1
|
Hewitt P, Hartmann A, Tornesi B, Ferry-Martin S, Valentin JP, Desert P, Gresham S, Demarta-Gatsi C, Venishetty VK, Kolly C. Importance of tailored non-clinical safety testing of novel antimalarial drugs: Industry best-practice. Regul Toxicol Pharmacol 2024; 154:105736. [PMID: 39515409 DOI: 10.1016/j.yrtph.2024.105736] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Malaria is an acute, debilitating parasitic illness. There were 249 million cases of malaria in 2022, resulting in 608,000 deaths globally, 76% of which were children ≤5 years. The unique nature of this disease (recurrences leading to re-treatments and numerous organ systems affected), specific clinical treatment regimens, poor compliance, and diversity of affected populations (predominantly pediatrics, women of childbearing potential, pregnant and lactating women), often makes standard testing approaches inadequate, and tailor-made safety assessments are more appropriate. We provide best practice recommendations based on company experience for the non-clinical safety assessment of antimalarial drugs, with a focus on small molecules since they represent the majority of drug candidates for this illness. We focus on specific testing considerations for repeat dose toxicity studies, including combination toxicity assessments, since new drug treatment regimens typically foresee short treatment durations to improve compliance (i.e., 1 day) with combinations of compounds to improve efficacy and limit potential resistance. Due to the target population, the timing of reproductive, developmental, and juvenile toxicity studies may be earlier than general testing roadmaps for other small molecule drugs. In conclusion, key recommendations presented should enable a more effective and efficient development path whilst protecting clinical trial participants and patients.
Collapse
Affiliation(s)
- Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany.
| | | | - Belen Tornesi
- Non-Clinical Pharmacology & Toxicology, Medicines for Malaria Venture, Geneva, Switzerland
| | - Sandrine Ferry-Martin
- Nonclinical Drug Safety, Merck Research Laboratories, Merck Sharp & Dohme, Clermont-Ferrand, France
| | - Jean-Pierre Valentin
- Early Clinical Development & Translational Science, Non-Clinical Safety Evaluation, UCB Pharma, Braine L'Alleud, Belgium
| | - Paul Desert
- Nonclinical Safety, Sanofi, Marcy l'Etoile, France
| | | | - Claudia Demarta-Gatsi
- Global Health R&D of Merck Healthcare, Ares Trading S.A., (a subsidiary of Merck KGaA, Darmstadt, Germany), Switzerland
| | | | - Carine Kolly
- Preclinical Safety, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
2
|
Lee L, Flach S, Xue H, Arivelu L, Golden L, Kong R, Darpo B. Lack of Concentration-QTc Relationship and Cardiac Risk With Vatiquinone Therapeutic and Supratherapeutic Doses. Clin Pharmacol Drug Dev 2024; 13:1227-1238. [PMID: 39415654 DOI: 10.1002/cpdd.1476] [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: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Vatiquinone, a 15-lipoxygenase inhibitor, is in development for patients with Friedreich ataxia. The study determined the effect of vatiquinone on electrocardiogram parameters. This was a 2-part, single-center, randomized, double-blinded, and placebo-controlled study. Part 1 used an adaptive approach to identify a supratherapeutic dose, while Part 2 evaluated the effect of vatiquinone on Fridericia corrected QT interval (QTcF). A safe and tolerated supratherapeutic dose of 1400 mg was identified. Concentration-QTcF analysis confirmed there was no statistically significant relationship between vatiquinone concentration and QTcF. QTcF effect (ie, ΔΔQTcF) exceeding 10 milliseconds was excluded for concentrations up to approximately 11,500 ng/mL. By-time-point analysis confirmed that least-squares mean ΔΔQTcF was below 10 milliseconds. Largest least-squares mean ΔΔQTcF of 1.5 milliseconds was observed at 2 hours after dosing. Vatiquinone did not have a clinically relevant effect on heart rate nor on cardiac conduction (PR interval and QRS interval). No new safety signals were found, as safety data are consistent with the known safety profile of vatiquinone. These findings altogether demonstrated that there is a minimal cardiac risk for vatiquinone concentrations up to the supratherapeutic dose level.
Collapse
Affiliation(s)
- Lucy Lee
- PTC Therapeutics, Inc., Warren, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
3
|
Harmer AR, Rolf MG. On the relationship between hERG inhibition and the magnitude of QTc prolongation: An in vitro to clinical translational analysis. Toxicol Appl Pharmacol 2024; 492:117135. [PMID: 39490450 DOI: 10.1016/j.taap.2024.117135] [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: 09/16/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Assessing the magnitude of QTc prolongation is crucial in drug development due to its association with Torsades de Pointes. Inhibition of the hERG channel, pivotal in cardiac repolarization, is a key factor in evaluating this risk. In this study, the relationship between hERG inhibition and QTc prolongation magnitude was investigated, with the aim to derive simple guidance on the required hERG margin to avoid a large (>20 ms) QTc prolongation. METHODS Data from literature and FDA sources were searched for compounds with hERG IC50 values alongside clinical QTc data with paired plasma concentrations, or compounds demonstrating a clinical concentration-QTc relationship. Relationships between hERG inhibition, hERG IC50 margin to unbound plasma Cmax, and QTc prolongation magnitude were calculated. RESULTS Analysis of 148 clinical QTc observations from 98 compounds revealed that compounds associated with QTc prolongation >10 ms typically exhibited hERG margins of ≤33-fold, while those exceeding 20 ms were generally associated with margins of ≤24-fold. QTc increases above 10 ms were not observed at hERG margins >100-fold. Based on 53 clinical concentration-QTc datasets, modest hERG inhibition levels of ∼4-6 % correlated with a 10 ms QTc prolongation, while ∼10-13 % inhibition corresponded to a 20 ms prolongation. CONCLUSIONS This study enhances understanding of the relationship between hERG inhibition and QTc prolongation magnitude, by conducting analysis across a wide range of 98 compounds. This information can be used to determine the optimal hERG margin, particularly for drug discovery projects with limited scope to completely design-out hERG activity.
Collapse
Affiliation(s)
- Alexander R Harmer
- The Discovery Centre, Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, UK.
| | - Michael G Rolf
- Gothenburg, Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Sweden
| |
Collapse
|
4
|
Best DD, Abernathy MM, Leishman DJ. A "one-step" approach to heart rate correction and statistical analysis applied to conscious dog QTc studies. Clin Transl Sci 2024; 17:e70046. [PMID: 39404505 PMCID: PMC11478927 DOI: 10.1111/cts.70046] [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] [Received: 07/11/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
A "one-step" method which combined the heart rate correction and statistical analysis for conscious nonhuman primate (NHP) QTc assessment was recently published. The principles of this method are applicable to other species. In the current analysis, we demonstrate the utility of the technique in conscious dog QTc studies. Two studies in male dogs (n = 8 and n = 7) implanted with telemetry devices were used. In both studies, treatments were randomized and all animals received all treatments. In the primary study, the effect on QTc of moxifloxacin was compared with vehicle. Each treatment (vehicle and moxifloxacin) was given on two separate occasions. In the second study, dogs were given vehicle or dofetilide. Conventional QTc analysis was compared with the "one-step" method. The effect on QTc relative to vehicle was determined along with the median minimal detectable difference. As expected, both moxifloxacin and dofetilide gave QTc increases with a maximum of ~ 20 ms. There was a significant increase in the sensitivity to detect a QTc effect when using the "one-step" method. The minimal detectable difference was 1.6 ms for the "one-step" method compared with 6.2 ms for the conventional method. These analyses are consistent with the increased sensitivity described for the "one-step" method applied to studies in NHP. The increased sensitivity should enhance the ability to support an integrated assessment of the QTc prolongation liability for new drugs.
Collapse
|
5
|
Boulay E, Authier S, Bartko T, Greiter-Wilke A, Leishman D, Li D, Nichols JV, Pierson J, Rossman EI, Valentin JP, Vicente J, Walisser J, Troncy E, Wisialowski TA. Assessment of corrected JT-peak (JTpc) and Tpeak-to-Tend (TpTec) as proarrhythmia biomarkers in non-human primates: Outcome from a HESI consortium. J Pharmacol Toxicol Methods 2024; 129:107543. [PMID: 39019200 DOI: 10.1016/j.vascn.2024.107543] [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: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
INTRODUCTION Corrected QT interval (QTc)is an established biomarker for drug-induced Torsade de Pointe (TdP), but with concerns for a false positive signal. Clinically, JTpc and TpTec have emerged as ECG sub-intervals to differentiate predominant hERG vs. mixed ion channel blocking drugs that prolong QTc. METHODS In a multicentric, prospective, controlled study, different proarrhythmic drug effects on QTc, JTpc and TpTec were characterized with cynomolgus monkeys using telemetry in a Lead II configuration for internal and external telemetry.Drugs and vehicle were administered orally (PO) to group size of 4 to 8 animals, in 4 laboratories. RESULTS In monkeys, dofetilide (0.03-0.3 mg/kg) was associated with exposure dependent QTc and JTpc increase, but no significant TpTec effect. Similarly, quinidine (2-50 mg/kg) increased QTc and JTpc but did not change TpTec. Mexiletine (1-15 mg/kg) and verapamil (50 mg/kg) did not induce any significant effect on QTc, JTpc or TpTec. DISCUSSION Clinically, predominant hERG blockers (dofetilide and quinidine) prolong QTc, JTpc and TpTec and are associated with increased risk for TdP. Results from this study demonstrate that ECG changes after dofetilide and quinidine administration to telemetered monkeys differ from the clinical response, lacking the expected effects on TpTec. Potential explanations for the lack of translation include physio-pharmacology species differences or ECG recording and analysis methodology variations. Mixed ion channel blockers verapamil and mexiletine administered to monkeys showed no significant QTc, JTpc or TpTec prolongation as expected based on the similar clinical response for these agents.
Collapse
Affiliation(s)
- Emmanuel Boulay
- Charles River Laboratories, Laval, Quebec, Canada; GREPAQ (Groupe de recherche en pharmacologie animale du Québec), Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Simon Authier
- Charles River Laboratories, Laval, Quebec, Canada; GREPAQ (Groupe de recherche en pharmacologie animale du Québec), Université de Montréal, St-Hyacinthe, Québec, Canada.
| | - Theresa Bartko
- Labcorp Early Development Laboratories Inc, Madison, WI, USA
| | | | | | | | - Jill V Nichols
- Labcorp Early Development Laboratories Inc, Madison, WI, USA
| | - Jennifer Pierson
- Health and Environmental Sciences Institute (HESI), Washington, DC, USA
| | | | | | - Jose Vicente
- Center for Drug Evaluation and Research, US Food & Drug Administration (FDA), Silver Spring, MD, USA
| | | | - Eric Troncy
- GREPAQ (Groupe de recherche en pharmacologie animale du Québec), Université de Montréal, St-Hyacinthe, Québec, Canada
| | | |
Collapse
|
6
|
Harrell AW, Reid K, Vahle J, Brouta F, Beilmann M, Young G, Beattie KA, Valentin JP, Shaid S, Brinck P. Endeavours made by trade associations, pharmaceutical companies and regulators in the replacement, reduction and refinement of animal experimentation in safety testing of pharmaceuticals. Regul Toxicol Pharmacol 2024; 152:105683. [PMID: 39117168 DOI: 10.1016/j.yrtph.2024.105683] [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: 04/11/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Following the European Commission decision to develop a roadmap to phase out animal testing and the signing of the US Modernisation Act, there is additional pressure on regulators and the pharmaceutical industry to abandon animal experimentation in safety testing. Often, endeavours already made by governments, regulators, trade associations, and industry to replace, reduce and refine animal experimentation (3Rs) are unnoticed. Herein, we review such endeavours to promote wider application and acceptance of 3Rs. ICH guidelines have stated 3Rs objectives and have enjoyed many successes driven by global consensus. Initiatives driven by US and European regulators such as the removal of the Abnormal Toxicity Test are neutralised by reticent regional regulators. Stream-lined testing requirements have been proposed for new modalities, oncology, impurity management and animal pharmacokinetics/metabolism. Use of virtual controls, value of the second toxicity species, information sharing and expectations for life-threatening diseases, human specific or well-characterised targets are currently being scrutinised. Despite much effort, progress falls short of the ambitious intent of decisionmakers. From a clinical safety and litigation perspective pharmaceutical companies and regulators are reluctant to step away from current paradigms unless replacement approaches are validated and globally accepted. Such consensus has historically been best achieved through ICH initiatives.
Collapse
Affiliation(s)
| | - Kirsty Reid
- European Federation of Pharmaceutical Industries and Associates, Leopold Plaza Building, Rue Du Trone 108, B-1050. Brussels, Belgium
| | - John Vahle
- Lilly Corporate Center, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Frederic Brouta
- UCB Biopharma SRL, Chemin Du Foriest, B-1420, Braine-l'Alleud, Belgium
| | - Mario Beilmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach, Germany
| | - Graeme Young
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK. UK
| | - Kylie A Beattie
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK. UK
| | | | - Shajahan Shaid
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK. UK
| | | |
Collapse
|
7
|
Lester RM, Engel C, van Haarst AD, Paglialunga S. Should You Run a Dedicated TQT Study? Sponsor and Regulatory Considerations on Substitution Pathways to Assess QT Liability. Clin Pharmacol Ther 2024; 116:42-51. [PMID: 38698592 DOI: 10.1002/cpt.3284] [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: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
Cardiac safety regulatory guidance for drug development has undergone several monumental shifts over the past decade as technological advancements, analysis models and study best practices have transformed this landscape. Once, clinical proarrhythmic risk assessment of a new chemical entity (NCE) was nearly exclusively evaluated in a dedicated thorough QT (TQT) study. However, since the introduction of the International Council for Harmonisation (ICH) E14/S7B Q&A 5.1 and 6.1 TQT substitutions, drug developers are offered an alternative pathway to evaluate proarrhythmic risk during an ascending dose study in healthy volunteers or during a powered patient study, respectively. In addition, the findings as well as the manner in which nonclinical studies are conducted (i.e., utilizing best practices) can dictate the need for a positive control in the clinical study and/or affect the labeling outcome. Drug sponsors are now faced with the option of pursuing a dedicated TQT study or requesting a TQT substitution. Potential factors influencing the choice of pathway include the NCE mechanism of action, pharmacokinetic properties, and safety profile, as well as business considerations. This tutorial will highlight the regulatory framework for integrated arrhythmia risk prediction models to outline drug safety, delineate potential reasons why a TQT substitution request may be rejected and discuss when a standalone TQT is recommended.
Collapse
|
8
|
Qu Y, Henderson KA, Harper TA, Vargas HM. Scientific Review of the Proarrhythmic Risks of Oligonucleotide Therapeutics: Are Dedicated ICH S7B/E14 Studies Needed for Low-Risk Modalities? Clin Pharmacol Ther 2024; 116:96-105. [PMID: 38362953 DOI: 10.1002/cpt.3204] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
Oligonucleotide therapeutics (ONTs) represent a new modality with unique pharmacological and chemical properties that modulate gene expression with a high degree of target specificity mediated by complementary Watson-Crick base pair hybridization. To date, the proarrhythmic assessment of ONTs has been influenced by International Conference on Harmonization (ICH) E14 and S7B guidance. To document current hERG/QTc evaluation practices, we reviewed US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) Approval Packages (source: PharmaPendium.com) and collated preclinical and clinical studies for 17 marketed ONTs. In addition, clinical QTc data from 12 investigational ONTs were obtained from the literature. Of the marketed ONTs, eight were tested in the hERG assay with no inhibitory effect identified at the top concentration (range: 34-3,000 μM) tested. Fourteen of the ONTs were evaluated in nonhuman primate cardiovascular studies with 11 of them in dedicated telemetry studies. No effect on QTc intervals were observed (at high exposure multiples) in all studies. Clinically, four ONTs were evaluated in TQT studies; an additional six ONTs were assessed by concentration-QTc interval analysis, and six by routine safety electrocardiogram monitoring. None of the clinical studies identified a QTc prolongation risk; the same was true for the 12 investigational ONTs. A search of the FDA Adverse Event Database indicated no association between approved ONTs and proarrhythmias. Overall, the collective weight of evidence from 29 ONTs demonstrate no clinical proarrhythmic risk based on data obtained from ICH S7B/E14 studies. Thus, new ONTs may benefit from reduced testing strategies because they have no proarrhythmic risk, a similar cardiac safety profile as monoclonal antibodies, proteins, and peptides.
Collapse
Affiliation(s)
- Yusheng Qu
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc., Thousand Oaks, California, USA
| | - Kim A Henderson
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc., Thousand Oaks, California, USA
| | - Tod A Harper
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc., Thousand Oaks, California, USA
| | - Hugo M Vargas
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc., Thousand Oaks, California, USA
| |
Collapse
|
9
|
Wisialowski TA, Ether N, Foley CM, Kleiman R, Koshman Y, Leishman D, Martel E, Nichols JV, Popp J, Rajamani S, Riley S, Rossman EI, Vargas HM. Improving the in vivo QTc assay: Nonclinical concentration-QTc modeling for risk assessment. J Pharmacol Toxicol Methods 2024; 128:107515. [PMID: 38777240 DOI: 10.1016/j.vascn.2024.107515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Affiliation(s)
| | | | | | | | | | | | - Eric Martel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | | | | | | | | | | |
Collapse
|
10
|
Bétat AM, Delaunois A, Delpy E, Loiseau M, Maurin A, Poizat G, Possémé C, Weinelt F, Drieu la Rochelle C, Martel E, Valentin JP. Results from a Joined Prospective Study to Evaluate the Sensitivity of the In Vivo Dog QT Assay in Line with the ICH E14/S7B Q&A Best Practices. Clin Pharmacol Ther 2024; 116:106-116. [PMID: 38709223 DOI: 10.1002/cpt.3283] [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: 01/08/2024] [Accepted: 04/12/2024] [Indexed: 05/07/2024]
Abstract
The ICH E14/S7B Q&As highlighted the need for best practices concerning the design, execution, analysis, interpretation, and reporting of the in vivo non-rodent QT assay as a component of the integrated risk assessment to potentially support a TQT waiver or substitute. We conducted a dog telemetry study to assess the effects on QTc of six reference compounds (five positive and one negative) previously evaluated by Darpo et al. (2015) in humans. The sensitivity of the assay to detect QTc increases was determined, and exposure-response analysis was performed, as done in clinical practice. By-timepoint analysis showed QTc prolongation induced by moxifloxacin, dofetilide, dolasetron, ondansetron, and quinine within human relevant plasma exposures ranges. Moreover, a hysteresis was observed for quinine. As expected, levocetirizine showed no statistically significant effect on QTc across a range of exposure, well exceeding the therapeutic Cmax. Power analyses confirmed the study ability to detect statistically significant QTc changes of less than 10 milliseconds with 80% probability, even with a sample size as low as n = 4 animals. Finally, concentration-QTc modeling enabled to predict the minimal plasma concentration needed to detect a 10 milliseconds QTc prolongation, including for quinine. The comparison with clinical available data supported the relevance of dogs under these experimental conditions as a robust translational predictor of drug-induced QTc prolongation in humans as a key pillar of the integrated risk assessment.
Collapse
Affiliation(s)
| | - Annie Delaunois
- Early Clinical Development & Translational Science Department, UCB Biopharma SRL, Braine-l'Alleud, Belgium
| | - Eric Delpy
- Non-Clinical Department, Biotrial Pharmacology, Rennes, France
| | | | - Anne Maurin
- Non-Clinical Department, Biotrial Pharmacology, Rennes, France
| | | | - Celine Possémé
- Non-Clinical Department, Biotrial Pharmacology, Rennes, France
| | - Ferdinand Weinelt
- Department of Drug Discovery Sciences-PKPD Modeling, Boehringer Ingelheim Pharma, Biberach/Riss, Germany
| | | | - Eric Martel
- Department of Drug Discovery Sciences-General Pharmacology, Boehringer Ingelheim Pharma, Biberach/Riss, Germany
| | - Jean-Pierre Valentin
- Early Clinical Development & Translational Science Department, UCB Biopharma SRL, Braine-l'Alleud, Belgium
| |
Collapse
|
11
|
Brennan RJ, Jenkinson S, Brown A, Delaunois A, Dumotier B, Pannirselvam M, Rao M, Ribeiro LR, Schmidt F, Sibony A, Timsit Y, Sales VT, Armstrong D, Lagrutta A, Mittlestadt SW, Naven R, Peri R, Roberts S, Vergis JM, Valentin JP. The state of the art in secondary pharmacology and its impact on the safety of new medicines. Nat Rev Drug Discov 2024; 23:525-545. [PMID: 38773351 DOI: 10.1038/s41573-024-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2024] [Indexed: 05/23/2024]
Abstract
Secondary pharmacology screening of investigational small-molecule drugs for potentially adverse off-target activities has become standard practice in pharmaceutical research and development, and regulatory agencies are increasingly requesting data on activity against targets with recognized adverse effect relationships. However, the screening strategies and target panels used by pharmaceutical companies may vary substantially. To help identify commonalities and differences, as well as to highlight opportunities for further optimization of secondary pharmacology assessment, we conducted a broad-ranging survey across 18 companies under the auspices of the DruSafe leadership group of the International Consortium for Innovation and Quality in Pharmaceutical Development. Based on our analysis of this survey and discussions and additional research within the group, we present here an overview of the current state of the art in secondary pharmacology screening. We discuss best practices, including additional safety-associated targets not covered by most current screening panels, and present approaches for interpreting and reporting off-target activities. We also provide an assessment of the safety impact of secondary pharmacology screening, and a perspective on opportunities and challenges in this rapidly developing field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mohan Rao
- Janssen Research & Development, San Diego, CA, USA
- Neurocrine Biosciences, San Diego, CA, USA
| | - Lyn Rosenbrier Ribeiro
- UCB Biopharma, Braine-l'Alleud, Belgium
- AstraZeneca, Cambridge, UK
- Grunenthal, Berkshire, UK
| | | | | | - Yoav Timsit
- Novartis Biomedical Research, Cambridge, MA, USA
- Blueprint Medicines, Cambridge, MA, USA
| | | | - Duncan Armstrong
- Novartis Biomedical Research, Cambridge, MA, USA
- Armstrong Pharmacology, Macclesfield, UK
| | | | | | - Russell Naven
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Novartis Biomedical Research, Cambridge, MA, USA
| | - Ravikumar Peri
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Alexion Pharmaceuticals, Wilmington, DE, USA
| | - Sonia Roberts
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - James M Vergis
- Faegre Drinker Biddle and Reath, LLP, Washington, DC, USA
| | | |
Collapse
|
12
|
Brown AP, Friedrichs GS, Tang HM, Traebert M, Weber V, Yao N, Yan GX. Electrophysiological Changes in the Rabbit Ventricular Wedge and Human-Induced Pluripotent Stem-Cell Derived (IPSC) Cardiomyocytes Translate to Severe Arrhythmia Observed in a Canine Toxicology Study, Not Predicted by Standard In Vitro Ion Channel Assays. Int J Toxicol 2024; 43:231-242. [PMID: 38327194 DOI: 10.1177/10915818241230900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
During drug discovery, small molecules are typically assayed in vitro for secondary pharmacology effects, which include ion channels relevant to cardiac electrophysiology. Compound A was an irreversible inhibitor of myeloperoxidase investigated for the treatment of peripheral artery disease. Oral doses in dogs at ≥5 mg/kg resulted in cardiac arrhythmias in a dose-dependent manner (at Cmax, free ≥1.53 μM) that progressed in severity with time. Nevertheless, a panel of 13 different cardiac ion channel (K, Na, and Ca) assays, including hERG, failed to identify pharmacologic risks of the molecule. Compound A and a related Compound B were evaluated for electrophysiological effects in the isolated rabbit ventricular wedge assay. Compounds A and B prolonged QT and Tp-e intervals at ≥1 and ≥.3 μM, respectively, and both prolonged QRS at ≥5 μM. Compound A produced early after depolarizations and premature ventricular complexes at ≥5 μM. These data indicate both compounds may be modulating hERG (Ikr) and Nav1.5 ion channels. In human IPSC cardiomyocytes, Compounds A and B prolonged field potential duration at ≥3 μM and induced cellular dysrhythmia at ≥10 and ≥3 μM, respectively. In a rat toxicology study, heart tissue: plasma concentration ratios for Compound A were ≥19X at 24 hours post-dose, indicating significant tissue distribution. In conclusion, in vitro ion channel assays may not always identify cardiovascular electrophysiological risks observed in vivo, which can be affected by tissue drug distribution. Risk for arrhythmia may increase with a "trappable" ion channel inhibitor, particularly if cardiac tissue drug levels achieve a critical threshold for pharmacologic effects.
Collapse
Affiliation(s)
- Alan P Brown
- Novartis Biomedical Research, Cambridge, MA, USA
| | | | | | | | | | - Nancy Yao
- Novartis Biomedical Research, East Hanover, NJ, USA
| | - Gan-Xin Yan
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| |
Collapse
|
13
|
Grandits T, Augustin CM, Haase G, Jost N, Mirams GR, Niederer SA, Plank G, Varró A, Virág L, Jung A. Neural network emulation of the human ventricular cardiomyocyte action potential for more efficient computations in pharmacological studies. eLife 2024; 12:RP91911. [PMID: 38598284 PMCID: PMC11006416 DOI: 10.7554/elife.91911] [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: 04/11/2024] Open
Abstract
Computer models of the human ventricular cardiomyocyte action potential (AP) have reached a level of detail and maturity that has led to an increasing number of applications in the pharmaceutical sector. However, interfacing the models with experimental data can become a significant computational burden. To mitigate the computational burden, the present study introduces a neural network (NN) that emulates the AP for given maximum conductances of selected ion channels, pumps, and exchangers. Its applicability in pharmacological studies was tested on synthetic and experimental data. The NN emulator potentially enables massive speed-ups compared to regular simulations and the forward problem (find drugged AP for pharmacological parameters defined as scaling factors of control maximum conductances) on synthetic data could be solved with average root-mean-square errors (RMSE) of 0.47 mV in normal APs and of 14.5 mV in abnormal APs exhibiting early afterdepolarizations (72.5% of the emulated APs were alining with the abnormality, and the substantial majority of the remaining APs demonstrated pronounced proximity). This demonstrates not only very fast and mostly very accurate AP emulations but also the capability of accounting for discontinuities, a major advantage over existing emulation strategies. Furthermore, the inverse problem (find pharmacological parameters for control and drugged APs through optimization) on synthetic data could be solved with high accuracy shown by a maximum RMSE of 0.22 in the estimated pharmacological parameters. However, notable mismatches were observed between pharmacological parameters estimated from experimental data and distributions obtained from the Comprehensive in vitro Proarrhythmia Assay initiative. This reveals larger inaccuracies which can be attributed particularly to the fact that small tissue preparations were studied while the emulator was trained on single cardiomyocyte data. Overall, our study highlights the potential of NN emulators as powerful tool for an increased efficiency in future quantitative systems pharmacology studies.
Collapse
Affiliation(s)
- Thomas Grandits
- Department of Mathematics and Scientific Computing, University of GrazGrazAustria
- NAWI Graz, University of GrazGrazAustria
| | - Christoph M Augustin
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| | - Gundolf Haase
- Department of Mathematics and Scientific Computing, University of GrazGrazAustria
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- HUN-REN-TKI, Research Group of PharmacologyBudapestHungary
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of NottinghamNottinghamUnited Kingdom
| | - Steven A Niederer
- Division of Imaging Sciences & Biomedical Engineering, King’s College LondonLondonUnited Kingdom
| | - Gernot Plank
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of GrazGrazAustria
- BioTechMed-GrazGrazAustria
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- HUN-REN-TKI, Research Group of PharmacologyBudapestHungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Alexander Jung
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of GrazGrazAustria
| |
Collapse
|
14
|
Kambayashi R, Goto A, Izumi-Nakaseko H, Takei Y, Sugiyama A. Characterization of cardiovascular profile of anti-influenza drug peramivir: A reverse-translational study using the isoflurane-anesthetized dog. J Pharmacol Sci 2024; 154:218-224. [PMID: 38395523 DOI: 10.1016/j.jphs.2024.02.002] [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: 11/11/2023] [Revised: 01/23/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
An injectable anti-influenza drug peramivir has been reported to induce QT-interval prolongation in some phase III studies, although its thorough QT/QTc study was negative. We investigated the discrepancy among those clinical studies using isoflurane-anesthetized beagle dogs (n = 4). Peramivir in doses of 1 mg/kg/10 min (sub-therapeutic dose) followed by 10 mg/kg/10 min (clinically-relevant dose) was intravenously administered. Peramivir prolonged QT interval/QTcV and Tpeak-Tend, and tended to delay ventricular repolarization in a reverse-frequency dependent manner, indicating IKr inhibition in vivo. Meanwhile, peramivir did not alter P-wave duration, PR interval or QRS width, indicating a lack of impact on cardiac conduction via Na+ or Ca2+ channel inhibition in vivo. Peramivir prolonged Tpeak-Tend and tended to prolong terminal repolarization period, which would develop substrates for initiating and maintaining spiral reentry, respectively. Meanwhile, peramivir did not prolong J-Tpeakc, which could not induce early afterdepolarization, a trigger inducing torsade de pointes. Thus, our results support that clinical dose exposure of peramivir can delay the ventricular repolarization in influenza patients. Peramivir has only a small potential to induce torsade de pointes in patients with the intact hearts, but caution should be paid on its use for patients formerly having the trigger for torsade de pointes.
Collapse
Affiliation(s)
- Ryuichi Kambayashi
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Ai Goto
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Hiroko Izumi-Nakaseko
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Yoshinori Takei
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Atsushi Sugiyama
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan.
| |
Collapse
|
15
|
Rusyn I, Wright FA. Ten years of using key characteristics of human carcinogens to organize and evaluate mechanistic evidence in IARC Monographs on the identification of carcinogenic hazards to humans: Patterns and associations. Toxicol Sci 2024; 198:141-154. [PMID: 38141214 PMCID: PMC10901152 DOI: 10.1093/toxsci/kfad134] [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: 12/25/2023] Open
Abstract
Systematic review and evaluation of mechanistic evidence using the Key Characteristics approach was proposed by the International Agency for Research on Cancer (IARC) in 2012 and used by the IARC Monographs Working Groups since 2015. Key Characteristics are 10 features of agents known to cause cancer in humans. From 2015 to 2022, a total of 19 Monographs (73 agents combined) used Key Characteristics for cancer hazard classification. We hypothesized that a retrospective analysis of applications of the Key Characteristics approach to cancer hazard classification using heterogenous mechanistic data on diverse agents would be informative for systematic reviews in decision-making. We extracted information on the conclusions, data types, and the role mechanistic data played in the cancer hazard classification from each Monograph. Statistical analyses identified patterns in the use of Key Characteristics, as well as trends and correlations among Key Characteristics, data types, and ultimate decisions. Despite gaps in data for many agents and Key Characteristics, several significant results emerged. Mechanistic data from in vivo animal, in vitro animal, and in vitro human studies were most impactful in concluding that an agent could cause cancer via a Key Characteristic. To exclude the involvement of a Key Characteristic, data from large-scale systematic in vitro testing programs such as ToxCast, were most informative. Overall, increased availability of systemized data streams, such as human in vitro data, would provide the basis for more confident and informed conclusions about both positive and negative associations and inform expert judgments on cancer hazard.
Collapse
Affiliation(s)
- Ivan Rusyn
- Department of Veterinary Pharmacology and Physiology, Texas A&M University, College Station, Texas 77843, USA
| | - Fred A Wright
- Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27606, USA
- Department of Biological Sciences, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27606, USA
| |
Collapse
|
16
|
Sang L, Zhou Z, Luo S, Zhang Y, Qian H, Zhou Y, He H, Hao K. An In Silico Platform to Predict Cardiotoxicity Risk of Anti-tumor Drug Combination with hiPSC-CMs Based In Vitro Study. Pharm Res 2024; 41:247-262. [PMID: 38148384 PMCID: PMC10879352 DOI: 10.1007/s11095-023-03644-4] [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: 07/18/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVE Antineoplastic agent-induced systolic dysfunction is a major reason for interruption of anticancer treatment. Although targeted anticancer agents infrequently cause systolic dysfunction, their combinations with chemotherapies remarkably increase the incidence. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a potent in vitro model to assess cardiovascular safety. However, quantitatively predicting the reduction of ejection fraction based on hiPSC-CMs is challenging due to the absence of the body's regulatory response to cardiomyocyte injury. METHODS Here, we developed and validated an in vitro-in vivo translational platform to assess the reduction of ejection fraction induced by antineoplastic drugs based on hiPSC-CMs. The translational platform integrates drug exposure, drug-cardiomyocyte interaction, and systemic response. The drug-cardiomyocyte interaction was implemented as a mechanism-based toxicodynamic (TD) model, which was then integrated into a quantitative system pharmacology-physiological-based pharmacokinetics (QSP-PBPK) model to form a complete translational platform. The platform was validated by comparing the model-predicted and clinically observed incidence of doxorubicin and trastuzumab-induced systolic dysfunction. RESULTS A total of 33,418 virtual patients were incorporated to receive doxorubicin and trastuzumab alone or in combination. For doxorubicin, the QSP-PBPK-TD model successfully captured the overall trend of systolic dysfunction incidences against the cumulative doses. For trastuzumab, the predicted incidence interval was 0.31-2.7% for single-agent treatment and 0.15-10% for trastuzumab-doxorubicin sequential treatment, covering the observations in clinical reports (0.50-1.0% and 1.5-8.3%, respectively). CONCLUSIONS In conclusion, the in vitro-in vivo translational platform is capable of predicting systolic dysfunction incidence almost merely depend on hiPSC-CMs, which could facilitate optimizing the treatment protocol of antineoplastic agents.
Collapse
Affiliation(s)
- Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Shizheng Luo
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yicui Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjie Qian
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
17
|
Grandits T, Augustin CM, Haase G, Jost N, Mirams GR, Niederer SA, Plank G, Varró A, Virág L, Jung A. Neural network emulation of the human ventricular cardiomyocyte action potential: a tool for more efficient computation in pharmacological studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553497. [PMID: 38234850 PMCID: PMC10793461 DOI: 10.1101/2023.08.16.553497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Computer models of the human ventricular cardiomyocyte action potential (AP) have reached a level of detail and maturity that has led to an increasing number of applications in the pharmaceutical sector. However, interfacing the models with experimental data can become a significant computational burden. To mitigate the computational burden, the present study introduces a neural network (NN) that emulates the AP for given maximum conductances of selected ion channels, pumps, and exchangers. Its applicability in pharmacological studies was tested on synthetic and experimental data. The NN emulator potentially enables massive speed-ups compared to regular simulations and the forward problem (find drugged AP for pharmacological parameters defined as scaling factors of control maximum conductances) on synthetic data could be solved with average root-mean-square errors (RMSE) of 0.47mV in normal APs and of 14.5mV in abnormal APs exhibiting early afterdepolarizations (72.5% of the emulated APs were alining with the abnormality, and the substantial majority of the remaining APs demonstrated pronounced proximity). This demonstrates not only very fast and mostly very accurate AP emulations but also the capability of accounting for discontinuities, a major advantage over existing emulation strategies. Furthermore, the inverse problem (find pharmacological parameters for control and drugged APs through optimization) on synthetic data could be solved with high accuracy shown by a maximum RMSE of 0.21 in the estimated pharmacological parameters. However, notable mismatches were observed between pharmacological parameters estimated from experimental data and distributions obtained from the Comprehensive in vitro Proarrhythmia Assay initiative. This reveals larger inaccuracies which can be attributed particularly to the fact that small tissue preparations were studied while the emulator was trained on single cardiomyocyte data. Overall, our study highlights the potential of NN emulators as powerful tool for an increased efficiency in future quantitative systems pharmacology studies.
Collapse
Affiliation(s)
- Thomas Grandits
- Department of Mathematics and Scientific Computing, University of Graz
- NAWI Graz, University of Graz
| | - Christoph M Augustin
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of Graz
- BioTechMed-Graz
| | - Gundolf Haase
- Department of Mathematics and Scientific Computing, University of Graz
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, University of Szeged
- HUN-REN-TKI, Research Group of Pharmacology
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham
| | - Steven A Niederer
- Division of Imaging Sciences & Biomedical Engineering, King's College London
| | - Gernot Plank
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of Graz
- BioTechMed-Graz
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged
- HUN-REN-TKI, Research Group of Pharmacology
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of Szeged
| | - Alexander Jung
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging - Division of Medical Physics and Biophysics, Medical University of Graz
| |
Collapse
|
18
|
Darpo B, Leishman DJ. The New S7B/E14 Q&A Document Provides Additional Opportunities to Replace the Thorough QT Study. J Clin Pharmacol 2023; 63:1256-1274. [PMID: 37455487 DOI: 10.1002/jcph.2309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Since 2015, concentration-QTc (C-QTc) analysis has been used to exclude the possibility that a drug has a concerning effect on the QTc interval. This has enabled the replacement of the designated thorough QT (TQT) study with serial electrocardiograms (ECGs) in routine clinical pharmacology studies, such as the first-in-human (FIH) study. The E14 revision has led to an increased proportion of FIH studies with the added objective of QT evaluation, with the intention of replacing the TQT study. With the more recent revision of the S7B/E14 Q&A document in February 2022, nonclinical assays/studies can be brought into the process of regulatory decisions at the time of marketing application. If the hERG (human ether-a-go-go-related gene) and the non-rodent in vivo study are conducted according to the described best practices and are negative, the previous requirement that a QTc effect of >10 milliseconds must be excluded in healthy subjects at plasma concentrations 2-fold above what can be seen in patients can be reduced to covering the concentrations seen in patients. For drugs that cannot be safely given in high doses to healthy subjects, ECG evaluation is often performed at the therapeutic dose in patients. If a QTc effect of >10 milliseconds can be excluded, an argument can be made that the drug should be considered as having a low likelihood of proarrhythmic effects due to delayedrepolarization, if supported by negative best practices hERG and in vivo studies. In this article, we describe what clinicians involved in early clinical development need to understand in terms of the hERG and in vivo studies to determine whether these meet best practices and therefore can be used in an integrated clinical/nonclinical QT/QTc risk assessment.
Collapse
|
19
|
Leishman DJ, Holdsworth DL, Lauver DA, Bailie MB, Roche BM. The "One-Step" approach for QT analysis increases the sensitivity of nonclinical QTc analysis. Clin Transl Sci 2023; 16:2253-2264. [PMID: 37726963 PMCID: PMC10651649 DOI: 10.1111/cts.13625] [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: 03/28/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023] Open
Abstract
Whether a compound prolongs cardiac repolarization independent of changes in beat rate is a critical question in drug research and development. Current practice is to resolve this in two steps. First, the QT interval is corrected for the influence of rate and then statistical significance is tested. There is renewed interest in improving the sensitivity of nonclinical corrected QT interval (QTc) assessment with modern studies having greater data density than previously utilized. The current analyses examine the effects of moxifloxacin or vehicle on the QT interval in nonhuman primates (NHPs) using a previously described one-step method. The primary end point is the statistical sensitivity of the assessment. Publications suggest that for a four animal crossover (4 × 4) in NHPs the minimal detectable difference (MDD) is greater than or equal to 10 ms, whereas in an eight animal crossover the MDD is ~6.5 ms. Using the one-step method, the MDD for the four animal NHP assessments was 3 ms. In addition, the one-step model accounted for day-to-day differences in the heart rate and QT-rate slope as well as drug-induced changes in these parameters. This method provides an increase in the sensitivity and reduces the number of animals necessary for detecting potential QT change and represents "best practice" in nonclinical QTc assessment in safety pharmacology studies.
Collapse
|
20
|
Rossman EI, Wisialowski TA, Vargas HM, Valentin JP, Rolf MG, Roche BM, Riley S, Pugsley MK, Nichols J, Li D, Leishman DJ, Kleiman RB, Greiter-Wilke A, Gintant GA, Engwall MJ, Delaunois A, Authier S. Best practice considerations for nonclinical in vivo cardiovascular telemetry studies in non-rodent species: Delivering high quality QTc data to support ICH E14/S7B Q&As. J Pharmacol Toxicol Methods 2023; 123:107270. [PMID: 37164235 DOI: 10.1016/j.vascn.2023.107270] [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: 02/20/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
The ICH E14/S7B Questions and Answers (Q&As) guideline introduces the concept of a "double negative" nonclinical scenario (negative hERG assay and negative in vivo QTc study) to demonstrate that a drug does not produce a clinically relevant QT prolongation (i.e., no QT liability). This nonclinical "double negative" data package, along with negative Phase 1 clinical QTc data, may be sufficient to substitute for a clinical Thorough QT (TQT) study in some specific cases. While standalone GLP in vivo cardiovascular studies in non-rodent species are standard practice during nonclinical drug development for small molecule programs, a variety of approaches to the design, conduct, analysis and interpretation are utilized across pharmaceutical companies and contract research organizations (CROs) that may, in some cases, negatively impact the stringent sensitivity needed to fulfill the new Q&As. Subject matter experts from both Pharma and CROs have collaborated to recommend best practices for more robust nonclinical cardiovascular telemetry studies in non-rodent species, with input from clinical and regulatory experts. The aim was to increase consistency and harmonization across the industry and to ensure delivery of high quality nonclinical QTc data to meet the proposed sensitivities defined within the revised ICH E14/S7B Q&As guideline (Q&As 5.1 and 6.1). The detailed best practice recommendations presented here cover the design and execution of the safety pharmacology cardiovascular study, including optimal methods for acquiring, analyzing, reporting, and interpreting the resulting QTc and pharmacokinetic data to allow for direct comparison to clinical exposures and assessment of safety margin for QTc prolongation.
Collapse
Affiliation(s)
- Eric I Rossman
- GSK, Nonclinical Safety, Safety Pharmacology, Collegeville, PA, USA.
| | - Todd A Wisialowski
- Pfizer Worldwide Research Development and Medical, Safety Pharmacology, Groton, CT, USA
| | - Hugo M Vargas
- Amgen Research, Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA
| | | | - Michael G Rolf
- AstraZeneca, Clinical Pharmacology & Safety Sciences, R&D, Gothenburg, Sweden
| | - Brian M Roche
- Charles River Laboratories, Global Safety Pharmacology, Ashland, OH, USA
| | - Steve Riley
- Pfizer Worldwide Research Development and Medical, Clinical Pharmacology, Groton, CT, USA
| | | | - Jill Nichols
- Labcorp Early Development Laboratories Inc., Madison, WI, USA
| | - Dingzhou Li
- Pfizer Global Product Development, Global Biometrics & Data Management, Groton, CT, USA
| | | | | | | | | | - Michael J Engwall
- Amgen Research, Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA
| | - Annie Delaunois
- UCB Biopharma SRL, Chemin du Foriest, B-1420 Braine-l'Alleud, Belgium
| | | |
Collapse
|
21
|
Rusyn I, Wright FA. Ten Years of Using Key Characteristics of Human Carcinogens to Organize and Evaluate Mechanistic Evidence in IARC Monographs on the Identification of Carcinogenic Hazards to Humans: Patterns and Associations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548354. [PMID: 37503163 PMCID: PMC10369858 DOI: 10.1101/2023.07.11.548354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Systematic review and evaluation of the mechanistic evidence only recently been instituted in cancer hazard identification step of decision-making. One example of organizing and evaluating mechanistic evidence is the Key Characteristics approach of the International Agency for Research on Cancer (IARC) Monographs on the Identification of Carcinogenic Hazards to Humans. The Key Characteristics of Human Carcinogens were proposed almost 10 years ago and have been used in every IARC Monograph since 2015. We investigated the patterns and associations in the use of Key Characteristics by the independent expert Working Groups. We examined 19 Monographs (2015-2022) that evaluated 73 agents. We extracted information on the conclusions by each Working Group on the strength of evidence for agent-Key Characteristic combinations, data types that were available for decisions, and the role mechanistic data played in the final cancer hazard classification. We conducted both descriptive and association analyses within and across data types. We found that IARC Working Groups were cautious when evaluating mechanistic evidence: for only ∼13% of the agents was strong evidence assigned for any Key Characteristic. Genotoxicity and cell proliferation were most data-rich, while little evidence was available for DNA repair and immortalization Key Characteristics. Analysis of the associations among Key Characteristics revealed that only chemical's metabolic activation was significantly co-occurring with genotoxicity and cell proliferation/death. Evidence from exposed humans was limited, while mechanistic evidence from rodent studies in vivo was often available. Only genotoxicity and cell proliferation/death were strongly associated with decisions on whether mechanistic data was impactful on the final cancer hazard classification. The practice of using the Key Characteristics approach is now well-established at IARC Monographs and other government agencies and the analyses presented herein will inform the future use of mechanistic evidence in regulatory decision-making.
Collapse
|
22
|
Pognan F, Beilmann M, Boonen HCM, Czich A, Dear G, Hewitt P, Mow T, Oinonen T, Roth A, Steger-Hartmann T, Valentin JP, Van Goethem F, Weaver RJ, Newham P. The evolving role of investigative toxicology in the pharmaceutical industry. Nat Rev Drug Discov 2023; 22:317-335. [PMID: 36781957 PMCID: PMC9924869 DOI: 10.1038/s41573-022-00633-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 02/15/2023]
Abstract
For decades, preclinical toxicology was essentially a descriptive discipline in which treatment-related effects were carefully reported and used as a basis to calculate safety margins for drug candidates. In recent years, however, technological advances have increasingly enabled researchers to gain insights into toxicity mechanisms, supporting greater understanding of species relevance and translatability to humans, prediction of safety events, mitigation of side effects and development of safety biomarkers. Consequently, investigative (or mechanistic) toxicology has been gaining momentum and is now a key capability in the pharmaceutical industry. Here, we provide an overview of the current status of the field using case studies and discuss the potential impact of ongoing technological developments, based on a survey of investigative toxicologists from 14 European-based medium-sized to large pharmaceutical companies.
Collapse
Affiliation(s)
- Francois Pognan
- Discovery and Investigative Safety, Novartis Pharma AG, Basel, Switzerland.
| | - Mario Beilmann
- Nonclinical Drug Safety Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Harrie C M Boonen
- Drug Safety, Dept of Exploratory Toxicology, Lundbeck A/S, Valby, Denmark
| | | | - Gordon Dear
- In Vitro In Vivo Translation, GlaxoSmithKline David Jack Centre for Research, Ware, UK
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Tomas Mow
- Safety Pharmacology and Early Toxicology, Novo Nordisk A/S, Maaloev, Denmark
| | - Teija Oinonen
- Preclinical Safety, Orion Corporation, Espoo, Finland
| | - Adrian Roth
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | | | - Freddy Van Goethem
- Predictive, Investigative & Translational Toxicology, Nonclinical Safety, Janssen Research & Development, Beerse, Belgium
| | - Richard J Weaver
- Innovation Life Cycle Management, Institut de Recherches Internationales Servier, Suresnes, France
| | - Peter Newham
- Clinical Pharmacology and Safety Sciences, AstraZeneca R&D, Cambridge, UK.
| |
Collapse
|
23
|
Engwall MJ, Baublits J, Chandra FA, Jones ZW, Wahlstrom J, Chui RW, Vargas HM. Evaluation of levocetirizine in beagle dog and cynomolgus monkey telemetry assays: Defining the no QTc effect profile by timepoint and concentration-QTc analysis. Clin Transl Sci 2023; 16:436-446. [PMID: 36369797 PMCID: PMC10014691 DOI: 10.1111/cts.13454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
In prior clinical studies, levocetirizine (LEVO) has demonstrated no effect on ventricular repolarization (QTc intervals), therefore it is a relevant negative control to assess in nonclinical assays to define low proarrhythmic risk. LEVO was tested in beagle dog and cynomolgus monkey (nonhuman primate [NHP]) telemetry models to understand the nonclinical-clinical translation of this negative control. One oral dose of vehicle, LEVO (10 mg/kg/species) or moxifloxacin (MOXI; 30 mg/kg/dog; 80 mg/kg/NHP) was administered to instrumented animals (N = 8/species) using a cross-over dosing design; MOXI was the in-study positive control. Corrected QT interval values (QTcI) were calculated using an individual animal correction factor. Blood samples were taken for drug exposure during telemetry and for pharmacokinetic (PK) analysis (same animals; different day) for exposure-response (C-QTc) modeling. Statistical analysis of QTc-by-timepoint data showed that LEVO treatment was consistent with vehicle, thus no effect on ventricular repolarization was observed over 24 h in both species. PK analysis indicated that LEVO-maximum concentration levels in dogs (range: 12,300-20,100 ng/ml) and NHPs (range: 4090-12,700 ng/ml) were ≥4-fold higher than supratherapeutic drug levels in clinical QTc studies. Slope analysis values in dogs (0.00019 ms/ng/ml) and NHPs (0.00016 ms/ng/ml) were similar to the human C-QTc relationship and indicated no relationship between QTc intervals and plasma levels of LEVO. MOXI treatment caused QTc interval prolongation (dog: 18 ms; NHP: 29 ms). The characterization of LEVO in these non-rodent telemetry studies further demonstrates the value and impact of the in vivo QTc assay to define a "no QTc effect" profile and support clinical safety assessment.
Collapse
Affiliation(s)
- Michael J Engwall
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| | - Joel Baublits
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| | - Fiona A Chandra
- Amgen Clinical Pharmacology Modeling & Simulation, Thousand Oaks, California, USA
| | - Zack W Jones
- Amgen Clinical Pharmacology Modeling & Simulation, Thousand Oaks, California, USA
| | - Jan Wahlstrom
- Pharmacokinetics, Amgen Research, Thousand Oaks, California, USA
| | - Ray W Chui
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| | - Hugo M Vargas
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| |
Collapse
|
24
|
Zhou X, Richardson DL, Dowlati A, Goel S, Sahebjam S, Strauss J, Chawla S, Wang D, Mould DR, Samnotra V, Faller DV, Venkatakrishnan K, Gupta N. Effect of Pevonedistat, an Investigational NEDD8-Activating Enzyme Inhibitor, on the QTc Interval in Patients With Advanced Solid Tumors. Clin Pharmacol Drug Dev 2023; 12:257-266. [PMID: 36382849 DOI: 10.1002/cpdd.1194] [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: 07/20/2022] [Accepted: 10/02/2022] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to assess the effect of pevonedistat, a neural precursor cell expressed, developmentally down-regulated protein 8 (NEDD8)-activating enzyme inhibitor, on the heart rate-corrected QT (QTc) interval in cancer patients. Patients were randomized 1:1 to receive pevonedistat 25 or 50 mg/m2 on day 1 and the alternate dose on day 8. Triplicate electrocardiograms were collected at intervals over 0-11 hours and at 24 hours via Holter recorders on days -1 (baseline), 1, and 8. Changes from time-matched baseline values were calculated for QTc by Fridericia (QTcF), PR, and QRS intervals. Serial time-matched blood samples for analysis of pevonedistat plasma pharmacokinetics were collected and a concentration-QTc analysis conducted. Safety was assessed by monitoring vital signs, physical examinations, and clinical laboratory tests. Forty-four patients were included in the QTc analysis. Maximum least square (LS) mean increase from time-matched baseline in QTcF was 3.2 milliseconds at 1 hour postdose for pevonedistat at 25 mg/m2 , while the LSs mean change from baseline in QTcF was -1.7 milliseconds 1 hour postdose at 50 mg/m2 . The maximum 2-sided 90% upper confidence bound was 6.7 and 2.9 milliseconds for pevonedistat at 25 and 50 mg/m2 , respectively. Pevonedistat did not result in clinically relevant effects on heart rate, nor on PR or QRS intervals. Results from pevonedistat concentration-QTc analysis were consistent with these findings. Administration of pevonedistat to cancer patients at a dose of up to 50 mg/m2 showed no evidence of QT prolongation, indicative of the lack of clinically meaningful effects on cardiac repolarization. ClinicalTrials.gov identifier: NCT03330106 (first registered on November 6, 2017).
Collapse
Affiliation(s)
- Xiaofei Zhou
- Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Debra L Richardson
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center and Sarah Cannon Research Institute, Oklahoma City, Oklahoma, USA
| | | | - Sanjay Goel
- Montefiore Medical Center, Bronx, New York, USA
| | - Solmaz Sahebjam
- University of South Florida H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | | | - Sant Chawla
- Sarcoma Oncology Center, Santa Monica, California, USA
| | - Ding Wang
- Henry Ford Hospital, Detroit, Michigan, USA
| | - Diane R Mould
- Projections Research Inc., Phoenixville, Pennsylvania, USA
| | - Vivek Samnotra
- Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Douglas V Faller
- Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | | | - Neeraj Gupta
- Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| |
Collapse
|
25
|
Vargas HM, Rossman EI, Wisialowski TA, Nichols J, Pugsley MK, Roche B, Gintant GA, Greiter-Wilke A, Kleiman RB, Valentin JP, Leishman DJ. Improving the in Vivo QTc assay: The value of implementing best practices to support an integrated nonclinical-clinical QTc risk assessment and TQT substitute. J Pharmacol Toxicol Methods 2023; 121:107265. [PMID: 36997076 DOI: 10.1016/j.vascn.2023.107265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Recent updates and modifications to the clinical ICH E14 and nonclinical ICH S7B guidelines, which both relate to the evaluation of drug-induced delayed repolarization risk, provide an opportunity for nonclinical in vivo electrocardiographic (ECG) data to directly influence clinical strategies, interpretation, regulatory decision-making and product labeling. This opportunity can be leveraged with more robust nonclinical in vivo QTc datasets based upon consensus standardized protocols and experimental best practices that reduce variability and optimize QTc signal detection, i.e., demonstrate assay sensitivity. The immediate opportunity for such nonclinical studies is when adequate clinical exposures (e.g., supratherapeutic) cannot be safely achieved, or other factors limit the robustness of the clinical QTc evaluation, e.g., the ICH E14 Q5.1 and Q6.1 scenarios. This position paper discusses the regulatory historical evolution and processes leading to this opportunity and details the expectations of future nonclinical in vivo QTc studies of new drug candidates. The conduct of in vivo QTc assays that are consistently designed, executed and analyzed will lead to confident interpretation, and increase their value for clinical QTc risk assessment. Lastly, this paper provides the rationale and basis for our companion article which describes technical details on in vivo QTc best practices and recommendations to achieve the goals of the new ICH E14/S7B Q&As, see Rossman et al., 2023 (this journal).
Collapse
|
26
|
Yamamiya I, Lester R, Sonnichsen D, Mina M, He Y, Benhadji KA. Effect of Futibatinib on Cardiac Repolarization: Results of a Randomized, Controlled, Double-Blind, QT/QTc, Phase 1 Study in Healthy Subjects. Clin Pharmacol Drug Dev 2023; 12:304-313. [PMID: 36404525 DOI: 10.1002/cpdd.1195] [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: 07/27/2022] [Accepted: 10/05/2022] [Indexed: 11/23/2022]
Abstract
Futibatinib, a fibroblast growth factor receptor (FGFR) 1-4 inhibitor, is being investigated for FGFR-aberrant tumors. A 4-period, crossover, phase 1 thorough QT/QTc study compared effects on Fridericia heart rate-corrected QT (QTcF) interval of single doses of futibatinib 20 and 80 mg (therapeutic and supratherapeutic doses, respectively), placebo, and moxifloxacin (positive control) in healthy subjects. The study objective was to assess the time-matched difference in change from baseline in QTcF (ddQTcF) between futibatinib and placebo. In addition, changes from baseline in QTcF and other electrocardiogram (ECG) parameters, pharmacokinetics, ECG morphology, and safety were assessed. Forty-eight subjects were randomized. ddQTcF upper limits of 2-sided 90%CIs remained <10 milliseconds (clinical threshold) for both futibatinib doses at all time points (range, 2.0-4.5 milliseconds). Assay sensitivity was demonstrated by lower limits of 2-sided 97.5%CIs of the dQTcF difference between moxifloxacin and placebo of >5 milliseconds. Futibatinib exposure increased in a dose-dependent manner, and no significant relationship was detected between plasma futibatinib concentration and ddQTcF. There were no significant effects on heart rate, other ECG parameters, or ECG morphology. No serious adverse events occurred. Futibatinib did not prolong QTcF or affect other cardiac measures at therapeutic or supratherapeutic doses.
Collapse
Affiliation(s)
| | | | - Daryl Sonnichsen
- Sonnichsen Pharmaceutical Associates, LLC, Collegeville, Pennsylvania, USA
| | - Mark Mina
- Taiho Oncology, Inc., Princeton, New Jersey, USA
| | - Yaohua He
- Taiho Oncology, Inc., Princeton, New Jersey, USA
| | | |
Collapse
|
27
|
Meyer C, Dierick I, Gauderat G, Langenhorst J, Sarr C, Leroux E, Chenel M, Fouliard S, Passier P. How to Successfully Generate an Alternative Approach to a Thorough QT Study: GLPG1972 as an Example. Clin Pharmacol Ther 2023; 113:310-320. [PMID: 35355254 PMCID: PMC10083980 DOI: 10.1002/cpt.2596] [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: 11/29/2021] [Accepted: 02/19/2022] [Indexed: 01/27/2023]
Abstract
During development of a drug, the requirement of evaluating the proarrhythmic risk and delayed repolarization needs to be fulfilled. Would it be possible to create an alternative to a thorough QT (TQT) study or is there a need to perform a dedicated TQT study? How is an alternative approach generated, what information is available, and which instructions are considered missing today to generate such an approach? This tutorial describes the considerations and path followed to create an early and feasible alternative to a TQT study using experience-based insights from a successful application to the US Food and Drug Administration for GLPG1972, an ADAMTS-5 inhibitor, and discusses the approach used in light of the current guidelines and literature.
Collapse
Affiliation(s)
| | | | | | | | | | - Emilie Leroux
- Translational Medicine DivisionServierSuresnesFrance
| | | | | | | |
Collapse
|
28
|
Suzuki N, Kambayashi R, Goto A, Izumi-Nakaseko H, Takei Y, Naito AT, Sugiyama A. Cardiovascular safety pharmacology of ivermectin assessed using the isoflurane-anesthetized beagle dogs: ICH S7B follow-up study. J Toxicol Sci 2023; 48:645-654. [PMID: 38044126 DOI: 10.2131/jts.48.645] [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: 12/05/2023]
Abstract
Antiparasitic ivermectin has been reported to induce cardiovascular adverse events, including orthostatic hypotension, tachycardia and cardiopulmonary arrest, of which the underlying pathophysiology remains unknown. Since its drug repurposing as an antiviral agent is underway at higher doses than those for antiparasitic, we evaluated the cardiovascular safety pharmacology of ivermectin using isoflurane-anesthetized beagle dogs (n=4). Ivermectin in doses of 0.1 followed by 1 mg/kg was intravenously infused over 10 min with an interval of 20 min, attaining peak plasma concentrations of 0.94 ± 0.04 and 8.82 ± 1.25 μg/mL, which were 29-31 and 276-288 times higher than those observed after its antiparasitic oral dose of 12 mg/body, respectively. The latter peak concentration was > 2 times greater than those inhibiting proliferation of dengue virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and hepatitis B virus in vitro. Ivermectin decreased heart rate without altering mean blood pressure, suggesting that ivermectin does not cause hypotension or tachycardia directly. Ivermectin hardly altered atrioventricular nodal or intraventricular conduction, indicating a lack of inhibitory action on Ca2+ or Na+ channel in vivo. Ivermectin prolonged QT interval/QTcV in a dose-related manner and tended to slow the repolarization speed in a reverse frequency-dependent manner, supporting previously described its IKr inhibition, which would explain Tpeak-Tend prolongation and heart-rate reduction in this study. Meanwhile, ivermectin did not significantly prolong J-Tpeakc or terminal repolarization period, indicating torsadogenic potential of ivermectin leading to the onset of cardiopulmonary arrest would be small. Thus, ivermectin has a broad range of cardiovascular safety profiles, which will help facilitate its drug repurposing.
Collapse
Affiliation(s)
- Nobuyuki Suzuki
- Department of Pharmacology, Faculty of Medicine, Toho University
- Division of Cellular Physiology, Department of Physiology, Toho University Graduate School of Medicine
| | | | - Ai Goto
- Department of Pharmacology, Faculty of Medicine, Toho University
| | | | - Yoshinori Takei
- Department of Pharmacology, Faculty of Medicine, Toho University
| | - Atsuhiko T Naito
- Division of Cellular Physiology, Department of Physiology, Toho University Graduate School of Medicine
| | - Atsushi Sugiyama
- Department of Pharmacology, Faculty of Medicine, Toho University
| |
Collapse
|
29
|
Qu Y, Kirby R, Davies R, Jinat A, Stabilini S, Wu B, Yu L, Gao B, Vargas HM. Time Is a Critical Factor When Evaluating Oligonucleotide Therapeutics in Human Ether-a-Go-Go-Related Gene Assays. Nucleic Acid Ther 2022; 33:132-140. [PMID: 36576986 PMCID: PMC10066779 DOI: 10.1089/nat.2022.0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In accord with International Conference on Harmonization S7B guidelines, an in vitro human ether-a-go-go-related gene (hERG) assay is one component of an integrated risk assessment for delayed ventricular repolarization. Function of hERG could be affected by direct (acute) mechanisms, or by indirect (chronic) mechanisms. Some approved oligonucleotide therapeutics had submitted hERG data to regulatory agents, which were all collected with the same protocol used for small-molecule testing (incubation time <20 min; acute), however, oligonucleotides have unique mechanisms and time courses of action (indirect). To reframe the hERG testing strategy for silencing RNA (siRNA), an investigation was performed to assess the time course for siRNA-mediated inhibition of hERG function and gene expression. Commercially available siRNAs of hERG were evaluated in a stable hERG-expressed cell line by whole-cell voltage clamp using automated electrophysiology and polymerase chain reaction. In the acute hERG study, no effects were observed after treatment with 100 nM siRNA for 20 min. The chronic effects of 100 nM siRNAs on hERG function were evaluated and recorded over 8-48 h following transfection. At 8 h there was no significant effect, whereas 77% reduction was observed at 48 h. Measurement of hERG mRNA levels demonstrated a 79% and 93% decrease of hERG mRNA at 8 and 48 h, respectively, consistent with inhibition of hERG transcription. The results indicate that an anti-hERG siRNA requires a long exposure time (48 h) in the hERG assay to produce a maximal reduction in hERG current; short exposures (20 min-8 h) had no effect. These findings imply that off-target profiling of novel oligonucleotides could benefit from using hERG protocol with long incubation times to de-risk potential off-target (indirect) effects on the hERG channel. This hERG assay modification may be important to consider if the findings are used to support an integrated nonclinical-clinical risk assessment for QTc (the duration of the QT interval adjusted for heart rate) prolongation.
Collapse
Affiliation(s)
- Yusheng Qu
- Amgen Research, Translational Safety and Bioanalytical Sciences, Amgen, Inc., Thousand Oaks, California, USA
| | - Robert Kirby
- Metrion Biosciences Ltd, Granta Center, Cambridge, United Kingdom
| | - Richard Davies
- Metrion Biosciences Ltd, Granta Center, Cambridge, United Kingdom
| | - Ayesha Jinat
- Metrion Biosciences Ltd, Granta Center, Cambridge, United Kingdom
| | | | - Bin Wu
- Hybrid Modality Engineering, Amgen, Inc., Thousand Oaks, California, USA
| | - Longchuan Yu
- Cardiometabolic Disorders, Amgen, Inc., Thousand Oaks, California, USA
| | - BaoXi Gao
- Amgen Research, Translational Safety and Bioanalytical Sciences, Amgen, Inc., Thousand Oaks, California, USA
| | - Hugo M Vargas
- Amgen Research, Translational Safety and Bioanalytical Sciences, Amgen, Inc., Thousand Oaks, California, USA
| |
Collapse
|
30
|
Li Y, Wan R, Liu J, Liu W, Ma L, Zhang H. In silico mechanisms of arsenic trioxide-induced cardiotoxicity. Front Physiol 2022; 13:1004605. [PMID: 36589437 PMCID: PMC9798418 DOI: 10.3389/fphys.2022.1004605] [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: 07/27/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
It has been found that arsenic trioxide (ATO) is effective in treating acute promyelocytic leukemia (APL). However, long QT syndrome was reported in patients receiving therapy using ATO, which even led to sudden cardiac death. The underlying mechanisms of ATO-induced cardiotoxicity have been investigated in some biological experiments, showing that ATO affects human ether-à-go-go-related gene (hERG) channels, coding rapid delayed rectifier potassium current (I Kr ), as well as L-type calcium (I CaL ) channels. Nevertheless, the mechanism by which these channel reconstitutions induced the arrhythmia in ventricular tissue remains unsolved. In this study, a mathematical model was developed to simulate the effect of ATO on ventricular electrical excitation at cellular and tissue levels by considering ATO's effects on I Kr and I CaL . The ATO-dose-dependent pore block model was incorporated into the I Kr model, and the enhanced degree of ATO to I CaL was based on experimental data. Simulation results indicated that ATO extended the action potential duration of three types of ventricular myocytes (VMs), including endocardial cells (ENDO), midmyocardial cells (MCELL), and epicardial cells (EPI), and exacerbated the heterogeneity among them. ATO could also induce alternans in all three kinds of VMs. In a cable model of the intramural ventricular strand, the effects of ATO are reflected in a prolonged QT interval of simulated pseudo-ECG and a wide vulnerable window, thus increasing the possibility of spiral wave formation in ventricular tissue. In addition to showing that ATO prolonged QT, we revealed that the heterogeneity caused by ATO is also an essential hazard factor. Based on this, a pharmacological intervention of ATO toxicity by resveratrol was undertaken. This study provides a further understanding of ATO-induced cardiotoxicity, which may help to improve the treatment for APL patients.
Collapse
Affiliation(s)
- Yacong Li
- Beijing Academy of Artificial Intelligence, Beijing, China
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China,*Correspondence: Jun Liu, ; Weichao Liu, ; Henggui Zhang,
| | - Weichao Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China,*Correspondence: Jun Liu, ; Weichao Liu, ; Henggui Zhang,
| | - Lei Ma
- Beijing Academy of Artificial Intelligence, Beijing, China,National Biomedical Imaging Center, Peking University, Beijing, China
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom,*Correspondence: Jun Liu, ; Weichao Liu, ; Henggui Zhang,
| |
Collapse
|
31
|
Yang X, Ribeiro AJS, Pang L, Strauss DG. Use of Human iPSC-CMs in Nonclinical Regulatory Studies for Cardiac Safety Assessment. Toxicol Sci 2022; 190:117-126. [PMID: 36099065 DOI: 10.1093/toxsci/kfac095] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a human-relevant platform for cardiac function assessment. Alternative assays using hiPSC-CMs are increasingly being employed for regulatory decision-making. A retrospective review revealed steady use of hiPSC-CM-based in vitro assays in nonclinical studies of drug-induced cardiotoxicity in regulatory submissions to the U.S. Food and Drug Administration (FDA). Most of the hiPSC-CMs data were obtained in exploratory studies and submitted as supportive evidence in concordance with other nonclinical data. Some of those studies were used to inform clinical trial design. This article provides an overview of the use of hiPSC-CMs in regulatory applications to FDA, with a focus on the integration of human-relevant in vitro data into proarrhythmic and non-proarrhythmic risk assessment. By identifying the regulatory submissions including hiPSC-CMs data, we explore their utility and discuss their limitations for predicting human cardiac safety in clinical trials. An important take-home message is that regulatory acceptance of hiPSC-CMs data is dependent on both the context of use and accurate data interpretation.
Collapse
Affiliation(s)
- Xi Yang
- Division of Pharmacology & Toxicology, Office of Cardiology, Hematology, Endocrinology, & Nephrology, Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration , Silver Spring, Maryland 20903, USA
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration , Silver Spring, Maryland 20903, USA
| | - Li Pang
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arizona 72079, USA
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration , Silver Spring, Maryland 20903, USA
| |
Collapse
|
32
|
Visone R, Lozano-Juan F, Marzorati S, Rivolta MW, Pesenti E, Redaelli A, Sassi R, Rasponi M, Occhetta P. Predicting human cardiac QT alterations and pro-arrhythmic effects of compounds with a 3D beating heart-on-chip platform. Toxicol Sci 2022; 191:47-60. [PMID: 36226800 PMCID: PMC9887672 DOI: 10.1093/toxsci/kfac108] [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] [Indexed: 02/03/2023] Open
Abstract
Determining the potential cardiotoxicity and pro-arrhythmic effects of drug candidates remains one of the most relevant issues in the drug development pipeline (DDP). New methods enabling to perform more representative preclinical in vitro studies by exploiting induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) are under investigation to increase the translational power of the outcomes. Here we present a pharmacological campaign conducted to evaluate the drug-induced QT alterations and arrhythmic events on uHeart, a 3D miniaturized in vitro model of human myocardium encompassing iPSC-CM and dermal fibroblasts embedded in fibrin. uHeart was mechanically trained resulting in synchronously beating cardiac microtissues in 1 week, characterized by a clear field potential (FP) signal that was recorded by means of an integrated electrical system. A drug screening protocol compliant with the new International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines was established and uHeart was employed for testing the effect of 11 compounds acting on single or multiple cardiac ion channels and well-known to elicit QT prolongation or arrhythmic events in clinics. The alterations of uHeart's electrophysiological parameters such as the beating period, the FP duration, the FP amplitude, and the detection of arrhythmic events prior and after drug administration at incremental doses were effectively analyzed through a custom-developed algorithm. Results demonstrated the ability of uHeart to successfully anticipate clinical outcome and to predict the QT prolongation with a sensitivity of 83.3%, a specificity of 100% and an accuracy of 91.6%. Cardiotoxic concentrations of drugs were notably detected in the range of the clinical highest blood drug concentration (Cmax), qualifying uHeart as a fit-to-purpose preclinical tool for cardiotoxicity studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | - Roberto Sassi
- Department of Computer Science, Università degli Studi di Milano, Milan, 20133, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | | |
Collapse
|
33
|
Appraisal of ICH E14/S7B Q&As adopted in February 2022 using thorough QT/QTc study data for α4-integrin antagonist carotegrast methyl in Japanese healthy subjects. J Pharmacol Sci 2022; 150:191-199. [DOI: 10.1016/j.jphs.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 12/14/2022] Open
|
34
|
Goto A, Sakamoto K, Kambayashi R, Izumi-Nakaseko H, Kawai S, Takei Y, Matsumoto A, Kanda Y, Sugiyama A. Validation of risk-stratification method for the chronic atrioventricular block cynomolgus monkey model and its mechanistic interpretation using 6 drugs with pharmacologically-distinct profile. Toxicol Sci 2022; 190:99-109. [PMID: 35993620 DOI: 10.1093/toxsci/kfac088] [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: 11/13/2022] Open
Abstract
Validation of risk-stratification method for the chronic atrioventricular block cynomolgus monkey model and its mechanistic interpretation were performed using 6 pharmacologically-distinct drugs. The following drugs were orally administered in conscious state, astemizole: 1, 5 and 10 mg/kg (n = 6); haloperidol: 1, 10 and 30 mg/kg (n = 5); amiodarone: 30 mg/kg (n = 4); famotidine: 10 mg/kg (n = 4); levofloxacin: 100 mg/kg (n = 4); and tolterodine: 0.2, 1 and 4.5 mg/kg (n = 4). Astemizole of 5 and 10 mg/kg significantly prolonged ΔΔQTcF, whereas no significant change was observed by the others. Torsade de pointes (TdP) was induced by astemizole of 5 and 10 mg/kg in 3/6 and 6/6, and by haloperidol of 10 and 30 mg/kg in 1/5 and 1/5, respectively, which was not observed in the others. Torsadogenic risk of the drugs was quantified using the criteria for the monkey model specified in our previous study. Namely, high-risk drugs induced TdP at ≤ 3times of their maximum clinical daily dose. Intermediate-risk drugs did not induce TdP at this dose range, but induced it at higher doses. Low/no-risk drugs never induced TdP at any dose tested. The magnitude of risk was intermediate for astemizole and haloperidol, and low/no risk for the others. The pre-specified, risk-stratification method for the monkey model may solve the issue existing between non-clinical models and patients with labile repolarization, which can reinforce the regulatory decision-making and labelling at time of marketing application of non-double-negative drug candidate (hERG assay positive and/or in vivo QT study positive).
Collapse
Affiliation(s)
- Ai Goto
- Department of Pharmacology, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Kengo Sakamoto
- Ina Research Inc, 2148-188 Nishiminowa, Ina-shi, Nagano, 399-4501, Japan
| | - Ryuichi Kambayashi
- Department of Pharmacology, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Hiroko Izumi-Nakaseko
- Department of Pharmacology, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Shinichi Kawai
- Department of Inflammation & Pain Control Research, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Yoshinori Takei
- Department of Pharmacology, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Akio Matsumoto
- Department of Aging Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Atsushi Sugiyama
- Department of Pharmacology, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan.,Department of Inflammation & Pain Control Research, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan.,Department of Aging Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| |
Collapse
|
35
|
van Bavel JJA, Beekman HDM, van Weperen VYH, van der Linde HJ, van der Heyden MAG, Vos MA. I Ks inhibitor JNJ303 prolongs the QT interval and perpetuates arrhythmia when combined with enhanced inotropy in the CAVB dog. Eur J Pharmacol 2022; 932:175218. [PMID: 36007604 DOI: 10.1016/j.ejphar.2022.175218] [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: 04/22/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Impaired IKs induced by drugs or due to a KCNQ1 mutation, diagnosed as long QT syndrome type 1 (LQT1) prolongs the QT interval and predisposes the heart to Torsade de Pointes (TdP) arrhythmias. The anesthetized chronic AV block (CAVB) dog is inducible for TdP after remodeling and IKr inhibitor dofetilide. We tested the proarrhythmic effect of IKs inhibition in the CAVB dog, and the proarrhythmic role of increased contractility herein. METHODS Dofetilide-inducible animals were included to test the proarrhythmic effect of 1) IKs inhibition by JNJ303 (0.63 mg/kg/10min i.v.; n = 4), 2) IKs inhibition combined with enhanced inotropy (ouabain, 0.045 mg/kg/1min i.v.; n = 6), and 3) the washout period of the anesthetic regime (n = 10). RESULTS JNJ303 prolonged the QTc interval (from 477 ± 53 ms to 565 ± 14 ms, P < 0.02) resembling standardized dofetilide-induced QTc prolongation. Single ectopic beats (n = 4) and ventricular tachycardia (VT) (n = 3) were present, increasing the arrhythmia score (AS) from 1.0 ± 0 to 7.1 ± 6.5. JNJ303 combined with ouabain increased contractile parameters (LVdP/dtmax from 1725 ± 273 to 4147 ± 611 mmHg/s, P < 0.01). Moreover, TdP arrhythmias were induced in 4/6 dogs and AS increased from 1.0 ± 0 to 20.2 ± 19.0 after JNJ303 and ouabain (P < 0.05). Finally, TdP arrhythmias were induced in 4/10 dogs during the anesthesia washout period and the AS increased from 1.1 ± 0.3 to 9.2 ± 11.2. CONCLUSION Mimicking LQT1 using IKs inhibitor JNJ303 prolongs the QTc interval and triggers ectopic beats and non-sustained VT in the CAVB dog. Induction of the more severe arrhythmic events (TdP) demands a combination of IKs inhibition with enhanced inotropy or ending the anesthetic regime.
Collapse
Affiliation(s)
- Joanne J A van Bavel
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Henriëtte D M Beekman
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Valerie Y H van Weperen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Henk J van der Linde
- Janssen Research & Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Marc A Vos
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
36
|
Klein MG, Krantz MJ, Fatima N, Watters A, Colon-Sanchez D, Geiger RM, Goldstein RE, Solhjoo S, Mehler PS, Flagg TP, Haigney MC. Methadone Blockade of Cardiac Inward Rectifier K + Current Augments Membrane Instability and Amplifies U Waves on Surface ECGs: A Translational Study. J Am Heart Assoc 2022; 11:e023482. [PMID: 35658478 PMCID: PMC9238736 DOI: 10.1161/jaha.121.023482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Methadone is associated with a disproportionate risk of sudden death and ventricular tachyarrhythmia despite only modest inhibition of delayed rectifier K+ current (IKr), the principal mechanism of drug-associated arrhythmia. Congenital defects of inward rectifier K+ current (IK1) have been linked to increased U-wave amplitude on ECG and fatal arrhythmia. We hypothesized that methadone may also be a potent inhibitor of IK1, contributing to delayed repolarization and manifesting on surface ECGs as augmented U-wave integrals. Methods and Results Using a whole-cell voltage clamp, methadone inhibited both recombinant and native IK1 with a half-maximal inhibitory concentration IC50) of 1.5 μmol/L, similar to that observed for IKr block (half-maximal inhibitory concentration of 2.9 μmol/L). Methadone modestly increased the action potential duration at 90% repolarization and slowed terminal repolarization at low concentrations. At higher concentrations, action potential duration at 90% repolarization lengthening was abolished, but its effect on terminal repolarization rose steadily and correlated with increased fluctuations of diastolic membrane potential. In parallel, patient ECGs were analyzed before and after methadone initiation, with 68% of patients having a markedly increased U-wave integral compared with premethadone (lead V3; mean +38%±15%, P=0.016), along with increased QT and TPeak to TEnd intervals, likely reflective of IKr block. Conclusions Methadone is a potent IK1 inhibitor that causes augmentation of U waves on surface ECG. We propose that increased membrane instability resulting from IK1 block may better explain methadone's arrhythmia risk beyond IKr inhibition alone. Drug-induced augmentation of U waves may represent evidence of blockade of multiple repolarizing ion channels, and evaluation of the effect of that agent on IK1 may be warranted.
Collapse
Affiliation(s)
- Michael G Klein
- Cardiology Division Department of Medicine Uniformed Services University Bethesda MD
| | - Mori J Krantz
- Denver Health Medical Center Cardiology Division Denver CO.,Department of Medicine University of Colorado School of Medicine Aurora CO.,Chief Science Advisor Cardiac Safety and Cardiovascular Imaging, Clario Inc. Philadelphia PA
| | - Naheed Fatima
- Department of Anatomy, Physiology & Genetics Uniformed Services University Bethesda MD
| | - Ashlie Watters
- ACUTE at Denver Health Denver CO.,Department of Medicine University of Colorado School of Medicine Aurora CO
| | - Dayan Colon-Sanchez
- Psychiatry Division Outpatient Behavioral Health Services, Denver Health, & University of Colorado School of Medicine Denver CO
| | - Robert M Geiger
- Department of Anatomy, Physiology & Genetics Uniformed Services University Bethesda MD
| | - Robert E Goldstein
- Cardiology Division Department of Medicine Uniformed Services University Bethesda MD
| | - Soroosh Solhjoo
- Cardiology Division Department of Medicine Uniformed Services University Bethesda MD.,Military Cardiovascular Outcomes Research (MiCOR) Bethesda MD
| | - Philip S Mehler
- ACUTE at Denver Health Denver CO.,Department of Medicine University of Colorado School of Medicine Aurora CO
| | - Thomas P Flagg
- Department of Anatomy, Physiology & Genetics Uniformed Services University Bethesda MD
| | - Mark C Haigney
- Cardiology Division Department of Medicine Uniformed Services University Bethesda MD.,Military Cardiovascular Outcomes Research (MiCOR) Bethesda MD
| |
Collapse
|
37
|
Valentin JP, Hoffmann P, Ortemann-Renon C, Koerner J, Pierson J, Gintant G, Willard J, Garnett C, Skinner M, Vargas HM, Wisialowski T, Pugsley MK. The Challenges of Predicting Drug-Induced QTc Prolongation in Humans. Toxicol Sci 2022; 187:3-24. [PMID: 35148401 PMCID: PMC9041548 DOI: 10.1093/toxsci/kfac013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The content of this article derives from a Health and Environmental Sciences Institute (HESI) consortium with a focus to improve cardiac safety during drug development. A detailed literature review was conducted to evaluate the concordance between nonclinical repolarization assays and the clinical thorough QT (TQT) study. Food and Drug Administration and HESI developed a joint database of nonclinical and clinical data, and a retrospective analysis of 150 anonymized drug candidates was reviewed to compare the performance of 3 standard nonclinical assays with clinical TQT study findings as well as investigate mechanism(s) potentially responsible for apparent discrepancies identified. The nonclinical assays were functional (IKr) current block (Human ether-a-go-go related gene), action potential duration, and corrected QT interval in animals (in vivo corrected QT). Although these nonclinical assays demonstrated good specificity for predicting negative clinical QT prolongation, they had relatively poor sensitivity for predicting positive clinical QT prolongation. After review, 28 discordant TQT-positive drugs were identified. This article provides an overview of direct and indirect mechanisms responsible for QT prolongation and theoretical reasons for lack of concordance between clinical TQT studies and nonclinical assays. We examine 6 specific and discordant TQT-positive drugs as case examples. These were derived from the unique HESI/Food and Drug Administration database. We would like to emphasize some reasons for discordant data including, insufficient or inadequate nonclinical data, effects of the drug on other cardiac ion channels, and indirect and/or nonelectrophysiological effects of drugs, including altered heart rate. We also outline best practices that were developed based upon our evaluation.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- Department of Investigative Toxicology, UCB Biopharma SRL, Braine-l’Alleud B-1420, Belgium
| | | | | | - John Koerner
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | - Jennifer Pierson
- Health and Environmental Sciences Institute, Washington, District of Columbia 20005, USA
| | | | - James Willard
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | - Christine Garnett
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | | | - Hugo M Vargas
- Department of Safety Pharmacology & Animal Research Center, Amgen, Thousand Oaks, California 91320, USA
| | - Todd Wisialowski
- Department of Safety Pharmacology, Pfizer, Groton, Connecticut 06340, USA
| | - Michael K Pugsley
- Department of Toxicology, Cytokinetics, South San Francisco, California 94080, USA
| |
Collapse
|
38
|
Gintant G. Assessing the Fidelity of Translation of Nonclinical Assays: A Pharma Perspective. Br J Pharmacol 2022; 179:2564-2576. [PMID: 35032025 DOI: 10.1111/bph.15796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/30/2022] Open
Abstract
Advances in nonclinical in vitro models, higher throughput approaches and the promise of human-derived preparations require methods to reliably assess the fidelity of translation of assays compared to in vivo models and clinical studies. This review discusses general principles and parameters useful to evaluate the value of nonclinical assays typically used to guide compound progression. I first consider the biological characteristics (including sensitivity and ability to recapitulate relevant responses) of models that form the foundation of an assay based on the questions posed. I then discuss the quantitative assessment of diagnostic performance and assay utility, including sensitivity and specificity, receiver-operator characteristic curves, positive and negative predictive values, likelihood ratios, along with advantages of combining two independent assays. Understanding the strengths and limitations of the biological model employed along with assay performance and context of use is essential to selecting the best assays supporting the best drug candidates.
Collapse
Affiliation(s)
- Gary Gintant
- Dept Integrative Pharmacology (ZR-13, Dept. AP-9A), AbbVie, North Chicago, IL, USA
| |
Collapse
|
39
|
Allegaert K, Salaets T, Ward RM, Annaert P, Smits A. QTc Intervals Are Prolonged in Late Preterm and Term Neonates during Therapeutic Hypothermia but Normalize Afterwards. CHILDREN (BASEL, SWITZERLAND) 2021; 8:1153. [PMID: 34943349 PMCID: PMC8700422 DOI: 10.3390/children8121153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND There are anecdotal reports on reversible QTc prolongation during therapeutic hypothermia (TH) for moderate to severe neonatal encephalopathy after asphyxia. As the QTc interval is a relevant biomarker for pharmacovigilance during medication development, a structured search and review on published neonatal QTc values to generate reference values is warranted to facilate medication development in this specific population. METHODS A structured search and literature assessment (PubMed, Embase, and Google Scholar) with 'Newborn/Infant, QT and hypothermia' was conducted (October 2021). Retrieved individual values were converted to QTc (Bazett) over postnatal age (day 1-7). RESULTS We retrieved 94 QTc intervals (during TH (n = 50, until day 3) or subsequent normothermia (n = 44, day 4-7)) in 33 neonates from 6 publications. The median (range) of QTc intervals during TH was 508 (430-678), and 410 (317-540) ms afterwards (difference 98 ms, or +28 ms/°C decrease). Four additional cohorts (without individual QTc intervals) confirmed the pattern and magnitude of the effect of body temperature on the QTc interval. CONCLUSIONS We highlighted a relevant non-maturational covariate (°C dependent TH) and generated reference values for the QTc interval in this specific neonatal subpopulation. This knowledge on QTc during TH should be considered and integrated in neonatal medication development.
Collapse
Affiliation(s)
- Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Department of Clinical Pharmacy, Erasmus MC, Postbus 2040, 3000 GA Rotterdam, The Netherlands
| | - Thomas Salaets
- Division of Pediatric Cardiology, Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Robert M. Ward
- Division of Neonatology and Clinical Pharmacology, Department of Pediatrics, University of Utah, Salt Lake City, UT 84132, USA;
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Neonatal Intensive Care Unit, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
40
|
Kilfoil P, Feng SL, Bassyouni A, Lee T, Leishman D, Li D, MacEwan DJ, Sharma P, Watt ED, Jenkinson S. Characterization of a high throughput human stem cell cardiomyocyte assay to predict drug-induced changes in clinical electrocardiogram parameters. Eur J Pharmacol 2021; 912:174584. [PMID: 34678241 DOI: 10.1016/j.ejphar.2021.174584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 01/12/2023]
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hIPSC-CM's) play an increasingly important role in the safety profiling of candidate drugs. For such models to have utility a clear understanding of clinical translation is required. In the present study we examined the ability of our hIPSC-CM model to predict the clinically observed effects of a diverse set of compounds on several electrocardiogram endpoints, including changes in QT and QRS intervals. To achieve this, compounds were profiled in a novel high throughput voltage-sensitive dye platform. Measurements were taken acutely (30 min) and chronically (24 h) to ensure that responses from compounds with slow onset kinetics or that affected surface ion channel expression would be captured. In addition, to avoid issues associated with changes in free drug levels due to protein binding, assays were run in serum free conditions. Changes in hIPSC-CM threshold APD90 values correlated with compound plasma exposures that produced a +10 ms change in clinical QTc (Pearson r2 = 0.80). In addition, randomForest modeling showed high predictivity in defining TdP risk (AUROC value = 0.938). Risk associated with QRS prolongation correlated with an increase in action potential rise-time (AUROC value = 0.982). The in-depth understanding of the clinical translatability of our hIPSC-CM model positions this assay to play a key role in defining cardiac risk early in drug development. Moreover, the ability to perform longer term studies enables the detection of compounds that may not be highlighted by more acute assay formats, such as inhibitors of hERG trafficking.
Collapse
Affiliation(s)
- Peter Kilfoil
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Shuyun Lily Feng
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Asser Bassyouni
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Tiffany Lee
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Derek Leishman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | | | - David J MacEwan
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L69 3GE, UK
| | - Parveen Sharma
- Department of Cardiovascular & Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, L69 3GE, UK
| | | | - Stephen Jenkinson
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA.
| |
Collapse
|
41
|
Fischer-Holzhausen S, Yamamoto K, Fjeldstad MP, Maleckar MM. Probing the Putative Role of K ATP Channels and Biological Variability in a Mathematical Model of Chondrocyte Electrophysiology. Bioelectricity 2021. [DOI: 10.1089/bioe.2021.0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Kei Yamamoto
- Department of Mathematics, University of Oslo, Oslo, Norway
- Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | | | - Mary M. Maleckar
- Computational Physiology, Simula Research Laboratory, Oslo, Norway
| |
Collapse
|
42
|
Smit T, Schickel E, Azimzadeh O, von Toerne C, Rauh O, Ritter S, Durante M, Schroeder IS. A Human 3D Cardiomyocyte Risk Model to Study the Cardiotoxic Influence of X-rays and Other Noxae in Adults. Cells 2021; 10:cells10102608. [PMID: 34685588 PMCID: PMC8533903 DOI: 10.3390/cells10102608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 02/06/2023] Open
Abstract
The heart tissue is a potential target of various noxae contributing to the onset of cardiovascular diseases. However, underlying pathophysiological mechanisms are largely unknown. Human stem cell-derived models are promising, but a major concern is cell immaturity when estimating risks for adults. In this study, 3D aggregates of human embryonic stem cell-derived cardiomyocytes were cultivated for 300 days and characterized regarding degree of maturity, structure, and cell composition. Furthermore, effects of ionizing radiation (X-rays, 0.1–2 Gy) on matured aggregates were investigated, representing one of the noxae that are challenging to assess. Video-based functional analyses were correlated to changes in the proteome after irradiation. Cardiomyocytes reached maximum maturity after 100 days in cultivation, judged by α-actinin lengths, and displayed typical multinucleation and branching. At this time, aggregates contained all major cardiac cell types, proven by the patch-clamp technique. Matured and X-ray-irradiated aggregates revealed a subtle increase in beat rates and a more arrhythmic sequence of cellular depolarisation and repolarisation compared to non-irradiated sham controls. The proteome analysis provides first insights into signaling mechanisms contributing to cardiotoxicity. Here, we propose an in vitro model suitable to screen various noxae to target adult cardiotoxicity by preserving all the benefits of a 3D tissue culture.
Collapse
Affiliation(s)
- Timo Smit
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany; (T.S.); (E.S.); (S.R.); (M.D.)
- Biology Department, Technische Universität Darmstadt, 64289 Darmstadt, Germany;
| | - Esther Schickel
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany; (T.S.); (E.S.); (S.R.); (M.D.)
| | - Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection (BfS), 85764 Neuherberg, Germany;
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Radiation Biology, 85764 Neuherberg, Germany
| | - Christine von Toerne
- Helmholtz Zentrum München-German Research Center for Environmental Health, Research Unit Protein Science, 80939 Munich, Germany;
| | - Oliver Rauh
- Biology Department, Technische Universität Darmstadt, 64289 Darmstadt, Germany;
| | - Sylvia Ritter
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany; (T.S.); (E.S.); (S.R.); (M.D.)
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany; (T.S.); (E.S.); (S.R.); (M.D.)
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, 64289 Darmstadt, Germany
| | - Insa S. Schroeder
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany; (T.S.); (E.S.); (S.R.); (M.D.)
- Correspondence:
| |
Collapse
|
43
|
Burnett SD, Blanchette AD, Chiu WA, Rusyn I. Cardiotoxicity Hazard and Risk Characterization of ToxCast Chemicals Using Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes from Multiple Donors. Chem Res Toxicol 2021; 34:2110-2124. [PMID: 34448577 PMCID: PMC8762671 DOI: 10.1021/acs.chemrestox.1c00203] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Heart disease remains a significant human health burden worldwide with a significant fraction of morbidity attributable to environmental exposures. However, the extent to which the thousands of chemicals in commerce and the environment may contribute to heart disease morbidity is largely unknown, because in contrast to pharmaceuticals, environmental chemicals are seldom tested for potential cardiotoxicity. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes have become an informative in vitro model for cardiotoxicity testing of drugs with the availability of cells from multiple individuals allowing in vitro testing of population variability. In this study, we hypothesized that a panel of iPSC-derived cardiomyocytes from healthy human donors can be used to screen for the potential cardiotoxicity hazard and risk of environmental chemicals. We conducted concentration-response testing of 1029 chemicals (drugs, pesticides, flame retardants, polycyclic aromatic hydrocarbons (PAHs), plasticizers, industrial chemicals, food/flavor/fragrance agents, etc.) in iPSC-derived cardiomyocytes from 5 donors. We used kinetic calcium flux and high-content imaging to derive quantitative measures as inputs into Bayesian population concentration-response modeling of the effects of each chemical. We found that many environmental chemicals pose a hazard to human cardiomyocytes in vitro with more than half of all chemicals eliciting positive or negative chronotropic or arrhythmogenic effects. However, most of the tested environmental chemicals for which human exposure and high-throughput toxicokinetics data were available had wide margins of exposure and, thus, do not appear to pose a significant human health risk in a general population. Still, relatively narrow margins of exposure (<100) were estimated for some perfuoroalkyl substances and phthalates, raising concerns that cumulative exposures may pose a cardiotoxicity risk. Collectively, this study demonstrated the value of using a population-based human in vitro model for rapid, high-throughput hazard and risk characterization of chemicals for which little to no cardiotoxicity data are available from guideline studies in animals.
Collapse
Affiliation(s)
- Sarah D. Burnett
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Alexander D. Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Weihsueh A. Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| |
Collapse
|
44
|
Rotordam MG, Obergrussberger A, Brinkwirth N, Takasuna K, Becker N, Horváth A, Goetze TA, Rapedius M, Furukawa H, Hasegawa Y, Oka T, Fertig N, Stoelzle-Feix S. Reliable identification of cardiac conduction abnormalities in drug discovery using automated patch clamp II: Best practices for Nav1.5 peak current in a high throughput screening environment. J Pharmacol Toxicol Methods 2021; 112:107125. [PMID: 34500078 DOI: 10.1016/j.vascn.2021.107125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/16/2021] [Accepted: 09/03/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION For reliable identification of cardiac safety risk, compounds should be screened for activity on cardiac ion channels in addition to hERG, including NaV1.5 and CaV1.2. We identified different parameters that might affect IC50s of compounds on NaV1.5 peak and late currents recorded using automated patch clamp (APC) and suggest outlines for best practices. METHODS APC instruments SyncroPatch 384 and Patchliner were used to record NaV1.5 peak and late current. Up to 24 CiPA compounds were used to investigate effects of voltage protocol, holding potential (-80 mV or - 95 mV) and temperature (23 ± 1 °C or 36 ± 1 °C) on IC50 values on hNaV1.5 overexpressed in HEK or CHO cells either as frozen cells or running cultures. RESULTS The IC50s of 18 compounds on the NaV1.5 peak current recorded on the SyncroPatch 384 using the CiPA step-ramp protocol correlated well with the literature. The use of frozen or cultured cells did not affect IC50s but voltage protocol and holding potential did cause differences in IC50 values. Temperature can affect Vhalf of inactivation and also compound potency. A compound incubation time of 5-6 min was sufficient for most compounds, however slow acting compounds such as terfenadine required longer to reach maximum effect. DISCUSSION We conclude that holding potential, voltage protocol and temperature can affect IC50 values and recommend the use of the CiPA step-ramp protocol at physiological temperature to record NaV1.5 peak and late currents for cardiac safety. Further recommendations include: a minimum compound incubation time of 5 min, a replicate number of 4 and the use of positive and negative controls for reliable IC50s.
Collapse
|
45
|
Lind L, Araujo JA, Barchowsky A, Belcher S, Berridge BR, Chiamvimonvat N, Chiu WA, Cogliano VJ, Elmore S, Farraj AK, Gomes AV, McHale CM, Meyer-Tamaki KB, Posnack NG, Vargas HM, Yang X, Zeise L, Zhou C, Smith MT. Key Characteristics of Cardiovascular Toxicants. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:95001. [PMID: 34558968 PMCID: PMC8462506 DOI: 10.1289/ehp9321] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
BACKGROUND The concept of chemical agents having properties that confer potential hazard called key characteristics (KCs) was first developed to identify carcinogenic hazards. Identification of KCs of cardiovascular (CV) toxicants could facilitate the systematic assessment of CV hazards and understanding of assay and data gaps associated with current approaches. OBJECTIVES We sought to develop a consensus-based synthesis of scientific evidence on the KCs of chemical and nonchemical agents known to cause CV toxicity along with methods to measure them. METHODS An expert working group was convened to discuss mechanisms associated with CV toxicity. RESULTS The group identified 12 KCs of CV toxicants, defined as exogenous agents that adversely interfere with function of the CV system. The KCs were organized into those primarily affecting cardiac tissue (numbers 1-4 below), the vascular system (5-7), or both (8-12), as follows: 1) impairs regulation of cardiac excitability, 2) impairs cardiac contractility and relaxation, 3) induces cardiomyocyte injury and death, 4) induces proliferation of valve stroma, 5) impacts endothelial and vascular function, 6) alters hemostasis, 7) causes dyslipidemia, 8) impairs mitochondrial function, 9) modifies autonomic nervous system activity, 10) induces oxidative stress, 11) causes inflammation, and 12) alters hormone signaling. DISCUSSION These 12 KCs can be used to help identify pharmaceuticals and environmental pollutants as CV toxicants, as well as to better understand the mechanistic underpinnings of their toxicity. For example, evidence exists that fine particulate matter [PM ≤2.5μm in aerodynamic diameter (PM2.5)] air pollution, arsenic, anthracycline drugs, and other exogenous chemicals possess one or more of the described KCs. In conclusion, the KCs could be used to identify potential CV toxicants and to define a set of test methods to evaluate CV toxicity in a more comprehensive and standardized manner than current approaches. https://doi.org/10.1289/EHP9321.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, University of Uppsala, Sweden
| | - Jesus A. Araujo
- Division of Cardiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), UCLA, Los Angeles, California, USA
- Department of Environmental Health Sciences, Fielding School of Public Health and Molecular Biology Institute, UCLA, Los Angeles, California, USA
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, USA
| | - Scott Belcher
- Department of Biological Sciences, North Carolina State University, North Carolina, USA
| | - Brian R. Berridge
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California, Davis, Davis, California, USA
| | - Weihsueh A. Chiu
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Vincent J. Cogliano
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency (EPA), Oakland, California, USA
| | - Sarah Elmore
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency (EPA), Oakland, California, USA
| | - Aimen K. Farraj
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Aldrin V. Gomes
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, California, USA
| | - Cliona M. McHale
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | | | - Nikki Gillum Posnack
- Children’s National Heart Institute and the Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC, USA
| | - Hugo M. Vargas
- Translational Safety & Bioanalytical Sciences, Amgen, Inc., Thousand Oaks, California, USA
| | - Xi Yang
- Division of Pharmacology and Toxicology, Office of Cardiology, Hematology, Endocrinology, and Nephrology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lauren Zeise
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency (EPA), Oakland, California, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Martyn T. Smith
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
46
|
Chi LH, Burrows AD, Anderson RL. Can preclinical drug development help to predict adverse events in clinical trials? Drug Discov Today 2021; 27:257-268. [PMID: 34469805 DOI: 10.1016/j.drudis.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/03/2021] [Accepted: 08/24/2021] [Indexed: 12/23/2022]
Abstract
The development of novel therapeutics is associated with high rates of attrition, with unexpected adverse events being a major cause of failure. Serious adverse events have led to organ failure, cancer development and deaths that were not expected outcomes in clinical trials. These life-threatening events were not identified during therapeutic development due to the lack of preclinical safety tests that faithfully represented human physiology. We highlight the successful application of several novel technologies, including high-throughput screening, organs-on-chips, microbiome-containing drug-testing platforms and humanised mouse models, for mechanistic studies and prediction of toxicity. We propose the incorporation of similar preclinical tests into future drug development to reduce the likelihood of hazardous therapeutics entering later-stage clinical trials.
Collapse
Affiliation(s)
- Lap Hing Chi
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Allan D Burrows
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Robin L Anderson
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
47
|
Schilling U, Henrich A, Muehlan C, Krause A, Dingemanse J, Ufer M. Impact of Daridorexant, a Dual Orexin Receptor Antagonist, on Cardiac Repolarization Following Bedtime Dosing: Results from a Thorough QT Study Using Concentration-QT Analysis. Clin Drug Investig 2021; 41:711-721. [PMID: 34331678 DOI: 10.1007/s40261-021-01062-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Daridorexant is a new dual orexin receptor antagonist currently in late-stage clinical development for the treatment of insomnia. This randomized, double-blind, placebo-controlled, four-period crossover study investigated the effect of daridorexant at a therapeutic and supratherapeutic dose on QT interval duration. METHODS Thirty-six healthy subjects received single oral doses of daridorexant (50 mg; 200 mg), moxifloxacin (400 mg; open label), and placebo. All treatments were administered at bedtime to mimic therapeutic practice. The primary analysis was based on linear mixed-effects concentration-QT modelling. Triplicate ECG data were extracted from Holter recordings at baseline and until 24 h post dosing at time points matching those for pharmacokinetic sampling. Plasma concentrations of daridorexant were determined over 24 h. RESULTS Assay sensitivity was demonstrated based on mean baseline- and placebo-corrected QT interval using Fridericia's formula (ΔΔQTcF) > 5 ms following moxifloxacin administration (p < 0.01). Following daridorexant administration, mean (90% confidence interval, CI) ΔΔQTcF was 1.40 ms (0.48; 2.32 ms) and 1.84 ms (-0.12; 3.79 ms) at the Cmax of 747 ng/mL (50 mg dose) and 1809 ng/mL (200 mg dose), respectively, i.e., the upper bounds of the CIs were < 10 ms defined as threshold of regulatory concern. Lack of relevant QT prolongation was confirmed by secondary by-time point analysis and absence of relevant findings in the categorical outlier analysis. Daridorexant was safe and well tolerated and its pharmacokinetics were consistent with previous data. CONCLUSION Daridorexant does not impair cardiac repolarization evidenced by absence of relevant QT prolongation at therapeutic and supratherapeutic doses. Clinical Trials Registration ID: NCT04250506.
Collapse
Affiliation(s)
- Uta Schilling
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Andrea Henrich
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Clemens Muehlan
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Andreas Krause
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| | - Jasper Dingemanse
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland.
| | - Mike Ufer
- Department of Clinical Pharmacology, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, 4123, Allschwil, Switzerland
| |
Collapse
|
48
|
Lester RM. Update on ICH E14/S7B Cardiac Safety Regulations: The Expanded Role of Preclinical Assays and the "Double-Negative" Scenario. Clin Pharmacol Drug Dev 2021; 10:964-973. [PMID: 34331518 PMCID: PMC8456868 DOI: 10.1002/cpdd.1003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/22/2021] [Indexed: 11/09/2022]
Abstract
For nearly 2 decades, regulators have adopted a harmonized approach to drug development, which has succeeded in bringing new pharmaceuticals to market without significant cardiac liability. Ushered in by technological advancements and better understanding of cellular electrophysiology, the initial paradigm detailed in the 2005 International Conference for Harmonization E14 and S7B documents has undergone evolutionary changes designed to streamline drug development and improve regulatory decision‐making and product labeling. The intent of this review is to summarize the new US Food and Drug Administration (FDA) Question and Answer update from August 2020 and key messaging from a subsequent FDA webinar describing best practices for preclinical and clinical data integration into a QT risk prediction model.
Collapse
|
49
|
Chui RW, Baublits J, Chandra FA, Jones ZW, Engwall MJ, Vargas HM. Evaluation of moxifloxacin in canine and non-human primate telemetry assays: Comparison of QTc interval prolongation by timepoint and concentration-QTc analysis. Clin Transl Sci 2021; 14:2379-2390. [PMID: 34173339 PMCID: PMC8604216 DOI: 10.1111/cts.13103] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 11/26/2022] Open
Abstract
The in vivo correct QT (QTc) assay is used by the pharmaceutical industry to characterize the potential for delayed ventricular repolarization and is a core safety assay mentioned in International Conference on Harmonization (ICH) S7B guideline. The typical telemetry study involves a dose‐response analysis of QTc intervals over time using a crossover (CO) design. This method has proven utility but does not include direct integration of pharmacokinetic (PK) data. An alternative approach has been validated and is used routinely in the clinical setting that pairs pharmacodynamic (PD) responses with PK exposure (e.g., concentration‐QTc (C‐QTc) analysis. The goal of our paper was to compare the QTc sensitivity of two experimental approaches in the conscious dog and non‐human primate (NHP) QTc assays. For timepoint analysis, a conventional design using eight animals (8 × 4 CO) to detect moxifloxacin‐induced QTc prolongation was compared to a PK/PD design in a subset (N = 4) of the same animals. The findings demonstrate that both approaches are equally sensitive in detecting threshold QTc prolongation on the order of 10 ms. Both QTc models demonstrated linearity in the QTc prolongation response to moxifloxacin dose escalation (6 to 46 ms). Further, comparison with human QTc findings with moxifloxacin showed agreement and consistent translation across the three species: C‐QTc slope values were 0.7‐ (dog) and 1.2‐ (NHP) fold of the composite human value. In conclusion, our data show that dog and NHP QTc telemetry with an integrated PK arm (C‐QTc) has the potential to supplement clinical evaluation and improve integrated QTc risk assessment.
Collapse
Affiliation(s)
- Ray W Chui
- Amgen Research, Thousand Oaks, California, USA
| | | | - Fiona A Chandra
- Amgen Translational Medicine, Thousand Oaks, California, USA
| | - Zack W Jones
- Amgen Translational Medicine, Thousand Oaks, California, USA
| | | | | |
Collapse
|
50
|
Darpo B, Ferber G. The New S7B/E14 Question and Answer Draft Guidance for Industry: Contents and Commentary. J Clin Pharmacol 2021; 61:1261-1273. [PMID: 33896027 PMCID: PMC9290990 DOI: 10.1002/jcph.1880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/20/2021] [Indexed: 11/10/2022]
Abstract
In August 2020, the International Council on Harmonisation (ICH) released a new draft document, which for the first time combined nonclinical (S7B) and clinical (E14) Questions and Answers (Q&As) into 1 document. FDA describes the revision as a “value proposition”: if the human ether‐à‐go‐go assay and the in vivo study are performed in a standardized way, the number of dedicated thorough QT (TQT) studies can be reduced. In this article, we describe and discuss the Q&As that relate to clinical ECG evaluation. If supported by negative standardized nonclinical assays, Q&A 5.1 will obviate the need for a TQT study in the case that a >2‐fold exposure margin vs high clinical scenario cannot be obtained. Q&A 6.1 addresses drugs that are poorly tolerated in healthy subjects and cannot be studied at high doses or in placebo‐controlled studies; it therefore mainly applies to oncology drugs. It will enable sponsors to claim that a new drug has a “low likelihood of proarrhythmic effects” in the case that the mean corrected QT effect is <10 milliseconds at the time of market application. The E14 2015 revision allowed application of concentration–corrected QT analysis on data from routinely performed clinical pharmacology studies, for example, the first‐in‐human study and the proportion of dedicated TQT studies has since steadily decreased. It can be foreseen that the proposed new revision will further reduce the number of TQT studies. To achieve harmonization across regulatory regions, it seems important to reach consensus within the International Council on Harmonisation group on the new threshold proposed in 6.1. For this purpose, the Implementation Working Group has asked for public comments.
Collapse
Affiliation(s)
- Borje Darpo
- Cardiac Safety, ERT, Rochester, New York, USA.,Cardiology, Karolinska Institute, Stockholm, Sweden
| | - Georg Ferber
- Statistik Georg Ferber GmbH, Riehen, Switzerland
| |
Collapse
|