1
|
de Leon J, Baldessarini RJ, Balon R, Bilbily J, Caroff SN, Citrome L, Correll CU, Cotes RO, Davis JM, DeLisi LE, Faden J, Freudenreich O, Goldsmith DR, Gurrera R, Josiassen RC, Kane JM, Kelly DL, Keshavan MS, Laitman RS, Lam YWF, Leung JG, Love RC, McCollum B, McGrane IR, Meyer J, Nasrallah HA, Nucifora FC, Rothschild AJ, Rubio JM, Sajatovic M, Sarpal DK, Schoretsanitis G, Shad M, Shelton C, Sher L, Singh B, Surya S, Zarzar TR, Sanz EJ, De las Cuevas C. Letter to the FDA Proposing Major Changes in the US Clozapine Package Insert Supported by Clozapine Experts Worldwide. Part I: A Review of the Pharmacokinetic Literature and Proposed Changes. J Clin Psychopharmacol 2025; 45:179-196. [PMID: 40198781 PMCID: PMC12105979 DOI: 10.1097/jcp.0000000000001987] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/02/2024] [Indexed: 04/10/2025]
Abstract
PURPOSE/BACKGROUND Clozapine was approved in the United States (US) using 1989 regulations and knowledge. After 30 years, many sections of the US package insert (PI) are outdated. METHODS We comprehensively reviewed the literature to propose PI updates. We present the information in 2 articles. In Part I, we focus on basic pharmacology based on 407 relevant articles. Part II focuses on clinical aspects and pharmacovigilance. FINDINGS/RESULTS Based on more recent expectations of Food and Drug Administration regulations, we reviewed clozapine basic pharmacology including the following: 1) clearance, 2) pharmacokinetics and pharmacodynamics, and 3) monitoring tools. We identified 9 major problems in the basic pharmacological sections of the PI including the following: 1) in vivo studies indicate that clozapine is dependent on CYP1A2 for its metabolism, 2) the minor role of CYP2D6 in clozapine metabolism requires removing the PI recommendation to lower clozapine doses in CYP2D6 poor metabolizers, 3) in nontoxic concentrations CYP3A4 has a minor role in clozapine metabolism and potent CYP3A4 inhibitors lack clinically relevant effects, 4) several drug-drug interactions need to be updated based on recent literature, 5) systemic inflammation may decrease clozapine metabolism and increase the risk of clozapine intoxication, 6) obesity may decrease clozapine metabolism, 7) patients of Asian and Indigenous American ancestry need lower clozapine doses, 8) personalized titration and c-reactive protein monitoring should be considered until prospective studies are available, and 9) the half-life section needs to be modified to acknowledge that single dosing at night is frequent in the US. IMPLICATIONS/CONCLUSIONS An improvement in the US clozapine PI may lead to improvement in PIs worldwide.
Collapse
Affiliation(s)
- Jose de Leon
- From the Department of Psychiatry, University of Kentucky, Lexington, KY
- Mental Health Research Center, Eastern State Hospital, Lexington, KY
- Biomedical Research Centre in Mental Health Net (CIBERSAM), Santiago Apostol Hospital, University of the Basque Country, Vitoria, Spain
| | - Ross J. Baldessarini
- International Consortium for Mood & Psychotic Disorders Research, Mailman Research Center, McLean Hospital, Belmont, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Richard Balon
- Departments of Psychiatry and Behavioral Neurosciences and Anesthesiology, Wayne State University, Detroit, MI
| | - John Bilbily
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO
| | - Stanley N. Caroff
- Behavioral Health Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Leslie Citrome
- New York Medical College, Department of Psychiatry and Behavioral Sciences, Valhalla, NY
| | - Christoph U. Correll
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY
- Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Robert O. Cotes
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - John M. Davis
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL
| | - Lynn E. DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA
| | - Justin Faden
- Department of Psychiatry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Oliver Freudenreich
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - David R. Goldsmith
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Ronald Gurrera
- Department of Psychiatry, Harvard Medical School, Boston, MA
- Mental Health Service, VA Boston Healthcare System, Boston, MA
| | | | - John M. Kane
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY
- Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY
| | - Deanna L. Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | | | | | - Y. W. Francis Lam
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | | | - Raymond C. Love
- Department of Practice, Sciences, and Health Outcomes Research, University of Maryland School of Pharmacy, Baltimore, MD
| | | | - Ian R. McGrane
- Department of Pharmacy Practice, University of Montana, Missoula, MT
| | - Jonathan Meyer
- Department of Psychiatry, University of California, San Diego, CA
| | - Henry A. Nasrallah
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Frederick C. Nucifora
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anthony J. Rothschild
- Department of Psychiatry, University of Massachusetts Chan Medical School and UMass Memorial HealthCare, Worcester, MA
| | - Jose M. Rubio
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY
- Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY
| | - Martha Sajatovic
- Department of Psychiatry and of Neurology, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Deepak K. Sarpal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Georgios Schoretsanitis
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY
- Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland
| | - Mujeeb Shad
- Department of Psychiatry, University of Nevada, Las Vegas, NV
| | - Charles Shelton
- From the Department of Psychiatry, University of Kentucky, Lexington, KY
- Eastern State Hospital, Lexington, KY
| | - Leo Sher
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Balwinder Singh
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MI
| | - Sandarsh Surya
- Department of Psychiatry and Health Behavior, Augusta University, Augusta, GA
| | - Theodore R. Zarzar
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Emilio J. Sanz
- Department of Physical Medicine and Pharmacology, School of Medicine, Universidad de La Laguna, Canary Islands, Spain
- Hospital Universitario de Canarias, Tenerife, Spain
| | - Carlos De las Cuevas
- Department of Internal Medicine, Dermatology and Psychiatry, School of Medicine, and Instituto Universitario de Neurociencia (IUNE), University of La Laguna, Canary Islands, Spain
| |
Collapse
|
2
|
Freiberger EC, Thompson MP, Zhang X, Underwood EB, Lynch TL, Jenkins GJ, Wagner DS. Utility of Common In Vitro Systems for Predicting Circulating Metabolites. Drug Metab Dispos 2024; 52:1373-1378. [PMID: 39433437 DOI: 10.1124/dmd.124.001732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024] Open
Abstract
In vitro systems such as cultured hepatocytes are used early in drug development as a proxy for in vivo data to predict metabolites in human and the potential preclinical species. These data support preclinical species selection for toxicity studies as well as provide early evidence for potential active and reactive metabolites that can be generated in human. Although in vivo data would be best to select preclinical species for a given compound, only in vitro systems are available when selecting toxicity study species. However, as with any in vitro system, the correlation to actual in vivo results can be variable. Understanding the reliability of predicting in vivo metabolites from the various available in vitro assays and determining which system may be most predictive would help de-risk drug development teams' selection process. In this manuscript, we address these questions: can in vitro systems predict circulating metabolites? If so, is predictivity quantitative or indicative of what levels may be seen circulating? Of the currently available in vitro systems, is one better than the others at generating predictive metabolites? To address the first two issues (general in vitro/in vivo predictivity, and whether any in vitro/in vivo correlations are quantitative), we used historical data from Abbott/AbbVie to compare in vitro metabolite profiles with metabolite profiles from in vivo absorption, distribution, metabolism, excretion, and clinical studies. In this retrospective analysis of historic metabolite profiling data, in vitro systems predicted ∼50% of circulating metabolites present in vivo, across preclinical species and human, with no correlation between apparent concentrations in vitro versus in vivo. To address the final question, we selected 10 commercially available compounds with published metabolism data and incubated them in five common in vitro systems (microsomes, liver S9, suspension hepatocytes, HμREL cocultured hepatocytes, and hepatocyte spheroids); the new in vitro metabolite profiling data were compared against published in vivo data to determine whether any individual system was more accurate in generating known major human circulating metabolites. Suspension hepatocytes and cocultured hepatocytes marginally outperformed the other systems. Current in vitro systems have value early in development when in vivo studies are not feasible and are required for regulatory filings to support preclinical toxicology species selection but should not be treated as wholly representative of a given drug's in vivo metabolism. SIGNIFICANCE STATEMENT: This is a comprehensive assessment of historic metabolism data quantitating the success rate of in vitro to in vivo predictivity. Reliability of in vitro systems for metabolite profiling is important for early drug development, and understanding predictivity will help give appropriate context to the data. New data were also generated to compare common in vitro liver models to determine whether any could be definitively identified as more predictive of human circulating metabolites than others.
Collapse
|
3
|
Savaryn JP, Coe K, Cerny MA, Colizza K, Moliner P, King L, Ma B, Atherton J, Auclair A, Cancilla MT, Eno M, Jurva U, Yue Q, Zhu SX, Freiberger E, Zhong G, Barlock B, Nachtigall J, Laboureur L, Pusalkar S, Guo R, Niehues M, Hauri S, Carreras ET, Maurer C, Prakash C, Jenkins GJ. The Current State of Biotransformation Science - Industry Survey of In Vitro and In Vivo Practices, Clinical Translation, and Future Trends. Pharm Res 2024; 41:2079-2093. [PMID: 39496990 PMCID: PMC11599300 DOI: 10.1007/s11095-024-03787-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
Embedded within the field of drug metabolism and pharmacokinetics (DMPK), biotransformation is a discipline that studies the origins, disposition, and structural identity of metabolites to provide a comprehensive safety assessment, including the assessment of exposure coverage in toxicological species. Spanning discovery and development, metabolite identification (metID) scientists employ various strategies and tools to address stage-specific questions aimed at guiding the maturation of early chemical matter into drug candidates. During this process, the identity of major (and minor) circulating human metabolites is ascertained to comply with the regulatory requirements such as the Metabolites in Safety Testing (MIST) guidance. Through the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), the "Translatability of MetID In Vitro Systems Working Group" was created within the Translational and ADME Sciences Leadership Group. The remit of this group was to objectively determine how accurate commonly employed in vitro systems have been with respect to prediction of circulating human metabolites, both qualitatively and quantitatively. A survey composed of 34 questions was conducted across 26 pharmaceutical companies to obtain a foundational understanding of current metID practices, preclinically and clinically, as well as to provide perspective on how successful these practices have been at predicting circulating human metabolites. The results of this survey are presented as an initial snapshot of current industry-based metID practices, including our perspective on how a harmonized framework for the conduct of in vitro metID studies could be established. Future perspectives from current practices to emerging advances with greater translational capability are also provided.
Collapse
Affiliation(s)
- John P Savaryn
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA.
| | - Kevin Coe
- J&J, Translational PKPD & Investigational Toxicology, San Diego, CA, USA
| | | | - Kevin Colizza
- GSK, DMPK Disposition and Biotransformation, Collegeville, PA, USA.
| | | | - Lloyd King
- UCB Biopharma, Dept. of DMPK, Slough, UK
| | - Bin Ma
- Genentech, Inc., Department of Drug Metabolism and Pharmacokinetics, South San Francisco, CA, USA
| | - Jim Atherton
- Incyte Research Institute, Translational Sciences, Wilmington, DE, USA
| | - Adam Auclair
- Boehringer Ingelheim Pharmaceuticals, Inc.,Drug Metabolism and Pharmacokinetics, Ridgefield, CT, USA
| | - Mark T Cancilla
- Merck & Co., Inc., Pharmacokinetics, Dynamics, Metabolism, and Bioanalysis, Rahway, NJ, USA
| | - Marsha Eno
- Eisai Inc., Global Drug Metabolism and Pharmacokinetics, Cambridge, MA, USA
| | - Ulrik Jurva
- AstraZeneca, Drug Metabolism and Pharmacokinetics (DMPK), Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Qin Yue
- Gilead Sciences, Inc., Drug Metabolism Dept, Foster City, CA, USA
| | - Sean Xiaochun Zhu
- Takeda Development Center Americas, Inc., Drug Metabolism and Pharmacokinetics & Modeling, Cambridge, MA, USA
| | - Elyse Freiberger
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA
| | - Guo Zhong
- Amgen, Pharmacokinetics and Drug Metabolism Department, South San Francisco, CA, USA
| | | | | | | | | | - Runcong Guo
- Beigene, DMPK, Department of Biology, Shanghai, China
| | - Michael Niehues
- Bayer AG, In Vitro ADME & Isotope Chemistry, Berlin, Germany
| | - Simon Hauri
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ester Tor Carreras
- Novartis Pharma AG, Novartis Institute for Biomedical Research, Basel, Switzerland
| | | | - Chandra Prakash
- DMPK/Clinical Pharmacology, Agios Pharmaceuticals, Cambridge, MA, USA
| | - Gary J Jenkins
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA
| |
Collapse
|
4
|
Ruan CJ, Olmos I, Ricciardi C, Schoretsanitis G, Vincent PD, Anıl Yağcıoğlu AE, Eap CB, Baptista T, Clark SR, Fernandez-Egea E, Kim SH, Lane HY, Leung J, Maroñas Amigo O, Motuca M, Every-Palmer S, Procyshyn RM, Rohde C, Suhas S, Schulte PFJ, Spina E, Takeuchi H, Verdoux H, Correll CU, Molden E, De Las Cuevas C, de Leon J. Exploring low clozapine C/D ratios, inverted clozapine-norclozapine ratios and undetectable concentrations as measures of non-adherence in clozapine patients: A literature review and a case series of 17 patients from 3 studies. Schizophr Res 2024; 268:293-301. [PMID: 37487869 DOI: 10.1016/j.schres.2023.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Up to 1/2 of outpatients prescribed clozapine may be partially/fully non-adherent, based on therapeutic drug monitoring (TDM). Three indices for measuring partial/full non-adherence are proposed a: 1) clozapine concentration/dose (C/D) ratio which drops to half or more of what is expected in the patient; 2) clozapine/norclozapine ratio that becomes inverted; and 3) clozapine concentration that becomes non-detectable. METHODS These 3 proposed indices are based on a literature review and 17 cases of possible non-adherence from 3 samples: 1) an inpatient study in a Chinese hospital, 2) an inpatient randomized clinical trial in a United States hospital, and 3) and a Uruguayan outpatient study. RESULTS The first index of non-adherence is a clozapine C/D ratio which is less than half the ratio corresponding to the patient's specific ancestry group and sex-smoking subgroup. Knowing the minimum therapeutic dose of the patient based on repeated TDM makes it much easier to establish non-adherence. The second index is inverted clozapine/norclozapine ratios in the absence of alternative explanations. The third index is undetectable concentrations. By using half-lives, the chronology of the 3 indices of non-adherence was modeled in two patients: 1) the clozapine C/D ratio dropped to ≥1/2 of what is expected from the patient (around day 2); 2) the clozapine/norclozapine ratio became inverted (around day 3); and 3) the clozapine concentration became undetectable by the laboratory (around days 9-11). CONCLUSION Prospective studies should further explore these proposed clozapine indices in average patients, poor metabolizers (3 presented) and ultrarapid metabolizers (2 presented).
Collapse
Affiliation(s)
- Can-Jun Ruan
- The National Clinical Research Centre for Mental Disorders & Beijing Key Lab of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Ismael Olmos
- Clinical Pharmacology Unit and Pharmacy Department, Vilardebó Hospital, Administración de Servicios de Salud, Montevideo, Uruguay.
| | - Carina Ricciardi
- Clinical Pharmacology Unit and Outpatient Clinic, Vilardebó Hospital, Administración de Servicios de Salud, Montevideo, Uruguay.
| | - Georgios Schoretsanitis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York, USA; Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY, USA.
| | - Philippe D Vincent
- Department of Pharmacy, Institut Universitaire en Santé Mentale de Montréal (IUSMM), Montreal, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Canada; IUSMM Research Center, Montreal, Canada.
| | | | - Chin B Eap
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland.
| | - Trino Baptista
- Department of Physiology, Los Andes University Medical School, Mérida, Venezuela; Medical School, Anáhuac University, Querétaro, Mexico; Neuroorigen, Querétaro, Mexico.
| | - Scott R Clark
- University of Adelaide, Discipline of Psychiatry, Adelaide, Australia.
| | - Emilio Fernandez-Egea
- Department of Psychiatry, University of Cambridge, Cambridge, UK; Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn Hospital, Fulbourn, Cambridge, UK.
| | - Se Hyun Kim
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Hsien-Yuan Lane
- Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan.
| | - Jonathan Leung
- Department of Pharmacy, Mayo Clinic, Rochester, MN, USA.
| | - Olalla Maroñas Amigo
- Genomic Medicine Group, Galician Public Foundation of Genomic Medicine (FPGMX), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain; Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain; Center for Biomedical Research in Rare Diseases Network, Carlos III Health Institute, Madrid, Spain.
| | - Mariano Motuca
- Department of Psychiatry, School of Medicine at Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Susanna Every-Palmer
- Department of Psychological Medicine, University of Otago Wellington, Wellington, New Zealand.
| | - Ric M Procyshyn
- Department of Psychiatry, University of British Columbia, Vancouver, Canada; British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, Canada.
| | - Christopher Rohde
- Department of Affective Disorders, Aarhus University Hospital - Psychiatry, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Satish Suhas
- Department of Psychiatry, National Institute of Mental Health and Neurosciences [NIMHANS], Bangalore, India.
| | - Peter F J Schulte
- Mental Health Services Noord-Holland-Noord, Alkmaar, Netherlands; Dutch Clozapine Collaboration Group, Castricum, Netherlands.
| | - Edoardo Spina
- Department of Clinical and Experimeta Medicine, University of Messina, Messina, Italy.
| | - Hiroyoshi Takeuchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.
| | - Hélène Verdoux
- Université Bordeaux, Inserm, Bordeaux Population Health Research Center, Team Pharmacoepidemiology, UMR 1219, F-33000 Bordeaux, France.
| | - Christoph U Correll
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York, USA; Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY, USA; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany.
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway; Department of Pharmacy, University of Oslo, Oslo, Norway.
| | - Carlos De Las Cuevas
- Department of Internal Medicine, Dermatology and Psychiatry, School of Medicine, University of La Laguna, Canary Islands, Spain; Instituto Universitario de Neurociencia (IUNE), Universidad de La Laguna, San Cristóbal de La Laguna, Spain.
| | - Jose de Leon
- Mental Health Research Center, Eastern State Hospital, Lexington, KY, USA; Biomedical Research Centre in Mental Health Net (CIBERSAM), Santiago Apóstol Hospital, University of the Basque Country, Vitoria, Spain.
| |
Collapse
|
5
|
Meyer JM. Making sense of norclozapine levels: 3 clinical axioms. Schizophr Res 2024; 268:289-292. [PMID: 37945386 DOI: 10.1016/j.schres.2023.10.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Laboratories commonly provide norclozapine concentrations when a plasma clozapine level is requested, but the appropriate use of this information for the treatment of individuals with schizophrenia is not always clear. Particularly vexing is the fact that norclozapine possesses pharmacological properties that are distinct from its parent compound and which contribute to clozapine's efficacy signal, yet the literature focuses primarily on the association of clozapine levels with symptomatic improvement. The purpose of this brief article is to highlight findings with respect to the need to track norclozapine levels, or the ratio of clozapine/norclozapine plasma levels, to optimize efficacy among inadequate responders to clozapine treatment. In addition, there will be a discussion of the specific type of information provided by the clozapine/norclozapine ratio on clozapine's clearance, and how this ratio is sometimes misinterpreted. There is clinical value from to be derived from norclozapine levels and the clozapine/norclozapine ratio for schizophrenia management, and the principles governing use of this information will be distilled into 3 succinct axioms to aid clinicians in managing their clozapine-treated patients with schizophrenia.
Collapse
|
6
|
Jiménez-Fernández S, Gurpegui M, Correll CU, de Leon J, Schoretsanitis G. A Systematic Review of Clozapine Concentration-Dose Ratio from Therapeutic Drug Monitoring Studies in Children and Adolescents Treated with Clozapine for Mental Disorders. Ther Drug Monit 2024; 46:170-180. [PMID: 38018845 PMCID: PMC10930379 DOI: 10.1097/ftd.0000000000001154] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/14/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Therapeutic drug monitoring of clozapine in children and adolescents has received insufficient attention. Calculation of concentration-to-dose (C/D) ratios from trough steady-state concentrations estimate drug clearance. METHODS A systematic electronic literature search was conducted in 3 article databases from inception until January 10, 2023, and articles reporting clozapine concentrations in children and adolescents were retrieved. The pharmacokinetic quality of the studies was assessed, and clozapine C/D ratios were calculated using the sample mean clozapine dose and concentration. RESULTS Of the 37 articles of potential interest, only 7 reported clozapine trough and steady-state concentrations. After excluding case reports and a study confounded by fluvoxamine, 4 studies on psychosis from Europe and the United States were included. The clozapine C/D ratios were similar to published adult values and ranged from 0.82 to 1.24 with a weighted mean of 1.08 ng/mL per mg/d. The weighted means were 334 mg/d for the dose and 380 ng/mL for the concentration. The stratified analysis of the weighted mean clozapine C/D ratios from 2 studies showed lower values in 52 male (1.05 ng/mL per mg/d) than in 46 female (1.46 ng/mL per mg/d) children and adolescents, with values similar to those reported for European adult nonsmokers. Two female adolescents had high clozapine C/D ratios (2.54 ng/mL per mg/d), an Asian American patient with borderline obesity and a patient with intellectual disability with low dosage (mean = 102 mg/d) and concentration (mean = 55 ng/mL). CONCLUSIONS Reports on clozapine therapeutic drug monitoring in children and adolescents are limited in number and quality. Future studies should focus on basic pharmacokinetic issues, such as stratification by sex, smoking, and relevant comedications with inductive or inhibitory properties.
Collapse
Affiliation(s)
- Sara Jiménez-Fernández
- Child and Adolescent Mental Health Unit, Virgen de Las Nieves University Hospital, Granada, Spain
- CTS-549 Research Group (Psychiatry and Neurosciences), Junta de Andalucía, Spain
| | - Manuel Gurpegui
- CTS-549 Research Group (Psychiatry and Neurosciences), Junta de Andalucía, Spain
- Granada Center for Psychiatric Studies, Granada, Spain
| | - Christoph U. Correll
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York
- Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, New York
| | - Jose de Leon
- Mental Health Research Center, Eastern State Hospital, Lexington, Kentucky
- Biomedical Research Centre in Mental Health Net (CIBERSAM), Santiago Apóstol Hospital, University of the Basque Country, Vitoria, Spain; and
| | - Georgios Schoretsanitis
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York
- Department of Psychiatry, Zucker School of Medicine at Northwell/Hofstra, Hempstead, New York
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland
| |
Collapse
|
7
|
Wang S, Ballard TE, Christopher LJ, Foti RS, Gu C, Khojasteh SC, Liu J, Ma S, Ma B, Obach RS, Schadt S, Zhang Z, Zhang D. The Importance of Tracking "Missing" Metabolites: How and Why? J Med Chem 2023; 66:15586-15612. [PMID: 37769129 DOI: 10.1021/acs.jmedchem.3c01293] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Technologies currently employed to find and identify drug metabolites in complex biological matrices generally yield results that offer a comprehensive picture of the drug metabolite profile. However, drug metabolites can be missed or are captured only late in the drug development process. This could be due to a variety of factors, such as metabolism that results in partial loss of the molecule, covalent bonding to macromolecules, the drug being metabolized in specific human tissues, or poor ionization in a mass spectrometer. These scenarios often draw a great deal of attention from chemistry, safety assessment, and pharmacology. This review will summarize scenarios of missing metabolites, why they are missing, and associated uncovering strategies from deeper investigations. Uncovering previously missed metabolites can have ramifications in drug development with toxicological and pharmacological consequences, and knowledge of these can help in the design of new drugs.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - T Eric Ballard
- Takeda Development Center Americas, Inc., 35 Landsdowne St, Cambridge, Massachusetts 02139, United States
| | - Lisa J Christopher
- Department of Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol-Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Robert S Foti
- Preclinical Development, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Chungang Gu
- Drug Metabolism and Pharmacokinetics, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shuguang Ma
- Drug Metabolism and Pharmacokinetics, Pliant Therapeutics, 260 Littlefield Avenue, South San Francisco, California 94080, United States
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Simone Schadt
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacher Strasse 124, 4070 Basel, Switzerland
| | - Zhoupeng Zhang
- DMPK Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
8
|
de Leon J, Schoretsanitis G, Smith RL, Molden E, Solismaa A, Seppälä N, Kopeček M, Švancer P, Olmos I, Ricciardi C, Iglesias-Garcia C, Iglesias-Alonso A, Spina E, Ruan CJ, Wang CY, Wang G, Tang YL, Lin SK, Lane HY, Kim YS, Kim SH, Rajkumar AP, González-Esquivel DF, Jung-Cook H, Baptista T, Rohde C, Nielsen J, Verdoux H, Quiles C, Sanz EJ, De las Cuevas C, Cohen D, Schulte PF, Ertuğrul A, Anıl Yağcıoğlu AE, Chopra N, McCollum B, Shelton C, Cotes RO, Kaithi AR, Kane JM, Farooq S, Ng CH, Bilbily J, Hiemke C, López-Jaramillo C, McGrane I, Lana F, Eap CB, Arrojo-Romero M, Rădulescu FŞ, Seifritz E, Every-Palmer S, Bousman CA, Bebawi E, Bhattacharya R, Kelly DL, Otsuka Y, Lazary J, Torres R, Yecora A, Motuca M, Chan SKW, Zolezzi M, Ouanes S, De Berardis D, Grover S, Procyshyn RM, Adebayo RA, Kirilochev OO, Soloviev A, Fountoulakis KN, Wilkowska A, Cubała WJ, Ayub M, Silva A, Bonelli RM, Villagrán-Moreno JM, Crespo-Facorro B, Temmingh H, Decloedt E, Pedro MR, Takeuchi H, Tsukahara M, Gründer G, Sagud M, Celofiga A, Ignjatovic Ristic D, Ortiz BB, Elkis H, Pacheco Palha AJ, Llerena A, Fernandez-Egea E, Siskind D, Weizman A, Masmoudi R, Mohd Saffian S, Leung JG, Buckley PF, Marder SR, et alde Leon J, Schoretsanitis G, Smith RL, Molden E, Solismaa A, Seppälä N, Kopeček M, Švancer P, Olmos I, Ricciardi C, Iglesias-Garcia C, Iglesias-Alonso A, Spina E, Ruan CJ, Wang CY, Wang G, Tang YL, Lin SK, Lane HY, Kim YS, Kim SH, Rajkumar AP, González-Esquivel DF, Jung-Cook H, Baptista T, Rohde C, Nielsen J, Verdoux H, Quiles C, Sanz EJ, De las Cuevas C, Cohen D, Schulte PF, Ertuğrul A, Anıl Yağcıoğlu AE, Chopra N, McCollum B, Shelton C, Cotes RO, Kaithi AR, Kane JM, Farooq S, Ng CH, Bilbily J, Hiemke C, López-Jaramillo C, McGrane I, Lana F, Eap CB, Arrojo-Romero M, Rădulescu FŞ, Seifritz E, Every-Palmer S, Bousman CA, Bebawi E, Bhattacharya R, Kelly DL, Otsuka Y, Lazary J, Torres R, Yecora A, Motuca M, Chan SKW, Zolezzi M, Ouanes S, De Berardis D, Grover S, Procyshyn RM, Adebayo RA, Kirilochev OO, Soloviev A, Fountoulakis KN, Wilkowska A, Cubała WJ, Ayub M, Silva A, Bonelli RM, Villagrán-Moreno JM, Crespo-Facorro B, Temmingh H, Decloedt E, Pedro MR, Takeuchi H, Tsukahara M, Gründer G, Sagud M, Celofiga A, Ignjatovic Ristic D, Ortiz BB, Elkis H, Pacheco Palha AJ, Llerena A, Fernandez-Egea E, Siskind D, Weizman A, Masmoudi R, Mohd Saffian S, Leung JG, Buckley PF, Marder SR, Citrome L, Freudenreich O, Correll CU, Müller DJ. Guía internacional para una dosificación más segura de la clozapina en adultos mediante el uso de 6 titulaciones personalizadas de dosis basados en la etnicidad, la proteína C reactiva y los niveles de clozapina. PSIQUIATRÍA BIOLÓGICA 2023; 30:100415. [DOI: 10.1016/j.psiq.2023.100415] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
|
9
|
Wen B, Zhang Y, Young GC, Kenworthy D, Pereira A, Pirhalla J, Doyle J, Jordon B, Zhan J, Johnson M. Investigation of Clinical Absorption, Distribution, Metabolism, and Excretion and Pharmacokinetics of the HIV-1 Maturation Inhibitor GSK3640254 Using an Intravenous Microtracer Combined with EnteroTracker for Biliary Sampling. Drug Metab Dispos 2022; 50:1442-1453. [PMID: 36153007 DOI: 10.1124/dmd.122.000955] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/17/2022] [Indexed: 11/22/2022] Open
Abstract
GSK3640254 is a next-generation maturation inhibitor in development for HIV-1 treatment, with pharmacokinetics (PK) supporting once-daily oral dosing in human. This open-label, nonrandomized, two-period clinical mass balance and excretion study was used to investigate the excretion balance, PK, and metabolism of GSK3640254. Five healthy men received a single intravenous microtracer of 100 μg [14C]GSK3640254 with a concomitant oral nonradiolabeled 200-mg tablet followed by an oral 85-mg dose of [14C]GSK3640254 14 days later. Complementary methods, including intravenous microtracing and accelerator mass spectrometry, allowed characterization of several parameters, including fraction absorbed, fraction escaping gut metabolism, hepatic extraction ratio, and renal clearance. Intravenous PK of GSK3640254 was characterized by low plasma clearance (1.04 l/h), moderate terminal phase half-life (21.7 hours), and low volume of distribution at steady state (28.7 L). Orally dosed GSK3640254 was absorbed (fraction absorbed, 0.26), with a high fraction escaping gut metabolism (0.898) and a low hepatic extraction ratio (0.00544), all consistent with low in vitro intrinsic clearance in liver microsomes and hepatocytes. No major metabolites in human plasma required further qualification in animal studies. Both unchanged parent GSK3640254 and its oxidative and conjugative metabolites were excreted into bile, with GSK3640254 likely subject to further metabolism through enterohepatic recirculation. Renal elimination of GSK3640254 as the parent drug or its metabolites was negligible, with >94% of total recovery of oral dose and >99% of the recovered radioactivity in feces. Altogether, the data suggest that systemically available GSK3640254 was slowly eliminated almost entirely by hepatobiliary secretion, primarily as conjugative and oxidative metabolites. SIGNIFICANCE STATEMENT: The combination of an intravenous 14C microtracer with duodenal bile sampling using EnteroTracker in a human absorption, distribution, metabolism, and excretion study enabled derivation of absorption and first-pass parameters, including fraction absorbed, proportion escaping first-pass extraction through the gut wall and liver, hepatic extraction, and other conventional clinical pharmacokinetic parameters. This approach identified hepatic metabolism and biliary excretion as a major elimination pathway for absorbed drug, which would be overlooked based solely on analyses of plasma, urine, and fecal matrices.
Collapse
Affiliation(s)
- Bo Wen
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Ying Zhang
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Graeme C Young
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - David Kenworthy
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Adrian Pereira
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Jill Pirhalla
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Janine Doyle
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Bethany Jordon
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Joyce Zhan
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| | - Mark Johnson
- GSK, Collegeville, Pennsylvania (B.W., Y.Z., J.P., J.D., J.Z.); GSK, Ware, United Kingdom (G.C.Y., D.K.); GSK, Stevenage, United Kingdom (A.P., B.J.); and ViiV Healthcare, Durham, North Carolina (M.J.)
| |
Collapse
|
10
|
de Leon J, Schoretsanitis G, Smith RL, Molden E, Solismaa A, Seppälä N, Kopeček M, Švancer P, Olmos I, Ricciardi C, Iglesias-Garcia C, Iglesias-Alonso A, Spina E, Ruan CJ, Wang CY, Wang G, Tang YL, Lin SK, Lane HY, Kim YS, Kim SH, Rajkumar AP, González-Esquivel DF, Jung-Cook H, Baptista T, Rohde C, Nielsen J, Verdoux H, Quiles C, Sanz EJ, De Las Cuevas C, Cohen D, Schulte PFJ, Ertuğrul A, Anıl Yağcıoğlu AE, Chopra N, McCollum B, Shelton C, Cotes RO, Kaithi AR, Kane JM, Farooq S, Ng CH, Bilbily J, Hiemke C, López-Jaramillo C, McGrane I, Lana F, Eap CB, Arrojo-Romero M, Rădulescu FŞ, Seifritz E, Every-Palmer S, Bousman CA, Bebawi E, Bhattacharya R, Kelly DL, Otsuka Y, Lazary J, Torres R, Yecora A, Motuca M, Chan SKW, Zolezzi M, Ouanes S, De Berardis D, Grover S, Procyshyn RM, Adebayo RA, Kirilochev OO, Soloviev A, Fountoulakis KN, Wilkowska A, Cubała WJ, Ayub M, Silva A, Bonelli RM, Villagrán-Moreno JM, Crespo-Facorro B, Temmingh H, Decloedt E, Pedro MR, Takeuchi H, Tsukahara M, Gründer G, Sagud M, Celofiga A, Ignjatovic Ristic D, Ortiz BB, Elkis H, Pacheco Palha AJ, LLerena A, Fernandez-Egea E, Siskind D, Weizman A, Masmoudi R, Mohd Saffian S, Leung JG, Buckley PF, Marder SR, et alde Leon J, Schoretsanitis G, Smith RL, Molden E, Solismaa A, Seppälä N, Kopeček M, Švancer P, Olmos I, Ricciardi C, Iglesias-Garcia C, Iglesias-Alonso A, Spina E, Ruan CJ, Wang CY, Wang G, Tang YL, Lin SK, Lane HY, Kim YS, Kim SH, Rajkumar AP, González-Esquivel DF, Jung-Cook H, Baptista T, Rohde C, Nielsen J, Verdoux H, Quiles C, Sanz EJ, De Las Cuevas C, Cohen D, Schulte PFJ, Ertuğrul A, Anıl Yağcıoğlu AE, Chopra N, McCollum B, Shelton C, Cotes RO, Kaithi AR, Kane JM, Farooq S, Ng CH, Bilbily J, Hiemke C, López-Jaramillo C, McGrane I, Lana F, Eap CB, Arrojo-Romero M, Rădulescu FŞ, Seifritz E, Every-Palmer S, Bousman CA, Bebawi E, Bhattacharya R, Kelly DL, Otsuka Y, Lazary J, Torres R, Yecora A, Motuca M, Chan SKW, Zolezzi M, Ouanes S, De Berardis D, Grover S, Procyshyn RM, Adebayo RA, Kirilochev OO, Soloviev A, Fountoulakis KN, Wilkowska A, Cubała WJ, Ayub M, Silva A, Bonelli RM, Villagrán-Moreno JM, Crespo-Facorro B, Temmingh H, Decloedt E, Pedro MR, Takeuchi H, Tsukahara M, Gründer G, Sagud M, Celofiga A, Ignjatovic Ristic D, Ortiz BB, Elkis H, Pacheco Palha AJ, LLerena A, Fernandez-Egea E, Siskind D, Weizman A, Masmoudi R, Mohd Saffian S, Leung JG, Buckley PF, Marder SR, Citrome L, Freudenreich O, Correll CU, Müller DJ. Correction: An International Adult Guideline for Making Clozapine Titration Safer by Using Six Ancestry-Based Personalized Dosing Titrations, CRP, and Clozapine Levels. PHARMACOPSYCHIATRY 2022; 55:e1. [PMID: 35052001 DOI: 10.1055/a-1625-6388] [Show More Authors] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AbstractThis international guideline proposes improving clozapine package inserts worldwide by using ancestry-based dosing and titration. Adverse drug reaction (ADR) databases suggest that clozapine is the third most toxic drug in the United States (US), and it produces four times higher worldwide pneumonia mortality than that by agranulocytosis or myocarditis. For trough steady-state clozapine serum concentrations, the therapeutic reference range is narrow, from 350 to 600 ng/mL with the potential for toxicity and ADRs as concentrations increase. Clozapine is mainly metabolized by CYP1A2 (female non-smokers, the lowest dose; male smokers, the highest dose). Poor metabolizer status through phenotypic conversion is associated with co-prescription of inhibitors (including oral contraceptives and valproate), obesity, or inflammation with C-reactive protein (CRP) elevations. The Asian population (Pakistan to Japan) or the Americas’ original inhabitants have lower CYP1A2 activity and require lower clozapine doses to reach concentrations of 350 ng/mL. In the US, daily doses of 300–600 mg/day are recommended. Slow personalized titration may prevent early ADRs (including syncope, myocarditis, and pneumonia). This guideline defines six personalized titration schedules for inpatients: 1) ancestry from Asia or the original people from the Americas with lower metabolism (obesity or valproate) needing minimum therapeutic dosages of 75–150 mg/day, 2) ancestry from Asia or the original people from the Americas with average metabolism needing 175–300 mg/day, 3) European/Western Asian ancestry with lower metabolism (obesity or valproate) needing 100–200 mg/day, 4) European/Western Asian ancestry with average metabolism needing 250–400 mg/day, 5) in the US with ancestries other than from Asia or the original people from the Americas with lower clozapine metabolism (obesity or valproate) needing 150–300 mg/day, and 6) in the US with ancestries other than from Asia or the original people from the Americas with average clozapine metabolism needing 300–600 mg/day. Baseline and weekly CRP monitoring for at least four weeks is required to identify any inflammation, including inflammation secondary to clozapine rapid titration.
Collapse
Affiliation(s)
- Jose de Leon
- Mental Health Research Center, Eastern State Hospital, Lexington, KY, USA
- Department of Psychiatry, University of Kentucky, Lexington, KY, USA
- Psychiatry and Neurosciences Research Group (CTS-549), Institute of Neurosciences, University of Granada, Granada, Spain
- Biomedical Research Centre in Mental Health Net (CIBERSAM), Santiago Apostol Hospital, University of the Basque Country, Vitoria, Spain
| | - Georgios Schoretsanitis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York, USA
| | - Robert L Smith
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Anssi Solismaa
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Psychiatry, Tampere University Hospital, Tampere, Finland
| | | | - Miloslav Kopeček
- National Institute of Mental Health, Klecany, Czech Republic
- Department of Psychiatry, Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Patrik Švancer
- National Institute of Mental Health, Klecany, Czech Republic
- Department of Psychiatry, Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Ismael Olmos
- Clinical Pharmacology Unit and Pharmacy Department, Vilardebó Hospital, Administración de Servicios de Salud, Montevideo, Uruguay
| | - Carina Ricciardi
- Clinical Pharmacology Unit and Outpatient Clinic, Vilardebó Hospital, Administración de Servicios de Salud, Montevideo, Uruguay
| | - Celso Iglesias-Garcia
- Universidad de Oviedo. CIBERSAM. INEUROPA. ISPA-FIMBA, Oviedo, Spain
- Hospital Valle del Nalón, Langreo, Spain
| | | | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Can-Jun Ruan
- Laboratory of Clinical Psychopharmacology, Beijing Anding Hospital, Capital Medical University, Beijing, China
- The National Clinical Research Centre for Mental Disorders & Beijing Key Lab of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Chuan-Yue Wang
- The National Clinical Research Centre for Mental Disorders & Beijing Key Lab of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Gang Wang
- The National Clinical Research Centre for Mental Disorders & Beijing Key Lab of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yi-Lang Tang
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
- Substance Abuse Treatment Program, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Shih-Ku Lin
- Department of Psychiatry, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| | - Yong Sik Kim
- Department of Neuropsychiatry, Nowon Eulji Medical Center, Eulji University, School of Medicine, Seoul, Korea
| | - Se Hyun Kim
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Anto P Rajkumar
- Department of Psychiatry, Christian Medical College, Vellore, India
- Institute of Mental Health, Jubilee Campus, University of Nottingham, Triumph Road, Nottingham, United Kingdom
| | | | - Helgi Jung-Cook
- Instituto Nacional de Neurología y Neurocirugía, México City, México
- Facultad de Química, Universidad Nacional Autónoma de México (UNAM), México City, México
| | - Trino Baptista
- Department of Physiology, Los Andes University Medical School, Mérida, Venezuela
| | - Christopher Rohde
- Department of Affective Disorders, Aarhus University Hospital - Psychiatry, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jimmi Nielsen
- Mental Health Centre Glostrup, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hélène Verdoux
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team Pharmacoepidemiology, UMR 1219, F-33000 Bordeaux, France
| | - Clelia Quiles
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, Team Pharmacoepidemiology, UMR 1219, F-33000 Bordeaux, France
| | - Emilio J Sanz
- Department of Physical Medicine and Pharmacology, School of Medicine, Universidad de La Laguna, Canary Islands, Spain
- Hospital Universitario de Canarias, Tenerife, Spain
| | - Carlos De Las Cuevas
- Department of Internal Medicine, Dermatology and Psychiatry, School of Medicine, and Instituto Universitario de Neurociencia (IUNE), University of La Laguna, Canary Islands, Spain
| | - Dan Cohen
- Dutch Clozapine Collaboration Group, Castricum, The Netherlands
- FACT-team in Heerhugowaard, Department of Severe Mental Illness, Mental Health Services North-Holland North, The Netherlands
| | - Peter F J Schulte
- Dutch Clozapine Collaboration Group, Castricum, The Netherlands
- Mental Health Team Alkmaar, Mental Health Services Noord-Holland-Noord, Alkmaar, The Netherlands
| | - Aygün Ertuğrul
- Department of Psychiatry, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | | | - Nitin Chopra
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | | | - Charles Shelton
- Department of Psychiatry, University of Kentucky, Lexington, KY, USA
- Eastern State Hospital, Lexington, Kentucky, USA
| | - Robert O Cotes
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - John M Kane
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York, USA
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Hempstead, New York, USA
| | - Saeed Farooq
- School of Medicine, Keele University, Staffordshire, and Midlands Partnership NHS Foundation Trust, Staffordshire, United Kingdom
| | - Chee H Ng
- Department of Psychiatry, The University of Melbourne, Melbourne, Victoria, Australia
| | - John Bilbily
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Christoph Hiemke
- Department of Psychiatry and Psychotherapy, University Medical Center of Mainz, Germany
| | - Carlos López-Jaramillo
- Grupo de Investigación en Psiquiatría GIPSI, Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
- Programa Trastornos del Ánimo, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Ian McGrane
- Department of Pharmacy Practice, University of Montana, Missoula, USA
| | - Fernando Lana
- Institute of Neuropsychiatry and Addictions (INAD), Parc de Salut Mar, Barcelona, Spain
- Department of Psychiatry, Autonomous University of Barcelona, Spain
| | - Chin B Eap
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Switzerland
| | - Manuel Arrojo-Romero
- Department of Psychiatry, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | - Flavian Ş Rădulescu
- Center for Drug Sciences, Faculty of Pharmacy, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland
| | - Susanna Every-Palmer
- Te Korowai Whāriki Central Regional Forensic Service, Capital and Coast District Health Board, Wellington, New Zealand
- Department of Psychological Medicine, University of Otago, Wellington, New Zealand
| | - Chad A Bousman
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, and Community Health Sciences University of Calgary, Alberta, Canada
| | - Emmanuel Bebawi
- Faculty of Medicine, University of Montreal, Montreal, Canada
- Department of Pharmacy, Hôpital du Sacré-Cœur de Montréal, Montreal, Canada
| | - Rahul Bhattacharya
- East London NHS Foundation Trust, London, United Kingdom
- Honorary Clinical Senior Lecturer, Barts and the London School of Medicine, Queen Mary University of London, United Kingdom
| | - Deanna L Kelly
- Department of Psychiatry, School of Medicine, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yuji Otsuka
- Department of Psychiatry, Asahi General Hospital, Chiba, Japan
| | - Judit Lazary
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - Rafael Torres
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Agustin Yecora
- Secretaría de Salud Mental y Adicciones, Ministerio de Salud de la Provincia de Jujuy, San Salvador de Jujuy, Argentina
| | - Mariano Motuca
- Instituto Vilapriño, Center for Studies, Assistance and Research in Neurosciences, Mendoza, Argentina
| | - Sherry K W Chan
- Department of Psychiatry, LSK Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR
| | - Monica Zolezzi
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Sami Ouanes
- Department of Psychiatry, Hamad Medical Corporation, Doha, Qatar
| | | | - Sandeep Grover
- Department of Psychiatry, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ric M Procyshyn
- British Columbia Mental Health and Substance Use Research Institute, Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | | | - Oleg O Kirilochev
- Department of Clinical Pharmacology, Astrakhan State Medical University, Astrakhan, Russian Federation
| | - Andrey Soloviev
- Department of Psychiatry and Clinical Psychology, Northern State Medical University, Arkhangelsk, Russia
| | - Konstantinos N Fountoulakis
- 3rd Department of Psychiatry, Division of Neurosciences, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece
| | - Alina Wilkowska
- Department of Psychiatry, Medical University of Gdańsk, Gdańsk, Poland
| | - Wiesław J Cubała
- Department of Psychiatry, Medical University of Gdańsk, Gdańsk, Poland
| | - Muhammad Ayub
- Department of Psychiatry, Queens University, Kingston, Canada
| | - Alzira Silva
- Psychiatry Department, Centro Hospitalar Universitário de S. João, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | | | - José M Villagrán-Moreno
- Department of Neurosciences, Jerez University Hospital, Andalusian Health Service, University of Cadiz, Jerez, Spain
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio-IBIS, Sevilla, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Sevilla, Spain
| | - Henk Temmingh
- Department of Psychiatry and Mental Health, University of Cape Town, Valkenberg Hospital, Western Cape, Cape Town, South Africa
| | - Eric Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | - Hiroyoshi Takeuchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Tsukahara
- Department of Psychiatry, Okayama Psychiatric Medical Center, Okayama, Japan
| | - Gerhard Gründer
- Central Institute of Mental Health, Department of Molecular Neuroimaging, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marina Sagud
- Department of Psychiatry, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department for Psychiatry and Psychological Medicine, University Hospital Center Zagreb, Croatia
| | - Andreja Celofiga
- Department of Psychiatry, University Medical Centre Maribor, Maribor, Slovenia
| | | | - Bruno B Ortiz
- Group of Resistant Schizophrenia (GER), Schizophrenia Program (Proesq), Federal University of Sao Paulo, SP, Brazil
| | - Helio Elkis
- Department and Institute of Psychiatry, University of São Paulo Medical School (FMUSP), Sao Paulo, Brazil
| | - António J Pacheco Palha
- Department and Institute of Psychiatry and Mental Health, Oporto Faculty of Medicine, Oporto, Portugal
- Casa de Salidedo Som Jesus (Psychiatric Hospital), Oporto, Portugal
| | - Adrián LLerena
- INUBE Biosanitary Research Institute of Extremadura. Extremadura University Hospital and Medical School, Badajoz, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Emilio Fernandez-Egea
- Cambridge Psychosis Centre, Cambrigeshire and Peterborough NHS Foundation Trust & Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Dan Siskind
- Metro South Addiction and Mental Health Service, Brisbane, Australia
- Queensland Centre for Mental Health Research and School of Clinical Medicine, University of Queensland, Brisbane, Australia
| | - Abraham Weizman
- Research Unit, Geha Mental Health Center and Molecular Psychiatry Laboratory, Felsenstein Medical Research Center, Tel Aviv, Israel
- Department of Psychiatry Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Rim Masmoudi
- Psychiatry "A" Department, Hedi Chaker University Hospital, Sfax, Tunisia
- Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Shamin Mohd Saffian
- Centre for Quality Management of Medicines, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Peter F Buckley
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Stephen R Marder
- Semel Institute for Neuroscience at UCLA, Los Angeles, CA, USA
- VA Desert Pacific Mental Illness Research, Education, and Clinical Center, Los Angeles, CA, USA
| | - Leslie Citrome
- New York Medical College, Department of Psychiatry and Behavioral Sciences, Valhalla, NY, USA
| | - Oliver Freudenreich
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christoph U Correll
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, New York, USA
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Hempstead, New York, USA
- Charité Universitätsmedizin Berlin, Department of Child and Adolescent Psychiatry, Berlin, Germany
| | - Daniel J Müller
- Addictions Division, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Chang R, Xu ST, Jin HW, Zhao YJ, Hu TY, Wang G, Wang J, Zhen L. Simultaneous determination of ZL-01, a novel nucleotide prodrug, and its metabolites in rat plasma by LC-MS/MS: Application to pharmacokinetic study. J Pharm Biomed Anal 2022; 208:114437. [PMID: 34735992 DOI: 10.1016/j.jpba.2021.114437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023]
Abstract
ZL-01 is a novel dual-prodrug which shows promise to be an antiviral candidate for hepatitis C virus. Here we have established a liquid chromatography tandem mass spectrometry (LC-MS/MS) method for simultaneous determination of ZL-01 and its four metabolites (M1, M7, M8, and M9) in rat plasma with special consideration of ex vivo ZL-01, M1, and M7 stability. Several factors affecting the stability were investigated. EDTA and citric acid solution (1 M) were added to plasma to maintain the stability of analytes. The protein-precipitation method was selected with acetonitrile containing sofosbuvir as internal standard (IS). Adequate separation of ZL-01 and its metabolites was achieved on XSelect HSS T3 (3.5 µm, 4.6 × 150 mm) column by a gradient-elution with a mobile phase consisting of 0.1% formic acid and acetonitrile at a flow rate of 0.5 mL/min. The detection was performed on a triple quadrupole tandem mass spectrometer by multiple reaction monitoring (MRM) mode to monitor the precursor-to-product ion transitions of m/z 599.2→418.5 for ZL-01, m/z 529.7→398.2 for M1, m/z 330.5→182.0 for M7, m/z 260.3→112.1 for M8, m/z 261.3→113.2 for M9 and m/z 530.4→243.4 for IS. The calibration curves exhibited good linearity (r>0.997) for all components. The lower limit of quantitation (LLOQ) was in the range of 1-2 ng/mL. The intra-day and inter-day precisions (RSD) at three different levels were both less than 10.2% and the accuracies (RE) ranged from -3.7-7.6%. The matrix effect and extraction recovery of them ranged from 84% to 110.3% and 88.3-106.3%. This LC-MS/MS method for the simultaneous quantitation of ZL-01 and its metabolites was developed successfully and applied in the pharmacokinetic studies of these in rats. Pharmacokinetic results indicated ZL-01 would be metabolized rapidly and M8 might be the main metabolites after oral absorption.
Collapse
Affiliation(s)
- Ruirui Chang
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Si-Tao Xu
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hao-Wen Jin
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yong-Jie Zhao
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tian-Yu Hu
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Jiankun Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Le Zhen
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
12
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Kowalski JP, Pelletier RD, McDonald MG, Kelly EJ, Rettie AE. Pharmacokinetics, metabolism and off-target effects in the rat of 8-[(1H- benzotriazol-1-yl)amino]octanoic acid, a selective inhibitor of human cytochrome P450 4Z1: β-oxidation as a potential augmenting pathway for inhibition. Xenobiotica 2021; 51:901-915. [PMID: 33993844 DOI: 10.1080/00498254.2021.1930281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
8-[(1H-1,2,3-benzotriazol-1-yl)amino]octanoic acid (8-BOA) was recently identified as a selective and potent mechanism-based inactivator (MBI) of breast cancer-associated CYP4Z1 and exhibited favourable inhibitory activity in vitro, thus meriting in vivo characterization.The pharmacokinetics and metabolism of 8-BOA in rats was examined after a single IV bolus dose of 10 mg/kg. A biphasic time-concentration profile resulted in relatively low clearance and a prolonged elimination half-life.The major circulating metabolites identified in plasma were products of β-oxidation; congeners losing two and four methylene groups accounted for >50% of metabolites by peak area. The -(CH2)2 product was characterized previously as a CYP4Z1 MBI and so represents an active metabolite that may contribute to the desired pharmacological effect.Ex vivo analysis of total CYP content in rat liver and kidney microsomes showed that off-target CYP inactivation was minimal; liver microsomal probe substrate metabolism also demonstrated low off-target inactivation. Standard clinical chemistries provided no indication of acute toxicity.In silico simulations using the free concentration of 8-BOA in plasma suggested that the in vivo dose used here may effectively inactivate CYP4Z1 in a xenografted tumour.
Collapse
Affiliation(s)
- John P Kowalski
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Drug Metabolism and Pharmacokinetics, Pfizer Boulder R&D, Boulder, CO, USA
| | - Robert D Pelletier
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Matthew G McDonald
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Drug Metabolism and Pharmacokinetics, Pfizer Boulder R&D, Boulder, CO, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Allan E Rettie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Future Studies on the Interaction Between Clozapine and Valproic Acid Should Aspire to Include Longitudinal Designs and Free Valproate Concentrations, and Should Consider that Inducer and/or Inhibitory Effects May Vary With Time, the Individual, and the Auto-Induction of Valproic Acid. Ther Drug Monit 2020; 42:159-161. [PMID: 31633608 DOI: 10.1097/ftd.0000000000000705] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Cerny MA, Kalgutkar AS, Obach RS, Sharma R, Spracklin DK, Walker GS. Effective Application of Metabolite Profiling in Drug Design and Discovery. J Med Chem 2020; 63:6387-6406. [PMID: 32097005 DOI: 10.1021/acs.jmedchem.9b01840] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
At one time, biotransformation was a descriptive activity in pharmaceutical development, viewed simply as structural elucidation of drug metabolites, completed only once compounds entered clinical development. Herein, we present our strategic approach using structural elucidation to enable chemistry design/SAR development. The approach considers four questions that often present themselves to medicinal chemists optimizing their compounds for candidate selection: (1) What are the important clearance mechanisms that mediate the disposition of my molecule? (2) Can metabolic liabilities be modulated in a favorable way? (3) Does my compound undergo bioactivation to a reactive metabolite? (4) Do any of the metabolites possess activity, either on- or off-target? An additional question necessary to support compound development relates to metabolites in safety testing (MIST) and our approach also addresses this question. The value in structural elucidation is derived from its application to better design molecules, guide their clinical development, and underwrite patient safety.
Collapse
Affiliation(s)
- Matthew A Cerny
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, 1 Portland Street, Cambridge Massachusetts 02139, United States
| | - R Scott Obach
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Raman Sharma
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Douglas K Spracklin
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory S Walker
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
16
|
Obach RS, Lin J, Kimoto E, Duvvuri S, Nicholas T, Kadar EP, Tremaine LM, Sawant-Basak A. Estimation of Circulating Drug Metabolite Exposure in Human Using In Vitro Data and Physiologically Based Pharmacokinetic Modeling: Example of a High Metabolite/Parent Drug Ratio. Drug Metab Dispos 2018; 46:89-99. [PMID: 29150544 DOI: 10.1124/dmd.117.078279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022] Open
Abstract
(R)-4-((4-(((4-((tetrahydrofuran-3-yl)oxy)benzo[d]isoxazol-3-yl)oxy)methyl)piperidin-1-yl)methyl)tetrahydro-2H-pyran-4-ol (TBPT), a serotonin-4 receptor partial agonist, is metabolized to two metabolites: an N-dealkylation product [(R)-3-(piperidin-4-ylmethoxy)-4-((tetrahydrofuran-3-yl)oxy)benzo[d]isoxazole (M1)] and a cyclized oxazolidine structure [7-(((4-(((R)-tetrahydrofuran-3-yl)oxy)benzo[d]isoxazol-3-yl)oxy)methyl)octahydro-3H (M2)]. After administration of TBPT to humans the exposure to M1 was low and the exposure to M2 was high, relative to the parent drug, despite this being the opposite in vitro. In this study, projection of the plasma metabolite/parent (M/P) ratios for M1 and M2 was attempted using in vitro metabolism, binding, and permeability data in static and dynamic physiologically based pharmacokinetic (PBPK) models. In the static model, the fraction of parent clearance yielding the metabolite (which also required taking into account secondary metabolites of M1 and M2), the clearance of the metabolites and parent, and an estimate of the availability of the metabolites from the liver were combined to yield estimated parent/metabolite ratios of 0.32 and 23 for M1 and M2, respectively. PBPK modeling that used in vitro and physicochemical data input yielded estimates of 0.26 and 20, respectively. The actual values were 0.12 for M1/TBPT and 58 for M2/TBPT. Thus, the ratio for M1 was overpredicted, albeit at values less than unity. The ratio for M2/TBPT was underpredicted, and the high ratio of 58 may exceed a limiting ceiling of the approach. Nevertheless, when considered in the context of determining whether a potential circulating metabolite may be quantitatively important prior to administration of a drug for the first time to humans, the approaches succeeded in highlighting the importance of M2 (M/P ratio >> 1) relative to M1, despite M1 being much greater than M2 in vitro.
Collapse
Affiliation(s)
- R Scott Obach
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Jian Lin
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Emi Kimoto
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Sridhar Duvvuri
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Timothy Nicholas
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Eugene P Kadar
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Larry M Tremaine
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| | - Aarti Sawant-Basak
- Department of Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut (RSO, JL, EK, EPK, and RSO), and Cambridge, Massachusetts (ASB); and Department of Clinical Pharmacology, Pfizer Inc., Cambridge, Massachusetts (SD and TN)
| |
Collapse
|
17
|
Nguyen HQ, Lin J, Kimoto E, Callegari E, Tse S, Obach RS. Prediction of Losartan-Active Carboxylic Acid Metabolite Exposure Following Losartan Administration Using Static and Physiologically Based Pharmacokinetic Models. J Pharm Sci 2017; 106:2758-2770. [DOI: 10.1016/j.xphs.2017.03.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 01/02/2023]
|
18
|
Luffer-Atlas D, Atrakchi A. A decade of drug metabolite safety testing: industry and regulatory shared learning. Expert Opin Drug Metab Toxicol 2017; 13:897-900. [DOI: 10.1080/17425255.2017.1364362] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Debra Luffer-Atlas
- Drug Disposition and Toxicology, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Aisar Atrakchi
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
19
|
Grime K, Pehrson R, Nordell P, Gillen M, Kühn W, Mant T, Brännström M, Svanberg P, Jones B, Brealey C. An S-warfarin and AZD1981 interaction: in vitro and clinical pilot data suggest the N-deacetylated amino acid metabolite as the primary perpetrator. Br J Clin Pharmacol 2016; 83:381-392. [PMID: 27558866 DOI: 10.1111/bcp.13102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/04/2016] [Accepted: 08/16/2016] [Indexed: 02/01/2023] Open
Abstract
AIM AZD1981 is an orally bioavailable chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTh2) receptor antagonist progressed to phase II trials for the treatment of allergic asthma. Previously performed in vitro human hepatocyte incubations identified N-deacetylated AZD1981 as a primary metabolite. We report on metabolite exposure from a clinical excretion balance, on in vitro studies performed to determine the likelihood of a metabolite-dependent drug-drug interaction (DDI) and on a clinical warfarin DDI study. The aim was to demonstrate that N-deacetylated AZD1981 is responsible for the observed interaction. METHODS The excretion and biotransformation of [14 C]-AZD1981 were studied in healthy male volunteers, and subsequently in vitro cytochrome P450 (CYP) inhibition and hepatocyte uptake investigations were carried out with metabolites and the parent drug. A clinical DDI study using coadministered twice-daily 100 mg and 400 mg AZD1981 with 25 mg warfarin was performed. RESULTS The excretion balance study showed N-deacetylated AZD1981 to be the most abundant metabolite present in plasma. In vitro data revealed the metabolite to be a weak CYP2C9 time-dependent inhibitor, subject to more active hepatic uptake than the parent molecule. Clinically, the S-warfarin area under the plasma concentration-time curve increased, on average, 1.4-fold [95% confidence interval (CI) 1.22, 1.50] and 2.4-fold (95% CI 2.11, 2.64) after 100 mg (n = 13) and 400 mg (n = 11) AZD1981 administration, respectively. In vitro CYP inhibition and hepatocyte uptake data were used to explain the interaction. CONCLUSIONS N-deacetylated AZD1981 can be added to the small list of drug metabolites reported as sole contributors to clinical drug-drug interactions, with weak time-dependent inhibition exacerbated by efficient hepatic uptake being the cause.
Collapse
Affiliation(s)
- Ken Grime
- Respiratory, Inflammation & Autoimmune Disease Department of DMPK, AstraZeneca R&D, Gothenburg, Sweden
| | - Rikard Pehrson
- Respiratory, Inflammation & Autoimmune Disease Department of DMPK, AstraZeneca R&D, Gothenburg, Sweden
| | - Pär Nordell
- Drug Safety and Metabolism, AstraZeneca R&D, Gothenburg, Sweden
| | - Michael Gillen
- AstraZeneca Early Clinical Development, Gaithersburg, MD, USA
| | - Wolfgang Kühn
- Quintiles Allergy, Respiratory, Infectious Diseases & Vaccines Therapeutic Science & Strategy Unit, Uppsala, Sweden
| | - Timothy Mant
- Quintiles Drug Research Unit at Guy's Hospital, London, UK
| | - Marie Brännström
- Respiratory, Inflammation & Autoimmune Disease Department of DMPK, AstraZeneca R&D, Gothenburg, Sweden
| | - Petter Svanberg
- Respiratory, Inflammation & Autoimmune Disease Department of DMPK, AstraZeneca R&D, Gothenburg, Sweden
| | - Barry Jones
- Drug Safety and Metabolism, AstraZeneca R&D, Gothenburg, Sweden
| | | |
Collapse
|
20
|
Genovino J, Sames D, Hamann LG, Touré BB. Die Erschließung von Wirkstoffmetaboliten durch übergangsmetallkatalysierte C-H-Oxidation: die Leber als Inspiration für die Synthese. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201602644] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Julien Genovino
- Pfizer Inc.; Worldwide Medicinal Chemistry, Cardiovascular, Metabolic, and Endocrine Diseases (CVMED); 558 Eastern Point Road Groton CT 06340 USA
| | - Dalibor Sames
- Columbia University; Department of Chemistry and Neurotechnology Center; 3000 Broadway MC3101 New York NY 10027 USA
| | - Lawrence G. Hamann
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC); 181 Massachusetts Avenue Cambridge MA 02139 USA
| | - B. Barry Touré
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC); 100 Technology Square Cambridge MA 02139 USA
| |
Collapse
|
21
|
Genovino J, Sames D, Hamann LG, Touré BB. Accessing Drug Metabolites via Transition-Metal Catalyzed C-H Oxidation: The Liver as Synthetic Inspiration. Angew Chem Int Ed Engl 2016; 55:14218-14238. [PMID: 27723189 DOI: 10.1002/anie.201602644] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/08/2016] [Indexed: 11/07/2022]
Abstract
Can classical and modern chemical C-H oxidation reactions complement biotransformation in the synthesis of drug metabolites? We have surveyed the literature in an effort to try to answer this important question of major practical significance in the pharmaceutical industry. Drug metabolites are required throughout all phases of the drug discovery and development process; however, their synthesis is still an unsolved problem. This Review, not intended to be comprehensive or historical, highlights relevant applications of chemical C-H oxidation reactions, electrochemistry and microfluidic technologies to drug templates in order to access drug metabolites, and also highlights promising reactions to this end. Where possible or appropriate, the contrast with biotransformation is drawn. In doing so, we have tried to identify gaps where they exist in the hope to spur further activity in this very important research area.
Collapse
Affiliation(s)
- Julien Genovino
- Pfizer Inc., Worldwide Medicinal Chemistry, Cardiovascular, Metabolic, and Endocrine Diseases (CVMED), 558 Eastern Point Road, Groton, CT, 06340, USA
| | - Dalibor Sames
- Columbia University, Department of Chemistry and Neurotechnology Center, 3000 Broadway MC3101, New York, NY, 10027, USA
| | - Lawrence G Hamann
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC), 181 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - B Barry Touré
- Novartis Institutes for Biomedical Sciences (NIBR), Global Discovery Chemistry (GDC), 100 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
22
|
Templeton IE, Chen Y, Mao J, Lin J, Yu H, Peters S, Shebley M, Varma MV. Quantitative Prediction of Drug-Drug Interactions Involving Inhibitory Metabolites in Drug Development: How Can Physiologically Based Pharmacokinetic Modeling Help? CPT Pharmacometrics Syst Pharmacol 2016; 5:505-515. [PMID: 27642087 PMCID: PMC5080647 DOI: 10.1002/psp4.12110] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 12/26/2022] Open
Abstract
This subteam under the Drug Metabolism Leadership Group (Innovation and Quality Consortium) investigated the quantitative role of circulating inhibitory metabolites in drug-drug interactions using physiologically based pharmacokinetic (PBPK) modeling. Three drugs with major circulating inhibitory metabolites (amiodarone, gemfibrozil, and sertraline) were systematically evaluated in addition to the literature review of recent examples. The application of PBPK modeling in drug interactions by inhibitory parent-metabolite pairs is described and guidance on strategic application is provided.
Collapse
Affiliation(s)
| | - Y Chen
- Genentech, South San Francisco, California, USA
| | - J Mao
- Genentech, South San Francisco, California, USA
| | - J Lin
- Pfizer Inc., Groton, Connecticut, USA
| | - H Yu
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | | | - M Shebley
- AbbVie Inc., North Chicago, Illinois, USA
| | - M V Varma
- Pfizer Inc., Groton, Connecticut, USA.
| |
Collapse
|
23
|
Li DQ, Zhang ZQ, Yang XL, Zhou CH, Qi JL. Online restricted-access material combined with high-performance liquid chromatography and tandem mass spectrometry for the simultaneous determination of vanillin and its vanillic acid metabolite in human plasma. J Sep Sci 2016; 39:3318-26. [PMID: 27384745 DOI: 10.1002/jssc.201600466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/16/2016] [Accepted: 06/29/2016] [Indexed: 12/15/2022]
Affiliation(s)
- De-qiang Li
- Department of Pharmacy; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Zhi-qing Zhang
- Department of Pharmacy; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Xiu-ling Yang
- Department of Pharmacy; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Chun-hua Zhou
- Department of Pharmacy; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Jin-long Qi
- Department of Pharmacology; Hebei Medical University; Shijiazhuang Hebei China
| |
Collapse
|
24
|
Eng H, Obach RS. Use of Human Plasma Samples to Identify Circulating Drug Metabolites that Inhibit Cytochrome P450 Enzymes. Drug Metab Dispos 2016; 44:1217-28. [PMID: 27271369 DOI: 10.1124/dmd.116.071084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/03/2016] [Indexed: 01/06/2023] Open
Abstract
Drug interactions elicited through inhibition of cytochrome P450 (P450) enzymes are important in pharmacotherapy. Recently, greater attention has been focused on not only parent drugs inhibiting P450 enzymes but also on possible inhibition of these enzymes by circulating metabolites. In this report, an ex vivo method whereby the potential for circulating metabolites to be inhibitors of P450 enzymes is described. To test this method, seven drugs and their known plasma metabolites were added to control human plasma at concentrations previously reported to occur in humans after administration of the parent drug. A volume of plasma for each drug based on the known inhibitory potency and time-averaged concentration of the parent drug was extracted and fractionated by high-pressure liquid chromatography-mass spectrometry, and the fractions were tested for inhibition of six human P450 enzyme activities (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). Observation of inhibition in fractions that correspond to the retention times of metabolites indicates that the metabolite has the potential to contribute to P450 inhibition in vivo. Using this approach, norfluoxetine, hydroxyitraconazole, desmethyldiltiazem, desacetyldiltiazem, desethylamiodarone, hydroxybupropion, erythro-dihydrobupropion, and threo-dihydrobupropion were identified as circulating metabolites that inhibit P450 activities at a similar or greater extent as the parent drug. A decision tree is presented outlining how this method can be used to determine when a deeper investigation of the P450 inhibition properties of a drug metabolite is warranted.
Collapse
|
25
|
Martin IJ, Hill SE, Baker JA, Deshmukh SV, Mulrooney EF. A Pharmacokinetic Modeling Approach to Predict the Contribution of Active Metabolites to Human Efficacious Dose. Drug Metab Dispos 2016; 44:1435-40. [PMID: 27260151 DOI: 10.1124/dmd.116.070391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/02/2016] [Indexed: 02/03/2023] Open
Abstract
A preclinical drug candidate, MRK-1 (Merck candidate drug parent compound), was found to elicit tumor regression in a mouse xenograft model. Analysis of samples from these studies revealed significant levels of two circulating metabolites, whose identities were confirmed by comparison with authentic standards using liquid chromatography-tandem mass spectrometry. These metabolites were found to have an in vitro potency similar to that of MRK-1 against the pharmacological target and were therefore thought to contribute to the observed efficacy. To predict this contribution in humans, a pharmacokinetic (PK) modeling approach was developed. At the mouse efficacious dose, the areas under the plasma concentration time curves (AUCs) of the active metabolites were normalized by their in vitro potency compared with MRK-1. These normalized metabolite AUCs were added to that of MRK-1 to yield a composite efficacious unbound AUC, expressed as "parent drug equivalents," which was used as the target AUC for predictions of the human efficacious dose. In vitro and preclinical PK studies afforded predictions of the PK of MRK-1 and the two active metabolites in human as well as the relative pathway flux to each metabolite. These were used to construct a PK model (Berkeley Madonna, version 8.3.18; Berkeley Madonna Inc., University of California, Berkeley, CA) and to predict the human dose required to achieve the target parent equivalent exposure. These predictions were used to inform on the feasibility of the human dose in terms of size, frequency, formulation, and likely safety margins, as well as to aid in the design of preclinical safety studies.
Collapse
Affiliation(s)
- Iain J Martin
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - Susan E Hill
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - James A Baker
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - Sujal V Deshmukh
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| | - Erin F Mulrooney
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts
| |
Collapse
|
26
|
Wang T, Yang L, Hua J, Xie H, Jiang X, Wang L. Simultaneous bioanalysis of rasagiline and its major metabolites in human plasma by LC–MS/MS: Application to a clinical pharmacokinetic study. J Pharm Biomed Anal 2016; 125:280-5. [DOI: 10.1016/j.jpba.2016.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 10/22/2022]
|
27
|
Iegre J, Hayes MA, Thompson RA, Weidolf L, Isin EM. Database Extraction of Metabolite Information of Drug Candidates: Analysis of 27 AstraZeneca Compounds with Human Absorption, Distribution, Metabolism, and Excretion Data. Drug Metab Dispos 2016; 44:732-40. [PMID: 26868617 DOI: 10.1124/dmd.115.067850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/10/2016] [Indexed: 02/13/2025] Open
Abstract
As part of the drug discovery and development process, it is important to understand the human metabolism of a candidate drug prior to clinical studies. Preclinical in vitro and in vivo experiments across species are conducted to build knowledge concerning human circulating metabolites in preparation for clinical studies; therefore, the quality of these experiments is critical. Within AstraZeneca, all metabolite identification (Met-ID) information is stored in a global database using ACDLabs software. In this study, the Met-ID information derived from in vitro and in vivo studies for 27 AstraZeneca drug candidates that underwent human absorption, distribution, metabolism, and excretion studies was extracted from the database. The retrospective analysis showed that 81% of human circulating metabolites were previously observed in preclinical in vitro and/or in vivo experiments. A detailed analysis was carried out to understand which human circulating metabolites were not captured in the preclinical experiments. Metabolites observed in human hepatocytes and rat plasma but not seen in circulation in humans (extraneous metabolites) were also investigated. The majority of human specific circulating metabolites derive from multistep biotransformation reactions that may not be observed in in vitro studies within the limited time frame in which cryopreserved hepatocytes are active. Factors leading to the formation of extraneous metabolites in preclinical studies seemed to be related to species differences with respect to transporter activity, secondary metabolism, and enzyme kinetics. This retrospective analysis assesses the predictive value of Met-ID experiments and improves our ability to discriminate between metabolites expected to circulate in humans and irrelevant metabolites seen in preclinical studies.
Collapse
Affiliation(s)
- Jessica Iegre
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit (J.I., M.A.H., L.W., E.M.I.) and Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit (R.A.T.), AstraZeneca, Mölndal, Sweden
| | - Martin A Hayes
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit (J.I., M.A.H., L.W., E.M.I.) and Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit (R.A.T.), AstraZeneca, Mölndal, Sweden
| | - Richard A Thompson
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit (J.I., M.A.H., L.W., E.M.I.) and Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit (R.A.T.), AstraZeneca, Mölndal, Sweden
| | - Lars Weidolf
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit (J.I., M.A.H., L.W., E.M.I.) and Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit (R.A.T.), AstraZeneca, Mölndal, Sweden
| | - Emre M Isin
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit (J.I., M.A.H., L.W., E.M.I.) and Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit (R.A.T.), AstraZeneca, Mölndal, Sweden
| |
Collapse
|
28
|
Methylation and its role in the disposition of tanshinol, a cardiovascular carboxylic catechol from Salvia miltiorrhiza roots (Danshen). Acta Pharmacol Sin 2015; 36:627-43. [PMID: 25891082 PMCID: PMC4422947 DOI: 10.1038/aps.2015.20] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/27/2015] [Indexed: 12/29/2022]
Abstract
AIM Tanshinol is an important catechol in the antianginal herb Salvia miltiorrhiza roots (Danshen). This study aimed to characterize tanshinol methylation. METHODS Metabolites of tanshinol were analyzed by liquid chromatography/mass spectrometry. Metabolism was assessed in vitro with rat and human enzymes. The major metabolites were synthesized for studying their interactions with drug metabolizing enzymes and transporters and their vasodilatory properties. Dose-related tanshinol methylation and its influences on tanshinol pharmacokinetics were also studied in rats. RESULTS Methylation, preferentially in the 3-hydroxyl group, was the major metabolic pathway of tanshinol. In rats, tanshinol also underwent considerable 3-O-sulfation, which appeared to be poor in human liver. These metabolites were mainly eliminated via renal excretion, which involved tubular secretion mainly by organic anion transporter (OAT) 1. The methylated metabolites had no vasodilatory activity. Entacapone-impaired methylation did not considerably increase systemic exposure to tanshinol in rats. The saturation of tanshinol methylation in rat liver could be predicted from the Michaelis constant of tanshinol for catechol-O-methyltransferase (COMT). Tanshinol had low affinity for human COMT and OATs; its methylated metabolites also had low affinity for the transporters. Tanshinol and its major human metabolite (3-O-methyltanshinol) exhibited negligible inhibitory activities against human cytochrome P450 enzymes, organic anion transporting polypeptides 1B1/1B3, multidrug resistance protein 1, multidrug resistance-associated protein 2, and breast cancer resistance protein. CONCLUSION Tanshinol is mainly metabolized via methylation. Tanshinol and its major human metabolite have low potential for pharmacokinetic interactions with synthetic antianginal agents. This study will help define the risk of hyperhomocysteinemia related to tanshinol methylation.
Collapse
|
29
|
Influence of HIV antiretrovirals on methadone N-demethylation and transport. Biochem Pharmacol 2015; 95:115-25. [PMID: 25801005 DOI: 10.1016/j.bcp.2015.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/12/2015] [Indexed: 02/02/2023]
Abstract
Drug interactions involving methadone and/or HIV antiretrovirals can be problematic. Mechanisms whereby antiretrovirals induce clinical methadone clearance are poorly understood. Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. This investigation evaluated human hepatocytes as a model for methadone induction, and tested the hypothesis that methadone and EDDP are substrates for human drug transporters. Human hepatocyte induction by several antiretrovirals of methadone N-demethylation, and CYP2B6 and CYP3A4 transcription, protein expression and catalytic activity, and pregnane X receptor (PXR) activation were evaluated. Methadone and EDDP uptake and efflux by overexpressed transporters were also determined. Methadone N-demethylation was generally not significantly increased by the antiretrovirals. CYP2B6 mRNA and activity (bupropion N-demethylation) were induced by several antiretrovirals, as were CYP3A4 mRNA and protein expression, but only indinavir increased CYP3A activity (alfentanil dealkylation). CYP upregulation appeared related to PXR activation. Methadone was not a substrate for uptake (OCT1, OCT2, OCT3, OATP1A2, OATP1B1, OATP1B3, OATP2B1) or efflux (P-gp, BCRP) transporters. EDDP was a good substrate for P-gp, BCRP, OCT1, OCT3, OATP1A2, and OATP1B1. OATP1A2- and OCT3-mediated EDDP uptake, and BCRP-mediated EDDP efflux transport, was inhibited by several antiretrovirals. Results show that hepatocyte methadone N-demethylation resembles expressed and liver microsomal metabolism more than clinical metabolism. Compared with clinical studies, hepatocytes underreport induction of methadone metabolism by HIV drugs. Hepatocytes are not a good predictive model for clinical antiretroviral induction of methadone metabolism and not a substitute for clinical studies. EDDP is a transporter substrate, and is susceptible to transporter-mediated interactions.
Collapse
|
30
|
Simultaneous determination of ornidazole and its main metabolites in human plasma by LC–MS/MS: application to a pharmacokinetic study. Bioanalysis 2014; 6:2343-56. [PMID: 25384588 DOI: 10.4155/bio.14.117] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Ornidazole is a 5-nitroimidazole antimicrobial agent used for almost 40 years. A novel LC–MS/MS assay was developed and validated for the simultaneous determination of ornidazole and its main metabolites (M3, M6, M16–1, and M16–2) in human plasma. Results: After extraction from 100 μl of plasma by protein precipitation with acetonitrile, all the analytes were separated on a Capcell PAK MG C18 column (100 × 4.6 mm, 5 μm) within 5.0 min and detected by ESI-MS/MS in the positive mode. The validation results met the acceptance criteria as per the US FDA and EMA guidelines. Conclusion: The validated method was successfully applied to a pharmacokinetic study after oral administration of 1000 mg ornidazole to six healthy Chinese volunteers.
Collapse
|
31
|
Smith BJ, Pithavala Y, Bu HZ, Kang P, Hee B, Deese AJ, Pool WF, Klamerus KJ, Wu EY, Dalvie DK. Pharmacokinetics, metabolism, and excretion of [14C]axitinib, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in humans. Drug Metab Dispos 2014; 42:918-31. [PMID: 24608633 DOI: 10.1124/dmd.113.056531] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The disposition of a single oral dose of 5 mg (100 μCi) of [(14)C]axitinib was investigated in fasted healthy human subjects (N = 8). Axitinib was rapidly absorbed, with a median plasma Tmax of 2.2 hours and a geometric mean Cmax and half-life of 29.2 ng/ml and 10.6 hours, respectively. The plasma total radioactivity-time profile was similar to that of axitinib but the AUC was greater, suggesting the presence of metabolites. The major metabolites in human plasma (0-12 hours), identified as axitinib N-glucuronide (M7) and axitinib sulfoxide (M12), were pharmacologically inactive, and with axitinib comprised 50.4%, 16.2%, and 22.5% of the radioactivity, respectively. In excreta, the majority of radioactivity was recovered in most subjects by 48 hours postdose. The median radioactivity excreted in urine, feces, and total recovery was 22.7%, 37.0%, and 59.7%, respectively. The recovery from feces was variable across subjects (range, 2.5%-60.2%). The metabolites identified in urine were M5 (carboxylic acid), M12 (sulfoxide), M7 (N-glucuronide), M9 (sulfoxide/N-oxide), and M8a (methylhydroxy glucuronide), accounting for 5.7%, 3.5%, 2.6%, 1.7%, and 1.3% of the dose, respectively. The drug-related products identified in feces were unchanged axitinib, M14/15 (mono-oxidation/sulfone), M12a (epoxide), and an unidentified metabolite, comprising 12%, 5.7%, 5.1%, and 5.0% of the dose, respectively. The proposed mechanism to form M5 involved a carbon-carbon bond cleavage via M12a, followed by rearrangement to a ketone intermediate and subsequent Baeyer-Villiger rearrangement, possibly through a peroxide intermediate. In summary, the study characterized axitinib metabolites in circulation and primary elimination pathways of the drug, which were mainly oxidative in nature.
Collapse
Affiliation(s)
- Bill J Smith
- Pharmacokinetics, Dynamics and Metabolism (B.J.S., H.-Z.B., P.K., W.F.P., E.Y.W., D.K.D.), Pfizer Oncology-Clinical Pharmacology (Y.P., B.H., K.J.K.), and Pharmaceutical Sciences (A.J.D.), Pfizer Inc., Worldwide Research and Development, La Jolla Laboratories, San Diego, CA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zamek-Gliszczynski MJ, Chu X, Polli JW, Paine MF, Galetin A. Understanding the transport properties of metabolites: case studies and considerations for drug development. Drug Metab Dispos 2014; 42:650-64. [PMID: 24346835 DOI: 10.1124/dmd.113.055558] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Recent analyses demonstrated that metabolites are unlikely to contribute significantly to clinical inhibition of cytochrome P450 (P450)-mediated drug metabolism, and that only ∼2% of this type of drug interaction could not be predicted from the parent drug alone. Due to generally increased polarity and decreased permeability, metabolites are less likely to interact with P450s, but their disposition is instead more likely to involve transporters. This commentary presents case studies illustrating the potential importance of transporters as determinants of metabolite disposition, and as sites of drug interactions, which may alter drug efficacy and safety. Many of these examples are hydrophilic phase II conjugates involved in enterohepatic cycling, where modulation of transporter-dependent disposition may alter pharmacokinetics/pharmacodynamics. The case studies suggest that characterization of metabolite disposition, toxicology, and pharmacology should not focus solely on metabolites with appreciable systemic exposure, but should take into consideration major excretory metabolites. A more thorough understanding of metabolite (phase I and II; circulating and excreted) transport properties during drug development may provide an improved understanding of complex drug-drug interactions (DDIs) that can alter drug and/or metabolite systemic and intracellular exposure. Knowledge and capability gaps remain in clinical translation of in vitro and animal data regarding metabolite disposition. To this end, useful experimental and modeling approaches are highlighted. Application of these tools may lead to a better understanding of metabolite victim and perpetrator DDI potential, and ultimately the establishment of approaches for prediction of pharmacodynamic and toxicodynamic consequences of metabolite transport modulation.
Collapse
Affiliation(s)
- Maciej J Zamek-Gliszczynski
- Drug Disposition, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana (M.J.Z.-G.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline Inc., Research Triangle Park, North Carolina (J.W.P.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.)
| | | | | | | | | |
Collapse
|
33
|
Haglund J, Halldin MM, Brunnström Å, Eklund G, Kautiainen A, Sandholm A, Iverson SL. Pragmatic Approaches to Determine the Exposures of Drug Metabolites in Preclinical and Clinical Subjects in the MIST Evaluation of the Clinical Development Phase. Chem Res Toxicol 2014; 27:601-10. [DOI: 10.1021/tx400449z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Johanna Haglund
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | | | - Åsa Brunnström
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | - Göran Eklund
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | | | - Anna Sandholm
- AstraZeneca R&D, DMPK Södertälje, SE-151 85 Södertälje, Sweden
| | | |
Collapse
|
34
|
A reflection on fit-for-purpose metabolite investigation at different stages of drug development. Bioanalysis 2014; 6:591-4. [DOI: 10.4155/bio.14.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|