1
|
Vinutha M, Sharma UR, Swamy G, Rohini S, Vada S, Janandri S, Haribabu T, Taj N, Gayathri SV, Jyotsna SK, Mudagal MP. COVID-19-related liver injury: Mechanisms, diagnosis, management; its impact on pre-existing conditions, cancer and liver transplant: A comprehensive review. Life Sci 2024; 356:123022. [PMID: 39214285 DOI: 10.1016/j.lfs.2024.123022] [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: 02/19/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
AIMS This review explores the mechanisms, diagnostic approaches, and management strategies for COVID-19-induced liver injury, with a focus on its impact on patients with pre-existing liver conditions, liver cancer, and those undergoing liver transplantation. MATERIALS AND METHODS A comprehensive literature review included studies on clinical manifestations of liver injury due to COVID-19. Key areas examined were direct viral effects, drug-induced liver injury, cytokine storms, and impacts on individuals with chronic liver diseases, liver transplants, and the role of vaccination. Data were collected from clinical trials, observational studies, case reports, and review literature. KEY FINDINGS COVID-19 can cause a spectrum of liver injuries, from mild enzyme elevations to severe hepatic dysfunction. Injury mechanisms include direct viral invasion, immune response alterations, drug toxicity, and hypoxia-reperfusion injury. Patients with chronic liver conditions (such as alcohol-related liver disease, nonalcoholic fatty liver disease, cirrhosis, and hepatocellular carcinoma) face increased risks of severe outcomes. The pandemic has worsened pre-existing liver conditions, disrupted cancer treatments, and complicated liver transplantation. Vaccination remains crucial for reducing severe disease, particularly in chronic liver patients and transplant recipients. Telemedicine has been beneficial in managing patients and reducing cross-infection risks. SIGNIFICANCE This review discusses the importance of improved diagnostic methods and management strategies for liver injury caused by COVID-19. It emphasizes the need for close monitoring and customized treatment for high-risk groups, advocating for future research to explore long-term effects, novel therapies, and evidence-based approaches to improve liver health during and after the pandemic.
Collapse
Affiliation(s)
- M Vinutha
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Uday Raj Sharma
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India.
| | - Gurubasvaraja Swamy
- Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - S Rohini
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Surendra Vada
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Suresh Janandri
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - T Haribabu
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Nageena Taj
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - S V Gayathri
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - S K Jyotsna
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| | - Manjunatha P Mudagal
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, India
| |
Collapse
|
2
|
Zhu L, Yang X, Wu S, Dong R, Yan Y, Lin N, Zhang B, Tan B. Hepatotoxicity of epidermal growth factor receptor - tyrosine kinase inhibitors (EGFR-TKIs). Drug Metab Rev 2024; 56:302-317. [PMID: 39120430 DOI: 10.1080/03602532.2024.2388203] [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: 03/27/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Drug-induced liver injury (DILI) is one of the most frequently adverse reactions in clinical drug use, usually caused by drugs or herbal compounds. Compared with other populations, cancer patients are more prone to abnormal liver function due to primary or secondary liver malignant tumor, radiation-induced liver injury and other reasons, making potential adverse reactions from liver damage caused by anticancer drugs of particular concernduring clinical treatment process. In recent years, the application of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) has changed the treatment status of a series of solid malignant tumors. Unfortunately, the increasing incidence of hepatotoxicitylimits the clinical application of EGFR-TKIs. The mechanisms of liver injury caused by EGFR-TKIs were complex. Despite more than a decade of research, other than direct damage to hepatocytes caused by inhibition of cellular DNA synthesis and resulting in hepatocyte necrosis, the rest of the specific mechanisms remain unclear, and few effective solutions are available. This review focuses on the clinical feature, incidence rates and the recent advances on the discovery of mechanism of hepatotoxicity in EGFR-TKIs, as well as rechallenge and therapeutic strategies underlying hepatotoxicity of EGFR-TKIs.
Collapse
Affiliation(s)
- Lulin Zhu
- Department of Pharmacy, Key Laboratory of Clinical CancerPharmacology andToxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Xinxin Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shanshan Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rong Dong
- Department of Pharmacy, Key Laboratory of Clinical CancerPharmacology andToxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Youyou Yan
- Department of Pharmacy, Key Laboratory of Clinical CancerPharmacology andToxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Nengming Lin
- Department of Pharmacy, Key Laboratory of Clinical CancerPharmacology andToxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Bo Zhang
- Department of Pharmacy, Key Laboratory of Clinical CancerPharmacology andToxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Biqin Tan
- Department of Pharmacy, Key Laboratory of Clinical CancerPharmacology andToxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
3
|
Khalil SM, Qin X, Hakenjos JM, Wang J, Hu Z, Liu X, Wang J, Maletic-Savatic M, MacKenzie KR, Matzuk MM, Li F. MALDI Imaging Mass Spectrometry Visualizes the Distribution of Antidepressant Duloxetine and Its Major Metabolites in Mouse Brain, Liver, Kidney, and Spleen Tissues. Drug Metab Dispos 2024; 52:673-680. [PMID: 38658163 PMCID: PMC11185819 DOI: 10.1124/dmd.124.001719] [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: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024] Open
Abstract
Imaging mass spectrometry (IMS) is a powerful tool for mapping the spatial distribution of unlabeled drugs and metabolites that may find application in assessing drug delivery, explaining drug efficacy, and identifying potential toxicity. This study focuses on determining the spatial distribution of the antidepressant duloxetine, which is widely prescribed despite common adverse effects (liver injury, constant headaches) whose mechanisms are not fully understood. We used high-resolution IMS with matrix-assisted laser desorption/ionization to examine the distribution of duloxetine and its major metabolites in four mouse organs where it may contribute to efficacy or toxicity: brain, liver, kidney, and spleen. In none of these tissues is duloxetine or its metabolites homogeneously distributed, which has implications for both efficacy and toxicity. We found duloxetine to be similarly distributed in spleen red pulp and white pulp but differentially distributed in different anatomic regions of the liver, kidney, and brain, with dose-dependent patterns. Comparison with hematoxylin and eosin staining of tissue sections reveals that the ion images of endogenous lipids help delineate anatomic regions in the brain and kidney, while heme ion images assist in differentiating regions within the spleen. These endogenous metabolites may serve as a valuable resource for examining the spatial distribution of other drugs in tissues when staining images are not available. These findings may facilitate future mechanistic studies of the therapeutic and adverse effects of duloxetine. In the current work, we did not perform absolute quantification of duloxetine, which will be reported in due course. SIGNIFICANCE STATEMENT: The study utilized imaging mass spectrometry to examine the spatial distribution of duloxetine and its primary metabolites in mouse brain, liver, kidney, and spleen. These results may pave the way for future investigations into the mechanisms behind duloxetine's therapeutic and adverse effects. Furthermore, the mass spectrometry images of specific endogenous metabolites such as heme could be valuable in analyzing the spatial distribution of other drugs within tissues in scenarios where histological staining images are unavailable.
Collapse
Affiliation(s)
- Saleh M Khalil
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xuan Qin
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - John M Hakenjos
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jian Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Zhaoyong Hu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xinli Liu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jin Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Mirjana Maletic-Savatic
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Martin M Matzuk
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Feng Li
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| |
Collapse
|
4
|
Kyriakaki I, Karanikola T, Lillis T, Kontonasaki E, Dabarakis N. Effect of direct oral anticoagulant dabigatran on early bone healing: An experimental study in rats. JOURNAL OF ADVANCED PERIODONTOLOGY & IMPLANT DENTISTRY 2023; 15:86-92. [PMID: 38357331 PMCID: PMC10862050 DOI: 10.34172/japid.2023.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/30/2023] [Indexed: 02/16/2024]
Abstract
Background Dabigatran belongs to the new generation of direct oral anticoagulants (DOACs). Its advantages are oral administration and no need for international normalized ratio (INR) monitoring. Although its use has increased, its potential side effects on bone healing and remodeling have not been fully investigated. The present study aimed to evaluate the possible effects of dabigatran on early bone healing. Methods Sixteen male Wistar rats were divided into two groups; in group A, 20-mg/kg dabigatran dose was administered orally daily for 15 days, while group B served as a control. Two circular bone defects (d=6 mm) were created on either side of the parietal bones. Two weeks after surgery and euthanasia of the animals, tissue samples (parietal bones that contained the defects) were harvested for histological and histomorphometric analysis. Statistical analysis was performed with a significance level of α=0.5. Results No statistically significant differences were found between the two groups regarding the regenerated bone (21.9% vs. 16.3%, P=0.172) or the percentage of bone bridging (63.3% vs. 53.5%, P=0.401). Conclusion Dabigatran did not affect bone regeneration, suggesting that it might be a safer drug compared to older anticoagulants known to lead to bone healing delay.
Collapse
Affiliation(s)
- Ioanna Kyriakaki
- Department of Dentoalveolar Surgery, Surgical Implantology and Roentgenology, Aristotle University, Thessaloniki, Greece
| | - Theodora Karanikola
- Private Practice, Clinical Instructor, Department of Oral Surgery, Implantology and Dental Radiology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodoros Lillis
- Department of Dentoalveolar Surgery, Surgical Implantology and Roentgenology, Aristotle University, Thessaloniki, Greece
| | - Eleana Kontonasaki
- Department of Prosthodontics, School of Dentistry, Aristotle University of Thessaloniki, Greece
| | - Nikolaos Dabarakis
- Department of Dentoalveolar Surgery, Surgical Implantology and Roentgenology, Aristotle University, Thessaloniki, Greece
| |
Collapse
|
5
|
Park JE, Ahn CH, Lee HJ, Sim DY, Park SY, Kim B, Shim BS, Lee DY, Kim SH. Antioxidant-Based Preventive Effect of Phytochemicals on Anticancer Drug-Induced Hepatotoxicity. Antioxid Redox Signal 2023; 38:1101-1121. [PMID: 36242510 DOI: 10.1089/ars.2022.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Drug-induced liver injury (DILI) or hepatotoxicity has been a hot issue to overcome on the safety and physiological function of the liver, since it is known to have biochemical, cellular, immunological, and molecular alterations in the liver mainly induced by alcohol, chemicals, drugs, heavy metals, and genetic factors. Recently efficient therapeutic and preventive strategies by some phytochemicals are of interest, targeting oxidative stress-mediated hepatotoxicity alone or in combination with anticancer drugs. Recent Advances: To assess DILI, the variety of in vitro and in vivo animal models has been developed mainly by using carbon tetrachloride, d-galactosamine, acetaminophen, and lipopolysaccharide. Also, the mechanisms on hepatotoxicity by several drugs and herbs have been explored in detail. Recent studies reveal that antioxidants including vitamins and some phytochemicals were reported to prevent against DILI. Critical Issues: Antioxidant therapy with some phytochemicals is noteworthy, since oxidative stress is critically involved in DILI via production of chemically reactive oxygen species or metabolites, impairment of mitochondrial respiratory chain, and induction of redox cycling. Future Directions: For efficient antioxidant therapy, DILI susceptibility, Human Leukocyte Antigen genetic factors, biomarkers, and pathogenesis implicated in hepatotoxicity should be further explored in association with oxidative stress-mediated signaling, while more randomized preclinical and clinical trials are required with optimal safe doses of drugs and/or phytochemicals alone or in combination for efficient clinical practice along with the development of advanced DILI diagnostic tools.
Collapse
Affiliation(s)
- Ji Eon Park
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyo-Jung Lee
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su Yeon Park
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bum Sang Shim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration (RDA), Eumseong, Republic of Korea
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Balon M, Tessier S, Damase-Michel C, Cottin J, Lambert A, Thompson MA, Benevent J, Lacroix I. Adverse drug reactions in pregnant women: Do they differ from those in non-pregnant women of childbearing age? Therapie 2023; 78:165-173. [PMID: 36517304 DOI: 10.1016/j.therap.2022.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022]
Abstract
Pharmacoepidemiological research in pregnant women has focused on adverse drug reactions for the course of pregnancy or for the unborn child, but little is known on the risks for the mother. We reported the results of a study that compared adverse drug reactions in pregnant women with non-pregnant women of childbearing age, and investigated whether which types of adverse reactions were more often reported in pregnant women and which drugs were more often involved. This study was carried out in the French pharmacovigilance database (BNPV). We compared adverse drug reactions reported between 1 January 2010 and 31 December 2019 in pregnant women with those reported in of non-pregnant women of childbearing age. We cross-matched each pregnant woman with three non-pregnant women of childbearing age according to geographic area, age and year the adverse reaction was reported. Data analysis revealed that serious adverse reactions were more frequently reported in pregnant women, including anaphylactic reactions. Other adverse reactions including tachycardia, hypotension and hepatic injury were also more frequent in pregnant women than in non-pregnant women of the same age. This could be explained by physiological changes in pregnancy that lead to greater sensitivity to certain adverse reactions. Some drugs, such as phloroglucinol, metoclopramide, iron, atosiban and nifedipine, were more frequently involved in adverse reactions in pregnant women. These drugs are specifically used during pregnancy, which may explain why they are over-represented in adverse reactions. This is the first comparative descriptive study on drug adverse reactions in pregnant women. Specific epidemiological and pharmacokinetic studies are necessary to confirm these results and better understand the differences observed to improve the monitoring of pregnant women exposed to certain drugs.
Collapse
Affiliation(s)
- Maylis Balon
- Unité "Médicaments, grossesse et allaitement", service de pharmacologie médicale et clinique, centre régional de pharmacovigilance, de pharmacoépidémiologie et d'informations sur le médicament (CRPV), CHU de Toulouse, faculté de médecine, Inserm 1295 CERPOP, 31000 Toulouse, France
| | - Samuel Tessier
- Unité "Médicaments, grossesse et allaitement", service de pharmacologie médicale et clinique, centre régional de pharmacovigilance, de pharmacoépidémiologie et d'informations sur le médicament (CRPV), CHU de Toulouse, faculté de médecine, Inserm 1295 CERPOP, 31000 Toulouse, France
| | - Christine Damase-Michel
- Unité "Médicaments, grossesse et allaitement", service de pharmacologie médicale et clinique, centre régional de pharmacovigilance, de pharmacoépidémiologie et d'informations sur le médicament (CRPV), CHU de Toulouse, faculté de médecine, Inserm 1295 CERPOP, 31000 Toulouse, France
| | - Judith Cottin
- Centre régional de pharmacovigilance, hospices civils de Lyon, 69424 Lyon, France
| | - Aude Lambert
- Centre régional de pharmacovigilance, hôpital civil, 67091 Strasbourg, France
| | - Marie-Andrée Thompson
- Centre régional de pharmacovigilance, service de pharmacologie médicale et toxicologie, CHU de Montpellier, 34295 Montpellier, France
| | - Justine Benevent
- Unité "Médicaments, grossesse et allaitement", service de pharmacologie médicale et clinique, centre régional de pharmacovigilance, de pharmacoépidémiologie et d'informations sur le médicament (CRPV), CHU de Toulouse, faculté de médecine, Inserm 1295 CERPOP, 31000 Toulouse, France
| | - Isabelle Lacroix
- Unité "Médicaments, grossesse et allaitement", service de pharmacologie médicale et clinique, centre régional de pharmacovigilance, de pharmacoépidémiologie et d'informations sur le médicament (CRPV), CHU de Toulouse, faculté de médecine, Inserm 1295 CERPOP, 31000 Toulouse, France.
| |
Collapse
|
7
|
Maliepaard M, Faber YS, van Bussel MTJ. Reported hepatotoxicity and hepatotoxicity guidance in the product information of protein kinase inhibitors in oncology registered at the European Medicines Agency. Pharmacol Res Perspect 2023; 11:e01067. [PMID: 36846954 PMCID: PMC9969339 DOI: 10.1002/prp2.1067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023] Open
Abstract
Protein kinase inhibitors (PKIs) used in oncology can induce severe and even fatal hepatotoxicity. Several PKIs are registered within a certain class to target a specific kinase. No systematic comparison of the reported hepatotoxicity and clinical guidance for monitoring and management of hepatotoxic events between the various PKI summaries of product characteristics (SmPC) is yet available. A systematic analysis of data on 21 hepatotoxicity parameters obtained from the SmPCs and European public assessment reports (EPARs) of European Medicines Agency-approved antineoplastic PKIs (n = 55) has been conducted. The median reported incidence (range) of all grades of aspartate aminotransferase (AST) elevations was 16.9% (2.0%-86.4%) for PKI monotherapy, with 2.1% (0.0%-10.3%) being grade 3/4 and for all grades alanine aminotransferase (ALT) elevations 17.6% (2.0%-85.5%), with 3.0% (0.0%-25.0%) being grade 3/4. Fatalities due to hepatotoxicity were reported for 22 out of 47 PKIs (monotherapy) and for 5 out of 8 PKIs (combination therapy). A maximum grade of grade 4 and grade 3 hepatotoxicity was reported for 45% (n = 25) and 6% (n = 3), respectively. Liver parameter monitoring recommendations were present in 47 of the 55 SmPCs. Dose reductions were recommended for 18 PKIs. Discontinuation was recommended for patients meeting Hy's law criteria (16 out of 55 SmPCs). Severe hepatotoxic events are reported in approximately 50% of the analyzed SmPCs and EPARs. Differences in the degree of hepatotoxicity are apparent. Although liver parameter monitoring recommendations are present in the vast majority of the analyzed PKI SmPCs, the clinical guidance for hepatotoxicity was not standardized.
Collapse
Affiliation(s)
- Marc Maliepaard
- Dutch Medicines Evaluation Board (CBG‐MEB)College ter Beoordeling van GeneesmiddelenUtrechtThe Netherlands,Department of Pharmacology and ToxicologyRadboud University Medical CentreNijmegenThe Netherlands
| | - Yoran S. Faber
- Dutch Medicines Evaluation Board (CBG‐MEB)College ter Beoordeling van GeneesmiddelenUtrechtThe Netherlands
| | - Mark T. J. van Bussel
- Dutch Medicines Evaluation Board (CBG‐MEB)College ter Beoordeling van GeneesmiddelenUtrechtThe Netherlands
| |
Collapse
|
8
|
Qin X, Xie C, Hakenjos JM, MacKenzie KR, Boyd SR, Barzi M, Bissig KD, Young DW, Li F. The roles of Cyp1a2 and Cyp2d in pharmacokinetic profiles of serotonin and norepinephrine reuptake inhibitor duloxetine and its metabolites in mice. Eur J Pharm Sci 2023; 181:106358. [PMID: 36513193 PMCID: PMC10395004 DOI: 10.1016/j.ejps.2022.106358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/03/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Duloxetine (DLX) is widely used to treat major depressive disorder. Little is known about the mechanistic basis for DLX-related adverse effects (e.g., liver injury). Human CYP1A2 and CYP2D6 mainly contributes to DLX metabolism, which was proposed to be involved in its adverse effects. Here, we investigated the roles of Cyp1a2 and Cyp2d on DLX pharmacokinetic profile and tissue distribution using a Cyp1a2 knockout (Cyp1a2-KO) mouse model together with a Cyp2d inhibitor (propranolol). Cyp1a2-KO has the few effects on the systematic exposure (area under the plasma concentration-time curve, AUC) and tissue disposition of DLX and its primary metabolites. Propranolol dramatically increased the AUCs of DLX by 3 folds and 1.5 folds in WT and Cyp1a2-KO mice, respectively. Meanwhile, Cyp2d inhibitor decreased the AUC of Cyp2d-involved DLX metabolites (e.g., M16). Mouse tissue distribution revealed that DLX and its major metabolites were the most abundant in kidney, followed by liver and lung with/without Cyp2d inhibitor. Cyp2d inhibitor significantly increased DLX levels in tissues (e.g., liver) in WT and KO mice and decreases the levels of M3, M15, M16 and M17, while it increased the levels of M4, M28 and M29 in tissues. Our findings indicated that Cyp2d play a fundamental role on DLX pharmacokinetic profile and tissue distribution in mice. Clinical studies suggested that CYP1A2 has more effects on DLX systemic exposure than CYP2D6. Further studies in liver humanized mice or clinical studies concerning CYP2D6 inhibitors-DLX interaction study could clarify the roles of CYP2D6 on DLX pharmacokinetics and toxicity in human.
Collapse
Affiliation(s)
- Xuan Qin
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - John M Hakenjos
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology & Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shelton R Boyd
- Department of Pharmacology & Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mercedes Barzi
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27708, USA
| | - Karl-Dimiter Bissig
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27708, USA
| | - Damian W Young
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology & Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology & Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Neuwirt E, Magnani G, Ćiković T, Wöhrle S, Fischer L, Kostina A, Flemming S, Fischenich NJ, Saller BS, Gorka O, Renner S, Agarinis C, Parker CN, Boettcher A, Farady CJ, Kesselring R, Berlin C, Backofen R, Rodriguez-Franco M, Kreutz C, Prinz M, Tholen M, Reinheckel T, Ott T, Groß CJ, Jost PJ, Groß O. Tyrosine kinase inhibitors can activate the NLRP3 inflammasome in myeloid cells through lysosomal damage and cell lysis. Sci Signal 2023; 16:eabh1083. [PMID: 36649377 DOI: 10.1126/scisignal.abh1083] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammasomes are intracellular protein complexes that promote an inflammatory host defense in response to pathogens and damaged or neoplastic tissues and are implicated in inflammatory disorders and therapeutic-induced toxicity. We investigated the mechanisms of activation for inflammasomes nucleated by NOD-like receptor (NLR) protiens. A screen of a small-molecule library revealed that several tyrosine kinase inhibitors (TKIs)-including those that are clinically approved (such as imatinib and crizotinib) or are in clinical trials (such as masitinib)-activated the NLRP3 inflammasome. Furthermore, imatinib and masitinib caused lysosomal swelling and damage independently of their kinase target, leading to cathepsin-mediated destabilization of myeloid cell membranes and, ultimately, cell lysis that was accompanied by potassium (K+) efflux, which activated NLRP3. This effect was specific to primary myeloid cells (such as peripheral blood mononuclear cells and mouse bone marrow-derived dendritic cells) and did not occur in other primary cell types or various cell lines. TKI-induced lytic cell death and NLRP3 activation, but not lysosomal damage, were prevented by stabilizing cell membranes. Our findings reveal a potential immunological off-target of some TKIs that may contribute to their clinical efficacy or to their adverse effects.
Collapse
Affiliation(s)
- Emilia Neuwirt
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Giovanni Magnani
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Tamara Ćiković
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.,Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, 81675 Munich, Germany
| | - Svenja Wöhrle
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Larissa Fischer
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anna Kostina
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Stephan Flemming
- Bioinformatics Group, Faculty of Engineering, University of Freiburg, 79110 Freiburg, Germany
| | - Nora J Fischenich
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Benedikt S Saller
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Steffen Renner
- Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Claudia Agarinis
- Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | | | - Andreas Boettcher
- Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | | | - Rebecca Kesselring
- Department for General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany
| | - Christopher Berlin
- Department for General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany
| | - Rolf Backofen
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Bioinformatics Group, Faculty of Engineering, University of Freiburg, 79110 Freiburg, Germany
| | | | - Clemens Kreutz
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Institute of Medical Biometry and Statistics (IMBI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Martina Tholen
- Institute for Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Reinheckel
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany.,Institute for Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Ott
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christina J Groß
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Philipp J Jost
- Division of Clinical Oncology, Department of Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Olaf Groß
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
10
|
Rakhshan A, Rahmati Kamel B, Saffaei A, Tavakoli-Ardakani M. Hepatotoxicity Induced by Azole Antifungal Agents: A Review Study. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2023; 22:e130336. [PMID: 38116543 PMCID: PMC10728840 DOI: 10.5812/ijpr-130336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/31/2023] [Accepted: 02/21/2023] [Indexed: 12/21/2023]
Abstract
Context Fungal infections are very common, and several medications are used to treat them. Azoles are prescribed widely to treat fungal infections. In addition to therapeutic effects, any drug can be accompanied by side effects in patients. One of the most important complications in this regard is liver injury. Therefore, hepatotoxicity induced by azole antifungal drugs were reviewed in this study. Evidence Acquisition English scientific papers were evaluated to review the effects of hepatotoxicity by azole antifungal agents, and the related studies' results were summarized using a table. The systematic search was implemented on electronic databases, including PubMed, Google Scholar, and Science Direct. Original articles and review articles that were published before April 1, 2022, were included in the study. Those articles without available full text or non-English articles were excluded. Also, articles that reported pediatric data were excluded. Results Most studies have reported the effects of hepatotoxicity by azole antifungal agents, and their mechanisms have been described. Conclusions Clinical evaluations regarding the hepatotoxicity of antifungal agents provided in the literature were reviewed. Therefore, it is recommended to prescribe these drugs with caution in high-risk patients suffering from liver diseases, and patients should be monitored for hepatotoxicity. However, more research is needed to evaluate the hepatotoxicity of azole antifungal agents and select appropriate drugs according to cost-effectiveness and the side effects' profiles, relying on lower incidence of this liver complication.
Collapse
Affiliation(s)
- Amin Rakhshan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bardia Rahmati Kamel
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Saffaei
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli-Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Soal V, Tiongko JL, Sedky K. Duloxetine and Hepatic Injury: A Case Presentation. Psychiatr Ann 2023. [DOI: 10.3928/00485713-20230105-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
12
|
Cao L, Zhao H, Qian M, Shao C, Zhang Y, Yang J. Construction of polysaccharide scaffold-based perfusion bioreactor supporting liver cell aggregates for drug screening. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2249-2269. [PMID: 35848470 DOI: 10.1080/09205063.2022.2102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Rebuilding a suitable microenvironment of liver cells is the key challenge to enhancing the expression of hepatic functions for drug screening in vitro. To improve the microenvironment by providing the specific adhesive ligands for hepatocytes in the three-dimensional dynamic culture, a perfusion bioreactor with a pectin/alginate blend porous scaffold was constructed in this study. The galactosyl component in the main chain of pectin was able to be specifically recognized by the asialoglycoprotein receptor on the surface of hepatocytes, and subsequently promoted the adhesion and aggregation of hepatocytes co-cultured with hepatic non-parenchymal cells. The bioreactor was optimized for 4 h of dynamic inoculation followed by perfusion at a flow rate of 2 mL/min, which provided adequate oxygen supply and good mass transfer to the liver cells. During dynamic cultured in the bioreactor for 14 days, more multicellular aggregates were formed and were evenly distributed in the pectin/alginate blend scaffolds. The expressions of intercellular interaction and hepatic functions of the hepatocytes in aggregates were significantly enhanced in the three-dimensional dynamic group. Furthermore, the bioreactor not only markedly upregulated the cell polarity markers expression of hepatocytes but also enhanced their metabolic capacity to acetaminophen, isoniazid, and tolbutamide, which exhibited a significant concentration-dependent manner. Therefore, the pectin/alginate blend scaffold-based perfusion bioreactor appeared to be a promising candidate in the field of drug development and liver regeneration research.
Collapse
Affiliation(s)
- Lei Cao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China.,Biological Sample Resource Sharing Center, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Huicun Zhao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Mengyuan Qian
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Chuxiao Shao
- Department of Hepatopancreatobiliary Surgery, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Yan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| |
Collapse
|
13
|
Inhibition of platelet-derived growth factor pathway suppresses tubulointerstitial injury in renal congestion. J Hypertens 2022; 40:1935-1949. [PMID: 35983805 PMCID: PMC9451920 DOI: 10.1097/hjh.0000000000003191] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Increased central venous pressure in congestive heart failure is responsible for renal dysfunction, which is mediated by renal venous congestion. Pericyte detachment from capillaries after renal congestion might trigger renal fibrogenesis via pericyte-myofibroblast transition (PMT). Platelet-derived growth factor receptors (PDGFRs), which are PMT indicators, were upregulated in our recently established renal congestion model. This study was designed to determine whether inhibition of the PDGFR pathway could suppress tubulointerstitial injury after renal congestion. METHODS The inferior vena cava between the renal veins was ligated in male Sprague-Dawley rats, inducing congestion only in the left kidney. Imatinib mesylate or vehicle were injected intraperitoneally daily from 1 day before the operation. Three days after the surgery, the effect of imatinib was assessed by physiological, morphological and molecular methods. The inhibition of PDGFRs against transforming growth factor-β1 (TGFB1)-induced fibrosis was also tested in human pericyte cell culture. RESULTS Increased kidney weight and renal fibrosis were observed in the congested kidneys. Upstream inferior vena cava (IVC) pressure immediately increased to around 20 mmHg after IVC ligation in both the imatinib and saline groups. Although vasa recta dilatation and pericyte detachment under renal congestion were maintained, imatinib ameliorated the increased kidney weight and suppressed renal fibrosis around the vasa recta. TGFB1-induced elevation of fibrosis markers in human pericytes was suppressed by PDGFR inhibitors at the transcriptional level. CONCLUSION The activation of the PDGFR pathway after renal congestion was responsible for renal congestion-induced fibrosis. This mechanism could be a candidate therapeutic target for renoprotection against renal congestion-induced tubulointerstitial injury.
Collapse
|
14
|
Xu J, Pan D, Liao W, Jia Z, Pan M, Weng J, Han X, Li S, Li Y, Liang K, Zhou S, Peng Q, Gao Y. Application of 3D Hepatic Plate-Like Liver Model for Statin-Induced Hepatotoxicity Evaluation. Front Bioeng Biotechnol 2022; 10:826093. [PMID: 35372314 PMCID: PMC8968918 DOI: 10.3389/fbioe.2022.826093] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Drug-induced liver injury is one of the main reasons of withdrawals of drugs in postmarketing stages. However, an experimental model(s) which can accurately recapitulates liver functions and reflects the level of drug hepatotoxicity is lack. In this study, we assessed drug hepatotoxicity using a novel three-dimensional hepatic plate-like hydrogel fiber (3D-P) co-culture system. Methods: During the 28-days culture period, the liver-specific functions, hepatocyte polarity, sensitivity of drug-induced toxicity of 3D-P co-culture system were evaluated with 2D co-culture, collagen sandwich co-culture, 3D hybrid hydrogel fiber co-culture and human primary hepatocytes as controls. High-content imaging and analysis (HCA) methods were used to explore the hepatotoxicity mechanism of five statins. Results: The 3D-P co-culture system showed enhancing liver-specific functions, cytochrome P450 enzymes (CYPs) metabolic activity and bile excretion, which were considered to result from improved hepatocyte polarity. Three of the statins may cause acute or chronic hepatotoxicity by via different mechanisms, such as cholestatic liver injury. Conclusion: Our 3D-P co-culture system is characterized by its biomimetic hepatic plate-like structure, long-term stable liver specificity, and prominent bile secretion function, making it applicable for acute/chronic drug hepatotoxicity assessments.
Collapse
Affiliation(s)
- Jiecheng Xu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Daogang Pan
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wei Liao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Zhidong Jia
- Guangzhou Overseas Chinese Hospital, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mingxin Pan
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xu Han
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Shao Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Kangyan Liang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qing Peng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Qing Peng, ; Yi Gao,
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
- *Correspondence: Qing Peng, ; Yi Gao,
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Drug-induced bile duct injury can be caused by a long list of agents. In most cases, damage is because of T-cell-mediated idiosyncratic reactions. Recently, a number of new agents, including not only drugs but also herbal supplements, have been incriminated and new mechanisms of bile duct injury have emerged. This review will focus on these new data. RECENT FINDINGS New members of drug families already known to be responsible for bile duct injury have been incriminated. New players have been identified, such as herbal supplements, like kratom, and recreational drugs, such as ketamine used outside the medical setting. Anticytokine monoclonal antibodies are rarely involved. In contrast, antineoplastic treatments are of growing concern, especially immune checkpoint inhibitors, which induce immune-related adverse effects because of the excessive stimulation of the immune system and its lack of regulation. SUMMARY Two patterns of bile duct injury are recognized. Drug-induced small-duct cholangiopathies target the smaller bile ducts; acute injuries eventually progress to chronic disease in the form of the vanishing bile duct syndrome. Drug-induced sclerosing cholangitis target large bile ducts, with a protracted chronic course; the onset of symptoms may be delayed after drug discontinuation; potentially severe, life-threatening complications can occur.
Collapse
|
16
|
Huang FR, Fang WT, Cheng ZP, Shen Y, Wang DJ, Wang YQ, Sun LN. Imatinib-induced hepatotoxicity via oxidative stress and activation of NLRP3 inflammasome: an in vitro and in vivo study. Arch Toxicol 2022; 96:1075-1087. [PMID: 35190838 DOI: 10.1007/s00204-022-03245-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
Imatinib (IM), a milestone drug used in the field of molecular targeted therapy, has been reported to cause serious adverse liver effects, including liver failure and even death. Immune-mediated injury and mitochondrial dysfunction are involved in drug-induced liver injury. However, the mechanism of IM-induced hepatotoxicity remains unclear and warrants further study. In our study, Sprague Dawley rats were administered IM by gavage with 50 mg/kg body weight (BW) once daily for 10 days. Drug-induced liver injury accompanied by inflammatory infiltration was observed in rats following IM exposure, and the expression of NOD-like receptor protein 3 (NLRP3) inflammasome-related proteins was significantly increased compared with that of the control. HepG2 cells were exposed to 0-100 μM IM for 24 h. The results showed that IM decreased cell viability in a dose-dependent manner. Moreover, IM induced a state of obvious oxidative stress and activation of nuclear factor kappa B (NF-κB) in cells, which resulted in the activation of NLRP3 inflammasomes, including caspase 1 cleavage and IL-1β release. These results were significantly reduced after the use of the antioxidants N-acetyl-l-cysteine or the NF-κB inhibitor pyrrolidine di-thio-carbamate. Furthermore, NLRP3 knockdown significantly reduced the release of inflammatory cytokines and improved cell viability. In summary, our data demonstrated that oxidative stress and NLRP3 inflammasome activation are involved in the process of IM-induced hepatotoxicity. The results of this study provide a reference for the prevention and treatment of IM-induced hepatotoxicity.
Collapse
Affiliation(s)
- Feng-Ru Huang
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China.,School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Wen-Tong Fang
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China
| | - Zi-Ping Cheng
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China
| | - Ye Shen
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China
| | - Dun-Jian Wang
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China
| | - Yong-Qing Wang
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China. .,School of Pharmacy, Nanjing Medical University, Nanjing, China.
| | - Lu-Ning Sun
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210009, China. .,School of Pharmacy, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
17
|
Saran C, Sundqvist L, Ho H, Niskanen J, Honkakoski P, Brouwer KLR. Novel Bile Acid-Dependent Mechanisms of Hepatotoxicity Associated with Tyrosine Kinase Inhibitors. J Pharmacol Exp Ther 2022; 380:114-125. [PMID: 34794962 PMCID: PMC9109172 DOI: 10.1124/jpet.121.000828] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023] Open
Abstract
Drug-induced liver injury (DILI) is the leading cause of acute liver failure and a major concern in drug development. Altered bile acid homeostasis via inhibition of the bile salt export pump (BSEP) is one mechanism of DILI. Dasatinib, pazopanib, and sorafenib are tyrosine kinase inhibitors (TKIs) that competitively inhibit BSEP and increase serum biomarkers for hepatotoxicity in ∼25-50% of patients. However, the mechanism(s) of hepatotoxicity beyond competitive inhibition of BSEP are poorly understood. This study examined mechanisms of TKI-mediated hepatotoxicity associated with altered bile acid homeostasis. Dasatinib, pazopanib, and sorafenib showed bile acid-dependent toxicity at clinically relevant concentrations, based on the C-DILI assay using sandwich-cultured human hepatocytes (SCHH). Among several bile acid-relevant genes, cytochrome P450 (CYP) 7A1 mRNA was specifically upregulated by 6.2- to 7.8-fold (dasatinib) and 5.7- to 9.3-fold (pazopanib), compared with control, within 8 hours. This was consistent with increased total bile acid concentrations in culture medium up to 2.3-fold, and in SCHH up to 1.4-fold, compared with control, within 24 hours. Additionally, protein abundance of sodium taurocholate co-transporting polypeptide (NTCP) was increased up to 2.0-fold by these three TKIs. The increase in NTCP protein abundance correlated with increased function; dasatinib and pazopanib increased hepatocyte uptake clearance (CLuptake) of taurocholic acid, a probe bile acid substrate, up to 1.4-fold. In conclusion, upregulation of CYP7A1 and NTCP in SCHH constitute novel mechanisms of TKI-associated hepatotoxicity. SIGNIFICANCE STATEMENT: Understanding the mechanisms of hepatotoxicity associated with tyrosine kinase inhibitors (TKIs) is fundamental to development of effective and safe intervention therapies for various cancers. Data generated in sandwich-cultured human hepatocytes, an in vitro model of drug-induced hepatotoxicity, revealed that TKIs upregulate bile acid synthesis and alter bile acid uptake and excretion. These findings provide novel insights into additional mechanisms of bile acid-mediated drug-induced liver injury, an adverse effect that limits the use and effectiveness of TKI treatment in some cancer patients.
Collapse
Affiliation(s)
- Chitra Saran
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S., L.S., H.H., P.H., K.L.R.B.); Department of Pharmacy, Uppsala University, Uppsala, Sweden (L.S.); and School of Pharmacy, University of Eastern Finland, Kuopio, Finland (J.N., P.H.)
| | - Louise Sundqvist
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S., L.S., H.H., P.H., K.L.R.B.); Department of Pharmacy, Uppsala University, Uppsala, Sweden (L.S.); and School of Pharmacy, University of Eastern Finland, Kuopio, Finland (J.N., P.H.)
| | - Henry Ho
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S., L.S., H.H., P.H., K.L.R.B.); Department of Pharmacy, Uppsala University, Uppsala, Sweden (L.S.); and School of Pharmacy, University of Eastern Finland, Kuopio, Finland (J.N., P.H.)
| | - Jonna Niskanen
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S., L.S., H.H., P.H., K.L.R.B.); Department of Pharmacy, Uppsala University, Uppsala, Sweden (L.S.); and School of Pharmacy, University of Eastern Finland, Kuopio, Finland (J.N., P.H.)
| | - Paavo Honkakoski
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S., L.S., H.H., P.H., K.L.R.B.); Department of Pharmacy, Uppsala University, Uppsala, Sweden (L.S.); and School of Pharmacy, University of Eastern Finland, Kuopio, Finland (J.N., P.H.)
| | - Kim L R Brouwer
- Department of Pharmacology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.S., L.S., H.H., P.H., K.L.R.B.); Department of Pharmacy, Uppsala University, Uppsala, Sweden (L.S.); and School of Pharmacy, University of Eastern Finland, Kuopio, Finland (J.N., P.H.)
| |
Collapse
|
18
|
|
19
|
Fang L, Wang S, Cao L, Yao K. Early intervention of acute liver injury related to venlafaxine: A case report. Medicine (Baltimore) 2021; 100:e28140. [PMID: 34889278 PMCID: PMC8663904 DOI: 10.1097/md.0000000000028140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/17/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Drug-induced liver injury (DILI) is the leading cause of acute liver injury (ALI), market withdrawal of a drug, and rejection of applications for marketing licenses. The incidence of DILI is very low, with a value between 1 and 19 per 100,000 patient years. All antidepressants may induce DILI even at low therapeutic doses. In this report, we present a case of ALI after venlafaxine administration. PATIENT CONCERNS A 27-year-old Chinese Han woman was admitted for depression. Several serum liver function indices in this patient were abnormal after antidepressant treatment. The Roussel Uclaf Causality Assessment Method (RUCAM) causality assessment score was 8, and the R value was 31.18. DIAGNOSES The patient was diagnosed with hepatocellular ALI, which was derived from venlafaxine-related adverse events. INTERVENTIONS First, all medications were stopped to block the progression of DILI. Then, a hepatoprotective strategy and proper psychological treatment were performed to recover the impaired hepatic function. OUTCOMES Liver function was fully recovered as indicated by liver function indices and ultrasound imaging. LESSONS The possibility of DILI should not be overlooked during the long-term use of antipsychotic drugs. In response, regular liver function monitoring should be performed in a timely manner to avoid missing diagnoses and delayed treatment. Furthermore, the necessary medical treatment needs to be conducted after the occurrence of ALI.
Collapse
Affiliation(s)
- Lin Fang
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| | - Shushan Wang
- Department of Pharmacy, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| | - Leiming Cao
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| | - Kun Yao
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| |
Collapse
|
20
|
Wang X, Lei J, Li Z, Yan L. Potential Effects of Coronaviruses on the Liver: An Update. Front Med (Lausanne) 2021; 8:651658. [PMID: 34646834 PMCID: PMC8502894 DOI: 10.3389/fmed.2021.651658] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
The coronaviruses that cause notable diseases, namely, severe acute respiratory syndrome (SARS), middle east respiratory syndrome (MERS) and coronavirus disease 2019 (COVID-19), exhibit remarkable similarities in genomic components and pathogenetic mechanisms. Although coronaviruses have widely been studied as respiratory tract pathogens, their effects on the hepatobiliary system have seldom been reported. Overall, the manifestations of liver injury caused by coronaviruses typically involve decreased albumin and elevated aminotransferase and bilirubin levels. Several pathophysiological hypotheses have been proposed, including direct damage, immune-mediated injury, ischemia and hypoxia, thrombosis and drug hepatotoxicity. The interaction between pre-existing liver disease and coronavirus infection has been illustrated, whereby coronaviruses influence the occurrence, severity, prognosis and treatment of liver diseases. Drugs and vaccines used for treating and preventing coronavirus infection also have hepatotoxicity. Currently, the establishment of optimized therapy for coronavirus infection and liver disease comorbidity is of significance, warranting further safety tests, animal trials and clinical trials.
Collapse
Affiliation(s)
- Xinyi Wang
- Thyroid and Parathyroid Surgery Center, West China Hospital of Sichuan University, Chengdu, China
- Liver Surgery Center, West China Hospital of Sichuan University, Chengdu, China
| | - Jianyong Lei
- Thyroid and Parathyroid Surgery Center, West China Hospital of Sichuan University, Chengdu, China
- Liver Surgery Center, West China Hospital of Sichuan University, Chengdu, China
| | - Zhihui Li
- Thyroid and Parathyroid Surgery Center, West China Hospital of Sichuan University, Chengdu, China
- Liver Surgery Center, West China Hospital of Sichuan University, Chengdu, China
| | - Lunan Yan
- Liver Surgery Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Ortiz GX, Lenhart G, Becker MW, Schwambach KH, Tovo CV, Blatt CR. Drug-induced liver injury and COVID-19: A review for clinical practice. World J Hepatol 2021; 13:1143-1153. [PMID: 34630881 PMCID: PMC8473488 DOI: 10.4254/wjh.v13.i9.1143] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/18/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) consists of a systemic disease that can present many complications. The infection presents broad clinical symptoms and a high rate of transmissibility. In addition to severe acute respiratory syndrome, the patients manifest complications beyond the respiratory system. The frequency of liver damage in COVID-19 patients ranges from 14.8% to 53% of patients. One should pay attention to drug-induced liver injury (DILI) in patients with COVID-19, especially considering the off-label use of drugs in prophylactic and therapeutic regimens applied on large scales. This review aims to present relevant information on the medication used so far in COVID-19 patients and its possible hepatotoxicity. We reviewed liver damage in patients with COVID-19 on PubMed and Virtual Health Library to investigate DILI cases. Four studies were selected, involving the medicines remdesivir, tocilizumab and a pharmacovigilance analysis study. The hepatotoxicity profile of drugs presented in the literature considers use in accordance to usual posology standards for treatment. However, drugs currently used in the management of COVID-19 follow different dosages and posology than those tested by the pharmaceutical industry. The deficiency of uniformity and standardization in the assessment of hepatotoxicity cases hinders the publication of information and the possibility of comparing information among healthcare professionals. It is suggested that severe liver injury in COVID-19 patients should be reported in pharmacovigilance institutions, and physicians should pay attention to any considerable abnormal liver test elevation as it can demonstrate unknown drug hepatotoxicity. Liver disorders in COVID-19 patients and the use of several concomitant off-label medications — with a potential risk of further damaging the liver - should at least be a warning sign for rapid identification and early intervention, thus preventing liver damage from contributing to severe impairment in patients.
Collapse
Affiliation(s)
- Gabriela Xavier Ortiz
- Graduate Program in Medicine Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| | - Gabriele Lenhart
- Multiprofessional Residency Integrated in Health, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| | - Matheus William Becker
- Graduate Program in Medicine Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| | - Karin Hepp Schwambach
- Graduate Program in Medicine Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| | - Cristiane Valle Tovo
- Internal Medicine Department, Graduate Program in Medicine-Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| | - Carine Raquel Blatt
- Pharmacoscience Department, Graduate Program in Medicine-Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| |
Collapse
|
22
|
Maggiore U, Palmisano A, Buti S, Claire Giudice G, Cattaneo D, Giuliani N, Fiaccadori E, Gandolfini I, Cravedi P. Chemotherapy, targeted therapy and immunotherapy: Which drugs can be safely used in the solid organ transplant recipients? Transpl Int 2021; 34:2442-2458. [PMID: 34555228 PMCID: PMC9298293 DOI: 10.1111/tri.14115] [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: 06/10/2021] [Revised: 08/04/2021] [Accepted: 09/03/2021] [Indexed: 11/29/2022]
Abstract
In solid organ transplant recipients, cancer is associated with worse prognosis than in the general population. Among the causes of increased cancer‐associated mortality, are the limitations in selecting the optimal anticancer regimen in solid organ transplant recipients, because of the associated risks of graft toxicity and rejection, drug‐to‐drug interactions, reduced kidney or liver function, and patient frailty and comorbid conditions. The advent of immunotherapy has generated further challenges, mainly because checkpoint inhibitors increase the risk of rejection, which may have life‐threatening consequences in recipients of life‐saving organs. In general, there are no safe or unsafe anticancer drugs. Rather, the optimal choice of the anticancer regimen results from a careful risk/benefit assessment, from the awareness of potential pharmacokinetic and pharmacodynamic drug‐to‐drug interactions, and of the risk of drug overexposure in patients with kidney or liver dysfunction. In this review, we summarize general principles that may help the oncologists and transplant physicians in the multidisciplinary management of recipients of solid organ transplantation with cancer who are candidates for chemotherapy, targeted therapy, or immunotherapy.
Collapse
Affiliation(s)
- Umberto Maggiore
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Nephrology Unit, University Hospital of Parma, Parma, Italy
| | | | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Dario Cattaneo
- Unit of Clinical Pharmacology, ASST Fatebenefratelli Sacco University Hospital, Milan, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Hematology Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Enrico Fiaccadori
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Nephrology Unit, University Hospital of Parma, Parma, Italy
| | - Ilaria Gandolfini
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Nephrology Unit, University Hospital of Parma, Parma, Italy
| | - Paolo Cravedi
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
23
|
García-Cortés M, Ortega-Alonso A, Andrade RJ. Safety of treating acute liver injury and failure. Expert Opin Drug Saf 2021; 21:191-203. [PMID: 34254839 DOI: 10.1080/14740338.2021.1955854] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Acute liver injury and progression to acute liver failure can be life-threatening conditions that require prompt careful clinical assessment and therapeutic management. AREAS COVERED The aim of this article is to review the safety and side effect profile of pharmacological therapies used in the treatment of acute liver injury with specific focus on hepatic toxicity. We performed an extensive literature search with the terms 'acute liver injury,' 'acute liver failure,' 'therapy,' 'safety,' 'adverse reactions' and 'drug induced liver injury.' A thorough discussion of the main drugs and devices used in patients with acute liver injury and acute liver failure, its safety profile and the management of complications associated to therapy of these conditions is presented. EXPERT OPINION Several pharmacological approaches are used in acute liver injury and acute liver failure in an empirical basis. Whilst steroids are frequently tried in serious drug-induced liver injury there is concern on a potential harmful effect of these agents because of the higher mortality in patients receiving the drug; hence, statistical approaches such as propensity score matching might help resolve this clinical dilemma. Likewise, properly designed clinical trials using old and new drugs for subjects with serious drug-induced liver injury are clearly needed.
Collapse
Affiliation(s)
- Miren García-Cortés
- Servicio De Aparato Digestivo, Instituto De Investigación Biomédica De Málaga-IBIMA. Hospital Universitario Virgen De La Victoria, Universidad De Málaga, Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas CIBERehd, Málaga, Spain
| | - Aida Ortega-Alonso
- Servicio De Aparato Digestivo, Instituto De Investigación Biomédica De Málaga-IBIMA. Hospital Universitario Virgen De La Victoria, Universidad De Málaga, Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas CIBERehd, Málaga, Spain
| | - Raúl J Andrade
- Servicio De Aparato Digestivo, Instituto De Investigación Biomédica De Málaga-IBIMA. Hospital Universitario Virgen De La Victoria, Universidad De Málaga, Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas CIBERehd, Málaga, Spain
| |
Collapse
|
24
|
Kirschner M, do Ó Hartmann N, Parmentier S, Hart C, Henze L, Bisping G, Griesshammer M, Langer F, Pabinger-Fasching I, Matzdorff A, Riess H, Koschmieder S. Primary Thromboprophylaxis in Patients with Malignancies: Daily Practice Recommendations by the Hemostasis Working Party of the German Society of Hematology and Medical Oncology (DGHO), the Society of Thrombosis and Hemostasis Research (GTH), and the Austrian Society of Hematology and Oncology (ÖGHO). Cancers (Basel) 2021; 13:2905. [PMID: 34200741 PMCID: PMC8230401 DOI: 10.3390/cancers13122905] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/20/2022] Open
Abstract
Patients with cancer, both hematologic and solid malignancies, are at increased risk for thrombosis and thromboembolism. In addition to general risk factors such as immobility and major surgery, shared by non-cancer patients, cancer patients are exposed to specific thrombotic risk factors. These include, among other factors, cancer-induced hypercoagulation, and chemotherapy-mediated endothelial dysfunction as well as tumor-cell-derived microparticles. After an episode of thrombosis in a cancer patient, secondary thromboprophylaxis to prevent recurrent thromboembolism has long been established and is typically continued as long as the cancer is active or actively treated. On the other hand, primary prophylaxis, even though firmly established in hospitalized cancer patients, has only recently been studied in ambulatory patients. This recent change is mostly due to the emergence of direct oral anticoagulants (DOACs). DOACs have a shorter half-life than vitamin K antagonists (VKA), and they overcome the need for parenteral application, the latter of which is associated with low-molecular-weight heparins (LMWH) and can be difficult for the patient to endure in the long term. Here, first, we discuss the clinical trials of primary thromboprophylaxis in the population of cancer patients in general, including the use of VKA, LMWH, and DOACs, and the potential drug interactions with pre-existing medications that need to be taken into account. Second, we focus on special situations in cancer patients where primary prophylactic anticoagulation should be considered, including myeloma, major surgery, indwelling catheters, or immobilization, concomitant diseases such as renal insufficiency, liver disease, or thrombophilia, as well as situations with a high bleeding risk, particularly thrombocytopenia, and specific drugs that may require primary thromboprophylaxis. We provide a novel algorithm intended to aid specialists but also family practitioners and nurses who care for cancer patients in the decision process of primary thromboprophylaxis in the individual patient.
Collapse
Affiliation(s)
- Martin Kirschner
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.K.); (N.d.Ó.H.)
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), 52074 Aachen, Germany
| | - Nicole do Ó Hartmann
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.K.); (N.d.Ó.H.)
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), 52074 Aachen, Germany
| | - Stefani Parmentier
- Oncology and Hematology, Tumor Center, St. Claraspital, 4058 Basel, Switzerland;
| | - Christina Hart
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Larissa Henze
- Department of Medicine, Clinic III—Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Guido Bisping
- Department of Medicine I, Mathias Spital Rheine, 48431 Rheine, Germany;
| | - Martin Griesshammer
- University Clinic for Hematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, University of Bochum, 32429 Minden, Germany;
| | - Florian Langer
- II.Medical Clinic and Polyclinic, Center for Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Ingrid Pabinger-Fasching
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| | - Axel Matzdorff
- Department of Internal Medicine II, Asklepios Clinic Uckermark, 16303 Schwedt, Germany;
| | - Hanno Riess
- Medical Department, Division of Oncology and Hematology, Campus Charité Mitte, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.K.); (N.d.Ó.H.)
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), 52074 Aachen, Germany
| |
Collapse
|
25
|
Zhang Y, Cai Y, Zhang SR, Li CY, Jiang LL, Wei P, He MF. Mechanism of hepatotoxicity of first-line tyrosine kinase inhibitors: Gefitinib and afatinib. Toxicol Lett 2021; 343:1-10. [PMID: 33571620 DOI: 10.1016/j.toxlet.2021.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/19/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
AIMS Both gefitinib and afatinib are epidermal growth factor tyrosine kinase inhibitors (EGFR-TKI) in the treatment of non-small cell lung cancer (NSCLC). It has been reported that gefitinib and afatinib could cause hepatotoxicity during the clinic treatment, therefore it is critical to investigate their hepatotoxicity systematically. In this study, zebrafish (Danio rerio) were used as model animals to compare the hepatotoxicity and their toxic mechanism. MAIN METHODS The zebrafish transgenic line [Tg (fabp10a: dsRed; ela3l:EGFP) was used in this study. After larvae developed at 3 days post fertilization (dpf), they were put into different concentrations of gefitinib and afatinib. At 6 dpf, the viability, liver area, fluorescence intensity, histopathology, apoptosis, transaminase reflecting liver function, the absorption of yolk sac, and the expression of relative genes were observed and analyzed respectively. KEY FINDINGS Both gefitinib and afatinib could induce the larvae hepatotoxicity dose-dependently. Based on the liver morphology, histopathology, apoptosis and function assessments, gefitinib showed higher toxicity, causing more serious liver damage. Both gefitinib and afatinib caused abnormal expressions of genes related to endoplasmic reticulum stress (ERS) pathway and apoptosis. For example, jnk, perk, bip, chop, ire1, bid, caspase3 and caspase9 were up-regulated, while xbp1s, grp78, bcl-2/bax, and caspase8 were down-regulated. The hepatotoxicity difference of gefitinib and afatinib might be due to the different expression level of related genes.
Collapse
Affiliation(s)
- Yao Zhang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Yang Cai
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Shi-Ru Zhang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Chong-Yong Li
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Ling-Ling Jiang
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 211816, China
| | - Pin Wei
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Ming-Fang He
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China.
| |
Collapse
|
26
|
|
27
|
González‐Muñoz M, Monserrat Villatoro J, Marín‐Serrano E, Stewart S, Bardón Rivera B, Marín J, Martínez de Soto L, Seco Meseguer E, Ramírez E. A case report of a drug-induced liver injury (DILI) caused by multiple antidepressants with causality established by the updated Roussel Uclaf causality assessment method (RUCAM) and in vitro testing. Clin Case Rep 2020; 8:3105-3109. [PMID: 33363890 PMCID: PMC7752543 DOI: 10.1002/ccr3.3348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/17/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022] Open
Abstract
A 56-year-old female patient was hospitalized because of a lack of response and poor tolerance to multiple antidepressants, which included an episode of DILI. During hospitalization, the patient suffered another episode of DILI. Causality was assessed both by RUCAM and Lymphocyte Transformation Test, allowing to identify a safer medication.
Collapse
Affiliation(s)
- Miguel González‐Muñoz
- Immunology DepartmentLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazMadridEspaña
| | - Jaime Monserrat Villatoro
- Clinical Pharmacology DepartmentFacultad de MedicinaLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazUniversidad Autónoma de MadridMadridEspaña
| | - Eva Marín‐Serrano
- Gastroenterology and Hepatology DepartmentLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazMadridEspaña
| | - Stefan Stewart
- Clinical Pharmacology DepartmentFacultad de MedicinaLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazUniversidad Autónoma de MadridMadridEspaña
| | - Belén Bardón Rivera
- Psiquiatry DepartmentLa Paz‐Cantoblanco‐Carlos III University Hospital, IdiPazMadridEspaña
| | - Jesús Marín
- Psiquiatry DepartmentLa Paz‐Cantoblanco‐Carlos III University Hospital, IdiPazMadridEspaña
| | - Lucía Martínez de Soto
- Clinical Pharmacology DepartmentFacultad de MedicinaLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazUniversidad Autónoma de MadridMadridEspaña
| | - Enrique Seco Meseguer
- Clinical Pharmacology DepartmentFacultad de MedicinaLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazUniversidad Autónoma de MadridMadridEspaña
| | - Elena Ramírez
- Clinical Pharmacology DepartmentFacultad de MedicinaLa Paz‐Cantoblanco‐Carlos III University HospitalIdiPazUniversidad Autónoma de MadridMadridEspaña
| |
Collapse
|
28
|
Potmešil P, Szotkowská R. Drug-induced liver injury after switching from tamoxifen to anastrozole in a patient with a history of breast cancer being treated for hypertension and diabetes. Ther Adv Chronic Dis 2020; 11:2040622320964152. [PMID: 33240477 PMCID: PMC7675855 DOI: 10.1177/2040622320964152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Anastrozole is a selective non-steroidal aromatase inhibitor that blocks the
conversion of androgens to estrogens in peripheral tissues. It is used as
adjuvant therapy for early-stage hormone-sensitive breast cancer in
postmenopausal women. Significant side effects of anastrozole include
osteoporosis and increased levels of cholesterol. To date, seven case reports on
anastrozole hepatotoxicity have been published. We report the case of an
81-year-old woman with a history of breast cancer, arterial hypertension, type 2
diabetes mellitus, hyperlipidemia, and chronic renal insufficiency. Four days
after switching hormone therapy from tamoxifen to anastrozole, icterus developed
along with a significant increase in liver enzymes (measured in the blood). The
patient was admitted to hospital, where a differential diagnosis of jaundice was
made and anastrozole was withdrawn. Subsequently, hepatic functions quickly
normalized. The observed liver injury was attributed to anastrozole since other
possible causes of jaundice were excluded. However, concomitant pharmacotherapy
could have contributed to the development of jaundice and hepatotoxicity, after
switching from tamoxifen to anastrozole since several the patient’s medications
were capable of inhibiting hepatobiliary transport of bilirubin, bile acids, and
metabolized drugs through inhibition of ATP-binding cassette proteins.
Telmisartan, tamoxifen, and metformin all block bile salt efflux pumps. The
efflux function of multidrug resistance protein 2 is known to be reduced by
telmisartan and tamoxifen and breast cancer resistance protein is known to be
inhibited by telmisartan and amlodipine. Moreover, the activity of
P-glycoprotein transporters are known to be decreased by telmisartan,
amlodipine, gliquidone, as well as the previously administered tamoxifen.
Finally, the role of genetic polymorphisms of cytochrome P450 enzymes and/or
drug transporters cannot be ruled out since the patient was not tested for
polymorphisms.
Collapse
Affiliation(s)
- Petr Potmešil
- Third Faculty of Medicine, Department of Pharmacology, Charles University, Prague, Czech Republic and Faculty of Medicine, Department of Pharmacology and Toxicology, Charles University, Pilsen, Czech Republic
| | - Radka Szotkowská
- 2nd Department of Internal Medicine, University Hospital Královské Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
29
|
Houron C, Danielou M, Mir O, Fromenty B, Perlemuter G, Voican CS. Multikinase inhibitor-induced liver injury in patients with cancer: A review for clinicians. Crit Rev Oncol Hematol 2020; 157:103127. [PMID: 33161366 DOI: 10.1016/j.critrevonc.2020.103127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Multikinase inhibitors (MKI) are targeted molecular agents that have revolutionized cancer management. However, there is a paucity of data concerning MKI-related liver injury risk and clinical guidelines for the management of liver toxicity in patients receiving MKI for cancer are scarce. DESIGN We conducted a PubMed search of articles in English published from January 2000 to December 2018 related to hepatotoxicity of the 29 FDA-approved MKIs at doses used in clinical practice. The search terms were the international non-proprietary name of each agent cross-referenced with «hepatotoxicity», «hepatitis», «hepatic adverse event», or «liver failure», and «phase II clinical trial», «phase III clinical trial», or «case report». RESULTS Following this search, 140 relevant studies and 99 case reports were considered. Although asymptomatic elevation of aminotransferase levels has been frequently observed in MKI clinical trials, clinically significant hepatotoxicity is a rare event. In most cases, the interval between treatment initiation and the onset of liver injury is between one week and two months. Liver toxicity is often hepatocellular and less frequently mixed. Life-threatening MKI-induced hepatic injury has been described, involving fulminant liver failure or death. Starting from existing data, a description of MKI-related liver events, grading of hepatotoxicity risk, and recommendations for management are also given for various MKI molecules. CONCLUSION All MKIs can potentially cause liver injury, which is sometimes irreversible. As there is still no strategy available to prevent MKI-related hepatotoxicity, early detection remains crucial. The surveillance of liver function during treatment may help in the early detection of hepatotoxicity. Furthermore, the exclusion of potential causes of hepatic injury is essential to avoid unnecessary MKI withdrawal.
Collapse
Affiliation(s)
- Camille Houron
- Faculté de Médecine Paris-Saclay, Université Paris-Saclay, F-94276, Le Kremlin-Bicêtre, France; INSERM U996, DHU Hepatinov, Labex LERMIT, F-92140, Clamart, France
| | - Marie Danielou
- Faculté de Médecine Paris-Saclay, Université Paris-Saclay, F-94276, Le Kremlin-Bicêtre, France; Service d'Hépato-Gastroentérologie et Nutrition, Hôpital Antoine-Béclère, AP-HP, Université Paris-Saclay, F-92140, Clamart, France
| | - Olivier Mir
- Gustave Roussy Cancer Campus, Department of Ambulatory Care, F-94805, Villejuif, France
| | - Bernard Fromenty
- INSERM, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer), UMR_A 1341, UMR_S 1241, F-35000, Rennes, France
| | - Gabriel Perlemuter
- Faculté de Médecine Paris-Saclay, Université Paris-Saclay, F-94276, Le Kremlin-Bicêtre, France; INSERM U996, DHU Hepatinov, Labex LERMIT, F-92140, Clamart, France; Service d'Hépato-Gastroentérologie et Nutrition, Hôpital Antoine-Béclère, AP-HP, Université Paris-Saclay, F-92140, Clamart, France.
| | - Cosmin Sebastian Voican
- Faculté de Médecine Paris-Saclay, Université Paris-Saclay, F-94276, Le Kremlin-Bicêtre, France; INSERM U996, DHU Hepatinov, Labex LERMIT, F-92140, Clamart, France; Service d'Hépato-Gastroentérologie et Nutrition, Hôpital Antoine-Béclère, AP-HP, Université Paris-Saclay, F-92140, Clamart, France
| |
Collapse
|
30
|
Ahmed Z, Singal AK, Kamath PS. Anticoagulants and Their Monitoring. Clin Liver Dis (Hoboken) 2020; 16:146-148. [PMID: 33163166 PMCID: PMC7609710 DOI: 10.1002/cld.946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/11/2020] [Indexed: 02/04/2023] Open
Affiliation(s)
- Zunirah Ahmed
- Division of Gastroenterology and HepatologyBaylor College of MedicineHoustonTX
| | - Ashwani K. Singal
- Division of Transplant HepatologyAvera Transplant InstituteUniversity of South Dakota Sanford School of MedicineSioux FallsSD
| | - Patrick S. Kamath
- Division of Gastroenterology and HepatologyMayo Clinic and Mayo Medical SchoolRochesterMN
| |
Collapse
|
31
|
Roth RA, Ganey PE. What have we learned from animal models of idiosyncratic, drug-induced liver injury? Expert Opin Drug Metab Toxicol 2020; 16:475-491. [PMID: 32324077 DOI: 10.1080/17425255.2020.1760246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Idiosyncratic, drug-induced liver injury (IDILI) continues to plague patients and restrict the use of drugs that are pharmacologically effective. Mechanisms of IDILI are incompletely understood, and a better understanding would reduce speculation and could help to identify safer drug candidates preclinically. Animal models have the potential to enhance knowledge of mechanisms of IDILI. AREAS COVERED Numerous hypotheses have emerged to explain IDILI pathogenesis, many of which center on the roles of the innate and/or adaptive immune systems. Animal models based on these hypotheses are reviewed in the context of their contributions to understanding of IDILI and their limitations. EXPERT OPINION Animal models of IDILI based on an activated adaptive immune system have to date failed to reproduce major liver injury that is of most concern clinically. The only models that have so far resulted in pronounced liver injury are based on the multiple determinant hypothesis or the inflammatory stress hypothesis. The liver pathogenesis in IDILI animal models involves various leukocytes and immune mediators such as cytokines. Insights from animal models are changing the way we view IDILI pathogenesis and are leading to better approaches to preclinical prediction of IDILI potential of new drug candidates.
Collapse
Affiliation(s)
- Robert A Roth
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University , East Lansing, MI, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University , East Lansing, MI, USA
| |
Collapse
|
32
|
Kato R, Ijiri Y, Hayashi T, Uetrecht J. Reactive metabolite of gefitinib activates inflammasomes: implications for gefitinib-induced idiosyncratic reaction. J Toxicol Sci 2020; 45:673-680. [DOI: 10.2131/jts.45.673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Ryuji Kato
- Department of Cardiovascular Pharmacotherapy and Toxicology, Osaka University of Pharmaceutical Sciences
| | - Yoshio Ijiri
- Department of Cardiovascular Pharmacotherapy and Toxicology, Osaka University of Pharmaceutical Sciences
| | - Tetsuya Hayashi
- Department of Cardiovascular Pharmacotherapy and Toxicology, Osaka University of Pharmaceutical Sciences
| | - Jack Uetrecht
- Department of Pharmaceutical Scinces, Faculty of Pharmacy, University of Toronto, Canada
| |
Collapse
|
33
|
Hu M, Yang J, Qu L, Deng X, Duan Z, Fu R, Liang L, Fan D. Ginsenoside Rk1 induces apoptosis and downregulates the expression of PD-L1 by targeting the NF-κB pathway in lung adenocarcinoma. Food Funct 2020; 11:456-471. [DOI: 10.1039/c9fo02166c] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ginsenoside Rk1 can function as an antitumor modulator that induces apoptosis in lung adenocarcinoma cells by inhibiting NF-κB transcription and triggering cell cycle arrest.
Collapse
Affiliation(s)
- Manling Hu
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Jing Yang
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Linlin Qu
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Xuqian Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Zhiguang Duan
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Rongzhan Fu
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Lihua Liang
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials
- School of Chemical Engineering
- Northwest University
- Xi'an
- China
| |
Collapse
|
34
|
Carretero-González A, Salamanca Santamaría J, Castellano D, de Velasco G. Three case reports: Temporal association between tyrosine-kinase inhibitor-induced hepatitis and immune checkpoint inhibitors in renal cell carcinoma. Medicine (Baltimore) 2019; 98:e18098. [PMID: 31764847 PMCID: PMC6882583 DOI: 10.1097/md.0000000000018098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
RATIONALE Hepatotoxicity is a well-known adverse effect of vascular endothelial growth factor receptor (VEGFR) tyrosine-kinase inhibitors (TKIs), usually employed for the treatment of metastatic renal cell carcinoma (mRCC). Immune checkpoint inhibitors (ICIs) have been shown to improve survival in specific patients with mRCC, but concerns have arisen over their safety profile, particularly as regards the risk of liver damage in those patients receiving TKIs sequentially or concurrently with these new drugs. Here, we report three cases of hepatitis presentation in patients receiving TKIs after ICIs that should potentially be considered in current clinical practice, where a combination of these hepatotoxic drugs is becoming increasingly used. PATIENTS CONCERNS All three patients were receiving TKIs therapy and presented with nonspecific clinical deterioration and liver enzyme elevation in different time frames according to the start of treatment. All were previously treated with ICIs. DIAGNOSES After performing imaging techniques and complementary laboratory tests for the differential diagnosis of hepatic injury, the diagnosis of potentially TKI-induced hepatitis was assumed in all these cases. Hepatic biopsy was performed only in the first case in order to confirm the diagnosis. INTERVENTIONS Potential toxic drugs were interrupted and steroids course with slow reduction regimen was administered in all these cases because of the previous use of ICIs. OUTCOMES The patients described improved with this conservative treatment without complications during the following weeks. Only one case presented a new episode of mild hepatic alteration while on treatment with following treatment. LESSONS Taking into account this new therapeutic context, stricter monitoring for potentially increased/altered adverse events should be indicated. Adequate patient selection and consideration of the safety profile of the different drugs used could help to optimize treatment in the near future.
Collapse
|
35
|
Abstract
Terlipressin, somatostatin, or octreotide are recommended as pharmacologic treatment of acute variceal hemorrhage. Nonselective β-blockers decrease the risk of variceal hemorrhage and hepatic decompensation, particularly in those 30% to 40% of patients with good hemodynamic response. Carvedilol, statins, and anticoagulants are promising agents in the management of portal hypertension. Recent advances in the pharmacologic treatment of portal hypertension have mainly focused on modifying an increased intrahepatic resistance through nitric oxide and/or modulation of vasoactive substances. Several novel pharmacologic agents for portal hypertension are being evaluated in humans.
Collapse
Affiliation(s)
- Chalermrat Bunchorntavakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Rajavithi Hospital, College of Medicine, Rangsit University, Rajavithi Road, Ratchathewi, Bangkok 10400, Thailand; Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, 2 Dulles, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - K Rajender Reddy
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, 2 Dulles, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Wang Z, Li W, Jing H, Ding M, Fu G, Yuan T, Huang W, Dai M, Tang D, Zeng M, Chen Y, Zhang H, Zhu X, Peng Y, Li Q, Yu WF, Yan HX, Zhai B. Generation of hepatic spheroids using human hepatocyte-derived liver progenitor-like cells for hepatotoxicity screening. Theranostics 2019; 9:6690-6705. [PMID: 31588244 PMCID: PMC6771233 DOI: 10.7150/thno.34520] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/04/2019] [Indexed: 02/06/2023] Open
Abstract
Rationale: The idiosyncratic drug-induced liver injury (iDILI) is a major cause of acute liver injury and a key challenge in late-stage drug development. Individual heterogeneity is considered to be an essential factor of iDILI. However, few in vitro model can predict heterogeneity in iDILI. We have previously shown that mouse and human hepatocytes can be converted to expandable liver progenitor-like cells in vitro (HepLPCs). However, the limited proliferation potential of human HepLPCs confines its industrial application. Here, we reported the generation of a novel hepatocyte model not only to provide unlimited cell sources for human hepatocytes but also to establish a tool for studying iDILI in vitro. Methods: Human primary hepatocytes were isolated by modified two-step perfusion technique. The chemical reprogramming culture condition together with gene-transfer were then used to generate the immortalized HepLPC cell lines (iHepLPCs). Growth curve, doubling time, and karyotype were analyzed to evaluate the proliferation characteristics of iHepLPCs. Modified Hepatocyte Maturation Medium and 3D spheroid culture were applied to re-differentiate iHepLPCs. Results: iHepLPCs exhibited efficient expansion for at least 40 population doublings, with a stable proliferative ability. They could easily differentiate back into metabolically functional hepatocytes in vitro within 10 days. Furthermore, under three-dimensional culture conditions, the formed hepatic spheroids showed multiple liver functions and toxicity profiles close to those of primary human hepatocytes. Importantly, we established a hepatocyte bank by generating a specific number of such cell lines. Screening for population heterogeneity allowed us to analyze the in vitro heterogeneous responses to hepatotoxicity induced by molecular targeted drugs. Conclusions: In light of the proliferative capacity and the heterogeneity they represented, these iHepLPCs cell lines may offer assistance in studying xenobiotic metabolism as well as liver diseases in vitro.
Collapse
|
37
|
Tangamornsuksan W, Kongkaew C, Scholfield CN, Subongkot S, Lohitnavy M. HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity: a systematic review and meta-analysis. THE PHARMACOGENOMICS JOURNAL 2019; 20:47-56. [PMID: 31383939 DOI: 10.1038/s41397-019-0092-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/28/2018] [Accepted: 07/18/2019] [Indexed: 01/21/2023]
Abstract
Associations between HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity have been reported. To consolidate the results from all available reports in scientific databases, systematic review and meta-analysis techniques were used to quantify these associations. Studies investigating associations between HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity were systematically searched in PubMed, Human Genome Epidemiology Network, and the Cochrane Library. Primary outcomes were the associations between HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity. Overall odds ratios (ORs) with the corresponding 95%CIs were calculated using a random-effect model to determine the associations between HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity. A clear association between HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity was identified in our analyses. The summary OR was 6.23 (95%CI = 4.11-9.45). Similar associations were also found in the subgroup analyses by lapatinib treatment regimens. ORs were 10.04 (95%CI = 6.15-16.39), 8.65 (95%CI = 4.52-16.58), and 3.88 (95%CI = 2.20-6.82) in the lapatinib group, lapatinib + trastuzumab group, and lapatinib + chemotherapy or lapatinib + trastuzumab + chemotherapy group, respectively. Since HLA-DRB1*07:01 is associated with lapatinib-induced hepatotoxicity, genetic screening of HLA-DRB1*07:01 in breast cancer patients prior to lapatinib therapy is warranted for patient safety. In addition, further studies should define the risk of HLA-DRB1*07:01 and lapatinib-induced hepatotoxicity in specific ethnicities.
Collapse
Affiliation(s)
- Wimonchat Tangamornsuksan
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Chuenjid Kongkaew
- Center of Excellence for Environmental Health & Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand.,Centre for Safety and Quality in Health, Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand.,Pharmacokinetic Research Unit, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand.,Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
| | - C N Scholfield
- Center of Excellence for Environmental Health & Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand.,Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand
| | - Suphat Subongkot
- Clinical Pharmacy Division, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Manupat Lohitnavy
- Center of Excellence for Environmental Health & Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand. .,Pharmacokinetic Research Unit, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand. .,Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand.
| |
Collapse
|
38
|
Yancheva N, Tzonev R. A case of late presentation of darunavir-related cholestatic hepatitis. Int J STD AIDS 2019; 30:620-622. [PMID: 30722751 DOI: 10.1177/0956462419826723] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report a case of darunavir-induced cholestatic hepatitis in a human immunodeficiency virus (HIV)-infected patient in the third year of his combined antiretroviral therapy. During the patient's monthly follow-up with regard to his HIV infection, elevated transaminase levels were detected. The patient was subsequently hospitalised at the AIDS and gastroenterology departments. All tested viral hepatitis markers were negative. A diagnosis of autoimmune hepatitis was also ruled out. The liver biopsy revealed cholestatic hepatitis. Darunavir withdrawal resulted in a progressive decrease in liver enzyme levels. We highlight the importance of recognising late development of liver injury secondary to the use of darunavir, and the importance of monitoring liver function in patients undergoing prolonged treatment involving darunavir.
Collapse
Affiliation(s)
- Nina Yancheva
- 1 Department of AIDS, Specialized Hospital for Active Treatment of Infectious and Parasitic Diseases "Prof. Iv. Kirov" Medical University, Sofia, Bulgaria
| | - Radin Tzonev
- 2 Department of Gastroenterology, Tokuda Acibadem Hospital, Sofia, Bulgaria
| |
Collapse
|
39
|
Beattie W, Yan B, Sood S. Acute severe hepatitis with alemtuzumab and rechallenge after a year. J Clin Neurosci 2018; 60:158-160. [PMID: 30348589 DOI: 10.1016/j.jocn.2018.10.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/06/2018] [Indexed: 01/21/2023]
Abstract
Alemtuzumab is a monoclonal antibody used as a disease modifying agent in relapsing and remitting multiple sclerosis. It has not previously been associated with drug induced liver injury. Here we present a case of a 49 year old female developing drug induced liver injury secondary to alemtuzumab, confirmed upon rechallenge. Our patient developed severe hepatitis within two days of starting alemtuzumab, both initially and upon rechallenge. The alanine aminotransferase peaked at 577 units per litre and 426 units per litre after initial dose of alemtuzumab and rechallenge respectively. The patient's liver function tests improved significantly between doses of alemtuzumab and again normalised within three months of the second dose, with no clinical manifestations of acute hepatic failure. A full hepatitis screen ruled out alternative causes of hepatitis, including autoimmune hepatitis and hepatotoxicity relating to other medications. Deliberate rechallenge with a medication thought to be associated with drug induced liver injury is uncommonly performed but provides substantial evidence for causality in the appropriate clinical context. The Roussel Uclaf Causality Assessment Method score was calculated as nine, indicating that drug induced liver injury due to alemtuzumab is highly probable. Drug induced liver injury is a potentially serious condition that should be monitored for during alemtuzumab treatment of relapsing and remitting multiple sclerosis. The role of a rechallenge should be weighed up on a case by case basis.
Collapse
Affiliation(s)
- William Beattie
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Victoria, Australia.
| | - Bernard Yan
- Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Siddharth Sood
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
40
|
Abstract
Various medications used to treat inflammatory bowel diseases have been implicated to cause hepatotoxicity. These include sulfasalazine, 5-aminosalicylic acids, fluoroquinolones, metronidazole, thiopurines, methotrexate, anti-tumor necrosis factor agents, and alpha-4 integrin inhibitors. Various types of liver injury have been reported in association with these medications including hypersensitivity reaction, hepatocellular or cholestatic disease, nodular regenerative hyperplasia, liver fibrosis/cirrhosis, portal hypertension and autoimmune liver injury. The revised Roussel Uclaf Causality Assessment Method (RUCAM) provides a scoring system to determine the likelihood of whether a drug caused liver injury. Unfortunately some of the reported liver injuries in association with these treatments have not undergone RUCAM assessment. Therefore, although some of the reports used in this review article show an association between a medication and the reported liver injury, they may not necessarily show causation. In this article, we address methods of monitoring to detect these injuries. We also discuss the prognosis and recommended management plans when liver injury occurs.
Collapse
|
41
|
Salas JR, Chen BY, Wong A, Duarte S, Angarita SAK, Lipshutz GS, Witte ON, Clark PM. Noninvasive Imaging of Drug-Induced Liver Injury with 18F-DFA PET. J Nucl Med 2018; 59:1308-1315. [PMID: 29496991 PMCID: PMC6071498 DOI: 10.2967/jnumed.117.206961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/21/2018] [Indexed: 12/19/2022] Open
Abstract
Drug-induced liver failure is a significant indication for a liver transplant, and unexpected liver toxicity is a major reason that otherwise effective therapies are removed from the market. Various methods exist for monitoring liver injury but are often inadequate to predict liver failure. New diagnostic tools are needed. Methods: We evaluate in a preclinical model whether 18F-2-deoxy-2-fluoroarabinose (18F-DFA), a PET radiotracer that measures the ribose salvage pathway, can be used to monitor acetaminophen-induced liver injury and failure. Mice treated with vehicle, 100, 300, or 500 mg/kg acetaminophen for 7 or 21 h were imaged with 18F-FDG and 18F-DFA PET. Hepatic radiotracer accumulation was correlated to survival and percentage of nonnecrotic tissue in the liver. Mice treated with acetaminophen and vehicle or N-acetylcysteine were imaged with 18F-DFA PET. 18F-DFA accumulation was evaluated in human hepatocytes engrafted into the mouse liver. Results: We show that hepatic 18F-DFA accumulation is 49%-52% lower in mice treated with high-dose acetaminophen than in mice treated with low-dose acetaminophen or vehicle. Under these same conditions, hepatic 18F-FDG accumulation was unaffected. At 21 h after acetaminophen treatment, hepatic 18F-DFA accumulation can distinguish mice that will succumb to the liver injury from those that will survive it (6.2 vs. 9.7 signal to background, respectively). Hepatic 18F-DFA accumulation in this model provides a tomographic representation of hepatocyte density in the liver, with a R2 between hepatic 18F-DFA accumulation and percentage of nonnecrotic tissue of 0.70. PET imaging with 18F-DFA can be used to distinguish effective from ineffective resolution of acetaminophen-induced liver injury with N-acetylcysteine (15.6 vs. 6.2 signal to background, respectively). Human hepatocytes, in culture or engrafted into a mouse liver, have levels of ribose salvage activity similar to those of mouse hepatocytes. Conclusion: Our findings suggest that PET imaging with 18F-DFA can be used to visualize and quantify drug-induced acute liver injury and may provide information on the progression from liver injury to hepatic failure.
Collapse
Affiliation(s)
- Jessica R Salas
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles California
- Crump Institute for Molecular Imaging, University of California, Los Angeles California
| | - Bao Ying Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles California
- Crump Institute for Molecular Imaging, University of California, Los Angeles California
| | - Alicia Wong
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles California
- Crump Institute for Molecular Imaging, University of California, Los Angeles California
| | - Sergio Duarte
- Department of Surgery, University of California, Los Angeles California
| | | | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles California
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles California; and
| | - Owen N Witte
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles California
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles California
| | - Peter M Clark
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles California
- Crump Institute for Molecular Imaging, University of California, Los Angeles California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles California
| |
Collapse
|
42
|
Ultra-performance LC–MS/MS study of the pharmacokinetic interaction of imatinib with selected vitamin preparations in rats. Bioanalysis 2018; 10:1099-1113. [DOI: 10.4155/bio-2018-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: The growing interest of cancerous patients in using vitamins, while on imatinib (IMA) therapy, increased the risk of their pharmacokinetic interactions. Methodology: Ultra-performance LC–MS/MS method was developed and validated for the determination of IMA following oral administration of selected vitamin preparations (vitamin A, E, D3 and C) in rat plasma using a hybrid sample preparation technique of protein precipitation followed by SPE. Results: The method showed good linear response for IMA over the concentration range 1–500 ng/ml. Co-administered vitamin preparations could affect IMA pharmacokinetic profiling through either an increase (vitamin A and E) or a decrease (vitamin C) in IMA bioavailability. Vitamin D3 produced no significant effect on IMA bioavailability. Conclusion: Particular concern should be paid when vitamin preparations are administered with IMA.
Collapse
|
43
|
Birch JC, Khatri G, Watumull LM, Arriaga YE, Leyendecker JR. Unintended Consequences of Systemic and Ablative Oncologic Therapy in the Abdomen and Pelvis. Radiographics 2018; 38:1158-1179. [PMID: 29995613 DOI: 10.1148/rg.2018170137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Human cancers are genetically complex and diverse. Although advances in oncologic therapy aim to define and target unique steps in carcinogenesis, oncologists often rely on less discriminate anticancer therapies that have consequences for normal tissues. Even many of the so-called targeted therapies currently employed can adversely affect normal cells, leading to complications that necessitate dose reductions or cessation of specific therapies. This article explores the unintended consequences of currently employed systemic and ablative anticancer therapies that might manifest at imaging examinations of the abdomen and pelvis, including cytotoxic, molecular targeted, and immunologic agents; ablation; and hematopoietic stem cell transplant. Each of these treatments can have both major and minor unintended effects in the targeted organ(s), in local or adjacent structures, or at distant sites. Timely detection and reporting of adverse consequences of anticancer therapies by the astute imager can result in critical treatment modifications and/or lifesaving interventions; therefore, knowledge of these unintended effects is paramount for radiologists interpreting the results of imaging examinations in cancer patients. ©RSNA, 2018.
Collapse
Affiliation(s)
- Julie C Birch
- From the Department of Radiology (J.C.B., G.K., L.M.W., J.R.L.) and Department of Internal Medicine, Division of Hematology/Oncology (Y.E.A.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Gaurav Khatri
- From the Department of Radiology (J.C.B., G.K., L.M.W., J.R.L.) and Department of Internal Medicine, Division of Hematology/Oncology (Y.E.A.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Lori M Watumull
- From the Department of Radiology (J.C.B., G.K., L.M.W., J.R.L.) and Department of Internal Medicine, Division of Hematology/Oncology (Y.E.A.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Yull E Arriaga
- From the Department of Radiology (J.C.B., G.K., L.M.W., J.R.L.) and Department of Internal Medicine, Division of Hematology/Oncology (Y.E.A.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| | - John R Leyendecker
- From the Department of Radiology (J.C.B., G.K., L.M.W., J.R.L.) and Department of Internal Medicine, Division of Hematology/Oncology (Y.E.A.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390
| |
Collapse
|
44
|
Barrett A, Moore M, Ferrins P, Thornton P, Murphy P, Quinn J. From a direct oral anticoagulant to warfarin: reasons why patients switch. Ir J Med Sci 2017; 187:719-721. [DOI: 10.1007/s11845-017-1730-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/07/2017] [Indexed: 12/20/2022]
|
45
|
Comuth WJ, Haase AM, Henriksen LØ, Malczynski J, van de Kerkhof D, Münster AMB. Cholestatic liver injury as a side-effect of dabigatran and the use of coagulation tests in dabigatran intoxication and after reversal by idarucizumab in bleeding and sepsis. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 78:1-5. [PMID: 29148292 DOI: 10.1080/00365513.2017.1402127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Idarucizumab, an antidote specific for dabigatran, became available recently. Dabigatran is not associated with increased risk of hepatotoxicity in comparison with warfarin, but it is seen as a rare side-effect. Cases of cholestatic liver injury due to dabigatran have not been reported previously. We present a case of severe gastro-intestinal bleeding with underlying dabigatran intoxication in a patient with renal failure and the effect of reversal of dabigatran using idaruzicumab on coagulation assays. International normalized ratio (INR) and activated partial thromboplastin time (APTT) results were elevated in a setting of sepsis, possibly due to liver failure. INR and APTT can be elevated if sepsis is complicated by disseminated intravascular coagulation (DIC) or liver failure, making it challenging to determine dabigatrans contribution to their prolongation. A rebound effect after administration of idarucizumab and slow elimination of dabigatran due to reduced kidney function could be detected using the Hemoclot® diluted thrombin time (dTT) in this situation, in contrast to with non-dilutional assays. Before admission, cholestatic liver injury started shortly after initiation of dabigatran etexilate therapy. As no other cause was found, this liver injury was likely to be drug-induced. Bleeding cessated promptly after administration of idarucizumab in dabigatran intoxication. In conclusion, the anticoagulant effect of dabigatran can be measured by Hemoclot® dTT in sepsis and cholestatic liver injury was seen as a possible rare side-effect of dabigatran treatment.
Collapse
Affiliation(s)
- Willemijn J Comuth
- a Department of Clinical Biochemistry , Hospital Unit West, Herning and Holstebro , Denmark.,b Department of Cardiology , Hospital Unit West , Herning , Denmark.,c Faculty of Health , Institute of Clinical Medicine, Aarhus University , Aarhus , Denmark
| | - Anne-Mette Haase
- d Department of Medicine , Hospital Unit West , Herning , Denmark
| | - Linda Ø Henriksen
- a Department of Clinical Biochemistry , Hospital Unit West, Herning and Holstebro , Denmark
| | - Jerzy Malczynski
- b Department of Cardiology , Hospital Unit West , Herning , Denmark
| | - Daan van de Kerkhof
- e Department of Clinical Biochemistry , Catharina Hospital , Eindhoven , the Netherlands
| | - Anna-Marie B Münster
- f Unit for Thrombosis Research , University of Southern Denmark , Esbjerg , Denmark.,g Department of Clinical Biochemistry , Hospital of South West Denmark , Esbjerg , Denmark
| |
Collapse
|
46
|
Abstract
Acute liver failure (ALF) is a life-threatening condition of heterogeneous etiology. Outcomes are better with early recognition and prompt initiation of etiology-specific therapy, intensive care protocols, and liver transplantation (LT). Prognostic scoring systems include the King's College Criteria and Model for End-stage Liver Disease score. Cerebral edema and intracranial hypertension are reasons for high morbidity and mortality; hypertonic saline is suggested for patients with a high risk for developing intracranial hypertension, and when it does, mannitol is recommended as first-line therapy. Extracorporeal liver support system may serve as a bridge to LT and may increase LT-free survival in select cases.
Collapse
Affiliation(s)
- Chalermrat Bunchorntavakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Rajavithi Hospital, College of Medicine, Rangsit University, Rajavithi Road, Ratchathewi, Bangkok 10400, Thailand; Division of Gastroenterology and Hepatology, Department of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania, 2 Dulles, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - K Rajender Reddy
- Division of Gastroenterology and Hepatology, Department of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania, 2 Dulles, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
|
48
|
Alessandrino F, Tirumani SH, Krajewski KM, Shinagare AB, Jagannathan JP, Ramaiya NH, Di Salvo DN. Imaging of hepatic toxicity of systemic therapy in a tertiary cancer centre: chemotherapy, haematopoietic stem cell transplantation, molecular targeted therapies, and immune checkpoint inhibitors. Clin Radiol 2017; 72:521-533. [PMID: 28476244 DOI: 10.1016/j.crad.2017.04.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 12/13/2022]
Abstract
The purpose of this review is to familiarise radiologists with the spectrum of hepatic toxicity seen in the oncology setting, in view of the different systemic therapies used in cancer patients. Drug-induced liver injury can manifest in various forms, and anti-neoplastic agents are associated with different types of hepatotoxicity. Although chemotherapy-induced liver injury can present as hepatitis, steatosis, sinusoidal obstruction syndrome, and chronic parenchymal damages, molecular targeted therapy-associated liver toxicity ranges from mild liver function test elevation to fulminant life-threatening acute liver failure. The recent arrival of immune checkpoint inhibitors in oncology has introduced a new range of immune-related adverse events, with differing mechanisms of liver toxicity and varied imaging presentation of liver injury. High-dose chemotherapy regimens for haematopoietic stem cell transplantation are associated with sinusoidal obstruction syndrome. Management of hepatic toxicity depends on the clinical scenario, the drug in use, and the severity of the findings. In this article, we will (1) present the most common types of oncological drugs associated with hepatic toxicity and associated liver injuries; (2) illustrate imaging findings of hepatic toxicities and the possible differential diagnosis; and (3) provide a guide for management of these conditions.
Collapse
Affiliation(s)
- F Alessandrino
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - S H Tirumani
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - K M Krajewski
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - A B Shinagare
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - J P Jagannathan
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - N H Ramaiya
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - D N Di Salvo
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|