1
|
Singh H, Kunkle BF, Troia AR, Suvarnakar AM, Waterman AC, Khin Y, Korkmaz SY, O'Connor CE, Lewis JH. Drug Induced Liver Injury: Highlights and Controversies in the 2023 Literature. Drug Saf 2025; 48:455-488. [PMID: 39921708 DOI: 10.1007/s40264-025-01514-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/10/2025]
Abstract
Drug-induced liver injury (DILI) remains an active field of clinical research and investigation with more than 4700 publications appearing in 2023 relating to hepatotoxicity of all causes and injury patterns. As in years past, we have attempted to identify and summarize highlights and controversies from the past year's literature. Several new and novel therapeutic agents were approved by the US Food and Drug Administration (FDA) in 2023, a number of which were associated with significant hepatotoxicity. Updates in the diagnosis and management of DILI using causality scores as well as newer artificial intelligence-based methods were published. Details of newly established hepatotoxins as well as updated information on previously documented hepatotoxic drugs is presented. Significant updates in treatment of DILI were also included as well as reports related to global DILI registries.
Collapse
Affiliation(s)
- Harjit Singh
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA.
| | - Bryce F Kunkle
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Angela R Troia
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | | | - Ade C Waterman
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Yadana Khin
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Serena Y Korkmaz
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Corinne E O'Connor
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - James H Lewis
- Division of Gastroenterology and Hepatology, Medstar Georgetown University Hospital, Washington, DC, USA
| |
Collapse
|
2
|
Pound P, Ram R, Archibald K. The UK's Early Access to Medicines Scheme 10 years on: an evaluation using publicly available data. JRSM Open 2025; 16:20542704251317916. [PMID: 40007990 PMCID: PMC11848878 DOI: 10.1177/20542704251317916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Objectives To investigate the drugs and indications that have passed through the UK's Early Access to Medicines Scheme (EAMS) to date, the type of evidence the regulator considers when accepting a drug into the EAMS, and potential risks to patients. Design Analysis of publicly available data: MHRA Public Assessment Reports; Electronic Medicines Compendium database; interactive Drug Analysis Profiles database; Eudravigilance database. Setting United Kingdom. Participants The 51 'scientific opinions' available on the MHRA website in June 2024. Main Outcome Measures Public Assessment Reports, pharmacovigilance data. Results After exclusions, there were 48 EAMS submissions, consisting of 48 indications and 32 drugs. 60% of indications were for cancer. Only 7% of EAMS submissions were based on double-blind, placebo-controlled randomised trials. The average sample size of studies conducted for the EAMS was 654. Most studies used surrogate (76%) and/or survival (57%) outcomes. Only 17% used subjective outcomes. For 17% of the indications, no ongoing studies were being conducted. Animal studies were conducted preclinically for all drugs and 35% also conducted in vitro studies. 47% of the drugs had elevated rates of suspected adverse reaction reports according to pharmacovigilance data. Conclusions We recommend that the EAMS drugs with elevated reporting rates are reviewed, that future studies of EAMS drugs use patient-centred outcomes, that preclinical studies make greater use of human biology-based approaches, that post-approval trials are conducted, and that future reviews of the EAMS centre the experience of patients.
Collapse
Affiliation(s)
| | - Rebecca Ram
- Safer Medicines Trust, Kingsbridge, TQ7 9AX, UK
| | | |
Collapse
|
3
|
He Q, Li M, Ji P, Zheng A, Yao L, Zhu X, Shin JG, Lauschke VM, Han B, Xiang X. Comparison of drug-induced liver injury risk between propylthiouracil and methimazole: A quantitative systems toxicology approach. Toxicol Appl Pharmacol 2024; 491:117064. [PMID: 39122118 DOI: 10.1016/j.taap.2024.117064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Propylthiouracil (PTU) and methimazole (MMI), two classical antithyroid agents possess risk of drug-induced liver injury (DILI) with unknown mechanism of action. This study aimed to examine and compare their hepatic toxicity using a quantitative system toxicology approach. The impact of PTU and MMI on hepatocyte survival, oxidative stress, mitochondrial function and bile acid transporters were assessed in vitro. The physiologically based pharmacokinetic (PBPK) models of PTU and MMI were constructed while their risk of DILI was calculated by DILIsym, a quantitative systems toxicology (QST) model by integrating the results from in vitro toxicological studies and PBPK models. The simulated DILI (ALT >2 × ULN) incidence for PTU (300 mg/d) was 21.2%, which was within the range observed in clinical practice. Moreover, a threshold dose of 200 mg/d was predicted with oxidative stress proposed as an important toxic mechanism. However, DILIsym predicted a 0% incidence of hepatoxicity caused by MMI (30 mg/d), suggesting that the toxicity of MMI was not mediated through mechanism incorporated into DILIsym. In conclusion, DILIsym appears to be a practical tool to unveil hepatoxicity mechanism and predict clinical risk of DILI.
Collapse
Affiliation(s)
- Qingfeng He
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Min Li
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peiying Ji
- Department of Pharmacy, Kong Jiang Hospital of Yangpu District, Shanghai 200093, China
| | - Aole Zheng
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Li Yao
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai 201100, China.
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
4
|
Pound P, Ritskes-Hoitinga M. Will Labour help us transition away from pre-clinical animal research? BMJ 2024; 386:q1600. [PMID: 39029938 DOI: 10.1136/bmj.q1600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Affiliation(s)
| | - Merel Ritskes-Hoitinga
- Institute for Risk Assessment Sciences, Veterinary Medicine, Utrecht University, Netherlands
| |
Collapse
|
5
|
Woodhead JL, Gebremichael Y, Macwan J, Qureshi IA, Bertz R, Wirtz V, Howell BA. Prediction of the liver safety profile of a first-in-class myeloperoxidase inhibitor using quantitative systems toxicology modeling. Xenobiotica 2024; 54:401-410. [PMID: 38874513 DOI: 10.1080/00498254.2024.2361027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
The novel myeloperoxidase inhibitor verdiperstat was developed as a treatment for neuroinflammatory and neurodegenerative diseases. During development, a computational prediction of verdiperstat liver safety was performed using DILIsym v8A, a quantitative systems toxicology (QST) model of liver safety.A physiologically-based pharmacokinetic (PBPK) model of verdiperstat was constructed in GastroPlus 9.8, and outputs for liver and plasma time courses of verdiperstat were input into DILIsym. In vitro experiments measured the likelihood that verdiperstat would inhibit mitochondrial function, inhibit bile acid transporters, and generate reactive oxygen species (ROS); these results were used as inputs into DILIsym, with two alternate sets of parameters used in order to fully explore the sensitivity of model predictions. Verdiperstat dosing protocols up to 600 mg BID were simulated for up to 48 weeks using a simulated population (SimPops) in DILIsym.Verdiperstat was predicted to be safe, with only very rare, mild liver enzyme increases as a potential possibility in highly sensitive individuals. Subsequent Phase 3 clinical trials found that ALT elevations in the verdiperstat treatment group were generally similar to those in the placebo group. This validates the DILIsym simulation results and demonstrates the power of QST modelling to predict the liver safety profile of novel therapeutics.
Collapse
|
6
|
Umemori Y, Handa K, Yoshimura S, Kageyama M, Iijima T. Development of a Novel In Silico Classification Model to Assess Reactive Metabolite Formation in the Cysteine Trapping Assay and Investigation of Important Substructures. Biomolecules 2024; 14:535. [PMID: 38785942 PMCID: PMC11117661 DOI: 10.3390/biom14050535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Predicting whether a compound can cause drug-induced liver injury (DILI) is difficult due to the complexity of drug mechanism. The cysteine trapping assay is a method for detecting reactive metabolites that bind to microsomes covalently. However, it is cumbersome to use 35S isotope-labeled cysteine for this assay. Therefore, we constructed an in silico classification model for predicting a positive/negative outcome in the cysteine trapping assay. We collected 475 compounds (436 in-house compounds and 39 publicly available drugs) based on experimental data performed in this study, and the composition of the results showed 248 positives and 227 negatives. Using a Message Passing Neural Network (MPNN) and Random Forest (RF) with extended connectivity fingerprint (ECFP) 4, we built machine learning models to predict the covalent binding risk of compounds. In the time-split dataset, AUC-ROC of MPNN and RF were 0.625 and 0.559 in the hold-out test, restrictively. This result suggests that the MPNN model has a higher predictivity than RF in the time-split dataset. Hence, we conclude that the in silico MPNN classification model for the cysteine trapping assay has a better predictive power. Furthermore, most of the substructures that contributed positively to the cysteine trapping assay were consistent with previous results.
Collapse
Affiliation(s)
| | - Koichi Handa
- DMPK Research Department, Teijin Institute for Bio-Medical Research, TEIJIN PHARMA LIMITED, 4-3-2 Asahigaoka, Hino-shi, Tokyo 191-8512, Japan; (Y.U.); (S.Y.); (M.K.); (T.I.)
| | | | | | | |
Collapse
|
7
|
Battista C, Shoda LKM, Watkins PB, Groettrup-Wolfers E, Rottmann A, Raschke M, Generaux GT. Quantitative Systems Toxicology Identifies Independent Mechanisms for Hepatotoxicity and Bilirubin Elevations Due to AKR1C3 Inhibitor BAY1128688. Clin Pharmacol Ther 2023; 114:1023-1032. [PMID: 37501650 DOI: 10.1002/cpt.3010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
BAY1128688 is a selective inhibitor of AKR1C3, investigated recently in a trial that was prematurely terminated due to drug-induced liver injury. These unexpected observations prompted use of the quantitative systems toxicology model, DILIsym, to determine possible mechanisms of hepatotoxicity. Using mechanistic in vitro toxicity data as well as clinical exposure data, DILIsym predicted the potential for BAY1128688 to cause liver toxicity (elevations in serum alanine aminotransferase (ALT)) and elevations in serum bilirubin. Initial simulations overpredicted hepatotoxicity and bilirubin elevations, so the BAY1128688 representation within DILIsym underwent optimization. The liver partition coefficient Kp was altered to align simulated bilirubin elevations with those observed clinically. Altering the mode of bile acid canalicular and basolateral efflux inhibition was necessary to accurately predict ALT elevations. Optimization results support that bilirubin elevations observed early during treatment are due to altered bilirubin metabolism and transporter inhibition, which is independent of liver injury. The modeling further supports that on-treatment ALT elevations result from inhibition of bile acid transporters, particularly the bile salt excretory pump, leading to accumulation of toxic bile acids. The predicted dose-dependent intrinsic hepatotoxicity may increase patient susceptibility to an adaptive immune response, accounting for ALT elevations observed after completion of treatment. These BAY1128688 simulations provide insight into the mechanisms behind hepatotoxicity and bilirubin elevations and may inform the potential risk posed by future compounds.
Collapse
Affiliation(s)
- Christina Battista
- DILIsym Services division, Simulations Plus, Inc., Durham, North Carolina, USA
| | - Lisl K M Shoda
- DILIsym Services division, Simulations Plus, Inc., Durham, North Carolina, USA
| | - Paul B Watkins
- Eshelman School of Pharmacy, Institute for Drug Safety Sciences, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Antje Rottmann
- Pharmaceuticals Division, Research & Early Development, Bayer AG, Berlin, Germany
| | - Marian Raschke
- Pharmaceuticals Division, Research & Early Development, Bayer AG, Berlin, Germany
| | | |
Collapse
|
8
|
Beaudoin JJ, Clemens L, Miedel MT, Gough A, Zaidi F, Ramamoorthy P, Wong KE, Sarangarajan R, Battista C, Shoda LKM, Siler SQ, Taylor DL, Howell BA, Vernetti LA, Yang K. The Combination of a Human Biomimetic Liver Microphysiology System with BIOLOGXsym, a Quantitative Systems Toxicology (QST) Modeling Platform for Macromolecules, Provides Mechanistic Understanding of Tocilizumab- and GGF2-Induced Liver Injury. Int J Mol Sci 2023; 24:9692. [PMID: 37298645 PMCID: PMC10253699 DOI: 10.3390/ijms24119692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Biologics address a range of unmet clinical needs, but the occurrence of biologics-induced liver injury remains a major challenge. Development of cimaglermin alfa (GGF2) was terminated due to transient elevations in serum aminotransferases and total bilirubin. Tocilizumab has been reported to induce transient aminotransferase elevations, requiring frequent monitoring. To evaluate the clinical risk of biologics-induced liver injury, a novel quantitative systems toxicology modeling platform, BIOLOGXsym™, representing relevant liver biochemistry and the mechanistic effects of biologics on liver pathophysiology, was developed in conjunction with clinically relevant data from a human biomimetic liver microphysiology system. Phenotypic and mechanistic toxicity data and metabolomics analysis from the Liver Acinus Microphysiology System showed that tocilizumab and GGF2 increased high mobility group box 1, indicating hepatic injury and stress. Tocilizumab exposure was associated with increased oxidative stress and extracellular/tissue remodeling, and GGF2 decreased bile acid secretion. BIOLOGXsym simulations, leveraging the in vivo exposure predicted by physiologically-based pharmacokinetic modeling and mechanistic toxicity data from the Liver Acinus Microphysiology System, reproduced the clinically observed liver signals of tocilizumab and GGF2, demonstrating that mechanistic toxicity data from microphysiology systems can be successfully integrated into a quantitative systems toxicology model to identify liabilities of biologics-induced liver injury and provide mechanistic insights into observed liver safety signals.
Collapse
Affiliation(s)
- James J. Beaudoin
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lara Clemens
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Mark T. Miedel
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Albert Gough
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Fatima Zaidi
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Kari E. Wong
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Christina Battista
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lisl K. M. Shoda
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Scott Q. Siler
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - D. Lansing Taylor
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Brett A. Howell
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lawrence A. Vernetti
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Kyunghee Yang
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| |
Collapse
|
9
|
Stern JI, Datta S, Chiang CC, Garza I, Vieira DL, Robertson CE. Narrative review of migraine management in patients with renal or hepatic disease. Headache 2023; 63:9-24. [PMID: 36709407 DOI: 10.1111/head.14437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 01/30/2023]
Abstract
OBJECTIVES/BACKGROUND Treatment of migraine in the setting of either renal or hepatic disease can be daunting for clinicians. Not only does the method of metabolism have to be considered, but also the method of elimination/excretion of the parent drug and any active or toxic metabolites. Furthermore, it is difficult to think about liver or kidney disease in isolation, as liver disease can sometimes contribute to impaired renal function and renal disease can sometimes impair hepatic metabolism, through the cytochrome P450 system. METHODS A detailed search for terms related to liver disease, renal disease, and migraine management was performed in PubMed, Ovid Medline, Embase, and the Cochrane Library.For each medication, product labels were retrieved and reviewed using the US FDA website, with additional review of IBM Micromedex, LiverTox, and the Renal Drug Handbook. RESULTS This manuscript provides an overview of migraine drug metabolism and how it can be affected by liver and renal impairment. It reviews the standard terminology recommended by the US Food and Drug Administration for the different stages of hepatic and renal failure. The available evidence regarding the use of abortive and preventative medicines in the setting of organ failure is discussed in detail, including more recent therapies such as lasmiditan, gepants, and calcitonin gene-related peptide antibodies. CONCLUSIONS For acute therapy, the use of NSAIDS should be limited, as these carry risk for both severe hepatic and renal disease. Triptans can be selectively used, often with dose guideline adjustments. Ubrogepant may be used in severe hepatic disease with dose adjustment and lasmiditan can be used in end stage renal disease. Though non-medicine strategies may be the most reasonable initial approach, many preventative medications can be used in the setting of hepatic and renal disease, often with dose adjustment. This review provides tables of guidelines, including reduced dosing recommendations, for the use of abortive and preventative migraine medications in hepatic and renal failure.
Collapse
Affiliation(s)
| | - Shae Datta
- Department of Neurology, NYU Langone Health, New York, New York, USA
| | | | - Ivan Garza
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Dorice L Vieira
- New York University Health Sciences Library, New York University Grossman School of Medicine, New York, New York, USA
| | | |
Collapse
|
10
|
Lin J, Li M, Mak W, Shi Y, Zhu X, Tang Z, He Q, Xiang X. Applications of In Silico Models to Predict Drug-Induced Liver Injury. TOXICS 2022; 10:788. [PMID: 36548621 PMCID: PMC9785299 DOI: 10.3390/toxics10120788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of the withdrawal of pre-marketed drugs, typically attributed to oxidative stress, mitochondrial damage, disrupted bile acid homeostasis, and innate immune-related inflammation. DILI can be divided into intrinsic and idiosyncratic DILI with cholestatic liver injury as an important manifestation. The diagnosis of DILI remains a challenge today and relies on clinical judgment and knowledge of the insulting agent. Early prediction of hepatotoxicity is an important but still unfulfilled component of drug development. In response, in silico modeling has shown good potential to fill the missing puzzle. Computer algorithms, with machine learning and artificial intelligence as a representative, can be established to initiate a reaction on the given condition to predict DILI. DILIsym is a mechanistic approach that integrates physiologically based pharmacokinetic modeling with the mechanisms of hepatoxicity and has gained increasing popularity for DILI prediction. This article reviews existing in silico approaches utilized to predict DILI risks in clinical medication and provides an overview of the underlying principles and related practical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qingfeng He
- Correspondence: (Q.H.); (X.X.); Tel.: +86-21-51980024 (X.X.)
| | - Xiaoqiang Xiang
- Correspondence: (Q.H.); (X.X.); Tel.: +86-21-51980024 (X.X.)
| |
Collapse
|
11
|
Moreno-Ajona D, Villar-Martínez MD, Goadsby PJ. New Generation Gepants: Migraine Acute and Preventive Medications. J Clin Med 2022; 11:1656. [PMID: 35329982 PMCID: PMC8953732 DOI: 10.3390/jcm11061656] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Migraine is a debilitating disease whose clinical and social impact is out of debate. Tolerability issues, interactions, contraindications, and inefficacy of the available medications make new options necessary. The calcitonin-gene-related peptide (CGRP) pathway has shown its importance in migraine pathophysiology and specific medications targeting this have become available. The first-generation CGRP receptor antagonists or gepants, have undergone clinical trials but their development was stopped because of hepatotoxicity. The new generation of gepants, however, are efficacious, safe, and well tolerated as per recent clinical trials. This led to the FDA-approval of rimegepant, ubrogepant, and atogepant. The clinical trials of the available gepants and some of the newer CGRP-antagonists are reviewed in this article.
Collapse
Affiliation(s)
- David Moreno-Ajona
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (D.M.-A.); (M.D.V.-M.)
- NIHR-Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre, King’s College Hospital, London SE5 9RS, UK
| | - María Dolores Villar-Martínez
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (D.M.-A.); (M.D.V.-M.)
- NIHR-Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre, King’s College Hospital, London SE5 9RS, UK
| | - Peter J. Goadsby
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (D.M.-A.); (M.D.V.-M.)
- NIHR-Wellcome Trust King’s Clinical Research Facility/SLaM Biomedical Research Centre, King’s College Hospital, London SE5 9RS, UK
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
12
|
Ribeiro dos Santos JB, Ribeiro da Silva MR. Small molecule CGRP receptor antagonists for the preventive treatment of migraine: A review. Eur J Pharmacol 2022; 922:174902. [DOI: 10.1016/j.ejphar.2022.174902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 01/29/2023]
|
13
|
Watkins PB. Liver Injury Due to Drugs and Viruses: Mechanistic Similarities and Implications for AAV Gene Therapy. Clin Pharmacol Ther 2021; 112:751-753. [PMID: 34910298 DOI: 10.1002/cpt.2500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/24/2021] [Indexed: 01/25/2023]
Affiliation(s)
- Paul B Watkins
- Institute for Drug Safety Sciences, University of North Carolina - Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
14
|
Clinton JW, Kiparizoska S, Aggarwal S, Woo S, Davis W, Lewis JH. Drug-Induced Liver Injury: Highlights and Controversies in the Recent Literature. Drug Saf 2021; 44:1125-1149. [PMID: 34533782 PMCID: PMC8447115 DOI: 10.1007/s40264-021-01109-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) remains an important, yet challenging diagnosis for physicians. Each year, additional drugs are implicated in DILI and this year was no different, with more than 1400 articles published on the subject. This review examines some of the most significant highlights and controversies in DILI-related research over the past year and their implications for clinical practice. Several new drugs were approved by the US Food and Drug Administration including a number of drugs implicated in causing DILI, particularly among the chemotherapeutic classes. The COVID-19 pandemic was also a major focus of attention in 2020 and we discuss some of the notable aspects of COVID-19-related liver injury and its implications for diagnosing DILI. Updates in diagnostic and causality assessments related to DILI such as the Roussel Uclaf Causality Assessment Method are included, mindful that there is still no single biomarker or diagnostic tool to unequivocally diagnose DILI. Glutamate dehydrogenase received renewed attention as being more specific than alanine aminotransferase. There were a few new reports of previously unrecognized hepatotoxins, including immune modulators and novel gene therapy drugs that we highlight. Updates and new developments of previously described hepatotoxins, such as immune checkpoint inhibitors and anti-tuberculosis drugs are reviewed. Finally, novel technologies such as organoid culture systems to better predict DILI preclinically may be coming of age and determinants of hepatocyte loss, such as calculating PALT are poised to improve our current means of estimating DILI severity and the risk of acute liver failure.
Collapse
Affiliation(s)
- Joseph William Clinton
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA.
| | - Sara Kiparizoska
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Soorya Aggarwal
- Division of Gastroenterology and Hepatology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Stephanie Woo
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - William Davis
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - James H Lewis
- Division of Gastroenterology and Hepatology, Medstar Georgetown University Hospital, Washington, DC, USA
| |
Collapse
|
15
|
Tosca EM, Bartolucci R, Magni P, Poggesi I. Modeling approaches for reducing safety-related attrition in drug discovery and development: a review on myelotoxicity, immunotoxicity, cardiovascular toxicity, and liver toxicity. Expert Opin Drug Discov 2021; 16:1365-1390. [PMID: 34181496 DOI: 10.1080/17460441.2021.1931114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction:Safety and tolerability is a critical area where improvements are needed to decrease the attrition rates during development of new drug candidates. Modeling approaches, when smartly implemented, can contribute to this aim.Areas covered:The focus of this review was on modeling approaches applied to four kinds of drug-induced toxicities: hematological, immunological, cardiovascular (CV) and liver toxicity. Papers, mainly published in the last 10 years, reporting models in three main methodological categories - computational models (e.g., quantitative structure-property relationships, machine learning approaches, neural networks, etc.), pharmacokinetic-pharmacodynamic (PK-PD) models, and quantitative system pharmacology (QSP) models - have been considered.Expert opinion:The picture observed in the four examined toxicity areas appears heterogeneous. Computational models are typically used in all areas as screening tools in the early stages of development for hematological, cardiovascular and liver toxicity, with accuracies in the range of 70-90%. A limited number of computational models, based on the analysis of drug protein sequence, was instead proposed for immunotoxicity. In the later stages of development, toxicities are quantitatively predicted with reasonably good accuracy using either semi-mechanistic PK-PD models (hematological and cardiovascular toxicity), or fully exploited QSP models (immuno-toxicity and liver toxicity).
Collapse
Affiliation(s)
- Elena M Tosca
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Roberta Bartolucci
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Paolo Magni
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Italo Poggesi
- Clinical Pharmacology & Pharmacometrics, Janssen Research & Development, Beerse, Belgium
| |
Collapse
|
16
|
Min KC, Kraft WK, Bondiskey P, Colón-González F, Liu W, Xu J, Panebianco D, Mixson L, Dockendorf MF, Matthews CZ, Boinpally R. Atogepant Is Not Associated With Clinically Meaningful Alanine Aminotransferase Elevations in Healthy Adults. Clin Transl Sci 2020; 14:599-605. [PMID: 33142014 PMCID: PMC7993278 DOI: 10.1111/cts.12917] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/04/2020] [Indexed: 11/26/2022] Open
Abstract
Atogepant is a potent, selective, oral calcitonin gene–related peptide (CGRP) receptor antagonist in development for migraine prevention. The chemical structure of atogepant is distinct from previous CGRP receptor antagonists, which were associated with elevated serum alanine aminotransferase (ALT) in clinical trials. Here, we report the safety, tolerability, and pharmacokinetics (PKs) of a once‐daily supratherapeutic dose (170 mg) of atogepant for 28 days from a randomized, double‐blind, placebo‐controlled phase I trial in healthy participants. Overall safety, hepatic safety, and plasma PK parameters were evaluated. Thirty‐four participants aged 23–55 years enrolled; 28 (82.4%) completed the study in accordance with the protocol. Multiple doses of 170 mg atogepant for 28 consecutive days were generally well‐tolerated. All adverse events (AEs; reported in 87.0% of the atogepant group; 72.7%, placebo) were mild in severity except one serious AE of subarachnoid hemorrhage due to a bicycle accident and not considered related to treatment. There were two discontinuations due to AEs, both with atogepant, one considered possibly related to treatment. Over 28 days of treatment, no participant receiving atogepant had an ALT elevation above 1.5 × upper limit of normal. Change from baseline in serum ALT levels was not different between atogepant and placebo. Atogepant is rapidly absorbed (median time to maximum plasma concentration, ~ 2 hours) with an apparent terminal half‐life of ~ 11 hours, and no evidence of accumulation after once‐daily dosing. Overall, atogepant at a high oral dose is safe and well‐tolerated in healthy participants with no clinically meaningful elevations in ALT.
Collapse
Affiliation(s)
- K Chris Min
- Formerly of Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Walter K Kraft
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | - Wen Liu
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Jialin Xu
- Formerly of Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | - Lori Mixson
- Formerly of Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | |
Collapse
|