1
|
Igarashi S, Shimaya K, Fujimaki K, Ko M, Maeda T, Hanabata N, Kanazawa K, Numao H, Munakata M, Sakuraba H. Relapsed refractory immune checkpoint inhibitor-induced colitis treated with tacrolimus and maintained with vedolizumab. Clin J Gastroenterol 2025; 18:463-469. [PMID: 40279047 DOI: 10.1007/s12328-025-02126-x] [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] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
A 58-year-old man was administered anti-programmed death ligand 1 and cytotoxic T-lymphocyte-associated protein 4 antibodies for primary lung cancer with brain metastasis. Subsequent development of diarrhea was diagnosed as immune-related adverse event (irAE) colitis. Initially, his condition improved with prednisolone (1 mg/kg/day). Therefore, the dose was reduced to 5 mg, and his condition was maintained with 5-aminosalicylic acid. However, diarrhea recurred after re-administration of another immune checkpoint inhibitor. Prednisolone (0.5 mg/kg/day) was ineffective for the first relapse. After two doses of infliximab, irAE colitis improved. One year later, diarrhea recurred after steroid tapering. Treatment comprising prednisolone (1 mg/kg/day), two doses of infliximab, granulocyte-monocyte apheresis, and ustekinumab for the second relapse failed to improve his condition. Tacrolimus was initiated 3 months after the second flare occurred, resulting in rapid symptom resolution. Due to frequently relapsed irAE colitis, vedolizumab was administered as maintenance therapy, allowing steroids and tacrolimus to be discontinued and maintaining remission. Tacrolimus may be effective for relapsed refractory irAE colitis, particularly for cases that cannot be successfully treated with infliximab and other treatments. Vedolizumab can be used as maintenance therapy for frequently relapsing irAE colitis.
Collapse
Affiliation(s)
- Shohei Igarashi
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan.
| | - Koji Shimaya
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Kota Fujimaki
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Masayoshi Ko
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Takato Maeda
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Norihiro Hanabata
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Kosuke Kanazawa
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Hiroshi Numao
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Masaki Munakata
- Department of Gastroenterology, Aomori Prefectural Central Hospital, 2-1-1, Higashi-Tsukurimichi, Aomori City, Aomori, Japan
| | - Hirotake Sakuraba
- Department of Gastroenterology, Hematology and Clinical Immunology, Hirosaki University Graduate School of Medicine, Hirosaki city, Aomori, Japan
| |
Collapse
|
2
|
Gomes GF, Batista CRÁ, Silva MCMD, Ferrari CZ, Koerich S, Brant ML, Saliba SW, Perez VG, Lima ALD, Ribeiro RDS, Candelario-Jalil E, Vieira LB, Fiebich BL, de Oliveira ACP. Tacrolimus mitigates pathological patterns in mouse models of Alzheimer's disease. Biomed Pharmacother 2025; 188:118168. [PMID: 40412360 DOI: 10.1016/j.biopha.2025.118168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Neuroinflammation and disruptions in glutamatergic neurotransmission are hallmark features of Alzheimer's disease (AD). Various compounds have been investigated for their potential to modulate these processes in this disease. Tacrolimus (FK506), a calcineurin inhibitor (CNI), has been suggested as a candidate for the treatment of AD, although its effects and possible mechanisms have not been extensively evaluated. Here we investigated whether tacrolimus treatment could mitigate cognitive deficits, neurotoxicity, and microgliosis in AD models, including Aβ1-42-induced intrahippocampal damage and middle-aged transgenic APP/PS1 mice, as well as improve glutamate release dysregulation in synaptosomes from the latter. A single dose of tacrolimus prevented the cognitive impairment induced by intrahippocampal microinjection of Aβ1-42 in the novel object recognition test (NORT), and reduced the neurodegeneration. Interestingly, in the APP/PS1 model, a 30-day treatment with the drug did not prevent memory impairment in the NORT, albeit it improved the social interaction and partially reduced microgliosis. Finally, tacrolimus restored the intrasynaptotosomal calcium levels and normalized impaired glutamate release in synaptosomes from APP/PS1 mice. These findings provide new evidence that both acute and chronic treatment with tacrolimus exerts neuroprotective effects, providing a foundation for the potential therapeutic application of this CNI in managing AD.
Collapse
Affiliation(s)
- Giovanni Freitas Gomes
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carla Ribeiro Álvares Batista
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Carolina Zaniboni Ferrari
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Suélyn Koerich
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Luiza Brant
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Soraya Wilke Saliba
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Victoria García Perez
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna Luiza Diniz Lima
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Roberta Dos Santos Ribeiro
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Luciene Bruno Vieira
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bernd L Fiebich
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
3
|
Bian L, Fu Y, Xu Z, Wu B, Liang J, Guo W, Lin G, Hu F. Bright nanoparticle-labeled lateral flow immunoassay for rapid quantitative detection of tacrolimus in human plasma. Mikrochim Acta 2025; 192:349. [PMID: 40358770 DOI: 10.1007/s00604-025-07200-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
Tacrolimus (Tac), a macrolide immunosuppressant widely employed in organ transplantation, requires therapeutic drug monitoring owing to the narrow treatment window. In this study, we established an innovative ultrabright nanoparticle-based lateral flow immunoassay (LFIA) for one-step rapid quantitative detection of Tac in human plasma. The aggregation-induced emission (AIE) luminogen, AIE490, was embedded in polystyrene nanoparticles to assemble the ultrabright AIE490NP as a highly sensitive fluorescence marker. Carboxyl-modified Tac was used to couple with carrier proteins to synthesize complete antigens coated on the test line. Under optimized conditions, the limit of detection for Tac is 0.589 ng/mL in human plasma. The linear range was 1-1000 ng/mL, completely covering the safe plasma concentration of Tac, 5-15 ng/mL. The intra- and inter-assay precisions of the established method are below 17.99% and 19.94%, respectively. In addition, for measuring the plasma concentration of Tac in 139 clinical samples, the results of the LFIA are consistent with those of LC-MS/MS (R2 = 0.8520, P < 0.0001). Therefore, the established LFIA demonstrates comparable reliability to the LC-MS/MS and holds great potential as a rapid quantitative detection method of Tac in human plasma.
Collapse
Affiliation(s)
- Lun Bian
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Yu Fu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Zihao Xu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Biru Wu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Junyu Liang
- Department of Clinical Laboratory, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wei Guo
- Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Guanfeng Lin
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
- Medical Big Data Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Fang Hu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China.
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
4
|
Zhang Y, Xue L, Hu L, Wang L, Pan H, Lin Y, Ding X, Huang Y, Miao L. Exploring the comprehensive factors influencing tacrolimus pharmacokinetics in early renal transplant recipients: A population pharmacokinetic analysis. Eur J Clin Pharmacol 2025; 81:785-799. [PMID: 40126611 DOI: 10.1007/s00228-025-03825-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025]
Abstract
PURPOSE To establish a population pharmacokinetic (PopPK) model of tacrolimus in the early stages after renal transplantation and evaluate the model's predictive performance with external data. METHODS Intravenous and oral tacrolimus were administered to 302 renal transplant recipients in the early posttransplantation stages. Related data were obtained from the electronic medical records. Single nucleotide polymorphisms in genes associated with tacrolimus pharmacokinetics were tested. The data were analyzed by NONMEM. The external data from 153 patients were subsequently used to evaluate model extrapolation. RESULTS A one-compartment model was used to determine tacrolimus pharmacokinetics. The estimated clearance (CL), volume of distribution (V) and bioavailability (F) of tacrolimus were 4.91 L/h, 77 L and 26.5%, respectively. CL and V decreased with increasing hematocrit. CL and F decreased with increasing operation time. Diltiazem and Wuzhi capsule resulted in 28.4% and 43.9% decreases in the CL, respectively. Omeprazole or esomeprazole resulted in a 9% increase in F. The value of F for patients expressing CYP3A5 was 36.6% lower than that for the patients who did not express CYP3A5. The evaluation of external data revealed that the proportion of individual prediction error within 20% of the observed tacrolimus concentration was greater than 77.3%. CONCLUSIONS A PopPK model for tacrolimus was established for early renal transplantation. CYP3A5 was a significant covariate for F. Fat-free mass was the best predictor of the influence of body size on CL and V. The model could be extrapolated to stable renal transplant recipients.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ling Xue
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Linkun Hu
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liangliang Wang
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Pan
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin Lin
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoliang Ding
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhua Huang
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Liyan Miao
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China.
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Zavrelova A, Zibridova K, Radocha J, Cermakova E, Rozsivalova P, Zak P, Visek B, Lanska M, Stevkova J, Merdita S, Slanar O, Sima M. Tacrolimus exposure during the three-month period following allogeneic stem cell transplantation predicts overall survival. Front Pharmacol 2025; 16:1517083. [PMID: 40351435 PMCID: PMC12061681 DOI: 10.3389/fphar.2025.1517083] [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] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Objectives The objective of this study was to investigate the relationship between both short-term and long-term tacrolimus exposure and overall survival after allogeneic stem cell transplantation and to propose individualized tacrolimus dosing based on the population pharmacokinetic model. Study design Tacrolimus exposure during the first 3 months of therapy after transplantation was calculated using therapeutic drug monitoring data from all patients who underwent allogeneic stem cell transplantation from 2016 to 2018. The optimal upper level was determined using ROC analysis, and the impact of cutoff tacrolimus exposure values on overall survival of patients was assessed together with other transplant variables using multivariate analysis. The population pharmacokinetic model was developed using a nonlinear mixed-effects modeling method, and the optimal tacrolimus dose was proposed. Results A total of 86 patients were included in the outcome analyses. Except for the disease risk category, age ≥55 years, and female-to-male donor, tacrolimus exposures of the area under the curve of trough concentrations (AUCtc) ≥ 222 ng h/mL, ≥258 ng h/mL, and ≥160 ng h/mL during the whole three-month period, second month, and third month of therapy, respectively, were also found to be statistically significant for overall survival in univariate analysis. These AUCtc values were independent variables for overall survival in multivariate analysis, with RR of 3.01 (P = 0.0056), 3.22 (P = 0.0058), and 2.93 (P = 0.0184) for the whole three-month period, second month, and third month of therapy, respectively. The disease risk category (RR 7.11; P < 0.0001), age (RR 2.45; P = 0.0214), and non-myeloablative conditioning (RR 3.39; P = 0.0014) were also significant factors influencing survival in multivariate analysis. Tacrolimus volume of distribution was 127.1 L and was not affected by any of the tested covariates, whereas clearance decreased with age according to the equation CL = 7.94 × e - 0.0085 × age and was reduced by 23% in patients who underwent repeat transplantation. Conclusion Except for the disease risk category, age, and non-myeloablative conditioning, exposure to tacrolimus is an independent predictor of overall survival and should not exceed trough levels of 10.7 ng/mL during the second month and 6.8 ng/mL during third month after transplantation. In order to reach this target, nomogram for estimation of the maximal initial tacrolimus daily dose was developed based on the population pharmacokinetic model.
Collapse
Affiliation(s)
- Alzbeta Zavrelova
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Katerina Zibridova
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Jakub Radocha
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Eva Cermakova
- Department of Medical Biophysics, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Petra Rozsivalova
- Department of Clinical Pharmacy, Hospital Pharmacy, University Hospital Hradec Kralove and Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Pavel Zak
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Benjamin Visek
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Miriam Lanska
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Jana Stevkova
- 4th Department of Internal Medicine – Hematology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Sara Merdita
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Ondrej Slanar
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Martin Sima
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
6
|
Zhong R, Li J, Zhang F, Guo Y. Drug-induced Guillain-Barré Syndrome: a disproportionality analysis based on the US FDA adverse event reporting system. Expert Opin Drug Saf 2025:1-9. [PMID: 40220275 DOI: 10.1080/14740338.2025.2493781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Guillain-Barré Syndrome (GBS) is a rare but severe neurological disorder often precipitated by infections, vaccines, and potentially by certain medications. Drug-induced GBS, though less commonly reported, presents significant diagnostic and therapeutic challenges. This study investigates the correlation between various medications and the onset of GBS. RESEARCH DESIGN AND METHODS We conducted a retrospective pharmacovigilance analysis using data from the FDA Adverse Event Reporting System (FAERS) from Q1 2004 to Q1 2024. The analysis focused on identifying drugs frequently associated with GBS and examining the time-to-onset and severity of these events. RESULTS From over 17 million adverse events, 1,869 cases were identified as drug-induced GBS. Monoclonal antibodies and immunomodulators were the most frequently implicated drug classes. The median time-to-onset for GBS was within the first 30 days following drug exposure. Approximately 51.8% of the cases resulted in severe outcomes, such as hospitalization or disability. Drugs such as brentuximab vedotin and efalizumab showed strong associations with GBS occurrences. CONCLUSIONS This study highlights the importance of monitoring for GBS symptoms following the administration of certain medications, particularly those that affect immune function, and underscores the need for healthcare providers to be aware of the potential neurological risks associated with these medications.
Collapse
Affiliation(s)
- Rujia Zhong
- Department of General Practice and International Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiahao Li
- Department of General Practice and International Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Science (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Feng Zhang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Guo
- Department of General Practice and International Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
7
|
Grebenik E, Zaichick S, Caraveo G. Calcineurin-mediated regulation of growth-associated protein 43 is essential for neurite and synapse formation and protects against α-synuclein-induced degeneration. Front Aging Neurosci 2025; 17:1566465. [PMID: 40259946 PMCID: PMC12009912 DOI: 10.3389/fnagi.2025.1566465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Introduction Elevated calcium (Ca2+) levels and hyperactivation of the Ca2+-dependent phosphatase calcineurin are key factors in α-synuclein (α-syn) pathobiology in Dementia with Lewy Bodies and Parkinson's Disease (PD). Calcineurin activity can be inhibited by FK506, an FDA-approved compound. Our previous work demonstrated that sub-saturating doses of FK506 provide neuroprotection against α-syn pathology in a rat model of α-syn neurodegeneration, an effect associated with the phosphorylation of growth-associated protein 43 (GAP-43). Methods To investigate the role of GAP-43 phosphorylation, we generated phosphomutants at the calcineurin-sensitive sites and expressed them in PC12 cells and primary rat cortical neuronal cultures to assess their effects on neurite morphology and synapse formation. Additionally, we performed immunoprecipitation mass spectrometry in HeLa cells to identify binding partners of these phosphorylation sites. Finally, we evaluated the ability of these phosphomutants to modulate α-syn toxicity. Results In this study, we demonstrate that calcineurin-regulated phosphorylation at S86 and T172 of GAP-43 is a crucial determinant of neurite branching and synapse formation. A phosphomimetic GAP-43 mutant at these sites enhances both processes and provides protection against α-syn-induced neurodegeneration. Conversely, the phosphoablative mutant prevents neurite branching and synapse formation while exhibiting increased interactions with ribosomal proteins. Discussion Our findings reveal a novel mechanism by which GAP-43 activity is regulated through phosphorylation at calcineurin-sensitive sites. These findings suggest that FK506's neuroprotective effects may be partially mediated through GAP-43 phosphorylation, providing a potential target for therapeutic intervention in synucleinopathies.
Collapse
Affiliation(s)
| | | | - Gabriela Caraveo
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
8
|
Coppinger C, Anderson PL. Considerations for drug-drug interactions between long-acting antiretrovirals and immunosuppressants for solid organ transplantation. Expert Opin Drug Metab Toxicol 2025; 21:343-346. [PMID: 39757464 DOI: 10.1080/17425255.2024.2448970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Affiliation(s)
- Corwin Coppinger
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
9
|
Keshavarzi M, Naraki K, Razavi BM, Hosseinzadeh H. Ameliorative and protective effects of coenzyme Q10 against natural and chemical toxicity: a narrative review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03992-5. [PMID: 40080152 DOI: 10.1007/s00210-025-03992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025]
Abstract
Coenzyme Q10 (CoQ10) or ubiquinone is the most known dietary and nutritional supplementation, which has various functions in the body such as involvement in adenosine triphosphate production, modulation of gene expression, antioxidant, and anti-inflammatory effects. It has been indicated that it is useful against cardiotoxicity, hepatotoxicity, neurotoxicity, nephrotoxicity, and so on, which are induced by various toxicants. In this review, we selected articles that include the protective effects of CoQ10 against the toxicity of various chemical and natural compounds including pharmaceuticals, metals, pesticides, etc. Scientific databases including PubMed/Medline, Science Direct, Scopus, and Google Scholar were searched to find relevant in vitro and in vivo studies. The underlying protective mechanisms for CoQ10 against natural and chemical compound toxicity included the enhancement of antioxidant enzyme activities such as superoxide dismutase, catalase, glutathione peroxidase, glutathione-S-transferase, and suppression of pro-inflammatory markers such as tumor necrosis factor-alpha, interleukin-1, and IL-6. Furthermore, it has anti-apoptotic potential by regulating the B-cell lymphoma, Bcl-2-associated X protein, and caspase3/9. Overall, these properties make CoQ10 a highly fascinating compound that may contribute to different aspects of health.
Collapse
Affiliation(s)
- Majid Keshavarzi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Karim Naraki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Yokota H, Akamine Y, Hatakeyama H, Kagaya H, Sakamoto S, Saito M, Takeda M, Sato K, Nakayama K, Kikuchi M. Effect of isavuconazole on the concentration of tacrolimus in a patient with genotype CYP3A5*1/*3: a case report. J Pharm Health Care Sci 2025; 11:20. [PMID: 40083032 PMCID: PMC11905633 DOI: 10.1186/s40780-025-00427-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Azole antifungals are the standard treatment for pulmonary mycosis, which may develop during long-term immunotherapy for kidney transplant. Isavuconazole (ISCZ) is a cytochrome P450 (CYP) 3 A inhibitor that has a risk of interacting with the immunosuppressive drug tacrolimus (TAC). We report a case of simple pulmonary aspergilloma with renal dysfunction due to increased trough levels of TAC after ISCZ coadministration. CASE PRESENTATION A male in his 60s was treated with TAC 3.0 mg/day orally to prevent graft rejection after kidney transplantation. He received a loading dose of ISCZ 600 mg/day orally for two days, followed by a maintenance dose of 200 mg/day for simple pulmonary aspergilloma. The TAC trough concentration increased markedly from 2.4 to 9.9 ng/mL on day 6 after coadministration. The creatinine level increased from 0.70 to 1.08 mg/dL, suggesting renal dysfunction due to TAC. Subsequently, the TAC dosage was reduced, leading to a decreased blood TAC concentration and improved renal function. The patient's genotype was CYP3A5*1/*3. CONCLUSIONS In the early stages of ISCZ treatment, the blood TAC concentration is higher, and CYP3A5 polymorphisms may partially explain the extent of this interaction. We recommend more careful monitoring of TAC and serum creatinine levels for approximately one week after ISCZ administration.
Collapse
Affiliation(s)
- Hayato Yokota
- Department of Pharmacy, Akita University Hospital, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Yumiko Akamine
- Department of Pharmacy, Akita University Hospital, 1-1-1 Hondo, Akita, 010-8543, Japan.
| | - Harumi Hatakeyama
- Department of Pharmacy, Akita University Hospital, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Hideaki Kagaya
- Department of Pharmacy, Akita University Hospital, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Sho Sakamoto
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsuru Saito
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Masahide Takeda
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhiro Sato
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Katsutoshi Nakayama
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Masafumi Kikuchi
- Department of Pharmacy, Akita University Hospital, 1-1-1 Hondo, Akita, 010-8543, Japan
| |
Collapse
|
11
|
Xu J, Guo G, Zhou S, Wang H, Chen Y, Lin R, Huang P, Lin C. Physiologically-based pharmacokinetic modeling to predict the exposure and provide dosage regimens of tacrolimus in pregnant women with infection disease. Eur J Pharm Sci 2025; 206:107003. [PMID: 39788164 DOI: 10.1016/j.ejps.2025.107003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Tacrolimus is extensively used for the prevention of graft rejection following solid organ transplantation in pregnant women. However, knowledge gaps in the dosage of tacrolimus for pregnant patients with different CYP3A5 genotypes and infection conditions have been identified. This study aimed to develop a pregnant physiologically based pharmacokinetic (PBPK) model to characterize the maternal and fetal pharmacokinetics of tacrolimus during pregnancy and explore and provide dosage adjustments. We developed PBPK models for nonpregnant patients and validated them via data from previous clinical studies using PK-Sim and Mobi software. To extrapolate to pregnancy, we considered anatomical, physiological, and metabolic alterations and simulated tacrolimus by adding six groups of IL-6 concentrations (0, 5, 25, 50, 500, and 5000 pg/mL). Models were verified by assessing goodness-of-fit plots and ratios of predicted-to-observed pharmacokinetic parameters. The developed PBPK models adequately describe the available clinical data; the fold errors of the predicted and observed values of the area under the curve and peak plasma concentration were between 0.59 and 1.64, and the average folding error and the absolute average folding error values for all concentration-time data points were 1.15 and 1.36, respectively. The simulation results indicated that the area under the steady-state concentration‒time curve and trough concentrations decreased from the first to the third trimester of pregnancy. The trough concentrations were not within the therapeutic range (4-11 ng/mL) in pregnant patients with the CYP3A5 genotype for most of the infection conditions and exceeded its effective concentration in all the CYP3A5 nonexpressers. Based on the model-derived dosing regimen, the tacrolimus trough concentration in pregnant patients with different CYP3A5 genotypes could fall into the therapeutic window, which provided a clinical practice reference for dosage adjustments during pregnancy.
Collapse
Affiliation(s)
- Jianwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Guimu Guo
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shuifang Zhou
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Han Wang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yuewen Chen
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Rongfang Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Pinfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Cuihong Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
12
|
Momper JD, Venkataramanan R, Jantz AS, Cibrik DM, Birdwell K, Nguyen T, Masters BM, Patel SJ. Evaluation of Effective Half-Life and Its Impact on Time to Steady State for Oral MeltDose Tacrolimus (LCPT) in De Novo Kidney Transplant Recipients. Ther Drug Monit 2025; 47:169-173. [PMID: 39446891 PMCID: PMC11702898 DOI: 10.1097/ftd.0000000000001270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
BACKGROUND For extended-release drug formulations, effective half-life (t 1/2eff ) is a relevant pharmacokinetic parameter to inform dosing strategies and time to reach steady state. Tacrolimus, an immunosuppressant commonly used for the prophylaxis of organ rejection in transplant patients, is available as both immediate- and extended-release formulations. To the best of our knowledge, the t 1/2eff of tacrolimus from these different formulations has not yet been assessed. The objective of this study was to characterize the t 1/2eff and terminal half-life (t 1/2z ) of an extended-release once-daily tacrolimus formulation (LCPT) and twice-daily immediate-release tacrolimus (IR-Tac). METHODS A noncompartmental analysis of pharmacokinetic data obtained from a phase 2 study in de novo kidney transplant recipients receiving either LCPT or IR-Tac was conducted. Intensive blood sampling was performed on days 1, 7, and 14, and tacrolimus whole blood concentrations were measured using a validated liquid chromatography with tandem mass spectrometry method. T 1/2eff was estimated using within-participant accumulation ratios. T 1/2z was estimated by linear regression of the terminal phase of the concentration versus time profile. RESULTS The median accumulation ratios of LCPT and IR-Tac on day 14 were 3.18 and 2.06, respectively.The median (interquartile range; IQR) t 1/2eff for LCPT at day 14 of dosing was 48.4 (37.4-77.9) hours, whereas the t 1/2z was 20.3 (17.6-22.9) hours. For IR-Tac, the median (IQR) t 1/2eff and t 1/2z on day 14 were 12.5 (8.8-23.0) hours and 12.2 (9.2-15.7) hours, respectively. CONCLUSIONS Consistent with its prolonged release of tacrolimus, LCPT demonstrated a higher accumulation ratio and a longer t 1/2eff compared with IR-Tac. These findings underscore the pharmacokinetic differences between different drug formulations of the same moiety and may help inform dose adjustments for LCPT in kidney transplantation.
Collapse
Affiliation(s)
- Jeremiah D. Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California;
| | - Raman Venkataramanan
- School of Pharmacy and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania;
| | - Arin S. Jantz
- Department of Pharmacy, Henry Ford Health, Detroit, Michigan;
| | - Diane M. Cibrik
- Department of Medicine, University of Kansas Health System, Kansas City, Kansas;
| | - Kelly Birdwell
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Tk Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California;
| | - Brian M. Masters
- Medical Affairs, Veloxis Pharmaceuticals, Inc., Cary, North Carolina.
| | - Samir J. Patel
- Medical Affairs, Veloxis Pharmaceuticals, Inc., Cary, North Carolina.
| |
Collapse
|
13
|
Pattanaik S, Monchaud C. Pharmacokinetic Boosting of Calcineurin Inhibitors in Transplantation: Pros, Cons, and Perspectives. Ther Drug Monit 2025; 47:118-140. [PMID: 39774591 DOI: 10.1097/ftd.0000000000001288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 09/27/2024] [Indexed: 01/11/2025]
Abstract
ABSTRACT The concept of pharmacokinetic (PK) boosting of calcineurin inhibitors (CNI) emerged after the FDA approval of cyclosporine-A. Several studies followed, and the proof of concept was well established by the late 1990s. This also continued for the next blockbuster immunosuppressant, tacrolimus. The driver for such research was an endeavor to save costs, as both drugs were expensive due to patent protection. Two CYP inhibitors, ketoconazole and diltiazem, have been extensively studied in this context and continue to be prescribed off-label along with the CNI. It has been observed that using ketoconazole reduces the dose requirement of tacrolimus by about 50% and 30% with diltiazem, which is in conformity with their pharmacological actions. Off-label co-prescription of these drugs with CNI is often encountered in low and middle-income countries. The foremost reason cited is economic. This article collates the evidence from the clinical studies that evaluate the PK-boosting effects of CNI and also reviews the gaps in the current evidence base. The current knowledge prevents the transplant community from making meaningful inferences about the risks and benefits of such strategies. Although the PK-boosting strategy can lead to serious adverse events, emerging evidence suggests that it may be advantageous for individuals with high CNI dose requirements. Hence, PK boosting may be an unmet need in the therapeutics of CNI. Nevertheless, there are several unanswered questions surrounding such use, and therefore, this merits testing in well-designed clinical studies. Moreover, drugs with better safer profiles and a history of successful PK boosting may be considered for evaluation with CNI.
Collapse
Affiliation(s)
- Smita Pattanaik
- Clinical Pharmacology Unit, Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Caroline Monchaud
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Limoges, Limoges, France
- INSERM UMR-1248 Pharmacologie et Transplantation, Université Limoges, Limoges, France; and
- FHU SUPORT, Limoges, France
| |
Collapse
|
14
|
Sun ZH, Zhao YC, Li JK, Peng F, Yu F, Zhang BK, Yan M. Population dynamics analysis of the interaction between tacrolimus and voriconazole in renal transplant recipients. Front Pharmacol 2025; 15:1502097. [PMID: 39944402 PMCID: PMC11813913 DOI: 10.3389/fphar.2024.1502097] [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] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/30/2024] [Indexed: 04/10/2025] Open
Abstract
Background The concurrent administration of tacrolimus and voriconazole in kidney transplant recipients can lead to drug interactions, potentially resulting in severe adverse reactions. This study aimed to establish a robust population pharmacokinetic model to explore the interaction between tacrolimus and voriconazole in greater depth. Methods Tacrolimus blood samples and laboratory data were prospectively collected from eligible patients enrolled between April 2023 and April 2024, following predefined inclusion and exclusion criteria. Using Phoenix (version 8.1), a pharmacokinetic prediction model was developed. Model performance was assessed using model fitting plots, bootstrap analysis, and visual predictive checks (VPC). Results This study ultimately included 51 eligible patients, with a total of 281 blood samples collected. Analysis revealed a significant negative correlation between voriconazole concentration (Cvrc) and tacrolimus volume of clearance rate (CL), a significant positive correlation between platelets (PLT) and tacrolimus clearance (CL), and a significant negative correlation between blood cells (RBC) and tacrolimus clearance (CL). Conclusion This study successfully established a population pharmacokinetic model for renal transplant patients concurrently receiving tacrolimus and voriconazole. The model demonstrated good predictive performance and offers valuable insights to clinicians for optimizing tacrolimus dosing in this patient population.
Collapse
Affiliation(s)
- Zhi-Hua Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yi-Chang Zhao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Jia-Kai Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Fenghua Peng
- Department of Urological Organ Transplantation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Bi-Kui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| |
Collapse
|
15
|
Yo JH, Palmer KR, Nikolic-Paterson D, Kerr PG, Marshall SA. Immunosuppressant drug tacrolimus inhibits HUVEC angiogenesis and production of placental growth factor. Placenta 2025; 159:146-153. [PMID: 39724756 DOI: 10.1016/j.placenta.2024.12.010] [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] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Tacrolimus is a cornerstone of immunosuppression in solid organ transplants, but its use is linked with the development of endothelial dysfunction. Pregnant solid organ transplant recipients are four to six times more likely to develop preeclampsia, which is also associated with endothelial dysfunction. Therefore, this in vitro study investigated the acute effects of tacrolimus on the expression of common angiogenic factors related to preeclampsia, and effects on angiogeneis in primary human tissues. METHODS Primary human umbilical vein endothelial cells (HUVECs) were exposed to tacrolimus (0, 5, 20, 50 ng/mL) for 24h alone, or in combination with tumour necrosis factor (TNF, 10 ng/mL) and high dose glucose (25 mM). Cell culture concentrations of sFlt-1, PlGF and activin A were measured. In addition, the effect of tacrolimus on markers of endothelial dysfunction and permeability were assessed, as were the effect of tacrolimus on tube formation. Angiogenic factors and mRNA markers of oxidative stress and inflammation were also assessed in primary placental tissue after an acute 24 h exposure to tacrolimus. RESULTS Tacrolimus exposure significantly reduced HUVEC secretion of PlGF, increased production of activin A, andreduced tubular structure formation without impacting cell permeability or viability. There was no change in ICAM1 or VCAM1 expression in HUVECs treated with tacrolimus treatment alone, however co-culture with TNF significantly increased expression of ICAM1 and VCAM1. In placental explants tacrolimus did not change angiogenic factor production or markers of inflammation or oxidative stress. CONCLUSION An acute tacrolimus exposure reduced PlGF secretion and impaired angiogenesis in primary endothelial cells, without affecting. These findings provide a potential mechanistic basis for tacrolimus to contribute to the endothelial dysfunction contributing to preeclampsia.
Collapse
Affiliation(s)
- Jennifer H Yo
- Department of Nephrology, Monash Health, Clayton, VIC, Australia; The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, VIC, Australia; Department of Medicine, School of Clinical Health Sciences, Monash University, Clayton, VIC, Australia.
| | - Kirsten R Palmer
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, VIC, Australia; Monash Women's, Monash Health, Clayton, VIC, Australia
| | | | - Peter G Kerr
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
| | - Sarah A Marshall
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
16
|
Gao Y, Ma J. The impact of CYP3A5, NR1I2, and POR polymorphisms on tacrolimus dose-adjusted concentration and clinical outcomes in adult allogeneic haematopoietic stem cell transplantation. Xenobiotica 2025; 55:16-24. [PMID: 39754510 DOI: 10.1080/00498254.2024.2448967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Polymorphisms in genes related to drug-metabolising enzymes may affect tacrolimus exposure. This study aimed to assess the influence of CYP3A5, NR1I2, and POR polymorphisms on tacrolimus pharmacokinetics and outcomes in allogeneic haematopoietic stem cell transplantation (HSCT).Forty-six adult patients receiving oral tacrolimus at an initial dose of 0.03 mg/kg/day for acute graft versus host disease (GVHD) prophylaxis after allogeneic HSCT were enrolled in this retrospective study. Genetic polymorphisms were detected in relation to concentration/dose (C/D) ratios of tacrolimus and the incidence of acute GVHD and acute kidney injury (AKI).The CYP3A5 *3/*3 genotype and co-administration of voriconazole were significantly associated with increased C/D ratios of tacrolimus (p < 0.05). NR1I2 8055CC presents a significantly higher tacrolimus C/D ratio compared with carriers of 8055CT and 8055TT genotypes in allogeneic HSCT recipients with the CYP3A5*1 allele (p = 0.033). Younger age and recipients with the CYP3A5*1 allele were significantly associated with higher incidence of II-IV acute GVHD post-transplantation.CYP3A5*3, NR1I2 8055C > T, and concomitant use of voriconazole are important determinants affecting tacrolimus pharmacokinetics. Moreover, CYP3A5*1 allele and younger age are independent risk factors for II-IV acute GVHD in HSCT recipients.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingjing Ma
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Ben Brahim B, Arenas Hoyos I, Zhang L, Vögelin E, Olariu R, Rieben R. Tacrolimus-loaded Drug Delivery Systems in Vascularized Composite Allotransplantation: Lessons and Opportunities for Local Immunosuppression. Transplantation 2025; 109:142-152. [PMID: 38773862 PMCID: PMC11627328 DOI: 10.1097/tp.0000000000005049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/26/2024] [Accepted: 03/19/2024] [Indexed: 05/24/2024]
Abstract
Long-term systemic immunosuppression is needed for vascularized composite allotransplantation (VCA). The high rate of acute rejection episodes in the first posttransplant year, the development of chronic rejection, and the adverse effects that come along with this treatment, currently prevent a wider clinical application of VCA. Opportunistic infections and metabolic disturbances are among the most observed side effects in VCA recipients. To overcome these challenges, local immunosuppression using biomaterial-based drug delivery systems (DDS) have been developed. The aim of these systems is to provide high local concentrations of immunosuppressive drugs while reducing their systemic load. This review provides a summary of recently investigated local DDS with different mechanisms of action such as on-demand, ultrasound-sensitive, or continuous drug delivery. In preclinical models, ranging from rodent to porcine and nonhuman primate models, this approach has been shown to reduce systemic tacrolimus (TAC) load and adverse effects, while prolonging graft survival. Localized immunosuppression using biomaterial-based DDS represents an encouraging approach to enhance graft survival and reduce toxic side effects of immunosuppressive drugs in VCA patients. Preclinical models using TAC-releasing DDS have demonstrated high local immunosuppressive effects with a low systemic burden. However, to reduce acute rejection events in translational animal models or in the clinical reality, the use of additional low-dose systemic TAC treatment may be envisaged. Patients may benefit through efficient graft immunosuppression and survival with negligible systemic adverse effects, resulting in better compliance and quality of life.
Collapse
Affiliation(s)
- Bilal Ben Brahim
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Isabel Arenas Hoyos
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Plastic and Hand Surgery, Inselspital Bern University Hospital, Bern, Switzerland
| | - Lei Zhang
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Plastic and Hand Surgery, Inselspital Bern University Hospital, Bern, Switzerland
| | - Esther Vögelin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Plastic and Hand Surgery, Inselspital Bern University Hospital, Bern, Switzerland
| | - Radu Olariu
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Plastic and Hand Surgery, Inselspital Bern University Hospital, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [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] [Scholar Register] [Received: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
19
|
Tolou-Ghamari Z. Tacrolimus, Cytochrome P450, Interactions with Food Variables in Organ Transplant Recipients; A Current and Comprehensive Review. Curr Drug Metab 2025; 25:547-553. [PMID: 39757635 DOI: 10.2174/0113892002328742241210102522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 01/07/2025]
Abstract
The well-established calcineurin inhibitor, tacrolimus, as an immunosuppressive agent, is widely prescribed after organ transplantation. Cytochrome P450 (CYP 450) isoforms are responsible for the metabolism of many features associated with food parameters like phytochemicals, juices, and fruits. This review article summarizes the findings of previous studies to help predict the efficacy or side effects of tacrolimus in the presence of food variables. From the commencement of databases associated with the topic of interest to 26 October 2024, all relevant articles were searched through PubMed, Scopus, and Web of Science. The suggested therapeutic range for tacrolimus trough concentration (C0 ) was reported as 5-15 ng/ml blood. Tacrolimus interaction with food variables could significantly change C0 after organ transplantation. For example, grapefruit juice could increase tacrolimus C0 due to CYP enzyme inhibition. Toxicity such as nephrotoxicity could result from turmeric and other herbal or food products. By inhibiting tacrolimus-metabolizing enzymes and transporters, a high intake of vegetables could increase the risk of adverse effects. Secondary metabolites of vegetables could lead to toxicity in patients with tacrolimus. Furthermore, grapefruit juice, citrus fruits, turmeric, and pomegranate juice could change clinical pharmacokinetics parameters such as Tmax, Cmax, AUC, and C0 of tacrolimus after organ transplantation. Bioavailability of tacrolimus might be decreased by induction of the CYP450 system and P-gp efflux pump due to cranberry, rooibos tea, and boldo. Increased inhibitory effect on CYP450 system and/or P-gp efflux pump by grapefruit juice, schisandra, berberine, turmeric, pomegranate juice, pomelo, and ginger could increase bioavailability of tacrolimus. A vigilant immunosuppressive strategy accompanied by scheduled therapeutic drug monitoring is recommended before and after transplant surgery.
Collapse
Affiliation(s)
- Zahra Tolou-Ghamari
- Deputy of Research and Technology, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
20
|
Dash K, Mishra M. The tradeoff between the efficacy of calcineurin inhibitors: prevention of allograft rejection vs. post-transplant renal and cardiovascular complications. Crit Rev Toxicol 2025; 55:63-79. [PMID: 39807635 DOI: 10.1080/10408444.2024.2433631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Solid organ transplantation has emerged as a crucial intervention in the field of medicine. During transplantation, our human body perceives the organ as an exogenous entity or graft, initiating an immune reaction to eliminate it. This immune response ultimately culminates in the rejection of the graft. So, to mitigate the possibility of graft rejection, implementing immune suppression is imperative. In this context, the utilization of calcineurin inhibitors (CNIs) assumes a pivotal role. Calcineurin inhibitors significantly preserve immunosuppression following solid organ transplantation. Calcineurin inhibitors have considerably improved short-term results in renal transplantation by reducing acute rejection rates. Concerning the limited therapeutic window of these medications, careful monitoring of pharmacological treatment and individual doses is required. However, a significant number of patients do experience CNI toxicity. Side effects of CNIs include renal failure, hypertension, respiratory disorders, gastrointestinal damage, gingivitis, and so on. Higher trough level of the drug causes acute nephrotoxicity, which is of three types: functional toxicity, tubular toxicity, and vascular toxicity. Acute nephrotoxicity, if untreated, leads to irreversible, progressive deterioration of allograft function, leading to chronic nephrotoxicity. Cardiovascular toxicity of CNIs includes atrial hypertension caused by vasoconstriction of the afferent arteriole, vascular remodeling, hypertrophy, dyslipidemia, and also the onset of diabetes. Such clinical complications further affect the patient's survivability and subjective well-being, possibly leading to graft loss. This review focuses on the most severe side effects of CNIs: renal and cardiovascular toxicity.
Collapse
Affiliation(s)
- Kalpanarani Dash
- Department of Life Sciences, Neural Developmental Biology Lab, National Institute of Technology, Rourkela, India
| | - Monalisa Mishra
- Department of Life Sciences, Neural Developmental Biology Lab, National Institute of Technology, Rourkela, India
| |
Collapse
|
21
|
Han L, Cui Y, Pan Y, Chen R, Jiao Z. External evaluation of tacrolimus population pharmacokinetic models in adult lung transplant patients: How to enhance the predictive ability of the model? Int Immunopharmacol 2024; 143:113225. [PMID: 39353393 DOI: 10.1016/j.intimp.2024.113225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Tacrolimus is the cornerstone of current immunosuppressive strategies after lung transplantation. However, its narrow therapeutic range and considerable pharmacokinetic variability pose challenges for individualized treatment. Several tacrolimus population pharmacokinetic (popPK) models have been developed for precision dosing in adult lung transplant patients. However, their applicability across different clinical settings remains uncertain. The aim of this study was to evaluate the external predictability of these models and identify influential factors. METHODS Published models were systematically retrieved and assessed based on an external dataset of 39 patients (1240 tacrolimus trough concentrations) using three approaches: (1) prediction-based diagnosis using dosing records and patient characteristics; (2) simulation-based diagnosis, with prediction- and variability-corrected visual predictive checks (pvcVPC) and normalized prediction distribution error tests (NPDE); and (3) Bayesian forecasting using one to four observations for posterior predictions. We also investigated the impact of model structure and covariates on predictability. RESULTS The predictive performance of six published models was externally evaluated, but none demonstrated satisfactory accuracy in prediction- and simulation-based diagnosis. Bayesian forecasting yielded satisfactory results with only one prior observation and optimal predictive performance with 2-3 priors for all included models. The structural model parameterized on plasma tacrolimus concentration outperformed others. Significant correlations were observed between prediction-error and daily tacrolimus dose, postoperative day, and voriconazole co-administration. CONCLUSIONS The overall predictive performance of all published models was unsatisfactory, making direct extrapolation inappropriate. However, Bayesian forecasting significantly improves predictive performance. Utilizing plasma tacrolimus concentration for parameter estimation can improve the predictive ability of tacrolimus popPK models.
Collapse
Affiliation(s)
- Lu Han
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yifan Cui
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yan Pan
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rui Chen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Bi F, Yuan T, Zhang B, Li J, Lin Y, Yang J. Establishment of Biopredictive Dissolution and Bioequivalence Safe Space Using the Physiologically Based Biopharmaceutics Modeling for Tacrolimus Extended-Release Capsules. AAPS PharmSciTech 2024; 26:13. [PMID: 39690309 DOI: 10.1208/s12249-024-03006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024] Open
Abstract
A slight variation in in vivo exposure for tacrolimus extended-release (ER) capsules, which have a narrow therapeutic index (NTI), significantly affects the pharmacodynamics of the drug. Generic drug bioequivalence (BE) standards are stricter, necessitating accurate assessment of the rate and extent of drug release. Therefore, an in vitro dissolution method with high in vivo predictive power is crucial for developing generic drugs. In this study, physiologically based biopharmaceutics modeling (PBBM) for 5 mg tacrolimus ER capsules was developed and validated. The reference and non-BE test formulations were assessed using the Flow-Through Cell apparatus (USP IV) with biorelevant media to establish a biopredictive dissolution method. Using PBBM, virtual bioequivalence trials with virtual batches were conducted to propose a BE safe space. These criteria can identify formulations that pass the internal quality control test but are likely non-BE. This study highlights the benefits of developing biopredictive dissolution methods that are based on biorelevant dissolution. The PBBM, constructed by integrating various drug parameters, combined with the developed biopredictive dissolution methods, is a convenient approach for BE evaluation of NTI drugs and a practical tool for developing new drugs.
Collapse
Affiliation(s)
- Fulin Bi
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Tong Yuan
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Baohong Zhang
- Logan Instruments (Shanghai) Co; Ltd, Shanghai, 201107, People's Republic of China
| | - Jixia Li
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Lin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jin Yang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
23
|
Aloui G, Charfi R, Daldoul M, Ben Hammamia S, Ben Sassi M, Zouari M, Eljeberi H, Daghfous R, Gaies E, Trabesli S. Optimizing tacrolimus therapeutic drug monitoring in Tunisian kidney transplant recipients: exploring the variability in bioavailability and the correlation between pharmacokinetic parameters. Drug Metab Pers Ther 2024; 39:215-220. [PMID: 39679494 DOI: 10.1515/dmpt-2024-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/18/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVES While the existing literature extensively covers the topic of tacrolimus variability, it remains crucial to gather data that are tailored to the Tunisian population. Our primary goal was to assess the variability in tacrolimus bioavailability using the Cp(0)/weight dosage ratio in Tunisian kidney transplant patients. We also aimed to determine the correlations between blood trough level (Cp(0)) and the area under the concentration-time curve (AUC0-12 h) in this cohort. METHODS This retrospective study included patients treated with oral tacrolimus for the prevention of organ rejection between 2009 and 2023. The correlation between parameters was analyzed through a Pearson coefficient and a regression model. We assessed the inter- and intraindividual variability by calculating the coefficient of variation for patients with at least three samples. RESULTS Analysis of 2,124 samples revealed a weak correlation (R=0.121) between Cp(0) and weight dosage. We found that 79.3 % of patients exhibited high variability in the Cp(0)/weight dosage ratio. A strong correlation (R=0.797) was found between Cp(0) and the AUC0-12 h. We also found that 47.6 % of patients showed high variability in the AUC0-12 h/Cp(0) ratio. CONCLUSIONS This study underscores the necessity for individualized therapeutic drug monitoring in Tunisian kidney transplant recipients due to the high variability in the Cp(0)/weight dosage ratio. The AUC0-12 h/Cp(0) ratio is proposed as a more consistent parameter for therapeutic drug monitoring, offering potential improvements in tacrolimus therapy management.
Collapse
Affiliation(s)
- Ghaith Aloui
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
| | - Rym Charfi
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| | - Mouna Daldoul
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| | - Syrine Ben Hammamia
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| | - Mouna Ben Sassi
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| | - Mohamed Zouari
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
| | - Hanene Eljeberi
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
| | - Riadh Daghfous
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| | - Emna Gaies
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| | - Sameh Trabesli
- Department of Clinical Pharmacology, National Centre Chalbi Belkahia of Pharmacovigilance, Tunis, Tunisia
- Faculty of Medicine of Tunis, EL Manar University, Tunis, Tunisia
| |
Collapse
|
24
|
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] [Scholar 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
|
25
|
Fernández-Alarcón B, Nolberger O, Vidal-Alabró A, Rigo-Bonnin R, Grinyó JM, Melilli E, Montero N, Manonelles A, Coloma A, Favà A, Codina S, Cruzado JM, Colom H, Lloberas N. Guiding the starting dose of the once-daily formulation of tacrolimus in " de novo" adult renal transplant patients: a population approach. Front Pharmacol 2024; 15:1456565. [PMID: 39364055 PMCID: PMC11447946 DOI: 10.3389/fphar.2024.1456565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/23/2024] [Indexed: 10/05/2024] Open
Abstract
Aims The once-daily extended-release tacrolimus formulation (ER-Tac) has demonstrated similar efficacy and safety to the twice-daily immediate-release formulation (IR-Tac), but few population-based pharmacokinetic models have been developed in de novo kidney transplant patients to optimize doses. Therefore, this study aimed i) at developing a population pharmacokinetic model for ER-Tac in de novo adult kidney transplant patients ii) and identifying genetic factors and time-varying covariates predictive of pharmacokinetic variability to guide tacrolimus dosage during the early post-transplant period. Methods A total of 1,067 blood tacrolimus concentrations from 138 kidney transplant patients were analyzed. A total of 29 out of 138 patients were intensively sampled for 24 h on the day 5 post-transplantation; meanwhile, for the remaining patients, concentrations were collected on days 5, 10, and 15 after transplantation. Tacrolimus daily doses and genetic and demographic characteristics were retrieved from the medical files. Biochemistry time-varying covariates were obtained on different days over the pharmacokinetic (PK) study. A simultaneous PK analysis of all concentrations was carried out using the non-linear mixed-effects approach with NONMEM 7.5. Results A two-compartment model with linear elimination and delayed absorption best described the tacrolimus pharmacokinetics. Between-patient variability was associated with oral blood clearance (CL/F) and the central compartment distribution volume (Vc/F). Tacrolimus concentrations standardized to a hematocrit value of 45% significantly improved the model (p < 0.001). This method outperformed the standard covariate modeling of the hematocrit-blood clearance relationship. The effect of the CYP3A5 genotype was statistically (p < 0.001) and clinically significant on CL/F. The CL/F of patients who were CYP3A5*1 carriers was 51% higher than that of CYP3A5*1 non-carriers. Age also influenced CL/F variability (p < 0.001). Specifically, CL/F declined by 0.0562 units per each increased year from the value estimated in patients who were 60 years and younger. Conclusion The 36% between-patient variability in CL/F was explained by CYP3A5 genotype, age, and hematocrit. Hematocrit standardization to 45% explained the variability of tacrolimus whole-blood concentrations, and this was of utmost importance in order to better interpret whole-blood tacrolimus concentrations during therapeutic drug monitoring. The dose requirements of CYP3A5*/1 carriers in patients aged 60 years or younger would be highest, while CYP3A5*/1 non-carriers older than 60 years would require the lowest doses.
Collapse
Affiliation(s)
- Beatriz Fernández-Alarcón
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Oscar Nolberger
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Anna Vidal-Alabró
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Raul Rigo-Bonnin
- Biochemistry Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Josep M. Grinyó
- Medicine Unit, Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Edoardo Melilli
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Nuria Montero
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Anna Manonelles
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Ana Coloma
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Alex Favà
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Sergi Codina
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Josep M. Cruzado
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| | - Helena Colom
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Nuria Lloberas
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, Barcelona, Spain
| |
Collapse
|
26
|
Zhao YC, Sun ZH, Li JK, Liu HY, Zhang BK, Xie XB, Fang CH, Sandaradura I, Peng FH, Yan M. Individualized dosing parameters for tacrolimus in the presence of voriconazole: a real-world PopPK study. Front Pharmacol 2024; 15:1439232. [PMID: 39318775 PMCID: PMC11419969 DOI: 10.3389/fphar.2024.1439232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Objectives Significant increase in tacrolimus exposure was observed during co-administration with voriconazole, and no population pharmacokinetic model exists for tacrolimus in renal transplant recipients receiving voriconazole. To achieve target tacrolimus concentrations, an optimal dosage regimen is required. This study aims to develop individualized dosing parameters through population pharmacokinetic analysis and simulate tacrolimus concentrations under different dosage regimens. Methods We conducted a retrospective study of renal transplant recipients who were hospitalized at the Second Xiangya Hospital of Central South University between January 2016 and March 2021. Subsequently, pharmacokinetic analysis and Monte Carlo simulation were employed for further analysis. Results Nineteen eligible patients receiving tacrolimus and voriconazole co-therapy were included in the study. We collected 167 blood samples and developed a one-compartment model with first-order absorption and elimination to describe the pharmacokinetic properties of tacrolimus. The final typical values for tacrolimus elimination rate constant (Ka), apparent volume of distribution (V/F), and apparent oral clearance (CL/F) were 8.39 h-1, 2690 L, and 42.87 L/h, respectively. Key covariates in the final model included voriconazole concentration and serum creatinine. Patients with higher voriconazole concentration had lower tacrolimus CL/F and V/F. In addition, higher serum creatinine levels were associated with lower tacrolimus CL/F. Conclusion Our findings suggest that clinicians can predict tacrolimus concentration and estimate optimal tacrolimus dosage based on voriconazole concentration and serum creatinine. The effect of voriconazole concentration on tacrolimus concentration was more significant than serum creatinine. These findings may inform clinical decision-making in the management of tacrolimus and voriconazole therapy in solid organ transplant recipients.
Collapse
Affiliation(s)
- Yi-Chang Zhao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Zhi-Hua Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jia-Kai Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Huai-Yuan Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Bi-Kui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Xu-Biao Xie
- Department of Urological Organ Transplantation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chun-Hua Fang
- Department of Urological Organ Transplantation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Indy Sandaradura
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, Sydney, NSW, Australia
| | - Feng-Hua Peng
- Department of Urological Organ Transplantation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| |
Collapse
|
27
|
Mohamed ME, Saqr A, Staley C, Onyeaghala G, Teigen L, Dorr CR, Remmel RP, Guan W, Oetting WS, Matas AJ, Israni AK, Jacobson PA. Pharmacomicrobiomics: Immunosuppressive Drugs and Microbiome Interactions in Transplantation. Transplantation 2024; 108:1895-1910. [PMID: 38361239 PMCID: PMC11327386 DOI: 10.1097/tp.0000000000004926] [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] [Scholar Register] [Indexed: 02/17/2024]
Abstract
The human microbiome is associated with human health and disease. Exogenous compounds, including pharmaceutical products, are also known to be affected by the microbiome, and this discovery has led to the field of pharmacomicobiomics. The microbiome can also alter drug pharmacokinetics and pharmacodynamics, possibly resulting in side effects, toxicities, and unanticipated disease response. Microbiome-mediated effects are referred to as drug-microbiome interactions (DMI). Rapid advances in the field of pharmacomicrobiomics have been driven by the availability of efficient bacterial genome sequencing methods and new computational and bioinformatics tools. The success of fecal microbiota transplantation for recurrent Clostridioides difficile has fueled enthusiasm and research in the field. This review focuses on the pharmacomicrobiome in transplantation. Alterations in the microbiome in transplant recipients are well documented, largely because of prophylactic antibiotic use, and the potential for DMI is high. There is evidence that the gut microbiome may alter the pharmacokinetic disposition of tacrolimus and result in microbiome-specific tacrolimus metabolites. The gut microbiome also impacts the enterohepatic recirculation of mycophenolate, resulting in substantial changes in pharmacokinetic disposition and systemic exposure. The mechanisms of these DMI and the specific bacteria or communities of bacteria are under investigation. There are little or no human DMI data for cyclosporine A, corticosteroids, and sirolimus. The available evidence in transplantation is limited and driven by small studies of heterogeneous designs. Larger clinical studies are needed, but the potential for future clinical application of the pharmacomicrobiome in avoiding poor outcomes is high.
Collapse
Affiliation(s)
- Moataz E Mohamed
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN
| | - Abdelrahman Saqr
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN
| | | | - Guillaume Onyeaghala
- Hennepin Healthcare Research Institute, Minneapolis, MN
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Levi Teigen
- Department of Food Science and Nutrition, University of Minnesota, St Paul, MN
| | - Casey R Dorr
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN
- Hennepin Healthcare Research Institute, Minneapolis, MN
- Department of Medicine, University of Minnesota, Minneapolis, MN
- Department of Medicine, Hennepin Healthcare, Minneapolis, MN
| | - Rory P Remmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - William S Oetting
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN
| | - Arthur J Matas
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Ajay K Israni
- Hennepin Healthcare Research Institute, Minneapolis, MN
- Department of Medicine, Hennepin Healthcare, Minneapolis, MN
- Department of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN
| | - Pamala A Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN
| |
Collapse
|
28
|
Jiang J, Luan J. Effect of CYP3A5 Gene Polymorphisms on Tacrolimus Blood Concentrations and Adverse Events in Allogeneic Hematopoietic Stem Cell Transplant Patients. Transplant Proc 2024; 56:1678-1682. [PMID: 39147616 DOI: 10.1016/j.transproceed.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Tacrolimus is the core basic immunosuppressant after transplantation. Cytochrome P450 3A5 (CYP3A5) is the main enzyme involved in tacrolimus metabolism, and rs776746A>G is the most frequently studied polymorphism in the CYP3A5 gene. The aim of this study was to investigate the effect of CYP3A5 gene polymorphisms on tacrolimus blood concentrations and acute graft versus host disease (GVHD) in patients with allogeneic hematopoietic stem cell transplantation (allo-HSCT). METHODS This study included adult patients who received allo-HSCT at the First Affiliated Hospital of Wannan Medical College from January 2021 to June 2022, and received postoperative treatment with tacrolimus. Tacrolimus blood levels were obtained by fully automatic chemiluminescence immunoassay analyzer. Polymerase chain reaction/restriction fragment length polymorphism was used to genotype for CYP3A5*3 allelic variants. RESULTS In a total of 50 transplant patients, 30 patients were detected with CYP3A5*3/*3 genotype, 15 patients with CYP3A5*1/*3 genotype, and 5 patients with CYP3A5*1/*1 genotype. The initial tacrolimus blood concentrations in allo-HSCT patients with CYP3A5*1/*1, *1/*3, and *3/*3 genes were 7.75, 8.61, and 10.19 ng/mL, respectively; The initial blood concentration/dose (C/D) ratios were 4.08, 4.42 and 5.66 ng/(mL·mg), respectively. The C/D ratios of allo-HSCT patients carrying CYP3A5*1/*1, *1/*3, and *3/*3 genes were 4.35 and 4.71 and 5.58, 4.19, 4.56 and 5.71 ng/(mL·mg) in the second and 3rd weeks after operation. These results showed that the blood concentration and C/D ratio of tacrolimus in patients with CYP3A5*3/*3 genotype were significantly higher than those in patients with CYP3A5*1/*3 or CYP3A5*1/*1 genotype. Moreover, the incidence of acute GVHD after allo-HSCT in patients with CYP3A5*1/*1 genotype was significantly higher than that in patients with CYP3A5*1/*3 or CYP3A5*3/*3 genotype. CONCLUSIONS Most patients carry the mutant allele CYP3A5*3. CYP3A5 gene polymorphisms affect tacrolimus blood concentrations and acute GVHD after allo-HSCT.
Collapse
Affiliation(s)
- Jia Jiang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China.
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| |
Collapse
|
29
|
Yang S, Wei J, Pan X, Li Z, Zhang X, Li Z, Dong X, Hua Z, Li X. Development and validation of individualized tacrolimus dosing software for Chinese pediatric liver transplantation patients: a population pharmacokinetic approach. Eur J Clin Pharmacol 2024; 80:1409-1420. [PMID: 38904798 DOI: 10.1007/s00228-024-03717-2] [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] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE We aim to describe the population pharmacokinetics (PPK) of tacrolimus in Chinese pediatric patients under 4 years old after liver transplantation and to develop individualized tacrolimus dosing software. METHODS A total of 663 blood concentrations from 85 patients aged 4.57 months to 3.97 years were collected in this study. PPK analysis was performed using a nonlinear mixed effects modeling approach with the software, Phoenix. Using C#, an individualized tacrolimus dosing software was created. The software was then used to predict the concentrations of another ten pediatric liver transplantation patients to verify the accuracy of said software. The predictive error (PE) and the absolute predictive error (APE) for each predicted time point were computed. RESULTS A one-compartment model with first-order elimination best fitted the data. The apparent volume of distribution (V/F) and apparent clearance (CL/F) were 198.65 L and 2.41 L/h. Postoperative days (POD), total bilirubin (TBIL), and the use of voriconazole significantly influenced tacrolimus apparent clearance. The incorporation of an increasing number of actual blood drug concentrations into the prediction resulted in a decrease in both PE (72%, 17%, 7%) and APE (87%, 53%, 26%). CONCLUSIONS A qualified PPK model of tacrolimus was developed in Chinese pediatric patients. The individualized tacrolimus dosing software could be used as a suitable tool for the personalization of tacrolimus dosing for pediatric patients after liver transplantation.
Collapse
Affiliation(s)
- Siyu Yang
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Jian Wei
- Department of Interventional Radiography, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xueqiang Pan
- Pharmacy Department of Beijing Health Vocational College, No. 128, Jiukeshu East Road, Tongzhou District, Beijing, 101101, China
| | - Ze Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xuanling Zhang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Zhe Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xianzhe Dong
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Zixin Hua
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xingang Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China.
| |
Collapse
|
30
|
Komenkul V, Sukarnjanaset W, Komolmit P, Wattanavijitkul T. External validation of population pharmacokinetic models of tacrolimus in Thai adult liver transplant recipients. Eur J Clin Pharmacol 2024; 80:1229-1240. [PMID: 38695888 DOI: 10.1007/s00228-024-03692-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/17/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Several population pharmacokinetic models of tacrolimus in liver transplant patients were built, and their predictability was evaluated in their settings. However, the extrapolation in the prediction was unclear. This study aimed to evaluate the predictive performance of published tacrolimus models in adult liver transplant recipients using data from the Thai population as an external dataset. METHODS The selected published models were systematically searched and evaluated for their quality. The external dataset of patients who underwent the first liver transplant and received immediate-release tacrolimus was used to assess the predictive performance of each selected model. Trough concentrations between 3 and 6 months were retrospectively collected to evaluate the predictability of each model using prediction-based diagnostics, simulation-based diagnostics, and Bayesian forecasting. RESULTS Sixty-seven patients with 360 trough concentrations and eight selected published models were included in this study. None of the models met the predictive precision criteria in prediction-based diagnostics. Meanwhile, four published population pharmacokinetic models showed a normal distribution in NPDE testing. Regarding Bayesian forecasting, all models improved their forecasts with at least one prior information data point. CONCLUSION Bayesian forecasting is more accurate and precise than other testing methods for predicting drug concentrations. However, none of the evaluated models provides satisfactory predictive performance for generalization to Thai liver transplant patients. This underscores the need for future research to develop population PK models tailored to the Thai population. Such efforts should consider the inclusion of nonlinear pharmacokinetics and region-specific factors, including genetic variability, to improve model accuracy and applicability.
Collapse
Affiliation(s)
- Virunya Komenkul
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Waroonrat Sukarnjanaset
- Department of Pharmaceutical Care, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Piyawat Komolmit
- Division of Gastro-enterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Liver Diseases, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Thitima Wattanavijitkul
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
31
|
Parant F, Delignette MC, Charpiat B, Lacaille L, Lebosse F, Monneret G, Mohkam K, Mabrut JY, Aubrun F, Heyer L, Antonini T. Tacrolimus Monitoring in Liver Transplant Recipients, Posttransplant Cholestasis: A Comparative Between 2 Commercial Immunoassays and a Liquid Chromatography-Tandem Mass Spectrometry Method. Ther Drug Monit 2024; 46:446-455. [PMID: 38648663 DOI: 10.1097/ftd.0000000000001201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/23/2023] [Indexed: 04/25/2024]
Abstract
BACKGROUND Cholestasis commonly occurs after orthotopic liver transplantation. It can be extrahepatic because of mechanical obstruction or intrahepatic because of various causes. During cholestasis episodes, blood concentrations of tacrolimus (TAC) metabolites may increase, potentially affecting TAC concentrations measured by immunoassays. This study aimed to simultaneously evaluate the analytical performance of 2 TAC immunoassays, a quantitative microsphere system (QMS) immunoassay, and chemiluminescence microparticle immunoassay, using liquid chromatography-tandem mass spectrometry (LC-MS/MS) as a reference method in liver transplant recipients. METHODS This single-center study included 265 patients who underwent orthotopic liver transplantation. In total, 942 blood samples were collected. TAC trough concentrations were measured using LC-MS/MS and 2 immunoassays in parallel. The plasma concentrations of conjugated bilirubin were measured in all samples. The results were analyzed using Bland-Altman plots and Passing-Bablok regressions. RESULTS The Bland-Altman plot analysis showed that the TAC QMS immunoassay has a significant bias (+37%) compared with LC-MS/MS, and this bias was higher in patients with cholestasis with hyperbilirubinemia (≤+70% in patients with conjugated bilirubin >150 µmol/L). In comparison, the chemiluminescence microparticle immunoassay showed acceptable analytical performance in patients with hyperbilirubinemia (bias <10%). CONCLUSIONS In agreement with previous findings, the TAC QMS immunoassay showed a positive bias compared with LC-MS/MS. This bias is remarkably high in patients with cholestasis and hyperbilirubinemia, suggesting the cross-reactivity of TAC metabolites with the monoclonal antibody used in the QMS immunoassay.
Collapse
Affiliation(s)
- François Parant
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-Sites (LBMMS), Hôpital Lyon-Sud-Hospices Civils de Lyon, Pierre-Bénite, France
| | | | - Bruno Charpiat
- Service Pharmaceutique, Hôpital de la Croix Rousse-Hospices Civils de Lyon, Lyon, France
| | - Louis Lacaille
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-Sites (LBMMS), Hôpital de la Croix Rousse-Hospices Civils de Lyon, Lyon, France
| | - Fanny Lebosse
- Service d'Hépatologie, Hôpital de la Croix Rousse-Hospices Civils de Lyon, Lyon, France
| | - Guillaume Monneret
- Laboratoire d'Immunologie, Hôpital Édouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Kayvan Mohkam
- Service de Chirurgie Générale, Digestive et Transplantations Hépatiques et Intestinales, Hôpital de la Croix Rousse-Hospices Civils de Lyon, France; and
| | - Jean-Yves Mabrut
- Service de Chirurgie Générale, Digestive et Transplantations Hépatiques et Intestinales, Hôpital de la Croix Rousse-Hospices Civils de Lyon, France; and
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Lyon, France
| | - Frederic Aubrun
- Service d'Anesthésie Réanimation, Hôpital de la Croix Rousse-Hospices Civils de Lyon, Lyon, France
| | - Laurent Heyer
- Service d'Anesthésie Réanimation, Hôpital de la Croix Rousse-Hospices Civils de Lyon, Lyon, France
| | - Teresa Antonini
- Service d'Hépatologie, Hôpital de la Croix Rousse-Hospices Civils de Lyon, Lyon, France
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, Lyon, France
| |
Collapse
|
32
|
Hu L, Liu J, Fan Q, Zhang S. Mung Bean (Vigna radiata L.) Soup Decreases Tacrolimus Blood Trough Level. Am J Ther 2024; 31:e489-e491. [PMID: 38976537 DOI: 10.1097/mjt.0000000000001696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Affiliation(s)
| | | | - Qiuyu Fan
- Rheumatology, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | | |
Collapse
|
33
|
D'Aragon F, Selzner M, Breau R, Masse MH, Lamontagne F, Masse M, Chassé M, Carrier FM, Cardinal H, Chaudhury P, Weiss M, Lauzier F, Turgeon AF, Frenette AJ, Bolduc B, Ducharme A, Lamarche C, Couture E, Holdsworth S, Bertholz L, Talbot H, Slessarev M, Luke P, Boyd JG, Shamseddin MK, Burns KEA, Zaltzman J, English S, Knoll G, Dhanani S, Healey A, Hanna S, Rochwerg B, Oczkowski SJW, Treleaven D, Meade M. Calcineurin Inhibitor in NEuRoloGically deceased donors to decrease kidney delayed graft function study: study protocol of the CINERGY Pilot randomised controlled trial. BMJ Open 2024; 14:e086777. [PMID: 38871657 PMCID: PMC11177676 DOI: 10.1136/bmjopen-2024-086777] [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] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Most solid organ transplants originate from donors meeting criteria for death by neurological criteria (DNC). Within the organ donor, physiological responses to brain death increase the risk of ischaemia reperfusion injury and delayed graft function. Donor preconditioning with calcineurin inhibition may reduce this risk. METHODS AND ANALYSIS We designed a multicentre placebo-controlled pilot randomised trial involving nine organ donation hospitals and all 28 transplant programmes in the Canadian provinces of Ontario and Québec. We planned to enrol 90 DNC donors and their approximately 324 organ recipients, totalling 414 participants. Donors receive an intravenous infusion of either tacrolimus 0.02 mg/kg over 4 hours prior to organ retrieval, or a matching placebo, while monitored in an intensive care unit for any haemodynamic changes during the infusion. Among all study organ recipients, we record measures of graft function for the first 7 days in hospital and we will record graft survival after 1 year. We examine the feasibility of this trial with respect to the proportion of all eligible donors enrolled and the proportion of all eligible transplant recipients consenting to receive a CINERGY organ transplant and to allow the use of their health data for study purposes. We will report these feasibility outcomes as proportions with 95% CIs. We also record any barriers encountered in the launch and in the implementation of this trial with detailed source documentation. ETHICS AND DISSEMINATION We will disseminate trial results through publications and presentations at participating sites and conferences. This study has been approved by Health Canada (HC6-24-c241083) and by the Research Ethics Boards of all participating sites and in Québec (MP-31-2020-3348) and Clinical Trials Ontario (Project #3309). TRIAL REGISTRATION NUMBER NCT05148715.
Collapse
Affiliation(s)
- Frederick D'Aragon
- Department of Anesthesiology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Markus Selzner
- Multi-Organ Transplant Program, Toronto General Hospital, Toronto, Quebec, Canada
- Department of General Surgery, University of Toronto and Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Ruth Breau
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Marie-Hélène Masse
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Francois Lamontagne
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Medicine, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mélanie Masse
- Department of Medicine, Universite de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Michael Chassé
- Department of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - François-Martin Carrier
- Department of Anesthesiology, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Héloïse Cardinal
- Department of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Prosanto Chaudhury
- Department of Surgery and Oncology, McGill University, Montreal, Québec, Canada
- Transplant Québec, Montréal, Québec, Canada
| | - Matthew Weiss
- Transplant Québec, Montreal, Québec, Canada
- Population Health and Optimal Health Practives Research Unit (Trauma - Emergency - Critical Care Medicine), Centre de Recherche du CHU de Québec - Université Laval, Quebec, Quebec, Canada
| | - Francois Lauzier
- Population Health and Optimal Health Practives Research Unit (Trauma - Emergency - Critical Care Medicine), Centre de Recherche du CHU de Québec - Université Laval, Quebec, Quebec, Canada
- Department of Medicine, Université Laval, Québec City, Québec, Canada
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Université Laval, Québec City, Québec, Canada
| | - Alexis F Turgeon
- Population Health and Optimal Health Practives Research Unit (Trauma - Emergency - Critical Care Medicine), Centre de Recherche du CHU de Québec - Université Laval, Quebec, Quebec, Canada
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Université Laval, Québec City, Québec, Canada
| | | | - Brigitte Bolduc
- Department of Pharmacy, Centre integre universitaire de sante et de services sociaux de l'Estrie Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Anique Ducharme
- Department of Medicine, Université de Montréal, Montreal, Québec, Canada
- Montreal Heart Institute, Montreal, Québec, Canada
| | - Caroline Lamarche
- Department of Medicine, Université de Montréal, Montreal, Québec, Canada
- Hôpital Maisonneuve-Rosemont Research Institute, Montréal, Québec, Canada
| | - Etienne Couture
- Department of Anesthesiology and Critical Care, Quebec Heart & Lung Institute, Université Laval, Quebec, Quebec, Canada
| | - Sandra Holdsworth
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada
| | - Liz Bertholz
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada
| | - Heather Talbot
- Canadian Donation and Transplant Research Program, Edmonton, Alberta, Canada
| | - Marat Slessarev
- Department of Medicine, Division of Critical Care, Western University, London, Ontario, Canada
| | - Patrick Luke
- Department of Surgery, Division of Urology, Western University, London, Ontario, Canada
| | - John Gordon Boyd
- Department of Medicine, Division of Neurology, Queen's University, Kingston, Ontario, Canada
- Department of Critical Care Medicine, Queen's University, Kingston, Ontario, Canada
| | - M Khaled Shamseddin
- Department of Medicine, Division of Nephrology, Queen's University, Kingston, Ontario, Canada
| | - Karen E A Burns
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute, University Health Toronto - St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jeffrey Zaltzman
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shane English
- Division of Critical Care, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Greg Knoll
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, Division of Nephrology, University of Ottawa, Ottawa, Ontario, Canada
| | - Sonny Dhanani
- Department of Pediatrics, Division of Critical Care, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Andrew Healey
- Department of Medicine, Division of Emergency Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Steven Hanna
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Bram Rochwerg
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Darin Treleaven
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Maureen Meade
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
Verona P, Edwards J, Hubert K, Avorio F, Re VL, Di Stefano R, Carollo A, Johnson H, Provenzani A. Tacrolimus-Induced Neurotoxicity After Transplant: A Literature Review. Drug Saf 2024; 47:419-438. [PMID: 38353884 DOI: 10.1007/s40264-024-01398-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 04/17/2024]
Abstract
Tacrolimus, a calcineurin inhibitor, is an immunosuppressant used globally to prevent rejection after organ transplantation. Although it significantly improves outcomes for solid organ transplant patients, it is associated with various side effects such as nephrotoxicity and neurotoxicity. Tacrolimus-induced neurotoxicity is frequently encountered in clinical practice and can present with a variety of symptoms that may occur even at therapeutic levels. Although tacrolimus-induced neurotoxicity is well documented, there is limited literature available on pharmacologic management. Twenty-eight case reports of tacrolimus-induced neurotoxicity were identified and analyzed in addition to other literature including reviews, retrospective studies, and animal model studies. The severity of cases of tacrolimus-induced neurotoxicity reported ranged from mild symptoms that could be managed with symptomatic treatment to conditions such as posterior reversible encephalopathy syndrome and chronic inflammatory demyelinating polyradiculoneuropathy that may require more immediate intervention. This information was utilized in addition to clinical experience to compile potential management options for prevention and treatment of neurotoxic adverse events. This review is limited by the utilization of primarily retrospective studies and case reports. The available literature on the subject is largely narrative and there are no guidelines on treatment of tacrolimus-induced neurotoxicity at the time of this research. This comprehensive review may guide further studies to investigate the pathophysiology of tacrolimus-induced neurotoxicity and to define patient-specific strategies for mitigation or minimization of neurotoxicity. This is especially important given that management of tacrolimus-induced neurotoxicity can include changes to immunosuppression that can result in an increased risk of rejection.
Collapse
Affiliation(s)
- Paige Verona
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jocelyn Edwards
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kassidy Hubert
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Federica Avorio
- Neurology Unit, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Vincenzina Lo Re
- Neurology Unit, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Roberta Di Stefano
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Via E.Tricomi n. 5, 90127, Palermo, Italy
| | - Anna Carollo
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Via E.Tricomi n. 5, 90127, Palermo, Italy
| | - Heather Johnson
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, 3501 Terrace Street, Pittsburgh, PA, USA
| | - Alessio Provenzani
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Via E.Tricomi n. 5, 90127, Palermo, Italy.
| |
Collapse
|
35
|
Karaterzi S, Tönshoff B, Ahlenstiel-Grunow T, Baghai M, Beck B, Büscher A, Eifler L, Giese T, Lezius S, Müller C, Oh J, Zapf A, Weber LT, Pape L. A multi-center interventional study to assess pharmacokinetics, effectiveness, and tolerability of prolonged-release tacrolimus after pediatric kidney transplantation: study protocol for a prospective, open-label, randomized, two-phase, two-sequence, single dose, crossover, phase III b trial. FRONTIERS IN NEPHROLOGY 2024; 4:1331510. [PMID: 38444519 PMCID: PMC10912931 DOI: 10.3389/fneph.2024.1331510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/25/2024] [Indexed: 03/07/2024]
Abstract
Background Tacrolimus, a calcineurin inhibitor (CNI), is currently the first-line immunosuppressive agent in kidney transplantation. The therapeutic index of tacrolimus is narrow due to due to the substantial impact of minor variations in drug concentration or exposure on clinical outcomes (i.e., nephrotoxicity), and it has a highly variable intra- and inter-individual bioavailability. Non-adherence to immunosuppressants is associated with rejection after kidney transplantation, which is the main cause of long-term graft loss. Once-daily formulations have been shown to significantly improve adherence compared to twice-daily dosing. Envarsus®, the once-daily prolonged-release formulation of tacrolimus, offers the same therapeutic efficacy as the conventional twice-daily immediate-release tacrolimus formulation (Prograf®) with improved bioavailability, a more consistent pharmacokinetic profile, and a reduced peak to trough, which may reduce CNI-related toxicity. Envarsus® has been approved as an immunosuppressive therapy in adults following kidney or liver transplantation but has not yet been approved in children. The objective of this study is to evaluate the pharmacokinetic profile, efficacy, and tolerability of Envarsus® in children and adolescents aged ≥ 8 and ≤ 18 years to assess its potential role as an additional option for immunosuppressive therapy in children after kidney transplantation. Methods/design The study is designed as a randomized, prospective crossover trial. Each patient undergoes two treatment sequences: sequence 1 includes 4 weeks of Envarsus® and sequence 2 includes 4 weeks of Prograf®. Patients are randomized to either group A (sequence 1, followed by sequence 2) or group B (sequence 2, followed by sequence 1). The primary objective is to assess equivalency between total exposure (of tacrolimus area under the curve concentration (AUC0-24)), immediate-release tacrolimus (Prograf®) therapy, and prolonged-release tacrolimus (Envarsus®) using a daily dose conversion factor of 0.7 for prolonged- versus immediate-release tacrolimus. Secondary objectives are the assessment of pharmacodynamics, pharmacogenetics, adherence, gut microbiome analyses, adverse events (including tacrolimus toxicity and biopsy-proven rejections), biopsy-proven rejections, difference in estimated glomerular filtration rate (eGFR), and occurrence of donor-specific antibodies (DSAs). Discussion This study will test the hypothesis that once-daily prolonged-release tacrolimus (Envarsus®) is bioequivalent to twice-daily intermediate-release tacrolimus after pediatric kidney transplantation and may reduce toxicity and facilitate medication adherence. This novel concept may optimize immunosuppressive therapy for more stable graft function and increased graft survival by avoiding T-cell mediated and/or antibody-mediated rejection due to improved adherence. In addition, the study will provide data on the pharmacodynamics and pharmacogenetics of prolonged-release tacrolimus in children and adolescents. Clinical Trial Registration EUDRA-CT 2019-003710-13 and ClinicalTrial.gov, identifier NCT06057545.
Collapse
Affiliation(s)
- Sinem Karaterzi
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | | | - Maral Baghai
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Bodo Beck
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Anja Büscher
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
| | - Lisa Eifler
- Children’s and Adolescents’ Hospital, Pediatric Nephrology, University Hospital of Cologne, Cologne, Germany
| | - Thomas Giese
- Department of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Susanne Lezius
- Institute of Medical Biometry and Epidemiology, University Hospital Eppendorf, Hamburg, Germany
| | - Carsten Müller
- Pharmacology at the Laboratory Diagnostics Centre, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jun Oh
- Department of Pediatric Nephrology and Gastroenterology, University Hospital Eppendorf, Hamburg, Germany
| | - Antonia Zapf
- Institute of Medical Biometry and Epidemiology, University Hospital Eppendorf, Hamburg, Germany
| | - Lutz T. Weber
- Children’s and Adolescents’ Hospital, Pediatric Nephrology, University Hospital of Cologne, Cologne, Germany
| | - Lars Pape
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
| |
Collapse
|
36
|
Li Z, Wang X, Li D, Cheng S, Li Z, Guo H, Dong Y, Zheng Y, Li X. Effects of CYP3A4*22 and POR*28 variations on the pharmacokinetics of tacrolimus in renal transplant recipients: a meta-analysis of 18 observational studies. BMC Nephrol 2024; 25:48. [PMID: 38321419 PMCID: PMC10848431 DOI: 10.1186/s12882-024-03467-4] [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] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/16/2024] [Indexed: 02/08/2024] Open
Abstract
PURPOSE This study aimed to investigate the association between cytochrome P450 (CYP) 3A4*22 and cytochrome P450 oxidoreductase (POR)*28 variations and the pharmacokinetics of tacrolimus. METHODS Cochrane Central Register of Controlled Trials (CENTRAL), Web of Science (SCI), MEDLINE, and Embase were systematically searched from inception to August 2022. The outcomes were weight-adjusted daily dose and dose-adjusted trough concentration (C0/Dose). RESULTS The study included 2931 renal transplant recipients from 18 publications. Weight-adjusted daily dose of CYP3A4*1/*1 carriers was 0.04 (WMD = 0.04, 95% CI: 0.02 to 0.06), 0.03 (WMD = 0.03, 95% CI: 0.02 to 0.05), 0.02 (WMD = 0.02, 95% CI: 0.01 to 0.03), or 0.02 mg/kg/day (WMD = 0.02, 95% CI: 0.00 to 0.04) higher than CYP3A4*22 carriers in Caucasians at 1 month, 3 months, 6 months, or 12 months post-transplantation. Conversely, C0/Dose was lower for CYP3A4*1/*1 carriers at 3 days (SMD = -0.35, 95% CI: -0.65 to -0.06), 1 month (SMD = -0.67, 95% CI: -1.16 to -0.18), 3 months (SMD = -0.60, 95% CI: -0.89 to -0.31), 6 months (SMD = -0.76, 95% CI: -1.49 to -0.04), or 12 months post-transplantation (SMD = -0.69, 95% CI: -1.37 to 0.00). Furthermore, C0/Dose of POR*1/*1 carriers was 22.64 (WMD = 22.64, 95% CI: 2.54 to 42.74) or 19.41 (ng/ml)/(mg/kg/day) (WMD = 19.41, 95% CI: 9.58 to 29.24) higher than POR*28 carriers in CYP3A5 expressers at 3 days or 7 days post-transplantation, and higher in Asians at 6 months post-transplantation (SMD = 0.96, 95% CI: 0.50 to 1.43). CONCLUSIONS CYP3A4*22 variant in Caucasians restrains the metabolism of tacrolimus, while POR*28 variant in CYP3A5 expressers enhances the metabolism of tacrolimus for renal transplant recipients. However, further well-designed prospective studies are necessary to substantiate these conclusions given some limitations.
Collapse
Affiliation(s)
- Ze Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Xiaozhen Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dandan Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Sheng Cheng
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Zhe Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Heng Guo
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Yiwen Dong
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Yingming Zheng
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Xingang Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China.
| |
Collapse
|
37
|
Zhumatayev S, Yalcin K, Celen SS, Karaman I, Daloglu H, Ozturkmen S, Uygun V, Karasu G, Yesilipek A. Comparison of tacrolimus vs. cyclosporine in pediatric hematopoietic stem cell transplantation for thalassemia. Pediatr Transplant 2024; 28:e14688. [PMID: 38317344 DOI: 10.1111/petr.14688] [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] [Scholar Register] [Received: 09/14/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 02/07/2024]
Abstract
OBJECTIVES Graft-versus-host disease (GvHD) is one of the leading causes of morbidity and mortality in patients undergoing allogeneic HSCT, and effective prevention of GvHD is critical for the success of the HSCT procedure. Calcineurin inhibitors (CNI) have been used for decades as the backbone of GvHD prophylaxis. In this study, the efficacy and safety of Cyclosporine A (CsA) and tacrolimus (TCR) were compared in pediatric HSCT for thalassemia. MATERIALS AND METHODS This is a retrospective analysis of 129 pediatric patients who underwent HSCT with the diagnosis of thalassemia at Medicalpark Göztepe and Antalya Hospitals between January 2017 and December 2020. RESULTS Despite the GvHD prophylaxis, grade II-IV acute GvHD developed in 29 patients. Of these patients, 12 had only gut, 10 had only skin, 6 had combined gut and skin, and one had only liver GvHD. Fifteen of these 29 patients were in the CsA group, and 14 of them were in the TCR group. There was no significant difference between the groups in terms of acute GvHD occurrence, GvHD stage, or involvement sites. In terms of CNI-related toxicity, neurotoxicity in 15 (CsA n = 9, TCR n = 6) and nephrotoxicity in 18 (CsA n = 4, TCR n = 14) patients were observed. While there was no difference between the two groups in terms of neurotoxicity, more nephrotoxicity developed in patients using TCR (p = .013). There was no significant difference between the groups in terms of engraftment syndrome, veno-occlusive disease, CMV reactivation, PRES, or graft rejection. CONCLUSION Regarding GvHD, there was no difference in efficacy between TCR and CsA usage. Patients taking TCR experienced noticeably higher nephrotoxicity in terms of adverse effects. This difference should be considered according to the patient's clinical situation while choosing a CNI.
Collapse
Affiliation(s)
- Suleimen Zhumatayev
- Department of Pediatric Hematology and Oncology, Goztepe Medical Park Hospital, Istanbul, Turkey
| | - Koray Yalcin
- Department of Pediatric Hematology and Oncology, Bahcesehir University, Goztepe Medical Park Hospital, Istanbul, Turkey
- Department of Medical Biotechnology, Institute of Health Science, Acibadem University, Istanbul, Turkey
| | - Safiye Suna Celen
- Department of Pediatric Hematology and Oncology, Bahcesehir University, Goztepe Medical Park Hospital, Istanbul, Turkey
| | - Irem Karaman
- Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Hayriye Daloglu
- Department of Pediatric Hematology and Oncology, Antalya Medical Park Hospital, Antalya, Turkey
| | - Seda Ozturkmen
- Department of Pediatric Hematology and Oncology, Antalya Medical Park Hospital, Antalya, Turkey
| | - Vedat Uygun
- Department of Pediatric Hematology and Oncology, Istinye University, Goztepe Medical Park Hospital, Istanbul, Turkey
| | - Gulsun Karasu
- Department of Pediatric Hematology and Oncology, Goztepe Medical Park Hospital, Istanbul, Turkey
- Department of Pediatric Hematology and Oncology, Antalya Medical Park Hospital, Antalya, Turkey
| | - Akif Yesilipek
- Department of Pediatric Hematology and Oncology, Goztepe Medical Park Hospital, Istanbul, Turkey
- Department of Pediatric Hematology and Oncology, Antalya Medical Park Hospital, Antalya, Turkey
| |
Collapse
|
38
|
Verdegaal AA, Goodman AL. Integrating the gut microbiome and pharmacology. Sci Transl Med 2024; 16:eadg8357. [PMID: 38295186 PMCID: PMC12121898 DOI: 10.1126/scitranslmed.adg8357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
The gut microbiome harbors trillions of organisms that contribute to human health and disease. These bacteria can also affect the properties of medical drugs used to treat these diseases, and drugs, in turn, can reshape the microbiome. Research addressing interdependent microbiome-host-drug interactions thus has broad impact. In this Review, we discuss these interactions from the perspective of drug bioavailability, absorption, metabolism, excretion, toxicity, and drug-mediated microbiome modulation. We survey approaches that aim to uncover the mechanisms underlying these effects and opportunities to translate this knowledge into new strategies to improve the development, administration, and monitoring of medical drugs.
Collapse
Affiliation(s)
- Andrew A. Verdegaal
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Andrew L. Goodman
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| |
Collapse
|
39
|
Poulin P, Nicolas JM, Bouzom F. A New Version of the Tissue Composition-Based Model for Improving the Mechanism-Based Prediction of Volume of Distribution at Steady-State for Neutral Drugs. J Pharm Sci 2024; 113:118-130. [PMID: 37634869 DOI: 10.1016/j.xphs.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023]
Abstract
In-vitro models are available in the literature for predicting the volume of distribution at steady-state (Vdss) of drugs. The mechanistic model refers to the tissue composition-based model (TCM), which includes important factors that govern Vdss such as drug physiochemistry and physiological data. The recognized TCM published by Rodgers and Rowland (TCM-RR) and a subsequent adjustment made by Simulations Plus Inc. (TCM-SP) have been shown to be generally less accurate with neutral compared to ionized drugs. Therefore, improving these models for neutral drugs becomes necessary. The objective of this study was to propose a new TCM for improving the prediction of Vdss for neutral drugs. The new TCM included two modifications of the published models (i) accentuate the effect of the blood-to-plasma ratio (BPR) that should cover permeated molecules across the biomembranes, which is lacking in these models for neutral compounds, and (ii) use a different approach to estimate the binding in tissues. The new TCM was validated with a large dataset of 202 commercial and proprietary compounds including preclinical and clinical data. All scenario datasets were predicted more accurately with the TCM-New, whereas all statistical parameters indicate that the TCM-New showed significant improvements in terms of accuracy over the TCM-RR and TCM-SP. Predictions of Vdss were frequently more accurate for the TCM-new with 83% within twofold error versus only 50% for the TCM-RR. And more than 95% of the predictions were within threefold error and patient interindividual differences can be predicted with the TCM-New, greatly exceeding the accuracy of the published models. Overall, the new TCM incorporating BPR significantly improved the Vdss predictions in animals and humans for neutral drugs, and, hence, has the potential to better support the drug discovery and facilitate the first-in-human predictions.
Collapse
Affiliation(s)
- Patrick Poulin
- Consultant Patrick Poulin Inc., Québec City, Québec, Canada; School of Public Health, Université de Montréal, Montréal, Québec, Canada.
| | | | - François Bouzom
- DMPK, Development Science, UCB Pharma, Braine I'Alleud, Belgium; Current: Simulations Plus, Inc., 42505 10th Street West, Lancaster, CA 93534, USA
| |
Collapse
|
40
|
Ohno Y, Oriyama T, Honda A, Kurokawa M, Takada T. Induction of CYP3A activity by dexamethasone may not be strong, even at high doses: insights from a case of tacrolimus co-administration. J Pharm Health Care Sci 2023; 9:39. [PMID: 38044431 PMCID: PMC10694947 DOI: 10.1186/s40780-023-00310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/24/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Dexamethasone (DEX) induces CYP3A activity in a concentration-dependent manner. However, no study has examined changes in the blood concentration of CYP3A substrate drugs when DEX is administered at high doses. Herein, we present a case in which tacrolimus (TAC), a typical CYP3A substrate drug, was co-administered with a chemotherapy regimen that included high-dose DEX. CASE PRESENTATION A 71-year-old woman underwent liver transplantation for hepatocellular carcinoma 18 years prior to her inclusion in this case study. She was receiving TAC orally at 2 mg/day and had a stable trough blood concentration of approximately 4 ng/mL and a trough blood concentration/dose (C/D) ratio of approximately 2. The patient was diagnosed with post-transplant lymphoproliferative disease (histological type: Burkitt's lymphoma) after admission. Thereafter, the patient received cyclophosphamide-prednisolone (CP), followed by two courses of R-HyperCVAD (rituximab, cyclophosphamide, doxorubicin, vincristine, and DEX) and R-MA (rituximab, methotrexate, and cytarabine) replacement therapy. DEX (33 mg/day) was administered intravenously on days 1-4 and days 11-14 of R-HyperCVAD treatment, and aprepitant (APR) was administered on days 1-5 in both courses. The TAC C/D ratio decreased to approximately 1 on day 11 during both courses, and then increased. Furthermore, a decreasing trend in the TAC C/D ratio was observed after R-MA therapy. The decrease in the TAC C/D ratio was attributed to APR administration rather than to DEX. CONCLUSION The induction of CYP3A activity by a high dose of DEX may not be strong. The pharmacokinetic information on DEX and in vitro enzyme activity induction studies also suggested that CYP3A activity induction is not prominent under high-dose DEX treatment.
Collapse
Affiliation(s)
- Yoshiyuki Ohno
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, Bunkyo-Ku, 113-8654, Japan.
| | - Toyohito Oriyama
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, Bunkyo-Ku, 113-8654, Japan
| | - Akira Honda
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, Bunkyo-Ku, 113-8654, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, Bunkyo-Ku, 113-8654, Japan
- Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Tokyo, Bunkyo-Ku, 113-8654, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, Bunkyo-Ku, 113-8654, Japan
| |
Collapse
|
41
|
El-Khateeb E, Chinnadurai R, Al Qassabi J, Scotcher D, Darwich AS, Kalra PA, Rostami-Hodjegan A. Using Prior Knowledge on Systems Through PBPK to Gain Further Insight into Routine Clinical Data on Trough Concentrations: The Case of Tacrolimus in Chronic Kidney Disease. Ther Drug Monit 2023; 45:743-753. [PMID: 37315152 PMCID: PMC10635338 DOI: 10.1097/ftd.0000000000001108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/23/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Routine therapeutic drug monitoring (TDM) relies heavily on measuring trough drug concentrations. Trough concentrations are affected not only by drug bioavailability and clearance, but also by various patient and disease factors and the volume of distribution. This often makes interpreting differences in drug exposure from trough data challenging. This study aimed to combine the advantages of top-down analysis of therapeutic drug monitoring data with bottom-up physiologically-based pharmacokinetic (PBPK) modeling to investigate the effect of declining renal function in chronic kidney disease (CKD) on the nonrenal intrinsic metabolic clearance ( CLint ) of tacrolimus as a case example. METHODS Data on biochemistry, demographics, and kidney function, along with 1167 tacrolimus trough concentrations for 40 renal transplant patients, were collected from the Salford Royal Hospital's database. A reduced PBPK model was developed to estimate CLint for each patient. Personalized unbound fractions, blood-to-plasma ratios, and drug affinities for various tissues were used as priors to estimate the apparent volume of distribution. Kidney function based on the estimated glomerular filtration rate ( eGFR ) was assessed as a covariate for CLint using the stochastic approximation of expectation and maximization method. RESULTS At baseline, the median (interquartile range) eGFR was 45 (34.5-55.5) mL/min/1.73 m 2 . A significant but weak correlation was observed between tacrolimus CLint and eGFR (r = 0.2, P < 0.001). The CLint declined gradually (up to 36%) with CKD progression. Tacrolimus CLint did not differ significantly between stable and failing transplant patients. CONCLUSIONS Kidney function deterioration in CKD can affect nonrenal CLint for drugs that undergo extensive hepatic metabolism, such as tacrolimus, with critical implications in clinical practice. This study demonstrates the advantages of combining prior system information (via PBPK) to investigate covariate effects in sparse real-world datasets.
Collapse
Affiliation(s)
- Eman El-Khateeb
- Certara UK Limited (Simcyp Division), Sheffield, United Kingdom
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Rajkumar Chinnadurai
- Northern Care Alliance NHS Foundation Trust, Salford, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jokha Al Qassabi
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom
- University of Technology and Applied Sciences, Muscat, Oman; and
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom
| | - Adam S. Darwich
- Logistics and Informatics in Health Care, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Philip A. Kalra
- Northern Care Alliance NHS Foundation Trust, Salford, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Amin Rostami-Hodjegan
- Certara UK Limited (Simcyp Division), Sheffield, United Kingdom
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
42
|
Mohammed Ali Z, Meertens M, Fernández B, Fontova P, Vidal-Alabró A, Rigo-Bonnin R, Melilli E, Cruzado JM, Grinyó JM, Colom H, Lloberas N. CYP3A5*3 and CYP3A4*22 Cluster Polymorphism Effects on LCP-Tac Tacrolimus Exposure: Population Pharmacokinetic Approach. Pharmaceutics 2023; 15:2699. [PMID: 38140040 PMCID: PMC10747255 DOI: 10.3390/pharmaceutics15122699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/24/2023] [Accepted: 11/26/2023] [Indexed: 12/24/2023] Open
Abstract
The aim of the study is to develop a population pharmacokinetic (PopPK) model and to investigate the influence of CYP3A5/CYP3A4 and ABCB1 single nucleotide polymorphisms (SNPs) on the Tacrolimus PK parameters after LCP-Tac formulation in stable adult renal transplant patients. The model was developed, using NONMEM v7.5, from full PK profiles from a clinical study (n = 30) and trough concentrations (C0) from patient follow-up (n = 68). The PK profile of the LCP-Tac formulation was best described by a two-compartment model with linear elimination, parameterized in elimination (CL/F) and distributional (CLD/F) clearances and central compartment (Vc/F) and peripheral compartment (Vp/F) distribution volumes. A time-lagged first-order absorption process was characterized using transit compartment models. According to the structural part of the base model, the LCP-Tac showed an absorption profile characterized by two transit compartments and a mean transit time of 3.02 h. Inter-individual variability was associated with CL/F, Vc/F, and Vp/F. Adding inter-occasion variability (IOV) on CL/F caused a statistically significant reduction in the model minimum objective function MOFV (p < 0.001). Genetic polymorphism of CYP3A5 and a cluster of CYP3A4/A5 SNPs statistically significantly influenced Tac CL/F. In conclusion, a PopPK model was successfully developed for LCP-Tac formulation in stable renal transplant patients. CYP3A4/A5 SNPs as a combined cluster including three different phenotypes (high, intermediate, and poor metabolizers) was the most powerful covariate to describe part of the inter-individual variability associated with apparent elimination clearance. Considering this covariate in the initial dose estimation and during the therapeutic drug monitoring (TDM) would probably optimize Tac exposure attainments.
Collapse
Affiliation(s)
- Zeyar Mohammed Ali
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy, University of Barcelona, 08007 Barcelona, Spain
| | - Marinda Meertens
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy, University of Barcelona, 08007 Barcelona, Spain
| | - Beatriz Fernández
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy, University of Barcelona, 08007 Barcelona, Spain
| | - Pere Fontova
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
| | - Anna Vidal-Alabró
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
| | - Raul Rigo-Bonnin
- Biochemistry Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain;
| | - Edoardo Melilli
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
| | - Josep M. Cruzado
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
| | - Josep M. Grinyó
- Department of Clinical Sciences, Medicine Unit, University of Barcelona, 08007 Barcelona, Spain;
| | - Helena Colom
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, School of Pharmacy, University of Barcelona, 08007 Barcelona, Spain
| | - Nuria Lloberas
- Nephrology Department, Hospital Universitari de Bellvitge-IDIBELL, 08908 Barcelona, Spain; (Z.M.A.); (M.M.); (B.F.); (P.F.); (A.V.-A.); (E.M.); (J.M.C.)
| |
Collapse
|
43
|
Zaichick S, Caraveo G. Harnessing IGF-1 and IL-2 as biomarkers for calcineurin activity to tailor optimal FK506 dosage in α-synucleinopathies. Front Mol Biosci 2023; 10:1292555. [PMID: 38094080 PMCID: PMC10716490 DOI: 10.3389/fmolb.2023.1292555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/16/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction: Rise in Calcium (Ca2+) and hyperactive Ca2+-dependent phosphatase calcineurin represent two key determinants of a-synuclein (a-syn) pathobiology implicated in Parkinson's Disease (PD) and other neurodegenerative diseases. Calcineurin activity can be inhibited with FK506, a Food and Drug Administration (FDA)-approved compound. Our previous work demonstrated a protective effect of low doses of FK506 against a-syn pathology in various models of a-syn related pathobiology. Methods: Control and a-syn-expressing mice (12-18 months old) were injected with vehicle or two single doses of FK506 administered 4 days apart. Cerebral cortex and serum from these mice were collected and assayed using a meso scale discovery quickplex SQ 120 for cytokines and Enzyme-linked immunosorbent assay for IGF-1. Results: In this study we present evidence that reducing calcineurin activity with FK506 in a-syn transgenic mice increased insulin growth factor (IGF-1), while simultaneously decreasing IL-2 levels in both cerebral cortex and serum. Discussion: The highly conserved Ca2+/calcineurin signaling pathway is known to be affected in a-syn-dependent human disease. FK506, an already approved drug for other uses, exhibits high brain penetrance and a proven safety profile. IL-2 and IGF-1 are produced throughout life and can be measured using standard clinical methods. Our findings provide two potential biomarkers that could guide a clinical trial of FK506 in PD patients, without posing significant logistical or regulatory challenges.
Collapse
Affiliation(s)
| | - Gabriela Caraveo
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
44
|
Coppola P, Butler A, Cole S, Kerwash E. Total and Free Blood and Plasma Concentration Changes in Pregnancy for Medicines Highly Bound to Plasma Proteins: Application of Physiologically Based Pharmacokinetic Modelling to Understand the Impact on Efficacy. Pharmaceutics 2023; 15:2455. [PMID: 37896215 PMCID: PMC10609738 DOI: 10.3390/pharmaceutics15102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Free drug concentrations are generally considered the pharmacologically active moiety and are important for cellular diffusion and distribution. Pregnancy-related changes in plasma protein binding and blood partitioning are due to decreases in plasma albumin, alpha-1-acid glycoprotein, and haematocrit; this may lead to increased free concentrations, tissue distribution, and clearance during pregnancy. In this paper we highlight the importance and challenges of considering changes in total and free concentrations during pregnancy. For medicines highly bound to plasma proteins, such as tacrolimus, efavirenz, clindamycin, phenytoin, and carbamazepine, differential changes in concentrations of free drug during pregnancy may be clinically significant and have important implications for dose adjustment. Therapeutic drug monitoring usually relies on the measurement of total concentrations; this can result in dose adjustments that are not necessary when changes in free concentrations are considered. We explore the potential of physiologically based pharmacokinetic (PBPK) models to support the understanding of the changes in plasma proteins binding, using tacrolimus and efavirenz as example drug models. The exposure to either drug was predicted to be reduced during pregnancy; however, the decrease in the exposure to the total tacrolimus and efavirenz were significantly larger than the reduction in the exposure to the free drug. These data show that PBPK modelling can support the impact of the changes in plasma protein binding and may be used for the simulation of free concentrations in pregnancy to support dosing decisions.
Collapse
|
45
|
Wanas H, Kamel MH, William EA, Fayad T, Abdelfattah ME, Elbadawy HM, Mikhael ES. The impact of CYP3A4 and CYP3A5 genetic variations on tacrolimus treatment of living-donor Egyptian kidney transplanted patients. J Clin Lab Anal 2023; 37:e24969. [PMID: 37789683 PMCID: PMC10681408 DOI: 10.1002/jcla.24969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Tacrolimus (TAC) is the mainstay of immunosuppressive regimen for kidney transplantations. Its clinical use is complex due to high inter-individual variations which can be partially attributed to genetic variations at the metabolizing enzymes CYP3A4 and CYP3A5. Two single nucleotide polymorphisms (SNPs), CYP3A4*22 and CYP3A5*3, have been reported as important causes of differences in pharmacokinetics that can affect efficacy and/or toxicity of TAC. OBJECTIVE Investigating the effect of CYP3A4*22 and CYP3A5*3 SNPs individually and in combination on the TAC concentration in Egyptian renal recipients. METHODS Overall, 72 Egyptian kidney transplant recipients were genotyped for CYP3A4*22 G>A and CYP3A5*3 T>C. According to the functional defect associated with CYP3A variants, patients were clustered into: poor (PM) and non-poor metabolizers (Non-PM). The impact on dose adjusted through TAC concentrations (C0) and daily doses at different time points after transplantation was evaluated. RESULTS Cyp3A4*1/*22 and PM groups require significantly lower dose of TAC (mg/kg) at different time points with significantly higher concentration/dose (C0/D) ratio at day 10 in comparison to Cyp3A4*1/*1 and Non-PM groups respectively. However, CyP3A5*3 heterozygous individuals did not show any significant difference in comparison to CyP3A5*1/*3 individuals. By comparing between PM and Non-PM, the PM group had a significantly lower rate of recipients not reaching target C0 at day 14. CONCLUSION This is the first study on Egyptian population to investigate the impact of CYP3A4*22 and CYP3A5*3 SNPs individually and in combination on the TAC concentration. This study and future multicenter studies can contribute to the individualization of TAC dosing in Egyptian patients.
Collapse
Affiliation(s)
- Hanaa Wanas
- Medical Pharmacology DepartmentFaculty of Medicine Cairo UniversityCairoEgypt
- Pharmacology and Toxicology Department, Faculty of PharmacyTaibah UniversityMadinahSaudi Arabia
| | - Mai Hamed Kamel
- Clinical and Chemical Pathology DepartmentFaculty of Medicine Cairo UniversityCairoEgypt
| | - Emad Adel William
- National Research Centre, Medical Research and Clinical Studies InstituteCairoEgypt
| | - Tarek Fayad
- Internal Medicine DepartmentFaculty of Medicine Cairo UniversityCairoEgypt
| | | | | | - Emily Samir Mikhael
- Clinical and Chemical Pathology DepartmentFaculty of Medicine Cairo UniversityCairoEgypt
| |
Collapse
|
46
|
Nguyen TD, Smith NM, Attwood K, Gundroo A, Chang S, Yonis M, Murray B, Tornatore KM. Bayesian optimization of tacrolimus exposure in stable kidney transplant patients. Pharmacotherapy 2023; 43:1032-1042. [PMID: 37452631 PMCID: PMC10592415 DOI: 10.1002/phar.2848] [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] [Scholar Register] [Received: 12/19/2022] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 07/18/2023]
Abstract
STUDY OBJECTIVE The objective was to compare tacrolimus AUC0-12 determined by Non-Compartmental Analysis (NCA) using intensive sampling to Maximum a Posteriori-Bayesian (MAP-Bayesian) estimates from robust (n = 9 samples/subject) and sparse (n = 2 samples/subject) sampling in 67 stable KTRs and a validation group of similar patients. DESIGN This open-label, prospective, single center 12-h PK study included nine serial samples collected in KTRs to determine steady-state NCA tacrolimus AUC0-12 . SETTING This study was conducted at a single site within a large, urban hospital in the western New York area. PATIENTS This study described tacrolimus pharmacokinetics in stable kidney transplant recipients on maintenance tacrolimus therapy. INTERVENTION Robust and sparse AUC0-12 estimates by a MAP-Bayesian approach were obtained using the Advanced Dosing Solutions (AdDS) and ADAPT5 freeware. Limited sampling strategies were evaluated using the original population PK model (n = 67), which was also assessed using a validation group (n = 15). AUC0-12 agreement was tested by paired t-tests with intraclass correlation coefficient (ICC) and Bland Altman analysis. MEASUREMENTS AND MAIN RESULTS A total of 35 Black and 32 White stable KTRs (estimated glomerular filtration rate [eGFR] = 55.2 ± 15.7 mL/min/1.73m2 ) received the tacrolimus dose of 3.4 ± 1.7 mg/study with troughs of 6.8 ± 1.8 ng/mL. The NCA-AUC0-12 was 123.8 ± 33.6 μg·h/L compared to MAP-Bayesian estimates for Robust-AUC0-12 of 124.7 ± 33.3 μg·h/L and optimal 2-specimen Sparse-AUC0-12 of 119.7 ± 32.7 μg·h/L for the training group. Comparison of Robust-AUC0-12 to NCA-AUC0-12 had an ICC of 0.96 (p = 0.99) while comparison of Robust-AUC0-12 to Sparse-AUC0-12 using Pre-dose trough [C(t0h )] and 1 h [C(t1h )] resulted in an ICC of 0.93 (p = 0.014). In the validation group, 5 Black and 10 White KTRs (eGFR = 56.4 ± 16.8 mL/min/1.73m2 ) received a mean tacrolimus dose of 1.9 ± 1.2 mg/study with a trough of 6.0 ± 1.7 ng/mL. The validation group's NCA-AUC0-12 (88.4 ± 33.1 μg·h/L) was comparable to Robust-AUC0-12 (85.1 ± 33.8 μg·h/L, ICC = 0.93; p = 0.12) and Sparse-AUC0-12 determined from C(t0h ) and C(t4h ) (86.7 ± 33.9 μg·h/L, ICC = 0.91; p = 0.61). CONCLUSION MAP-Bayesian estimation for patient-specific AUC0-12 using sparse, two-specimen sampling is comparable to NCA and may enhance tacrolimus TDM in stable KTRs.
Collapse
Affiliation(s)
- Thomas D. Nguyen
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| | - Nicholas M. Smith
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| | - Kris Attwood
- Biostatistics, School of Public Health and Health Professions, Buffalo, New York, USA
| | - Aijaz Gundroo
- Nephrology Division; Medicine, School of Medicine, and Biomedical Sciences, Buffalo, New York, USA
| | - Shirley Chang
- Nephrology Division; Medicine, School of Medicine, and Biomedical Sciences, Buffalo, New York, USA
- Erie County Medical Center, Buffalo, New York, USA
| | - Mahfuz Yonis
- Nephrology Division; Medicine, School of Medicine, and Biomedical Sciences, Buffalo, New York, USA
- Erie County Medical Center, Buffalo, New York, USA
| | - Brian Murray
- Nephrology Division; Medicine, School of Medicine, and Biomedical Sciences, Buffalo, New York, USA
- Erie County Medical Center, Buffalo, New York, USA
| | - Kathleen M. Tornatore
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- Nephrology Division; Medicine, School of Medicine, and Biomedical Sciences, Buffalo, New York, USA
| |
Collapse
|
47
|
Henkel L, Jehn U, Thölking G, Reuter S. Tacrolimus-why pharmacokinetics matter in the clinic. FRONTIERS IN TRANSPLANTATION 2023; 2:1160752. [PMID: 38993881 PMCID: PMC11235362 DOI: 10.3389/frtra.2023.1160752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/07/2023] [Indexed: 07/13/2024]
Abstract
The calcineurin inhibitor (CNI) Tacrolimus (Tac) is the most prescribed immunosuppressant drug after solid organ transplantation. After renal transplantation (RTx) approximately 95% of recipients are discharged with a Tac-based immunosuppressive regime. Despite the high immunosuppressive efficacy, its adverse effects, narrow therapeutic window and high intra- and interpatient variability (IPV) in pharmacokinetics require therapeutic drug monitoring (TDM), which makes treatment with Tac a major challenge for physicians. The C/D ratio (full blood trough level normalized by daily dose) is able to classify patients receiving Tac into two major metabolism groups, which were significantly associated with the clinical outcomes of patients after renal or liver transplantation. Therefore, the C/D ratio is a simple but effective tool to identify patients at risk of an unfavorable outcome. This review highlights the challenges of Tac-based immunosuppressive therapy faced by transplant physicians in their daily routine, the underlying causes and pharmacokinetics (including genetics, interactions, and differences between available Tac formulations), and the latest data on potential solutions to optimize treatment of high-risk patients.
Collapse
Affiliation(s)
- Lino Henkel
- Department of Medicine D, University of Münster, Münster, Germany
| | - Ulrich Jehn
- Department of Medicine D, University of Münster, Münster, Germany
| | - Gerold Thölking
- Department of Medicine D, University of Münster, Münster, Germany
- Department of Internal Medicine and Nephrology, University Hospital of Münster Marienhospital Steinfurt, Steinfurt, Germany
| | - Stefan Reuter
- Department of Medicine D, University of Münster, Münster, Germany
| |
Collapse
|
48
|
Kim JS, Shim S, Yee J, Choi KH, Gwak HS. Effects of CYP3A4*22 polymorphism on trough concentration of tacrolimus in kidney transplantation: a systematic review and meta-analysis. Front Pharmacol 2023; 14:1201083. [PMID: 37564175 PMCID: PMC10409991 DOI: 10.3389/fphar.2023.1201083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
Purpose: Tacrolimus (Tac) is a widely used immunosuppressive agent in kidney transplantation. Cytochrome P450 (CYP), especially CYP3A4 enzymes are responsible for the metabolism of drugs. However, the correlation between plasma Tac concentration and CYP3A4*22 gene variants is controversial. This meta-analysis aims to evaluate the association between CYP3A4*22 polymorphism and the dose-adjusted trough concentration (C0/D) of Tac in adult kidney transplant patients. Methods: We conducted a literature review for qualifying studies using the PubMed, Web of Science, and Embase databases until July 2023. For the continuous variables (C0/D and daily dose), mean difference (MD) and corresponding 95% confidence intervals (CIs) were calculated to evaluate the association between the CYP3A4 * 22 and Tac pharmacokinetics. We performed an additional analysis on the relationship of CYP3A5*3 with Tac PKs and analyzed the effects of CYP3A4*22 in CYP3A5 non-expressers. Results: Overall, eight eligible studies with 2,683 renal transplant recipients were included in this meta-analysis. The CYP3A4*22 allele was significantly associated with a higher C0/D (MD 0.57 ng/mL/mg (95% CI: 0.28 to 0.86; p = 0.0001) and lower mean daily dose requirement (MD -2.02 mg/day, 95% CI: -2.55 to -1.50; p < 0.00001). An additional meta-analysis demonstrated that carrying the CYP3A5*3 polymorphism greatly impacted Tac blood concentration. From the result with CYP3A5 non-expressers, CYP3A4*22 showed significant effects on the Tac C0/D and dose requirement even after adjusting the effect of CYP3A5*3. Conclusion: Patients with CYP3A4*22 allele showed significantly higher plasma C0/D of Tac and required lower daily dose to achieve the therapeutic trough level after kidney transplantation. These findings of our meta-analysis may provide further evidence for the effects of genetic polymorphism in CYP3A4 on the PKs of Tac, which will improve individualized treatment in a clinical setting.
Collapse
Affiliation(s)
- Jung Sun Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sunyoung Shim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jeong Yee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Kyung Hee Choi
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Hye Sun Gwak
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
49
|
Degraeve AL, Haufroid V, Loriot A, Gatto L, Andries V, Vereecke L, Elens L, Bindels LB. Gut microbiome modulates tacrolimus pharmacokinetics through the transcriptional regulation of ABCB1. MICROBIOME 2023; 11:138. [PMID: 37408070 DOI: 10.1186/s40168-023-01578-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Following solid organ transplantation, tacrolimus (TAC) is an essential drug in the immunosuppressive strategy. Its use constitutes a challenge due to its narrow therapeutic index and its high inter- and intra-pharmacokinetic (PK) variability. As the contribution of the gut microbiota to drug metabolism is now emerging, it might be explored as one of the factors explaining TAC PK variability. Herein, we explored the consequences of TAC administration on the gut microbiota composition. Reciprocally, we studied the contribution of the gut microbiota to TAC PK, using a combination of in vivo and in vitro models. RESULTS TAC oral administration in mice resulted in compositional alterations of the gut microbiota, namely lower evenness and disturbance in the relative abundance of specific bacterial taxa. Compared to controls, mice with a lower intestinal microbial load due to antibiotics administration exhibit a 33% reduction in TAC whole blood exposure and a lower inter-individual variability. This reduction in TAC levels was strongly correlated with higher expression of the efflux transporter ABCB1 (also known as the p-glycoprotein (P-gp) or the multidrug resistance protein 1 (MDR1)) in the small intestine. Conventionalization of germ-free mice confirmed the ability of the gut microbiota to downregulate ABCB1 expression in a site-specific fashion. The functional inhibition of ABCB1 in vivo by zosuquidar formally established the implication of this efflux transporter in the modulation of TAC PK by the gut microbiota. Furthermore, we showed that polar bacterial metabolites could recapitulate the transcriptional regulation of ABCB1 by the gut microbiota, without affecting its functionality. Finally, whole transcriptome analyses pinpointed, among others, the Constitutive Androstane Receptor (CAR) as a transcription factor likely to mediate the impact of the gut microbiota on ABCB1 transcriptional regulation. CONCLUSIONS We highlight for the first time how the modulation of ABCB1 expression by bacterial metabolites results in changes in TAC PK, affecting not only blood levels but also the inter-individual variability. More broadly, considering the high number of drugs with unexplained PK variability transported by ABCB1, our work is of clinical importance and paves the way for incorporating the gut microbiota in prediction algorithms for dosage of such drugs. Video Abstract.
Collapse
Affiliation(s)
- Alexandra L Degraeve
- Department of Integrated PharmacoMetrics, PharmacoGenomics and PharmacoKinetics, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vincent Haufroid
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
- Department of Clinical Chemistry, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Axelle Loriot
- Computational Biology and Bioinformatics Unit (CBIO), de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Laurent Gatto
- Computational Biology and Bioinformatics Unit (CBIO), de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vanessa Andries
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent, Belgium
| | - Lars Vereecke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent, Belgium
| | - Laure Elens
- Department of Integrated PharmacoMetrics, PharmacoGenomics and PharmacoKinetics, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
- WELBIO department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
50
|
Rower JE, McKnite A, Hong B, Daly KP, Hope KD, Cabrera AG, Molina KM. External assessment and refinement of a population pharmacokinetic model to guide tacrolimus dosing in pediatric heart transplant. Pharmacotherapy 2023; 43:650-658. [PMID: 37328271 PMCID: PMC10527671 DOI: 10.1002/phar.2836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 06/18/2023]
Abstract
STUDY OBJECTIVE The immunosuppressant tacrolimus is a first-line agent to prevent graft rejection following pediatric heart transplant; however, it suffers from extensive inter-patient variability and a narrow therapeutic window. Personalized tacrolimus dosing may improve transplant outcomes by more efficiently achieving and maintaining therapeutic tacrolimus concentrations. We sought to externally validate a previously published population pharmacokinetic (PK) model that was constructed with data from a single site. DATA SOURCE Data were collected from Seattle, Texas, and Boston Children's Hospitals, and assessed using standard population PK modeling techniques in NONMEMv7.2. MAIN RESULTS While the model was not successfully validated for use with external data, further covariate searching identified weight (p < 0.0001 on both volume and elimination rate) as a model-significant covariate. This refined model acceptably predicted future tacrolimus concentrations when guided by as few as three concentrations (median prediction error = 7%; median absolute prediction error = 27%). CONCLUSION These findings support the potential clinical utility of a population PK model to provide personalized tacrolimus dosing guidance.
Collapse
Affiliation(s)
- Joseph E. Rower
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, USA
- Center for Human Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, USA
| | - Autumn McKnite
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, USA
| | - Borah Hong
- Division of Pediatric Cardiology, University of Washington and Seattle Children’s Hospital, Seattle, Washington, USA
| | - Kevin P. Daly
- Department of Pediatric Cardiology, Harvard Medical School/Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Kyle D. Hope
- Lillie Frank Abercrombie Division of Pediatric Cardiology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Antonio G. Cabrera
- Lillie Frank Abercrombie Division of Pediatric Cardiology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Cardiology, University of Utah/Intermountain Primary Children’s Hospital, Salt Lake City, Utah, USA
| | - Kimberly M. Molina
- Division of Pediatric Cardiology, University of Utah/Intermountain Primary Children’s Hospital, Salt Lake City, Utah, USA
| |
Collapse
|