1
|
Ford M, Thomson PJ, Lister A, Snoeys J, Leclercq L, Cuyckens F, Naisbitt DJ, Meng X. Bioactivation of the β-Amyloid Precursor Protein-Cleaving Enzyme 1 Inhibitor Atabecestat Leads to Protein Adduct Formation on Glutathione S-Transferase Pi. Chem Res Toxicol 2025; 38:812-815. [PMID: 40326176 DOI: 10.1021/acs.chemrestox.5c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Exposure to atabecestat is associated with liver injury, which subsequently led to its withdrawal from development. Previous studies of patients with atabecestat induced liver injury identified T cells responsive to atabecestat and its metabolites, indicating that immune-mediated mechanisms are involved. As irreversible protein modification is suspected to drive immunogenicity, this study aimed to characterize potential atabecestat protein adducts using HSA, GSTA1, and GSTP as model proteins. We have shown that atabecestat only formed a cysteine adduct on GSTP in the presence of metabolic systems, highlighting the important role of bioactivation in adduct formation and selectivity for the binding interaction.
Collapse
Affiliation(s)
- Megan Ford
- Department Pharmacology and Therapeutics, University of Liverpool, Liverpool L693GE, U.K
| | - Paul J Thomson
- Department Pharmacology and Therapeutics, University of Liverpool, Liverpool L693GE, U.K
| | - Adam Lister
- Department Pharmacology and Therapeutics, University of Liverpool, Liverpool L693GE, U.K
| | - Jan Snoeys
- Translational Pharmacokinetics Pharmacodynamics and Investigative Toxicology, Johnson & Johnson, 2340 Beerse, Belgium
| | - Laurent Leclercq
- Translational Pharmacokinetics Pharmacodynamics and Investigative Toxicology, Johnson & Johnson, 2340 Beerse, Belgium
| | - Filip Cuyckens
- Translational Pharmacokinetics Pharmacodynamics and Investigative Toxicology, Johnson & Johnson, 2340 Beerse, Belgium
| | - Dean J Naisbitt
- Department Pharmacology and Therapeutics, University of Liverpool, Liverpool L693GE, U.K
| | - Xiaoli Meng
- Department Pharmacology and Therapeutics, University of Liverpool, Liverpool L693GE, U.K
| |
Collapse
|
2
|
Leclercq L, de Vries R, Koppen V, Verboven P, Cuyckens F, Wynant I, Vermeulen WAA, Naisbitt D, Ford M, Meng X, Sakamoto S, Fukushima T, Snoeys J. CYP3A4-Mediated Bioactivation of the β-Amyloid Precursor Protein-Cleaving Enzyme 1 Inhibitor JNJ-54861911 Results in Redox-Neutral Addition of Glutathione via Catalysis by Glutathione S-Transferase α1, Identified as the Major Target Protein in Human Hepatocytes. Chem Res Toxicol 2025; 38:58-72. [PMID: 39662968 DOI: 10.1021/acs.chemrestox.4c00279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
The β-amyloid precursor protein-cleaving enzyme 1 (BACE1) inhibitor JNJ-54861911, a candidate for the treatment of Alzheimer's disease, was withdrawn from clinical trials due to drug-induced liver injury (DILI). This paper describes our investigation of the metabolism of JNJ-54861911 to understand the potential contribution to the observed DILI. In human hepatocytes, JNJ-54861911 is metabolized by CYP450 3A4 to a reactive intermediate (RI), which undergoes glutathione (GSH) addition at C6 of the 2-amino-4-methyl-1,3-thiazin-4-yl moiety via glutathione S-transferase α1 (GSTA1) catalysis. Despite the preponderant role of CYP3A4 as an enabler, the adduct has the same level of oxidation as that of JNJ-54861911. The exact mechanism of RI formation might involve a sulfoxide (with further reduction) or tautomeric forms of JNJ-54861911 bearing a reactive thiazinium cation activating both the C2 and C6 positions. The cell pellet from the human hepatocyte incubated with 14C-JNJ-54861911 was analyzed via gel electrophoresis, resulting in the identification of a major protein adduct on GSTA1, a cross-link resulting from the addition of GSH and lysine 120 to JNJ-54861911, most likely on position C6 and on the nitrile, respectively. Ultimately, this major adduct might only account for 15-25% of the total covalent binding (CVB). Other important contributors to CVB might exist, like the bioactivation of the major diaminothiazine metabolite (DIAT). The level of covalent binding (CVB) burden (fraction of the dose resulting in CVB) is clearly below 1 mg/day, with a low daily dose of 25 mg. Despite this limited magnitude of CVB, it could still contribute to the liver enzyme elevations observed in approximately 20% of the patients and to the few cases of severe immune-mediated DILI. The latter could occur through a haptenization phenomenon and/or by inducing stress in hepatocytes. Such stress may activate an innate immune response, which, in turn, promotes the adaptive immune response.
Collapse
Affiliation(s)
- Laurent Leclercq
- Translational Pharmacokinetics, Pharmacodynamics and Investigative Toxicology, Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Ronald de Vries
- Advanced Material Characterization & Investigations (AMCI), Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Valérie Koppen
- Chemical and Pharmaceutical Development & Supply, Analytical Development, Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Peter Verboven
- Translational Pharmacokinetics, Pharmacodynamics and Investigative Toxicology, Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Filip Cuyckens
- Translational Pharmacokinetics, Pharmacodynamics and Investigative Toxicology, Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Inneke Wynant
- Translational Pharmacokinetics, Pharmacodynamics and Investigative Toxicology, Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Wim A A Vermeulen
- Advanced Material Characterization & Investigations (AMCI), Janssen Research & Development, LLC, 2340 Beerse, Belgium
| | - Dean Naisbitt
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, U.K
| | - Megan Ford
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, U.K
| | - Xiaoli Meng
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, U.K
| | - Shingo Sakamoto
- DMPK & Analytical Sciences 2, Laboratory for Drug Discovery and Development, Shionogi, Toyonaka City 5610825, Osaka, Japan
| | - Tamio Fukushima
- Non-Clinical Safety, Laboratory for Drug Discovery and Development, Shionogi, Toyonaka City 6510825, Osaka, Japan
| | - Jan Snoeys
- Translational Pharmacokinetics, Pharmacodynamics and Investigative Toxicology, Janssen Research & Development, LLC, 2340 Beerse, Belgium
| |
Collapse
|
3
|
Ford M, Thomson PJ, Snoeys J, Meng X, Naisbitt DJ. Selective HLA Class II Allele-Restricted Activation of Atabecestat Metabolite-Specific Human T-Cells. Chem Res Toxicol 2024; 37:1712-1727. [PMID: 39348529 PMCID: PMC11497358 DOI: 10.1021/acs.chemrestox.4c00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/02/2024]
Abstract
Elevations in hepatic enzymes were detected in several trial patients exposed to the Alzheimer's drug atabecestat, which resulted in termination of the drug development program. Characterization of hepatic T-lymphocyte infiltrates and diaminothiazine (DIAT) metabolite-responsive, human leukocyte antigen (HLA)-DR-restricted, CD4+ T-lymphocytes in the blood of patients confirmed an immune pathogenesis. Patients with immune-mediated liver injury expressed a restricted panel of HLA-DRB1 alleles including HLA-DRB1*12:01, HLA-DRB1*13:02, and HLA-DRB1*15:01. Thus, the objectives of this study were to (i) generate DIAT-responsive T-cell clones from HLA-genotyped drug-naive donors, (ii) characterize pathways of DIAT-specific T-cell activation, and (iii) assess HLA allele restriction of the DIAT-specific T-cell response. Sixteen drug-naive donors expressing the HLA-DR molecules outlined above were recruited, and T-cell clones were generated. Cellular phenotype, function, and HLA-allele restriction were assessed using culture assays. Peptides displayed by HLA class II molecules in the presence and absence of atabecestat were analyzed by mass spectrometry. Several DIAT-responsive CD4+ clones, displaying no reactivity toward the parent drug, were successfully generated from donors expressing HLA-DRB1*12:01, HLA-DRB1*13:02, and HLA-DRB1*15:01 but not from other donors expressing other HLA-DRB1 alleles. T-cell clones were activated following direct binding of DIAT to HLA-DR proteins expressed on the surface of antigen presenting cells. DIAT binding did not alter the HLA-DRB1 peptide binding repertoire, indicative of a binding interaction with the HLA-associated peptide rather than with the HLA protein itself. DIAT-specific T-cell responses displayed HLA-DRB1*12:01, HLA-DRB1*13:02, and HLA-DRB1*15:01 restriction. These data demonstrate that DIAT displays a degree of selectivity toward HLA protein and associated peptides, with expression of certain alleles increasing and that of others decreasing, the likelihood that a drug-specific T-cell response develops.
Collapse
Affiliation(s)
- Megan Ford
- Centre
for Drug Safety Science, Department of Pharmacology and Therapeutics,
Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Paul J. Thomson
- Centre
for Drug Safety Science, Department of Pharmacology and Therapeutics,
Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, U.K.
- AstraZeneca,
The Discovery Centre, Cambridge Biomedical
Campus, Cambridge CB2 0AA, U.K.
| | - Jan Snoeys
- Translational
PK PD and Investigative Toxicology, Janssen
Research & Development, Division of Janssen Pharmaceutica NV, Beerse 2340, Belgium
| | - Xiaoli Meng
- Centre
for Drug Safety Science, Department of Pharmacology and Therapeutics,
Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Dean J. Naisbitt
- Centre
for Drug Safety Science, Department of Pharmacology and Therapeutics,
Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, U.K.
| |
Collapse
|
4
|
Preethy H A, Rajendran K, Sukumar AJ, Krishnan UM. Emerging paradigms in Alzheimer's therapy. Eur J Pharmacol 2024; 981:176872. [PMID: 39117266 DOI: 10.1016/j.ejphar.2024.176872] [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: 03/08/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Alzheimer's disease is a neurodegenerative disorder that affects elderly, and its incidence is continuously increasing across the globe. Unfortunately, despite decades of research, a complete cure for Alzheimer's disease continues to elude us. The current medications are mainly symptomatic and slow the disease progression but do not result in reversal of all disease pathologies. The growing body of knowledge on the factors responsible for the onset and progression of the disease has resulted in the identification of new targets that could be targeted for treatment of Alzheimer's disease. This has opened new vistas for treatment of Alzheimer's disease that have moved away from chemotherapeutic agents modulating a single target to biologics and combinations that acted on multiple targets thereby offering better therapeutic outcomes. This review discusses the emerging directions in therapeutic interventions against Alzheimer's disease highlighting their merits that promise to change the treatment paradigm and challenges that limit their clinical translation.
Collapse
Affiliation(s)
- Agnes Preethy H
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Anitha Josephine Sukumar
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
5
|
Pallardy M, Bechara R, Whritenour J, Mitchell-Ryan S, Herzyk D, Lebrec H, Merk H, Gourley I, Komocsar WJ, Piccotti JR, Balazs M, Sharma A, Walker DB, Weinstock D. Drug hypersensitivity reactions: review of the state of the science for prediction and diagnosis. Toxicol Sci 2024; 200:11-30. [PMID: 38588579 PMCID: PMC11199923 DOI: 10.1093/toxsci/kfae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Drug hypersensitivity reactions (DHRs) are a type of adverse drug reaction that can occur with different classes of drugs and affect multiple organ systems and patient populations. DHRs can be classified as allergic or non-allergic based on the cellular mechanisms involved. Whereas nonallergic reactions rely mainly on the innate immune system, allergic reactions involve the generation of an adaptive immune response. Consequently, drug allergies are DHRs for which an immunological mechanism, with antibody and/or T cell, is demonstrated. Despite decades of research, methods to predict the potential for a new chemical entity to cause DHRs or to correctly attribute DHRs to a specific mechanism and a specific molecule are not well-established. This review will focus on allergic reactions induced by systemically administered low-molecular weight drugs with an emphasis on drug- and patient-specific factors that could influence the development of DHRs. Strategies for predicting and diagnosing DHRs, including potential tools based on the current state of the science, will also be discussed.
Collapse
Affiliation(s)
- Marc Pallardy
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, Orsay, 91400, France
| | - Rami Bechara
- Université Paris-Saclay, INSERM, CEA, Center for Research in Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB), Le Kremlin Bicêtre, 94270, France
| | - Jessica Whritenour
- Pfizer Worldwide Research, Development and Medical, Groton, Connecticut 06340, USA
| | - Shermaine Mitchell-Ryan
- The Health and Environmental Science Institute, Immunosafety Technical Committee, Washington, District of Columbia 20005, USA
| | - Danuta Herzyk
- Merck & Co., Inc, West Point, Pennsylvania 19486, USA
| | - Herve Lebrec
- Amgen Inc., Translational Safety and Bioanalytical Sciences, South San Francisco, California 94080, USA
| | - Hans Merk
- Department of Dermatology and Allergology, RWTH Aachen University, Aachen, 52062, Germany
| | - Ian Gourley
- Janssen Research & Development, LLC, Immunology Clinical Development, Spring House, Pennsylvania 19002, USA
| | - Wendy J Komocsar
- Immunology Business Unit, Eli Lilly and Company, Indianapolis, Indiana 46225, USA
| | | | - Mercedesz Balazs
- Genentech, Biochemical and Cellular Pharmacology, South San Francisco, California 94080, USA
| | - Amy Sharma
- Pfizer, Drug Safety Research & Development, New York 10017, USA
| | - Dana B Walker
- Novartis Institute for Biomedical Research, Preclinical Safety-Translational Immunology and Clinical Pathology, Cambridge, Massachusetts 02139, USA
| | - Daniel Weinstock
- Janssen Research & Development, LLC, Preclinical Sciences Translational Safety, Spring House, Pennsylvania 19002, USA
| |
Collapse
|
6
|
Gu X, Qi L, Qi Q, Zhou J, Chen S, Wang L. Monoclonal antibody therapy for Alzheimer's disease focusing on intracerebral targets. Biosci Trends 2024; 18:49-65. [PMID: 38382942 DOI: 10.5582/bst.2023.01288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Due to the complexity of the disorder and the presence of the blood-brain barrier (BBB), its drug discovery and development are facing enormous challenges, especially after several failures of monoclonal antibody (mAb) trials. Nevertheless, the Food and Drug Administration's approval of the mAb aducanumab has ushered in a new day. As we better understand the disease's pathogenesis and identify novel intracerebral therapeutic targets, antibody-based therapies have advanced over the past few years. The mAb drugs targeting β-amyloid or hyperphosphorylated tau protein are the focus of the current research. Massive neuronal loss and glial cell-mediated inflammation are also the vital pathological hallmarks of AD, signaling a new direction for research on mAb drugs. We have elucidated the mechanisms by which AD-specific mAbs cross the BBB to bind to targets. In order to investigate therapeutic approaches to treat AD, this review focuses on the promising mAbs targeting intracerebral dysfunction and related strategies to cross the BBB.
Collapse
Affiliation(s)
- Xiaolei Gu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Long Qi
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Qing Qi
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Song Chen
- Postdoctoral Station of Xiamen University, Fujian, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
7
|
Thomson P, Fragkas N, Kafu LM, Aithal GP, Lucena MI, Terracciano L, Meng X, Pirmohamed M, Brees D, Kullak‐Ublick GA, Odermatt A, Hammond T, Kammüller M, Naisbitt DJ. Patients with naproxen-induced liver injury display T-cell memory responses toward an oxidative (S)-O-desmethyl naproxen metabolite but not the acyl glucuronide. Allergy 2024; 79:200-214. [PMID: 37515456 PMCID: PMC10952231 DOI: 10.1111/all.15830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Exposure to nonsteroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen (IBU) and naproxen (NAP) is associated with idiosyncratic drug-induced liver injury (DILI). Carboxylate bioactivation into reactive metabolites (e.g., acyl glucuronides, AG) and resulting T-cell activation is hypothesized as causal for this adverse event. However, conclusive evidence supporting this is lacking. METHODS In this work, we identify CD4+ and CD8+ T-cell hepatic infiltration in a biopsy from an IBU DILI patient. Lymphocyte transformation test and IFN-γ ELIspot, conducted on peripheral blood mononuclear cells (PBMCs) of patients with NAP-DILI, were used to explore drug-specific T-cell activation. T-cell clones (TCC) were generated and tested for drug specificity, phenotype/function, and pathways of T-cell activation. Cells were exposed to NAP, its oxidative metabolite 6-O-desmethyl NAP (DM-NAP), its AG or synthesized NAP-AG human-serum albumin adducts (NAP-AG adduct). RESULTS CD4+ and CD8+ T-cells from patients expressing a range of different Vβ receptors were stimulated to proliferate and secrete IFN-γ and IL-22 when exposed to DM-NAP, but not NAP, NAP-AG or the NAP-AG adduct. Activation of the CD4+ TCC was HLA-DQ-restricted and dependent on antigen presenting cells (APC); most TCC were activated with DM-NAP-pulsed APC, while fixation of APC blocked the T-cell response. Cross-reactivity was not observed with structurally-related drugs. CONCLUSION Our results confirm hepatic T-cell infiltrations in NSAID-induced DILI, and show a T-cell memory response toward DM-NAP indicating an immune-mediated basis for the adverse event. Whilst bioactivation at the carboxylate group is widely hypothesized to be pathogenic for NSAID associated DILI, we found no evidence of this with NAP.
Collapse
Affiliation(s)
- Paul Thomson
- Molecular& Clinical PharmacologyUniversity of LiverpoolLiverpoolUK
| | - Nik Fragkas
- Novartis Institutes for BioMedical ResearchBaselSwitzerland
| | - Laila M. Kafu
- Molecular& Clinical PharmacologyUniversity of LiverpoolLiverpoolUK
| | - Guruprasad P. Aithal
- NIHR Nottingham Biomedical Research Centre and Nottingham Digestive Diseases Centre, Translational Medical Sciences, West Block, Queen's Medical CentreUniversity of NottinghamNottinghamUK
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Aparato Digestivo y Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga‐IBIMA, Hospital Universitario Virgen de la VictoriaUniversidad de Málaga, CIBERehdMalagaSpain
| | | | - Xiaoli Meng
- Molecular& Clinical PharmacologyUniversity of LiverpoolLiverpoolUK
| | - Munir Pirmohamed
- Molecular& Clinical PharmacologyUniversity of LiverpoolLiverpoolUK
| | | | - Gerd A. Kullak‐Ublick
- University Hospital ZurichUniversity of ZurichZurichSwitzerland
- Novartis Global Drug DevelopmentBaselSwitzerland
| | - Alex Odermatt
- Division of Molecular & Systems Toxicology, Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
| | - Thomas Hammond
- Division of Molecular & Systems Toxicology, Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
- Oncology Safety, Clinical Pharmacology and Safety Sciences R&DCambridgeUK
| | | | - Dean J. Naisbitt
- Molecular& Clinical PharmacologyUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
8
|
Li QS, Francke S, Snoeys J, Thipphawong J, Romano G, Novak GP. Genome-wide association study of abnormal elevation of ALT in patients exposed to atabecestat. BMC Genomics 2023; 24:513. [PMID: 37658353 PMCID: PMC10472559 DOI: 10.1186/s12864-023-09625-6] [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: 04/17/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Atabecestat, a potent brain penetrable BACE1 inhibitor that reduces CSF amyloid beta (Aβ), was developed as an oral treatment for Alzheimer's disease (AD). Elevated liver enzyme adverse events were reported in three studies although only one case met Hy's law criteria to predict serious hepatotoxicity. METHOD We performed a case-control genome-wide association study (GWAS) to identify genetic risk variants associated with liver enzyme elevation using 42 cases with alanine transaminase (ALT) above three times the upper limit of normal (ULN) and 141 controls below ULN. Additionally, we performed a GWAS using continuous maximal ALT/ULN (expressed as times the ULN) upon exposure to atabecestat as the outcome measure (n = 285). RESULTS No variant passed the genome-wide significance threshold (p = 5 × 10- 8) in the case-control GWAS. We identified suggestive association signals in genes (NLRP1, SCIMP, and C1QBP) implicated in the inflammatory processes. Among the genes implicated by position mapping using variants suggestively associated (p < 1 × 10- 5) with ALT elevation case-control status, gene sets involved in innate immune response (adjusted p-value = 0.05) and regulation of cytokine production (adjusted p-value = 0.04) were enriched. One genomic region in the intronic region of GABRG3 passed the genome-wide significance threshold in the continuous max(ALT/ULN) GWAS, and this variant was nominally associated with ALT elevation case status (p = 0.009). CONCLUSION The suggestive GWAS signals in the case-control GWAS analysis suggest the potential role of inflammation in atabecestat-induced liver enzyme elevation.
Collapse
Affiliation(s)
- Qingqin S Li
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA.
- JRD Data Science, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA.
| | - Stephan Francke
- Computational Science Translational Platforms, Janssen Research & Development, LLC, Spring House, PA, 19477, USA
- Pharmacogenomics & Biomarker in Clinical Development, Cary, NC, USA
| | - Jan Snoeys
- Translational Pharmacokinetics Pharmacodynamics and Investigative Toxicology, Janssen Research & Development, Beerse, Belgium
| | - John Thipphawong
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
| | - Gary Romano
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
- Passage Bio, Philadelphia, PA, USA
| | - Gerald P Novak
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
| |
Collapse
|
9
|
Bazzari FH, Bazzari AH. BACE1 Inhibitors for Alzheimer's Disease: The Past, Present and Any Future? MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248823. [PMID: 36557955 PMCID: PMC9785888 DOI: 10.3390/molecules27248823] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and the most common cause of dementia in the elderly. The complexity of AD has hindered the development of either a cure or a disease-modifying therapy to halt the disease progression. Numerous hypotheses were presented in order to explain the mechanisms underlying the pathogenesis of AD. Introduced in 1992, the "Amyloid Cascade Hypothesis" had a huge impact on the field and inspired the rise of various drug candidates, especially amyloid-beta (Aβ)-directed drugs; including beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors. Adopted by a number of pharmaceutical companies, the development of BACE1 inhibitors has gained momentum in the past decade with promising results from experimental and early clinical-phase studies. Nevertheless, nearly all BACE1 inhibitors failed in later phases of clinical trials, due to safety and/or efficacy issues, and others were discontinued early in favor of second-generation small-molecule candidates. This paper aims to provide a comprehensive review of all BACE1 inhibitors to ever reach clinical trials, and we discuss the challenges and different perspectives on whether BACE1 inhibitors are to be reconsidered or revitalized in the future.
Collapse
Affiliation(s)
- Firas H. Bazzari
- Faculty of Pharmacy, Jerash University, Al-Urdon St., Jerash 26150, Jordan
- Correspondence:
| | - Amjad H. Bazzari
- Department of Basic Scientific Sciences, Faculty of Arts & Sciences, Applied Science Private University, Amman 11931, Jordan
| |
Collapse
|
10
|
Thomson P, Hammond S, Naisbitt DJ. Pathology of drug hypersensitivity reactions and mechanisms of immune tolerance. Clin Exp Allergy 2022; 52:1379-1390. [PMID: 36177544 DOI: 10.1111/cea.14235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/08/2022] [Accepted: 09/25/2022] [Indexed: 01/26/2023]
Abstract
Immune-mediated type IV adverse drug reactions are idiosyncratic in nature, generally not related to the primary or secondary pharmacology of the drug. Due to their complex nature and rarity, these iatrogenic reactions are seldom predicted or encountered during preclinical/early clinical development stages, and often precipitate upon exposure to wider populations (i.e. phase III onwards). They confer a burden on the healthcare sector in both a clinical and financial sense presenting a severe impediment to the drug discovery and development process. Research over the past 50 years has improved our understanding of these reactions markedly as both in vitro and in vivo studies have placed the role of the immune system, in particular; drug-responsive T cells, firmly in the spotlight as the mediators of these reactions. Indeed, the role of different populations of T cells in adverse events and the interaction of drug molecules with HLA proteins expressed on the surface of antigen-presenting cells is of considerable interest. Herein, this review examines the pathways of immune-mediated adverse events including the various T cell subtypes implicated and the mechanisms of T cell activation. Additionally, we address the enigma of immunological tolerance and explore the role tolerance plays in determination of susceptibility to such adverse events even in individuals carrying immunogenic liabilities.
Collapse
Affiliation(s)
- Paul Thomson
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - Sean Hammond
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK.,ApconiX, Alderley Park, Alderley Edge, UK
| | - Dean J Naisbitt
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
11
|
The "Cerebrospinal Fluid Sink Therapeutic Strategy" in Alzheimer's Disease-From Theory to Design of Applied Systems. Biomedicines 2022; 10:biomedicines10071509. [PMID: 35884814 PMCID: PMC9313192 DOI: 10.3390/biomedicines10071509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a global health problem, with incidence and prevalence considered to increase during the next decades. However, no currently available effective treatment exists despite numerous clinical trials in progress. Moreover, although many hypotheses are accepted regarding the pathophysiological mechanisms of AD onset and evolution, there are still many unknowns about the disorder. A relatively new approach, based on the amyloid-beta dynamics among different biological compartments, is currently intensely discussed, as it seems to offer a promising solution with significant therapeutic impact. Known as the “cerebrospinal-fluid-sink therapeutic strategy”, part of the “three-sink therapeutic strategy”, this theoretical model focuses on the dynamics of amyloid-beta among the three main liquid compartments of the human body, namely blood, cerebrospinal fluid, and the (brain) interstitial fluid. In this context, this article aims to describe in detail the abovementioned hypothesis, by reviewing in the first part the most relevant anatomical and physiological aspects of amyloid-beta dynamics. Subsequently, explored therapeutic strategies based on the clearance of amyloid-beta from the cerebrospinal fluid level are presented, additionally highlighting their limitations. Finally, the originality and novelty of this work rely on the research experience of the authors, who focus on implantable devices and their utility in AD treatment.
Collapse
|
12
|
Fernandez‐Santamaria R, Ariza A, Fernandez TD, Cespedes JA, Labella M, Mayorga C, Torres MJ. Advances and highlights in T and B cell responses to drug antigens. Allergy 2022; 77:1129-1138. [PMID: 34617287 DOI: 10.1111/all.15126] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 03/31/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022]
Abstract
The immunological mechanisms involved in drug hypersensitivity reactions (DHRs) are complex, and despite important advances, multiple aspects remain poorly understood. These not fully known aspects are mainly related to the factors that drive towards either a tolerant or a hypersensitivity response and specifically regarding the role of B and T cells. In this review, we focus on recent findings on this knowledge area within the last 2 years. We highlight new evidences of covalent and non-covalent interactions of drug antigen with proteins, as well as the very first characterization of naturally processed flucloxacillin-haptenated human leukocyte antigen (HLA) ligands. Moreover, we have analysed new insights into the identification of risk factors associated with the development of DHRs, such as the role of oxidative metabolism of drugs in the activation of the immune system and the discovery of new associations between DHRs and HLA variants. Finally, evidence of IgG-mediated anaphylaxis in humans and the involvement of specific subpopulations of effector cells associated with different clinical entities are also topics explored in this review. All these recent findings are relevant for the underlying pathology mechanisms and advance the field towards a more precise diagnosis, management and treatment approach for DHRs.
Collapse
Affiliation(s)
| | - Adriana Ariza
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA‐ARADyAL Málaga Spain
| | - Tahia D. Fernandez
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA‐ARADyAL Málaga Spain
- Departamento de Biología Celular Genética y Fisiología Universidad de Málaga Málaga Spain
| | - José A Cespedes
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA‐ARADyAL Málaga Spain
| | - Marina Labella
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA‐ARADyAL Málaga Spain
- Allergy Unit Hospital Regional Universitario de Málaga‐ARADyAL Málaga Spain
| | - Cristobalina Mayorga
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA‐ARADyAL Málaga Spain
- Allergy Unit Hospital Regional Universitario de Málaga‐ARADyAL Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
| | - María J Torres
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA‐ARADyAL Málaga Spain
- Allergy Unit Hospital Regional Universitario de Málaga‐ARADyAL Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
- Departamento de Medicina Universidad de Málaga Málaga Spain
| |
Collapse
|
13
|
Zhou X, Yu L, Zhou M, Hou P, Yi L, Mi M. Dihydromyricetin ameliorates liver fibrosis via inhibition of hepatic stellate cells by inducing autophagy and natural killer cell-mediated killing effect. Nutr Metab (Lond) 2021; 18:64. [PMID: 34147124 PMCID: PMC8214786 DOI: 10.1186/s12986-021-00589-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022] Open
Abstract
Background This study investigated the mechanisms underlying the preventive effect of dihydromyricetin (DHM) against liver fibrosis involving hepatic stellate cells (HSCs) and hepatic natural killer (NK) cells. Methods A carbon tetrachloride (CCl4)-induced liver fibrosis model was established in C57BL/6 mice to study the antifibrotic effect of DHM based on serum biochemical parameters, histological and immunofluorescence stainings, and the expression of several fibrosis-related markers. Based on the immunoregulatory role of DHM, the effect of DHM on NK cell activation ex vivo was evaluated by flow cytometry. Then, we investigated whether DHM-induced autophagy was involved in HSCs inactivation using enzyme-linked immunosorbent assays, transmission electron microscopy, and western blot analysis. Thereafter, the role of DHM in NK cell-mediated killing was studied by in vitro coculture of NK cells and HSCs, with subsequent analysis by flow cytometry. Finally, the mechanism by which DHM regulates NK cells was studied by western blot analysis. Results DHM ameliorated liver fibrosis in C57BL/6 mice, as characterized by decreased serum alanine transaminase and aspartate transaminase levels, decreased expressions of collagen I alpha 1 (CoL-1α1), collagen I alpha 2 (CoL-1α2), tissue inhibitor of metalloproteinases 1 (TIMP-1), α-smooth muscle actin (α-SMA) and desmin, as well as increased expression of matrix metalloproteinase 1 (MMP1). Interestingly, HSCs activation was significantly inhibited by DHM in vivo and in vitro. As expected, DHM also upregulated autophagy-related indicators in liver from CCl4-treated mice. DHM also prevented TGF-β1-induced activation of HSCs in vitro by initiating autophagic flux. In contrast, the autophagy inhibitor 3-methyladenine markedly abolished the antifibrotic effect of DHM. Surprisingly, the frequency of activated intrahepatic NK cells was significantly elevated by DHM ex vivo. Furthermore, DHM enhanced NK cell-mediated killing of HSCs by increasing IFN-γ expression, which was abolished by an anti-IFN-γ neutralizing antibody. Mechanistically, DHM-induced IFN-γ expression was through AhR-NF-κB/STAT3 pathway in NK cells. Conclusion These results demonstrated that DHM can ameliorate the progression of liver fibrosis and inhibition of HSCs activation by inducing autophagy and enhancing NK cell-mediated killing through the AhR-NF-κB/STAT3-IFN-γ signaling pathway, providing new insights into the preventive role of DHM in liver fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12986-021-00589-6.
Collapse
Affiliation(s)
- Xi Zhou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO. 30th Gao Tan Yan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Li Yu
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO. 30th Gao Tan Yan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Min Zhou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO. 30th Gao Tan Yan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Pengfei Hou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO. 30th Gao Tan Yan Street, Shapingba District, Chongqing, 400038, People's Republic of China
| | - Long Yi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO. 30th Gao Tan Yan Street, Shapingba District, Chongqing, 400038, People's Republic of China.
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University (Army Medical University), NO. 30th Gao Tan Yan Street, Shapingba District, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
14
|
Hammond S, Thomson P, Meng X, Naisbitt D. In-Vitro Approaches to Predict and Study T-Cell Mediated Hypersensitivity to Drugs. Front Immunol 2021; 12:630530. [PMID: 33927714 PMCID: PMC8076677 DOI: 10.3389/fimmu.2021.630530] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/17/2021] [Indexed: 01/11/2023] Open
Abstract
Mitigating the risk of drug hypersensitivity reactions is an important facet of a given pharmaceutical, with poor performance in this area of safety often leading to warnings, restrictions and withdrawals. In the last 50 years, efforts to diagnose, manage, and circumvent these obscure, iatrogenic diseases have resulted in the development of assays at all stages of a drugs lifespan. Indeed, this begins with intelligent lead compound selection/design to minimize the existence of deleterious chemical reactivity through exclusion of ominous structural moieties. Preclinical studies then investigate how compounds interact with biological systems, with emphasis placed on modeling immunological/toxicological liabilities. During clinical use, competent and accurate diagnoses are sought to effectively manage patients with such ailments, and pharmacovigilance datasets can be used for stratification of patient populations in order to optimise safety profiles. Herein, an overview of some of the in-vitro approaches to predict intrinsic immunogenicity of drugs and diagnose culprit drugs in allergic patients after exposure is detailed, with current perspectives and opportunities provided.
Collapse
Affiliation(s)
- Sean Hammond
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
- ApconiX, Alderley Park, Alderley Edge, United Kingdom
| | - Paul Thomson
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Dean Naisbitt
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|