1
|
Schoeberl B, Musante CJ, Ramanujan S. Future Directions for Quantitative Systems Pharmacology. Handb Exp Pharmacol 2025. [PMID: 39812657 DOI: 10.1007/164_2024_737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
In this chapter, we envision the future of Quantitative Systems Pharmacology (QSP) which integrates closely with emerging data and technologies including advanced analytics, novel experimental technologies, and diverse and larger datasets. Machine learning (ML) and Artificial Intelligence (AI) will increasingly help QSP modelers to find, prepare, integrate, and exploit larger and diverse datasets, as well as build, parameterize, and simulate models. We picture QSP models being applied during all stages of drug discovery and development: During the discovery stages, QSP models predict the early human experience of in silico compounds created by generative AI. In preclinical development, QSP will integrate with non-animal "new approach methodologies" and reverse-translated datasets to improve understanding of and translation to the human patient. During clinical development, integration with complementary modeling approaches and multimodal patient data will create multidimensional digital twins and virtual populations for clinical trial simulations that guide clinical development and point to opportunities for precision medicine. QSP can evolve into this future by (1) pursuing high-impact applications enabled by novel experimental and quantitative technologies and data types; (2) integrating closely with analytical and computational advancements; and (3) increasing efficiencies through automation, standardization, and model reuse. In this vision, the QSP expert will play a critical role in designing strategies, evaluating data, staging and executing analyses, verifying, interpreting, and communicating findings, and ensuring the ethical, safe, and rational application of novel data types, technologies, and advanced analytics including AI/ML.
Collapse
|
2
|
Beattie KA, Verma M, Brennan RJ, Clausznitzer D, Damian V, Leishman D, Spilker ME, Boras B, Li Z, Oziolor E, Rieger TR, Sher A. Quantitative systems toxicology modeling in pharmaceutical research and development: An industry-wide survey and selected case study examples. CPT Pharmacometrics Syst Pharmacol 2024; 13:2036-2051. [PMID: 39412216 PMCID: PMC11646944 DOI: 10.1002/psp4.13227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 07/15/2024] [Accepted: 08/07/2024] [Indexed: 12/17/2024] Open
Abstract
Quantitative systems toxicology (QST) models are increasingly being applied for predicting and understanding toxicity liabilities in pharmaceutical research and development. A European Federation of Pharmaceutical Industries and Associations (EFPIA)-wide survey was completed by 15 companies. The results provide insights into the current use of QST models across the industry. 73% of responding companies with more than 10,000 employees utilize QST models. The most applied QST models are for liver, cardiac electrophysiology, and bone marrow/hematology. Responders indicated particular interest in QST models for the central nervous system (CNS), kidney, lung, and skin. QST models are used to support decisions in both preclinical and clinical stages of pharmaceutical development. The survey suggests high demand for QST models and resource limitations were indicated as a common obstacle to broader use and impact. Increased investment in QST resources and training may accelerate application and impact. Case studies of QST model use in decision-making within EFPIA companies are also discussed. This article aims to (i) share industry experience and learnings from applying QST models to inform decision-making in drug discovery and development programs, and (ii) share approaches taken during QST model development and validation and compare these with recommendations for modeling best practices and frameworks proposed in the literature. Discussion of QST-specific applications in relation to these modeling frameworks is relevant in the context of the recently proposed International Council for Harmonization (ICH) M15 guideline on general principles for Model-Informed Drug Development (MIDD).
Collapse
Affiliation(s)
| | - Meghna Verma
- Systems Medicine, Clinical Pharmacology and Quantitative PharmacologyR&D BioPharmaceuticals, AstraZenecaGaithersburgMarylandUSA
| | | | - Diana Clausznitzer
- Quantitative, Translational and ADME SciencesAbbVie DeutschlandLudwigshafenGermany
| | - Valeriu Damian
- Computational SciencesGSKUpper ProvidencePennsylvaniaUSA
| | - Derek Leishman
- Translational and Quantitative ToxicologyEli Lilly and CompanyIndianapolisIndianaUSA
| | - Mary E. Spilker
- Pharmacokinetics, Dynamics and MetabolismPfizer Research and Development, Pfizer Inc.La JollaCaliforniaUSA
| | - Britton Boras
- Pharmacokinetics, Dynamics and MetabolismPfizer Research and Development, Pfizer Inc.La JollaCaliforniaUSA
| | - Zhenhong Li
- Translational Modeling and SimulationPfizer Research and Development, Pfizer Inc.CambridgeMassachusettsUSA
| | - Elias Oziolor
- Drug Safety Research and DevelopmentPfizer Research and Development, Pfizer Inc.GrotonConnecticutUSA
| | - Theodore R. Rieger
- Pharmacometrics and Systems PharmacologyPfizer Research and Development, Pfizer Inc.CambridgeMassachusettsUSA
| | - Anna Sher
- Clinical Pharmacology Modeling and SimulationGSKWalthamMassachusettsUSA
| |
Collapse
|
3
|
Ambe K, Nakamori M, Tohno R, Suzuki K, Sasaki T, Tohkin M, Yoshinari K. Machine Learning-Based In Silico Prediction of the Inhibitory Activity of Chemical Substances Against Rat and Human Cytochrome P450s. Chem Res Toxicol 2024; 37:1843-1850. [PMID: 39427263 DOI: 10.1021/acs.chemrestox.4c00168] [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: 10/22/2024]
Abstract
The prediction of cytochrome P450 inhibition by a computational (quantitative) structure-activity relationship approach using chemical structure information and machine learning would be useful for toxicity research as a simple and rapid in silico tool. However, there are few in silico models focusing on the species differences between rat and human in the P450s inhibition. This study aimed to establish in silico models to classify chemical substances as inhibitors or non-inhibitors of various rat and human P450s, using only molecular descriptors. Using the in-house test results from our in vitro experiments, we used 326 substances for model construction and internal validation data. Apart from the 326 substances, 60 substances were used as external validation data set. We focused on seven rat P450s (CYP1A1, CYP1A2, CYP2B1, CYP2C6, CYP2D1, CYP2E1, and CYP3A2) and 11 human P450s (CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4). Most of the models established using XGBoost showed an area under the receiver operating characteristic curve (ROC-AUC) of 0.8 or more in the internal validation. When we set an applicability domain for the models and confirmed their generalization performance through external validation, most of the models showed an ROC-AUC of 0.7 or more. Interestingly, for CYP1A1 and CYP1A2, we discovered that a human P450 inhibitory activity model can predict rat P450 inhibitory activity and vice versa. These models are the first attempts to predict inhibitory activity against a wide variety of P450s in both rats and humans using chemical structure information. Our experimental results and in silico models would be helpful to support information for species similarities and differences in chemical-induced toxicity.
Collapse
Affiliation(s)
- Kaori Ambe
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Mizuki Nakamori
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Riku Tohno
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Kotaro Suzuki
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Takamitsu Sasaki
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 4228526, Japan
| | - Masahiro Tohkin
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 4228526, Japan
| |
Collapse
|
4
|
Tarasiuk O, Invernizzi C, Alberti P. In vitro neurotoxicity testing: lessons from chemotherapy-induced peripheral neurotoxicity. Expert Opin Drug Metab Toxicol 2024; 20:1037-1052. [PMID: 39246127 DOI: 10.1080/17425255.2024.2401584] [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: 04/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Chemotherapy induced peripheral neurotoxicity (CIPN) is a long-lasting, or even permanent, late toxicity caused by largely used anticancer drugs. CIPN affects a growing population of cancer survivors and diminishes their quality of life since there is no curative/preventive treatment. Among several reasons for this unmet clinical need, there is an incomplete knowledge on mechanisms leading to CIPN. Therefore, bench side research is still greatly needed: in vitro studies are pivotal to both evaluate neurotoxicity mechanisms and potential neuroprotection strategies. AREAS COVERED Advantages and disadvantages of in vitro approaches are addressed with respect to their applicability to the CIPN field. Different cell cultures and techniques to assess neurotoxicity/neuroprotection are described. PubMed search-string: (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (in vitro) AND (((((model) OR SH-SY5Y) OR PC12) OR iPSC) OR DRG neurons); (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (model) AND (((neurite elongation) OR cell viability) OR morphology). No articles published before 1990 were selected. EXPERT OPINION CIPN is an ideal experimental setting to test axonal damage and, in general, peripheral nervous system mechanisms of disease and neuroprotection. Therefore, starting from robust preclinical data in this field, potentially, relevant biological rationale can be transferred to other human spontaneous diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Chiara Invernizzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- Neuroscience, School of Medicine and Surgery, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
- Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
5
|
He Q, Li M, Ji P, Zheng A, Yao L, Zhu X, Shin JG, Lauschke VM, Han B, Xiang X. Comparison of drug-induced liver injury risk between propylthiouracil and methimazole: A quantitative systems toxicology approach. Toxicol Appl Pharmacol 2024; 491:117064. [PMID: 39122118 DOI: 10.1016/j.taap.2024.117064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Propylthiouracil (PTU) and methimazole (MMI), two classical antithyroid agents possess risk of drug-induced liver injury (DILI) with unknown mechanism of action. This study aimed to examine and compare their hepatic toxicity using a quantitative system toxicology approach. The impact of PTU and MMI on hepatocyte survival, oxidative stress, mitochondrial function and bile acid transporters were assessed in vitro. The physiologically based pharmacokinetic (PBPK) models of PTU and MMI were constructed while their risk of DILI was calculated by DILIsym, a quantitative systems toxicology (QST) model by integrating the results from in vitro toxicological studies and PBPK models. The simulated DILI (ALT >2 × ULN) incidence for PTU (300 mg/d) was 21.2%, which was within the range observed in clinical practice. Moreover, a threshold dose of 200 mg/d was predicted with oxidative stress proposed as an important toxic mechanism. However, DILIsym predicted a 0% incidence of hepatoxicity caused by MMI (30 mg/d), suggesting that the toxicity of MMI was not mediated through mechanism incorporated into DILIsym. In conclusion, DILIsym appears to be a practical tool to unveil hepatoxicity mechanism and predict clinical risk of DILI.
Collapse
Affiliation(s)
- Qingfeng He
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Min Li
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peiying Ji
- Department of Pharmacy, Kong Jiang Hospital of Yangpu District, Shanghai 200093, China
| | - Aole Zheng
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Li Yao
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai 201100, China.
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
6
|
Ahmad F, Muhmood T. Clinical translation of nanomedicine with integrated digital medicine and machine learning interventions. Colloids Surf B Biointerfaces 2024; 241:114041. [PMID: 38897022 DOI: 10.1016/j.colsurfb.2024.114041] [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: 02/01/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Nanomaterials based therapeutics transform the ways of disease prevention, diagnosis and treatment with increasing sophistications in nanotechnology at a breakneck pace, but very few could reach to the clinic due to inconsistencies in preclinical studies followed by regulatory hinderances. To tackle this, integrating the nanomedicine discovery with digital medicine provide technologies as tools of specific biological activity measurement. Hence, overcome the redundancies in nanomedicine discovery by the on-site data acquisition and analytics through integrating intelligent sensors and artificial intelligence (AI) or machine learning (ML). Integrated AI/ML wearable sensors directly gather clinically relevant biochemical information from the subject's body and process data for physicians to make right clinical decision(s) in a time and cost-effective way. This review summarizes insights and recommend the infusion of actionable big data computation enabled sensors in burgeoning field of nanomedicine at academia, research institutes, and pharmaceutical industries, with a potential of clinical translation. Furthermore, many blind spots are present in modern clinically relevant computation, one of which could prevent ML-guided low-cost new nanomedicine development from being successfully translated into the clinic was also discussed.
Collapse
Affiliation(s)
- Farooq Ahmad
- State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources, College of Chemistry, Xinjiang University, Urumqi 830017, China.
| | - Tahir Muhmood
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga 4715-330, Portugal.
| |
Collapse
|
7
|
Stresser DM, Kopec AK, Hewitt P, Hardwick RN, Van Vleet TR, Mahalingaiah PKS, O'Connell D, Jenkins GJ, David R, Graham J, Lee D, Ekert J, Fullerton A, Villenave R, Bajaj P, Gosset JR, Ralston SL, Guha M, Amador-Arjona A, Khan K, Agarwal S, Hasselgren C, Wang X, Adams K, Kaushik G, Raczynski A, Homan KA. Towards in vitro models for reducing or replacing the use of animals in drug testing. Nat Biomed Eng 2024; 8:930-935. [PMID: 38151640 DOI: 10.1038/s41551-023-01154-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Affiliation(s)
- David M Stresser
- Quantitative, Translational & ADME Sciences, AbbVie, North Chicago, IL, USA.
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), .
- IQ Microphysiological Systems Affiliate (IQ-), .
| | - Anna K Kopec
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Drug Safety Research & Development, Pfizer, Inc., Groton, CT, USA
| | - Philip Hewitt
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Chemical and Preclinical Safety, Merck KGaA, Darmstadt, Germany
| | - Rhiannon N Hardwick
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Discovery Toxicology, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, San Diego, CA, USA
| | - Terry R Van Vleet
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology and Pathology, AbbVie, North Chicago, IL, USA
| | - Prathap Kumar S Mahalingaiah
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology and Pathology, AbbVie, North Chicago, IL, USA
| | - Denice O'Connell
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- Global Animal Welfare, AbbVie, North Chicago, IL, USA
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
| | - Gary J Jenkins
- Quantitative, Translational & ADME Sciences, AbbVie, North Chicago, IL, USA
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Translational and ADME Sciences Leadership Group (TALG)
| | - Rhiannon David
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, UK
| | - Jessica Graham
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- Product Quality & Occupational Toxicology, Genentech, Inc., South San Francisco, CA, USA
- IQ DruSafe
- Safety Assessment, Genentech, Inc., South San Francisco, CA, USA
| | - Donna Lee
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
- Safety Assessment, Genentech, Inc., South San Francisco, CA, USA
| | - Jason Ekert
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- UCB Pharma, Cambridge, MA, USA
| | - Aaron Fullerton
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology, Genentech, Inc., South San Francisco, CA, USA
| | - Remi Villenave
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Piyush Bajaj
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA, USA
| | - James R Gosset
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer, Inc, Cambridge, MA, USA
| | - Sherry L Ralston
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
- Preclinical Safety, AbbVie, North Chicago, IL, USA
| | - Manti Guha
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Discovery Biology, Incyte, Wilmington, DE, USA
| | - Alejandro Amador-Arjona
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Discovery Biology, Incyte, Wilmington, DE, USA
| | - Kainat Khan
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, UK
| | - Saket Agarwal
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Investigative Toxicology, Early Development, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Catrin Hasselgren
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ DruSafe
- Predictive Toxicology, Genentech, Inc., South San Francisco, CA, USA
| | - Xiaoting Wang
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Translational Safety & Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Khary Adams
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ 3Rs (Replacement, Reduction, Refinement) Translational and Predictive Sciences Leadership Group
- Laboratory Animal Resources, Incyte, Wilmington, DE, USA
| | - Gaurav Kaushik
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Arkadiusz Raczynski
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ)
- IQ Microphysiological Systems Affiliate (IQ-)
- Preclinical Safety Assessment, Vertex Pharmaceuticals, Inc, Boston, MA, USA
| | - Kimberly A Homan
- International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), .
- IQ Microphysiological Systems Affiliate (IQ-), .
- Complex in vitro Systems Group, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
8
|
Chen C, Lavezzi SM, McDougall D. The estimation and translation uncertainties in applying NOAEL to clinical dose escalation. Clin Transl Sci 2024; 17:e13831. [PMID: 38808564 PMCID: PMC11134224 DOI: 10.1111/cts.13831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/08/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
The systemic exposure at the no-observed-adverse-effect-level (NOAEL) estimated from animals is an important criterion commonly applied to guard the safety of participants in clinical trials of investigational drugs. However, the discrepancy in toxicity profile between species is widely recognized. The objective of the work reported here was to assess, via simulation, the level of uncertainty in the NOAEL estimated from an animal species and the effectiveness of applying its associated exposure value to minimizing the toxicity risk to human. Simulations were conducted for dose escalation of an investigational new chemical entity with hypothetical exposure-response models for the dose-limiting toxicity under a variety of conditions, in terms of between-species relative sensitivity to the toxicity and the between-subject variability in the key parameters determining the sensitivity and pharmacokinetics. Results show a high uncertainty in the NOAEL estimation. Notably, even when the animal species and humans are assumed to have the same sensitivity, which may not be realistic, limiting clinical dose to the exposure at the NOAEL that has been identified in the animals carries a high risk of either causing toxicity or under-dosing, hence undermining the therapeutic potential of the drug candidate. These findings highlight the importance of understanding the mechanism of the toxicity profile and its cross-species translatability, as well as the importance of understanding the dose requirement for achieving adequate pharmacology.
Collapse
Affiliation(s)
- Chao Chen
- Clinical Pharmacology Modelling and SimulationGSKLondonUK
| | - Silvia Maria Lavezzi
- Clinical Pharmacology, Modelling and SimulationParexel InternationalDublinIreland
| | - David McDougall
- Clinical Pharmacology, Modelling and SimulationParexel InternationalBrisbaneQueenslandAustralia
| |
Collapse
|
9
|
Bassan A, Steigerwalt R, Keller D, Beilke L, Bradley PM, Bringezu F, Brock WJ, Burns-Naas LA, Chambers J, Cross K, Dorato M, Elespuru R, Fuhrer D, Hall F, Hartke J, Jahnke GD, Kluxen FM, McDuffie E, Schmidt F, Valentin JP, Woolley D, Zane D, Myatt GJ. Developing a pragmatic consensus procedure supporting the ICH S1B(R1) weight of evidence carcinogenicity assessment. FRONTIERS IN TOXICOLOGY 2024; 6:1370045. [PMID: 38646442 PMCID: PMC11027748 DOI: 10.3389/ftox.2024.1370045] [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: 01/13/2024] [Accepted: 03/04/2024] [Indexed: 04/23/2024] Open
Abstract
The ICH S1B carcinogenicity global testing guideline has been recently revised with a novel addendum that describes a comprehensive integrated Weight of Evidence (WoE) approach to determine the need for a 2-year rat carcinogenicity study. In the present work, experts from different organizations have joined efforts to standardize as much as possible a procedural framework for the integration of evidence associated with the different ICH S1B(R1) WoE criteria. The framework uses a pragmatic consensus procedure for carcinogenicity hazard assessment to facilitate transparent, consistent, and documented decision-making and it discusses best-practices both for the organization of studies and presentation of data in a format suitable for regulatory review. First, it is acknowledged that the six WoE factors described in the addendum form an integrated network of evidence within a holistic assessment framework that is used synergistically to analyze and explain safety signals. Second, the proposed standardized procedure builds upon different considerations related to the primary sources of evidence, mechanistic analysis, alternative methodologies and novel investigative approaches, metabolites, and reliability of the data and other acquired information. Each of the six WoE factors is described highlighting how they can contribute evidence for the overall WoE assessment. A suggested reporting format to summarize the cross-integration of evidence from the different WoE factors is also presented. This work also notes that even if a 2-year rat study is ultimately required, creating a WoE assessment is valuable in understanding the specific factors and levels of human carcinogenic risk better than have been identified previously with the 2-year rat bioassay alone.
Collapse
Affiliation(s)
| | | | - Douglas Keller
- Independent Consultant, Kennett Square, PA, United States
| | - Lisa Beilke
- Toxicology Solutions, Inc., Marana, AZ, United States
| | | | - Frank Bringezu
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - William J. Brock
- Brock Scientific Consulting, LLC, Hilton Head, SC, United States
| | | | | | | | | | | | - Douglas Fuhrer
- BioXcel Therapeutics, Inc., New Haven, CT, United States
| | | | - Jim Hartke
- Gilead Sciences, Inc., Foster City, CA, United States
| | | | | | - Eric McDuffie
- Neurocrine Bioscience, Inc., San Diego, CA, United States
| | | | | | | | - Doris Zane
- Gilead Sciences, Inc., Foster City, CA, United States
| | | |
Collapse
|
10
|
Gall L, Jardi F, Lammens L, Piñero J, Souza TM, Rodrigues D, Jennen DGJ, de Kok TM, Coyle L, Chung S, Ferreira S, Jo H, Beattie KA, Kelly C, Duckworth CA, Pritchard DM, Pin C. A dynamic model of the intestinal epithelium integrates multiple sources of preclinical data and enables clinical translation of drug-induced toxicity. CPT Pharmacometrics Syst Pharmacol 2023; 12:1511-1528. [PMID: 37621010 PMCID: PMC10583244 DOI: 10.1002/psp4.13029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023] Open
Abstract
We have built a quantitative systems toxicology modeling framework focused on the early prediction of oncotherapeutic-induced clinical intestinal adverse effects. The model describes stem and progenitor cell dynamics in the small intestinal epithelium and integrates heterogeneous epithelial-related processes, such as transcriptional profiles, citrulline kinetics, and probability of diarrhea. We fitted a mouse-specific version of the model to quantify doxorubicin and 5-fluorouracil (5-FU)-induced toxicity, which included pharmacokinetics and 5-FU metabolism and assumed that both drugs led to cell cycle arrest and apoptosis in stem cells and proliferative progenitors. The model successfully recapitulated observations in mice regarding dose-dependent disruption of proliferation which could lead to villus shortening, decrease of circulating citrulline, increased diarrhea risk, and transcriptional induction of the p53 pathway. Using a human-specific epithelial model, we translated the cytotoxic activity of doxorubicin and 5-FU quantified in mice into human intestinal injury and predicted with accuracy clinical diarrhea incidence. However, for gefitinib, a specific-molecularly targeted therapy, the mice failed to reproduce epithelial toxicity at exposures much higher than those associated with clinical diarrhea. This indicates that, regardless of the translational modeling approach, preclinical experimental settings have to be suitable to quantify drug-induced clinical toxicity with precision at the structural scale of the model. Our work demonstrates the usefulness of translational models at early stages of the drug development pipeline to predict clinical toxicity and highlights the importance of understanding cross-settings differences in toxicity when building these approaches.
Collapse
Affiliation(s)
- Louis Gall
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&DAstraZenecaCambridgeUK
| | - Ferran Jardi
- Preclinical Sciences & Translational SafetyJanssen Pharmaceutica NVBeerseBelgium
| | - Lieve Lammens
- Preclinical Sciences & Translational SafetyJanssen Pharmaceutica NVBeerseBelgium
| | - Janet Piñero
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM)UPFBarcelonaSpain
| | - Terezinha M. Souza
- Department of Toxicogenomics, GROW School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Daniela Rodrigues
- Department of Toxicogenomics, GROW School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Danyel G. J. Jennen
- Department of Toxicogenomics, GROW School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Theo M. de Kok
- Department of Toxicogenomics, GROW School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Luke Coyle
- Boehringer Ingelheim International GmbHRidgefieldConnecticutUSA
| | | | | | - Heeseung Jo
- Simcyp DivisionCertara UK LimitedSheffieldUK
| | - Kylie A. Beattie
- Target and Systems Safety, Non‐Clinical Safety, In Vivo/In Vitro TranslationGSKStevenageUK
| | - Colette Kelly
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Carrie A. Duckworth
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - D. Mark Pritchard
- Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&DAstraZenecaCambridgeUK
| |
Collapse
|
11
|
Beaudoin JJ, Clemens L, Miedel MT, Gough A, Zaidi F, Ramamoorthy P, Wong KE, Sarangarajan R, Battista C, Shoda LKM, Siler SQ, Taylor DL, Howell BA, Vernetti LA, Yang K. The Combination of a Human Biomimetic Liver Microphysiology System with BIOLOGXsym, a Quantitative Systems Toxicology (QST) Modeling Platform for Macromolecules, Provides Mechanistic Understanding of Tocilizumab- and GGF2-Induced Liver Injury. Int J Mol Sci 2023; 24:9692. [PMID: 37298645 PMCID: PMC10253699 DOI: 10.3390/ijms24119692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Biologics address a range of unmet clinical needs, but the occurrence of biologics-induced liver injury remains a major challenge. Development of cimaglermin alfa (GGF2) was terminated due to transient elevations in serum aminotransferases and total bilirubin. Tocilizumab has been reported to induce transient aminotransferase elevations, requiring frequent monitoring. To evaluate the clinical risk of biologics-induced liver injury, a novel quantitative systems toxicology modeling platform, BIOLOGXsym™, representing relevant liver biochemistry and the mechanistic effects of biologics on liver pathophysiology, was developed in conjunction with clinically relevant data from a human biomimetic liver microphysiology system. Phenotypic and mechanistic toxicity data and metabolomics analysis from the Liver Acinus Microphysiology System showed that tocilizumab and GGF2 increased high mobility group box 1, indicating hepatic injury and stress. Tocilizumab exposure was associated with increased oxidative stress and extracellular/tissue remodeling, and GGF2 decreased bile acid secretion. BIOLOGXsym simulations, leveraging the in vivo exposure predicted by physiologically-based pharmacokinetic modeling and mechanistic toxicity data from the Liver Acinus Microphysiology System, reproduced the clinically observed liver signals of tocilizumab and GGF2, demonstrating that mechanistic toxicity data from microphysiology systems can be successfully integrated into a quantitative systems toxicology model to identify liabilities of biologics-induced liver injury and provide mechanistic insights into observed liver safety signals.
Collapse
Affiliation(s)
- James J. Beaudoin
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lara Clemens
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Mark T. Miedel
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Albert Gough
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Fatima Zaidi
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Kari E. Wong
- Metabolon Inc., Durham, NC 27713, USA (P.R.); (K.E.W.); (R.S.)
| | | | - Christina Battista
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lisl K. M. Shoda
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Scott Q. Siler
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - D. Lansing Taylor
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Brett A. Howell
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| | - Lawrence A. Vernetti
- Department of Computational and Systems Biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA (A.G.); (D.L.T.)
| | - Kyunghee Yang
- DILIsym Services Division, Simulations Plus Inc., Research Triangle Park, Durham, NC 27709, USA (S.Q.S.)
| |
Collapse
|
12
|
Aquines O, Saavedra-Hernández A, Urbina-Arias N, Melchor-Martínez EM, Sosa-Hernández JE, Robledo-Padilla F, Iqbal HMN, Parra-Saldívar R. In Silico Modeling Study of Curcumin Diffusion and Cellular Growth. APPLIED SCIENCES 2022; 12:9749. [DOI: 10.3390/app12199749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Curcumin can enhance cutaneous wound healing by improving fibroblast proliferation. However, its therapeutic properties are dose-dependent: high concentrations produce cytotoxic effects, whereas low concentrations benefit cell proliferation. Similarly, the type of administration and its moderation are key aspects, as an erroneous distribution may result in null or noxious activity to the organism. In silico models for curcumin diffusion work as predictive tools for evaluating curcumin’s cytotoxic effects and establishing therapeutic windows. A 2D fibroblast culture growth model was created based on a model developed by Gérard and Goldbeter. Similarly, a curcumin diffusion model was developed by adjusting experimental release values obtained from Aguilar-Rabiela et al. and fitted to Korsmeyer–Peppas and Peleg’s hyperbolic models. The release of six key curcumin concentrations was achieved. Both models were integrated using Morpheus software, and a scratch-wound assay simulated curcumin’s dose-dependent effects on wound healing. The most beneficial effect was achieved at 0.25 μM, which exhibited the lowest cell-division period, the highest confluence (~60% for both release models, 447 initial cells), and the highest final cell population. The least beneficial effect was found at 20 μM, which inhibited cell division and achieved the lowest confluence (~34.30% for both release models, 447 initial cells). Confluence was shown to decrease as curcumin concentration increased, since higher concentrations of curcumin have inhibitory and cytotoxic effects.
Collapse
Affiliation(s)
- Osvaldo Aquines
- Department of Physics and Mathematics, Universidad de Monterrey, Av. Morones Prieto 4500, San Pedro Garza García 66238, N.L., Mexico
| | - Annel Saavedra-Hernández
- Department of Biomedical Engineering, Universidad de Monterrey, Av. Morones Prieto 4500, San Pedro Garza García 66238, N.L., Mexico
| | - Natalia Urbina-Arias
- Department of Biomedical Engineering, Universidad de Monterrey, Av. Morones Prieto 4500, San Pedro Garza García 66238, N.L., Mexico
| | - Elda M. Melchor-Martínez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Juan Eduardo Sosa-Hernández
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Felipe Robledo-Padilla
- Department of Physics and Mathematics, Universidad de Monterrey, Av. Morones Prieto 4500, San Pedro Garza García 66238, N.L., Mexico
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| | - Roberto Parra-Saldívar
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, N.L., Mexico
| |
Collapse
|
13
|
Madden JC, Enoch SJ, Paini A, Cronin MTD. A Review of In Silico Tools as Alternatives to Animal Testing: Principles, Resources and Applications. Altern Lab Anim 2020; 48:146-172. [PMID: 33119417 DOI: 10.1177/0261192920965977] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Across the spectrum of industrial sectors, including pharmaceuticals, chemicals, personal care products, food additives and their associated regulatory agencies, there is a need to develop robust and reliable methods to reduce or replace animal testing. It is generally recognised that no single alternative method will be able to provide a one-to-one replacement for assays based on more complex toxicological endpoints. Hence, information from a combination of techniques is required. A greater understanding of the time and concentration-dependent mechanisms, underlying the interactions between chemicals and biological systems, and the sequence of events that can lead to apical effects, will help to move forward the science of reducing and replacing animal experiments. In silico modelling, in vitro assays, high-throughput screening, organ-on-a-chip technology, omics and mathematical biology, can provide complementary information to develop a complete picture of the potential response of an organism to a chemical stressor. Adverse outcome pathways (AOPs) and systems biology frameworks enable relevant information from diverse sources to be logically integrated. While individual researchers do not need to be experts across all disciplines, it is useful to have a fundamental understanding of what other areas of science have to offer, and how knowledge can be integrated with other disciplines. The purpose of this review is to provide those who are unfamiliar with predictive in silico tools, with a fundamental understanding of the underlying theory. Current applications, software, barriers to acceptance, new developments and the use of integrated approaches are all discussed, with additional resources being signposted for each of the topics.
Collapse
Affiliation(s)
- Judith C Madden
- School of Pharmacy and Biomolecular Sciences, 4589Liverpool John Moores University, Liverpool, UK
| | - Steven J Enoch
- School of Pharmacy and Biomolecular Sciences, 4589Liverpool John Moores University, Liverpool, UK
| | - Alicia Paini
- 99013European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, 4589Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
14
|
Slavov S, Beger RD. Quantitative structure–toxicity relationships in translational toxicology. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Watkins PB. The DILI-sim Initiative: Insights into Hepatotoxicity Mechanisms and Biomarker Interpretation. Clin Transl Sci 2020; 12:122-129. [PMID: 30762301 PMCID: PMC6440570 DOI: 10.1111/cts.12629] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/16/2022] Open
Abstract
The drug‐induced liver injury (DILI)‐sim Initiative is a public‐private partnership involving scientists from industry, academia, and the US Food and Drug Administration (FDA). The Initiative uses quantitative systems toxicology (QST) to build and refine a model (DILIsym) capable of understanding and predicting liver safety liabilities in new drug candidates and to optimize interpretation of liver safety biomarkers used in clinical studies. Insights gained to date include the observation that most dose‐dependent hepatotoxicity can be accounted for by combinations of just three mechanisms (oxidative stress, interference with mitochondrial respiration, and alterations in bile acid homeostasis) and the importance of noncompetitive inhibition of bile acid transporters. The effort has also provided novel insight into species and interpatient differences in susceptibility, structure‐activity relationships, and the role of nonimmune mechanisms in delayed idiosyncratic hepatotoxicity. The model is increasingly used to evaluate new drug candidates and several clinical trials are underway that will test the model's ability to prospectively predict liver safety. With more refinement, in the future, it may be possible to use the DILIsym predictions to justify reduction in the size of some clinical trials. The mature model could also potentially assist physicians in managing the liver safety of their patients as well as aid in the diagnosis of DILI.
Collapse
Affiliation(s)
- Paul B Watkins
- Institute for Drug Safety Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
16
|
Wu Q, Taboureau O, Audouze K. Development of an adverse drug event network to predict drug toxicity. Curr Res Toxicol 2020; 1:48-55. [PMID: 34345836 PMCID: PMC8320634 DOI: 10.1016/j.crtox.2020.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 11/28/2022] Open
Abstract
Despite of their therapeutic effects, drug's exposure may have negative effects on human health such as adverse drug reaction (ADR) and side effects (SE). Adverse drug events (ADEs), that correspond to an event occurring during the drug treatment (i.e. ADR and SE), is not necessarily caused by the drug itself, as this is the case with medical errors and social factors. Due to the complexity of the biological systems, not all ADEs are known for marketed drugs. Therefore, new and effective methods are needed to determine potential risks, including the development of computational strategies. We present an ADE association network based on 90,827 drug-ADE associations between 930 unique drug and 6221 unique ADE, on which we implemented a scoring system based on a pull-down approach for prediction of drug-ADE combination. Based on our network, ADEs proposed for three drugs, safinamide, sonidegib, rufinamide are further discussed. The model was able to identify, already known drug-ADE associations that are supported by the literature and FDA reports, and also to predict uncharacterized associations such as dopamine dysregulation syndrome, or nicotinic acid deficiency for the drugs safinamide and sonidegib respectively, illustrating the power of such integrative toxicological approach.
Collapse
Key Words
- ADE, adverse drug event
- ADR, adverse drug reaction
- AOP, adverse outcome pathway
- Adverse event network
- Computational toxicology
- FAERS, FDA Adverse Event Reporting System
- FDA, Food and Drug Administration
- HMS-PCI, high-throughput mass spectrometric protein complex identification
- LRT, Likelihood Ratio Test
- MedDRA, Medical Dictionary for Regulatory Activities
- Network science
- PPAN, protein-protein association network
- PT, Preferred Term
- Predictive toxicity
- QSAR, Quantitative structure-activity relationships
- SE, side effect
- SOC, System Organ Class
- System toxicology
- TAP–MS, tandem-affinity-purification method coupled to mass spectrometry
- pullS, pull-down score
- wS, weighted score
Collapse
Affiliation(s)
- Qier Wu
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Olivier Taboureau
- Université de Paris, BFA, CNRS UMR 8251, ERL Inserm U1133, CNRS UMR 8251, F-75013 Paris, France
| | - Karine Audouze
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| |
Collapse
|
17
|
Longo DM, Shoda LKM, Howell BA, Coric V, Berman RM, Qureshi IA. Assessing Effects of BHV-0223 40 mg Zydis Sublingual Formulation and Riluzole 50 mg Oral Tablet on Liver Function Test Parameters Utilizing DILIsym. Toxicol Sci 2020; 175:292-300. [PMID: 32040174 PMCID: PMC7253195 DOI: 10.1093/toxsci/kfaa019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
For patients with amyotrophic lateral sclerosis who take oral riluzole tablets, approximately 50% experience alanine transaminase (ALT) levels above upper limit of normal (ULN), 8% above 3× ULN, and 2% above 5× ULN. BHV-0223 is a novel 40 mg rapidly sublingually disintegrating (Zydis) formulation of riluzole, bioequivalent to conventional riluzole 50 mg oral tablets, that averts the need for swallowing tablets and mitigates first-pass hepatic metabolism, thereby potentially reducing risk of liver toxicity. DILIsym is a validated multiscale computational model that supports evaluation of liver toxicity risks. DILIsym was used to compare the hepatotoxicity potential of oral riluzole tablets (50 mg BID) versus BHV-0223 (40 mg BID) by integrating clinical data and in vitro toxicity data. In a simulated population (SimPops), ALT levels > 3× ULN were predicted in 3.9% (11/285) versus 1.4% (4/285) of individuals with oral riluzole tablets and sublingual BHV-0223, respectively. This represents a relative risk reduction of 64% associated with BHV-0223 versus conventional riluzole tablets. Mechanistic investigations revealed that oxidative stress was responsible for the predicted ALT elevations. The validity of the DILIsym representation of riluzole and assumptions is supported by its ability to predict rates of ALT elevations for riluzole oral tablets comparable with that observed in clinical data. Combining a mechanistic, quantitative representation of hepatotoxicity with interindividual variability in both susceptibility and liver exposure suggests that sublingual BHV-0223 confers diminished rates of liver toxicity compared with oral tablets of riluzole, consistent with having a lower overall dose of riluzole and bypassing first-pass liver metabolism.
Collapse
Affiliation(s)
- Diane M Longo
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
- To whom correspondence should be addressed at 6 Davis Drive, PO Box 12317, Research Triangle Park, NC 27709. E-mail:
| | - Lisl K M Shoda
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| | - Brett A Howell
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| | - Vladimir Coric
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut 06510
| | - Robert M Berman
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut 06510
| | - Irfan A Qureshi
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut 06510
| |
Collapse
|
18
|
Watkins PB. Quantitative Systems Toxicology Approaches to Understand and Predict Drug-Induced Liver Injury. Clin Liver Dis 2020; 24:49-60. [PMID: 31753250 DOI: 10.1016/j.cld.2019.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The DILI-sim Initiative is a public-private partnership using quantitative systems toxicology to build a model (DILIsym) capable of understanding and predicting liver safety liabilities in drug candidates. The effort has provided insights into mechanisms underlying dose-dependent drug-induced liver injury (DILI) and interpatient differences in susceptibility to dose-dependent DILI. DILIsym may be useful in identifying drugs capable of causing idiosyncratic hepatotoxicity. DILIsym is used to optimize interpretation of traditional and newer serum biomarkers of DILI. DILIsym results are considered in drug development decisions. In the future, it may be possible to use DILsym predictions to justify reduction in size of some clinical trials.
Collapse
Affiliation(s)
- Paul B Watkins
- Institute for Drug Safety Sciences, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, 6 Davis Drive, PO Box 12137, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
19
|
Kenna JG, Taskar KS, Battista C, Bourdet DL, Brouwer KLR, Brouwer KR, Dai D, Funk C, Hafey MJ, Lai Y, Maher J, Pak YA, Pedersen JM, Polli JW, Rodrigues AD, Watkins PB, Yang K, Yucha RW. Can Bile Salt Export Pump Inhibition Testing in Drug Discovery and Development Reduce Liver Injury Risk? An International Transporter Consortium Perspective. Clin Pharmacol Ther 2019; 104:916-932. [PMID: 30137645 PMCID: PMC6220754 DOI: 10.1002/cpt.1222] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022]
Abstract
Bile salt export pump (BSEP) inhibition has emerged as an important mechanism that may contribute to the initiation of human drug‐induced liver injury (DILI). Proactive evaluation and understanding of BSEP inhibition is recommended in drug discovery and development to aid internal decision making on DILI risk. BSEP inhibition can be quantified using in vitro assays. When interpreting assay data, it is important to consider in vivo drug exposure. Currently, this can be undertaken most effectively by consideration of total plasma steady state drug concentrations (Css,plasma). However, because total drug concentrations are not predictive of pharmacological effect, the relationship between total exposure and BSEP inhibition is not causal. Various follow‐up studies can aid interpretation of in vitro BSEP inhibition data and may be undertaken on a case‐by‐case basis. BSEP inhibition is one of several mechanisms by which drugs may cause DILI, therefore, it should be considered alongside other mechanisms when evaluating possible DILI risk.
Collapse
Affiliation(s)
| | - Kunal S Taskar
- Mechanistic Safety and Disposition, IVIVT, GlaxoSmithKline, Ware, Hertfordshire, UK
| | - Christina Battista
- DILIsym Services Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA
| | - David L Bourdet
- Drug Metabolism and Pharmacokinetics, Theravance Biopharma, South San Francisco, California, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - David Dai
- Clinical Pharmacology, Research and Development Sciences, Agios Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Christoph Funk
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Michael J Hafey
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| | - Jonathan Maher
- Safety Assessment, Genentech, South San Francisco, California, USA
| | - Y Anne Pak
- Lilly Research Laboratory, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jenny M Pedersen
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Novum, Huddinge, Sweden
| | - Joseph W Polli
- Mechanistic Safety and Drug Disposition, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | | | - Paul B Watkins
- Institute for Drug Safety Sciences, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kyunghee Yang
- DILIsym Services Inc., a Simulations Plus Company, Research Triangle Park, North Carolina, USA
| | - Robert W Yucha
- Takeda Pharmaceuticals, Global Drug Metabolism and Pharmacokinetics, Cambridge, Massachusetts, USA
| | | |
Collapse
|
20
|
Polak S, Tylutki Z, Holbrook M, Wiśniowska B. Better prediction of the local concentration-effect relationship: the role of physiologically based pharmacokinetics and quantitative systems pharmacology and toxicology in the evolution of model-informed drug discovery and development. Drug Discov Today 2019; 24:1344-1354. [PMID: 31132414 DOI: 10.1016/j.drudis.2019.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/04/2019] [Accepted: 05/21/2019] [Indexed: 12/15/2022]
Abstract
Model-informed drug discovery and development (MID3) is an umbrella term under which sit several computational approaches: quantitative systems pharmacology (QSP), quantitative systems toxicology (QST) and physiologically based pharmacokinetics (PBPK). QSP models are built using mechanistic knowledge of the pharmacological pathway focusing on the putative mechanism of drug efficacy; whereas QST models focus on safety and toxicity issues and the molecular pathways and networks that drive these adverse effects. These can be mediated through exaggerated on-target or off-target pharmacology, immunogenicity or the physiochemical nature of the compound. PBPK models provide a mechanistic description of individual organs and tissues to allow the prediction of the intra- and extra-cellular concentration of the parent drug and metabolites under different conditions. Information on biophase concentration enables the prediction of a drug effect in different organs and assessment of the potential for drug-drug interactions. Together, these modelling approaches can inform the exposure-response relationship and hence support hypothesis generation and testing, compound selection, hazard identification and risk assessment through to clinical proof of concept (POC) and beyond to the market.
Collapse
Affiliation(s)
- Sebastian Polak
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland; Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.
| | - Zofia Tylutki
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland; Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Mark Holbrook
- Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Barbara Wiśniowska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland
| |
Collapse
|
21
|
Matsuzaka Y, Uesawa Y. Optimization of a Deep-Learning Method Based on the Classification of Images Generated by Parameterized Deep Snap a Novel Molecular-Image-Input Technique for Quantitative Structure-Activity Relationship (QSAR) Analysis. Front Bioeng Biotechnol 2019; 7:65. [PMID: 30984753 PMCID: PMC6447703 DOI: 10.3389/fbioe.2019.00065] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/07/2019] [Indexed: 12/22/2022] Open
Abstract
Numerous chemical compounds are distributed around the world and may affect the homeostasis of the endocrine system by disrupting the normal functions of hormone receptors. Although the risks associated with these compounds have been evaluated by acute toxicity testing in mammalian models, the chronic toxicity of many chemicals remains due to high cost of the compounds and the testing, etc. However, computational approaches may be promising alternatives and reduce these evaluations. Recently, deep learning (DL) has been shown to be promising prediction models with high accuracy for recognition of images, speech, signals, and videos since it greatly benefits from large datasets. Recently, a novel DL-based technique called DeepSnap was developed to conduct QSAR analysis using three-dimensional images of chemical structures. It can be used to predict the potential toxicity of many different chemicals to various receptors without extraction of descriptors. DeepSnap has been shown to have a very high capacity in tests using Tox21 quantitative qHTP datasets. Numerous parameters must be adjusted to use the DeepSnap method but they have not been optimized. In this study, the effects of these parameters on the performance of the DL prediction model were evaluated in terms of the loss in validation as an indicator for evaluating the performance of the DL using the toxicity information in the Tox21 qHTP database. The relations of the parameters of DeepSnap such as (1) number of molecules per SDF split into (2) zoom factor percentage, (3) atom size for van der waals percentage, (4) bond radius, (5) minimum bond distance, and (6) bond tolerance, with the validation loss following quadratic function curves, which suggests that optimal thresholds exist to attain the best performance with these prediction models. Using the parameter values set with the best performance, the prediction model of chemical compounds for CAR agonist was built using 64 images, at 105° angle, with AUC of 0.791. Thus, based on these parameters, the proposed DeepSnap-DL approach will be highly reliable and beneficial to establish models to assess the risk associated with various chemicals.
Collapse
Affiliation(s)
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
22
|
Analyzing the Mechanisms Behind Macrolide Antibiotic-Induced Liver Injury Using Quantitative Systems Toxicology Modeling. Pharm Res 2019; 36:48. [PMID: 30734107 PMCID: PMC6373306 DOI: 10.1007/s11095-019-2582-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/27/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Macrolide antibiotics are commonly prescribed treatments for drug-resistant bacterial infections; however, many macrolides have been shown to cause liver enzyme elevations and one macrolide, telithromycin, has been pulled from the market by its provider due to liver toxicity. This work seeks to assess the mechanisms responsible for the toxicity of macrolide antibiotics. METHODS Five macrolides were assessed in in vitro systems designed to test for bile acid transporter inhibition, mitochondrial dysfunction, and oxidative stress. The macrolides were then represented in DILIsym, a quantitative systems pharmacology (QST) model of drug-induced liver injury, placing the in vitro results in context with each compound's predicted liver exposure and known biochemistry. RESULTS DILIsym results suggest that solithromycin and clarithromycin toxicity is primarily due to inhibition of the mitochondrial electron transport chain (ETC) while erythromycin toxicity is primarily due to bile acid transporter inhibition. Telithromycin and azithromycin toxicity was not predicted by DILIsym and may be caused by mechanisms not currently incorporated into DILIsym or by unknown metabolite effects. CONCLUSIONS The mechanisms responsible for toxicity can be significantly different within a class of drugs, despite the structural similarity among the drugs. QST modeling can provide valuable insight into the nature of these mechanistic differences.
Collapse
|
23
|
|
24
|
Machine Learning Models for the Prediction of Chemotherapy-Induced Peripheral Neuropathy. Pharm Res 2019; 36:35. [DOI: 10.1007/s11095-018-2562-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023]
|
25
|
Piñero J, Furlong LI, Sanz F. In silico models in drug development: where we are. Curr Opin Pharmacol 2018; 42:111-121. [PMID: 30205360 DOI: 10.1016/j.coph.2018.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
The use and utility of computational models in drug development has significantly grown in the last decades, fostered by the availability of high throughput datasets and new data analysis strategies. These in silico approaches are demonstrating their ability to generate reliable predictions as well as new knowledge on the mode of action of drugs and the mechanisms underlying their side effects, altogether helping to reduce the costs of drug development. The aim of this review is to provide a panorama of developments in the field in the last two years.
Collapse
Affiliation(s)
- Janet Piñero
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Laura I Furlong
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
26
|
Bloomingdale P, Nguyen VA, Niu J, Mager DE. Boolean network modeling in systems pharmacology. J Pharmacokinet Pharmacodyn 2018; 45:159-180. [PMID: 29307099 PMCID: PMC6531050 DOI: 10.1007/s10928-017-9567-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/29/2017] [Indexed: 01/01/2023]
Abstract
Quantitative systems pharmacology (QSP) is an emerging discipline that aims to discover how drugs modulate the dynamics of biological components in molecular and cellular networks and the impact of those perturbations on human pathophysiology. The integration of systems-based experimental and computational approaches is required to facilitate the advancement of this field. QSP models typically consist of a series of ordinary differential equations (ODE). However, this mathematical framework requires extensive knowledge of parameters pertaining to biological processes, which is often unavailable. An alternative framework that does not require knowledge of system-specific parameters, such as Boolean network modeling, could serve as an initial foundation prior to the development of an ODE-based model. Boolean network models have been shown to efficiently describe, in a qualitative manner, the complex behavior of signal transduction and gene/protein regulatory processes. In addition to providing a starting point prior to quantitative modeling, Boolean network models can also be utilized to discover novel therapeutic targets and combinatorial treatment strategies. Identifying drug targets using a network-based approach could supplement current drug discovery methodologies and help to fill the innovation gap across the pharmaceutical industry. In this review, we discuss the process of developing Boolean network models and the various analyses that can be performed to identify novel drug targets and combinatorial approaches. An example for each of these analyses is provided using a previously developed Boolean network of signaling pathways in multiple myeloma. Selected examples of Boolean network models of human (patho-)physiological systems are also reviewed in brief.
Collapse
Affiliation(s)
- Peter Bloomingdale
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 431 Kapoor Hall, Buffalo, NY, 14214, USA
| | - Van Anh Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 431 Kapoor Hall, Buffalo, NY, 14214, USA
| | - Jin Niu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 431 Kapoor Hall, Buffalo, NY, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 431 Kapoor Hall, Buffalo, NY, 14214, USA.
| |
Collapse
|