequencies (MAFs), linkage disequilibrium (LD) and haplotype distributions among these populations. 65 of the drug-response related alleles, which are considered as minor (variant) in global population, are present as the major alleles (frequency ≥0.5) in at least one or more populations. Populations that belong to the same super-population have similar distribution pattern for majority of the variant alleles. These drug response related variant allele frequencies and their pairwise LD measure (r2) can clearly distinguish the populations in a way that correspond to the known evolutionary history of human and current geographic distributions, while D' cannot. The data presented here may aid in identifying drugs that are more appropriate and/or require pharmacogenetic testing in these populations. Our findings emphasize on the importance of distinct, ethnicity-specific clinical guidelines, especially for the African populations, to avoid ADRs and ensure effective drug treatment.
Collapse
Affiliation(s)
- Tamim Ahsan
- Department of Genetic Engineering & Biotechnology, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | | | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
35
|
More Severe Deficits in Performance Status at Time of Liver Transplant is Associated With Significantly Higher Risk of Death Following Liver Transplantation. J Clin Gastroenterol 2019; 53:e392-e399. [PMID: 30762610 DOI: 10.1097/mcg.0000000000001187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Abstract
GOAL To evaluate the impact of Karnofsky Performance Status score (KPSS) at the time of liver transplantation (LT) on post-LT survival. BACKGROUND While the Model for End-Stage Liver Disease (MELD) score is used to prioritize individuals for LT, it does not specifically incorporate functional status into patient assessment for LT. METHODS Using 2005 to 2016 United Network for Organ Sharing data, all adults (age 18 y and above) undergoing LT were identified. The association of KPSS at the time of LT (KPSS 1: functional status 80% to 100%, KPSS 2: 60% to 70%, KPSS 3: 40% to 50%, KPSS 4: 10% to 30%) with post-LT survival was evaluated using Kaplan-Meier methods and adjusted multivariate logistic regression models. RESULTS Among 66,397 LT recipients (68% male, 72% non-Hispanic white, 22% hepatocellular carcinoma, median age: 55 to 57), women were more likely to be KPSS 4 at the time of LT compared with men (27.95% vs. 22.79%; P<0.001) and African Americans (25.43% vs. 23.03%; P<0.001) and Hispanics (31.69% vs. 23.03%; P<0.001) were more likely to be KPSS 4 than non-Hispanic whites. Worse KPSS at LT correlated with higher post-LT mortality [compared with KPSS 1: Hazard Ratio (HR) for KPSS 2: 1.16, 95% confidence interval (CI): 1.10-1.22; HR for KPSS 3: 1.40; 95% CI: 1.32-1.49; HR for KPSS 4: 1.67; 95% CI: 1.55-1.79]. This increased mortality seen with worse KPSS was observed among all liver disease etiologies and in patients with and without hepatocellular carcinoma. CONCLUSIONS Worse functional status at the time of LT is strongly associated with higher risk of mortality following LT, emphasizing the importance of optimizing performance status in the preoperative period.
Collapse
|
36
|
Mohamed ME, Schladt DP, Guan W, Wu B, van Setten J, Keating B, Iklé D, Remmel RP, Dorr CR, Mannon RB, Matas AJ, Israni AK, Oetting WS, Jacobson PA. Tacrolimus troughs and genetic determinants of metabolism in kidney transplant recipients: A comparison of four ancestry groups. Am J Transplant 2019; 19:2795-2804. [PMID: 30953600 PMCID: PMC6763344 DOI: 10.1111/ajt.15385] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/20/2018] [Revised: 03/04/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Tacrolimus trough and dose requirements vary dramatically between individuals of European and African American ancestry. These differences are less well described in other populations. We conducted an observational, prospective, multicenter study from which 2595 kidney transplant recipients of European, African, Native American, and Asian ancestry were studied for tacrolimus trough, doses, and genetic determinants of metabolism. We studied the well-known variants and conducted a CYP3A4/5 gene-wide analysis to identify new variants. Daily doses, and dose-normalized troughs were significantly different between the four groups (P < .001). CYP3A5*3 (rs776746) was associated with higher dose-normalized tacrolimus troughs in all groups but occurred at different allele frequencies and had differing effect sizes. The CYP3A5*6 (rs10264272) and *7 (rs413003343) variants were only present in African Americans. CYP3A4*22 (rs35599367) was not found in any of the Asian ancestry samples. We identified seven suggestive variants in the CYP3A4/5 genes associated with dose-normalized troughs in Native Americans (P = 1.1 × 10-5 -8.8 × 10-6 ) and one suggestive variant in Asian Americans (P = 5.6 × 10-6 ). Tacrolimus daily doses and dose-normalized troughs vary significantly among different ancestry groups. We identified potential new variants important in Asians and Native Americans. Studies with larger populations should be conducted to assess the importance of the identified suggestive variants.
Collapse
Affiliation(s)
- Moataz E. Mohamed
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA,Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | | | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Baolin Wu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Brendan Keating
- Department of Surgery, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Rory P. Remmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Casey R. Dorr
- Hennepin Healthcare Research Institute, Minneapolis, MN, USA,Department of Medicine, University of Minnesota, Hennepin Healthcare, Minneapolis, MN
| | | | - Arthur J. Matas
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Ajay K. Israni
- Hennepin Healthcare Research Institute, Minneapolis, MN, USA,Department of Medicine, University of Minnesota, Hennepin Healthcare, Minneapolis, MN,Department of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN, USA
| | - William S. Oetting
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Pamala A. Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
37
|
Largeau B, Guellec CBL, Longuet H, Lesne P, Bouvarel A, Préteseille L, Marquet P, Halimi JM, Büchler M, Gatault P, Noble J. Comparison of Tacrolimus Starting Doses Based on CYP3A5 Phenotype or Genotype in Kidney Transplant Recipients. Prog Transplant 2019; 29:300-308. [PMID: 31514576 DOI: 10.1177/1526924819873905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Selection of expected phenotypes (ie, expressers/non-expressers) is currently used in CYP3A5*3 genotype-based tacrolimus dosing. The authors assessed whether a dosing regimen based on the 3 CYP3A5 genotypes may reduce the occurrence of inadequate exposure. METHODS Tacrolimus whole blood trough levels (C 0) were retrieved from a retrospective cohort of 100 kidney transplant recipients treated with a starting dose of 0.15 (non-expressers) or 0.30 (expressers) mg/kg/d. The authors evaluated the occurrence of overexposures (12 < C 0 < 20 ng/mL) or toxic concentrations (C 0 ≥ 20 ng/mL). These results were used to set up a new strategy based on the 3 distinct CYP3A5 genotypes, which relevance was evaluated in a prospective cohort of 107 patients. RESULTS In the retrospective cohort, non-expressers exhibited frequent overexposure (63.6%) or toxic C 0 (20.8%). Among expressers, none of the homozygous *1 carriers exhibited overexposure contrary to 25% of the heterozygotes. Based on these results, new tacrolimus starting doses were set at 0.10, 0.20, and 0.30 mg/kg/d for CYP3A5*3/*3, CYP3A5*1/*3, and CYP3A5*1/*1 genotypes, respectively. Tacrolimus overexposure was reduced in the CYP3A5*3/*3 group (63.6% vs 40%, P = .0038). None of the heterozygous patients exhibited toxic tacrolimus C 0. Clinical outcomes were not different between the 2 periods, whatever the genotype. Our results indicate that the best tacrolimus exposure was obtained for doses of 0.10, 0.20, and 0.20 mg/kg/d for CYP3A5*3/3, CYP3A5*1/*3, and CYP3A5*1/*1, respectively. CONCLUSIONS Our results confirm that selecting tacrolimus dosing regimen according to the expected phenotype is appropriate, but that lower than currently recommended doses may be preferable.
Collapse
Affiliation(s)
- Bérenger Largeau
- CHRU de Tours, Laboratoire de Biochimie et Biologie Moléculaire, Tours, France
| | - Chantal Barin-Le Guellec
- Université de Tours, Université de Limoges, INSERM, Individual profiling and prevention of risks with immunosuppressive therapies and transplantation (IPPRITT) - UMR 1248, CHRU de Tours, Laboratoire de Biochimie et Biologie Moléculaire, Tours, France
| | - Hélène Longuet
- CHRU de Tours, Service de Néphrologie-Hypertension artérielle, Dialyses et Transplantation Rénale, FHU SUPORT, Tours, France
| | - Philippe Lesne
- CHRU de Tours, Laboratoire de Biochimie et Biologie Moléculaire, Tours, France
| | - Antoine Bouvarel
- CHRU de Tours, Laboratoire de Biochimie et Biologie Moléculaire, Tours, France
| | - Laura Préteseille
- CHRU de Tours, Laboratoire de Biochimie et Biologie Moléculaire, Tours, France
| | - Pierre Marquet
- Université de Limoges, INSERM, Individual profiling and prevention of risks with immunosuppressive therapies and transplantation (IPPRITT) - UMR 1248, CHU de Limoges, Service de Pharmacologie et Toxicologie, FHU SUPORT, Limoges, France
| | - Jean-Michel Halimi
- Université de Tours, Transplantation, immunologie et inflammation (T2I) - EA4245, CHRU de Tours, Service de Néphrologie-Hypertension artérielle, Dialyses et Transplantation Rénale, FHU SUPORT, Tours, France
| | - Matthias Büchler
- Université de Tours, Transplantation, immunologie et inflammation (T2I) - EA4245, CHRU de Tours, Service de Néphrologie-Hypertension artérielle, Dialyses et Transplantation Rénale, FHU SUPORT, Tours, France
| | - Philippe Gatault
- Université de Tours, Transplantation, immunologie et inflammation (T2I) - EA4245, CHRU de Tours, Service de Néphrologie-Hypertension artérielle, Dialyses et Transplantation Rénale, FHU SUPORT, Tours, France
| | - Johan Noble
- CHRU de Tours, Service de Néphrologie-Hypertension artérielle, Dialyses et Transplantation Rénale, FHU SUPORT, Tours, France
| |
Collapse
|
38
|
Elmaasarani Z, Mardis C, Gylten L, Taber DJ, Fleming J, Patel N, Baliga P, Rao V, Rohan V, DuBay D, Pilch NA. Protocol-based nurse coordinator management of ambulatory tacrolimus dosing in de novo renal transplant recipients-A single-center experience with a large African American population. Clin Transplant 2019; 33:e13701. [PMID: 31461791 DOI: 10.1111/ctr.13701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/29/2019] [Revised: 07/12/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Transplant nurse (RN) coordinators review tacrolimus levels frequently and would be capable of making dose adjustments autonomously if not limited by their license. Collaborative practice agreements could be an answer; thus, the aim of this evaluation was to determine if an RN-driven protocol could be used safely and effectively to manage tacrolimus in ambulatory kidney transplant (KTX) recipients. METHODS This was a retrospective review of all solitary adult KTX recipients between August 1, 2016, and July 29, 2017. The primary objective was to evaluate protocol adherence and frequency of use, and secondary objectives were to evaluate the utility of the protocol both overall and based on ethnicity. RESULTS A total of 173 patients were included in the evaluation (59% African American [AA], 41% non-African American [non-AA). RN coordinators followed the protocol for 75% of tacrolimus adjustments; however, they only responded to 27% of the overall levels. There was no difference in 180-day tacrolimus-associated readmission (15% AA vs 5% non-AA, P = .06), biopsy-proven acute rejection (4% AA vs 7% non-AA, P = .363), or hyperkalemia (34% AA vs 32% non-AA, P = .87) between groups. CONCLUSIONS Transplant nurse coordinators are capable of accurately following a protocol for tacrolimus dosage adjustment in a large, racially diverse kidney transplant center.
Collapse
Affiliation(s)
- Zana Elmaasarani
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
| | | | - Logan Gylten
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
| | - David J Taber
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA.,Department of Pharmacy, Ralph H Johnson VAMC, Charleston, SC, USA
| | - James Fleming
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Neha Patel
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
| | - Prabhakar Baliga
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Vinaya Rao
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Vinayak Rohan
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Derek DuBay
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Nicole A Pilch
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
39
|
Morris AA. Utilizing gene expression profiling to understand immunologic mechanisms that impact racial disparities after heart transplant. J Heart Lung Transplant 2019; 38:830-832. [PMID: 31352998 DOI: 10.1016/j.healun.2019.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/14/2019] [Accepted: 06/16/2019] [Indexed: 11/29/2022] Open
|
40
|
Prytuła A, Cransberg K, Raes A. Drug-metabolizing enzymes CYP3A as a link between tacrolimus and vitamin D in renal transplant recipients: is it relevant in clinical practice? Pediatr Nephrol 2019; 34:1201-1210. [PMID: 30058048 DOI: 10.1007/s00467-018-4030-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/30/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 01/08/2023]
Abstract
CYP3A enzymes are involved in the metabolism of calcineurin inhibitor tacrolimus as well as vitamin D. In this review, we summarize the clinical aspects of CYP3A-mediated metabolism of tacrolimus and vitamin D with emphasis on the influence of single-nucleotide polymorphisms on tacrolimus disposition. We describe the utility of 4β hydroxycholesterol as a marker of CYP3A activity. Then, we discuss the possible interaction between calcineurin inhibitors and vitamin D in solid organ transplant recipients. Also, we review other mechanisms which may contribute to side effects of calcineurin inhibitors on bone. Lastly, suggestions for future research and clinical perspectives are discussed.
Collapse
Affiliation(s)
- Agnieszka Prytuła
- Paediatric Nephrology and Rheumatology Department, Ghent University Hospital, C Heymanslaan 10, 9000, Ghent, Belgium.
| | - Karlien Cransberg
- Paediatric Nephrology Department, Erasmus MC- Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Ann Raes
- Paediatric Nephrology and Rheumatology Department, Ghent University Hospital, C Heymanslaan 10, 9000, Ghent, Belgium.,Safepedrug Unit, Ghent, Belgium
| |
Collapse
|
41
|
Daya M, Barnes KC. African American ancestry contribution to asthma and atopic dermatitis. Ann Allergy Asthma Immunol 2019; 122:456-462. [PMID: 30772392 PMCID: PMC6500742 DOI: 10.1016/j.anai.2019.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/15/2018] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Asthma and atopic dermatitis (AD) are complex diseases with striking disparities across racial and ethnic groups, which may be partly attributable to genetic factors. Here we summarize current knowledge from asthma and AD genome-wide association studies (GWAS) and pharmacogenetic studies in African ancestry populations. DATA SOURCES GWAS catalog; PUBMed. STUDY SELECTIONS GWAS catalog studies with trait annotations "asthma" and "atopic eczema" and African ancestry individuals in the discovery dataset; the recent CAAPA asthma GWAS; reports on pharmacogenetic studies in asthma and AD. RESULTS Although GWASs have revolutionized gene discovery for multiple complex traits, African Americans continue to be severely underrepresented in sufficiently powered genetics studies. Indeed, of the 16 asthma and 21 AD loci that reached genomewide significance in Europeans, very few have replicated in African ancestry populations. Challenges in comparing results from European vs African ancestry cohorts include modest sample size, differences in risk allele frequency, effect size, correlation between genetic variants, and environmental exposure in evolutionary history. African Americans also constitute a small percentage of dermatological and respiratory-focused clinical trials. Pharmacogenetic studies have similarly been focused largely on non-Hispanic whites, despite compelling evidence that genetic variation from different ancestral backgrounds may alter therapeutic efficacy of asthma and AD drugs. CONCLUSION Large-scale genetic studies of asthma and AD in African Americans are essential to reduce research and health disparities and empower scientific discoveries.
Collapse
Affiliation(s)
- Michelle Daya
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado Denver, Aurora, Colorado.
| |
Collapse
|
42
|
van Hoogdalem EJ, Jones Iii JP, Constant J, Achira M. Science-based Ethnic Bridging in Drug Development; Review of Recent Precedence and Suggested Steps Forward. ACTA ACUST UNITED AC 2019; 14:197-207. [PMID: 30961506 DOI: 10.2174/1574884714666190408125206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/18/2019] [Revised: 02/01/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure, safety and/or efficacy of drugs are subject to potential differences between human races or ethnicities, as acknowledged by regulatory guidance and by label texts of various, but not all approved drugs. OBJECTIVE The objective of the present review was to assess recent regulatory precedence on drug use and race or ethnicity, with the goal of identifying opportunities for increasing the informative value of clinical ethnic or racial bridging in drug development. METHODS Recently, (January 2014-July 2018) FDA approved drug product label texts and approval packages were reviewed for claims, comments and underlying data on use of the product in specific ethnic or racial groups. RESULTS Among the 266 FDA-approved products, no product with unambiguous race- or ethnicity specific dosing instructions was retrieved. A small majority (55%) was approved with a claim or comment on race or ethnicity, and of these, a large majority (87%) was based on population pharmacokinetic data analysis. Statements were often related to incidence of a genotype for drug metabolizing enzyme or for other risk factors, or were related to body weight. Absence of clinically relevant exposure differences were often justified in terms of exposure ratios that notably exceeded the typical 0.80-1.25 no-effect boundary. CONCLUSIONS Recent precedence reflected a pragmatic, descriptive approach of racial or ethnic bridging, apparently meeting current regulatory expectations, whilst not resulting in strict guidance to prescribers. We recommend further work on defining the objectives of bridging studies, as well as criteria for their design and data analysis. Regarding the latter, we recommend investigating the value of prospectively defined tests for similarity with appropriate follow-up analysis in the case where the test has failed.
Collapse
Affiliation(s)
| | - John P Jones Iii
- PRA Health Sciences, Scientific Affairs - Clinical Pharmacology, Blue Bell, PA, United States
| | - John Constant
- PRA Health Sciences, Scientific Affairs, Victoria, BC, Canada
| | - Meguru Achira
- Clinical Pharmacology, Takeda PRA Development Center KK, Osaka 540-8645, Japan
| |
Collapse
|
43
|
Ampong DN. Landmarks of pharmacogenomics and some considerations for clinical practice. Ther Adv Psychopharmacol 2019; 9:2045125319896650. [PMID: 35186262 PMCID: PMC8851126 DOI: 10.1177/2045125319896650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/20/2019] [Accepted: 11/26/2019] [Indexed: 11/15/2022] Open
Abstract
Since the completion of the Human Genome Project 28 years ago, myriad genomics applications have risen in areas such as agriculture, livestock, infectious agents, forensics, bioenergy, ancestry, health, disease, and medicine. This was driven partly by the US government's ability to use a unique program to facilitate genome sequencing to the point where the cost of sequencing a whole human genome is not prohibitive. However, application of this knowledge of the double helix twisted DNA at the bedside in psychiatric clinical practice has little to report, despite US Food and Drug Administration (FDA) approval of nearly 40 psychotropic drugs, as well as specific guidelines for their application. Patients with treatment-resistant mental illness, history of unresponsiveness to psychotropic medications, and history or family history of serious adverse effects to psychotropic drugs may qualify for pharmacogenomics (PGx) testing with insurance reimbursement, or a low, out-of-pocket, payment of not greater than US $300. Psychiatric mental health nurse practitioners and providers who utilize PGx will not only improve patient care outcomes, but also contribute to the acceleration of the potential diagnostic and preventive capabilities of PGx testing.
Collapse
Affiliation(s)
- David Nana Ampong
- College of Health, University of Alaska Anchorage, 3211 Providence Driver, Anchorage, AK 99508, USA
| |
Collapse
|
44
|
Guo Y, Busch MP, Seielstad M, Endres-Dighe S, Westhoff CM, Keating B, Hoppe C, Bordbar A, Custer B, Butterworth AS, Kanias T, Mast AE, Kleinman S, Lu Y, Page GP. Development and evaluation of a transfusion medicine genome wide genotyping array. Transfusion 2019; 59:101-111. [PMID: 30456907 PMCID: PMC7032526 DOI: 10.1111/trf.15012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Many aspects of transfusion medicine are affected by genetics. Current single-nucleotide polymorphism (SNP) arrays are limited in the number of targets that can be interrogated and cannot detect all variation of interest. We designed a transfusion medicine array (TM-Array) for study of both common and rare transfusion-relevant variations in genetically diverse donor and recipient populations. STUDY DESIGN AND METHODS The array was designed by conducting extensive bioinformatics mining and consulting experts to identify genes and genetic variation related to a wide range of transfusion medicine clinical relevant and research-related topics. Copy number polymorphisms were added in the alpha globin, beta globin, and Rh gene clusters. RESULTS The final array contains approximately 879,000 SNP and copy number polymorphism markers. Over 99% of SNPs were called reliably. Technical replication showed the array to be robust and reproducible, with an error rate less than 0.03%. The array also had a very low Mendelian error rate (average parent-child trio accuracy of 0.9997). Blood group results were in concordance with serology testing results, and the array accurately identifies rare variants (minor allele frequency of 0.5%). The array achieved high genome-wide imputation coverage for African-American (97.5%), Hispanic (96.1%), East Asian (94.6%), and white (96.1%) genomes at a minor allele frequency of 5%. CONCLUSIONS A custom array for transfusion medicine research has been designed and evaluated. It gives wide coverage and accurate identification of rare SNPs in diverse populations. The TM-Array will be useful for future genetic studies in the diverse fields of transfusion medicine research.
Collapse
Affiliation(s)
- Yuelong Guo
- RTI International, Research Triangle Park, North Carolina
| | - Michael P Busch
- Vitalant Research Institute (formerly Blood Systems Research Institute), San Francisco, California
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California
| | - Mark Seielstad
- Vitalant Research Institute (formerly Blood Systems Research Institute), San Francisco, California
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California
| | | | | | - Brendan Keating
- Penn Transplant Institute, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carolyn Hoppe
- UCSF Benioff Children's Hospital Oakland, Oakland, California
| | | | - Brian Custer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- NIHR Blood and Transplant Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| | - Tamir Kanias
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alan E Mast
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Steve Kleinman
- University of British Columbia, Victoria, British Columbia, Canada
| | - Yontao Lu
- Affymetrix Incorporated, Santa Clara, California
| | | |
Collapse
|
45
|
Identification of genetic variants associated with tacrolimus metabolism in kidney transplant recipients by extreme phenotype sampling and next generation sequencing. THE PHARMACOGENOMICS JOURNAL 2018; 19:375-389. [PMID: 30442921 PMCID: PMC6522337 DOI: 10.1038/s41397-018-0063-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/16/2018] [Revised: 09/11/2018] [Accepted: 09/27/2018] [Indexed: 12/26/2022]
Abstract
An extreme phenotype sampling (EPS) model with targeted next-generation sequencing (NGS) identified genetic variants associated with tacrolimus (Tac) metabolism in subjects from the Deterioration of Kidney Allograft Function (DeKAF) Genomics cohort which included 1,442 European Americans (EA) and 345 African Americans (AA). This study included 48 subjects separated into 4 groups of 12 (AA high, AA low, EA high, EA low). Groups were selected by the extreme phenotype of dose-normalized Tac trough concentrations after adjusting for common genetic variants and clinical factors. NGS spanned >3 Mb of 28 genes and identified 18,661 genetic variants (3,961 previously unknown). A group of 125 deleterious variants, by SIFT analysis, were associated with Tac troughs in EAs (burden test, p=0.008), CYB5R2 was associated with Tac troughs in AAs (SKAT, p=0.00079). In CYB5R2, rs61733057 (increased allele frequency in AAs) was predicted to disrupt protein function by SIFT and PolyPhen2 analysis. The variants merit further validation.
Collapse
|
46
|
Campagne O, Mager DE, Tornatore KM. Population Pharmacokinetics of Tacrolimus in Transplant Recipients: What Did We Learn About Sources of Interindividual Variabilities? J Clin Pharmacol 2018; 59:309-325. [PMID: 30371942 DOI: 10.1002/jcph.1325] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2018] [Accepted: 09/18/2018] [Indexed: 12/24/2022]
Abstract
Tacrolimus, a calcineurin inhibitor, is a common immunosuppressant prescribed after organ transplantation and has notable inter- and intrapatient pharmacokinetic variability. The sources of variability have been investigated using population pharmacokinetic modeling over the last 2 decades. This article provides an updated synopsis on published nonlinear mixed-effects analyses developed for tacrolimus in transplant recipients. The objectives were to establish a detailed overview of the current data and to investigate covariate relationships determined by the models. Sixty-three published analyses were reviewed, and data regarding the study design, modeling approach, and resulting findings were extracted and summarized. Most of the studies investigated tacrolimus pharmacokinetics in adult and pediatric renal and liver transplants after administration of the immediate-release formulation. Model structures largely depended on the study sampling strategy, with ∼50% of studies developing a 1-compartment model using trough concentrations and a 2-compartment model with delayed absorption from intensive sampling. The CYP3A5 genotype, as a covariate, consistently impacted tacrolimus clearance, and dosing adjustments were required to achieve similar drug exposure among patients. Numerous covariates were identified as sources of interindividual variability on tacrolimus pharmacokinetics with limited consistency across these studies, which may be the result of the study designs. Additional analyses are required to further evaluate the potential impact of these covariates and the clinical implementation of these models to guide tacrolimus dosing recommendations. This article may be useful for guiding the design of future population pharmacokinetic studies and provides recommendations for the selection of an existing optimal model to individualize tacrolimus therapy.
Collapse
Affiliation(s)
- Olivia Campagne
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA.,Faculty of Pharmacy, Universités Paris Descartes-Paris Diderot, Paris, France
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Kathleen M Tornatore
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Immunosuppressive Pharmacology Research Program, Translational Pharmacology Research Core, NYS Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
47
|
Asleh R, Snipelisky D, Hathcock M, Kremers W, Liu D, Batzler A, Jenkins G, Kushwaha S, Pereira NL. Genomewide association study reveals novel genetic loci associated with change in renal function in heart transplant recipients. Clin Transplant 2018; 32:e13395. [PMID: 30160337 DOI: 10.1111/ctr.13395] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/29/2018] [Revised: 08/13/2018] [Accepted: 08/23/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Renal dysfunction occurs commonly after heart transplantation (HTx) with wide inter-individual variability but whether a genetic predisposition exists in these patients is unknown. Genomewide association studies (GWAS) have not been performed to assess the association of genetic variation with change in renal function after HTx. METHODS Clinical and demographic data of patients who underwent HTx and provided blood samples and consent for genetic analysis were included. Genotyping was performed using Illumina Infinium Human CoreExome v1.0 analysis kit. A GWAS utilizing linear regression models was performed with estimated glomerular filtration rate (eGFR) at 1 year as the phenotype after adjusting for baseline eGFR prior to HTx and conversion from calcineurin inhibitor to sirolimus as primary immunosuppression therapy. RESULTS A total of 251 HTx recipients were genotyped for 314,903 single nucleotide polymorphisms (SNPs). The mean (SD) age was 50 (12.5) years; most patients were of European origin (n = 243, 96.8%) and males (n = 179, 71.3%). After adjustment for potential confounders, two variants, rs17033285 (P = 4.3 × 10-7 ) and rs4917601 (P = 6.46 × 10-7 ), in a long non-coding RNA (lncRNA) gene LINC01121 and a pseudogene BTBD7P2, were identified to have a significant association with change in GFR at 1 year after HTx. CONCLUSIONS Our first of its kind GWAS demonstrates that genetic variation affects renal function after HTx independent of other risk factors. Agnostic genetic approaches such as these may lead to identification of novel biological pathways such as the role of lncRNAs in the development of renal dysfunction post-HTx.
Collapse
Affiliation(s)
- Rabea Asleh
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - David Snipelisky
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Matthew Hathcock
- Department of Biomedical Statistic and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Walter Kremers
- Department of Biomedical Statistic and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Anthony Batzler
- Department of Biomedical Statistic and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Gregory Jenkins
- Department of Biomedical Statistic and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Sudhir Kushwaha
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Naveen L Pereira
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
48
|
Dave S, Dodge JL, Terrault NA, Sarkar M. Racial and Ethnic Differences in Graft Loss Among Female Liver Transplant Recipients. Transplant Proc 2018; 50:1413-1423. [PMID: 29880364 DOI: 10.1016/j.transproceed.2018.02.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2017] [Revised: 01/29/2018] [Accepted: 02/17/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Racial differences in post-liver transplantation (LT) outcomes are identified in predominantly male cohorts. Despite known sex differences in a spectrum of liver-related outcomes, it is not known how race influences graft outcomes in women. METHODS Using the Scientific Registry of Transplant Recipients, we examined race and ethnicity and graft loss (death or retransplant) in women transplanted from 2002 to 2012. Covariates included recipient and donor characteristics, socioeconomics, and medical comorbidities. RESULTS The eligible cohort (n = 15,860) included 11,051 Caucasians, 2171 Hispanics, 1876 African Americans (AAs), and 762 Asian women with median follow-up of 3.1 years. Five-year graft survival was lower in AA women (60%) compared with Caucasians (71%), Hispanics (70%), and Asians (73%) (P < .001). Graft loss was 45% higher among AA women <40 years at transplant compared with AA women aged 50 to 59 (hazard ratio 1.45, 95% confidence interval 1.17-1.81) and aged 60 to 69 years (hazard ratio 1.33, 95% confidence interval 1.03-1.71), and risk increased after age 60 among Caucasians (P < .001 for race-age interactions). Increased graft loss among young AA women was limited to the first 2 years post-LT (P = .002). CONCLUSION Younger AA women are at particularly high risk for graft loss, which predominates in the first 2 years post-LT. Prospective studies of immunosuppression adherence and pharmacokinetics, particularly in relation to patient age, may help to explain the mechanisms underlying the higher rates of graft loss in younger AA women.
Collapse
Affiliation(s)
- S Dave
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - J L Dodge
- Department of Surgery, Division of Transplant, University of California-San Francisco, San Francisco, California, USA
| | - N A Terrault
- Division of Gastroenterology and Hepatology, University of California-San Francisco, San Francisco, California, USA; Department of Surgery, Division of Transplant, University of California-San Francisco, San Francisco, California, USA
| | - M Sarkar
- Division of Gastroenterology and Hepatology, University of California-San Francisco, San Francisco, California, USA.
| |
Collapse
|
49
|
Dafoe DC, Tantisattamo E, Reddy U. Precision Medicine and Personalized Approach to Renal Transplantation. Semin Nephrol 2018; 38:346-354. [DOI: 10.1016/j.semnephrol.2018.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
|
50
|
Campagne O, Mager DE, Brazeau D, Venuto RC, Tornatore KM. Tacrolimus Population Pharmacokinetics and Multiple CYP3A5 Genotypes in Black and White Renal Transplant Recipients. J Clin Pharmacol 2018; 58:1184-1195. [PMID: 29775201 DOI: 10.1002/jcph.1118] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2017] [Accepted: 02/13/2018] [Indexed: 01/08/2023]
Abstract
Tacrolimus exhibits inter-patient pharmacokinetic variability attributed to CYP3A5 isoenzymes and the efflux transporter, P-glycoprotein. Most black renal transplant recipients require higher tacrolimus doses compared to whites to achieve similar troughs when race-adjusted recommendations are used. An established guideline provides tacrolimus genotype dosing recommendations based on CYP3A5*1(W/T) and loss of protein function variants: CYP3A5*3 (rs776746), CYP3A5*6 (rs10264272), CYP3A5*7 (rs41303343) and may provide more comprehensive race-adjusted dosing recommendations. Our objective was to develop a tacrolimus population pharmacokinetic model evaluating demographic, clinical, and genomic factors in stable black and white renal transplant recipients. A secondary objective investigated race-based tacrolimus regimens and genotype-specific dosing. Sixty-seven recipients receiving oral tacrolimus and mycophenolic acid ≥6 months completed a 12-hour pharmacokinetic study. CYP3A5*3,*6,*7 and ABCB1 1236C>T, 2677G>T/A, 3435C>T polymorphisms were characterized. Patients were classified as extensive, intermediate, and poor metabolizers using a novel CYP3A5*3*6*7 metabolic composite. Modeling and simulation was performed with computer software (NONMEM 7.3, ICON Development Solutions; Ellicott City, Maryland). A 2-compartment model with first-order elimination and absorption with lag time best described the data. The CYP3A5*3*6*7 metabolic composite was significantly associated with tacrolimus clearance (P value < .05), which was faster in extensive (mean: 45.0 L/hr) and intermediate (29.5 L/hr) metabolizers than poor metabolizers (19.8 L/hr). Simulations support CYP3A5*3*6*7 genotype-based tacrolimus dosing to enhance general race-adjusted regimens, with dose increases of 1.5-fold and 2-fold, respectively, in intermediate and extensive metabolizers for comparable exposures to poor metabolizers. This model offers a novel approach to determine tacrolimus dosing adjustments that maintain comparable therapeutic exposure between black and white recipients with different CYP3A5 genotypes.
Collapse
Affiliation(s)
- Olivia Campagne
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA.,Faculty of Pharmacy, Universités Paris Descartes-Paris Diderot, Paris, France
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Daniel Brazeau
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, USA
| | - Rocco C Venuto
- Erie County Medical Center, Division of Nephrology, Department of Medicine, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Kathleen M Tornatore
- Erie County Medical Center, Division of Nephrology, Department of Medicine, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Immunosuppressive Pharmacology Research Program, Translational Pharmacology Research Core, NYS Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|