1
|
Beaudoin JJ, Clemens L, Miedel MT, Gough A, Zaidi F, Ramamoorthy P, Wong KE, Sarangarajan R, Battista C, Shoda LKM, Siler SQ, Taylor DL, Howell BA, Vernetti LA, Yang K. The Combination of a Human Biomimetic Liver Microphysiology System with BIOLOGXsym, a Quantitative Systems Toxicology (QST) Modeling Platform for Macromolecules, Provides Mechanistic Understanding of Tocilizumab- and GGF2-Induced Liver Injury. Int J Mol Sci 2023; 24:9692. [PMID: 37298645 PMCID: PMC10253699 DOI: 10.3390/ijms24119692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Biologics address a range of unmet clinical needs, but the occurrence of biologics-induced liver injury remains a major challenge. Development of cimaglermin alfa (GGF2) was terminated due to transient elevations in serum aminotransferases and total bilirubin. Tocilizumab has been reported to induce transient aminotransferase elevations, requiring frequent monitoring. To evaluate the clinical risk of biologics-induced liver injury, a novel quantitative systems toxicology modeling platform, BIOLOGXsym™, representing relevant liver biochemistry and the mechanistic effects of biologics on liver pathophysiology, was developed in conjunction with clinically relevant data from a human biomimetic liver microphysiology system. Phenotypic and mechanistic toxicity data and metabolomics analysis from the Liver Acinus Microphysiology System showed that tocilizumab and GGF2 increased high mobility group box 1, indicating hepatic injury and stress. Tocilizumab exposure was associated with increased oxidative stress and extracellular/tissue remodeling, and GGF2 decreased bile acid secretion. BIOLOGXsym simulations, leveraging the in vivo exposure predicted by physiologically-based pharmacokinetic modeling and mechanistic toxicity data from the Liver Acinus Microphysiology System, reproduced the clinically observed liver signals of tocilizumab and GGF2, demonstrating that mechanistic toxicity data from microphysiology systems can be successfully integrated into a quantitative systems toxicology model to identify liabilities of biologics-induced liver injury and provide mechanistic insights into observed liver safety signals.
Collapse
Affiliation(s)
- James J. Beaudoin
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lara Clemens
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Mark T. Miedel
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Albert Gough
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Fatima Zaidi
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Kari E. Wong
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Christina Battista
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lisl K. M. Shoda
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Scott Q. Siler
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - D. Lansing Taylor
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Brett A. Howell
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lawrence A. Vernetti
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Kyunghee Yang
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| |
Collapse
|
2
|
Quantitative Systems Toxicology and Drug Development: The DILIsym Experience. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2486:181-196. [PMID: 35437723 DOI: 10.1007/978-1-0716-2265-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
DILIsym® is a Quantitative Systems Toxicology (QST) model that has been developed over the last decade by a public-private partnership to predict the liver safety liability in new drug candidates. DILIsym integrates the quantitative abilities of parent and relevant metabolites to cause oxidative stress, mitochondrial dysfunction, and alter bile acid homeostasis. Like the prediction of drug-drug interactions, the data entered into DILIsym are assessed in the laboratory in human experimental systems, and combined with estimates of liver exposure to predict the outcome. DILIsym is now frequently used in decision-making within the pharmaceutical industry and its modeling results are increasingly included in regulatory communications and NDA submissions. DILIsym can be used to identify dominant mechanisms underlying liver toxicity and this information is increasingly being used to identify patient-specific risk factors, including certain disease states. DILIsym is also increasingly used to optimize the interpretation of liver injury biomarkers. DILIsym provides an example of how QST modeling can help speed the delivery of safer new drugs to the patients who need them.
Collapse
|
3
|
Ghufran H, Azam M, Mehmood A, Butt H, Riazuddin S. Standardization of diethylnitrosamine-induced hepatocellular carcinoma rat model with time based molecular assessment. Exp Mol Pathol 2021; 123:104715. [PMID: 34699901 DOI: 10.1016/j.yexmp.2021.104715] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
This study was intended (1) to develop a robust animal model for hepatocellular carcinoma (HCC) research, in which HCC tumors develop in a background of fibrosis or cirrhosis; and (2) to explore time-dependent regulatory changes in key molecular markers during disease advancement and HCC development. With the aim of establishing such HCC model, male Sprague-Dawley rats were injected with diethylnitrosamine (DEN) at a dose of 30 mg/kg twice a week for 10 weeks then once a week from 12th to 16th weeks. The rats were kept under observation until 18th week. At defined time intervals (2nd, 4th, 12th, and 18th week), serum biomarkers and microscopic components of tissue samples were used to investigate the chronic progression of liver disease, while gene and protein analysis was used to monitor expression patterns during HCC development. DEN-intoxicated rats manifested inflammation at week 4, fibrosis at week 12 and cirrhosis with early HCC tumors at week 18. Molecular analysis revealed that key markers of inflammation (Il-1β, Il-6, and Tnf-α), fibrosis (Tgf-β1, Col1α1, Col3α1, and Timp-1), and angiogenesis (Hif1-α and Vegf) were promptly (P ≤ 0.001) up-regulated at week 4, week 12 and week 18, respectively. Oxidative stress (iNos, Cyp2e1, and Sod1) and pro-apoptotic (Bax) markers showed significant upregulation from week 4 to week 12. However, Sod1 and Bax expressions dropped after week 12 and reached a minimum at 18th week. Strikingly, expressions of anti-apoptotic (Bcl-2) and cell proliferation (Pcna, Hgf, and Afp) markers were abruptly increased at week 18. Collectively, we describe an 18-week HCC model in DEN-intoxicated rats that exhibit chronic inflammation, oxidative imbalance, advance fibrosis/cirrhosis, halted apoptosis, and angiogenic sprouting, progressively.
Collapse
Affiliation(s)
- Hafiz Ghufran
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Maryam Azam
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Hira Butt
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Sheikh Riazuddin
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan; Allama Iqbal Medical Research Centre, Jinnah Burn and Reconstructive Surgery Centre, Lahore, Pakistan.
| |
Collapse
|
4
|
Abstract
The incidence rate of drug-induced liver injury has been high with the extensive use of drugs and the development and application of new drugs. The pathogenesis of drug-induced liver injury is not fully understood, so there is no significant breakthrough in its treatment. The diagnosis of drug-induced liver injury still depends on drug history, clinical manifestations, imaging, biochemical tests, and liver biopsy. This article reviews the recent progress in the understanding of the incidence rate, classification, risk factors, and serum markers of drug-induced liver injury.
Collapse
Affiliation(s)
- Zhao-Chun Chi
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266011, Shandong Province, China
| |
Collapse
|
5
|
Yang E, Choi H, Park JS, Noh YW, Choi CM, Lee WJ, Ko JW, Kim J. A first-in-human study of KMRC011, a potential treatment for acute radiation syndrome, to explore tolerability, pharmacokinetics, and pharmacodynamics. Clin Transl Sci 2021; 14:2161-2170. [PMID: 34080313 PMCID: PMC8604209 DOI: 10.1111/cts.13073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 11/27/2022] Open
Abstract
KMRC011 is a novel Toll-like receptor 5 agonist under development as a treatment for acute radiation syndrome (ARS). The aim of this first-in-human study was to investigate the tolerability, pharmacokinetics, and pharmacodynamics of a single intramuscular dose of KMRC011 in healthy subjects. A randomized, single-blind, placebo-controlled, single dose-escalation study was conducted with the starting dose of 5 μg. Eight (4 only for 5 μg cohort) subjects per cohort were randomly assigned to KMRC011 or placebo in a 3:1 ratio. Dose-limiting toxicity (DLT) was assessed throughout the study. Serum concentrations of KMRC011, granulocyte colony-stimulating factor (G-CSF), and interleukin-6 (IL-6) were measured up to 48 h postdose. Based on safety review, the dose of KMRC011 escalated up to 20 μg, and consequently, a total of 4 dose levels (5, 10, 15, and 20 μg) were explored. The most common adverse event was injection site reaction, showing no dose-related trend. Three DLTs (2 cases of hepatic enzyme increased and 1 of pyrexia) were observed; 1 in the 15 μg cohort and 2 in the 20 μg cohort. A developed method could not detect any KMRC011 in serum. KMRC011 15 μg and 20 μg showed significant increases of G-CSF, IL-6, and absolute neutrophil counts, compared with the placebo. A single intramuscular administration of KMRC011 ranging from 5 to 15 μg was tolerated in healthy subjects. Doses of KMRC011 equal to or greater than 15 μg exerted TLR5 agonist-like activities by increasing serum G-CSF and IL-6. It suggests that KMRC011 has the potential for a treatment for ARS.
Collapse
Affiliation(s)
- Eunsol Yang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Hyejung Choi
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Jin-Sol Park
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju-si, Korea
| | | | - Woo-Jong Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon-si, Korea
| | - Jae-Wook Ko
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Jungryul Kim
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea.,Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
6
|
Application of the DILIsym® Quantitative Systems Toxicology drug-induced liver injury model to evaluate the carcinogenic hazard potential of acetaminophen. Regul Toxicol Pharmacol 2020; 118:104788. [PMID: 33153971 DOI: 10.1016/j.yrtph.2020.104788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/11/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
In 2019, the California Office of Environmental Health Hazard Assessment (OEHHA) initiated a review of the carcinogenic hazard potential of acetaminophen. The objective of the analysis herein was to inform this review by assessing whether variability in patient baseline characteristics (e.g. baseline glutathione (GSH) levels, pharmacokinetics, and capacity of hepatic antioxidants) leads to potential differences in carcinogenic hazard potential at different dosing schemes: maximum labeled doses of 4 g/day, repeated doses above the maximum labeled dose (>4-12 g/day), and acute overdoses of acetaminophen (>15 g). This was achieved by performing simulations of acetaminophen exposure in thousands of diverse virtual patients scenarios using the DILIsym® Quantitative Systems Toxicology (QST) model. Simulations included assessments of the dose and exposure response for toxicity and mode of cell death based on evaluations of the kinetics of changes of: GSH, N-acetyl-p-benzoquinone-imine (NAPQI), protein adducts, mitochondrial dysfunction, and hepatic cell death. Results support that, at therapeutic doses, cellular GSH binds to NAPQI providing sufficient buffering capacity to limit protein adduct formation and subsequent oxidative stress. Simulations evaluating repeated high-level supratherapeutic exposures or acute overdoses indicate that cell death precedes DNA damage that could result in carcinogenicity and thus acetaminophen does not present a carcinogenicity hazard to humans at any dose.
Collapse
|
7
|
Watkins PB. DILIsym: Quantitative systems toxicology impacting drug development. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
8
|
Watkins PB. The DILI-sim Initiative: Insights into Hepatotoxicity Mechanisms and Biomarker Interpretation. Clin Transl Sci 2020; 12:122-129. [PMID: 30762301 PMCID: PMC6440570 DOI: 10.1111/cts.12629] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/16/2022] Open
Abstract
The drug‐induced liver injury (DILI)‐sim Initiative is a public‐private partnership involving scientists from industry, academia, and the US Food and Drug Administration (FDA). The Initiative uses quantitative systems toxicology (QST) to build and refine a model (DILIsym) capable of understanding and predicting liver safety liabilities in new drug candidates and to optimize interpretation of liver safety biomarkers used in clinical studies. Insights gained to date include the observation that most dose‐dependent hepatotoxicity can be accounted for by combinations of just three mechanisms (oxidative stress, interference with mitochondrial respiration, and alterations in bile acid homeostasis) and the importance of noncompetitive inhibition of bile acid transporters. The effort has also provided novel insight into species and interpatient differences in susceptibility, structure‐activity relationships, and the role of nonimmune mechanisms in delayed idiosyncratic hepatotoxicity. The model is increasingly used to evaluate new drug candidates and several clinical trials are underway that will test the model's ability to prospectively predict liver safety. With more refinement, in the future, it may be possible to use the DILIsym predictions to justify reduction in the size of some clinical trials. The mature model could also potentially assist physicians in managing the liver safety of their patients as well as aid in the diagnosis of DILI.
Collapse
Affiliation(s)
- Paul B Watkins
- Institute for Drug Safety Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
9
|
Smith AK, Ropella GEP, McGill MR, Krishnan P, Dutta L, Kennedy RC, Jaeschke H, Hunt CA. Contrasting model mechanisms of alanine aminotransferase (ALT) release from damaged and necrotic hepatocytes as an example of general biomarker mechanisms. PLoS Comput Biol 2020; 16:e1007622. [PMID: 32484845 PMCID: PMC7292418 DOI: 10.1371/journal.pcbi.1007622] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/12/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022] Open
Abstract
Interpretations of elevated blood levels of alanine aminotransferase (ALT) for drug-induced liver injury often assume that the biomarker is released passively from dying cells. However, the mechanisms driving that release have not been explored experimentally. The usefulness of ALT and related biomarkers will improve by developing mechanism-based explanations of elevated levels that can be expanded and elaborated incrementally. We provide the means to challenge the ability of closely related model mechanisms to generate patterns of simulated hepatic injury and ALT release that scale (or not) to be quantitatively similar to the wet-lab validation targets, which are elevated plasma ALT values following acetaminophen (APAP) exposure in mice. We build on a published model mechanism that helps explain the generation of characteristic spatiotemporal features of APAP hepatotoxicity within hepatic lobules. Discrete event and agent-oriented software methods are most prominent. We instantiate and leverage a small constellation of concrete model mechanisms. Their details during execution help bring into focus ways in which particular sources of uncertainty become entangled with cause-effect details within and across several levels. We scale ALT amounts in virtual mice directly to target plasma ALT values in individual mice. A virtual experiment comprises a set of Monte Carlo simulations. We challenge the sufficiency of four potentially explanatory theories for ALT release. The first of the tested model theories failed to achieve the initial validation target, but each of the three others succeeded. Results for one of the three model mechanisms matched all target ALT values quantitatively. It explains how ALT externalization is the combined consequence of lobular-location-dependent drug-induced cellular damage and hepatocyte death. Falsification of one (or more) of the model mechanisms provides new knowledge and incrementally shrinks the constellation of model mechanisms. The modularity and biomimicry of our explanatory models enable seamless transition from mice to humans.
Collapse
Affiliation(s)
- Andrew K. Smith
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| | | | - Mitchell R. McGill
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Preethi Krishnan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| | - Lopamudra Dutta
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| | - Ryan C. Kennedy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - C. Anthony Hunt
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| |
Collapse
|
10
|
Church RJ, Watkins PB. Serum biomarkers of drug-induced liver injury: Current status and future directions. J Dig Dis 2019; 20:2-10. [PMID: 30378260 DOI: 10.1111/1751-2980.12684] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Drug-induced liver injury (DILI), which is caused by drugs and herbal or dietary supplements, remains a serious concern for drug developers, regulators, and clinicians; however, serum biomarkers utilized to detect and monitor DILI have not changed in decades and have limitations. Data-driven mathematical modeling that incorporates the release and clearance kinetics of traditional biomarkers has improved their use in the prediction of liver safety liabilities for new drug candidates. Several newer biomarkers have shown promise in terms of liver specificity, predicting the outcome of DILI events, and providing insight into its underlying mechanisms. For these new biomarkers to be qualified for regulatory acceptance, it will require their assessment in large numbers of patients who are receiving a wide range of compounds and who develop a broad spectrum of liver injuries. The ongoing and evolving international biomarker consortia should play a major role in this effort, which is likely to transform the assessment of liver safety in clinical trials and in the clinic.
Collapse
Affiliation(s)
- Rachel J Church
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Paul B Watkins
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
Taylor DL, Gough A, Schurdak ME, Vernetti L, Chennubhotla CS, Lefever D, Pei F, Faeder JR, Lezon TR, Stern AM, Bahar I. Harnessing Human Microphysiology Systems as Key Experimental Models for Quantitative Systems Pharmacology. Handb Exp Pharmacol 2019; 260:327-367. [PMID: 31201557 PMCID: PMC6911651 DOI: 10.1007/164_2019_239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Two technologies that have emerged in the last decade offer a new paradigm for modern pharmacology, as well as drug discovery and development. Quantitative systems pharmacology (QSP) is a complementary approach to traditional, target-centric pharmacology and drug discovery and is based on an iterative application of computational and systems biology methods with multiscale experimental methods, both of which include models of ADME-Tox and disease. QSP has emerged as a new approach due to the low efficiency of success in developing therapeutics based on the existing target-centric paradigm. Likewise, human microphysiology systems (MPS) are experimental models complementary to existing animal models and are based on the use of human primary cells, adult stem cells, and/or induced pluripotent stem cells (iPSCs) to mimic human tissues and organ functions/structures involved in disease and ADME-Tox. Human MPS experimental models have been developed to address the relatively low concordance of human disease and ADME-Tox with engineered, experimental animal models of disease. The integration of the QSP paradigm with the use of human MPS has the potential to enhance the process of drug discovery and development.
Collapse
Affiliation(s)
- D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark E Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chakra S Chennubhotla
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Lefever
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Fen Pei
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Faeder
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy R Lezon
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Chung J, Longo DM, Watkins PB. A Rapid Method to Estimate Hepatocyte Loss Due to Drug‐Induced Liver Injury. Clin Pharmacol Ther 2018; 105:746-753. [DOI: 10.1002/cpt.1254] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/15/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Jae‐Yong Chung
- Department of Clinical Pharmacology and Therapeutics Seoul National University College of Medicine and Bundang Hospital Seongnam Korea
| | - Diane M. Longo
- DILIsym Services, Inc. Research Triangle Park North Carolina USA
| | - Paul B. Watkins
- Institute for Drug Safety Sciences Eshelman School of Pharmacy University of North Carolina Chapel Hill North Carolina USA
| |
Collapse
|
13
|
Barnhill MS, Real M, Lewis JH. Latest advances in diagnosing and predicting DILI: what was new in 2017? Expert Rev Gastroenterol Hepatol 2018; 12:1033-1043. [PMID: 30111182 DOI: 10.1080/17474124.2018.1512854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Drug-induced liver injury (DILI) remains an increasingly recognized cause of hepatotoxicity and liver failure worldwide. In 2017, we continued to learn about predicting, diagnosing, and prognosticating drug hepatotoxicity. Areas covered: In this review, we selected from over 1200 articles from 2017 to synopsize updates in DILI. There were new HLA haplotypes associated with medications including HLA-C0401 and HLA-B*14. There has been continued work with quantitative systems pharmacology, particularly with the DILIsym® initiative, which employs mathematical representations of DILI mechanisms to predict hepatotoxicity in simulated populations. Additionally, knowledge regarding microRNAs (miRNAs) continues to expand. Some new miRNAs this past year include miRNA-223 and miRNA-605. Aside from miRNAs, other biomarkers for diagnosis, prognosis, and even prediction of DILI were explored. Studies on K18, OPN, and MCSFR have correlated DILI and liver-associated death within 6 months. Conversely, a new prognostic panel using apolipoportein-A1 and haptoglobin has been proposed to predict recovery. Further study of CDH5 has also provided researchers a possible new biomarker for prediction and susceptibility to DILI. Expert commentary: Although research on DILI remains quite promising, there is yet to be a reliable, simple method to predict, diagnose, and risk assess this form of hepatotoxicity.
Collapse
Affiliation(s)
- Michele S Barnhill
- a Department of Internal Medicine , Medstar Georgetown University Hospital , Washington , DC , USA
| | - Mark Real
- a Department of Internal Medicine , Medstar Georgetown University Hospital , Washington , DC , USA
| | - James H Lewis
- b Department of Gastroenterology , Medstar Georgetown University Hospital , Washington , DC , USA
| |
Collapse
|
14
|
Battista C, Howell BA, Siler SQ, Watkins PB. An Introduction to DILIsym® Software, a Mechanistic Mathematical Representation of Drug-Induced Liver Injury. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-1-4939-7677-5_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
15
|
Church RJ, Watkins PB. In silico modeling to optimize interpretation of liver safety biomarkers in clinical trials. Exp Biol Med (Maywood) 2017; 243:300-307. [PMID: 29096561 DOI: 10.1177/1535370217740853] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Current strategies to delineate the risk of serious drug-induced liver injury associated with drugs rely on assessment of serum biomarkers that have been utilized for many decades. In particular, serum alanine aminotransferase and total bilirubin levels are typically used to assess hepatic integrity and function, respectively. Parallel measurement of these biomarkers is utilized to identify patients with drug-induced hepatocellular jaundice ("Hy's Law" cases) which carries at least a 10% risk of death or liver transplant. However, current guidelines regarding use of these biomarkers in clinical trials can put study subjects at risk for life-threatening drug-induced liver injury, or result in over estimation of risk that may halt development of safe drugs. In addition, pharmaceutical companies are increasingly being required to conduct large and expensive clinical trials to "defend" the safety of their new drug when results from smaller trials are inconclusive. Innovative approaches and some novel biomarkers are now being employed to maximize the value of traditional biochemical tests. DILIsym®, a product of the DILIsim Initiative, utilizes serial serum alanine aminotransferase values, along with serum biomarkers of apoptosis vs necrosis, to estimate percent hepatocyte loss and total bilirubin elevations resulting from loss of global liver function. The results from analyses conducted with DILIsym have been reported to the FDA to support the safety of entolimod and cimaglermin alfa after elevations in serum alanine aminotransferase and/or bilirubin halted clinical development. DILIsym can also be utilized to determine whether rises in serum conjugated and unconjugated bilirubin are consistent with mechanisms unrelated to toxicity ( i.e. inhibition of bilirubin transport or metabolism). In silico modeling of traditional and novel drug-induced liver injury biomarker data obtained in clinical trials may be the most efficient and accurate way to define the liver safety profile of new drug candidates. Impact statement Blood tests used in clinical trials to detect and monitor drug-induced liver injury (DILI) have not changed in half a century. These tests have several shortcomings: their use has not completely prevented clinical trial participants from risk of life-threatening DILI, they can give false positive results that halt the development of safe drug candidates, and they can create liver safety "concerns" that require large additional clinical trials to accurately define DILI risk. This review highlights the use of in silico modeling to improve interpretation of the blood tests currently available to detect DILI risk in new drug candidates. This approach is increasingly being applied in clinical trials to more precisely assess the degree of hepatocellular injury and its functional impact. This new approach holds the promise of more accurately defining DILI risk in smaller clinical trials.
Collapse
Affiliation(s)
- Rachel J Church
- 1 UNC Institute for Drug Safety Sciences, 2331 Research Triangle Park , NC 27709, USA.,2 Division of Pharmacotherapy and Experimental Therapeutics, 15521 UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill , NC 27599, USA
| | - Paul B Watkins
- 1 UNC Institute for Drug Safety Sciences, 2331 Research Triangle Park , NC 27709, USA.,2 Division of Pharmacotherapy and Experimental Therapeutics, 15521 UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill , NC 27599, USA
| |
Collapse
|
16
|
Church RJ, Watkins PB. The transformation in biomarker detection and management of drug-induced liver injury. Liver Int 2017; 37:1582-1590. [PMID: 28386997 PMCID: PMC5632128 DOI: 10.1111/liv.13441] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury (DILI) is a major concern for patients, care givers and the pharmaceutical industry. Interpretation of the serum biomarkers routinely used to detect and monitor DILI, which have not changed in almost 50 years, can be improved with recently proposed models employing quantitative systems pharmacology. In addition, several newer serum biomarkers are showing great promise. Studies in rodents indicate that the ratio of the caspase cleaved fragment of cytokeratin 18 to total K18 in serum (termed the "apoptotic index") estimates the relative proportions of apoptosis vs necrosis during drug-induced liver injury. Glutamate dehydrogenase can reliably differentiate liver from muscle injury and, when serum is properly prepared, may also detect mitochondrial toxicity as a mechanism of liver injury. MicroRNA-122 is liver-specific, but recent data suggests it can be actively released from hepatocytes in the absence of overt toxicity limiting enthusiasm for it as a DILI biomarker. Finally, damage associated molecular patterns, particularly high mobility group box 1 and its various modified forms, are promising biomarkers of innate immune activation, which may be useful in distinguishing benign elevations in aminotransferases from those that portend clinically important liver injury. These new biomarkers are already being measured in early clinical trials, but broad acceptance will require widespread archiving of serum from diverse clinical trials and probably pre-competitive analysis efforts. We believe that utilization of a panel of traditional and newer biomarkers in conjunction with quantitative systems pharmacology modelling approaches will transform DILI detection and risk management.
Collapse
Affiliation(s)
- Rachel J. Church
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Paul B. Watkins
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| |
Collapse
|
17
|
Longo DM, Generaux GT, Howell BA, Siler SQ, Antoine DJ, Button D, Caggiano A, Eisen A, Iaci J, Stanulis R, Parry T, Mosedale M, Watkins PB. Refining Liver Safety Risk Assessment: Application of Mechanistic Modeling and Serum Biomarkers to Cimaglermin Alfa (GGF2) Clinical Trials. Clin Pharmacol Ther 2017; 102:961-969. [PMID: 28419467 PMCID: PMC5697568 DOI: 10.1002/cpt.711] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/14/2017] [Accepted: 04/09/2017] [Indexed: 02/06/2023]
Abstract
Cimaglermin alfa (GGF2) is a recombinant human protein growth factor in development for heart failure. Phase I trials were suspended when two cimaglermin alfa‐treated subjects experienced concomitant elevations in serum aminotransferases and total bilirubin, meeting current US Food and Drug Administration criteria for a serious liver safety signal (i.e., “Hy's Law”). We assayed mechanistic biomarkers in archived clinical trial serum samples which confirmed the hepatic origin of the aminotransferase elevations in these two subjects and identified apoptosis as the major mode of hepatocyte death. Using a mathematical model of drug‐induced liver injury (DILIsym) and a simulated population, we estimated that the maximum hepatocyte loss in these two subjects was <13%, which would not result in liver dysfunction sufficient to significantly increase serum bilirubin levels. We conclude that the two subjects should not be considered Hy's Law cases and that mechanistic biomarkers and modeling can aid in refining liver safety risk assessment in clinical trials.
Collapse
Affiliation(s)
- D M Longo
- DILIsym Services Inc., Research Triangle Park, North Carolina, USA
| | - G T Generaux
- DILIsym Services Inc., Research Triangle Park, North Carolina, USA
| | - B A Howell
- DILIsym Services Inc., Research Triangle Park, North Carolina, USA
| | - S Q Siler
- DILIsym Services Inc., Research Triangle Park, North Carolina, USA
| | - D J Antoine
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, Liverpool University, Liverpool, UK
| | - D Button
- Acorda Therapeutics Inc., New York, New York, USA
| | - A Caggiano
- Acorda Therapeutics Inc., New York, New York, USA
| | - A Eisen
- Acorda Therapeutics Inc., New York, New York, USA
| | - J Iaci
- Acorda Therapeutics Inc., New York, New York, USA
| | - R Stanulis
- Acorda Therapeutics Inc., New York, New York, USA
| | - T Parry
- Acorda Therapeutics Inc., New York, New York, USA
| | - M Mosedale
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - P B Watkins
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| |
Collapse
|
18
|
|
19
|
Abd El Monein Solaiman A, Mahmoud Elagawany A. Histological study of adult male albino rats’ hepatocytes after formaldehyde administration and the possible protective role of dill oil. THE EGYPTIAN JOURNAL OF HISTOLOGY 2015; 38:493-503. [DOI: 10.1097/01.ehx.0000470835.13588.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
20
|
Singh H, Prakash A, Kalia AN, Majeed ABA. Synergistic hepatoprotective potential of ethanolic extract of Solanum xanthocarpum and Juniperus communis against paracetamol and azithromycin induced liver injury in rats. J Tradit Complement Med 2015; 6:370-376. [PMID: 27774421 PMCID: PMC5067840 DOI: 10.1016/j.jtcme.2015.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 07/20/2015] [Accepted: 07/28/2015] [Indexed: 11/18/2022] Open
Abstract
Previously explored combination therapies mostly involved the use of bioactive molecules. It is believed that herbal compounds containing multiple plant products have synergistic hepatoprotective effects and could enhance the desired actions. To investigate the combination of ethanolic fruits extract of Solanum xanthocarpum (SX) and Juniperus communis (JC) against Paracetamol (PCM) and Azithromycin (AZM) induced liver toxicity in rats. Liver toxicity was induced by combine oral administration of PCM (250 mg/kg) and AZM (200 mg/kg) for 7 days in Wistar rats. Fruit extract of SX (200 and 400 mg/kg) and JC (200 and 400 mg/kg) were administered daily for 14 days. The hepatoprotective activity was assessed using liver functional test, oxidative parameters and histopathological examination. The results demonstrated that combine administration of AZM and PCM significantly produced liver toxicity by increasing the serum level of hepatic enzymes and oxidative parameters in liver of rats. Histopathological examination also indicated that AZM and PCM produced liver damage in rats. Chronic treatment of SX and JC extract significantly and dose-dependently attenuated the liver toxicity by normalizing the biochemical factors and no gross histopathological changes were observed in liver of rats. Furthermore, combine administration of lower dose of SX and JC significantly potentiated their hepatoprotective effect which was significant as compared to their effect per se. The results clearly indicated that SX and JC extract has hepatoprotective potential against AZM and PCM induced liver toxicity due to their synergistic anti-oxidant properties.
Collapse
Affiliation(s)
- Hem Singh
- Department of Pharmacology, Indo Soviet Friendship (ISF) College of Pharmacy, Moga 142001, Punjab, India; Dr Rajendra Prasad Government Medical College, Tanda Hospital Rd, Kangra, Himachal Pradesh 176001, India
| | - Atish Prakash
- Department of Pharmacology, Indo Soviet Friendship (ISF) College of Pharmacy, Moga 142001, Punjab, India; Faculty of Pharmacy, Campus Puncak Alam, Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - A N Kalia
- Department of Pharmacology, Indo Soviet Friendship (ISF) College of Pharmacy, Moga 142001, Punjab, India
| | - Abu Bakar Abdul Majeed
- Faculty of Pharmacy, Campus Puncak Alam, Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
21
|
Yang K, Woodhead JL, Shoda LK, Yang Y, Watkins PB, Brouwer KL, Howell BA, Siler SQ. Mechanistic Modeling of Drug‐Induced Liver Injury (DILI). METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2015:173-198. [DOI: 10.1002/9783527673643.ch09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Al- Daihan S, Shafi Bhat R. Impact of Propionic Acid on Liver Damage in Rats. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2015; 4:188-95. [PMID: 26629488 PMCID: PMC4644531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Propionic acid (PA) is a short chain fatty acid, a common food preservative and metabolic end product of enteric bacteria in the gut. The present study was undertaken to investigate the effect of PA on liver injury in male rats. Male western albino rats were divided into two groups. The first group served as normal control, the second was treated with PA. The activities of serum hepatospecific markers such as aspartate transaminase, alanine transaminase, and alkaline phosphatase were estimated. Antioxidant status in liver tissues was estimated by determining the level of lipid peroxidation and activities of enzymatic and non-enzymatic antioxidants. Sodium and potassium levels were also measured in liver tissue. PA treatment caused significant changes in all hepatospecific markers. Biochemical analysis of liver homogenates from PA-treated rats showed an increase in oxidative stress markers like lipid peroxidation and lactate dehydrogenase, coupled with a decrease in glutathione, vitamin C and glutathione S- transferase. However, PA exposure caused no change in sodium and potassium levels in liver tissue. Our study demonstrated that PA persuade hepatic damage in rats.
Collapse
Affiliation(s)
| | - Ramesa Shafi Bhat
- Corresponding author: Biochemistry Department, Science College, King Saud University, P,O box 22452, Zip code 11495, Riyadh, Saudi Arabia. E-mail: ,
| |
Collapse
|
23
|
Woodhead JL, Yang K, Siler SQ, Watkins PB, Brouwer KLR, Barton HA, Howell BA. Exploring BSEP inhibition-mediated toxicity with a mechanistic model of drug-induced liver injury. Front Pharmacol 2014; 5:240. [PMID: 25426072 PMCID: PMC4224072 DOI: 10.3389/fphar.2014.00240] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/22/2014] [Indexed: 01/15/2023] Open
Abstract
Inhibition of the bile salt export pump (BSEP) has been linked to incidence of drug-induced liver injury (DILI), presumably by the accumulation of toxic bile acids in the liver. We have previously constructed and validated a model of bile acid disposition within DILIsym®, a mechanistic model of DILI. In this paper, we use DILIsym® to simulate the DILI response of the hepatotoxic BSEP inhibitors bosentan and CP-724,714 and the non-hepatotoxic BSEP inhibitor telmisartan in humans in order to explore whether we can predict that hepatotoxic BSEP inhibitors can cause bile acid accumulation to reach toxic levels. We also simulate bosentan in rats in order to illuminate potential reasons behind the lack of toxicity in rats compared to the toxicity observed in humans. DILIsym® predicts that bosentan, but not telmisartan, will cause mild hepatocellular ATP decline and serum ALT elevation in a simulated population of humans. The difference in hepatotoxic potential between bosentan and telmisartan is consistent with clinical observations. However, DILIsym® underpredicts the incidence of bosentan toxicity. DILIsym® also predicts that bosentan will not cause toxicity in a simulated population of rats, and that the difference between the response to bosentan in rats and in humans is primarily due to the less toxic bile acid pool in rats. Our simulations also suggest a potential synergistic role for bile acid accumulation and mitochondrial electron transport chain (ETC) inhibition in producing the observed toxicity in CP-724,714, and suggest that CP-724,714 metabolites may also play a role in the observed toxicity. Our work also compares the impact of competitive and noncompetitive BSEP inhibition for CP-724,714 and demonstrates that noncompetitive inhibition leads to much greater bile acid accumulation and potential toxicity. Our research demonstrates the potential for mechanistic modeling to contribute to the understanding of how bile acid transport inhibitors cause DILI.
Collapse
Affiliation(s)
- Jeffrey L Woodhead
- The Hamner-UNC Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences Research Triangle Park, NC, USA
| | - Kyunghee Yang
- The Hamner-UNC Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences Research Triangle Park, NC, USA
| | - Scott Q Siler
- The Hamner-UNC Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences Research Triangle Park, NC, USA
| | - Paul B Watkins
- The Hamner-UNC Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences Research Triangle Park, NC, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC-Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Hugh A Barton
- Pharmacokinetics, Dynamics, and Metabolism, Worldwide Research and Development, Pfizer, Inc. Groton CT, USA
| | - Brett A Howell
- The Hamner-UNC Institute for Drug Safety Sciences, The Hamner Institutes for Health Sciences Research Triangle Park, NC, USA
| |
Collapse
|
24
|
Senthilkumar R, Chandran R, Parimelazhagan T. Hepatoprotective effect of Rhodiola imbricata rhizome against paracetamol-induced liver toxicity in rats. Saudi J Biol Sci 2014; 21:409-16. [PMID: 25313275 PMCID: PMC4191600 DOI: 10.1016/j.sjbs.2014.04.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 01/20/2023] Open
Abstract
Rhodiola imbricata is a perennial herb of the family Crassulaceae, which has significant traditional usage as medicine and is also known to biosynthesize phytochemicals such as flavonoids, coumarins and phenyl glycosides. The present investigation was aimed to estimate the hepatoprotective activity of R. imbricata rhizome acetone extract against paracetamol (2 g/kg) induced liver toxicity. Paracetamol was administered to induce hepatic damage in Wistar rats. 200 and 400 mg/kg doses of rhizome acetone extract and silymarin (25 mg/kg) were used as treatment groups. The blood samples were analyzed for biochemical markers of hepatic injury and tissue samples were subjected for estimation of liver antioxidants and histopathological studies. Analysis of the extract treated rats (400 mg/kg) showed an elevation of superoxide dismutase (0.326 units/min/mg protein), catalase (185.03 μmole of H2O2 consumed/min/mg protein), glutothione peroxidase (19.26 mg GSH consumed/min/mg protein) and reduced glutathione (16.2 μmole of GSH/mg protein). Moreover, the biochemical parameters in serum like alkaline phosphatase, serum glutamic oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT) and lipid profiles were also improved in treated groups compared to the control. The oral administration of different doses of rhizome acetone extract significantly protected the hepatic cells from damage. The hematological and biochemical parameters were also normal in extract treated rats compared to the control and standard (silymarin) groups. The HPLC analysis revealed the presence of some important phenolic compounds which could be responsible for the hepatoprotective activity. This study proved that R. imbricata could be taken as a good natural source of the hepatoprotective agent.
Collapse
Affiliation(s)
| | | | - Thangaraj Parimelazhagan
- Department of Botany, Bioprospecting Laboratory, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|