1
|
Choi MA, Rose S, Langouët S. Per- and polyfluoroalkyl substances as potentiators of hepatotoxicity in an exposome framework: Current challenges of environmental toxicology. Toxicology 2025; 515:154167. [PMID: 40300710 DOI: 10.1016/j.tox.2025.154167] [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: 02/05/2025] [Revised: 04/17/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Chronic liver diseases, including metabolic dysfunction-associated steatosic liver disease (MASLD) and hepatocellular carcinoma (HCC), are on the rise, potentially due to daily exposure to complex mixtures of chemical compounds forming part of the exposome. Understanding the mechanisms involved in hepatotoxicity of these mixtures is essential to identify common molecular targets that may highlight potential interactions at the molecular level. We illustrated this issue with two families of environmental contaminants, per- and polyfluoroalkyl substances (PFAS) and heterocyclic aromatic amines (HAAs), both of which could be involved in the progression of chronic liver diseases, and whose toxicity may be potentiated by interactions at the level of xenobiotic metabolism. In the study of exposome effects on chronic liver disease, New Approach Methodologies (NAMs) including omics analyses and data from various in vitro, in vivo and in silico approaches, are crucial for improving predictivity of toxicological studies in humans while reducing animal experimentation. Additionally, the development of complex in vitro human liver cell models, such as organoids, is essential to avoid interspecies differences that minimize the risk for humans. All together, these approaches will contribute to construct Adverse Outcome Pathways (AOPs) and could be applied not only to PFAS mixtures but also to other chemical families, providing valuable insights into mixture hepatotoxicity prediction in the study of the exposome. A better understanding of toxicological mechanisms will clarify the role of environmental contaminant mixtures in the development of MASLD and HCC, supporting risk assessment for better treatment, monitoring and prevention strategies.
Collapse
Affiliation(s)
- Minna A Choi
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France
| | - Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France.
| |
Collapse
|
2
|
Subramaniam V, Abrahan C, Higgins BR, Chisolm SJ, Sweeney B, Duraivel S, Balzano-Nogueira L, Monjure T, Wang CY, Palmer GD, Angelini TE. A functional human liver tissue model: 3D bioprinted co-culture discoids. BIOMATERIALS ADVANCES 2025; 173:214288. [PMID: 40106895 DOI: 10.1016/j.bioadv.2025.214288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
To reduce costs and delays related to developing new and effective drugs, there is a critical need for improved human liver tissue models. Here we describe an approach for 3D bioprinting functional human liver tissue models, in which we fabricate disc-shaped structures (discoids) 200 μm in thickness and 1-3 mm in diameter from mixtures of cells and collagen-1, embedded in a highly permeable support medium made from packed polyethylene glycol (PEG) microgels. We demonstrate that the method is precise, accurate, and scalable; up to 100 tissues/h can be manufactured with a variability and error in diameter of about 4 %. Histologic and immunohistochemical evaluation of printed discs reveal self-organization, cell cohesion, and key liver marker expression. Over the course of three weeks in culture, the tissues stably synthesize albumin and urea at high levels, outperforming spheroid tissue models. We find the tissues express >100 genes associated with molecular absorption, distribution, metabolism, and excretion (ADME) at levels within the range of human liver. The liver tissue models exhibit enzymatic formation of metabolites after exposure to multiple test compounds. Together, these results demonstrate the promise of 3D printed discoids for pharmacological and toxicological applications.
Collapse
Affiliation(s)
- Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Carolina Abrahan
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, FL, United States of America
| | - Brett R Higgins
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States of America
| | - Steven J Chisolm
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Baleigh Sweeney
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Senthilkumar Duraivel
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, United States of America
| | - Leandro Balzano-Nogueira
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States of America
| | - Tia Monjure
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Chih-Yi Wang
- Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Glyn D Palmer
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America; Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America; J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
3
|
Pike CM, Levi JA, Boone LA, Peddibhotla S, Johnson J, Zwarycz B, Bunger MK, Thelin W, Boazak EM. High-throughput assay for predicting diarrhea risk using a 2D human intestinal stem cell-derived model. Toxicol In Vitro 2025; 106:106040. [PMID: 40086646 DOI: 10.1016/j.tiv.2025.106040] [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: 10/09/2024] [Revised: 01/29/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Gastrointestinal toxicities (GITs) in clinical trials often lead to dose-limitations that reduce drug efficacy and delay treatment optimization. Preclinical animal models do not accurately replicate human physiology, leaving few options for early detection of GITs, such as diarrhea, before human studies. Chemotherapeutic agents, known to cause clinical diarrhea, frequently target mitotic cells. Therefore, we hypothesized a model utilizing proliferative cell populations derived from human intestinal crypts would predict clinical diarrhea occurrence with high accuracy. Here, we describe the development of a diarrhea prediction assay utilizing RepliGut® Planar, a primary intestinal stem cell-derived platform. To evaluate the ability of this model to predict clinical diarrhea risk, we assessed toxicity of 30 marketed drugs by measuring cell proliferation (EdU incorporation), cell abundance (nuclei quantification), and barrier formation (TEER) in cells derived from three human donors. Dose response curves were generated for each drug, and the IC15 to Cmax ratio was used to identify a threshold for assay positivity. This model accurately predicted diarrhea potential, achieving an accuracy of 91 % for proliferation, 90 % for abundance, and 88 % for barrier formation. In vitro toxicity screening using primary proliferative cells may reduce clinical diarrhea and ultimately lead to safer and more effective treatments for patients.
Collapse
|
4
|
Hu Y, Ma W, Xu G, Zhao W, Yu Z. Valorization of crocodile head for anti-inflammatory peptides: In silico screening and cellular validation. Food Res Int 2025; 211:116457. [PMID: 40356184 DOI: 10.1016/j.foodres.2025.116457] [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: 11/14/2024] [Revised: 03/14/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
In this study, two novel anti-inflammatory peptides were identified from the hydrolysates of crocodile heads using computer-assisted techniques and in vitro cellular experiments. Their interaction mechanisms were elucidated through molecular docking and molecular dynamics simulations. A total of 67 peptides were identified via LC-MS/MS, with AKLDLEEVIK and DFLDLPSIER emerging as promising candidates for anti-inflammatory activity. Results of in vitro cellular experiments demonstrated that these peptides significantly inhibited LPS-stimulated macrophage hyperactivation, resulting in reduced release of nitric oxide (NO) and pro-inflammatory cytokines, i.e., TNF-α, IL-1β, and IL-6. Molecular dynamics (MD) simulations and docking analyses revealed that AKLDLEEVIK and DFLDLPSIER displayed high affinity for the TLR4-MD2 complex, with stable and tight interactions. Key residues Arg90, Ser118, Cys133, and Arg264 were identified as critical for the recognition and interaction between the peptides and TLR4-MD2. This study provides a theoretical foundation for the development of anti-inflammatory functional foods derived from crocodile heads.
Collapse
Affiliation(s)
- Yongyong Hu
- School of Food Science and Engineering, Hainan University, Haikou 570228, PR China
| | - Wenhao Ma
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Ge Xu
- School of Food Science and Engineering, Hainan University, Haikou 570228, PR China
| | - Wenzhu Zhao
- School of Food Science and Engineering, Hainan University, Haikou 570228, PR China.
| | - Zhipeng Yu
- School of Food Science and Engineering, Hainan University, Haikou 570228, PR China
| |
Collapse
|
5
|
Lv W, Zhang X, Zhang H, Alitanhua, Xiao Y. Safety of Nutmeg powder by oral exposure: Toxicity prediction and in vivo evaluation. Food Chem Toxicol 2025; 200:115364. [PMID: 40024564 DOI: 10.1016/j.fct.2025.115364] [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: 12/18/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/04/2025]
Abstract
Nutmeg (Myristica fragrans Houtt.) is widely cultivated in tropical regions and valued for its culinary and medicinal uses. However, its safety profile remains underexplored. This study aimed to evaluate the safety of nutmeg powder through in-silico toxicity predictions, acute oral toxicity, and a 13-week repeated-dose toxicity assessment. In the in-silico study, bioactive compounds identified via High-Performance Liquid Chromatography (HPLC) were analyzed for toxicity parameters using the ProTox II server. For acute toxicity, a single dose of nutmeg powder was administered to mice, with no mortality observed over 14 days, and an LD50 greater than 5000 mg/kg body weight was determined. In the sub-chronic toxicity study, rats received nutmeg powder at 1400 mg/kg and 3500 mg/kg (35 and 87.5 times the human therapeutic dosage, respectively) for 90 days. A 28-day recovery phase was included to assess delayed or reversible effects. The results indicate that a dose of 1400 mg/kg leads to liver damage and hemosiderin deposition in the spleen, while a dose of 3500 mg/kg causes liver and kidney damage, as well as hemosiderin deposition in the spleen. These findings highlight the potential toxicity of long-term nutmeg consumption and underscore the need for further research to ensure its safety in preclinical.
Collapse
Affiliation(s)
- Wei Lv
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Xin Zhang
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Hongli Zhang
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Alitanhua
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Yunfeng Xiao
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China; Key Laboratory of Drug Safety Evaluation, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China.
| |
Collapse
|
6
|
O'Mahony ET, Arian CM, Aryeh KS, Wang K, Thummel KE, Kelly EJ. Human intestinal enteroids: Nonclinical applications for predicting oral drug disposition, toxicity, and efficacy. Pharmacol Ther 2025:108879. [PMID: 40398537 DOI: 10.1016/j.pharmthera.2025.108879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/19/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
The application of human enteroid systems presents a significant opportunity within the drug development pipeline, highlighting considerable potential for advancements in the characterization and evaluation of new molecular entities. Derived from LGR5+ crypt-based columnar cells, enteroid systems more accurately recapitulate the microanatomy and physiological processes of the human intestinal mucosa compared to traditionally used systems. They contain the complement of major mucosal epithelial cell types, maintain the genetic identity of the donor and intestinal segment they were derived from, and exhibit biological functions and specific activities that are seen in vivo. In this review, we examine the applications of human enteroid systems in nonclinical drug development and compare findings to existing and emerging in vitro models of the small intestine. Specifically, we explore enteroid systems in the context of predicting oral drug disposition, focusing on apparent permeability, intestinal first-pass metabolism, and drug interactions, as well as their utility in assessing drug-induced gastrointestinal toxicity and screening therapeutic efficacy against enteric diseases. Additionally, we highlight aspects of enteroid systems that warrant further study.
Collapse
Affiliation(s)
- Eimear T O'Mahony
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Christopher M Arian
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Kayenat S Aryeh
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Kai Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America; Center of Excellence for Natural Product Drug Interaction Research, Spokane, WA, United States of America
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America; Kidney Research Institute, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
7
|
Seal S, Mahale M, García-Ortegón M, Joshi CK, Hosseini-Gerami L, Beatson A, Greenig M, Shekhar M, Patra A, Weis C, Mehrjou A, Badré A, Paisley B, Lowe R, Singh S, Shah F, Johannesson B, Williams D, Rouquie D, Clevert DA, Schwab P, Richmond N, Nicolaou CA, Gonzalez RJ, Naven R, Schramm C, Vidler LR, Mansouri K, Walters WP, Wilk DD, Spjuth O, Carpenter AE, Bender A. Machine Learning for Toxicity Prediction Using Chemical Structures: Pillars for Success in the Real World. Chem Res Toxicol 2025; 38:759-807. [PMID: 40314361 DOI: 10.1021/acs.chemrestox.5c00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Machine learning (ML) is increasingly valuable for predicting molecular properties and toxicity in drug discovery. However, toxicity-related end points have always been challenging to evaluate experimentally with respect to in vivo translation due to the required resources for human and animal studies; this has impacted data availability in the field. ML can augment or even potentially replace traditional experimental processes depending on the project phase and specific goals of the prediction. For instance, models can be used to select promising compounds for on-target effects or to deselect those with undesirable characteristics (e.g., off-target or ineffective due to unfavorable pharmacokinetics). However, reliance on ML is not without risks, due to biases stemming from nonrepresentative training data, incompatible choice of algorithm to represent the underlying data, or poor model building and validation approaches. This might lead to inaccurate predictions, misinterpretation of the confidence in ML predictions, and ultimately suboptimal decision-making. Hence, understanding the predictive validity of ML models is of utmost importance to enable faster drug development timelines while improving the quality of decisions. This perspective emphasizes the need to enhance the understanding and application of machine learning models in drug discovery, focusing on well-defined data sets for toxicity prediction based on small molecule structures. We focus on five crucial pillars for success with ML-driven molecular property and toxicity prediction: (1) data set selection, (2) structural representations, (3) model algorithm, (4) model validation, and (5) translation of predictions to decision-making. Understanding these key pillars will foster collaboration and coordination between ML researchers and toxicologists, which will help to advance drug discovery and development.
Collapse
Affiliation(s)
- Srijit Seal
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Manas Mahale
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Mumbai 400098, India
| | | | - Chaitanya K Joshi
- Department of Computer Science and Technology, University of Cambridge, Cambridge CB3 0FD, U.K
| | | | - Alex Beatson
- Axiom Bio, San Francisco, California 94107, United States
| | - Matthew Greenig
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Mrinal Shekhar
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | | | | | | | - Adrien Badré
- Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Brianna Paisley
- Eli Lilly & Company, Indianapolis, Indiana 46285, United States
| | | | - Shantanu Singh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Falgun Shah
- Non Clinical Drug Safety, Merck Inc., West Point, Pennsylvania 19486, United States
| | | | | | - David Rouquie
- Toxicology Data Science, Bayer SAS Crop Science Division, Valbonne Sophia-Antipolis 06560, France
| | - Djork-Arné Clevert
- Pfizer, Worldwide Research, Development and Medical, Machine Learning & Computational Sciences, Berlin 10922, Germany
| | | | | | - Christos A Nicolaou
- Computational Drug Design, Digital Science & Innovation, Novo Nordisk US R&D, Lexington, Massachusetts 02421, United States
| | - Raymond J Gonzalez
- Non Clinical Drug Safety, Merck Inc., West Point, Pennsylvania 19486, United States
| | - Russell Naven
- Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | | | | | - Kamel Mansouri
- NIH/NIEHS/DTT/NICEATM, Research Triangle Park, North Carolina 27709, United States
| | | | | | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala 751 24, Sweden
- Phenaros Pharmaceuticals AB, Uppsala 75239, Sweden
| | - Anne E Carpenter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andreas Bender
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
8
|
Zhou Y, Zhong Y, Lauschke VM. Evaluating the synergistic use of advanced liver models and AI for the prediction of drug-induced liver injury. Expert Opin Drug Metab Toxicol 2025; 21:563-577. [PMID: 39893552 DOI: 10.1080/17425255.2025.2461484] [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: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Drug-induced liver injury (DILI) is a leading cause of acute liver failure. Hepatotoxicity typically occurs only in a subset of individuals after prolonged exposure and constitutes a major risk factor for the termination of drug development projects. AREAS COVERED We provide an overview of available human liver models for DILI research and discuss how they have been used to aid in early risk assessments and to mitigate the risk of project closures due to DILI in clinical stages. We summarize the different data that can be provided by such models and illustrate how these diverse data types can be interfaced with machine learning strategies to improve predictions of liver safety liabilities. EXPERT OPINION Advanced human liver models closely mimic human liver phenotypes and functions for many weeks, allowing for the recapitulation of hepatotoxicity events in vitro. Integration of the biochemical, histological, and toxicogenomic output data from these models with physicochemical compound properties using different machine learning architectures holds promise to enhance preclinical DILI predictions. However, to realize this aim, it is important to benchmark the available liver models on test sets of DILI positive and negative compounds and to carefully annotate and share the resulting data.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Yi Zhong
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Liao J, Lu B, Yang J, Wang X, Li S, Fu H, Gao F. In vivo toxic and lethal cardiorespiratory effects of a synthetic quaternary ammonium salt derivative of haloperidol in mice. Animal Model Exp Med 2025; 8:842-853. [PMID: 39853902 DOI: 10.1002/ame2.12531] [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: 11/08/2024] [Accepted: 12/10/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND To investigate the toxicity of N-n-butyl haloperidol iodide (F2), a quaternary ammonium salt derivative of haloperidol, in mice for potential therapeutic purposes. METHODS The acute median lethal dose (LD50) of F2 was determined using the Bliss method following intravenous administration in mice. Routine surface electrocardiograms (ECGs) and arterial blood pressures (aBPs) were recorded under general anesthesia in untreated and pharmacologically vagotomized mice injected with F2. Sublethal doses of F2 were tested for their effects on aBP, heart rate, and biochemical parameters such as lactate dehydrogenase (LDH), blood urea nitrogen (BUN), and serum lactate levels. Histopathological changes in the heart, lungs, liver, and kidneys were evaluated after F2 administration. RESULTS The acute LD50 of F2 was determined to be 5.11 mg/kg. A 10 mg/kg dose of F2 caused severe hypotension, second-degree atrioventricular block, progressive prolongation of Pmurr intervals, and death due to cardiac asystole. Similar ECG and aBP changes were observed in atropine-pretreated mice, indicating that cholinergic effects do not play a major role in F2-induced toxicity. Sublethal doses of F2 (1.2 and 2.4 mg/kg) caused dose-dependent decreases in aBP and increases in heart rate. F2 induced significant, dose-dependent increases in LDH, BUN, and serum lactate levels. Histopathological analysis revealed acute lung lesions at 10 mg/kg, with no significant changes observed in the heart, liver, or kidneys. CONCLUSION Acute intravenous injection of F2 exhibits dose-dependent cardiopulmonary toxicity, characterized by severe hypotension, arrhythmias, and biochemical changes. These findings highlight the potential risks of F2 and the need for further evaluation of its safety profile for therapeutic use.
Collapse
Affiliation(s)
- Jilin Liao
- Department of Pharmacy, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| | - Binger Lu
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Department of Pharmacy, First Affiliated Hospital, Shantou University Medical College, Guangdong, China
| | - Jinhua Yang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaowan Wang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| | - Shuxian Li
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| | - Hongbo Fu
- Department of Pharmacy, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
10
|
Waikar SS, Mogg R, Baker AF, Frendl G, Topper M, Adler S, Sultana S, Zhao R, King NMP, Piccoli SP, Sauer JM, Hoffmann S, Nunes I, Sistare FD. Urinary Kidney Injury Biomarker Profiles in Healthy Individuals and After Nephrotoxic and Ischemic Injury. Clin Pharmacol Ther 2025; 117:1272-1283. [PMID: 39754474 DOI: 10.1002/cpt.3531] [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: 08/16/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Two observational studies were conducted to support an initiative to qualify translational kidney safety biomarkers as clinical drug development tools that identify tubular injury prior to changes in estimated glomerular filtration rate (eGFR). Normal healthy volunteers provided three morning spot urine collections over 4 weeks. Patients undergoing surgical resection and intrathoracic cisplatin for malignant pleural mesothelioma provided urine samples pre- and postoperatively at 4, 8, and 12 hours and daily for 6 days. Using receiver-operating characteristics curves, "statistically significant thresholds" established peak longitudinal changes for 8 biomarkers to differentiate mesothelioma patients who developed acute kidney injury (AKI) from normal healthy volunteers. We also assessed "medically significant thresholds" to differentiate mesothelioma patients who did vs. did not develop AKI. Statistically and medically significant thresholds for a fold-change from baseline of urine creatinine (UCr)-normalized values were established for 6 biomarkers: clusterin (2.2, 5.1); osteopontin (3.1, 7.1); N-acetyl-ß-D-glucosaminidase (2.7, 8.1); kidney injury molecule-1 (4.3, 7.5); cystatin C (1.8, 4.5); neutrophil gelatinase-associated lipocalin (2.9, 7.8). For urine albumin and total protein, thresholds were established based on UCr-normalized absolute values: (> upper limit normal, > 10× upper limit normal). Statistically significant thresholds for all biomarkers outperformed eGFR at discriminating mesothelioma subjects exposed to cisplatin from healthy volunteers, demonstrating their utility for enhancing safe drug development. Medically significant thresholds provide perspective on when patients begin to exhibit AKI. These studies have established guideposts for confirmatory studies with additional cohorts and nephrotoxicants to formally qualify the selected biomarkers with worldwide regulatory authorities.
Collapse
Affiliation(s)
- Sushrut S Waikar
- Department of Medicine and Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Robin Mogg
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | - Gyorgy Frendl
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | | | - Runqi Zhao
- Department of Medicine and Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Steve Hoffmann
- Foundation for the National Institutes of Health, North Bethesda, Maryland, USA
| | | | | |
Collapse
|
11
|
Li S, Bao S, Cai L, Li B, Sun Y, Sun Y. Acute and subacute toxicity evaluation of ZhenzhuXiaoji decoction in preclinical models: implications for safe clinical use. Front Pharmacol 2025; 16:1554732. [PMID: 40303917 PMCID: PMC12037581 DOI: 10.3389/fphar.2025.1554732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Background ZhenzhuXiaoji Decoction (ZZXJD) is a traditional Chinese medicine formulation composed of five herbs: Ligustrum lucidum, Curcuma zedoaria, Prunella vulgaris, Hedyotis diffusa, and Glycyrrhiza uralensis, developed for the treatment of hepatocellular carcinoma (HCC). Although early studies have demonstrated the therapeutic potential of ZZXJD, its safety profile, particularly regarding potential toxicity, remains underexplored. This study aims to evaluate both the pharmacological effects and toxicity of ZZXJD in preclinical models to determine its clinical applicability. Study design and Methods This study employed in vitro and in vivo experiments to assess the pharmacological effects and safety of ZZXJD. HHL-5 and HEK-293 cell lines were treated with ZZXJD at varying concentrations (125, 250, 500, and 1,000 μg/mL) for 24, 48, and 72 h to evaluate its effects on cell viability, apoptosis, and necrosis. Acute and subacute toxicity studies were conducted in male and female mice, including assessments of behavioral changes, body weight, organ weight, and liver/kidney functions. Additionally, routine blood tests were performed to identify potential immunostimulatory effects. Results In vitro experiments demonstrated that ZZXJD inhibited the proliferation of HHL-5 and HEK-293 cells in a dose-dependent manner and induced apoptosis and necrosis. In subacute toxicity studies, mice in the low and mid-dose groups exhibited no significant behavioral changes, whereas the high-dose group showed transient alterations in liver and kidney function markers, particularly in female mice. These changes were reversible following treatment cessation. Blood tests indicated increased lymphocyte and monocyte counts in treated male mice; however, these increases were not statistically significant. Organ weight and histopathological analyses revealed no significant signs of toxicity at therapeutic doses. Conclusion Treatment with ZZXJD at standard therapeutic dosage did not produce acute or subacute toxic effects on liver or kidney functions in vivo, suggesting its safety for continued use in cancer treatment. However, reversible abnormalities in liver and kidney function markers were observed at higher doses. Thus, regular monitoring of liver and kidney functions is recommended during clinical use, especially when higher doses are employed.
Collapse
Affiliation(s)
- Songzhe Li
- Department of Biology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shuchang Bao
- Department of Biology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lingyun Cai
- Department of Biology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Baolong Li
- Department of Biology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yue Sun
- Department of Biology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
- Nanjing Seventeen Retirement Center for Retired Cadres, Nanjing, China
| | - Yang Sun
- Department of Biology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
12
|
Han J, Zhung W, Jang I, Lee J, Kang MJ, Lee TD, Kwack SJ, Kim KB, Hwang D, Lee B, Kim HS, Kim WY, Lee S. HepatoToxicity Portal (HTP): an integrated database of drug-induced hepatotoxicity knowledgebase and graph neural network-based prediction model. J Cheminform 2025; 17:48. [PMID: 40200282 PMCID: PMC11980326 DOI: 10.1186/s13321-025-00992-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
Liver toxicity poses a critical challenge in drug development due to the liver's pivotal role in drug metabolism and detoxification. Accurately predicting liver toxicity is crucial but is hindered by scattered information sources, a lack of curation standards, and the heterogeneity of data perspectives. To address these challenges, we developed the HepatoToxicity Portal (HTP), which integrates an expert-curated knowledgebase (HTP-KB) and a state-of-the-art machine learning model for toxicity prediction (HTP-Pred). The HTP-KB consolidates hepatotoxicity data from nine major databases, carefully reviewed by hepatotoxicity experts and categorized into three levels: in vitro, in vivo, and clinical, using the Medical Dictionary for Regulatory Activities (MedDRA) terminology. The knowledgebase includes information on 8,306 chemicals. This curated dataset was used to build a hepatotoxicity prediction module by fine-tuning a GNN-based foundation model, which was pre-trained with approximately 10 million chemicals in the PubChem database. Our model demonstrated excellent performance, achieving an area under the ROC curve (AUROC) of 0.761, surpassing existing methods for hepatotoxicity prediction. The HTP is publicly accessible at https://kobic.re.kr/htp/ , offering both curated data and prediction services through an intuitive interface, thus effectively supporting drug development efforts.Scientific contributionsHTP-KB consolidates comprehensive curated information on liver toxicity gathered from nine sources. HTP-Pred utilizes advanced deep learning techniques, significantly enhancing predictive accuracy. Together, these tools provide valuable resources for researchers and practitioners in drug development, accessible through a user-friendly interface.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Bio-Information Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Wonho Zhung
- Department of Chemistry, KAIST, Daejeon, 34141, Republic of Korea
| | - Insoo Jang
- Korea Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Joongwon Lee
- Department of Chemistry, KAIST, Daejeon, 34141, Republic of Korea
| | - Min Ji Kang
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Timothy Dain Lee
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Jun Kwack
- Department Bio Health Science, College of Natural Sciences, Changwon National University, Changwon, 51140, Republic of Korea
| | - Kyu-Bong Kim
- Center for Human Risk Assessment and College of Pharmacy, Dankook University, 119 Dandae-Ro, Cheonan, 31116, Republic of Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byungwook Lee
- Korea Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Woo Youn Kim
- Department of Chemistry, KAIST, Daejeon, 34141, Republic of Korea.
| | - Sanghyuk Lee
- Department of Bio-Information Science, Ewha Womans University, Seoul, 03760, Republic of Korea.
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
13
|
Kim SY, van de Wetering M, Clevers H, Sanders K. The future of tumor organoids in precision therapy. Trends Cancer 2025:S2405-8033(25)00073-1. [PMID: 40185656 DOI: 10.1016/j.trecan.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Tumoroids are cultures of patient-derived tumor cells, which are grown in 3D in the presence of an extracellular matrix extract and specific growth factors. Tumoroids can be generated from adult as well as pediatric cancers, including epithelial cancers, sarcomas, and brain cancers. Tumoroids retain multi-omic characteristics of their corresponding tumor and recapitulate interpatient and intratumor heterogeneity. Retrospective and prospective studies have demonstrated that tumoroids predict patient responses to anticancer therapies, making them a promising tool for precision oncology. However, several challenges remain before tumoroids can be fully integrated into clinical decision-making, including success rates of tumoroid establishment and turnaround times. This review discusses the current advances, challenges, and future directions of tumoroid-based models in cancer research and precision therapy.
Collapse
Affiliation(s)
- Seok-Young Kim
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, The Netherlands; Current address: Roche Pharmaceutical Research and Early Development (pRED) of F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Karin Sanders
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Godjo A, Donald DM, Ansaldi L, Darwish IAA, Byrne JL, Kakouli-Duarte T. Effects of hexavalent chromium on the biology of Steinernema feltiae: evaluating sublethal endpoints for ecotoxicity testing. PLoS One 2025; 20:e0320329. [PMID: 40168372 PMCID: PMC11960951 DOI: 10.1371/journal.pone.0320329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/17/2025] [Indexed: 04/03/2025] Open
Abstract
Essential information about the effects of a pollutant on an ecosystem can be obtained by observing how it influences a bioindicator organism. Hexavalent chromium (Cr VI+) naturally occurs in Irish agricultural soils at levels of up to 250 mg/kg, which raises concerns about potential negative impacts on human health and the surrounding areas. This research aimed to assess the sublethal effect concentrations (up to 300 ppm) of Cr VI + on the entomopathogenic nematode (EPN) Steinernema feltiae focusing on endpoints such as nematode movement and host finding ability in contaminated sand and pathogenicity, percentage penetration, sex ratio and reproduction in Galleria mellonella. To achieve that, an Irish isolate of S. feltiae [strain SB 12 (1)], was used in all experiments. The attraction of nematodes to the insect host was tested using PVC tubes of various lengths, containing sand with various concentrations of Cr VI + (50-300 ppm in increments of 50). The replication was tenfold and the insect mortality was recorded at the end of the experiment. Results showed that there was a significant effect of Cr VI + on the pathogenicity, movement and host finding ability of the nematodes in contaminated sand, and on the percentage of penetration in an insect host. However, no significant effects among the studied Cr VI + concentrations were observed in S. feltiae reproduction in G. mellonella. Similarly, the presence of the toxicant (at low concentration of 12ppm) did not affect the growth of the nematode symbiotic bacteria in liquid and solid media (TSA and NBTA). Reproduction, unlike the other sublethal parameters tested, appeared not to be an optimal endpoint for assessing soil Cr VI + risk contamination. Overall, this study confirms the excellent potential of S. feltiae to be used as a suitable sentinel organism in assessing the risk of Cr VI + soil contamination especially in the contexts of agriculture and soil health.
Collapse
Affiliation(s)
- Anique Godjo
- Department of Applied Science, Molecular Ecology and Nematode Research Group, enviroCORE, South East Technological University, Carlow, Ireland
| | - Darren Mc Donald
- Department of Applied Science, Molecular Ecology and Nematode Research Group, enviroCORE, South East Technological University, Carlow, Ireland
| | - Lucile Ansaldi
- Department of Applied Science, Molecular Ecology and Nematode Research Group, enviroCORE, South East Technological University, Carlow, Ireland
| | - Islam A. A. Darwish
- Department of Applied Science, Molecular Ecology and Nematode Research Group, enviroCORE, South East Technological University, Carlow, Ireland
| | - John L. Byrne
- Department of Applied Science, Molecular Ecology and Nematode Research Group, enviroCORE, South East Technological University, Carlow, Ireland
| | - Thomais Kakouli-Duarte
- Department of Applied Science, Molecular Ecology and Nematode Research Group, enviroCORE, South East Technological University, Carlow, Ireland
| |
Collapse
|
15
|
Sultana N, Izawa T, Kamei T, Fujiwara S, Ito Y, Takami Y, Kuwamura M. Application of humanized mice to toxicology studies: properties of chimeric mice with humanized liver (PXB-mice) for hepatotoxicity. J Toxicol Pathol 2025; 38:183-189. [PMID: 40190627 PMCID: PMC11966125 DOI: 10.1293/tox.2024-0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/23/2025] [Indexed: 04/09/2025] Open
Abstract
Chimeric mice with humanized liver are considered a useful tool to predict drug pharmacokinetics and in vivo toxicity in humans. The PXB-mouse is one of such chimeric (humanized) mouse models with more than 70% of human hepatocytes in their liver, which can produce human albumin with human-type bile secretion and express human xenobiotic metabolizing enzymes. However, data are limited regarding the properties of such humanized mice in hepatotoxicity studies. This study aimed to explore the distinctive characteristics of chimeric PXB-mice with humanized liver that can influence susceptibility to hepatotoxicity. Morphologically, the PXB-mice have a diffuse hepatic macrovesicular and microvesicular steatosis in the transplanted human hepatocytes, which can be suppressed after human growth hormone treatment. The humanized liver of the PXB-mice has a metabolic zonation of glutamine synthetase, cytochrome P450 2E1, and argininosuccinate synthase 1, similar to normal liver in rodents and humans. The transplanted human hepatocytes in the PXB liver have a markedly decreased N-cadherin expression compared with normal human liver. Scanning electron microscopy revealed formation of septum-like structures encircling the transplanted human hepatocytes in the PXB liver, which consists of an accumulation of fibers in the space of Disse under transmission electron microscopy and is immunolabeled for laminin. Overall, the present report demonstrated the morphological and immunohistochemical characteristics of the PXB-mice with humanized liver along with some abnormalities in the cell adhesion of the transplanted human hepatocytes. These findings would be useful for hepatotoxicity studies using humanized animal models.
Collapse
Affiliation(s)
- Nazneen Sultana
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Tomomi Kamei
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Sho Fujiwara
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Yuri Ito
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Yuki Takami
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Orai-Kita, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
16
|
Kunze M, Malfatti F. Towards a Conceptual Framework to Better Understand the Advantages and Limitations of Model Organisms. Eur J Neurosci 2025; 61:e70071. [PMID: 40165014 DOI: 10.1111/ejn.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Model organisms (MO) are widely used in neuroscience to study brain processes, behavior, and the biological foundation of human diseases. However, the use of MO has also been criticized for low reliability and insufficient success rate in the development of therapeutic approaches, because the success of MO use also led to overoptimistic and simplistic applications, which sometimes resulted in wrong conclusions. Here, we develop a conceptual framework of MO to support scientists in their practical work and to foster discussions about their power and limitations. For this purpose, we take advantage of concepts developed in the philosophy of science and adjust them for practical application by neuroscientists. We suggest that MO can be best understood as tools that are used to gain information about a group of species or a phenomenon in a species of interest. These learning processes are made possible by some properties of MO, which facilitate the process of acquisition of understanding or provide practical advantages, and the possibility to transfer information between species. However, residual uncertainty in the reliability of information transfer remains, and incorrect generalizations can be side-effects of epistemic benefits, which we consider as representational and epistemic risks. This suggests that to use MO most effectively, scientists should analyze the similarity relation between the involved species, weigh advantages and risks of certain epistemic benefits, and invest in carefully designed validation experiments. Altogether, our analysis illustrates how scientists can benefit from philosophical concepts for their research practice.
Collapse
Affiliation(s)
- Markus Kunze
- Center for Brain Research, Department of Pathobiology of the Nervous System, Medical University of Vienna, Vienna, Austria
| | - Federica Malfatti
- Institut für Christliche Philosophie, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Bajaj P, Brennan RJ, Laurent S, Sauzeat S, Dufault M, Richards B, Adkins K. Transcriptomic analysis in liver spheroids identifies a dog-specific mechanism of hepatotoxicity for amcenestrant. Toxicol Sci 2025; 204:228-241. [PMID: 39886943 DOI: 10.1093/toxsci/kfaf012] [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] [Indexed: 02/01/2025] Open
Abstract
Therapeutic drugs can sometimes cause adverse effects in a nonclinical species that do not translate to other species, including human. Species-specific (rat, dog, and human) in vitro liver spheroids were employed to understand the human relevance of cholestatic liver injury observed with a selective estrogen receptor degrader (amcenestrant) in dog, but not in rat, during preclinical development. Amcenestrant showed comparable cytotoxicity in liver spheroids from all 3 species; however, its M5 metabolite (RA15400562) showed dog preferential cytotoxicity after 7 days of treatment. Whole genome transcript profiles generated from liver spheroids revealed downregulation of genes related to bile acid synthesis and transport indicative of strong farnesoid X receptor (FXR) antagonism following treatment with both amcenestrant and its M5 metabolite in the dog but not in rat or human. In human spheroids, upregulation of genes for detoxification enzymes indicative of pregnane X receptor (PXR) agonism was observed following amcenestrant treatment, whereas in the dog these genes were downregulated. The M5 metabolite showed gene dysregulation indicating PXR agonism in both rat and human, and antagonism in dog. Analysis of liver samples from a 3-mo dog toxicity study conducted with amcenestrant showed downregulation of several genes associated with PXR and FXR, corroborating the in vitro results. These results support the hypothesis that dogs are uniquely susceptible to cholestatic hepatotoxicity following administration of amcenestrant due to species-specific antagonism of FXR and highlight the value of in vitro liver spheroids to investigating mechanisms of toxicity and possible species differences.
Collapse
Affiliation(s)
- Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, United States
| | - Richard J Brennan
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, United States
| | | | | | - Michael Dufault
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA 02141, United States
| | - Brenda Richards
- Genetic Medicine Unit, Sanofi, Waltham, MA 02451, United States
| | - Karissa Adkins
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, United States
| |
Collapse
|
18
|
Shepherd J. Biomimetic Approaches in the Development of Optimised 3D Culture Environments for Drug Discovery in Cardiac Disease. Biomimetics (Basel) 2025; 10:204. [PMID: 40277603 PMCID: PMC12024959 DOI: 10.3390/biomimetics10040204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/09/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide, yet despite massive investment in drug discovery, the progress of cardiovascular drugs from lab to clinic remains slow. It is a complex, costly pathway from drug discovery to the clinic and failure becomes more expensive as a drug progresses along this pathway. The focus has begun to shift to optimisation of in vitro culture methodologies, not only because these must be undertaken are earlier on in the drug discovery pathway, but also because the principles of the 3Rs have become embedded in national and international legislation and regulation. Numerous studies have shown myocyte cell behaviour to be much more physiologically relevant in 3D culture compared to 2D culture, highlighting the advantages of using 3D-based models, whether microfluidic or otherwise, for preclinical drug screening. This review aims to provide an overview of the challenges in cardiovascular drug discovery, the limitations of traditional routes, and the successes in the field of preclinical models for cardiovascular drug discovery. It focuses on the particular role biomimicry can play, but also the challenges around implementation within commercial drug discovery.
Collapse
Affiliation(s)
- Jenny Shepherd
- School of Engineering, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
19
|
Fukunaga I, Takebe T. In vitro liver models for toxicological research. Drug Metab Pharmacokinet 2025; 62:101478. [PMID: 40203632 DOI: 10.1016/j.dmpk.2025.101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Drug-induced liver injury (DILI) presents a major challenge not only in new drug development but also in post-marketing withdrawals and the safety of food, cosmetics, and chemicals. Experimental model organisms such as the rodents have been widely used for preclinical toxicological testing. However, the tension exists associated with the ethical and sustainable use of animals in part because animals do not necessarily inform the human-specific ADME (adsorption, dynamics, metabolism and elimination) profiling. To establish alternative models in humans, in vitro hepatic tissue models have been proposed, ranging from primary hepatocytes, immortal hepatocytes, to the development of new cell resources such as stem cell-derived hepatocytes. Given the evolving number of novel alternative methods, understanding possible combinations of cell sources and culture methods will be crucial to develop the context-of-use assays. This review primarily focuses on 3D liver organoid models for conducting. We will review the relevant cell sources, bioengineering methods, selection of training compounds, and biomarkers towards the rationale design of in vitro toxicology testing.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
20
|
Rijia A, Krishnamoorthi R, Mahalingam PU, Kaviyadharshini M, Rajeswari M, Kumar KKS, Rasmi M, Chung YK, Fang JY. Unveiling the anticancer potential and toxicity of Ganoderma applanatum wild mushroom derived bioactive compounds: An in vitro, in vivo and in silico evaluation. Bioorg Chem 2025; 156:108233. [PMID: 39908734 DOI: 10.1016/j.bioorg.2025.108233] [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: 01/06/2025] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
This study explores the anticancer potential of methanolic extract from Ganoderma applanatum, focusing on its cytotoxicity across various cancer cell lines and its safety and efficacy in an in vivo hepatocellular carcinoma (HCC) model, along with molecular docking analysis of its bioactive compounds targeting B-cell lymphoma 2 (Bcl-2) protein. The MTT assay revealed significant cytotoxicity of the extract against epidermoid carcinoma (A431), human alveolar carcinoma (A549), and hepatocellular carcinoma (HepG2) cell lines, with the extract exhibiting the highest potency (IC50 of 95.65 µg/ml) against HepG2 cells. Apoptosis induction and DNA degradation in HepG2 cells were confirmed through mitochondrial membrane potential analysis, ethidium bromide/acridine orange staining, and DNA fragmentation assays. In vivo studies on Wistar albino rats showed that administration of the extract up to 1000 mg/ml did not significantly affect body weight or hematological parameters, suggesting a favorable safety profile. Histopathological examination revealed normal liver architecture at most doses, with mild inflammation observed at the highest dose (1000 mg/ml). The G. applanatum extract were showed reducing liver weight and improving body weight in a Diethylnitrosamine (DEN)-induced HCC model was comparable to cyclophosphamide, indicating its potential as a less toxic alternative or adjunct to conventional chemotherapy. Additionally, the extract reduced elevated serum liver enzymes, demonstrating hepatoprotective effects. Molecular docking of nine bioactive compounds from G. applanatum identified 2h-3,11c-(epoxymethano)phenanthro[10,1-bc]pyran as a promising candidate for further investigation. These findings suggest G. applanatum as a novel anticancer agent with the potential for natural, effective cancer therapy.
Collapse
Affiliation(s)
- Akbar Rijia
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India
| | - Raman Krishnamoorthi
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.
| | - Pambayan Ulagan Mahalingam
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India.
| | | | - Murugan Rajeswari
- Department of Chemistry, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India
| | - Konda Kannan Satheesh Kumar
- Department of Chemistry, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India
| | - Madhusoodhanan Rasmi
- Department of Microbiology, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Yu-Kuo Chung
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| |
Collapse
|
21
|
Ailabouni A, Prasad B. Organic cation transporters 2: Structure, regulation, functions, and clinical implications. Drug Metab Dispos 2025; 53:100044. [PMID: 40020559 DOI: 10.1016/j.dmd.2025.100044] [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: 10/16/2024] [Accepted: 01/21/2025] [Indexed: 03/03/2025] Open
Abstract
The SLC22A2 gene encodes organic cation transporter 2 (OCT2), which is predominantly expressed in renal proximal tubule cells. OCT2 is critical for the active renal excretion of various cationic drugs and endogenous metabolites. OCT2 expression varies across species, with higher levels in mice and monkeys compared with humans and rats. The human OCT2 protein consists of 555 amino acids and contains 12 transmembrane domains. OCT2 functions as a uniporter, facilitating the bidirectional transport of organic cations into renal tubular cells, driven by the inside-negative membrane potential. Its expression is regulated by sex hormones, contributing to potential sex differences in Oct2 activity in rodents. OCT2 has been linked to tissue toxicity, such as cisplatin-induced nephrotoxicity. Factors such as genetic variants, age, disease states, and the coadministration of drugs, including tyrosine kinase inhibitors, contribute to interindividual variability in OCT2 activity. This, in turn, impacts the systemic exposure and elimination of drugs and endogenous substances. Regulatory agencies recommend evaluating the potential of a drug to inhibit OCT2 through in vitro and clinical drug-drug interaction (DDI) studies, often using metformin as a probe substrate. Emerging tools like transporter biomarkers and physiologically based pharmacokinetic modeling hold promise in predicting OCT2-mediated DDIs. While several OCT2 biomarkers, such as N1-methylnicotinamide, have been proposed, their reliability in predicting renal DDIs remains uncertain and requires further study. Ultimately, a better understanding of the factors influencing OCT2 activity is essential for achieving precision medicine and minimizing renal and systemic toxicity. SIGNIFICANCE STATEMENT: Organic cation transporter 2 (OCT2) is essential for the active tubular secretion of xenobiotics and endogenous cationic substances in the kidneys. This article offers a comprehensive overview of the tissue distribution, interspecies differences, and factors affecting its activity-critical for evaluating tissue toxicity and systemic exposure to cationic substances. Using OCT2 biomarkers and integrating OCT2 activity and expression data into physiologically based pharmacokinetic models are valuable tools for predicting OCT2 function and its clinical implications.
Collapse
Affiliation(s)
- Anoud Ailabouni
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington.
| |
Collapse
|
22
|
Kim H, Park HJ. Current hPSC-derived liver organoids for toxicity testing: Cytochrome P450 enzymes and drug metabolism. Toxicol Res 2025; 41:105-121. [PMID: 40013078 PMCID: PMC11850699 DOI: 10.1007/s43188-024-00275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 02/28/2025] Open
Abstract
Drug-induced hepatotoxicity is the leading cause of attrition of drug candidates and withdrawal of marketed drugs owing to safety concerns. In most hepatotoxicity cases, the parent drugs are metabolized by cytochrome P450 (CYP) enzymes, generating reactive metabolites that bind to intracellular organelles and proteins, ultimately causing hepatocellular damage. A major limitation of animal models, which are widely used for toxicity assessment, is the discrepancy in CYP-mediated drug metabolism and toxicological outcomes owing to species differences between humans and animals. Two-dimensional (2D) hepatocytes were first developed as a promising alternative model using human pluripotent stem cells (hPSCs). However, their CYP expression was similar to that of the fetal liver, and they lacked CYP-mediated hepatic metabolism. CYP expression in hPSC-derived hepatic models is closely correlated with liver maturity. Therefore, liver organoids that are more mature than hPSC-derived hepatic models and mimic the structure and physiological functions of the human liver have emerged as new alternatives. In this review, we explored the role and essentiality of CYPs in human hepatotoxicity, their expression, and epigenetic regulation in hPSC-derived hepatocytes and liver organoids, as well as the current state of liver organoid technology in terms of CYP expression and activity, drug metabolism, and toxicity. We also discussed the current challenges and future directions for the practical use of liver organoids. In conclusion, we highlight the importance of methods and metrics for accurately assessing CYP expression and activity in liver organoids to enable the development of feasible models that reproduce hepatotoxicity in humans.
Collapse
Affiliation(s)
- Hyemin Kim
- Division of Advanced Predictive Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Han-Jin Park
- Division of Advanced Predictive Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Sakolish C, Tsai HHD, Lin HC, Bajaj P, Villenave R, Ferguson SS, Stanko JP, Becker RA, Hewitt P, Chiu WA, Rusyn I. Comparative Analysis of Proximal Tubule Cell Sources for In Vitro Studies of Renal Proximal Tubule Toxicity. Biomedicines 2025; 13:563. [PMID: 40149543 PMCID: PMC11940618 DOI: 10.3390/biomedicines13030563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: The kidneys are essential for eliminating drugs and chemicals from the human body and renal epithelial cells are particularly vulnerable to damage caused by xenobiotics and their metabolites. Drug-induced kidney toxicity is a major cause of drug attrition during preclinical and clinical development and the ability to predict renal toxicity remains a pressing challenge, necessitating more predictive in vitro models. However, the abundance of commercially available renal proximal tubule epithelial cell (RPTEC) sources complicates the selection of the most predictive cell types. Methods: This study compared a wide range of RPTEC sources, including primary cells (Lonza) and various RPTEC lines from different vendors, such as ciPTECs (Cell4Pharma), TERT1/RPTECs (ATCC), and HEK293 (GenoMembrane), including OAT1-overexpressing variants. HepG2 cells were included for a comparison of organ specificity. The different cells were cultured in 96- or 384-well plates and exposed to 12 drugs for 72 h at a concentration yielding a response (0.3-300 µM) to evaluate their ability to predict clinical outcomes. The CellTiterGlo® assay was used to measure cell viability, and transcriptome data from unexposed cells was analyzed using the TempO-seq® S1500+ platform. Results: Gene expression data showed that the primary kidney cells most closely matched the transcriptome of the human kidney medulla, followed by the TERT1 and ciPTEC lines, with the HEK lines showing the lowest similarity. The RPTEC sources showed clustering by cell type, with OAT1 overexpression driving changes in metabolic, detoxification, and immune pathways, especially in TERT1 cells. Cell viability data were used to determine points of departure (PODs) which were compared to human serum Cmax values to assess safety margins. The TERT1 and ciPTEC RPTEC lines demonstrated the highest predictive performance for nephrotoxicity, with OAT1 overexpression significantly enhancing sensitivity, accuracy, and overall predictive power (MCC scores: 0.764 and 0.667, respectively). In contrast, HepG2 cells showed the lowest performance across all metrics, highlighting the critical role of cell type and transporter expression in nephrotoxicity prediction. Conclusions: This study highlights important differences among RPTEC sources and their utility in drug safety studies of the renal proximal tubule. We show that while improved cell options for renal proximal tubule are needed, OAT1-overexpressing RPTECs are a superior model to the background cell type.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Han-Hsuan D. Tsai
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Hsing-Chieh Lin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, USA;
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland;
| | - Stephen S. Ferguson
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (S.S.F.); (J.P.S.)
| | - Jason P. Stanko
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (S.S.F.); (J.P.S.)
| | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293 Darmstadt, Germany;
| | - Weihsueh A. Chiu
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (C.S.); (H.-H.D.T.); (H.-C.L.); (W.A.C.)
| |
Collapse
|
24
|
Elmore SA, Haseman JK, Pemberton M, Gollapudi BB, Cohen SM. A critical evaluation of rodent carcinogenicity studies on butyl methacrylate demonstrates a lack of carcinogenic potential. Crit Rev Toxicol 2025; 55:105-123. [PMID: 40152788 DOI: 10.1080/10408444.2025.2451885] [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: 11/15/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 03/29/2025]
Abstract
Assessments of a chemical agent's carcinogenicity based on rodent bioassays rely on their appropriate interpretation. This involves attention to study details, including reliable histopathologic diagnoses, and proper statistical analyses, including consideration of multiple comparisons, concurrent and historical controls. A major factor is evaluation of their likely mode of action and the human relevance of any identified tumors. We present a critical evaluation of the assessment of the 2-year inhalation bioassays of n-butyl methacrylate (n-BMA) in rats and mice performed by the Japan Bioassay Research Center (JBRC) and an assessment of the International Agency for Research on Cancer (IARC) review and classification as Group 2B, possible human carcinogen. The tumors of concern for assessment of its carcinogenicity included mononuclear cell leukemia (MCL) in male rats, thyroid C-cell tumors in female rats, liver tumors and histiocytic sarcomas in male mice, and hemangiosarcomas in female mice. Our review of these studies raises concerns regarding the accuracy of histopathology diagnoses and human relevance of MCL. Most critically, the statistical evaluation/interpretation of all tumor types indicates no carcinogenic effects, since the frequency of increases (at p < 0.05) in tumor incidences in the study is totally consistent with chance expectation (i.e. not treatment related). Furthermore, the plausibility of n-BMA being carcinogenic is questionable since it is non-genotoxic, and the weight of evidence including read-across to the close structural analog methyl methacrylate indicates no concern for cancer. After thorough review of these bioassays, we conclude that there is no convincing evidence of carcinogenicity for n-BMA, contrary to the conclusion of the JBRC and the decision by the IARC.
Collapse
Affiliation(s)
| | | | | | | | - Samuel M Cohen
- Department of Pathology, Microbiology, and Immunology and the Buffett Cancer Center, and the Havlik-Wall Professor of Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
25
|
Ed-Dahmani I, El Fadili M, Nouioura G, Kandsi F, Atki YE, Abuelizz HA, Conte R, Zahra Lafdil F, Taleb A, Abdellaoui A, Taleb M. Ferula communis leaf extract: antioxidant capacity, UHPLC-MS/MS analysis, and in vivo and in silico toxicity investigations. Front Chem 2025; 12:1485463. [PMID: 39925381 PMCID: PMC11803407 DOI: 10.3389/fchem.2024.1485463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/03/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Ferula communis has demonstrated an abundance of pharmacological and antioxidative qualities. Methods This study investigates the antioxidant activity of F. communis leaf aqueous extract, total polyphenol and flavonoid concentrations, and ultra-high-performance liquid chromatography (UHPLC) composition and then evaluates the toxicity of the plant's leaves in vitro and in silico. The major compound of the studied extract, namely, p-hydroxybenzoic acid, was chosen for a molecular docking technique to discover the inhibition mechanism toward antioxidant proteins. In addition, a detailed molecular dynamics simulation was carried out to examine the thermodynamic stability of the produced intermolecular interactions. The antioxidant capacity of the extracts of F. communis was evaluated using 2,2-diphenylpicryl hydroxyl (DPPH) radical and ferric reducing antioxidant power (FRAP) procedures. Acute toxicity was tested on albino mice at doses of 200, 300, and 400 mg/kg. Results The results show that the polyphenol and flavonoid contents of the extract are significant (0.257 ± 0.003 mg Eq AG/mg and 0.32 ± 0.04 mg Eq Q/mg, respectively). The antioxidant activity illustrates that the extracts have notable activity in DPPH and FRAP assays. The toxicity study revealed that the mice's behavior, body weight, and organ weights (liver and kidneys) were unaffected by Ferula communis leaf extract administration compared to controls. UHPLC-tandem mass spectrometry (MS/MS) analysis of the extract highlights the presence of 11 compounds, the most abundant of which is p-hydroxybenzoic acid, representing 53.65%. The predicted pharmacokinetic characteristics of absorption, distribution, metabolism, excretion, and toxicity (ADMET) attest to the well-absorbed nature of the isolated compounds, with human intestinal absorption (HIA) varying from 42% for arbutin (M3) to 100% for ursolic acid (M4). Conclusion In conclusion, the leaves of Ferula communis are a good source of natural antioxidants and phenolic compounds. Thus, this study demonstrates that this plant has a wide range of applications, including natural food preservatives, pharmaceuticals, and cosmetics, as evidenced by ongoing research.
Collapse
Affiliation(s)
- Imad Ed-Dahmani
- Laboratory of Engineering, Electrochemistry, Modelling and Environment, Sidi Mohamed Ben Abdellah University, Faculty of Sciences Fez, Fez, Morocco
| | - Mohamed El Fadili
- LIMAS Laboratory, Faculty of Sciences Dhar El Mehraz, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| | - Ghizlane Nouioura
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El-Mehraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Fahd Kandsi
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda, Morocco
| | - Yassine El Atki
- High Institute of Nursing Professions and Health Techniques Errachidia, Errachidia, Morocco
| | - Hatem A. Abuelizz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Naples, Italy
| | - Fatima Zahra Lafdil
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda, Morocco
| | - Abdeslam Taleb
- Laboratory of Water and Environmental Engineering, Hassan II University of Casablanca, Casablanca, Morocco
| | - Abdelfattah Abdellaoui
- Laboratory of Engineering, Electrochemistry, Modelling and Environment, Sidi Mohamed Ben Abdellah University, Faculty of Sciences Fez, Fez, Morocco
| | - Mustapha Taleb
- Laboratory of Engineering, Electrochemistry, Modelling and Environment, Sidi Mohamed Ben Abdellah University, Faculty of Sciences Fez, Fez, Morocco
| |
Collapse
|
26
|
Klein JA, Heidmann JD, Kiyota T, Fullerton A, Homan KA, Co JY. The differentiation state of small intestinal organoid models influences prediction of drug-induced toxicity. Front Cell Dev Biol 2025; 13:1508820. [PMID: 39917568 PMCID: PMC11799252 DOI: 10.3389/fcell.2025.1508820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025] Open
Abstract
Drug-induced intestinal toxicity (GIT) is a frequent dose-limiting adverse event that can impact patient compliance and treatment outcomes. In vivo, there are proliferative and differentiated cell types critical to maintaining intestinal homeostasis. Traditional in vitro models using transformed cell lines do not capture this cellular complexity, and often fail to predict intestinal toxicity. Primary tissue-derived intestinal organoids, on the other hand, are a scalable Complex in vitro Model (CIVM) that recapitulates major intestinal cell lineages and function. Intestinal organoid toxicity assays have been shown to correlate with clinical incidence of drug-induced diarrhea, however existing studies do not consider how differentiation state of the organoids impacts assay readouts and predictivity. We employed distinct proliferative and differentiated organoid models of the small intestine to assess whether differentiation state alone can alter toxicity responses to small molecule compounds in cell viability assays. In doing so, we identified several examples of small molecules which elicit differential toxicity in proliferative and differentiated organoid models. This proof of concept highlights the need to consider which cell types are present in CIVMs, their differentiation state, and how this alters interpretation of toxicity assays.
Collapse
Affiliation(s)
- Jessica A. Klein
- Complex In Vitro Systems, Translational Safety, Genentech Inc., South San Francisco, CA, United States
| | - Julia D. Heidmann
- Investigative Toxicology, Translational Safety, Genentech Inc., South San Francisco, CA, United States
| | - Tomomi Kiyota
- Investigative Toxicology, Translational Safety, Genentech Inc., South San Francisco, CA, United States
| | - Aaron Fullerton
- Investigative Toxicology, Translational Safety, Genentech Inc., South San Francisco, CA, United States
| | - Kimberly A. Homan
- Complex In Vitro Systems, Translational Safety, Genentech Inc., South San Francisco, CA, United States
| | - Julia Y. Co
- Complex In Vitro Systems, Translational Safety, Genentech Inc., South San Francisco, CA, United States
| |
Collapse
|
27
|
Gao Y, Bissoyi A, Guo Q, Gibson MI. Induced Extracellular Ice Nucleation Protects Cocultured Spheroid Interior and Exterior during Cryopreservation. ACS Biomater Sci Eng 2025; 11:208-212. [PMID: 39315639 PMCID: PMC11733914 DOI: 10.1021/acsbiomaterials.4c00958] [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: 05/24/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
Spheroids and other 3D cellular models more accurately recapitulate physiological responses when compared to 2D models and represent potential alternatives to animal testing. The cryopreservation of spheroids remains challenging, limiting their wider use. Standard DMSO-only cryopreservation results in supercooling to low subzero temperatures, reducing viability, shedding surface cells, and perforating spheroid interiors. Here, cocultured spheroids with differentially labeled outer cell layers allow spatial evaluation of the protective effect of macromolecular ice nucleators by microscopy and histology. Extracellular nucleation is shown to reduce damage to both interior and exterior regions of the spheroids, which will support the development of "off-the-shelf" 3D models.
Collapse
Affiliation(s)
- Yanan Gao
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Akalabya Bissoyi
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess
Street, Manchester M1 7DN, United Kingdom
| | - Qiongyu Guo
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Matthew I. Gibson
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
- Division
of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, United
Kingdom
- Department
of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
- Manchester
Institute of Biotechnology, University of
Manchester, 131 Princess
Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
28
|
Bonafé Allende JC, Ambrosioni F, Ruiz Moreno FN, Marin C, Romero VL, Virgolini MB, Maletto BA, Jimenez Kairuz AF, Alvarez Igarzabal CI, Picchio ML. Pyrogallol-rich supramolecular hydrogels with enzyme-sensitive microdomains for controlled topical delivery of hydrophobic drugs. BIOMATERIALS ADVANCES 2025; 166:214075. [PMID: 39476684 DOI: 10.1016/j.bioadv.2024.214075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 11/13/2024]
Abstract
Skin wound treatments require efficient and targeted delivery of therapeutic agents to promote fast tissue regeneration and prevent infections. Hydrogels are one of the most popular products in the wound care market, although their use as medicated wound dressings remains a massive challenge when hydrophobic drugs are needed due to the hydrophilic nature of these soft materials. In this study, we developed innovative, dynamic hydrogels based on polyvinyl alcohol (PVA), pyrogallol as a hydrogen bond crosslinker, and casein micelles as hydrophobic reservoirs of silver sulfadiazine (SSD) for enzyme-activated smart delivery at wound sites. The hydrogel formulation was optimized for mechanical strength, viscoelastic behavior, water absorption capacity, and drug-loading efficiency. In vitro drug delivery studies revealed a sustainable release profile of SSD for over 24 h from the micelles within the hydrogel network. Furthermore, biocompatibility evaluation using mouse fibroblast L929 cells demonstrated that the hydrogel did not inhibit cell viability, while in vivo experiments on Caenorhabditis elegans (C. elegans) proved its safety in complex organisms. This versatile hydrogel also has anti-inflammatory and antibacterial effects stemming from the therapeutic polyphenol, which could benefit the healing process. The combination of PVA, pyrogallol, and casein-based nanocarriers could offer an approach to wound healing, providing a new platform for hosting hydrophobic therapeutic substances. Overall, this hydrogel system shows great promise in wound care and could broaden the applications of this family of soft materials for treating various skin injuries.
Collapse
Affiliation(s)
- Juan Cruz Bonafé Allende
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Instituto De Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA-CONICET), Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina
| | - Franco Ambrosioni
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA-CONICET). Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina
| | - Federico N Ruiz Moreno
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Constanza Marin
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Verónica L Romero
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Farmacología Otto Orsingher, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina
| | - Miriam B Virgolini
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Farmacología Otto Orsingher, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Belkys A Maletto
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Alvaro F Jimenez Kairuz
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA-CONICET). Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina
| | - Cecilia I Alvarez Igarzabal
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina; Instituto De Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA-CONICET), Haya de la Torre y Av. Medina Allende, X5000HUA Córdoba, Argentina.
| | - Matías L Picchio
- POLYMAT, Applied Chemistry Department, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizábal, 3, 20018 Donostia-San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain; Facultad Regional Villa María (Universidad Tecnológica Nacional), Av. Universidad 450, Villa María, 5900, Córdoba, Argentina.
| |
Collapse
|
29
|
Carratt SA, Zuch de Zafra CL, Oziolor E, Rana P, Vansell NR, Mangipudy R, Vaidya VS. An industry perspective on the FDA Modernization Act 2.0/3.0: potential next steps for sponsors to reduce animal use in drug development. Toxicol Sci 2025; 203:28-34. [PMID: 39298459 DOI: 10.1093/toxsci/kfae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
Pharmaceutical developers are encouraged to adopt the best practices of being purposefully thoughtful about the use of animals, seeking alternatives wherever possible. They should engage with health authorities to increase their familiarity with the methods, study designs, data outputs, and the context of use for new approach methodologies (NAMs). Although current state of technology does not yet provide adequate models to fully replace in vivo studies, many models are sufficiently good for an augmented approach that will enhance our understanding of in vitro to in vivo correlations and advance the long-term goal of reducing animal use through innovative NAMs. The goal of future nonclinical safety packages is to advance the utilization of such enabling technologies toward appropriate human risk characterization. Establishing confidence in NAMs is a critical first step. For example, sponsors may include both "traditional" and NAM-based nonclinical safety data in regulatory submissions to establish confidence with health authorities. In addition, regulators should create a "safe harbor" for hybrid nonclinical data packages to facilitate iterative learning, refinement, and implementation of NAM-based safety assessment strategies. Sponsors are urged to contribute to NAMs evolution through consortia participation, peer-reviewed publications, and documenting animal reduction in studies/programs, accelerating the eventual elimination of animal use in pharmaceutical development, as envisioned in the FDA Modernization Act 3.0.
Collapse
Affiliation(s)
- Sarah A Carratt
- Drug Safety Research and Development, Pfizer Research and Development, Bothell, WA 98021, United States
| | - Christina L Zuch de Zafra
- Drug Safety Research and Development, Pfizer Research and Development, San Francisco, CA 94080, United States
| | - Elias Oziolor
- Drug Safety Research and Development, Pfizer Research and Development, Groton, CT 06340, United States
| | - Payal Rana
- Drug Safety Research and Development, Pfizer Research and Development, Groton, CT 06340, United States
| | - Nichole R Vansell
- Drug Safety Research and Development, Pfizer Research and Development, Groton, CT 06340, United States
| | - Raja Mangipudy
- Drug Safety Research and Development, Pfizer Research and Development, Groton, CT 06340, United States
| | - Vishal S Vaidya
- Drug Safety Research and Development, Pfizer Research and Development, Cambridge, MA 02138, United States
| |
Collapse
|
30
|
Islam R, Deb A, Ghosh AJ, Dutta D, Ray A, Dutta A, Ghosh S, Sarkar S, Bahadur M, Kumar A, Saha T. Toxicological profiling of methanolic seed extract of Abutilon indicum (L.) Sweet: in-vitro and in-vivo analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118655. [PMID: 39097211 DOI: 10.1016/j.jep.2024.118655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Abutilon indicum, a shrub of the Malvaceae family, is found abundantly in tropical countries like India. A. indicum is widely used for its high medicinal properties. Traditionally, A. indicum seed powder is consumed to treat piles, constipation, chronic cystitis, gonorrhea, gleet, and pregnancy-related problems. Despite having numerous medicinal properties and widespread traditional use of A. indicum seeds, scientific validation, and toxicity studies have yet to be documented. AIMS OF THE STUDY The primary objective of this study is to conduct a comprehensive study on phytochemical profiling, in-vitro cytotoxicity, mutagenicity, and in-vivo acute and sub-acute toxicity, and genotoxicity on animal models of methanolic extract of A. indicum seed (MAS). MATERIALS AND METHODS The qualitative analysis of MAS was explored through FTIR and HR LC-MS. For in-vitro cytotoxicity, the HEK-293 cell line was used, and the TA100 (Staphylococcus typhimurium) bacterial strain was used for the Ames mutagenicity test. A single oral dose of 250, 500, 1000, or 2000 mg/kg body weight of MAS was given to each male and female rat for acute toxicity study and observed for 14 days for any toxicity signs. In the sub-acute toxicity study, 250, 500, or 1000 mg/kg body weight of MAS was administered orally to each rat for 28 days. The experimental animals were weighed weekly, and general behavior was monitored regularly. After 28 days of the experiment, the rats were sacrificed, and different serum biochemical, hematological, and histological analyses were performed. The blood samples of different doses of MAS were used for genotoxicity study through comet assay. RESULTS FTIR analysis found different functional groups, which indicated the presence of phenolics, flavonoids, and alkaloids. HR LC-MS analysis depicts several components with different biological functions. The cell cytotoxicity and Ames mutagenicity results showed minimal toxicity and mutagenicity up to a certain dose. The acute toxicity study conducted in Wistar albino rats demonstrated zero mortality among the animals, and the LD50 value for seed extract was determined to be 2000 mg/kg body weight. Sub-acute toxicity assessments indicated that the administration of seed extract resulted in no adverse effects at dosages of 250 and 500 mg/kg body weight. However, at higher doses, specifically 1000 mg/kg body weight, the liver of the experimental rats exhibited some toxic effects. In the genotoxicity study, minimal DNA damage was found in 250 and 500 mg/kg doses, respectively, but slightly greater DNA damage was found in 1000 mg/kg doses in both male and female rats. CONCLUSIONS The consumption of A. indicum seed powder is deemed safe; however, doses exceeding 500 mg/kg body weight may raise concerns regarding use. These findings pave the path for the creation of innovative medicines with improved efficacy and safety profiles.
Collapse
Affiliation(s)
- Rejuan Islam
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Arijit Deb
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Amlan Jyoti Ghosh
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Debojit Dutta
- Genetics and Moleular Biology Labratoty, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Arpita Ray
- Genetics and Moleular Biology Labratoty, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Ankita Dutta
- Advanced Nanoscale Molecular Oncology Laboratory, Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Supriyo Ghosh
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Sagar Sarkar
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India; Department of Zoology, Siliguri College, Darjeeling, West Bengal, 734001, India
| | - Min Bahadur
- Genetics and Moleular Biology Labratoty, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India
| | - Anoop Kumar
- Advanced Nanoscale Molecular Oncology Laboratory, Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| | - Tilak Saha
- Immunology and Microbiology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, West Bengal, 734013, India.
| |
Collapse
|
31
|
Maache S, Tahraoui A, Soulo N, Benlabchir AA, Nouioura G, Bouslamti M, Aldisi D, Saghrouchni H, Giesy JP, Aboul-Soud M, Lyoussi B, Elarabi I. Sub-chronic and acute toxicity of aqueous extracts Salvia blancoana subsp. mesatlantica (Maire) Figuerola to rodents. Toxicol Rep 2024; 13:101847. [PMID: 39703767 PMCID: PMC11655812 DOI: 10.1016/j.toxrep.2024.101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024] Open
Abstract
Background Salvia blancoana subsp. mesatlantica (Maire) Figuerola (SBm) is a plant endemic to Morocco and is one of the less studied species of Salvia. Herbal therapy is becoming more and more popular, especially in underdeveloped nations where access to medicinal herbs is affordable. However, some plants demonstrated toxic effects in animals and humans. Objective Our study aimed to evaluate the SBm-extract for both acute and sub-chronic toxicity. Methods Aqueous extracts were obtained from the aerial parts of SBm collected from Immouzer Kander commune (Middle Atlas, Morocco). Total Phenolic Content (TPC) and Flavones and flavonols Content (FFC), Antioxidant activity (2,2-diphenyl-1-picrylhydrazyl (DPPH) and reducing power) was determined, and chemical composition was determined by High-Performance Liquid Chromatography with Diode Array Detection (HPLC-DAD). Toxicity tests were conducted on mice and rats. Results In acute toxicity, Swiss albino mice (mass of 25-35 g) received SBm-extract orally and intraperitoneally at doses (0.5-11 g/kg, bm). The sub-chronic toxicity was tested in Wistar albino rats (mass of 200-240 g) for 90 days at doses of 0, 250, 500, or 1000 mg/kg, bm. Values of TPC and FFC were estimated to be 157.56 ± 0.32 mg GAE/g DW and 7.89 ± 0.05 mg QE/g DW, respectively. DPPH scavenging (IC50) was estimated to be 26.9 ± 0.08 µg/mL while reducing power was 12.41 ± 0.03 µg/mL. No toxicity or deaths were observed in acute tests after oral exposure, while intraperitoneal administration resulted in dose-dependent acute toxicity, with an LD50 value of 6.82 g/kg. In sub-chronic tests, most hematological and biochemical parameters remained unchanged, except for transient fluctuations in specific blood constituents and a transitory reduction in serum glucose levels observed at elevated dosages. Histopathological investigation revealed no organ abnormalities. The SBm-extract exhibited minimal toxicity, supporting its safe use. Conclusions Despite the relevant results of this study, future studies need to confirm these findings and expand our understanding of the safety characteristics of Salvia. Further investigations are needed to explore the effects of other solvents on the extraction of bioactive compounds from the underground and aerial parts of this endemic species. Evaluation of other biological properties such as anti-microbial, anti-cancer, and anti-inflammatory activities are needed.
Collapse
Affiliation(s)
- Souad Maache
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Adel Tahraoui
- Regional Center of Education and Training Careers (CRMEF), Fez-Meknes, Morocco
| | - Najoua Soulo
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Abdessamad Ait Benlabchir
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ghizlane Nouioura
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mohammed Bouslamti
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Dara Aldisi
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Hamza Saghrouchni
- Department of Biotechnology, Institute of Natural and Applied Sciences, Çukurova University, 01330 Balcalı, Adana, Türkiye
| | - John P. Giesy
- Toxicology Centre, University of Saskatchewan, Saskatoon, Canada
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
- Department of Integrative Biology and Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA
| | - Mourad.A.M. Aboul-Soud
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Badiaa Lyoussi
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ilham Elarabi
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health, and Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
32
|
Camara Dit Pinto S, Cherkaoui J, Ghosh D, Cazaubon V, Benzeroual KE, Levine SM, Cherkaoui M, Sood GK, Anandasabapathy S, Dhingra S, Vierling JM, Gallo NR. A virtual scalable model of the Hepatic Lobule for acetaminophen hepatotoxicity prediction. NPJ Digit Med 2024; 7:340. [PMID: 39604584 PMCID: PMC11603025 DOI: 10.1038/s41746-024-01349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Addressing drug-induced liver injury is crucial in drug development, often causing Phase III trial failures and market withdrawals. Traditional animal models fail to predict human liver toxicity accurately. Virtual twins of human organs present a promising solution. We introduce the Virtual Hepatic Lobule, a foundational element of the Living Liver, a multi-scale liver virtual twin. This model integrates blood flow dynamics and an acetaminophen-induced injury model to predict hepatocyte injury patterns specific to patients. By incorporating metabolic zonation, our predictions align with clinical zonal hepatotoxicity observations. This methodology advances the development of a human liver virtual twin, aiding in the prediction and validation of drug-induced liver injuries.
Collapse
Affiliation(s)
- Stelian Camara Dit Pinto
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Jalal Cherkaoui
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
- Institut National des Sciences Appliquées, Lyon, France
| | - Debarshi Ghosh
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Valentine Cazaubon
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Kenza E Benzeroual
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | | | - Mohammed Cherkaoui
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Gagan K Sood
- Division of Gastroenterology, Baylor College of Medicine, Houston, TX, USA
| | | | - Sadhna Dhingra
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - John M Vierling
- Departments of Medicine and Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Nicolas R Gallo
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA.
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| |
Collapse
|
33
|
Sciandrone B, Kentsop RAD, Pensotti R, Ottolina G, Mascheretti I, Mattana M, Regonesi ME. Toxicological Analysis of the Arylnaphthalene Lignan Justicidin B Using a Caenorhabditis elegans Model. Molecules 2024; 29:5516. [PMID: 39683676 DOI: 10.3390/molecules29235516] [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: 10/20/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
The screening of plant-derived compounds with anti-cancer properties is a promising strategy to meet the growing need for new, safe and effective anti-cancer drugs. Justicidin B is a plants secondary metabolite that displays anti-cancer properties in several tumor cells. Therefore, it represents a good candidate. We used the 3R-compliant organism Caenorhabditis elegans to evaluate the safety of justicidin B produced by in vitro-grown adventitious roots of Linum lewisii. We showed that a dose of 100 µg/mL justicidin B does not affect worm vitality in either short-term or chronic administration; in contrast, the 200 µg/mL dose induces a lifespan reduction, but only in short-term daily treatment. We attributed this effect to its accumulation in lipofuscin granules in the pharynx as observed through confocal analysis. HPLC analysis confirmed the higher accumulation justicidin B with a 200 µg/mL dose but also revealed the presence of metabolic derivatives that could be responsible for the toxicity. We also demonstrated that the 100 µg/mL dose does not affect worm fertility or development. Our results highlight the safety of justicidin B, supporting its employment in cancer therapy, and encourage the use of a C. elegans model as an appropriate tool to assess compounds' toxicity before moving to more complex organisms.
Collapse
Affiliation(s)
- Barbara Sciandrone
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | | | - Roberta Pensotti
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Gianluca Ottolina
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council, 20131 Milan, Italy
| | - Iride Mascheretti
- Institute of Agricultural Biology and Biotechnology, National Research Council, 20133 Milan, Italy
| | - Monica Mattana
- Institute of Agricultural Biology and Biotechnology, National Research Council, 20133 Milan, Italy
| | - Maria Elena Regonesi
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
- Milan Center of Neuroscience (NeuroMI), 20126 Milan, Italy
| |
Collapse
|
34
|
Monticello TM, Potter DM, Huang Q, Hart TK, Shuey D, Troth S, Vergis JM, Tassew N, Glascott P. Do longer duration nonclinical toxicology studies provide predictive clinical safety value? The IQ consortium longer duration nonclinical to clinical translational database. Toxicol Appl Pharmacol 2024; 492:117087. [PMID: 39243825 DOI: 10.1016/j.taap.2024.117087] [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: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
The IQ Consortium's DruSafe Leadership Group previously reported results of a nonclinical to clinical translational database for First-In-Human (FIH)-enabling animal toxicology studies. We have completed an additional translational database populated with longer duration (>1 month) animal toxicology studies and longer duration (Phase 2 and beyond) clinical trials. The blinded database was composed of 127 molecules. Animal and clinical data were categorized by organ system and animal model (e.g. rodent, dog). The 2 × 2 contingency table (true positive, false positive, true negative, false negative) was used for statistical analysis and both the positive predictive value (PPV) and negative predictive value (NPV) were determined. As also reported in the FIH database, the NPV was the strongest predictive performance measure at 96 %. The PPV was lower than the FIH database with the rodent at 29 %, dog at 21 % and NHP at 20 %. No new additional target organs were observed in 62 % of the entries. A new target organ was identified in 38 % of the entries, with the majority in a rodent (26 %) and fewer in the dog (8 %) or NHP (12 %). However, new target organ data resulted in only a PPV of 13 %, suggesting that current ICH requirements for longer duration animal general toxicology studies should be re-evaluated and better aligned with the 3Rs. A newer paradigm could include an appropriately justified single animal model for longer duration studies, in addition to utilizing New Approach Methods (NAMs) that would provide translational safety data, but additional research is needed.
Collapse
Affiliation(s)
- Thomas M Monticello
- Translational Safety and Bioanalytical Analysis, Amgen, Thousand Oaks, CA 91320, United States of America
| | - David M Potter
- Independent Statistical Consultant, Bristol, RI 02809, United States of America
| | - Qihong Huang
- Boehringer Ingelheim, Ridgefield, CT, 06879, United States of America
| | - Timothy K Hart
- GlaxoSmithKline, King of Prussia, PA, 19406, United States of America
| | - Dana Shuey
- Toxicology, Incyte Corporation, Wilmington, DE 19803, United States of America
| | - Sean Troth
- Nonclinical Drug Safety, Merck and Co., Inc., Rahway, NJ, 07065, United States of America
| | - James M Vergis
- IQ Consortium, Washington, DC, 20005, United States of America
| | - Nardos Tassew
- Safety Assessment, Genentech, South San Francisco, CA, 92056, United States of America
| | - Peter Glascott
- Preclinical Safety, Sanofi, Bridgewater, NJ, 08807, United States of America.
| |
Collapse
|
35
|
Pandit VA, Singhal SK, Deshmane GB, Sahasrabuddhe RA, Karandikar MN, Pawar MS, Kulkarni VS, Dawane JS, Wele AA. Toxicity profile of standardized Krishna Vajra Abhraka bhasma made from biotite mica. J Ayurveda Integr Med 2024; 15:100980. [PMID: 39616794 DOI: 10.1016/j.jaim.2024.100980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/02/2024] [Accepted: 05/17/2024] [Indexed: 12/25/2024] Open
Abstract
INTRODUCTION The Bhasmas, herbo-minerals, are reported to produce adverse effects when used clinically. Therefore, the present study aimed to assess the safety of Krishna Vajra Abhraka Bhasma (KVB), through acute and subacute toxicity testing. MATERIALS AND METHODS After ethics committee approval, total 66 Wistar albino rats weighing 180-200 gms were used. Experiments were conducted as per CCSEA guidelines. In acute toxicity study, a single limit dose of 2000 mg/kg of KVB with honey was administered to six female rats. Subacute toxicity studies involved 60 Wistar albino rats of either sex, randomly divided into six groups and received KVB in low (highest clinical dose extrapolated to rats), medium (double the low dose) and high (double the medium dose) dose administered orally for 28 days. RESULTS No mortality or adverse effects on rat's general behaviour or internal organs was observed in acute toxicity study The results of subacute study indicated no significant differences in body and organ weights, hematological or renal function between control and treated groups. However, mid and high doses of KVB revealed reversible, dose-dependent hepatotoxicity through biochemical and histopathological evaluation, while the satellite high dose group did not exhibit this effect, confirming it as drug-induced hepatotoxicity. CONCLUSION The acute toxicity results positioned KVB in class V (safe) of the GHS classification. Subacute toxicity findings suggested that while low therapeutic doses were non-toxic, mid and high doses induced reversible hepatotoxicity, emphasizing the importance of careful dosage considerations and patient monitoring in Ayurvedic practice. The study contributes valuable insights into the safety profile of KVB, addressing concerns for both users and practitioners of Ayurvedic medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Asmita Ashish Wele
- Department of Rasashastra & Bhaishajyakalpana, Dr. D.Y. Patil College of Ayurved & Research Centre, Dr. D.Y. Patil Vidyapeeth (Deemed to be University), Pune- 411018, India.
| |
Collapse
|
36
|
Dolatimehr A, Mahyar A, Barough SPH, Mahmoodi M. Insights into the efficiencies of different biological treatment systems for pharmaceuticals removal: A review. WATER ENVIRONMENT RESEARCH : A RESEARCH PUBLICATION OF THE WATER ENVIRONMENT FEDERATION 2024; 96:e11153. [PMID: 39539062 DOI: 10.1002/wer.11153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/29/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
This review presents a comprehensive analysis of current research on biological treatment processes for removing pharmaceutical compounds (PhCs) from wastewater. Unlike previous studies on this topic, our study specifically delves into the effectiveness and drawbacks of various treatment approaches such as traditional wastewater treatment facilities (WWTP), membrane bioreactors (MBRs), constructed wetlands (CW), and moving bed biofilm reactors (MBBR). Through the examination and synthesis of information gathered from more than 200 research studies, we have created a comprehensive database that delves into the effectiveness of eliminating 19 particular PhCs, including commonly studied compounds such as acetaminophen, ibuprofen, diclofenac, naproxen, ketoprofen, indomethacin, salicylic acid, codeine, and fenoprofen, amoxicillin, azithromycin, ciprofloxacin, ofloxacin, tetracycline, atenolol, propranolol, and metoprolol. This resource provides a depth and scope of information that was previously lacking in this area of study. Notably, among these pharmaceuticals, azithromycin demonstrated the highest removal rates across all examined treatment systems, with the exception of WWTPs, while carbamazepine consistently exhibited the lowest removal efficiencies across various systems. The analysis showcases the diverse results in removal efficiency impacted by factors such as system configuration, operation specifics, and environmental circumstances. The findings emphasize the critical need for continued innovation and research, specifically recommending the integration of advanced oxidation processes (AOPs) with existing biological treatment methods to improve the breakdown of recalcitrant compounds like carbamazepine. PRACTITIONER POINTS: Persistent pharmaceuticals harm aquatic ecosystems and human health. Biological systems show varying pharmaceutical removal efficiencies. Enhancing HRT and SRT improves removal but adds complexity and costs. Tailored treatment approaches needed based on contaminants and conditions.
Collapse
Affiliation(s)
- Armin Dolatimehr
- Master of Water and Hydraulics, Independent Researcher, Islamic Azad University, Berlin, Germany
| | - Ali Mahyar
- Brandenburg University of Technology (Cottbus-Senftenberg) Volmerstr, Berlin, Germany
| | | | - Mohammadreza Mahmoodi
- Department of Civil, Architectural and Environmental Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
37
|
Poh WT, Stanslas J. The new paradigm in animal testing - "3Rs alternatives". Regul Toxicol Pharmacol 2024; 153:105705. [PMID: 39299677 DOI: 10.1016/j.yrtph.2024.105705] [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: 03/25/2024] [Revised: 07/07/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Regulatory studies have revolutionised over time. Today, the focus has shifted from animal toxicity testing to non-animal for regulatory safety testing. This move is in line with the international 3Rs (Replacement, Reduction, and Refinement) principle and has also changed the regulator's perspective. The 3R principle has stimulated changes in policy, regulations, and new approaches to safety assessment in drug development in many countries. The 3Rs approach has led to the discovery and application of new technologies and more human-relevant in vitro approaches that minimise the use of animals including non-human primates, in research and improve animal welfare. In 2016, the European Medicines Agency published the Guidelines on the principles of regulatory acceptance of 3Rs testing approaches, followed by a conceptual paper in 2023 to align with current 3R standards. Additionally, the United States Food and Drug Administration passed new legislation in 2023 that no longer requires all new human drugs to be tested on animals, which will change the current testing paradigm. This review paper provides the adoption of the 3Rs and the current regulatory perspective regarding their implementation.
Collapse
Affiliation(s)
- Wen Tsin Poh
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
38
|
Panchuk I, Smirnikhina S. Toolbox for creating three-dimensional liver models. Biochem Biophys Res Commun 2024; 731:150375. [PMID: 39018971 DOI: 10.1016/j.bbrc.2024.150375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Research within the hepato-biliary system and hepatic function is currently experiencing heightened interest, this is due to the high frequency of relapse rates observed in chronic conditions, as well as the imperative for the development of innovative therapeutic strategies to address both inherited and acquired diseases within this domain. The most commonly used sources for studying hepatocytes include primary human hepatocytes, human hepatic cancer cell lines, and hepatic-like cells derived from induced pluripotent stem cells. However, a significant challenge in primary hepatic cell culture is the rapid decline in their phenotypic characteristics, dedifferentiation and short cultivation time. This limitation creates various problems, including the inability to maintain long-term cell cultures, which can lead to failed experiments in drug development and the creation of relevant disease models for researchers' purposes. To address these issues, the creation of a powerful 3D cell model could play a pivotal role as a personalized disease model and help reduce the use of animal models during certain stages of research. Such a cell model could be used for disease modelling, genome editing, and drug discovery purposes. This review provides an overview of the main methods of 3D-culturing liver cells, including a discussion of their characteristics, advantages, and disadvantages.
Collapse
Affiliation(s)
- Irina Panchuk
- Research Centre for Medical Genetics, Moscow, Russian Federation.
| | | |
Collapse
|
39
|
Driche EH, Badji B, Mathieu F, Zitouni A. In-vitro antibacterial and antibiofilm activities and in-silico analysis of a potent cyclic peptide from a novel Streptomyces sp. strain RG-5 against antibiotic-resistant and biofilm-forming pathogenic bacteria. Arch Microbiol 2024; 206:450. [PMID: 39476249 DOI: 10.1007/s00203-024-04174-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/10/2024]
Abstract
The proliferation of multidrug-resistant and biofilm-forming pathogenic bacteria poses a serious threat to public health. The limited effectiveness of current antibiotics motivates the search for new antibacterial compounds. In this study, a novel strain, RG-5, was isolated from desert soil. This strain exhibited potent antibacterial and antibiofilm properties against multidrug-resistant and biofilm-forming pathogenic bacteria. Through phenotypical characterizations, 16S rRNA gene sequence and phylogenetic analysis, the strain was identified as Streptomyces pratensis with 99.8% similarity. The active compound, RG5-1, was extracted, purified by reverse phase silica column HPLC, identified by ESI-MS spectrometry, and confirmed by 1H and 13C NMR analysis as 2,5-Piperazinedione, 3,6-bis(2-methylpropyl), belonging to cyclic peptides. This compound showed interesting minimum inhibitory concentrations (MICs) of 04 to 15 µg/mL and minimum biofilm inhibitory concentrations (MBICs 50%) of ½ MIC against the tested bacteria. Its molecular mechanism of action was elucidated through a molecular docking study against five drug-protein targets. The results demonstrated that the compound RG5-1 has a strong affinity and interaction patterns with glucosamine-6-phosphate synthase at - 6.0 kcal/mol compared to reference inhibitor (- 5.4 kcal/mol), medium with penicillin-binding protein 1a (- 6.1 kcal/mol), and LasR regulator protein of quorum sensing (- 5.4 kcal/mol), confirming its antibacterial and antibiofilm activities. The compound exhibited minimal toxicity and favorable physicochemical and pharmacological properties. This is the first report that describes its production from Streptomyces, its activities against biofilm-forming and multidrug-resistant bacteria, and its mechanism of action. These findings indicate that 2,5-piperazinedione, 3,6-bis(2-methylpropyl) has the potential to be a promising lead compound in the treatment of antibiotic-resistant and biofilm-forming pathogens.
Collapse
Affiliation(s)
- El-Hadj Driche
- Laboratory of Molecular Biology, Genomics and Bioinformatics (LBMGB), Department of Biology, Faculty of Nature and Life Sciences (SNV), Hassiba Benbouali University of Chlef, Ouled Fares, Chlef, 02180, Algeria.
- Laboratory of Biology of Microbial Systems (LBSM), Higher Normal School of Kouba B.P. 92, Kouba, Alger, 16050, Algeria.
| | - Boubekeur Badji
- Laboratory of Biology of Microbial Systems (LBSM), Higher Normal School of Kouba B.P. 92, Kouba, Alger, 16050, Algeria
| | - Florence Mathieu
- Chemical Engineering Laboratory, LGC, UMR 5503 (CNRS/INPT/UPS), University of Toulouse, Toulouse, France
| | - Abdelghani Zitouni
- Laboratory of Biology of Microbial Systems (LBSM), Higher Normal School of Kouba B.P. 92, Kouba, Alger, 16050, Algeria
| |
Collapse
|
40
|
Kastrinou-Lampou V, Rodríguez-Pérez R, Poller B, Huth F, Gáborik Z, Mártonné-Tóth B, Temesszentandrási-Ambrus C, Schadt HS, Kullak-Ublick GA, Arand M, Camenisch G. Identification of reversible OATP1B1 and time-dependent CYP3A4 inhibition as the major risk factors for drug-induced cholestasis (DIC). Arch Toxicol 2024; 98:3409-3424. [PMID: 39023798 DOI: 10.1007/s00204-024-03794-3] [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: 09/28/2023] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Abstract
Hepatic bile acid regulation is a multifaceted process modulated by several hepatic transporters and enzymes. Drug-induced cholestasis (DIC), a main type of drug-induced liver injury (DILI), denotes any drug-mediated condition in which hepatic bile flow is impaired. Our ability in translating preclinical toxicological findings to human DIC risk is currently very limited, mainly due to important interspecies differences. Accordingly, the anticipation of clinical DIC with available in vitro or in silico models is also challenging, due to the complexity of the bile acid homeostasis. Herein, we assessed the in vitro inhibition potential of 47 marketed drugs with various degrees of reported DILI severity towards all metabolic and transport mechanisms currently known to be involved in the hepatic regulation of bile acids. The reported DILI concern and/or cholestatic annotation correlated with the number of investigated processes being inhibited. Furthermore, we employed univariate and multivariate statistical methods to determine the important processes for DILI discrimination. We identified time-dependent inhibition (TDI) of cytochrome P450 (CYP) 3A4 and reversible inhibition of the organic anion transporting polypeptide (OATP) 1B1 as the major risk factors for DIC among the tested mechanisms related to bile acid transport and metabolism. These results were consistent across multiple statistical methods and DILI classification systems applied in our dataset. We anticipate that our assessment of the two most important processes in the development of cholestasis will enable a risk assessment for DIC to be efficiently integrated into the preclinical development process.
Collapse
Affiliation(s)
- Vlasia Kastrinou-Lampou
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland
- Preclinical Safety, BioMedical Research, Novartis, Basel, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Birk Poller
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland
| | - Felix Huth
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland
| | - Zsuzsanna Gáborik
- SOLVO Biotechnology, Charles River Laboratories Hungary, 1117, Budapest, Hungary
| | - Beáta Mártonné-Tóth
- SOLVO Biotechnology, Charles River Laboratories Hungary, 1117, Budapest, Hungary
| | | | - Heiko S Schadt
- Preclinical Safety, BioMedical Research, Novartis, Basel, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Mechanistic Safety, CMO & Patient Safety, Global Drug Development, Novartis, Basel, Switzerland
| | - Michael Arand
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Gian Camenisch
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland.
| |
Collapse
|
41
|
Dey S, Bhat A, Janani G, Shandilya V, Gupta R, Mandal BB. Microfluidic human physiomimetic liver model as a screening platform for drug induced liver injury. Biomaterials 2024; 310:122627. [PMID: 38823194 DOI: 10.1016/j.biomaterials.2024.122627] [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: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The pre-clinical animal models often fail to predict intrinsic and idiosyncratic drug induced liver injury (DILI), thus contributing to drug failures in clinical trials, black box warnings and withdrawal of marketed drugs. This suggests a critical need for human-relevant in vitro models to predict diverse DILI phenotypes. In this study, a porcine liver extracellular matrix (ECM) based biomaterial ink with high printing fidelity, biocompatibility and tunable rheological and mechanical properties is formulated for supporting both parenchymal and non-parenchymal cells. Further, we applied 3D printing and microfluidic technology to bioengineer a human physiomimetic liver acinus model (HPLAM), recapitulating the radial hepatic cord-like structure with functional sinusoidal microvasculature network, biochemical and biophysical properties of native liver acinus. Intriguingly, the human derived hepatic cells incorporated HPLAM cultured under physiologically relevant microenvironment, acts as metabolic biofactories manifesting enhanced hepatic functionality, secretome levels and biomarkers expression over several weeks. We also report that the matured HPLAM reproduces dose- and time-dependent hepatotoxic response of human clinical relevance to drugs typically recognized for inducing diverse DILI phenotypes as compared to conventional static culture. Overall, the developed HPLAM emulates in vivo like functions and may provide a useful platform for DILI risk assessment to better determine safety and human risk.
Collapse
Affiliation(s)
- Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Amritha Bhat
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - G Janani
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Vartik Shandilya
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Biman B Mandal
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
42
|
Segovia-Zafra A, Villanueva-Paz M, Serras AS, Matilla-Cabello G, Bodoque-García A, Di Zeo-Sánchez DE, Niu H, Álvarez-Álvarez I, Sanz-Villanueva L, Godec S, Milisav I, Bagnaninchi P, Andrade RJ, Lucena MI, Fernández-Checa JC, Cubero FJ, Miranda JP, Nelson LJ. Control compounds for preclinical drug-induced liver injury assessment: Consensus-driven systematic review by the ProEuroDILI network. J Hepatol 2024; 81:630-640. [PMID: 38703829 DOI: 10.1016/j.jhep.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/10/2024] [Accepted: 04/21/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND & AIMS Idiosyncratic drug-induced liver injury (DILI) is a complex and unpredictable event caused by drugs, and herbal or dietary supplements. Early identification of human hepatotoxicity at preclinical stages remains a major challenge, in which the selection of validated in vitro systems and test drugs has a significant impact. In this systematic review, we analyzed the compounds used in hepatotoxicity assays and established a list of DILI-positive and -negative control drugs for validation of in vitro models of DILI, supported by literature and clinical evidence and endorsed by an expert committee from the COST Action ProEuroDILI Network (CA17112). METHODS Following 2020 PRISMA guidelines, original research articles focusing on DILI which used in vitro human models and performed at least one hepatotoxicity assay with positive and negative control compounds, were included. Bias of the studies was assessed by a modified 'Toxicological Data Reliability Assessment Tool'. RESULTS A total of 51 studies (out of 2,936) met the inclusion criteria, with 30 categorized as reliable without restrictions. Although there was a broad consensus on positive compounds, the selection of negative compounds lacked clarity. 2D monoculture, short exposure times and cytotoxicity endpoints were the most tested, although there was no consensus on drug concentrations. CONCLUSIONS Extensive analysis highlighted the lack of agreement on control compounds for in vitro DILI assessment. Following comprehensive in vitro and clinical data analysis together with input from the expert committee, an evidence-based consensus-driven list of 10 positive and negative control drugs for validation of in vitro models of DILI is proposed. IMPACT AND IMPLICATIONS Prediction of human toxicity early in the drug development process remains a major challenge, necessitating the development of more physiologically relevant liver models and careful selection of drug-induced liver injury (DILI)-positive and -negative control drugs to better predict the risk of DILI associated with new drug candidates. Thus, this systematic study has crucial implications for standardizing the validation of new in vitro models of DILI. By establishing a consensus-driven list of positive and negative control drugs, the study provides a scientifically justified framework for enhancing the consistency of preclinical testing, thereby addressing a significant challenge in early hepatotoxicity identification. Practically, these findings can guide researchers in evaluating safety profiles of new drugs, refining in vitro models, and informing regulatory agencies on potential improvements to regulatory guidelines, ensuring a more systematic and efficient approach to drug safety assessment.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Marina Villanueva-Paz
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Ana Sofia Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Gonzalo Matilla-Cabello
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Ana Bodoque-García
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - Daniel E Di Zeo-Sánchez
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Hao Niu
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - Ismael Álvarez-Álvarez
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Laura Sanz-Villanueva
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy VIC, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Sergej Godec
- Department of Anaesthesiology and Surgical Intensive Care, University Medical Centre Ljubljana, Ljubljana, Slovenia; Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of oxidative stress research, Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Pierre Bagnaninchi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Raúl J Andrade
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Plataforma de Investigación Clínica y Ensayos Clínicos UICEC-IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain
| | - M Isabel Lucena
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Plataforma de Investigación Clínica y Ensayos Clínicos UICEC-IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain.
| | - José C Fernández-Checa
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Medicine, Keck School of Division of Gastrointestinal and Liver disease, University of Southern California, Los Angeles, CA, United States.
| | - Francisco Javier Cubero
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Joana Paiva Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Leonard J Nelson
- Institute for Bioengineering, School of Engineering, Faraday Building, The University of Edinburgh, Scotland, United Kingdom
| |
Collapse
|
43
|
Tecchio KB, Alves FDM, Alves JD, Barbosa CDS, Salgado MAR, Santos VJDSVD, Varotti FDP, Campos-Junior PHDA, Viana GHR, Santos FVD. Evaluation of the in vivo acute toxicity and in vitro genotoxicity and mutagenicity of synthetic β-carboline alkaloids with selective cytotoxic activity against ovarian and breast cancer cell lines. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 899:503808. [PMID: 39326936 DOI: 10.1016/j.mrgentox.2024.503808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 09/28/2024]
Abstract
The aim of this study was to evaluate the in vitro cytotoxic, genotoxic, and mutagenic potential and to determine the in silico ADME parameters of two synthetic β-carboline alkaloids developed as prototypes of antitumor agents (NQBio-06 and NQBio-21). Additionally, acute toxicity of the compounds was evaluated in mice. The results from the MTT assay showed that NQBio-06 presented higher cytotoxicity in the ovarian cancer cell line TOV-21 G (IC50 = 2.5 µM, selectivity index = 23.7). NQBio-21 presented an IC50 of 6.9 µM and a selectivity index of 14.5 against MDA-MB-231 breast cancer cells. Comet assay results showed that NQBio-06 did not induce chromosomal breaks in vitro, but NQBio-21 was genotoxic with and without metabolic activation (S9 fraction). Micronucleus assay showed that both compounds were mutagenic. In addition, metabolic activation enhanced this effect in vitro. The in silico predictions showed that the compounds met the criteria set by Lipinski's rules, had strong prediction for intestinal absorption, and were possible substrates for P-glycoprotein. The in vivo results demonstrated that both the compounds exhibited low acute toxicity. These results suggest that the mechanisms underlying the cytotoxicity of NQBio-06 and NQBio-21 are related to DNA damage induction and that the use of S9 enhanced these effects. In vivo analysis showed signs of toxicity after a single administration of the compounds in mice. These findings highlight the potential of β-carboline compounds as sources for the development of new anticancer chemotherapeutic agents.
Collapse
Affiliation(s)
- Kimberly Brito Tecchio
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil; Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Fernanda de Moura Alves
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Janaina Domingas Alves
- Laboratório de Pesquisa em Reprodução, Departamento de Ciências Naturais, Universidade Federal de São João del Rei, Campus Dom Bosco, São João del-Rei, MG 36301-160, Brazil
| | - Camila de Souza Barbosa
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Mariana Alves Rezende Salgado
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Vanessa Jaqueline da Silva Vieira Dos Santos
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil; Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Fernando de Pilla Varotti
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Paulo Henrique de Almeida Campos-Junior
- Laboratório de Pesquisa em Reprodução, Departamento de Ciências Naturais, Universidade Federal de São João del Rei, Campus Dom Bosco, São João del-Rei, MG 36301-160, Brazil
| | - Gustavo Henrique Ribeiro Viana
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil
| | - Fabio Vieira Dos Santos
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil; Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Campus Centro Oeste, Divinópolis, MG 35501-296, Brazil.
| |
Collapse
|
44
|
Godschalk R, Faulk C, LaRocca J, van Benthem J, Marchetti F. Epigenotoxicity: Decoding the epigenetic imprints of genotoxic agents and their implications for regulatory genetic toxicology. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024. [PMID: 39262275 DOI: 10.1002/em.22626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
Regulatory genetic toxicology focuses on DNA damage and subsequent gene mutations. However, genotoxic agents can also affect epigenetic marks, and incorporation of epigenetic data into the regulatory framework may thus enhance the accuracy of risk assessment. Additionally, epigenetic alterations may identify non-genotoxic carcinogens that are not captured with the current battery of tests. Epigenetic alterations could also explain long-term consequences and potential transgenerational effects in the absence of DNA mutations. Therefore, at the 2022 International Workshops on Genotoxicity Testing (IWGT) in Ottawa (Ontario, Canada), an expert workgroup explored whether including epigenetic endpoints would improve regulatory genetic toxicology. Here we summarize the presentations and the discussions on technical advancements in assessing epigenetics, how the assessment of epigenetics can enhance strategies for detecting genotoxic and non-genotoxic carcinogens and the correlation between epigenetic alterations with other relevant apical endpoints.
Collapse
Affiliation(s)
- Roger Godschalk
- Department of Pharmacology and Toxicology, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | | | | | - Jan van Benthem
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
45
|
Jiao D, Xie L, Xing W. A pumpless liver-on-a-chip for drug hepatotoxicity analysis. Analyst 2024; 149:4675-4686. [PMID: 39086194 DOI: 10.1039/d4an00602j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
This study presents the development and validation of an innovative microfluidic liver-on-a-chip device utilizing gravity-driven perfusion for the evaluation of drug hepatotoxicity. This research involved the construction of a hydrogel-based coculture chip that integrates liver parenchymal and stellate cells within a tri-channel configuration. The assembly and operation of the liver-on-a-chip and its accompanying custom rocker were straightforward. The cells in the chip maintained high viability and continuously synthesized liver albumin over extended culture durations. Acetaminophen (APAP), a hepatic injury-inducing drug, was utilized as a positive control in hepatic toxicity assays on the chip. The liver chip exhibited hepatotoxic responses comparable to those observed in 2D models. Furthermore, in this study we evaluated the effects of two plant-derived natural compounds, aristolochic acid I (AA) and its analog aristolactam AII (AL), in both 2D cell models and the liver-on-a-chip system. AA, known for its hepatorenal toxicity, was observed to cause hepatotoxicity in both the 2D models and on the chip. The flow cytometry and mRNA sequencing results confirmed the propensity of these compounds to induce liver cell apoptosis. Notably, AL, previously considered nontoxic, provoked a significant decrease in the hepatic functionality marker albumin exclusively in the liver chip but not in 2D models, indicating the liver chip's enhanced sensitivity to toxic substances. In summary, this pumpless liver-on-a-chip is a simple yet powerful tool for drug hepatotoxicity studies.
Collapse
Affiliation(s)
- Dian Jiao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lan Xie
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing, China.
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Wanli Xing
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing, China.
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| |
Collapse
|
46
|
Nicosia N, Kwiecień I, Bednarski M, Głuch-Lutwin M, Mordyl B, Mika K, Hambaryan R, Miller A, Alesci A, Lauriano ER, Zammit P, Ragusa S, Trojan E, Fumia A, Sapa J, Miceli N, Kotańska M. Anti-diabetes and neuroprotection potential and primary safety studies of Isatis tinctoria L. hydroalcoholic leaf extract. Fitoterapia 2024; 177:106138. [PMID: 39053741 DOI: 10.1016/j.fitote.2024.106138] [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: 05/01/2024] [Revised: 07/21/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE Natural plant raw materials, previously underestimated in therapeutics, are becoming the subject of research for new applications in medicine. In our research, the hydroalcoholic extract of Isatis tinctoria leaf, rich in flavonoid compounds such as vicenin-2 and quercetin, was examined as a potential antidiabetic and neuroprotective agent. METHODS The effect of the extract and its main flavonoid compounds on protein glycation, alpha-glucosidase activity, and acetylcholinesterase activity was tested. In vitro, in the mouse hippocampal neuronal cell line and in vivo, using a mouse model, the safety of the extract was screened for. RESULTS Our experiments demonstrated significant inhibition of protein glycation, alpha-glucosidase activity, acetylcholinesterase activity, and β-amyloid aggregation by the extract, in a concentration-dependent manner. The extract had a strong reducing effect and did not exhibit cytotoxicity up to a concentration of 25 mg/mL. Intraperitoneal administration of the extract to mice did not have negative effects on body mass, locomotor activity, coordination, and liver cell integrity. CONCLUSIONS Our research sheds new light on this raw material and deepens knowledge of its activity. This may result in the recognition of its therapeutic effects and even in its introduction in the modern treatment of diseases characterized by pathological changes associated with hyperglycemia, oxidation, and inflammation.
Collapse
Affiliation(s)
- Noemi Nicosia
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland; PhD Program in Neuroscience, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy; Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy; Foundation "Prof. Antonio Imbesi", University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy
| | - Inga Kwiecień
- Department of Medicinal Plant and Mushroom Biotechnology, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland
| | - Marek Bednarski
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Barbara Mordyl
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Kamil Mika
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland
| | - Rimma Hambaryan
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland
| | - Anthea Miller
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland; Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Alessio Alesci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Eugenia R Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Paula Zammit
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland; University of Malta, Msida MSD 2080, Malta
| | - Salvatore Ragusa
- PLANTA/Autonomous Center for Research, Documentation and Training, Via Serraglio Vecchio 28, 90123 Palermo, Italy
| | - Ewa Trojan
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland; Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Str., 31-343 Kraków, Poland
| | - Angelo Fumia
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy
| | - Jacek Sapa
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland
| | - Natalizia Miceli
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Cracow, Poland.
| |
Collapse
|
47
|
Daley MC, Moreau M, Bronk P, Fisher J, Kofron CM, Mende U, McMullen P, Choi BR, Coulombe K. In vitro to in vivo extrapolation from 3D hiPSC-derived cardiac microtissues and physiologically based pharmacokinetic modeling to inform next-generation arrhythmia risk assessment. Toxicol Sci 2024; 201:145-157. [PMID: 38897660 PMCID: PMC11347779 DOI: 10.1093/toxsci/kfae079] [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] [Indexed: 06/21/2024] Open
Abstract
Proarrhythmic cardiotoxicity remains a substantial barrier to drug development as well as a major global health challenge. In vitro human pluripotent stem cell-based new approach methodologies have been increasingly proposed and employed as alternatives to existing in vitro and in vivo models that do not accurately recapitulate human cardiac electrophysiology or cardiotoxicity risk. In this study, we expanded the capacity of our previously established 3D human cardiac microtissue model to perform quantitative risk assessment by combining it with a physiologically based pharmacokinetic model, allowing a direct comparison of potentially harmful concentrations predicted in vitro to in vivo therapeutic levels. This approach enabled the measurement of concentration responses and margins of exposure for 2 physiologically relevant metrics of proarrhythmic risk (i.e. action potential duration and triangulation assessed by optical mapping) across concentrations spanning 3 orders of magnitude. The combination of both metrics enabled accurate proarrhythmic risk assessment of 4 compounds with a range of known proarrhythmic risk profiles (i.e. quinidine, cisapride, ranolazine, and verapamil) and demonstrated close agreement with their known clinical effects. Action potential triangulation was found to be a more sensitive metric for predicting proarrhythmic risk associated with the primary mechanism of concern for pharmaceutical-induced fatal ventricular arrhythmias, delayed cardiac repolarization due to inhibition of the rapid delayed rectifier potassium channel, or hERG channel. This study advances human-induced pluripotent stem cell-based 3D cardiac tissue models as new approach methodologies that enable in vitro proarrhythmic risk assessment with high precision of quantitative metrics for understanding clinically relevant cardiotoxicity.
Collapse
Affiliation(s)
- Mark C Daley
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, United States
| | | | - Peter Bronk
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, United States
| | | | - Celinda M Kofron
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, United States
| | - Ulrike Mende
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, United States
| | | | - Bum-Rak Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Kareen Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, United States
| |
Collapse
|
48
|
Garcia KR, Menezes RCR, Dos Santos V, Koester LS, Dallegrave E. Toward a greener multifunctional pharmaceutical excipient: in vivo safety evaluation of nanofibrillated cellulose from tobacco stalk. Drug Chem Toxicol 2024; 47:507-515. [PMID: 38326987 DOI: 10.1080/01480545.2024.2311288] [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: 08/03/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 02/09/2024]
Abstract
Tobacco stalk is a cellulose-rich material and a sustainable alternative to be applied as a plant-based nanofibrillated cellulose (NFC) source. NFC use has garnered attention in the development of oral pharmaceutical forms, despite concerns about its safety due to the adverse effects of nicotine on health. Therefore, we aimed at establishing the safety of NFC derived from tobacco stalk for its potential use as a novel pharmaceutical excipient, exploring its potential functions for tablet production. We conducted acute and subchronic oral toxicity tests in adult female Wistar rats. Initially, individual animals received sequential doses (175-5,000 mg·kg-1) for 24 hours followed by a careful observation of any toxic effects. Subsequently, 20 rats were divided into four groups for a subchronic assay, evaluating toxicity signs, body weight changes, hematological, biochemical, and histopathological parameters. No deaths or other clinical toxicity signs were observed in either the acute or the subchronic assays. We noticed a significant reduction in body weight gain (p < 0.05) after 14 days. We found statistical differences for hematological and biochemical parameters, unrelated to dosage. There were no observed toxic effects, and tobacco stalk ingestion did not adversely affect organ morphology in the histopathological evaluation. The oral administration of NFC at 5,000 mg·kg-1 per day for 28 days was well-tolerated by treated rats, with no reported deaths. In conclusion, NFC derived from tobacco stalk has shown to be a sustainable and safe alternative for use as an excipient at experimental doses, demonstrating compatibility with its proposed applications.
Collapse
Affiliation(s)
- Keth Ribeiro Garcia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Rafaella Câmara Rocha Menezes
- Programa de Pós-Graduação em Ciências da Nutrição, Laboratório de Pesquisa em Toxicologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, Brazil
| | - Venina Dos Santos
- Programa de Pós-Graduação em Engenharia de Processos e Tecnologias, Universidade de Caxias do Sul (UCS), Rua Francisco Getúlio Vargas, Caxias do Sul, RS, Brazil
| | - Letícia Scherer Koester
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Eliane Dallegrave
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Pesquisa em Toxicologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, Brazil
| |
Collapse
|
49
|
Pike CM, Levi JA, Boone LA, Peddibhotla S, Johnson J, Zwarycz B, Bunger MK, Thelin W, Boazak EM. High-Throughput Assay for Predicting Diarrhea Risk Using a 2D Human Intestinal Stem Cell-Derived Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610072. [PMID: 39257790 PMCID: PMC11383669 DOI: 10.1101/2024.08.28.610072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Gastrointestinal toxicities (GITs) are the most prevalent adverse events (AE) reported in clinical trials, often resulting in dose-limitations that reduce drug efficacy and delay development and treatment optimization. Preclinical animal models do not accurately replicate human GI physiology, leaving few options for early detection of GI side effects prior to human studies. Development of an accurate model that predicts GIT earlier in drug discovery programs would better support successful clinical trial outcomes. Chemotherapeutics, which exhibit high rates of clinical GIT, frequently target mitotic cells. Therefore, we hypothesized that a model utilizing proliferative cell populations derived from human intestinal crypts would predict the occurrence of clinical GITs with high accuracy. Here, we describe the development of a multiparametric assay utilizing the RepliGut® Planar system, an intestinal stem cell-derived platform cultured in an accessible high throughput Transwell™ format. This assay addresses key physiological elements of GIT by assessing cell proliferation (EdU incorporation), cell abundance (DAPI quantification), and barrier function (TEER). Using this approach, we demonstrate that primary proliferative cell populations reproducibly respond to marketed chemotherapeutics at physiologic concentrations. To determine the ability of this model to predict clinical diarrhea risk, we evaluated a set of 30 drugs with known clinical diarrhea incidence in three human donors, comparing results to known plasma drug concentrations. This resulted in highly accurate predictions of diarrhea potential for each endpoint (balanced accuracy of 91% for DAPI, 90% for EdU, 88% for TEER) with minimal variation across human donors. In vitro toxicity screening using primary proliferative cells may enable improved safety evaluations, reducing the risk of AEs in clinical trials and ultimately lead to safer and more effective treatments for patients.
Collapse
|
50
|
Kang S, Chen EC, Cifuentes H, Co JY, Cole G, Graham J, Hsia R, Kiyota T, Klein JA, Kroll KT, Nieves Lopez LM, Norona LM, Peiris H, Potla R, Romero-Lopez M, Roth JG, Tseng M, Fullerton AM, Homan KA. Complex in vitromodels positioned for impact to drug testing in pharma: a review. Biofabrication 2024; 16:042006. [PMID: 39189069 DOI: 10.1088/1758-5090/ad6933] [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: 12/22/2023] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.
Collapse
Affiliation(s)
- Serah Kang
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Eugene C Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Helen Cifuentes
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julia Y Co
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Gabrielle Cole
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica Graham
- Product Quality & Occupational Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of Americaica
| | - Rebecca Hsia
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Tomomi Kiyota
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica A Klein
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Katharina T Kroll
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Lenitza M Nieves Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Leah M Norona
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Heshan Peiris
- Human Genetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Ratnakar Potla
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Monica Romero-Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julien G Roth
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Min Tseng
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Aaron M Fullerton
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Kimberly A Homan
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| |
Collapse
|