1
|
Berglund LJ. Modulating the PI3K Signalling Pathway in Activated PI3K Delta Syndrome: a Clinical Perspective. J Clin Immunol 2023; 44:34. [PMID: 38148368 PMCID: PMC10751257 DOI: 10.1007/s10875-023-01626-0] [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: 09/25/2023] [Accepted: 11/09/2023] [Indexed: 12/28/2023]
Abstract
Activated phosphoinositide-3-kinase (PI3K) δ syndrome (APDS) is an inborn error of immunity characterised by immune dysregulation. Since the discovery of genetic mutations resulting in PI3Kδ overactivation, treatment of APDS patients has begun to focus on modulation of the PI3K pathway in addition to supportive therapies. The mTOR inhibitor sirolimus has been used effectively for some clinical manifestations of this condition, however the arrival of specific PI3Kδ inhibitor leniolisib has shown promising early results and may provide a more targeted approach. This review summarizes key aspects of PI3K pathway biology and discusses potential options for nuanced modulation of the PI3K pathway in APDS from a clinical perspective, highlighting differences from PI3K inhibition in haematological malignancies.
Collapse
Affiliation(s)
- Lucinda J Berglund
- Faculty of Medicine, University of Sydney, Sydney, NSW, Australia.
- Department of Immunopathology, Westmead Hospital, NSW Health Pathology, Westmead, Sydney, NSW, Australia.
| |
Collapse
|
2
|
Shen G, Moua KTY, Perkins K, Johnson D, Li A, Curtin P, Gao W, McCune JS. Precision sirolimus dosing in children: The potential for model-informed dosing and novel drug monitoring. Front Pharmacol 2023; 14:1126981. [PMID: 37021042 PMCID: PMC10069443 DOI: 10.3389/fphar.2023.1126981] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/14/2023] [Indexed: 04/07/2023] Open
Abstract
The mTOR inhibitor sirolimus is prescribed to treat children with varying diseases, ranging from vascular anomalies to sporadic lymphangioleiomyomatosis to transplantation (solid organ or hematopoietic cell). Precision dosing of sirolimus using therapeutic drug monitoring (TDM) of sirolimus concentrations in whole blood drawn at the trough (before the next dose) time-point is the current standard of care. For sirolimus, trough concentrations are only modestly correlated with the area under the curve, with R 2 values ranging from 0.52 to 0.84. Thus, it should not be surprising, even with the use of sirolimus TDM, that patients treated with sirolimus have variable pharmacokinetics, toxicity, and effectiveness. Model-informed precision dosing (MIPD) will be beneficial and should be implemented. The data do not suggest dried blood spots point-of-care sampling of sirolimus concentrations for precision dosing of sirolimus. Future research on precision dosing of sirolimus should focus on pharmacogenomic and pharmacometabolomic tools to predict sirolimus pharmacokinetics and wearables for point-of-care quantitation and MIPD.
Collapse
Affiliation(s)
- Guofang Shen
- Department of Hematologic Malignancies Translational Sciences, City of Hope, and Department of Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, United States
| | - Kao Tang Ying Moua
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, United States
| | - Kathryn Perkins
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, United States
| | - Deron Johnson
- Clinical Informatics, City of Hope Medical Center, Duarte, CA, United States
| | - Arthur Li
- Division of Biostatistics, City of Hope, Duarte, CA, United States
| | - Peter Curtin
- Department of Hematologic Malignancies Translational Sciences, City of Hope, and Department of Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, United States
| | - Wei Gao
- Division of Engineering and Applied Science, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Jeannine S. McCune
- Department of Hematologic Malignancies Translational Sciences, City of Hope, and Department of Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, United States
| |
Collapse
|
3
|
Zhai Q, van der Lee M, van Gelder T, Swen JJ. Why We Need to Take a Closer Look at Genetic Contributions to CYP3A Activity. Front Pharmacol 2022; 13:912618. [PMID: 35784699 PMCID: PMC9243486 DOI: 10.3389/fphar.2022.912618] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cytochrome P450 3A (CYP3A) subfamily enzymes are involved in the metabolism of 40% of drugs in clinical use. Twin studies have indicated that 66% of the variability in CYP3A4 activity is hereditary. Yet, the complexity of the CYP3A locus and the lack of distinct drug metabolizer phenotypes has limited the identification and clinical application of CYP3A genetic variants compared to other Cytochrome P450 enzymes. In recent years evidence has emerged indicating that a substantial part of the missing heritability is caused by low frequency genetic variation. In this review, we outline the current pharmacogenomics knowledge of CYP3A activity and discuss potential future directions to improve our genetic knowledge and ability to explain CYP3A variability.
Collapse
|
4
|
The Interplay between Vitamin D, Exposure of Anticholinergic Antipsychotics and Cognition in Schizophrenia. Biomedicines 2022; 10:biomedicines10051096. [PMID: 35625833 PMCID: PMC9138360 DOI: 10.3390/biomedicines10051096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 01/21/2023] Open
Abstract
Vitamin D deficiency is a frequent finding in schizophrenia and may contribute to neurocognitive dysfunction, a core element of the disease. However, there is limited knowledge about the neuropsychological profile of vitamin D deficiency-related cognitive deficits and their underlying molecular mechanisms. As an inductor of cytochrome P450 3A4, a lack of vitamin D might aggravate cognitive deficits by increased exposure to anticholinergic antipsychotics. This cross-sectional study aims to assess the relationship between 25-OH-vitamin D-serum concentrations, anticholinergic drug exposure and neurocognitive functioning (Brief Assessment of Cognition in Schizophrenia, BACS, and Trail Making Test, TMT) in 141 patients with schizophrenia. The anticholinergic drug exposure was estimated by adjusting the concentration of each drug for its individual muscarinic receptor affinity. Using regression analysis, we observed a positive relationship between vitamin D levels and processing speed (TMT-A and BACS Symbol Coding) as well as executive functioning (TMT-B and BACS Tower of London). Moreover, a negative impact of vitamin D on anticholinergic drug exposure emerged, but the latter did not significantly affect cognition. When other cognitive items were included as regressors, the impact of vitamin D remained only significant for the TMT-A. Among the different cognitive impairments in schizophrenia, vitamin D deficiency may most directly affect processing speed, which in turn may aggravate deficits in executive functioning. This finding is not explained by a cytochrome P450-mediated increased exposure to anticholinergic antipsychotics.
Collapse
|
5
|
Pavek P, Dusek J, Smutny T, Lochman L, Kucera R, Skoda J, Smutna L, Kamaraj R, Soucek P, Vrzal R, Dvorak Z. Gene expression profiling of 1α,25(OH)
2
D
3
treatment in 2D/3D human hepatocyte models reveals CYP3A4 induction but minor changes in other xenobiotic‐metabolizing genes. Mol Nutr Food Res 2022; 66:e2200070. [DOI: 10.1002/mnfr.202200070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Petr Pavek
- Department of Pharmacology and Toxicology Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Jan Dusek
- Department of Pharmacology and Toxicology Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Tomas Smutny
- Department of Pharmacology and Toxicology Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Lukas Lochman
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Radim Kucera
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Josef Skoda
- Department of Pharmacology and Toxicology Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Lucie Smutna
- Department of Pharmacology and Toxicology Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Rajamanikkam Kamaraj
- Department of Pharmacology and Toxicology Faculty of Pharmacy in Hradec Kralove Charles University Hradec Kralove Czech Republic
| | - Pavel Soucek
- Toxicogenomics Unit National Institute of Public Health Prague Czech Republic
- Laboratory of Pharmacogenomics Biomedical Center Faculty of Medicine in Pilsen Charles University Pilsen Czech Republic
| | - Radim Vrzal
- Department of Cell Biology and Genetics Faculty of Science Palacky University Olomouc Czech Republic
| | - Zdenek Dvorak
- Department of Cell Biology and Genetics Faculty of Science Palacky University Olomouc Czech Republic
| |
Collapse
|
6
|
Gaebler AJ, Finner‐Prével M, Lammertz S, Schaffrath S, Eisner P, Stöhr F, Röcher E, Winkler L, Kaleta P, Lenzen L, Augustin M, Hovancakova J, Schwemmer L, Stormanns E, Keskin F, Hendricks F, Paulzen M, Gründer G, Schneider F, Mathiak K, Augustin M, Cordes J, Demirel E, Dielentheis T, Dreher J, Eisner P, Finner‐Prével M, Gaebler AJ, Gründer G, Hendricks F, Hovancakova J, Kaleta P, Keskin F, Kirchner M, Kirner‐Veselinovic A, Lammertz S, Lange C, Larcher F, Lenzen LM, Mathiak K, Meisenzahl‐Lechner E, Muysers J, Neff A, Paulzen M, Plum M, Röcher E, Ruttmann A, Schaffrath S, Schneider F, Schwemmer L, Stöhr F, Stormanns E, Trauzeddel A, Winkler L. Vitamin D’s negative impact on antipsychotic drug exposure may counteract its potential benefits in schizophrenia. Br J Clin Pharmacol 2022; 88:3193-3200. [DOI: 10.1111/bcp.15223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 10/19/2021] [Accepted: 12/22/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- Arnim Johannes Gaebler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Michelle Finner‐Prével
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Sarah Lammertz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Sabrina Schaffrath
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Patrick Eisner
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Felix Stöhr
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Erik Röcher
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Lina Winkler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Peter Kaleta
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Laura Lenzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Marc Augustin
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
- Protestant University of Applied Sciences Bochum Germany
| | - Jana Hovancakova
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Lara Schwemmer
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Eva Stormanns
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | | | | | - Michael Paulzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
- Alexianer Hospital Aachen Germany
| | - Gerhard Gründer
- Central Institute of Mental Health, Department of Molecular Neuroimaging, Medical Faculty Mannheim University of Heidelberg Mannheim Germany
| | - Frank Schneider
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
- University Hospital Düsseldorf Germany
| | - Klaus Mathiak
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Marc Augustin
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
- Protestant University of Applied Sciences Bochum Germany
| | - Joachim Cordes
- LVR Klinikum Düsseldorf University Hospital Düsseldorf Germany
| | - Emir Demirel
- LVR Klinikum Düsseldorf University Hospital Düsseldorf Germany
| | | | | | - Patrick Eisner
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Michelle Finner‐Prével
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Arnim Johannes Gaebler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Gerhard Gründer
- Central Institute of Mental Health, Department of Molecular Neuroimaging, Medical Faculty Mannheim University of Heidelberg Mannheim Germany
| | | | - Jana Hovancakova
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Peter Kaleta
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | | | | | - André Kirner‐Veselinovic
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Sarah Lammertz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | | | - Federico Larcher
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Laura Marianne Lenzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Klaus Mathiak
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | | | | | | | - Michael Paulzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
- Alexianer Hospital Aachen Germany
| | | | - Erik Röcher
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Axel Ruttmann
- LVR Klinikum Düsseldorf University Hospital Düsseldorf Germany
| | - Sabrina Schaffrath
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Frank Schneider
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
- University Hospital Düsseldorf Germany
| | - Lara Schwemmer
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Felix Stöhr
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | - Eva Stormanns
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | | | - Lina Winkler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine RWTH Aachen Germany
- JARA ‐ Translational Brain Medicine Aachen Germany
| | | |
Collapse
|
7
|
Grangeon A, Clermont V, Barama A, Gaudette F, Turgeon J, Michaud V. Determination of CYP450 Expression Levels in the Human Small Intestine by Mass Spectrometry-Based Targeted Proteomics. Int J Mol Sci 2021; 22:ijms222312791. [PMID: 34884595 PMCID: PMC8657875 DOI: 10.3390/ijms222312791] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022] Open
Abstract
The human small intestine can be involved in the first-pass metabolism of drugs. Under this condition, members of the CYP450 superfamily are expected to contribute to drug presystemic biotransformation. The aim of this study was to quantify protein expression levels of 16 major CYP450 isoforms in tissue obtained from nine human organ donors in seven subsections of the small intestine, i.e., duodenum (one section, N = 7 tissue samples), jejunum (three subsections (proximal, mid and distal), N = 9 tissue samples) and ileum (three subsections, (proximal, mid and distal), N = 9 tissue samples), using liquid chromatography tandem mass spectrometry (LC-MS/MS) based targeted proteomics. CYP450 absolute protein expression levels were compared to mRNA levels and enzyme activities by using established probe drugs. Proteins corresponding to seven of sixteen potential CYP450 isoforms were detected and quantified in various sections of the small intestine: CYP2C9, CYP2C19, CYP2D6, CYP2J2, CYP3A4, CYP3A5 and CYP4F2. Wide inter-subject variability was observed, especially for CYP2D6. CYP2C9 (p = 0.004) and CYP2C19 (p = 0.005) expression levels decreased along the small intestine. From the duodenum to the ileum, CYP2J2 (p = 0.001) increased, and a trend was observed for CYP3A5 (p = 0.13). CYP3A4 expression was higher in the jejunum than in the ileum (p = 0.03), while CYP4F2 expression was lower in the duodenum compared to the jejunum and the ileum (p = 0.005). CYP450 protein levels were better correlated with specific isoform activities than with mRNA levels. This study provides new data on absolute CYP450 quantification in human small intestine that could improve physiologically based pharmacokinetic models. These data could better inform drug absorption profiles while considering the regional expression of CYP450 isoforms.
Collapse
Affiliation(s)
- Alexia Grangeon
- CRCHUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, 900 St. Denis Street, Montreal, QC H2X 0A9, Canada; (A.G.); (V.C.); (F.G.)
| | - Valérie Clermont
- CRCHUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, 900 St. Denis Street, Montreal, QC H2X 0A9, Canada; (A.G.); (V.C.); (F.G.)
| | - Azemi Barama
- CHUM, Centre Hospitalier de l’Université de Montréal, 1000 St. Denis Street, Montreal, QC H2X 0C1, Canada;
| | - Fleur Gaudette
- CRCHUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, 900 St. Denis Street, Montreal, QC H2X 0A9, Canada; (A.G.); (V.C.); (F.G.)
| | - Jacques Turgeon
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de la Polytechnique, Montreal, QC H3T 1J4, Canada; or
- Precision Pharmacotherapy Research and Development Institute, Tabula Rasa HealthCare, 13485 Veterans Way, Orlando, FL 32827, USA
| | - Veronique Michaud
- CRCHUM, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, 900 St. Denis Street, Montreal, QC H2X 0A9, Canada; (A.G.); (V.C.); (F.G.)
- Faculty of Pharmacy, Université de Montréal, 2940 Chemin de la Polytechnique, Montreal, QC H3T 1J4, Canada; or
- Precision Pharmacotherapy Research and Development Institute, Tabula Rasa HealthCare, 13485 Veterans Way, Orlando, FL 32827, USA
- Correspondence: or
| |
Collapse
|
8
|
Annu K, Yasuda K, Caufield WV, Freeman BB, Schuetz EG. Vitamin D levels do not cause vitamin-drug interactions with dexamethasone or dasatinib in mice. PLoS One 2021; 16:e0258579. [PMID: 34669728 PMCID: PMC8528301 DOI: 10.1371/journal.pone.0258579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/30/2021] [Indexed: 11/28/2022] Open
Abstract
Vitamin D3 (VD3) induces intestinal CYP3A that metabolizes orally administered anti-leukemic chemotherapeutic substrates dexamethasone (DEX) and dasatinib potentially causing a vitamin-drug interaction. To determine the impact of VD3 status on systemic exposure and efficacy of these chemotherapeutic agents, we used VD3 sufficient and deficient mice and performed pharmacokinetic and anti-leukemic efficacy studies. Female C57BL/6J and hCYP3A4 transgenic VD3 deficient mice had significantly lower duodenal (but not hepatic) mouse Cyp3a11 and hCYP3A4 expression compared to VD3 sufficient mice, while duodenal expression of Mdr1a, Bcrp and Mrp4 were significantly higher in deficient mice. When the effect of VD3 status on DEX systemic exposure was compared following a discontinuous oral DEX regimen, similar to that used to treat pediatric acute lymphoblastic leukemia patients, male VD3 deficient mice had significantly higher mean plasma DEX levels (31.7 nM) compared to sufficient mice (12.43 nM) at days 3.5 but not at any later timepoints. Following a single oral gavage of DEX, there was a statistically, but not practically, significant decrease in DEX systemic exposure in VD3 deficient vs. sufficient mice. While VD3 status had no effect on oral dasatinib's area under the plasma drug concentration-time curve, VD3 deficient male mice had significantly higher dasatinib plasma levels at t = 0.25 hr. Dexamethasone was unable to reverse the poorer survival of VD3 sufficient vs. deficient mice to BCR-ABL leukemia. In conclusion, although VD3 levels significantly altered intestinal mouse Cyp3a in female mice, DEX plasma exposure was only transiently different for orally administered DEX and dasatinib in male mice. Likewise, the small effect size of VD3 deficiency on single oral dose DEX clearance suggests that the clinical significance of VD3 levels on DEX systemic exposure are likely to be limited.
Collapse
Affiliation(s)
- Kavya Annu
- Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Integrated Biomedical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Kazuto Yasuda
- Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - William V. Caufield
- Preclinical Pharmacokinetic Shared Resource, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Burgess B. Freeman
- Preclinical Pharmacokinetic Shared Resource, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Erin G. Schuetz
- Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
9
|
Lin YS, Thummel KE, Thompson BD, Totah RA, Cho CW. Sources of Interindividual Variability. Methods Mol Biol 2021; 2342:481-550. [PMID: 34272705 DOI: 10.1007/978-1-0716-1554-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The efficacy, safety, and tolerability of drugs are dependent on numerous factors that influence their disposition. A dose that is efficacious and safe for one individual may result in sub-therapeutic or toxic blood concentrations in others. A significant source of this variability in drug response is drug metabolism, where differences in presystemic and systemic biotransformation efficiency result in variable degrees of systemic exposure (e.g., AUC, Cmax, and/or Cmin) following administration of a fixed dose.Interindividual differences in drug biotransformation have been studied extensively. It is recognized that both intrinsic factors (e.g., genetics, age, sex, and disease states) and extrinsic factors (e.g., diet , chemical exposures from the environment, and the microbiome) play a significant role. For drug-metabolizing enzymes, genetic variation can result in the complete absence or enhanced expression of a functional enzyme. In addition, upregulation and downregulation of gene expression, in response to an altered cellular environment, can achieve the same range of metabolic function (phenotype), but often in a less predictable and time-dependent manner. Understanding the mechanistic basis for variability in drug disposition and response is essential if we are to move beyond the era of empirical, trial-and-error dose selection and into an age of personalized medicine that will improve outcomes in maintaining health and treating disease.
Collapse
Affiliation(s)
- Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
| | - Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Brice D Thompson
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Christi W Cho
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
10
|
Effects of vitamin D on drugs: Response and disposal. Nutrition 2020; 74:110734. [PMID: 32179384 DOI: 10.1016/j.nut.2020.110734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/29/2019] [Accepted: 01/01/2020] [Indexed: 12/11/2022]
Abstract
Vitamin D supplementation and vitamin D deficiency are common in clinical experience and in daily life. Vitamin D not only promotes calcium absorption and immune regulation, but also changes drug effects (pharmacodynamics and adverse reactions) and drug disposal in vivo when combined with various commonly used clinical drugs. The extensive physiological effects of vitamin D may cause synergism effects or alleviation of adverse reactions, and vitamin D's affect on drugs in vivo disposal through drug transporters or metabolic enzymes may also lead to changes in drug effects. Herein, the effects of vitamin D combined with commonly used drugs were reviewed from the perspective of drug efficacy and adverse reactions. The effects of vitamin D on drug transport and metabolism were summarized and analyzed. Hopefully, more attention will be paid to vitamin D supplementation and deficiency in clinical treatment and drug research and development.
Collapse
|
11
|
Turner RM, Pirmohamed M. Statin-Related Myotoxicity: A Comprehensive Review of Pharmacokinetic, Pharmacogenomic and Muscle Components. J Clin Med 2019; 9:jcm9010022. [PMID: 31861911 PMCID: PMC7019839 DOI: 10.3390/jcm9010022] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023] Open
Abstract
Statins are a cornerstone in the pharmacological prevention of cardiovascular disease. Although generally well tolerated, a small subset of patients experience statin-related myotoxicity (SRM). SRM is heterogeneous in presentation; phenotypes include the relatively more common myalgias, infrequent myopathies, and rare rhabdomyolysis. Very rarely, statins induce an anti-HMGCR positive immune-mediated necrotizing myopathy. Diagnosing SRM in clinical practice can be challenging, particularly for mild SRM that is frequently due to alternative aetiologies and the nocebo effect. Nevertheless, SRM can directly harm patients and lead to statin discontinuation/non-adherence, which increases the risk of cardiovascular events. Several factors increase systemic statin exposure and predispose to SRM, including advanced age, concomitant medications, and the nonsynonymous variant, rs4149056, in SLCO1B1, which encodes the hepatic sinusoidal transporter, OATP1B1. Increased exposure of skeletal muscle to statins increases the risk of mitochondrial dysfunction, calcium signalling disruption, reduced prenylation, atrogin-1 mediated atrophy and pro-apoptotic signalling. Rare variants in several metabolic myopathy genes including CACNA1S, CPT2, LPIN1, PYGM and RYR1 increase myopathy/rhabdomyolysis risk following statin exposure. The immune system is implicated in both conventional statin intolerance/myotoxicity via LILRB5 rs12975366, and a strong association exists between HLA-DRB1*11:01 and anti-HMGCR positive myopathy. Epigenetic factors (miR-499-5p, miR-145) have also been implicated in statin myotoxicity. SRM remains a challenge to the safe and effective use of statins, although consensus strategies to manage SRM have been proposed. Further research is required, including stringent phenotyping of mild SRM through N-of-1 trials coupled to systems pharmacology omics- approaches to identify novel risk factors and provide mechanistic insight.
Collapse
|
12
|
Krogstad V, Peric A, Robertsen I, Kringen MK, Wegler C, Angeles PC, Hjelmesæth J, Karlsson C, Andersson S, Artursson P, Åsberg A, Andersson TB, Christensen H. A Comparative Analysis of Cytochrome P450 Activities in Paired Liver and Small Intestinal Samples from Patients with Obesity. Drug Metab Dispos 2019; 48:8-17. [DOI: 10.1124/dmd.119.087940] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022] Open
|
13
|
Role of vitamin D receptor in the regulation of CYP3A gene expression. Acta Pharm Sin B 2019; 9:1087-1098. [PMID: 31867158 PMCID: PMC6900549 DOI: 10.1016/j.apsb.2019.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/28/2019] [Accepted: 03/15/2019] [Indexed: 12/17/2022] Open
Abstract
Vitamin D3 (VD3) is a multifunctional nutrient which can be either synthesized or absorbed from the diet. It plays a pivotal role in systemic calcium and phosphate homeostasis, as well as in various physiological and pathological processes. VD3 is converted to the active form, 1α,25-dihydroxyvitamin D3 (1,25-D3), by cytochrome P450 2R1 (CYP2R1)/CYP27A1 and CYP27B1 sequentially, and deactivated by multiple enzymes including CYP3A4. On the other hand, 1,25-D3 is capable of activating the transcription of CYP3A genes in humans, mice and rats. The vitamin D receptor (VDR)-mediated transactivation of human CYP3A4 and CYP3A5 resembles that known for pregnane X receptor (PXR). Activated VDR forms a heterodimer with retinoid X receptor α (RXRα), recruits co-activators, translocates to the cell nucleus, binds to the specific vitamin D responsive elements (VDRE), and activates the gene transcription. In mice, intestinal Cyp3a11 mRNA levels, but not those of hepatic CYP3As, were induced by in vivo administration of VDR and PXR agonists. In rats, intestinal Cyp3a1 and Cyp3a2 mRNAs were induced by 1,25-D3 or lithocholic acid (LCA), whereas hepatic Cyp3a2, but not Cyp3a1 and Cyp3a9, was modulated to 1,25-D3 treatment. In general, the VDR-mediated regulation of CYP3A presents species and organ specificity.
Collapse
|
14
|
Ramsden D, Fung C, Hariparsad N, Kenny JR, Mohutsky M, Parrott NJ, Robertson S, Tweedie DJ. Perspectives from the Innovation and Quality Consortium Induction Working Group on Factors Impacting Clinical Drug-Drug Interactions Resulting from Induction: Focus on Cytochrome 3A Substrates. Drug Metab Dispos 2019; 47:1206-1221. [PMID: 31439574 DOI: 10.1124/dmd.119.087270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
A recent publication from the Innovation and Quality Consortium Induction Working Group collated a large clinical data set with the goal of evaluating the accuracy of drug-drug interaction (DDI) prediction from in vitro data. Somewhat surprisingly, comparison across studies of the mean- or median-reported area under the curve ratio showed appreciable variability in the magnitude of outcome. This commentary explores the possible drivers of this range of outcomes observed in clinical induction studies. While recommendations on clinical study design are not being proposed, some key observations were informative during the aggregate analysis of clinical data. Although DDI data are often presented using median data, individual data would enable evaluation of how differences in study design, baseline expression, and the number of subjects contribute. Since variability in perpetrator pharmacokinetics (PK) could impact the overall DDI interpretation, should this be routinely captured? Maximal induction was typically observed after 5-7 days of dosing. Thus, when the half-life of the inducer is less than 30 hours, are there benefits to a more standardized study design? A large proportion of CYP3A4 inducers were also CYP3A4 inhibitors and/or inactivators based on in vitro data. In these cases, using CYP3A selective substrates has limitations. More intensive monitoring of changes in area under the curve over time is warranted. With selective CYP3A substrates, the net effect was often inhibition, whereas less selective substrates could discern induction through mechanisms not susceptible to inhibition. The latter included oral contraceptives, which raise concerns of reduced efficacy following induction. Alternative approaches for modeling induction, such as applying biomarkers and physiologically based pharmacokinetic modeling (PBPK), are also considered. SIGNIFICANCE STATEMENT: The goal of this commentary is to stimulate discussion on whether there are opportunities to optimize clinical drug-drug interaction study design. The overall aim is to reduce, understand and contextualize the variability observed in the magnitude of induction across reported clinical studies. A large clinical CYP3A induction dataset was collected and further analyzed to identify trends and gaps. Reporting individual victim PK data, characterizing perpetrator PK and including additional PK assessments for mixed-mechanism perpetrators may provide insights into how these factors impact differences observed in clinical outcomes. The potential utility of biomarkers and PBPK modeling are discussed in considering future directions.
Collapse
Affiliation(s)
- Diane Ramsden
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Conrad Fung
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Niresh Hariparsad
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Jane R Kenny
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Michael Mohutsky
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Neil J Parrott
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Sarah Robertson
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| | - Donald J Tweedie
- Alnylam Pharmaceuticals, Cambridge, Massachusetts (D.R.); Vertex Pharmaceuticals, Boston, Massachusetts (C.F., N.H., S.R.); Genentech, South San Francisco, California (J.R.K.); Eli Lilly and Company, Indianapolis, Indiana (M.M.); Roche Innovation Center, Basel, Switzerland (N.J.P.); and Merck & Co., Inc., Kenilworth, New Jersey (D.T.)
| |
Collapse
|
15
|
A Pilot Study towards the Impact of Type 2 Diabetes on the Expression and Activities of Drug Metabolizing Enzymes and Transporters in Human Duodenum. Int J Mol Sci 2019; 20:ijms20133257. [PMID: 31269743 PMCID: PMC6651059 DOI: 10.3390/ijms20133257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/13/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
To characterize effects of type 2 diabetes (T2D) on mRNA expression levels for 10 Cytochromes P450 (CYP450s), two carboxylesterases, and three drug transporters (ABCB1, ABCG2, SLCO2B1) in human duodenal biopsies. To compare drug metabolizing enzyme activities of four CYP450 isoenzymes in duodenal biopsies from patients with or without T2D. mRNA levels were quantified (RT-qPCR) in human duodenal biopsies obtained from patients with (n = 20) or without (n = 16) T2D undergoing a scheduled gastro-intestinal endoscopy. CYP450 activities were determined following incubation of biopsy homogenates with probe substrates for CYP2B6 (bupropion), CYP2C9 (tolbutamide), CYP2J2 (ebastine), and CYP3A4/5 (midazolam). Covariables related to inflammation, T2D, demographic, and genetics were investigated. T2D had no major effects on mRNA levels of all enzymes and transporters assessed. Formation rates of metabolites (pmoles mg protein−1 min−1) determined by LC-MS/MS for CYP2C9 (0.48 ± 0.26 vs. 0.41 ± 0.12), CYP2J2 (2.16 ± 1.70 vs. 1.69 ± 0.93), and CYP3A (5.25 ± 3.72 vs. 5.02 ± 4.76) were not different between biopsies obtained from individuals with or without T2D (p > 0.05). No CYP2B6 specific activity was measured. TNF-α levels were higher in T2D patients but did not correlate with any changes in mRNA expression levels for drug metabolizing enzymes or transporters in the duodenum. T2D did not modulate expression or activity of tested drug metabolizing enzymes and transporters in the human duodenum. Previously reported changes in drug oral clearances in patients with T2D could be due to a tissue-specific disease modulation occurring in the liver and/or in other parts of the intestines.
Collapse
|
16
|
Prytuła A, Cransberg K, Raes A. Drug-metabolizing enzymes CYP3A as a link between tacrolimus and vitamin D in renal transplant recipients: is it relevant in clinical practice? Pediatr Nephrol 2019; 34:1201-1210. [PMID: 30058048 DOI: 10.1007/s00467-018-4030-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 01/08/2023]
Abstract
CYP3A enzymes are involved in the metabolism of calcineurin inhibitor tacrolimus as well as vitamin D. In this review, we summarize the clinical aspects of CYP3A-mediated metabolism of tacrolimus and vitamin D with emphasis on the influence of single-nucleotide polymorphisms on tacrolimus disposition. We describe the utility of 4β hydroxycholesterol as a marker of CYP3A activity. Then, we discuss the possible interaction between calcineurin inhibitors and vitamin D in solid organ transplant recipients. Also, we review other mechanisms which may contribute to side effects of calcineurin inhibitors on bone. Lastly, suggestions for future research and clinical perspectives are discussed.
Collapse
Affiliation(s)
- Agnieszka Prytuła
- Paediatric Nephrology and Rheumatology Department, Ghent University Hospital, C Heymanslaan 10, 9000, Ghent, Belgium.
| | - Karlien Cransberg
- Paediatric Nephrology Department, Erasmus MC- Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Ann Raes
- Paediatric Nephrology and Rheumatology Department, Ghent University Hospital, C Heymanslaan 10, 9000, Ghent, Belgium.,Safepedrug Unit, Ghent, Belgium
| |
Collapse
|
17
|
Naito T, Ohshiro J, Sato H, Torikai E, Suzuki M, Ogawa N, Kawakami J. Relationships between concomitant biologic DMARDs and prednisolone administration and blood tacrolimus exposure or serum CYP3A4/5-related markers in rheumatoid arthritis patients. Clin Biochem 2019; 69:8-14. [DOI: 10.1016/j.clinbiochem.2019.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/19/2019] [Accepted: 05/06/2019] [Indexed: 11/29/2022]
|
18
|
Clermont V, Grangeon A, Barama A, Turgeon J, Lallier M, Malaise J, Michaud V. Activity and mRNA expression levels of selected cytochromes P450 in various sections of the human small intestine. Br J Clin Pharmacol 2019; 85:1367-1377. [PMID: 30817016 DOI: 10.1111/bcp.13908] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022] Open
Abstract
AIMS To characterize mRNA expression levels (17 cytochromes P450) and activity (9 isoforms) of major cytochromes P450 expressed throughout the human small intestine. METHODS Tissue samples were obtained from 9 deceased subjects and intestinal sections (n = 10) were isolated for each subject. Relative mRNA expression levels were determined using quantitative real-time PCR. Intestinal microsomes were prepared from 5 subsections: duodenum, jejunum (proximal and mid-jejunum) and ileum (proximal and mid-ileum) regions. In vitro incubations were performed with various cytochrome P450 probe substrates: bupropion (CYP2B6), repaglinide (CYP2C8), tolbutamide (CYP2C9), S-mephenytoin (CYP2C19), bufuralol (CYP2D6), chlorzoxazone (CYP2E1), ebastine (CYP2J2), midazolam (CYP3A4/5) and lauric acid (CYP4A11). Metabolite formation was assessed using validated liquid chromatography-tandem mass spectrometry assays. RESULTS Cytochrome P450 mRNA levels ranked as follows: CYP3A4 > CYP2C9 > CYP2C19 > CYP2J2 > CYP4F2. Cytochrome P450 mRNA transcripts showed different patterns in their relative expression from 1 region to the other but CYP3A4, CYP2C9, CYP2C19 and CYP2J2 displayed the highest levels of mRNA expression (>5%) in all intestinal sections. Cytochrome P450 activities were greater in proximal part of the small intestine with the jejunum showing the greatest drug-metabolism activity. Spearman's correlation analyses indicated that cytochrome P450 mRNA expressions and corresponding cytochrome P450 activities in the human intestine were moderately associated for CYP2C19, CYP2D6 and CYP4A11 (rs = 0.44-0.56). CONCLUSIONS Our study provides new and additional information on the expression and activities of selected cytochromes P450 in various sections of the human small intestine.
Collapse
Affiliation(s)
- Valérie Clermont
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Alexia Grangeon
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Azemi Barama
- Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Jacques Turgeon
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Michel Lallier
- Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Jacques Malaise
- Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Veronique Michaud
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
19
|
Fuhr U, Hsin CH, Li X, Jabrane W, Sörgel F. Assessment of Pharmacokinetic Drug-Drug Interactions in Humans: In Vivo Probe Substrates for Drug Metabolism and Drug Transport Revisited. Annu Rev Pharmacol Toxicol 2018; 59:507-536. [PMID: 30156973 DOI: 10.1146/annurev-pharmtox-010818-021909] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pharmacokinetic parameters of selective probe substrates are used to quantify the activity of an individual pharmacokinetic process (PKP) and the effect of perpetrator drugs thereon in clinical drug-drug interaction (DDI) studies. For instance, oral caffeine is used to quantify hepatic CYP1A2 activity, and oral dagibatran etexilate for intestinal P-glycoprotein (P-gp) activity. However, no probe substrate depends exclusively on the PKP it is meant to quantify. Lack of selectivity for a given enzyme/transporter and expression of the respective enzyme/transporter at several sites in the human body are the main challenges. Thus, a detailed understanding of the role of individual PKPs for the pharmacokinetics of any probe substrate is essential to allocate the effect of a perpetrator drug to a specific PKP; this is a prerequisite for reliably informed pharmacokinetic models that will allow for the quantitative prediction of perpetrator effects on therapeutic drugs, also in respective patient populations not included in DDI studies.
Collapse
Affiliation(s)
- Uwe Fuhr
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Chih-Hsuan Hsin
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Xia Li
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Wafaâ Jabrane
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Fritz Sörgel
- Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany
| |
Collapse
|
20
|
Gao C, Liao MZ, Han LW, Thummel KE, Mao Q. Hepatic Transport of 25-Hydroxyvitamin D 3 Conjugates: A Mechanism of 25-Hydroxyvitamin D 3 Delivery to the Intestinal Tract. Drug Metab Dispos 2018; 46:581-591. [PMID: 29467214 PMCID: PMC5896369 DOI: 10.1124/dmd.117.078881] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022] Open
Abstract
Vitamin D3 is an important prohormone critical for maintaining calcium and phosphate homeostasis in the body and regulating drug-metabolizing enzymes and transporters. 25-Hydroxyvitamin D3 (25OHD3), the most abundant circulating metabolite of vitamin D3, is further transformed to the biologically active metabolite 1α,25-dihydroxyvitamin D3 (1α,25-(OH)2D3) by CYP27B1 in the kidney and extrarenal tissues, and to nonactive metabolites by other cytochrome P450 enzymes. In addition, 25OHD3 undergoes sulfation and glucuronidation in the liver, forming two major conjugative metabolites, 25OHD3-3-O-sulfate (25OHD3-S) and 25OHD3-3-O-glucuronide (25OHD3-G), both of which were detected in human blood and bile. Considering that the conjugates excreted into the bile may be circulated to and reabsorbed from the intestinal lumen, deconjugated to 25OHD3, and then converted to 1α,25-(OH)2D3, exerting local intestinal cellular effects, it is crucial to characterize enterohepatic transport mechanisms of 25OHD3-S and 25OHD3-G, and thereby understand and predict mechanisms of interindividual variability in mineral homeostasis. In the present study, with plasma membrane vesicle and cell-based transport studies, we showed that 25OHD3-G is a substrate of multidrug resistance proteins 2 and 3, OATP1B1, and OATP1B3, and that 25OHD3-S is probably a substrate of breast cancer resistance protein, OATP2B1, and OATP1B3. We also demonstrated sinusoidal and canalicular efflux of both conjugates using sandwich-cultured human hepatocytes. Given substantial expression of these transporters in liver hepatocytes and intestinal enterocytes, this study demonstrates for the first time that transporters could play important roles in the enterohepatic circulation of 25OHD3 conjugates, providing an alternative pathway of 25OHD3 delivery to the intestinal tract, which could be critical for vitamin D receptor-dependent gene regulation in enterocytes.
Collapse
Affiliation(s)
- Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Lyrialle W Han
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Allegra S, Cusato J, De Francia S, Arduino A, Longo F, Pirro E, Massano D, De Nicolò A, Piga A, D'Avolio A. Role of CYP24A1, VDR and GC gene polymorphisms on deferasirox pharmacokinetics and clinical outcomes. THE PHARMACOGENOMICS JOURNAL 2017; 18:506-515. [PMID: 29160302 DOI: 10.1038/tpj.2017.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/01/2017] [Accepted: 08/04/2017] [Indexed: 02/06/2023]
Abstract
β-Thalassemia patients develop deficiency in vitamin D absorption and liver hydroxylation, resulting in extremely low calcitriol levels. We explored the role of single-nucleotide polymorphisms (SNPs) involved in vitamin D metabolism, transport and activity on deferasirox pharmacokinetics and outcomes (effectiveness trough levels (Ctrough) and the area under the curve (AUC) cutoffs of 20 μg ml-1 and 360 μg ml-1 h-1, respectively; nonresponse AUC limit of 250 μg ml-1 h-1). Ninety-nine β-thalassemic patients were enrolled. Drug plasma Ctrough and AUC were measured by the high-performance liquid chromatography system coupled with an ultraviolet determination method. Allelic discrimination for VDR, CYP24A1, CYP27B1 and GC gene SNPs was performed by real-time PCR. CYP24A1 22776 TT significantly influenced Cmin and negatively predicted it in regression analysis. CYP24A1 3999 CC was associated with Ctrough and Cmin and was a negative predictor of Tmax, whereas CYP24A1 8620 GG seemed to have a role in Ctrough, AUC, t1/2 and Cmin, and was an AUC negative predictor factor. Considering treatment outcome, Cdx2 and GC 1296 were retained in regression analysis as AUC efficacy cutoff negative predictors.
Collapse
Affiliation(s)
- S Allegra
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - J Cusato
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - S De Francia
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Italy
| | - A Arduino
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - F Longo
- Department of Paediatrics, Centre for Microcitemie, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Italy
| | - E Pirro
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Italy
| | - D Massano
- Department of Paediatrics, Centre for Microcitemie, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Italy
| | - A De Nicolò
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - A Piga
- Department of Paediatrics, Centre for Microcitemie, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Italy
| | - A D'Avolio
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| |
Collapse
|
22
|
Yan L, Wang Y, Liu J, Nie Y, Zhong XB, Kan Q, Zhang L. Alterations of Histone Modifications Contribute to Pregnane X Receptor-Mediated Induction of CYP3A4 by Rifampicin. Mol Pharmacol 2017; 92:113-123. [PMID: 28546420 DOI: 10.1124/mol.117.108225] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/22/2017] [Indexed: 01/28/2023] Open
Abstract
CYP3A4 is one of the major drug-metabolizing enzymes in human and is responsible for the metabolism of 60% of clinically used drugs. Many drugs are able to induce the expression of CYP3A4, which usually causes drug-drug interactions and adverse drug reactions. This study aims to explore the role of histone modifications in rifampicin-induced expression of CYP3A4 in LS174T cells. We found that the induction of CYP3A4 mRNA (4- to 15-fold) by rifampicin in LS174T cells was associated with increased levels of histone H3 lysine 4 trimethylation (H3K4me3, above 1.8-fold) and H3 acetylation (above 2-fold) and a decreased level of histone H3 lysine 27 trimethylation (H3K27me3, about 50%) in the CYP3A4 promoter. Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Silencing NCOA6 or p300 by short-hairpin RNAs resulted in inhibition of the CYP3A4 induction as well as altered levels of H3K4me3, H3K27me3, or H3 acetylation in the CYP3A4 promoter. Knockdown of pregnane X receptor (PXR) expression not only suppressed the recruitment of NCOA6 and p300 but also abolished the changes caused by rifampicin in H3K4me3, H3K27me3, and H3 acetylation levels in the CYP3A4 promoter. Moreover, rifampicin treatment enhanced the nuclear accumulation and interactions between PXR and NCOA6/p300. In conclusion, we show that the alterations of histone modifications contribute to the PXR-mediated induction of CYP3A4 by rifampicin.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Yiting Wang
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Jingyang Liu
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Yali Nie
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Xiao-Bo Zhong
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Quancheng Kan
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Lirong Zhang
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| |
Collapse
|
23
|
Hohmann N, Kreuter R, Blank A, Weiss J, Burhenne J, Haefeli WE, Mikus G. Autoinhibitory properties of the parent but not of the N-oxide metabolite contribute to infusion rate-dependent voriconazole pharmacokinetics. Br J Clin Pharmacol 2017; 83:1954-1965. [PMID: 28370390 DOI: 10.1111/bcp.13297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/21/2017] [Accepted: 03/26/2017] [Indexed: 12/23/2022] Open
Abstract
AIMS The pharmacokinetics of voriconazole show a nonlinear dose-exposure relationship caused by inhibition of its own CYP3A-dependent metabolism. Because the magnitude of autoinhibition also depends on voriconazole concentrations, infusion rate might modulate voriconazole exposure. The impact of four different infusion rates on voriconazole pharmacokinetics was investigated. METHODS Twelve healthy participants received 100 mg voriconazole intravenous over 4 h, 400 mg over 6 h, 4 h, and 2 h in a crossover design. Oral midazolam (3 μg) was given at the end of infusion. Blood and urine samples were collected up to 48 h. Voriconazole and its N-oxide metabolite were quantified using high-performance liquid chromatography coupled to tandem mass spectrometry. Midazolam estimated metabolic clearance (eCLmet) was calculated using a limited sampling strategy. Voriconazole-N-oxide inhibition of cytochrome P450 (CYP) isoforms 2C19 and 3A4 were assessed with the P450-Glo luminescence assay. RESULTS Area under the concentration-time curve for 400 mg intravenous voriconazole was 16% (90% confidence interval: 12-20%) lower when administered over 6 h compared to 2 h infusion. Dose-corrected area under the concentration-time curve for 100 mg over 4 h was 34% lower compared to 400 mg over 4 h. Midazolam eCLmet was 516 ml min-1 (420-640) following 100 mg 4 h-1 voriconazole, 152 ml min-1 (139-166) for 400 mg 6 h-1 , 192 ml min-1 (167-220) for 400 mg 4 h-1 , and 202 ml min-1 (189-217) for 400 mg 2 h-1 . Concentration giving 50% CYP inhibition of voriconazole N-oxide was 146 ± 23 μmol l-1 for CYP3A4, and 40.2 ± 4.2 μmol l-1 for CYP2C19. CONCLUSIONS Voriconazole pharmacokinetics is modulated by infusion rate, an autoinhibitory contribution voriconazole metabolism by CYP3A and 2C19 and to a lesser extent its main N-oxide metabolite for CYP2C19. To avoid reduced exposure, the infusion rate should be 2 h.
Collapse
Affiliation(s)
- Nicolas Hohmann
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Rebecca Kreuter
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Antje Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| |
Collapse
|
24
|
Pharmacokinetics, Pharmacodynamics and Pharmacogenomics of Immunosuppressants in Allogeneic Haematopoietic Cell Transplantation: Part I. Clin Pharmacokinet 2016; 55:525-50. [PMID: 26563168 DOI: 10.1007/s40262-015-0339-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although immunosuppressive treatments and target concentration intervention (TCI) have significantly contributed to the success of allogeneic haematopoietic cell transplantation (alloHCT), there is currently no consensus on the best immunosuppressive strategies. Compared with solid organ transplantation, alloHCT is unique because of the potential for bidirectional reactions (i.e. host-versus-graft and graft-versus-host). Postgraft immunosuppression typically includes a calcineurin inhibitor (cyclosporine or tacrolimus) and a short course of methotrexate after high-dose myeloablative conditioning, or a calcineurin inhibitor and mycophenolate mofetil after reduced-intensity conditioning. There are evolving roles for the antithymyocyte globulins (ATGs) and sirolimus as postgraft immunosuppression. A review of the pharmacokinetics and TCI of the main postgraft immunosuppressants is presented in this two-part review. All immunosuppressants are characterized by large intra- and interindividual pharmacokinetic variability and by narrow therapeutic indices. It is essential to understand immunosuppressants' pharmacokinetic properties and how to use them for individualized treatment incorporating TCI to improve outcomes. TCI, which is mandatory for the calcineurin inhibitors and sirolimus, has become an integral part of postgraft immunosuppression. TCI is usually based on trough concentration monitoring, but other approaches include measurement of the area under the concentration-time curve (AUC) over the dosing interval or limited sampling schedules with maximum a posteriori Bayesian personalization approaches. Interpretation of pharmacodynamic results is hindered by the prevalence of studies enrolling only a small number of patients, variability in the allogeneic graft source and variability in postgraft immunosuppression. Given the curative potential of alloHCT, the pharmacodynamics of these immunosuppressants deserves to be explored in depth. Development of sophisticated systems pharmacology models and improved TCI tools are needed to accurately evaluate patients' exposure to drugs in general and to immunosuppressants in particular. Sequential studies, first without and then with TCI, should be conducted to validate the clinical benefit of TCI in homogenous populations; randomized trials are not feasible, because there are higher-priority research questions in alloHCT. In Part I of this article, we review the alloHCT process to facilitate optimal design of pharmacokinetic and pharmacodynamics studies. We also review the pharmacokinetics and TCI of calcineurin inhibitors and methotrexate.
Collapse
|
25
|
Meng C, Wei Z, Zhang Y, Yan L, He H, Zhang L, Xing Q. Regulation of cytochrome P450 3A4 by small vault RNAb derived from the non-coding vault RNA1 of multidrug resistance-linked vault particle. Mol Med Rep 2016; 14:387-93. [PMID: 27177257 DOI: 10.3892/mmr.2016.5228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 01/25/2016] [Indexed: 11/06/2022] Open
Abstract
Cytochrome P450 3A4 (CYP3A4) is the most abundant cytochrome P450 enzyme in human liver and intestine, contributing to the metabolism of >60% of all pharmaceuticals. The expression levels of hepatic CYP3A4 show great inter‑individual variation. However, the detailed regulatory mechanism of CYP3A4 expression has remained largely elusive. It has been reported that the non‑coding RNA small vault (sv)RNAb targets the 3' untranslated region (3'UTR) of CYP3A4 in MCF7 cells. However, to date, the role of svRNAb has not been examined in human liver tissue and hepatic cell lines such as HepG2, which was the aim of the present study. Polymerase chain reaction analysis indicated that the expression of CYP3A4 was significantly different within a study cohort (n=19). In addition, a significant negative correlation was observed between svRNAb and CYP3A4 expression in human liver tissue samples. Furthermore, a luciferase assay on HepG2 cells verified that svRNAb directly targets CYP3A4 and regulates the expression of CYP3A4 by interacting with the validated binding sites of the CYP3A4 3'UTR. The results provided insight into the variation of the expression of CYP3A4 among individuals and provided a novel method for the adjustment of personalized drug treatment. Furthermore, the present study provided a mechanism of the regulatory role of svRNAb in multidrug‑resistant cells.
Collapse
Affiliation(s)
- Chunjie Meng
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Zhiyun Wei
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yiting Zhang
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Liang Yan
- Department of Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan 450006, P.R. China
| | - Hang He
- Department of Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan 450006, P.R. China
| | - Lirong Zhang
- Department of Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan 450006, P.R. China
| | - Qinghe Xing
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
26
|
Hohmann N, Haefeli WE, Mikus G. CYP3A activity: towards dose adaptation to the individual. Expert Opin Drug Metab Toxicol 2016; 12:479-97. [PMID: 26950050 DOI: 10.1517/17425255.2016.1163337] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Co-medication, gene polymorphisms and co-morbidity are main causes for high variability in expression and function of the CYP3A isoenzymes. Pharmacokinetic variability is a major source of interindividual variability of drug effect and response of CYP3A substrates. While CYP3A genotyping is of limited use, direct testing of enzyme function ('phenotyping') may be more promising to achieve individualized dosing of CYP3A substrates. AREAS COVERED We will discuss available phenotyping strategies for CYP3A isoenzymes and causes of intra- and interindividual variability of CYP3A. The impact of phenotyping on the dose selection and pharmacokinetics of CYP3A substrates (docetaxel, irinotecan, tyrosine kinase inhibitors, ciclosporin, tacrolimus) are reviewed. Pubmed searches were conducted during March-November 2015 to retrieve articles related to CYP3A enzyme, phenotyping, drug interactions with CYP3A probe substrates, and phenotyping-guided dosing algorithms. EXPERT OPINION While ample data is available on the choice appropriate phenotyping drugs (midazolam, alfentanil, aplrazolam, buspirone, triazolam), less clinical trial data is available concerning strategies to usefully guide dosing in the clinical practice. Implementation into the clinical routine necessitates further research to identify (1) an easy-to-use and cheap test for CYP3A activity that (2) adequately predicts drug exposure to (3) allow a sound decision on dose adaptation and hence (4) improve clinical outcome and/or reduce the intensity or frequency of adverse drug effects.
Collapse
Affiliation(s)
- Nicolas Hohmann
- a Department of Clinical Pharmacology and Pharmacoepidemiology , University Hospital Heidelberg , Heidelberg , Germany
| | - Walter E Haefeli
- a Department of Clinical Pharmacology and Pharmacoepidemiology , University Hospital Heidelberg , Heidelberg , Germany
| | - Gerd Mikus
- a Department of Clinical Pharmacology and Pharmacoepidemiology , University Hospital Heidelberg , Heidelberg , Germany
| |
Collapse
|
27
|
Ekström L, Skilving I, Ovesjö ML, Aklillu E, Nylén H, Rane A, Diczfalusy U, Björkhem-Bergman L. miRNA-27b levels are associated with CYP3A activity in vitro and in vivo. Pharmacol Res Perspect 2015; 3:e00192. [PMID: 27022466 PMCID: PMC4777245 DOI: 10.1002/prp2.192] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/17/2015] [Indexed: 01/21/2023] Open
Abstract
Previous in vitro studies have shown that microRNA‐27b (miR‐27b) may regulate mRNA levels of CYP3A4, vitamin D receptor (VDR), and Peroxisome proliferator‐activated receptor α (PPARα) as well as CYP3A4 protein expression and activity. In vitro studies have also shown that vitamin D may affect the expression of CYP3A4. The primary aim of this pilot study was to investigate the association between miR‐27b and CYP3A expression and activity. The secondary aim was to investigate the association between 25‐hydroxy vitamin D in serum and CYP3A activity. Mi‐RNA‐27b was quantified using real‐time PCR in serum samples (n = 28) and 25‐hydroxyvitamin D was measured and correlated with the levels of the endogenous CYP3A activity marker 4β‐hydroxycholesterol. In addition, the correlation between miR‐27b and CYP3A activity, measured by dextromethorphan N‐demethylation and 6β‐hydroxylation of testosterone and the gene expression of CYP3A4, VDR and PPARα were assessed in 20 human liver samples. A significant association between circulatory miR‐27b levels and 4β‐hydroxycholesterol ratio was found; P = 0.04, and between hepatic miR‐27b levels and CYP3A activity, measured by dextromethorphan N‐demethylation in human liver (P = 0.04). There was no association between hepatic miR‐27b and mRNA levels of CYP3A4, VDR or PPARα. There was a significant association between serum 25‐hydroxyvitamin D levels and 4β‐hydroxycholesterol ratio, P = 0.002. In conclusion, this pilot‐study supports the hypothesis that miR‐27b levels as well as 25‐hydroxyvitamin D may affect CYP3A activity in vivo. The results indicate that miR‐27b exerts its inhibitory effect on a translational level rather than affecting mRNA levels.
Collapse
Affiliation(s)
- Lena Ekström
- Division of Clinical Pharmacology Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Ilona Skilving
- Division of Clinical Pharmacology Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Marie-Louise Ovesjö
- Division of Clinical Pharmacology Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Eleni Aklillu
- Division of Clinical Pharmacology Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Hanna Nylén
- Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Anders Rane
- Division of Clinical Pharmacology Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Ulf Diczfalusy
- Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden
| | - Linda Björkhem-Bergman
- Division of Clinical Microbiology Department of Laboratory Medicine Karolinska Institutet Karolinska University Hospital, Huddinge SE-141 86 Stockholm Sweden; Palliative Home Care and Hospice Ward ASIH Stockholm Södra Långbro Park Bergtallsvägen 12SE-125 59 Älvsjö Sweden
| |
Collapse
|
28
|
Shi X, Wang L, Du S, Wang H, Feng T, Jin T, Kang L. Genetic polymorphism of pharmacogenomic VIP variants in the Deng people from the Himalayas in Southeast Tibet. Biomarkers 2015; 20:275-86. [PMID: 26329523 DOI: 10.3109/1354750x.2015.1068859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Little is known about polymorphic distribution of pharmacogenes among ethnicities, including the Deng people. In this study, we recruited 100 unrelated, healthy Deng people and genotyped them with respect to 76 different single-nucleotide polymorphisms by the PharmGKB database. Our results first indicated that the polymorphic distribution of pharmacogenes of the Deng people is most similar to CHD, suggesting that Deng people have a closest genetic relationship with CHD. Our data will enrich the database of pharmacogenomics and provide a theoretical basis for safer drug administration and individualized treatment plans, promoting the development of personalized medicine.
Collapse
Affiliation(s)
- Xugang Shi
- a Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University , Xianyang , Shaanxi , China
| | | | | | | | | | | | | |
Collapse
|
29
|
Turner RM, Park BK, Pirmohamed M. Parsing interindividual drug variability: an emerging role for systems pharmacology. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:221-41. [PMID: 25950758 PMCID: PMC4696409 DOI: 10.1002/wsbm.1302] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/08/2015] [Accepted: 04/15/2015] [Indexed: 12/25/2022]
Abstract
There is notable interindividual heterogeneity in drug response, affecting both drug efficacy and toxicity, resulting in patient harm and the inefficient utilization of limited healthcare resources. Pharmacogenomics is at the forefront of research to understand interindividual drug response variability, but although many genotype-drug response associations have been identified, translation of pharmacogenomic associations into clinical practice has been hampered by inconsistent findings and inadequate predictive values. These limitations are in part due to the complex interplay between drug-specific, human body and environmental factors influencing drug response and therefore pharmacogenomics, whilst intrinsically necessary, is by itself unlikely to adequately parse drug variability. The emergent, interdisciplinary and rapidly developing field of systems pharmacology, which incorporates but goes beyond pharmacogenomics, holds significant potential to further parse interindividual drug variability. Systems pharmacology broadly encompasses two distinct research efforts, pharmacologically-orientated systems biology and pharmacometrics. Pharmacologically-orientated systems biology utilizes high throughput omics technologies, including next-generation sequencing, transcriptomics and proteomics, to identify factors associated with differential drug response within the different levels of biological organization in the hierarchical human body. Increasingly complex pharmacometric models are being developed that quantitatively integrate factors associated with drug response. Although distinct, these research areas complement one another and continual development can be facilitated by iterating between dynamic experimental and computational findings. Ultimately, quantitative data-derived models of sufficient detail will be required to help realize the goal of precision medicine.
Collapse
Affiliation(s)
- Richard M Turner
- The Wolfson Centre for Personalised Medicine, Institute for Translational Medicine, University of Liverpool, Liverpool, UK
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute for Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
30
|
No impact of vitamin D on the CYP3A biomarker 4β-hydroxycholesterol in patients with abnormal glucose regulation. PLoS One 2015; 10:e0121984. [PMID: 25835492 PMCID: PMC4383380 DOI: 10.1371/journal.pone.0121984] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/04/2015] [Indexed: 01/21/2023] Open
Abstract
Purpose To investigate the effect of vitamin D3 on hepatic Cytochrome P450 enzyme (CYP) 3A4 in patients with abnormal glucose regulation using the endogenous marker 4β-hydroxycholesterol (4β-OHC):cholesterol ratio. Methods The present study took advantage of a trial primarily aiming to investigate the effect of vitamin D3 on beta cell function and insulin sensitivity in patients with abnormal glucose regulation. 44 subjects were randomized to receive vitamin D3, 30000 IU given orally once weekly or placebo for 8 weeks. The two sample t-test was used to test the means of the intra-individual differences of 4β-OHC:cholesterol ratio between the two groups. Results Mean (SD) 4β-OHC in the whole group of patients before and after the intervention was 26 (11) ng/ml and 26 (12). Mean (SD) 4β-OHC:cholesterol ratio in the whole group of patients before and after the intervention was 0.12 (0.046) and 0.13 (0.047). In the Vitamin D group mean (SD) serum 25-OH-vitamin D3 increased from 46 (16) to 85nM (13) during the corresponding time period. To investigate the impact of vitamin D3 on hepatic CYP3A4 we calculated the mean intra-individual differences in 4β-OHC:cholesterol ratio (delta 4β-OHC:cholesterol ratio) before versus after the intervention in the two treatment groups. The difference (95% CI) between delta 4β-OHC:cholesterol ratio in the control group and intervention group was -0.0010 (-0.0093, 0.0072), a difference being not statistically significant (p = 0.80). Conclusions We provide further evidence that vitamin D3 may not substantially affect hepatic CYP3A4. This does not exclude the possibility of an impact of intestinal first-pass metabolism of orally administered drugs which should be investigated. Trial Registration ClinicalTrials.gov NCT01497132
Collapse
|
31
|
Betts S, Björkhem-Bergman L, Rane A, Ekström L. Expression of CYP3A4 and CYP3A7 in Human Foetal Tissues and its Correlation with Nuclear Receptors. Basic Clin Pharmacol Toxicol 2015; 117:261-6. [PMID: 25689036 DOI: 10.1111/bcpt.12392] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/09/2015] [Indexed: 12/24/2022]
Abstract
Previous reports have suggested that the nuclear receptors vitamin D receptor (VDR), peroxisome proliferator-activated receptor α (PPARα), pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are involved in the regulation of the drug-metabolizing enzyme cytochrome P450 (CYP) 3A4 expression in adults. The aim of this study was to investigate the gene expression of CYP3A4 and the foetal CYP3A7 in human foetal tissues and their relation to gene expression and genetic variations in the nuclear receptors VDR, PPARα, PXR and CAR. We determined the relative expression of CYP3A4 and CYP3A7 and these nuclear receptors in foetal livers, intestines and adrenals, using quantitative PCR. In addition, the expression of these enzymes was also analysed in adult liver. There was a high interindividual variability in CYP3A4 and CYP3A7, 49 times and 326 times, respectively. Both CYP3A4 and CYP3A7 had the highest expression in the liver. There were significant correlations (p < 0.001) between the nuclear receptors studied and the expression of CYP3A4 and CYP3A7 in foetal liver, as well as the expression of CYP3A4 in foetal intestine. Polymorphisms in the VDR gene, rs1544410 and rs1523130 (TaqI), in the PXR gene, rs1523130, and in the PPARα gene, rs4253728, were not correlated with CYP3A4 or CYP3A7 expression. However, C-homozygous individuals of the TaqI VDR polymorphism had 60% lower VDR gene expression (p < 0.05), than individuals carrying one or two T alleles. In conclusion, differences in the expression of nuclear receptors might determine the variability in CYP3A4 and CYP3A7 expression observed in foetal liver.
Collapse
Affiliation(s)
- Stina Betts
- Division of Clinical Pharmacology, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Linda Björkhem-Bergman
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anders Rane
- Division of Clinical Pharmacology, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lena Ekström
- Division of Clinical Pharmacology, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
32
|
Prakash C, Zuniga B, Song CS, Jiang S, Cropper J, Park S, Chatterjee B. Nuclear Receptors in Drug Metabolism, Drug Response and Drug Interactions. NUCLEAR RECEPTOR RESEARCH 2015; 2:101178. [PMID: 27478824 PMCID: PMC4963026 DOI: 10.11131/2015/101178] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Orally delivered small-molecule therapeutics are metabolized in the liver and intestine by phase I and phase II drug-metabolizing enzymes (DMEs), and transport proteins coordinate drug influx (phase 0) and drug/drug-metabolite efflux (phase III). Genes involved in drug metabolism and disposition are induced by xenobiotic-activated nuclear receptors (NRs), i.e. PXR (pregnane X receptor) and CAR (constitutive androstane receptor), and by the 1α, 25-dihydroxy vitamin D3-activated vitamin D receptor (VDR), due to transactivation of xenobiotic-response elements (XREs) present in phase 0-III genes. Additional NRs, like HNF4-α, FXR, LXR-α play important roles in drug metabolism in certain settings, such as in relation to cholesterol and bile acid metabolism. The phase I enzymes CYP3A4/A5, CYP2D6, CYP2B6, CYP2C9, CYP2C19, CYP1A2, CYP2C8, CYP2A6, CYP2J2, and CYP2E1 metabolize >90% of all prescription drugs, and phase II conjugation of hydrophilic functional groups (with/without phase I modification) facilitates drug clearance. The conjugation step is mediated by broad-specificity transferases like UGTs, SULTs, GSTs. This review delves into our current understanding of PXR/CAR/VDR-mediated regulation of DME and transporter expression, as well as effects of single nucleotide polymorphism (SNP) and epigenome (specified by promoter methylation, histone modification, microRNAs, long non coding RNAs) on the expression of PXR/CAR/VDR and phase 0-III mediators, and their impacts on variable drug response. Therapeutic agents that target epigenetic regulation and the molecular basis and consequences (overdosing, underdosing, or beneficial outcome) of drug-drug/drug-food/drug-herb interactions are also discussed. Precision medicine requires understanding of a drug's impact on DME and transporter activity and their NR-regulated expression in order to achieve optimal drug efficacy without adverse drug reactions. In future drug screening, new tools such as humanized mouse models and microfluidic organs-on-chips, which mimic the physiology of a multicellular environment, will likely replace the current cell-based workflow.
Collapse
Affiliation(s)
- Chandra Prakash
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- William Carey University College of Osteopathic Medicine, 498 Tucsan Ave, Hattiesburg, Mississipi 39401
| | - Baltazar Zuniga
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- University of Texas at Austin, 2100 Comal Street, Austin, Texas 78712
| | - Chung Seog Song
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Shoulei Jiang
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Jodie Cropper
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Sulgi Park
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Bandana Chatterjee
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- South Texas Veterans Health Care System, Audie L Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, Texas 78229
| |
Collapse
|
33
|
Nylén H, Björkhem-Bergman L, Ekström L, Roh HK, Bertilsson L, Eliasson E, Lindh JD, Diczfalusy U. Plasma levels of 25-hydroxyvitamin D3 and in vivo markers of cytochrome P450 3A activity in Swedes and Koreans: effects of a genetic polymorphism and oral contraceptives. Basic Clin Pharmacol Toxicol 2014; 115:366-71. [PMID: 24655660 DOI: 10.1111/bcpt.12230] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/26/2014] [Indexed: 01/21/2023]
Abstract
In vitro studies have shown that vitamin D may induce several cytochrome P450 (CYP) enzymes in general and CYP3A4 in particular. The primary aim of this study was to investigate the relationship between plasma levels of 25-hydroxyvitamin D3 and suggested in vivo markers of CYP3A activity in healthy volunteers from Sweden and Korea. Plasma concentrations of 25-hydroxyvitamin D3 were analysed in samples from three previously performed studies, and the correlation between these levels and suggested in vivo markers of CYP3A activity was investigated by means of nonparametric correlation. In addition, we studied the modulating effects of three vitamin D receptor promoter polymorphisms on the association between 25-hydroxyvitamin D3 and CYP3A enzyme activity in Swedish subjects. The plasma levels of 25-hydroxyvitamin D3 were not significantly associated with CYP3A phenotypes in any of the three studies, but after accounting for the vitamin D receptor polymorphism rs4516035, there was a significant positive association between 25-hydroxyvitamin D3 and CYP3A activity (p = 0.004). Swedes (n = 65) had significantly higher 25-hydroxyvitamin D3 levels than Koreans (n = 67), 75 nM compared with 31 nM (p < 0.001). Swedish women taking oral contraceptives (OC) (n = 19) had somewhat higher plasma levels of 25-hydroxyvitamin D3 compared with Swedish women not taking oral contraceptives (n = 21), 89 and 72 nM, respectively (p = 0.02). In conclusion, our results suggest that the overall influence on the CYP3A activity by 25-hydroxyvitamin D3 is of marginal importance.
Collapse
Affiliation(s)
- Hanna Nylén
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The efficacy, safety, and tolerability of drugs are dependent on numerous factors that influence their disposition. A dose that is efficacious and safe for one individual may result in sub-therapeutic or toxic blood concentrations in other individuals. A major source of this variability in drug response is drug metabolism, where differences in pre-systemic and systemic biotransformation efficiency result in variable degrees of systemic exposure (e.g., AUC, C max, and/or C min) following administration of a fixed dose.Interindividual differences in drug biotransformation have been studied extensively. It is well recognized that both intrinsic (such as genetics, age, sex, and disease states) and extrinsic (such as diet, chemical exposures from the environment, and even sunlight) factors play a significant role. For the family of cytochrome P450 enzymes, the most critical of the drug metabolizing enzymes, genetic variation can result in the complete absence or enhanced expression of a functional enzyme. In addition, up- and down-regulation of gene expression, in response to an altered cellular environment, can achieve the same range of metabolic function (phenotype), but often in a less reliably predictable and time-dependent manner. Understanding the mechanistic basis for drug disposition and response variability is essential if we are to move beyond the era of empirical, trial-and-error dose selection and into an age of personalized medicine that brings with it true improvements in health outcomes in the therapeutic treatment of disease.
Collapse
Affiliation(s)
- Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
35
|
De Mattia E, Dreussi E, Cecchin E, Toffoli G. Pharmacogenetics of the nuclear hormone receptors: the missing link between environment and drug effects? Pharmacogenomics 2013; 14:2035-54. [DOI: 10.2217/pgs.13.214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In the last decade, genetic variations in ABC/SLC transporters and phase I/II enzymes have raised pharmacogenetic markers as being predictive to the attention of researchers in the field of personalized medicine in oncology. However, it is becoming evident that the sequence variations in these genes cannot address by themselves the sharp interindividual variability in drug effects. Recently, nuclear receptors (NRs), including pregnane X receptor, constitutive androstane receptor, retinoid X receptor, farnesoid X receptor, liver X receptor, vitamin D receptor, peroxisome proliferator-activated receptors and HNF4A, have demonstrated key roles in regulating transporter and metabolic gene expression in response to xeno/endobiotics, as well as antineoplastic drugs. These findings attracted interest to the genetics of the NRs for their possible role in influencing the metabolism and pharmacological profiles of chemotherapeutics. In this review, we aim to summarize the most recent findings in the innovative field of NR pharmacogenetics and findings in how they could integrate with more traditional markers in order to improve drug treatment personalization.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| | - Eva Dreussi
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| |
Collapse
|
36
|
Teft WA, Kim RB. Sunny outlook for personalized medicine: tamoxifen and beyond. Pharmacogenomics 2013; 14:1533-6. [PMID: 24088122 DOI: 10.2217/pgs.13.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Wendy A Teft
- Division of Clinical Pharmacology, Department of Medicine, University of Western Ontario, London Health Sciences Centre-UH, 339 Windermere Road, London, ON N6A 5A5, Canada
| | | |
Collapse
|
37
|
Chaudhry AS, Thirumaran RK, Yasuda K, Yang X, Fan Y, Strom SC, Schuetz EG. Genetic variation in aldo-keto reductase 1D1 (AKR1D1) affects the expression and activity of multiple cytochrome P450s. Drug Metab Dispos 2013; 41:1538-47. [PMID: 23704699 PMCID: PMC4162005 DOI: 10.1124/dmd.113.051672] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/23/2013] [Indexed: 02/02/2023] Open
Abstract
Human liver gene regulatory (Bayesian) network analysis was previously used to identify a cytochrome P450 (P450) gene subnetwork with Aldo-keto reductase 1D1 (AKR1D1) as a key regulatory driver of this subnetwork. This study assessed the biologic importance of AKR1D1 [a key enzyme in the synthesis of bile acids, ligand activators of farnesoid X receptor (FXR), pregnane X receptor (PXR), and constitutive androstane receptor (CAR), known transcriptional regulators of P450s] to hepatic P450 expression. Overexpression of AKR1D1 in primary human hepatocytes led to increased expression of CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2B6. Conversely, AKR1D1 knockdown decreased expression of these P450s. We resequenced AKR1D1 from 98 donor livers and identified a 3'-untranslated region (UTR) (rs1872930) single nucleotide polymorphism (SNP) significantly associated with higher AKR1D1 mRNA expression. AKR1D1 3'-UTR-luciferase reporter studies showed that the variant allele resulted in higher luciferase activity, suggesting that the SNP increases AKR1D1 mRNA stability and/or translation efficiency. Consistent with AKR1D1's putative role as a driver of the P450 subnetwork, the AKR1D1 3'-UTR SNP was significantly associated with increased hepatic mRNA expression of multiple P450s (CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2B6) and CYP3A4, CYP2C8, CYP2C19, and CYP2B6 activities. After adjusting for multiple testing, the association remained significant for AKR1D1, CYP2C9, and CYP2C8 mRNA expression and CYP2C8 activity. These results provide new insights into the variation in expression and activity of P450s that can account for interindividual differences in drug metabolism/efficacy and adverse drug events. In conclusion, we provide the first experimental evidence supporting a role for AKR1D1 as a key genetic regulator of the P450 network.
Collapse
Affiliation(s)
- Amarjit S Chaudhry
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Zheng XE, Lipka S, Li T, Shahzad G, Levine E, Vlacancich R, Takeshige U, Mustacchia P. The relationship of vitamin D status, smoking, and colorectal adenoma: a retrospective study in an ethnically diverse community. J Steroid Biochem Mol Biol 2013; 136:280-3. [PMID: 23000288 DOI: 10.1016/j.jsbmb.2012.09.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 09/05/2012] [Accepted: 09/12/2012] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Circulating 25-hydroxyvitamin D (25OHD) level is suggested to be negatively correlated with risk of colorectal cancer (CRC) and colorectal adenoma (CRA), but most of the epidemiological data were originated amongst Caucasians and African Americans. This study aimed to investigate the relationship between vitamin D status, smoking and CRA in an ethnically diverse community with a high Hispanic density. METHODS In this retrospective study, we included 233 patients who underwent complete colonoscopies from 2009 to 2011, and their serum 25OHD levels in the winter season had been measured. Among them, 65 adenoma cases and 168 adenoma-free controls were identified and evaluated for the association of CRA with smoking, ethnicity and serum 25OHD level using unstratified and stratified multivariate logistic regression analyses. RESULTS In our study participants, the mean serum 25OHD level and the percentage of Hispanics were lower in the adenoma group versus the control group, while no black-white difference was noted in the CRA prevalence. When adjusted for 25OHD level, the lower rate of adenoma in Hispanics compared to non-Hispanics was attenuated and became statistically insignificant. A mild protective effect of vitamin D (6% reduction) on the CRA risk was found significant for active smokers, but not for non-smokers. A detrimental impact of smoking in the CRA risk was only shown among non-Hispanic patients, but not among Hispanics irrespective of vitamin D status. CONCLUSIONS Our data suggest a marked distinction between Hispanics and non-Hispanics in the risk of CRA. The reduced adenoma prevalence among Hispanics vs. non-Hispanics could be partially explained by vitamin D status, cigarette smoking and their interactions. Future larger-sized multi-center studies on vitamin D status and ethnicity, as well as dietary, behavioral, genetic factors and their interactions for CRA and CRC are needed. This article is part of a Special Issue entitled 'Vitamin D Workshop'.
Collapse
Affiliation(s)
- Xi Emily Zheng
- Department of Medicine, Division of Gastroenterology, Nassau University Medical Center, East Meadow, NY 11554, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang Z, Schuetz EG, Xu Y, Thummel KE. Interplay between vitamin D and the drug metabolizing enzyme CYP3A4. J Steroid Biochem Mol Biol 2013; 136:54-8. [PMID: 22985909 PMCID: PMC3549031 DOI: 10.1016/j.jsbmb.2012.09.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/04/2012] [Accepted: 09/07/2012] [Indexed: 12/19/2022]
Abstract
Cytochrome P450 3A4 (CYP3A4) is a multifunctional enzyme involved in both xenobiotic and endobiotic metabolism. This review focuses on two aspects: regulation of CYP3A4 expression by vitamin D and metabolism of vitamin D by CYP3A4. Enterohepatic circulation of vitamin D metabolites and their conjugates will be also discussed. The interplay between vitamin D and CYP3A4 provides new insights into our understanding of how enzyme induction can contribute to vitamin D deficiency. This article is part of a Special Issue entitled 'Vitamin D Workshop'.
Collapse
Affiliation(s)
- Zhican Wang
- Departments of Pharmaceutics, University of Washington, Seattle, WA
| | - Erin G. Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yang Xu
- Departments of Pharmaceutics, University of Washington, Seattle, WA
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, Thousand Oaks, CA
| | | |
Collapse
|
40
|
Lindh JD, Björkhem-Bergman L, Eliasson E. Vitamin D and drug-metabolising enzymes. Photochem Photobiol Sci 2013; 11:1797-801. [PMID: 22903070 DOI: 10.1039/c2pp25194a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Experimental studies on the molecular regulation of human drug metabolism have revealed that vitamin D up-regulates transcription of several key enzymes, such as CYP3A4, through the vitamin D receptor pathway in intestinal and hepatic cells. Recent data suggest that this results in seasonal changes with higher clearance of orally administered drugs during periods with high UV-B radiation and vitamin D levels. Taken together, vitamin D status might contribute to inter- and intraindividual differences in drug metabolism, but the therapeutic impact of these findings remains to be established.
Collapse
Affiliation(s)
- Jonatan D Lindh
- Karolinska Institutet, Department of Laboratory Medicine, Clinical Pharmacology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | |
Collapse
|
41
|
Teft WA, Gong IY, Dingle B, Potvin K, Younus J, Vandenberg TA, Brackstone M, Perera FE, Choi YH, Zou G, Legan RM, Tirona RG, Kim RB. CYP3A4 and seasonal variation in vitamin D status in addition to CYP2D6 contribute to therapeutic endoxifen level during tamoxifen therapy. Breast Cancer Res Treat 2013; 139:95-105. [PMID: 23580071 DOI: 10.1007/s10549-013-2511-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 03/28/2013] [Indexed: 12/11/2022]
Abstract
Tamoxifen is a widely utilized adjuvant anti-estrogen agent for hormone receptor-positive breast cancer, known to undergo CYP2D6-mediated bioactivation to endoxifen. However, little is known regarding additional genetic and non-genetic determinants of optimal endoxifen plasma concentration. Therefore, 196 breast cancer patients on tamoxifen were enrolled in this prospective study over a 24-month period. Blood samples were collected for pharmacogenetic and drug-level analysis of tamoxifen and metabolites. Regression analysis indicated that besides CYP2D6, the recently described CYP3A4*22 genotype, seasonal variation, and concomitant use of CYP2D6-inhibiting antidepressants were significant predictors of endoxifen concentration. Of note, genetic variation explained 33 % of the variability while non-genetic variables accounted for 13 %. Given the proposed notion of a sub-therapeutic endoxifen concentration for predicting breast cancer recurrence, we set the therapeutic threshold at 18 nM, the 20th percentile for endoxifen level among enrolled patients in this cohort. Nearly 70 % of CYP2D6 poor metabolizers as well as extensive metabolizers on potent CYP2D6-inhibiting antidepressants exhibited endoxifen levels below 18 nM, while carriers of CYP3A4*22 were twofold less likely to be in sub-therapeutic range. Unexpectedly, endoxifen levels were 20 % lower during winter months than mean levels across seasons, which was also associated with lower vitamin D levels. CYP3A4*22 genotype along with sunshine exposure and vitamin D status may be unappreciated contributors of tamoxifen efficacy. The identified covariates along with demographic variables were integrated to create an endoxifen concentration prediction algorithm to pre-emptively evaluate the likelihood of individual patients falling below the optimal endoxifen concentration.
Collapse
Affiliation(s)
- Wendy A Teft
- Division of Clinical Pharmacology, Department of Medicine, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Björkhem-Bergman L, Nylén H, Norlin AC, Lindh JD, Ekström L, Eliasson E, Bergman P, Diczfalusy U. Serum levels of 25-hydroxyvitamin D and the CYP3A biomarker 4β-hydroxycholesterol in a high-dose vitamin D supplementation study. Drug Metab Dispos 2013; 41:704-8. [PMID: 23386704 DOI: 10.1124/dmd.113.051136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The primary aim was to study the relationship between individual serum levels of 25-hydroxyvitamin D and 4β-hydroxycholesterol, which is an endogenous biomarker of the drug-metabolizing CYP3A enzymes. In addition, the relationship between this biomarker and inflammation, measured as C-reactive protein (CRP), was investigated. Serum samples were used from a recently performed clinical trial in patients with antibody deficiency or increased susceptibility to respiratory tract infections that were randomized to either placebo or high-dose (4000 IU/day) vitamin D for 12 months. One hundred sixteen patients were included in the final analyses, and serum samples collected 6 months after study start were analyzed. At this time point, 25-hydroxyvitamin D levels were found to range between 10 and 284 nM. Individual levels of 25-hydroxyvitamin D as well as CRP were compared with 4β-hydroxycholesterol levels. In addition, all participants were genotyped for two polymorphisms (Taq1 and Foq1) in the vitamin D receptor gene. There was no significant correlation between individual serum levels of 25-hydroxyvitamin D and 4β-hydroxycholesterol. However, a moderate, but statistically significant, negative correlation between CRP and 4β-hydroxycholesterol levels was observed. This study in patients with highly variable serum levels of 25-hydroxyvitamin D could not reveal any relationship between vitamin D and 4β-hydroxycholesterol, an endogenous biomarker of CYP3A activity. However, the negative correlation between CRP and 4β-hydroxycholesterol supports earlier experimental results that inflammation may suppress hepatic CYP3A activity, a finding of potentially high clinical relevance that warrants further exploration.
Collapse
Affiliation(s)
- Linda Björkhem-Bergman
- Karolinska Institutet, Department of Laboratory Medicine, Divisions of Clinical Pharmacology, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|