1
|
Jangjou A, Moqadas M, Mohsenian L, Kamyab H, Chelliapan S, Alshehery S, Ali MA, Dehbozorgi F, Yadav KK, Khorami M, Zarei Jelyani N. Awareness raising and dealing with methanol poisoning based on effective strategies. ENVIRONMENTAL RESEARCH 2023; 228:115886. [PMID: 37072082 DOI: 10.1016/j.envres.2023.115886] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023]
Abstract
Intoxication with methanol most commonly occurs as a consequence of ingesting, inhaling, or coming into contact with formulations that include methanol as a base. Clinical manifestations of methanol poisoning include suppression of the central nervous system, gastrointestinal symptoms, and decompensated metabolic acidosis, which is associated with impaired vision and either early or late blindness within 0.5-4 h after ingestion. After ingestion, methanol concentrations in the blood that are greater than 50 mg/dl should raise some concern. Ingested methanol is typically digested by alcohol dehydrogenase (ADH), and it is subsequently redistributed to the body's water to attain a volume distribution that is about equivalent to 0.77 L/kg. Moreover, it is removed from the body as its natural, unchanged parent molecules. Due to the fact that methanol poisoning is relatively uncommon but frequently involves a large number of victims at the same time, this type of incident occupies a special position in the field of clinical toxicology. The beginning of the COVID-19 pandemic has resulted in an increase in erroneous assumptions regarding the preventative capability of methanol in comparison to viral infection. More than 1000 Iranians fell ill, and more than 300 of them passed away in March of this year after they consumed methanol in the expectation that it would protect them from a new coronavirus. The Atlanta epidemic, which involved 323 individuals and resulted in the deaths of 41, is one example of mass poisoning. Another example is the Kristiansand outbreak, which involved 70 people and resulted in the deaths of three. In 2003, the AAPCC received reports of more than one thousand pediatric exposures. Since methanol poisoning is associated with high mortality rates, it is vital that the condition be addressed seriously and managed as quickly as feasible. The objective of this review was to raise awareness about the mechanism and metabolism of methanol toxicity, the introduction of therapeutic interventions such as gastrointestinal decontamination and methanol metabolism inhibition, the correction of metabolic disturbances, and the establishment of novel diagnostic/screening nanoparticle-based strategies for methanol poisoning such as the discovery of ADH inhibitors as well as the detection of the adulteration of alcoholic drinks by nanoparticles in order to prevent methanol poisoning. In conclusion, increasing warnings and knowledge about clinical manifestations, medical interventions, and novel strategies for methanol poisoning probably results in a decrease in the death load.
Collapse
Affiliation(s)
- Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Moqadas
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Mohsenian
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Faculty of Architecture and Urbanism, UTE University, Calle Rumipamba S/N and Bourgeois, Quito, Ecuador; Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600 077, India; Process Systems Engineering Centre (PROSPECT), Faculty of Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia.
| | - Shreeshivadasan Chelliapan
- Engineering Department, Razak Faculty of Technology and Informatics, Universiti Teknologi Malaysia, Jln Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Sultan Alshehery
- Department of Mechanical Engineering King Khalid University, zip code - 62217, Saudi Arabia
| | - Mohammed Azam Ali
- Department of Mechanical Engineering King Khalid University, zip code - 62217, Saudi Arabia
| | - Farbod Dehbozorgi
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Krishna Kumar Yadav
- Faculty of Science and Technology, Madhyanchal Professional University, Ratibad, Bhopal, 462044, India; Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Nasiriyah, 64001, Iraq
| | - Masoud Khorami
- Department of Civil Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Najmeh Zarei Jelyani
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran; Emergency Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Beers J, Authement AK, Isoherranen N, Jackson KD. Cytosolic Enzymes Generate Cannabinoid Metabolites 7-Carboxycannabidiol and 11-Nor-9-carboxytetrahydrocannabinol. ACS Med Chem Lett 2023; 14:614-620. [PMID: 37197460 PMCID: PMC10184666 DOI: 10.1021/acsmedchemlett.3c00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
The cannabinoids cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) undergo extensive oxidative metabolism in the liver. Although cytochromes P450 form the primary, pharmacologically active, hydroxylated metabolites of CBD and THC, less is known about the enzymes that generate the major in vivo circulating metabolites of CBD and THC, 7-carboxy-CBD and 11-carboxy-THC, respectively. The purpose of this study was to elucidate the enzymes involved in forming these metabolites. Cofactor dependence experiments with human liver subcellular fractions revealed that 7-carboxy-CBD and 11-carboxy-THC formation is largely dependent on cytosolic NAD+-dependent enzymes, with lesser contributions from NADPH-dependent microsomal enzymes. Experiments with chemical inhibitors provided evidence that 7-carboxy-CBD formation is mainly dependent on aldehyde dehydrogenases and 11-carboxy-THC formation is mediated also in part by aldehyde oxidase. This study is the first to demonstrate the involvement of cytosolic drug-metabolizing enzymes in generating major in vivo metabolites of CBD and THC and addresses a knowledge gap in cannabinoid metabolism.
Collapse
Affiliation(s)
- Jessica
L. Beers
- Division
of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Aurora K. Authement
- Department
of Pharmaceutics, University of Washington
School of Pharmacy, Seattle, Washington 98195, United States
| | - Nina Isoherranen
- Department
of Pharmaceutics, University of Washington
School of Pharmacy, Seattle, Washington 98195, United States
| | - Klarissa D. Jackson
- Division
of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
3
|
Sharma R, Dowling MS, Futatsugi K, Kalgutkar AS. Mitigating a Bioactivation Liability with an Azetidine-Based Inhibitor of Diacylglycerol Acyltransferase 2 (DGAT2) En Route to the Discovery of the Clinical Candidate Ervogastat. Chem Res Toxicol 2023. [PMID: 37148271 DOI: 10.1021/acs.chemrestox.3c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We recently disclosed SAR studies on systemically acting, amide-based inhibitors of diacylglycerol acyltransferase 2 (DGAT2) that addressed metabolic liabilities with the liver-targeted DGAT2 inhibitor PF-06427878. Despite strategic placement of a nitrogen atom in the dialkoxyaromatic ring in PF-06427878 to evade oxidative O-dearylation, metabolic intrinsic clearance remained high due to extensive piperidine ring oxidation as exemplified with compound 1. Piperidine ring modifications through alternate N-linked heterocyclic ring/spacer combination led to azetidine 2 that demonstrated lower intrinsic clearance. However, 2 underwent a facile cytochrome P450 (CYP)-mediated α-carbon oxidation followed by azetidine ring scission, resulting in the formation of ketone (M2) and aldehyde (M6) as stable metabolites in NADPH-supplemented human liver microsomes. Inclusion of GSH or semicarbazide in microsomal incubations led to the formation of Cys-Gly-thiazolidine (M3), Cys-thiazolidine (M5), and semicarbazone (M7) conjugates, which were derived from reaction of the nucleophilic trapping agents with aldehyde M6. Metabolites M2 and M5 were biosynthesized from NADPH- and l-cysteine-fortified human liver microsomal incubations with 2, and proposed metabolite structures were verified using one- and two-dimensional NMR spectroscopy. Replacement of the azetidine substituent with a pyridine ring furnished 8, which mitigated the formation of the electrophilic aldehyde metabolite, and was a more potent DGAT2 inhibitor than 2. Further structural refinements in 8, specifically introducing amide bond substituents with greater metabolic stability, led to the discovery of PF-06865571 (ervogastat) that is currently in phase 2 clinical trials for the treatment of nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Raman Sharma
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S Dowling
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kentaro Futatsugi
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, 1 Portland St, Cambridge, Massachusetts 02139, United States
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, 1 Portland St, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
4
|
A novel approach for the bioanalysis of short-lived aldehydes. Bioanalysis 2022; 14:1317-1326. [PMID: 36541259 DOI: 10.4155/bio-2022-0187] [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: 12/24/2022] Open
Abstract
Background: The instability of aldehydes in biological matrices is associated with their reactions with thiol and amino moieties in proteins. This chemical reaction is reversible by nature and highly pH dependent. Method: A novel approach that includes protein precipitation with an acidic solution of acetonitrile/water/formic acid (85/14/1; v/v/v) was developed to efficiently recover Aldehyde-1 from plasma by shifting the equilibrium toward the formation of the free form. Results: This enabled the support of two GLP studies where Aldehyde-1 was administered to mice. The recovery of Aldehyde-1 from plasma exceeded 88% at three concentration levels. Plasma stability was confirmed at ambient conditions for 24 h and in the freezer for at least 43 (-20°C) and 64 (-70°C) days.
Collapse
|
5
|
Traccis F, Presciuttini R, Pani PP, Sinclair JMA, Leggio L, Agabio R. Alcohol-medication interactions: A systematic review and meta-analysis of placebo-controlled trials. Neurosci Biobehav Rev 2021; 132:519-541. [PMID: 34826511 DOI: 10.1016/j.neubiorev.2021.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 09/20/2021] [Accepted: 11/15/2021] [Indexed: 11/28/2022]
Abstract
Alcohol and other xenobiotics may limit the therapeutic effects of medications. We aimed at investigating alcohol-medication interactions (AMI) after the exclusion of confounding effects related to other xenobiotics. We performed a systematic review and meta-analysis of controlled studies comparing the effects induced by alcohol versus placebo on pharmacodynamic and/or pharmacokinetic parameters of approved medications. Certainty in the evidence of AMI was assessed when at least 3 independent studies and at least 200 participants were available. We included 107 articles (3097 participants): for diazepam, cannabis, opioids, and methylphenidate, we found significant AMI and enough data to assign the certainty of evidence. Alcohol consumption significantly increases the peak plasma concentration of diazepam (low certainty; almost 290 participants), cannabis (high certainty; almost 650 participants), opioids (low certainty; 560 participants), and methylphenidate (moderate certainty; 290 participants). For most medications, we found some AMI but not enough data to assign them the certainty grades; for some medications, we found no differences between alcohol and placebo in any outcomes evaluated. Our results add further evidence for interactions between alcohol and certain medications after the exclusion of confounding effects related to other xenobiotics. Physicians should advise patients who use these specific medications to avoid alcohol consumption. Further studies with appropriate control groups, enough female participants to investigate sex differences, and elderly population are needed to expand our knowledge in this field. Short phrases suitable for indexing terms.
Collapse
Affiliation(s)
- Francesco Traccis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| | - Riccardo Presciuttini
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| | - Pier Paolo Pani
- Health Social Services Public Health Trust Sardinia, Cagliari, Italy.
| | | | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Basic Research, National Institutes of Health, Baltimore and Bethesda, MD, United States; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, United States; Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States; Division of Addiction Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States; Department of Neuroscience, Georgetown University, Washington, DC, United States.
| | - Roberta Agabio
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
6
|
Non-cytochrome P450 enzymes involved in the oxidative metabolism of xenobiotics: Focus on the regulation of gene expression and enzyme activity. Pharmacol Ther 2021; 233:108020. [PMID: 34637840 DOI: 10.1016/j.pharmthera.2021.108020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Oxidative metabolism is one of the major biotransformation reactions that regulates the exposure of xenobiotics and their metabolites in the circulatory system and local tissues and organs, and influences their efficacy and toxicity. Although cytochrome (CY)P450s play critical roles in the oxidative reaction, extensive CYP450-independent oxidative metabolism also occurs in some xenobiotics, such as aldehyde oxidase, xanthine oxidoreductase, flavin-containing monooxygenase, monoamine oxidase, alcohol dehydrogenase, or aldehyde dehydrogenase-dependent oxidative metabolism. Drugs form a large portion of xenobiotics and are the primary target of this review. The common reaction mechanisms and roles of non-CYP450 enzymes in metabolism, factors affecting the expression and activity of non-CYP450 enzymes in terms of inhibition, induction, regulation, and species differences in pharmaceutical research and development have been summarized. These non-CYP450 enzymes are detoxifying enzymes, although sometimes they mediate severe toxicity. Synthetic or natural chemicals serve as inhibitors for these non-CYP450 enzymes. However, pharmacokinetic-based drug interactions through these inhibitors have rarely been reported in vivo. Although multiple mechanisms participate in the basal expression and regulation of non-CYP450 enzymes, only a limited number of inducers upregulate their expression. Therefore, these enzymes are considered non-inducible or less inducible. Overall, this review focuses on the potential xenobiotic factors that contribute to variations in gene expression levels and the activities of non-CYP450 enzymes.
Collapse
|
7
|
Fukami T, Yokoi T, Nakajima M. Non-P450 Drug-Metabolizing Enzymes: Contribution to Drug Disposition, Toxicity, and Development. Annu Rev Pharmacol Toxicol 2021; 62:405-425. [PMID: 34499522 DOI: 10.1146/annurev-pharmtox-052220-105907] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most clinically used drugs are metabolized in the body via oxidation, reduction, or hydrolysis reactions, which are considered phase I reactions. Cytochrome P450 (P450) enzymes, which primarily catalyze oxidation reactions, contribute to the metabolism of over 50% of clinically used drugs. In the last few decades, the function and regulation of P450s have been extensively studied, whereas the characterization of non-P450 phase I enzymes is still incomplete. Recent studies suggest that approximately 30% of drug metabolism is carried out by non-P450 enzymes. This review summarizes current knowledge of non-P450 phase I enzymes, focusing on their roles in controlling drug efficacy and adverse reactions as an important aspect of drug development. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, and WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, and WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
8
|
Jee A, Sernoskie SC, Uetrecht J. Idiosyncratic Drug-Induced Liver Injury: Mechanistic and Clinical Challenges. Int J Mol Sci 2021; 22:ijms22062954. [PMID: 33799477 PMCID: PMC7998339 DOI: 10.3390/ijms22062954] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (IDILI) remains a significant problem for patients and drug development. The idiosyncratic nature of IDILI makes mechanistic studies difficult, and little is known of its pathogenesis for certain. Circumstantial evidence suggests that most, but not all, IDILI is caused by reactive metabolites of drugs that are bioactivated by cytochromes P450 and other enzymes in the liver. Additionally, there is overwhelming evidence that most IDILI is mediated by the adaptive immune system; one example being the association of IDILI caused by specific drugs with specific human leukocyte antigen (HLA) haplotypes, and this may in part explain the idiosyncratic nature of these reactions. The T cell receptor repertoire likely also contributes to the idiosyncratic nature. Although most of the liver injury is likely mediated by the adaptive immune system, specifically cytotoxic CD8+ T cells, adaptive immune activation first requires an innate immune response to activate antigen presenting cells and produce cytokines required for T cell proliferation. This innate response is likely caused by either a reactive metabolite or some form of cell stress that is clinically silent but not idiosyncratic. If this is true it would make it possible to study the early steps in the immune response that in some patients can lead to IDILI. Other hypotheses have been proposed, such as mitochondrial injury, inhibition of the bile salt export pump, unfolded protein response, and oxidative stress although, in most cases, it is likely that they are also involved in the initiation of an immune response rather than representing a completely separate mechanism. Using the clinical manifestations of liver injury from a number of examples of IDILI-associated drugs, this review aims to summarize and illustrate these mechanistic hypotheses.
Collapse
Affiliation(s)
- Alison Jee
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | | | - Jack Uetrecht
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada;
- Correspondence:
| |
Collapse
|
9
|
Di L, Balesano A, Jordan S, Shi SM. The Role of Alcohol Dehydrogenase in Drug Metabolism: Beyond Ethanol Oxidation. AAPS JOURNAL 2021; 23:20. [DOI: 10.1208/s12248-020-00536-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/17/2020] [Indexed: 02/08/2023]
|
10
|
Scimone C, Alibrandi S, Donato L, Giofrè SV, Rao G, Sidoti A, D'Angelo R. Antiretroviral treatment leading to secondary trimethylaminuria: Genetic associations and successful management with riboflavin. J Clin Pharm Ther 2020; 46:304-309. [PMID: 33247860 DOI: 10.1111/jcpt.13315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Trimethylaminuria is a metabolic disorder characterized by excessive excretion of trimethylamine in body fluids following FMO3 gene mutations. Secondary forms of the disease may be due to consumption of trimethylamine precursor-rich foods or metabolism of some xenobiotics. CASE SUMMARY A HIV patient developed secondary trimethylaminuria following antiretroviral treatment. Riboflavin supplementation ameliorated his phenotype. 1 H-NMR confirmed increased urine level of TMA. Several genes involved in choline catabolism harboured missense mutations. Riboflavin supplement improved enzymatic activity of mutated enzymes promoting TMA clearance. WHAT IS NEW AND CONCLUSION Antiretrovirals may increase the concentration of TMA precursors. The present study reports antiretroviral treatment as risk factor for such secondary trimethylaminuria. Riboflavin is an effective treatment.
Collapse
Affiliation(s)
- Concetta Scimone
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy.,Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T, Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy.,Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Luigi Donato
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy.,Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T, Palermo, Italy
| | - Salvatore V Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Giacomo Rao
- Prevention and Research division, INAIL, Rome, Italy
| | - Antonina Sidoti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy.,Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T, Palermo, Italy
| | - Rosalia D'Angelo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy.,Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T, Palermo, Italy
| |
Collapse
|
11
|
Shanu-Wilson J, Evans L, Wrigley S, Steele J, Atherton J, Boer J. Biotransformation: Impact and Application of Metabolism in Drug Discovery. ACS Med Chem Lett 2020; 11:2087-2107. [PMID: 33214818 DOI: 10.1021/acsmedchemlett.0c00202] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
Biotransformation has a huge impact on the efficacy and safety of drugs. Ultimately the effects of metabolism can be the lynchpin in the discovery and development cycle of a new drug. This article discusses the impact and application of biotransformation of drugs by mammalian systems, microorganisms, and recombinant enzymes, covering active and reactive metabolites, the impact of the gut microbiome on metabolism, and how insights gained from biotransformation studies can influence drug design from the combined perspectives of a CRO specializing in a range of biotransformation techniques and pharma biotransformation scientists. We include a commentary on how biology-driven approaches can complement medicinal chemistry strategies in drug optimization and the in vitro and surrogate systems available to explore and exploit biotransformation.
Collapse
Affiliation(s)
- Julia Shanu-Wilson
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Abingdon, Oxfordshire OX14 4SD, U.K
| | - Liam Evans
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Abingdon, Oxfordshire OX14 4SD, U.K
| | - Stephen Wrigley
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Abingdon, Oxfordshire OX14 4SD, U.K
| | - Jonathan Steele
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Abingdon, Oxfordshire OX14 4SD, U.K
| | - James Atherton
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, Delaware 19803, United States
| | - Jason Boer
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, Delaware 19803, United States
| |
Collapse
|
12
|
Gampe C, Verma VA. Curse or Cure? A Perspective on the Developability of Aldehydes as Active Pharmaceutical Ingredients. J Med Chem 2020; 63:14357-14381. [DOI: 10.1021/acs.jmedchem.0c01177] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Christian Gampe
- Genentech, 1 DNA Way, South San Francisco, 94080 California, United States
| | - Vishal A. Verma
- Genentech, 1 DNA Way, South San Francisco, 94080 California, United States
| |
Collapse
|
13
|
Naisbitt DJ, Olsson‐Brown A, Gibson A, Meng X, Ogese MO, Tailor A, Thomson P. Immune dysregulation increases the incidence of delayed-type drug hypersensitivity reactions. Allergy 2020; 75:781-797. [PMID: 31758810 DOI: 10.1111/all.14127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/05/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022]
Abstract
Delayed-type, T cell-mediated, drug hypersensitivity reactions are a serious unwanted manifestation of drug exposure that develops in a small percentage of the human population. Drugs and drug metabolites are known to interact directly and indirectly (through irreversible protein binding and processing to the derived adducts) with HLA proteins that present the drug-peptide complex to T cells. Multiple forms of drug hypersensitivity are strongly linked to expression of a single HLA allele, and there is increasing evidence that drugs and peptides interact selectively with the protein encoded by the HLA allele. Despite this, many individuals expressing HLA risk alleles do not develop hypersensitivity when exposed to culprit drugs suggesting a nonlinear, multifactorial relationship in which HLA risk alleles are one factor. This has prompted a search for additional susceptibility factors. Herein, we argue that immune regulatory pathways are one key determinant of susceptibility. As expression and activity of these pathways are influenced by disease, environmental and patient factors, it is currently impossible to predict whether drug exposure will result in a health benefit, hypersensitivity or both. Thus, a concerted effort is required to investigate how immune dysregulation influences susceptibility towards drug hypersensitivity.
Collapse
Affiliation(s)
- Dean J. Naisbitt
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Anna Olsson‐Brown
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Andrew Gibson
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Monday O. Ogese
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Arun Tailor
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| | - Paul Thomson
- MRC Centre for Drug Safety Science Department of Clinical and Molecular Pharmacology The University of Liverpool Liverpool UK
| |
Collapse
|
14
|
Fodor J, Riley BT, Kass I, Buckle AM, Borg NA. The Role of Conformational Dynamics in Abacavir-Induced Hypersensitivity Syndrome. Sci Rep 2019; 9:10523. [PMID: 31324847 PMCID: PMC6642150 DOI: 10.1038/s41598-019-47001-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/04/2019] [Indexed: 02/06/2023] Open
Abstract
Abacavir is an antiretroviral drug used to reduce human immunodeficiency virus (HIV) replication and decrease the risk of developing acquired immune deficiency syndrome (AIDS). However, its therapeutic value is diminished by the fact that it is associated with drug hypersensitivity reactions in up to 8% of treated patients. This hypersensitivity is strongly associated with patients carrying human leukocyte antigen (HLA)-B*57:01, but not patients carrying closely related alleles. Abacavir’s specificity to HLA-B*57:01 is attributed to its binding site within the peptide-binding cleft and subsequent influence of the repertoire of peptides that can bind HLA-B*57:01. To further our understanding of abacavir-induced hypersensitivity we used molecular dynamics (MD) to analyze the dynamics of three different peptides bound to HLA-B*57:01 in the presence and absence of abacavir or abacavir analogues. We found that abacavir and associated peptides bind to HLA-B*57:01 in a highly diverse range of conformations that are not apparent from static crystallographic snapshots, but observed no difference in either the conformations, nor degree of flexibility when compared to abacavir-unbound systems. Our results support hypersensitivity models in which abacavir-binding alters the conformational ensemble of neopeptides, so as to favour exposed peptide surfaces that are no longer recognized as self by circulating CD8+ T cells, and are conducive to TCR binding. Our findings highlight the need to also consider the role of dynamics in understanding drug-induced hypersensitivities at the molecular and mechanistic level. This additional insight can help inform the chemical modification of abacavir to prevent hypersensitivity reactions in HLA-B*57:01+ HIV patients whilst retaining potent antiretroviral activity.
Collapse
Affiliation(s)
- James Fodor
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Blake T Riley
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Itamar Kass
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.,The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Ashley M Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Natalie A Borg
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
15
|
Li H, Toth E, Cherrington NJ. Alcohol Metabolism in the Progression of Human Nonalcoholic Steatohepatitis. Toxicol Sci 2018; 164:428-438. [PMID: 29718361 PMCID: PMC6659028 DOI: 10.1093/toxsci/kfy106] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Alcohol metabolism is a well-characterized biological process that is dominated by the alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) families. Nonalcoholic steatohepatitis (NASH) is the advanced inflammatory stage of nonalcoholic fatty liver disease (NAFLD) and is known to alter the metabolism and disposition of numerous drugs. The purpose of this study was to investigate the alterations in alcohol metabolism processes in response to human NASH progression. Expression and function of ADHs, ALDHs, and catalase were examined in normal, steatosis, NASH (fatty) and NASH (not fatty) human liver samples. ALDH4A1 mRNA was significantly decreased in both NASH groups, while no significant changes were observed in the mRNA levels of other alcohol-related enzymes. The protein levels of ADH1A, ADH1B, and ADH4 were each decreased in the NASH groups, which was consistent with a decreased overall ADH activity. The protein level of ALDH2 was significantly increased in both NASH groups, while ALDH1A1 and ALDH1B1 were only decreased in NASH (fatty) samples. ALDH activity represented by oxidation of acetaldehyde was decreased in the NASH (fatty) group. The protein level of catalase was decreased in both NASH groups, though activity was unchanged. Furthermore, the significant accumulation of 4-hydroxynonenal protein adduct in NASH indicated significant oxidative stress and a potential reduction in ALDH activity. Collectively, ADH and ALDH expression and function are profoundly altered in the progression of NASH, which may have a notable impact on ADH- and ALDH-associated cellular metabolism processes and lead to significant alterations in drug metabolism mediated by these enzymes.
Collapse
Affiliation(s)
- Hui Li
- Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Erica Toth
- Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | | |
Collapse
|
16
|
Fődi T, Ignácz G, Decsi B, Béni Z, Túrós GI, Kupai J, Weiser DB, Greiner I, Huszthy P, Balogh GT. Biomimetic Synthesis of Drug Metabolites in Batch and Continuous-Flow Reactors. Chemistry 2018; 24:9385-9392. [DOI: 10.1002/chem.201800892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Tamás Fődi
- Compound Profiling Laboratory; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
- Department of Organic Chemistry and Technology; Budapest University of Technology and Economics; Műegyetem rkp. 3 1111 Budapest Hungary
| | - Gergő Ignácz
- Compound Profiling Laboratory; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
- Department of Organic Chemistry and Technology; Budapest University of Technology and Economics; Műegyetem rkp. 3 1111 Budapest Hungary
| | - Balázs Decsi
- Compound Profiling Laboratory; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
- Department of Organic Chemistry and Technology; Budapest University of Technology and Economics; Műegyetem rkp. 3 1111 Budapest Hungary
| | - Zoltán Béni
- Spectroscopic Research Department; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
| | - György I. Túrós
- Medicinal Chemistry Laboratory II; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
| | - József Kupai
- Department of Organic Chemistry and Technology; Budapest University of Technology and Economics; Műegyetem rkp. 3 1111 Budapest Hungary
| | - Diána Balogh Weiser
- Department of Organic Chemistry and Technology; Budapest University of Technology and Economics; Műegyetem rkp. 3 1111 Budapest Hungary
- Department of Physical Chemistry and Materials Science; Budapest University of Technology and Economics; Budafoki út 8 1111 Budapest Hungary
| | - István Greiner
- Research Directorate, Chemical Works; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
| | - Péter Huszthy
- Department of Organic Chemistry and Technology; Budapest University of Technology and Economics; Műegyetem rkp. 3 1111 Budapest Hungary
| | - György T. Balogh
- Compound Profiling Laboratory; Gedeon Richter Plc; Gyömrői út 19-21 1103 Budapest Hungary
| |
Collapse
|
17
|
Shi SM, Di L. The role of carbonyl reductase 1 in drug discovery and development. Expert Opin Drug Metab Toxicol 2017; 13:859-870. [DOI: 10.1080/17425255.2017.1356820] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | - Li Di
- Pfizer Inc., Groton, CT, USA
| |
Collapse
|
18
|
Wu X, Zhang Q, Guo J, Jia Y, Zhang Z, Zhao M, Yang Y, Wang B, Hu J, Sheng L, Li Y. Metabolism of F18, a Derivative of Calanolide A, in Human Liver Microsomes and Cytosol. Front Pharmacol 2017; 8:479. [PMID: 28769808 PMCID: PMC5515859 DOI: 10.3389/fphar.2017.00479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/04/2017] [Indexed: 12/02/2022] Open
Abstract
10-Chloromethyl-11-demethyl-12-oxo-calanolide (F18), an analog of calanolide A, is a novel potent nonnucleoside reverse transcriptase inhibitor against HIV-1. Here, we report the metabolic profile and the results of associated biochemical studies of F18 in vitro and in vivo. The metabolites of F18 were identified based on liquid chromatography-electrospray ionization mass spectrometry and/or nuclear magnetic resonance. Twenty-three metabolites of F18 were observed in liver microsomes in vitro. The metabolism of F18 involved 4-propyl chain oxidation, 10-chloromethyl oxidative dechlorination and 12-carbonyl reduction. Three metabolites (M1, M3-1, and M3-2) were also found in rat blood after oral administration of F18 and the reduction metabolites M3-1 and M3-2 were found to exhibit high potency for the inhibition of HIV-1 in vitro. The oxidative metabolism of F18 was mainly catalyzed by cytochrome P450 3A4 in human microsomes, whereas flavin-containing monooxygenases and 11β-hydroxysteroid dehydrogenase were found to be involved in its carbonyl reduction. In human cytosol, multiple carbonyl reductases, including aldo-keto reductase 1C, short-chain dehydrogenases/reductases and quinone oxidoreductase 1, were demonstrated to be responsible for F18 carbonyl reduction. In conclusion, the in vitro metabolism of F18 involves multiple drug metabolizing enzymes, and several metabolites exhibited anti-HIV-1 activities. Notably, the described results provide the first demonstration of the capability of FMOs for carbonyl reduction.
Collapse
Affiliation(s)
- Xiangmeng Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Qinghao Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Jiamei Guo
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yufei Jia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Ziqian Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Manman Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yakun Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Baolian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Li Sheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| |
Collapse
|
19
|
Dixit VA, Lal LA, Agrawal SR. Recent advances in the prediction of non‐
CYP450
‐mediated drug metabolism. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2017. [DOI: 10.1002/wcms.1323] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vaibhav A. Dixit
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management (SPTM)Shri Vile Parle Kelavani Mandal's (SVKM's), Narsee Monjee Institute of Management Studies (NMIMS)ShirpurIndia
| | - L. Arun Lal
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management (SPTM)Shri Vile Parle Kelavani Mandal's (SVKM's), Narsee Monjee Institute of Management Studies (NMIMS)ShirpurIndia
| | - Simran R. Agrawal
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management (SPTM)Shri Vile Parle Kelavani Mandal's (SVKM's), Narsee Monjee Institute of Management Studies (NMIMS)ShirpurIndia
| |
Collapse
|
20
|
Smith JM, Flexner C. The challenge of polypharmacy in an aging population and implications for future antiretroviral therapy development. AIDS 2017; 31 Suppl 2:S173-S184. [PMID: 28471948 DOI: 10.1097/qad.0000000000001401] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
: It is estimated that by 2030 nearly three-quarters of persons living with HIV will be 50 years and older. The aging HIV population presents a new clinical concern for HIV providers: adverse effects from polypharmacy. An aging population means more comorbidities and potentially more drug-drug interactions for providers to manage. This review discusses major comorbidities including cardiovascular disease, anticoagulation, hypertension, diabetes mellitus and malignancy and considerations for drug-interactions with antiretrovirals.
Collapse
|
21
|
Kalgutkar AS. Liabilities Associated with the Formation of “Hard” Electrophiles in Reactive Metabolite Trapping Screens. Chem Res Toxicol 2016; 30:220-238. [DOI: 10.1021/acs.chemrestox.6b00332] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Amit S. Kalgutkar
- Pharmacokinetics, Dynamics, and Metabolism − New Chemical
Entities, Pfizer Worldwide Research and Development, 610 Main
Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
22
|
Cho T, Uetrecht J. How Reactive Metabolites Induce an Immune Response That Sometimes Leads to an Idiosyncratic Drug Reaction. Chem Res Toxicol 2016; 30:295-314. [DOI: 10.1021/acs.chemrestox.6b00357] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tiffany Cho
- Faculty
of Pharmacy, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | - Jack Uetrecht
- Faculty
of Pharmacy, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| |
Collapse
|
23
|
Tailor A, Waddington JC, Meng X, Park BK. Mass Spectrometric and Functional Aspects of Drug–Protein Conjugation. Chem Res Toxicol 2016; 29:1912-1935. [DOI: 10.1021/acs.chemrestox.6b00147] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Arun Tailor
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - James C. Waddington
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Xiaoli Meng
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - B. Kevin Park
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| |
Collapse
|
24
|
Gan J, Ma S, Zhang D. Non-cytochrome P450-mediated bioactivation and its toxicological relevance. Drug Metab Rev 2016; 48:473-501. [DOI: 10.1080/03602532.2016.1225756] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
25
|
Shi J, Xie C, Liu H, Krausz KW, Bewley CA, Zhang S, Tang L, Zhou Z, Gonzalez FJ. Metabolism and Bioactivation of Fluorochloridone, a Novel Selective Herbicide, in Vivo and in Vitro. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:9652-60. [PMID: 27443216 PMCID: PMC6169518 DOI: 10.1021/acs.est.6b02113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Fluorochloridone (FLC) is a herbicide used worldwide that is thought to be safe. However, due to its potential genotoxicity, cytotoxicity, and even systematic toxicity, there are increasing concerns about human exposure to this compound. Thus, the metabolism and bioactivation of FLC was investigated. After oral administration to mice, 27 metabolites were identified by ultrahigh performance liquid chromatography-electrospray ionization-quadrupole time-of-flight-mass spectrometry and with further structural identification by nuclear magnetic resonance spectroscopy. Hydroxylation and oxidative dechlorination were the major phase I pathways, while glutathione (GSH) and N-acetylcysteine conjugations were two major phase II pathways, indicating the formation of a reactive intermediate. In vitro microsomal and cytosolic studies revealed that a GSH conjugate (M13) was the predominant metabolite of FLC formed through a nucleophilic SN2 substitution of 3-Cl by GSH; this pathway is NADPH independent and accelerated by glutathione S-transferase (GST). Further, a kinetic study showed that M13 formation in both human liver microsomes and cytosols obeyed typical Michaelis-Menten kinetics. The maximum clearance (Vmax/Km) of GSH conjugation in human liver microsomes was approximately 5.5-fold higher than human liver cytosol, thus implying that microsomal GST was mainly responsible for M13 formation. These findings are important for understanding the potential hazard of human exposure to FLC.
Collapse
Affiliation(s)
- Jingmin Shi
- Pharmacology and Toxicology, Department/Center for Drug Safety Evaluation, Shanghai Institute for Food and Drug Control, Shanghai 201203, PR China
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Hongbing Liu
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kristopher W. Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Carole A. Bewley
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Suhui Zhang
- Pharmacology and Toxicology, Department/Center for Drug Safety Evaluation, Shanghai Institute for Food and Drug Control, Shanghai 201203, PR China
| | - Liming Tang
- Pharmacology and Toxicology, Department/Center for Drug Safety Evaluation, Shanghai Institute for Food and Drug Control, Shanghai 201203, PR China
- Corresponding Authors: (F.J.G.)., (L.T.)
| | - Zhijun Zhou
- School of Public Health, Fudan University, Shanghai 200032, PR China
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
- Corresponding Authors: (F.J.G.)., (L.T.)
| |
Collapse
|
26
|
Foti RS, Dalvie DK. Cytochrome P450 and Non-Cytochrome P450 Oxidative Metabolism: Contributions to the Pharmacokinetics, Safety, and Efficacy of Xenobiotics. ACTA ACUST UNITED AC 2016; 44:1229-45. [PMID: 27298339 DOI: 10.1124/dmd.116.071753] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 12/16/2022]
Abstract
The drug-metabolizing enzymes that contribute to the metabolism or bioactivation of a drug play a crucial role in defining the absorption, distribution, metabolism, and excretion properties of that drug. Although the overall effect of the cytochrome P450 (P450) family of drug-metabolizing enzymes in this capacity cannot be understated, advancements in the field of non-P450-mediated metabolism have garnered increasing attention in recent years. This is perhaps a direct result of our ability to systematically avoid P450 liabilities by introducing chemical moieties that are not susceptible to P450 metabolism but, as a result, may introduce key pharmacophores for other drug-metabolizing enzymes. Furthermore, the effects of both P450 and non-P450 metabolism at a drug's site of therapeutic action have also been subject to increased scrutiny. To this end, this Special Section on Emerging Novel Enzyme Pathways in Drug Metabolism will highlight a number of advancements that have recently been reported. The included articles support the important role of non-P450 enzymes in the clearance pathways of U.S. Food and Drug Administration-approved drugs over the past 10 years. Specific examples will detail recent reports of aldehyde oxidase, flavin-containing monooxygenase, and other non-P450 pathways that contribute to the metabolic, pharmacokinetic, or pharmacodynamic properties of xenobiotic compounds. Collectively, this series of articles provides additional support for the role of non-P450-mediated metabolic pathways that contribute to the absorption, distribution, metabolism, and excretion properties of current xenobiotics.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| | - Deepak K Dalvie
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| |
Collapse
|
27
|
Nishiya Y, Nakai D, Urasaki Y, Takakusa H, Ohsuki S, Iwano Y, Yasukochi T, Takayama T, Bazyo S, Oza C, Kurihara A, Savage RE, Izumi T. Stereoselective hydroxylation by CYP2C19 and oxidation by ADH4 in the in vitro metabolism of tivantinib. Xenobiotica 2016; 46:967-76. [PMID: 26899628 DOI: 10.3109/00498254.2016.1144896] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
1. In prior studies, it has been shown that tivantinib is extensively metabolized in humans to many oxidative metabolites and glucuronides. In order to identify the responsible enzymes, we investigated the in vitro metabolism of tivantinib and its four major circulating metabolites. 2. The primary isoforms involved in the elimination of tivantinib were CYP2C19 and CYP3A4/5. CYP2C19 showed catalytic activity for the formation of M5 (hydroxylated metabolite), but not for M4 (a stereoisomer of M5), whereas CYP3A4/5 catalyzed the formation of both metabolites. For the elimination of M4, M5 and M8 (keto-metabolite), CYP3A4/5 was the major cytochrome P450 isoform and UGT1A9 was mainly involved in the glucuronidation of M4 and M5. 3. ADH4 was identified as one of the major alcohol dehydrogenase isoforms contributing to the formation of M6 (sequential keto-metabolite of M4 and M5) and M8. The substrate preference of ADH for M4, and not M5, was observed in the formation of M6. 4. In conclusion, CYP2C19, CYP3A4/5, UGT1A9 and ADH4 were the primary drug metabolizing enzymes involved in the in vitro metabolism of tivantinib and its metabolites. The stereoselective hydroxylation by CYP2C19 and substrate stereoselectivity of ADH4-catalyzed oxidation in the in vitro metabolism of tivantinib was discovered.
Collapse
Affiliation(s)
- Yumi Nishiya
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Daisuke Nakai
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Yoko Urasaki
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Hideo Takakusa
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Satoru Ohsuki
- b Organic Synthesis Department, Daiichi Sankyo RD Novare Co., Ltd. , Tokyo , Japan
| | - Yuji Iwano
- b Organic Synthesis Department, Daiichi Sankyo RD Novare Co., Ltd. , Tokyo , Japan
| | - Takanori Yasukochi
- b Organic Synthesis Department, Daiichi Sankyo RD Novare Co., Ltd. , Tokyo , Japan
| | - Tomoko Takayama
- c Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. , Tokyo , Japan
| | - Shohei Bazyo
- c Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. , Tokyo , Japan
| | - Chikahiro Oza
- c Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. , Tokyo , Japan
| | - Atsushi Kurihara
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | | | - Takashi Izumi
- e Research Function, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| |
Collapse
|
28
|
Inoue K, Fukuda K, Yoshimura T, Kusano K. Comparison of the Reactivity of Trapping Reagents toward Electrophiles: Cysteine Derivatives Can Be Bifunctional Trapping Reagents. Chem Res Toxicol 2015; 28:1546-55. [PMID: 26172216 DOI: 10.1021/acs.chemrestox.5b00129] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Trapping reagents are powerful tools to detect unstable reactive metabolites. There are a variety of trapping reagents based on chemical reactivity to electrophiles, and we investigated the reactivity of thiol and amine trapping reagents to metabolically generated electrophiles and commercially available electrophilic compounds. Glutathione (GSH) and N-acetylcysteine (Nac) trapped soft electrophiles, and amine derivatives such as semicarbazide (SC) and methoxyamine (MeA) reacted as hard nucleophiles to trap aldehydes as imine derivatives. Cysteine (Cys) and homocysteine (HCys) captured both soft electrophiles and hard electrophilic aldehydes. There were no qualitative differences in trapping soft electrophiles among Cys, HCys, GSH, and Nac, although quantitative reactivity to trap soft electrophiles varied likely depending on the pKa values of their thiol group. In the reactivity with aldehydes, Cys and HCys showed relatively lower reactivity as compared with SC and MeA. Nonetheless, they can trap aldehydes, and the resulting conjugates were stable and detected easily because their amino group formed imines after reaction with aldehydes, which are successively attacked by the intramolecular thiol group to form stable ring structures. This report demonstrated that Cys and HCys are advantageous to evaluate the formations of both soft electrophiles and aldehyde-type derivatives from a lot of drug candidates at early drug discovery by their unique structural characteristics.
Collapse
Affiliation(s)
- Kazuko Inoue
- Drug Metabolism and Pharmacokinetics Japan, Eisai Product Creation Systems, Eisai Co., Ltd., Tsukuba, Japan
| | - Katsuyuki Fukuda
- Drug Metabolism and Pharmacokinetics Japan, Eisai Product Creation Systems, Eisai Co., Ltd., Tsukuba, Japan
| | - Tsutomu Yoshimura
- Drug Metabolism and Pharmacokinetics Japan, Eisai Product Creation Systems, Eisai Co., Ltd., Tsukuba, Japan
| | - Kazutomi Kusano
- Drug Metabolism and Pharmacokinetics Japan, Eisai Product Creation Systems, Eisai Co., Ltd., Tsukuba, Japan
| |
Collapse
|
29
|
Grillo MP. Detecting reactive drug metabolites for reducing the potential for drug toxicity. Expert Opin Drug Metab Toxicol 2015; 11:1281-302. [PMID: 26005795 DOI: 10.1517/17425255.2015.1048222] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION A number of withdrawn drugs are known to undergo bioactivation by a range of drug metabolizing enzymes to chemically reactive metabolites that bind covalently to protein and DNA resulting in organ toxicity and carcinogenesis, respectively. An important goal in drug discovery is to identify structural sites of bioactivation within discovery molecules for providing strategic modifications that eliminate or minimize reactive metabolite formation, while maintaining target potency, selectivity and desired pharmacokinetic properties leading to the development of efficacious and nontoxic drugs. AREAS COVERED This review covers experimental techniques currently used to detect reactive drug metabolites and provides recent examples where information from mechanistic in vitro studies was successfully used to redesign candidate drugs leading to blocked or minimized bioactivation. Reviewed techniques include in vitro radiolabeled drug covalent binding to protein and reactive metabolite trapping with reagents such as glutathione, cyanide, semicarbazide and DNA bases. Case studies regarding reactive metabolite detection using a combination of varied techniques, including liquid chromatography-tandem mass spectrometry and NMR analyses and subsequent structural modification are discussed. EXPERT OPINION Information derived from state-of-art mechanistic drug metabolism studies can be used successfully to direct medicinal chemistry towards the synthesis of candidate drugs devoid of bioactivation liabilities, while maintaining desired pharmacology and pharmacokinetic properties.
Collapse
Affiliation(s)
- Mark P Grillo
- MyoKardia , 333 Allerton Ave, South San Francisco, CA 94080 , USA
| |
Collapse
|
30
|
Sullivan A, Gibson A, Park BK, Naisbitt DJ. Are drug metabolites able to cause T-cell-mediated hypersensitivity reactions? Expert Opin Drug Metab Toxicol 2014; 11:357-68. [DOI: 10.1517/17425255.2015.992780] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Peng YS, Liu B, Wang RF, Zhao QT, Xu W, Yang XW. Hepatic metabolism: a key component of herbal drugs research. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2014; 17:89-106. [PMID: 25296190 DOI: 10.1080/10286020.2014.960856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Liver is the largest metabolic organ for a wide range of endogenous and exogenous compounds and plays a crucial part in the pharmacokinetics and pharmacodynamics through various metabolic reactions. This review provides a progressive description of hepatic metabolism of herbal drugs with respect to metabolic types and investigational methods. In addition, the problems encountered during the research process are discussed.
Collapse
Affiliation(s)
- Yu-Shuai Peng
- a School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing 100102 , China
| | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Meng X, Lawrenson AS, Berry NG, Maggs JL, French NS, Back DJ, Khoo SH, Naisbitt DJ, Park BK. Abacavir Forms Novel Cross-Linking Abacavir Protein Adducts in Patients. Chem Res Toxicol 2014; 27:524-35. [DOI: 10.1021/tx400406p] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xiaoli Meng
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Alexandre S. Lawrenson
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United Kingdom
| | - Neil G. Berry
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United Kingdom
| | - James L. Maggs
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Neil S. French
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - David J. Back
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Saye H. Khoo
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Dean J. Naisbitt
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - B. Kevin Park
- MRC Centre
for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| |
Collapse
|
34
|
Pirmohamed M, Drummond NS, Naisbitt DJ, Park BK. Drug hypersensitivity reactions in patients with HIV disease. Expert Rev Clin Immunol 2014; 3:395-410. [DOI: 10.1586/1744666x.3.3.395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
35
|
Bioactivation to an aldehyde metabolite—Possible role in the onset of toxicity induced by the anti-HIV drug abacavir. Toxicol Lett 2014; 224:416-23. [DOI: 10.1016/j.toxlet.2013.10.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/25/2013] [Accepted: 10/28/2013] [Indexed: 12/20/2022]
|
36
|
Earnshaw CJ, Pecaric-Petkovic T, Park BK, Naisbitt DJ. T cell responses to drugs and drug metabolites. EXPERIENTIA SUPPLEMENTUM (2012) 2014; 104:137-63. [PMID: 24214623 DOI: 10.1007/978-3-0348-0726-5_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Understanding the chemical mechanisms by which drugs and drug metabolites interact with cells of the immune system is pivotal to our knowledge of drug hypersensitivity as a whole.In this chapter, we will discuss the currently accepted mechanisms where there is scientific and clinical evidence to support the ways in which drugs and their metabolites interact with T cells. We will also discuss bioanalytical platforms, such as mass spectrometry, and in vitro test assays such as the lymphocyte transformation test that can be used to study drug hypersensitivity; the combination of such techniques can be used to relate the chemistry of drug antigen formation to immune function. Ab initio T cell priming assays are also discussed with respect to predicting the potential of a drug to cause hypersensitivity reactions in humans in relation to the chemistry of the drug and its ability to form haptens, antigens and immunogens in patients.
Collapse
Affiliation(s)
- C J Earnshaw
- Department of Molecular and Clinical Pharmacology, Medical Research Council Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ahston Street, Liverpool, L69 3GE, UK,
| | | | | | | |
Collapse
|
37
|
Bell CC, Santoyo Castelazo A, Yang EL, Maggs JL, Jenkins RE, Tugwood J, O’Neill PM, Naisbitt DJ, Park BK. Oxidative Bioactivation of Abacavir in Subcellular Fractions of Human Antigen Presenting Cells. Chem Res Toxicol 2013; 26:1064-72. [DOI: 10.1021/tx400041v] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Catherine C. Bell
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Anahi Santoyo Castelazo
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Emma L. Yang
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, United
Kingdom
| | - James L. Maggs
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Rosalind E. Jenkins
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Jonathan Tugwood
- Paterson Institute
for Cancer
Research, The University of Manchester,
Manchester M20 4BX, United Kingdom
| | - Paul M. O’Neill
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, United
Kingdom
| | - Dean J. Naisbitt
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - B. Kevin Park
- MRC Centre
for Drug Safety Science,
Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| |
Collapse
|
38
|
Monitoring abacavir bioactivation in humans: Screening for an aldehyde metabolite. Toxicol Lett 2013; 219:59-64. [DOI: 10.1016/j.toxlet.2013.02.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 02/21/2013] [Accepted: 02/23/2013] [Indexed: 11/19/2022]
|
39
|
Bell CC, Faulkner L, Martinsson K, Farrell J, Alfirevic A, Tugwood J, Pirmohamed M, Naisbitt DJ, Park BK. T-Cells from HLA-B*57:01+ Human Subjects Are Activated with Abacavir through Two Independent Pathways and Induce Cell Death by Multiple Mechanisms. Chem Res Toxicol 2013; 26:759-66. [DOI: 10.1021/tx400060p] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Catherine C. Bell
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - Lee Faulkner
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - Klara Martinsson
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - John Farrell
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - Ana Alfirevic
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - Jonathan Tugwood
- Paterson Institute for Cancer
Research, The University of Manchester,
Wilmslow Road, Manchester M20 4BX, England
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - Dean J. Naisbitt
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - B. Kevin Park
- MRC Centre for Drug Safety Science,
Department of Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| |
Collapse
|
40
|
Charneira C, Grilo NM, Pereira SA, Godinho ALA, Monteiro EC, Marques MM, Antunes AMM. N-terminal valine adduct from the anti-HIV drug abacavir in rat haemoglobin as evidence for abacavir metabolism to a reactive aldehyde in vivo. Br J Pharmacol 2013; 167:1353-61. [PMID: 22725138 DOI: 10.1111/j.1476-5381.2012.02079.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study was to obtain evidence for the activation of the nucleoside reverse transcriptase inhibitor abacavir to reactive aldehyde metabolites in vivo. Protein haptenation by these reactive metabolites may be a factor in abacavir-induced toxic events. EXPERIMENTAL APPROACH The formation of N-terminal valine adducts from the abacavir-derived aldehydes was investigated in the haemoglobin of Wistar rats treated with eight daily doses (120 mg·kg(-1)) of abacavir. The analyses were conducted by high-performance liquid chromatography-electrospray ionization-tandem mass spectrometry upon comparison with synthetic standards. KEY RESULTS An N-terminal valine haemoglobin adduct derived from an α,β-unsaturated aldehyde metabolite of abacavir was identified in vivo for the first time. CONCLUSIONS AND IMPLICATIONS This preliminary work on abacavir metabolism provides the first unequivocal evidence for the formation of an α,β-unsaturated aldehyde metabolite in vivo and of its ability to form haptens with proteins. The methodology described herein can be used to assess the formation of this metabolite in human samples and has the potential to become a valuable pharmacological tool for mechanistic studies of abacavir toxicity. In fact, the simplicity of the method suggests that the abacavir adduct with the N-terminal valine of haemoglobin could be used to investigate abacavir-induced toxicity for accurate risk/benefit estimations.
Collapse
Affiliation(s)
- C Charneira
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade Técnica de Lisboa, Lisboa, Portugal
| | | | | | | | | | | | | |
Collapse
|
41
|
Diao X, Deng P, Xie C, Li X, Zhong D, Zhang Y, Chen X. Metabolism and Pharmacokinetics of 3-n-Butylphthalide (NBP) in Humans: The Role of Cytochrome P450s and Alcohol Dehydrogenase in Biotransformation. Drug Metab Dispos 2012; 41:430-44. [DOI: 10.1124/dmd.112.049684] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
42
|
Stachulski AV, Baillie TA, Kevin Park B, Scott Obach R, Dalvie DK, Williams DP, Srivastava A, Regan SL, Antoine DJ, Goldring CEP, Chia AJL, Kitteringham NR, Randle LE, Callan H, Castrejon JL, Farrell J, Naisbitt DJ, Lennard MS. The Generation, Detection, and Effects of Reactive Drug Metabolites. Med Res Rev 2012; 33:985-1080. [DOI: 10.1002/med.21273] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories; University of Liverpool; Liverpool; L69 7ZD; UK
| | - Thomas A. Baillie
- School of Pharmacy; University of Washington; Box 357631; Seattle; Washington; 98195-7631
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; Groton; Connecticut 06340
| | - Deepak K. Dalvie
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; La Jolla; California 94121
| | - Dominic P. Williams
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Abhishek Srivastava
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Sophie L. Regan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Daniel J. Antoine
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Christopher E. P. Goldring
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Alvin J. L. Chia
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Neil R. Kitteringham
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Laura E. Randle
- School of Pharmacy and Biomolecular Sciences, Faculty of Science; Liverpool John Moores University; James Parsons Building, Byrom Street; Liverpool L3 3AF; UK
| | - Hayley Callan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - J. Luis Castrejon
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - John Farrell
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Martin S. Lennard
- Academic Unit of Medical Education; University of Sheffield; 85 Wilkinson Street; Sheffield S10 2GJ; UK
| |
Collapse
|
43
|
Abacavir induces loading of novel self-peptides into HLA-B*57: 01: an autoimmune model for HLA-associated drug hypersensitivity. AIDS 2012; 26:F21-9. [PMID: 22617051 DOI: 10.1097/qad.0b013e328355fe8f] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Abacavir drug hypersensitivity in HIV-treated patients is associated with HLA-B57:01 expression. To understand the immunochemistry of abacavir drug reactions, we investigated the effects of abacavir on HLA-B57:01 epitope-binding in vitro and the quality and quantity of self-peptides presented by HLA-B57:01 from abacavir-treated cells. DESIGN AND METHODS An HLA-B57:01-specific epitope-binding assay was developed to test for effects of abacavir, didanosine or flucloxacillin on self-peptide binding. To examine whether abacavir alters the peptide repertoire in HLA-B57:01, a B-cell line secreting soluble human leucocyte antigen (sHLA) was cultured in the presence or absence of abacavir, peptides were eluted from purified human leucocyte antigen (HLA), and the peptide epitopes comparatively mapped by mass spectroscopy to identify drug-unique peptides. RESULTS Abacavir, but not didansosine or flucloxacillin, enhanced binding of the FITC-labeled self-peptide LF9 to HLA-B57:01 in a dose-dependent manner. Endogenous peptides isolated from abacavir-treated HLA-B57:01 B cells showed amino acid sequence differences compared with peptides from untreated cells. Novel drug-induced peptides lacked typical carboxyl (C) terminal amino acids characteristic of the HLA-B57:01 peptide motif and instead contained predominantly isoleucine or leucine residues. Drug-induced peptides bind to soluble HLA-B57:01 with high affinity that was not altered by abacavir addition. CONCLUSION Our results support a model of drug-induced autoimmunity in which abacavir alters the quantity and quality of self-peptide loading into HLA-B57:01. Drug-induced loading of novel self-peptides into HLA, possibly by abacavir either altering the binding cleft or modifying the peptide-loading complex, generates an array of neo-antigen peptides that drive polyclonal T-cell autoimmune responses and multiorgan systemic toxicity.
Collapse
|
44
|
Adam J, Eriksson KK, Schnyder B, Fontana S, Pichler WJ, Yerly D. Avidity determines T-cell reactivity in abacavir hypersensitivity. Eur J Immunol 2012; 42:1706-16. [DOI: 10.1002/eji.201142159] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 03/09/2012] [Accepted: 03/30/2012] [Indexed: 12/12/2022]
Affiliation(s)
- Jacqueline Adam
- Clinic for Rheumatology and Clinical Immunology/Allergology; University Hospital of Bern; Switzerland
| | - Klara K. Eriksson
- Clinic for Rheumatology and Clinical Immunology/Allergology; University Hospital of Bern; Switzerland
| | - Benno Schnyder
- Clinic for Rheumatology and Clinical Immunology/Allergology; University Hospital of Bern; Switzerland
| | - Stefano Fontana
- Regional Blood Transfusion Service of the Swiss Red Cross; Bern; Switzerland
| | - Werner J. Pichler
- Clinic for Rheumatology and Clinical Immunology/Allergology; University Hospital of Bern; Switzerland
| | - Daniel Yerly
- Clinic for Rheumatology and Clinical Immunology/Allergology; University Hospital of Bern; Switzerland
| |
Collapse
|
45
|
Hvenegaard MG, Bang-Andersen B, Pedersen H, Jørgensen M, Püschl A, Dalgaard L. Identification of the Cytochrome P450 and Other Enzymes Involved in the In Vitro Oxidative Metabolism of a Novel Antidepressant, Lu AA21004. Drug Metab Dispos 2012; 40:1357-65. [DOI: 10.1124/dmd.112.044610] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
46
|
Abstract
Bupropion is metabolized extensively in humans by oxidative and reductive processes. CYP2B6 mediates oxidation of bupropion to hydroxybupropion, but the enzyme(s) catalyzing carbonyl reduction of bupropion to erythro- and threohydrobupropion in human liver is unknown. The objective of this study was to examine the enzyme kinetics of bupropion reduction in human liver. In human liver cytosol, the reduction of bupropion to erythro-and threohydrobupropion was NADPH dependent with Cl(int) values of 0.08 and 0.60 µL·min(-1)mg(-1) protein, respectively. Bupropion reduction in liver microsomes was also NADPH dependent with Cl(int) values of 10.4 and 280 µL·min(-1)mg(-1) protein, respectively. Formation of erythro-and threohydrobupropion in microsomes exceeded that in cytosol by 70 and 170 fold, respectively. Menadione, an inhibitor of cytosolic carbonyl reducing enzymes (e.g. CBRs), inhibited erythro-and threohydrobupropion formation in cytosol with IC(50) of 30 and 54 µM, respectively. In microsomes 18β-glycyrrhetinic acid, an inhibitor of microsomal carbonyl reductases (e.g. 11β-HSDs), inhibited their formation with IC(50) of 25 and 26 nM, respectively. Our findings, in agreement with recent human placental studies, show that carbonyl reducing enzymes in hepatic microsomes are significant players in bupropion reduction. Contrary to past studies, we found that threohydrobupropion (not hydroxybupropion) is the major microsomal generated hepatic metabolite of bupropion.
Collapse
Affiliation(s)
- Jillissa C Molnari
- Department of Pharmaceutical, Biomedical and Administrative Sciences, College of Pharmacy and Health Sciences, Drake University, Des Moines, IA, USA
| | | |
Collapse
|
47
|
HIV-Antiretroviral Therapy Induced Liver, Gastrointestinal, and Pancreatic Injury. Int J Hepatol 2012; 2012:760706. [PMID: 22506127 PMCID: PMC3312274 DOI: 10.1155/2012/760706] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 12/30/2011] [Accepted: 01/01/2012] [Indexed: 01/11/2023] Open
Abstract
The present paper describes possible connections between antiretroviral therapies (ARTs) used to treat human immunodeficiency virus (HIV) infection and adverse drug reactions (ADRs) encountered predominantly in the liver, including hypersensitivity syndrome reactions, as well as throughout the gastrointestinal system, including the pancreas. Highly active antiretroviral therapy (HAART) has a positive influence on the quality of life and longevity in HIV patients, substantially reducing morbidity and mortality in this population. However, HAART produces a spectrum of ADRs. Alcohol consumption can interact with HAART as well as other pharmaceutical agents used for the prevention of opportunistic infections such as pneumonia and tuberculosis. Other coinfections that occur in HIV, such as hepatitis viruses B or C, cytomegalovirus, or herpes simplex virus, further complicate the etiology of HAART-induced ADRs. The aspect of liver pathology including liver structure and function has received little attention and deserves further evaluation. The materials used provide a data-supported approach. They are based on systematic review and analysis of recently published world literature (MedLine search) and the experience of the authors in the specified topic. We conclude that therapeutic and drug monitoring of ART, using laboratory identification of phenotypic susceptibilities, drug interactions with other medications, drug interactions with herbal medicines, and alcohol intake might enable a safer use of this medication.
Collapse
|
48
|
Charneira C, Godinho ALA, Oliveira MC, Pereira SA, Monteiro EC, Marques MM, Antunes AMM. Reactive Aldehyde Metabolites from the Anti-HIV Drug Abacavir: Amino Acid Adducts as Possible Factors in Abacavir Toxicity. Chem Res Toxicol 2011; 24:2129-41. [DOI: 10.1021/tx200337b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Catarina Charneira
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| | - Ana L. A. Godinho
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| | - M. Conceição Oliveira
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| | - Sofia A. Pereira
- Centro de Estudos de Doenças Crónicas (CEDOC), Departamento de Farmacologia, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Emília C. Monteiro
- Centro de Estudos de Doenças Crónicas (CEDOC), Departamento de Farmacologia, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - M. Matilde Marques
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| | - Alexandra M. M. Antunes
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade Técnica de Lisboa, 1049-001 Lisboa, Portugal
| |
Collapse
|
49
|
Bharadwaj M, Illing P, Theodossis A, Purcell AW, Rossjohn J, McCluskey J. Drug hypersensitivity and human leukocyte antigens of the major histocompatibility complex. Annu Rev Pharmacol Toxicol 2011; 52:401-31. [PMID: 22017685 DOI: 10.1146/annurev-pharmtox-010611-134701] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The human leukocyte antigen (HLA) genes are the most polymorphic in the human genome and are critical in regulating specific immunity, hence their historical discovery as "immune response" genes. HLA allotypes are also implicated in unwanted immune reactions, including drug hypersensitivity syndrome, in which small therapeutic drugs interact with antigenic peptides to drive T cell responses restricted by host HLA. Abacavir, allo-purinol, and carbamazepine are three commonly used drugs that cause a T cell-mediated hypersensitivity that is HLA linked, with each drug exhibiting striking specificity for presentation by defined HLA allotypes. Recent findings have begun to unearth the mechanistic basis for these HLA associations, and here we review recent advances in the field of HLA-associated drug hypersensitivities.
Collapse
Affiliation(s)
- Mandvi Bharadwaj
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | | | | | |
Collapse
|
50
|
Sanaei-Zadeh H, Zamani N, Shahmohammadi F. Can fomepizole be substituted by abacavir in the treatment of methanol poisoning? J Med Toxicol 2011; 7:179-80. [PMID: 21484534 DOI: 10.1007/s13181-011-0154-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Hossein Sanaei-Zadeh
- Department of Forensic Medicine and Toxicology, School of Medicine (Pardis Hemmat), Tehran University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|