1
|
Valentin JP, Leishman D. Points to consider for revising the ICH S7A guideline on safety and secondary pharmacology. Regul Toxicol Pharmacol 2025; 159:105795. [PMID: 40057171 DOI: 10.1016/j.yrtph.2025.105795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025]
Abstract
Although the ICH S7A guideline on safety pharmacology largely achieved its objective, a proportion of remaining adverse drug reactions and attrition can be attributed in part to gaps in safety and secondary pharmacology assessments. Advances in science, technology, drug development paradigm and regulatory practices necessitate revisiting and evolving ICH S7A to address these limitations. The anticipated completion of the ICH S7B Q&As by end of 2025 provides an opportunity to integrate its outcomes with ICH S7A into a comprehensive, modality-agnostic sustainable over time framework. Such consolidation could streamline guidance, enhance usability, and align regulatory expectations globally. This proposed revision should aim to address key aspects of safety and secondary pharmacology, including the definition of adversity, the integration of human-relevant in vitro and in silico models, and the adoption of state-of-the-art in vivo platforms. Further considerations should include the development of principles for model and assay validation, the promotion of integrated risk assessment frameworks, and incorporation of weight of evidence approaches. Revised guideline would also emphasize sustainable practices by adapting to evolving therapeutic modalities, while reducing reliance on animal testing through New Approach Methodologies. The revision seeks to enhance the benefit-risk evaluation of drug candidates, refine clinical monitoring, foster regulatory acceptance, and streamline drug development. This comprehensive update has the potential to not only optimize drug safety evaluations but also to align industry practices with modern scientific advancements and ethical considerations, ensuring a more robust and efficient pathway for therapeutic innovation.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- Non-Clinical Safety Evaluation, Early Clinical Development & Translational Science, UCB, Braine L'Alleud, Belgium.
| | - Derek Leishman
- Drug Disposition, Toxicology and PKPD, Eli Lilly and Company, Indianapolis, IN, 46285, USA.
| |
Collapse
|
2
|
Shuttleworth JG, Lei CL, Windley MJ, Hill AP, Preston SP, Mirams GR. Evaluating the predictive accuracy of ion-channel models using data from multiple experimental designs. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2025; 383:20240211. [PMID: 40078148 PMCID: PMC11904619 DOI: 10.1098/rsta.2024.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 03/14/2025]
Abstract
Mathematical models are increasingly being relied upon to provide quantitatively accurate predictions of cardiac electrophysiology. Many such models concern the behaviour of particular subcellular components (namely, ion channels) which, together, allow the propagation of electrical signals through heart-muscle tissue; that is, the firing of action potentials. In particular, IKr, a voltage-sensitive potassium ion-channel current, is of interest owing to the central pore of its primary protein having a propensity to blockage by various small molecules. We use newly collected data obtained from an ensemble of voltage-clamp experiment designs (protocols) to validate the predictive accuracy of various dynamical models of IKr. To do this, we fit models to each protocol individually and quantify the error in the resultant model predictions for other protocols. This allows the comparison of predictive accuracy for IKr models under a diverse collection of previously unexplored dynamics. Our results highlight heterogeneity between parameter estimates obtained from different cells, suggesting the presence of latent effects not yet accounted for in our models. This heterogeneity has a significant effect on our parameter estimates and suggests routes for model improvement.This article is part of the theme issue 'Uncertainty quantification for healthcare and biological systems (Part 1)'.
Collapse
Affiliation(s)
- Joseph G Shuttleworth
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Chon Lok Lei
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Monique J Windley
- Computational Cardiology Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Facility of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Adam P Hill
- Computational Cardiology Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Facility of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Simon P Preston
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
3
|
Takawale R, Singh D, Nikam VS. Cardiac physiological changes induced by cardiovascular drugs from different chemical classes in zebrafish mirrored in mice: A predictive tool for comprehensive risk assessment. Indian J Pharmacol 2025; 57:12-20. [PMID: 40324826 DOI: 10.4103/ijp.ijp_566_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/01/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE Our study investigated the impact of various cardiovascular drug on the cardiac physiology of zebrafish embryos and validated these findings in mice. BACKGROUND Cardiotoxicity has significantly contributed to the high drug attrition rate over the last two decades, underscoring the cardiac risk assessment in drug discovery and development. Although regulatory authority's guidelines specified the cell-based assays for the safety assessment of drugs, the current requirements fall short due to a lack of in vivo biology. The use of zebrafish experimental system has surged in developmental and pathophysiological investigation due to their striking resemblance to mammals. Hence, we used the zebrafish model system for cardiovascular drug studies and validated it in the mice model. MATERIALS AND METHODS The zebrafish embryos of 72 hours post-fertilization (hpf) were exposed to different CVS drug and, recorded their heart rate, and further validated in mice. RESULTS We observed that exposure to amlodipine (a calcium channel blocker), atenolol (a class II antiarrhythmic), and amiodarone (a class III antiarrhythmic) led to dose-dependent reductions in heart rate in zebrafish embryos, with effects varying based on drug concentration and mechanism of action. Specifically, amiodarone treatment resulted in a dose-dependent decrease in heart rate (0.001-100 μM) and atrioventricular block starting at a 10 μM concentration. Each class of cardiovascular drug demonstrated unique cardiac effects in zebrafish embryos, reflecting similar patterns in mice treated with these drugs. CONCLUSIONS Our findings highlight the zebrafish model's utility for early-phase cardiac risk assessment in drug discovery due to its high throughput capabilities and other beneficial features.
Collapse
Affiliation(s)
- Rohan Takawale
- Department of Pharmacology, Smt. Kashibai Navale College of Pharmacy, Savitribai Phule Pune University, Pune, Maharashtra, India
| | | | | |
Collapse
|
4
|
Yang KHS, Isaev D, Oz M. Additive Inhibition of HERG Channels Expressed in Xenopus Oocytes by Antipsychotic Drugs and Citrus Juice Flavonoid Naringenin. Pharmacology 2024; 110:122-126. [PMID: 39326403 DOI: 10.1159/000541005] [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: 04/04/2024] [Accepted: 08/14/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Citrus juice has been shown to cause QT prolongation in electrocardiograms of healthy volunteers, and naringenin, a major flavonoid found in citrus juice, has been identified as the potent inhibitor of human ether-a-go-go-related gene (HERG) channels as the cause of QT prolongation. Inhibition of HERG channels and prolongation of QT interval by antipsychotic drugs such as haloperidol, chlorpromazine, and clozapine have also been shown. However, naringenin's effect on HERG channel function in conjunction with antipsychotic medications has not been investigated. METHODS In the present study, we evaluated the effect of combining naringenin with antipsychotics on the function of HERG channels expressed in Xenopus oocytes. RESULTS When 30 µm naringenin was added to antipsychotic drugs (1 µm haloperidol, 10 µm chlorpromazine, or 10 µm clozapine), significantly greater HERG inhibition was demonstrated, compared to the inhibition caused by antipsychotic drugs alone. Co-application studies also showed that the magnitudes of inhibitions caused by naringenin + antipsychotics were similar to that predicted by the allotopic interaction model, suggesting that naringenin and antipsychotics bind to the HERG channel at different sites. CONCLUSION The results suggest that there is an additive interaction between antipsychotics and naringenin. Due to the potential for repolarization heterogeneity and a decrease in repolarization reserve, this additive HERG inhibition may increase the risk of arrhythmias.
Collapse
Affiliation(s)
- Keun-Hang Susan Yang
- Department of Biological Sciences, Schmid College of Science and Technology, Chapman University, One University Drive, Orange, California, USA
| | - Dmytro Isaev
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Murat Oz
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
5
|
Liu J, Khan MKH, Guo W, Dong F, Ge W, Zhang C, Gong P, Patterson TA, Hong H. Machine learning and deep learning approaches for enhanced prediction of hERG blockade: a comprehensive QSAR modeling study. Expert Opin Drug Metab Toxicol 2024; 20:665-684. [PMID: 38968091 DOI: 10.1080/17425255.2024.2377593] [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: 02/27/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Cardiotoxicity is a major cause of drug withdrawal. The hERG channel, regulating ion flow, is pivotal for heart and nervous system function. Its blockade is a concern in drug development. Predicting hERG blockade is essential for identifying cardiac safety issues. Various QSAR models exist, but their performance varies. Ongoing improvements show promise, necessitating continued efforts to enhance accuracy using emerging deep learning algorithms in predicting potential hERG blockade. STUDY DESIGN AND METHOD Using a large training dataset, six individual QSAR models were developed. Additionally, three ensemble models were constructed. All models were evaluated using 10-fold cross-validations and two external datasets. RESULTS The 10-fold cross-validations resulted in Mathews correlation coefficient (MCC) values from 0.682 to 0.730, surpassing the best-reported model on the same dataset (0.689). External validations yielded MCC values from 0.520 to 0.715 for the first dataset, exceeding those of previously reported models (0-0.599). For the second dataset, MCC values fell between 0.025 and 0.215, aligning with those of reported models (0.112-0.220). CONCLUSIONS The developed models can assist the pharmaceutical industry and regulatory agencies in predicting hERG blockage activity, thereby enhancing safety assessments and reducing the risk of adverse cardiac events associated with new drug candidates.
Collapse
Affiliation(s)
- Jie Liu
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Md Kamrul Hasan Khan
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Wenjing Guo
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Fan Dong
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Weigong Ge
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Chaoyang Zhang
- School of Computing Sciences and Computer Engineering, University of Southern Mississippi, Hattiesburg, MS, USA
| | - Ping Gong
- Environmental Laboratory, US Army Engineer Research and Development Center, Vicksburg, MS, USA
| | - Tucker A Patterson
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Huixiao Hong
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| |
Collapse
|
6
|
Chaves AA, Ferraro JW, Yu J, Moye MJ, Yee KL, Li F, Steve DL, Lengel DJ, Regan CP. Characterization of ascending dose canine telemetry model supports its use in E14/S7B QT integrated risk assessments. J Pharmacol Toxicol Methods 2024; 128:107525. [PMID: 38851600 DOI: 10.1016/j.vascn.2024.107525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
INTRODUCTION Nonclinical evaluation of the cardiovascular effects of novel chemical or biological entities (NCE, NBEs) is crucial for supporting first-in-human clinical trials. One important aspect of these evaluations is the assessment of potential QT/QTc prolongation risk, as drug-induced QT prolongation can have catastrophic effects. The recent publication of E14/S7B Q&As allows for the situational incorporation of nonclinical QTc data as part of an integrated risk assessment for a Thorough QT (TQT) waiver application provided certain best practice criteria are met. Recent publications provided detailed characterization of nonclinical QTc telemetry data collected from the commonly used Latin square study design. METHODS To understand whether data from alternate telemetry study designs were sufficient to serve as part of the E14/S7B integrated risk assessment, we report the performance and translational sensitivity to identify clinical risk of QTc prolongation risk for an ascending dose telemetry design. RESULTS The data demonstrated low variability in QTci interval within animals from day to day, indicating a well-controlled study environment and limited concern for uncontrolled effects across dosing days. Historical study variances of the ascending dose design with n = 4 subjects, measured by least significant difference (LSD) and root mean square error (RMSE) values, were low enough to detect a + 10 ms QTci interval change, and the median minimum detectable difference (MDD) for QTci interval changes was <10 ms. Furthermore, concentration-QTci (C-QTci) assessments to determine +10 ms QTci increases for known hERG inhibitors were comparable to clinical CC values listed in the E14/S7B training materials, supporting the use of the ascending dose design in an E14/S7B integrated risk assessment. DISCUSSION These findings suggest that the ascending dose design can be a valuable tool in nonclinical evaluation of QT/QTc prolongation risk and the support of TQT waiver applications.
Collapse
Affiliation(s)
- Alysia A Chaves
- Safety and Exploratory Pharmacology (SEP), Merck & Co., Inc., West Point, PA, USA.
| | - Jude W Ferraro
- Safety and Exploratory Pharmacology (SEP), Merck & Co., Inc., West Point, PA, USA
| | - Jing Yu
- Biometrics Research, Merck & Co., Inc., West Point, PA, USA
| | - Matthew J Moye
- Translational Medicine-Quantitative Pharmacology and Pharmacometrics (TMED-QP2), Merck & Co., Inc., West Point, PA, USA
| | - Ka Lai Yee
- Translational Medicine-Quantitative Pharmacology and Pharmacometrics (TMED-QP2), Merck & Co., Inc., West Point, PA, USA
| | - Fangbiao Li
- Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., West Point, PA, USA
| | - Desiree L Steve
- Safety and Exploratory Pharmacology (SEP), Merck & Co., Inc., West Point, PA, USA
| | - David J Lengel
- Safety and Exploratory Pharmacology (SEP), Merck & Co., Inc., West Point, PA, USA
| | - Christopher P Regan
- Safety and Exploratory Pharmacology (SEP), Merck & Co., Inc., West Point, PA, USA
| |
Collapse
|
7
|
Xi NM, Huang DP. Drug safety assessment by machine learning models. J Biopharm Stat 2024:1-12. [PMID: 38888177 DOI: 10.1080/10543406.2024.2365976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
The evaluation of drug-induced Torsades de pointes (TdP) risks is crucial in drug safety assessment. In this study, we discuss machine learning approaches in the prediction of drug-induced TdP risks using preclinical data. Specifically, a random forest model was trained on the dataset generated by the rabbit ventricular wedge assay. The model prediction performance was measured on 28 drugs from the Comprehensive In Vitro Proarrhythmia Assay initiative. Leave-one-drug-out cross-validation provided an unbiased estimation of model performance. Stratified bootstrap revealed the uncertainty in the asymptotic model prediction. Our study validated the utility of machine learning approaches in predicting drug-induced TdP risks from preclinical data. Our methods can be extended to other preclinical protocols and serve as a supplementary evaluation in drug safety assessment.
Collapse
Affiliation(s)
- Nan Miles Xi
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL, USA
| | - Dalong Patrick Huang
- Office of Biostatistics, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
8
|
Isaev D, Susan Yang KH, Oz M. Additive Effects of Citrus Juice Flavonoid Naringenin and Statins on HERG Channels Expressed in Xenopus Oocytes. Med Princ Pract 2024; 33:000538780. [PMID: 38593751 PMCID: PMC11324219 DOI: 10.1159/000538780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/08/2024] [Indexed: 04/11/2024] Open
Abstract
OBJECTIVE Naringenin, a major flavonoid found in citrus juice, has been shown to inhibit HERG channels and cause QT prolongation. Statins, the most commonly used class of cholesterol reducing drugs, have also been reported to inhibit HERG channels and prolong QT interval in patients using these drugs. However, the interaction between naringenin and statins on the function of HERG channels has not been studied. MATERIALS AND METHODS In the present study, we expressed HERG channels in Xenopus oocytes and tested the effects of naringenin and statins separately and combined on HERG channels. RESULTS When 30 µM naringenin was added to statins (1 µM rosuvastatin or 3 µM atorvastatin), significantly greater inhibition of HERG was demonstrated, compared to the inhibition caused by statins alone. CONCLUSIONS The results indicate that an additive interaction occurs between naringenin and statins; this could pose an increased risk of arrhythmias by decreasing repolarization reserve.
Collapse
Affiliation(s)
- Dmytro Isaev
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Keun-Hang Susan Yang
- Department of Biological Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | - Murat Oz
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
9
|
Bender AM, Parr LC, Livingston WB, Lindsley CW, Merryman WD. 2B Determined: The Future of the Serotonin Receptor 2B in Drug Discovery. J Med Chem 2023; 66:11027-11039. [PMID: 37584406 PMCID: PMC11073569 DOI: 10.1021/acs.jmedchem.3c01178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The cardiotoxicity associated with des-ethyl-dexfenfluramine (norDF) and related agonists of the serotonin receptor 2B (5-HT2B) has solidified the receptor's place as an "antitarget" in drug discovery. Conversely, a growing body of evidence has highlighted the utility of 5-HT2B antagonists for the treatment of pulmonary arterial hypertension (PAH), valvular heart disease (VHD), and related cardiopathies. In this Perspective, we summarize the link between the clinical failure of fenfluramine-phentermine (fen-phen) and the subsequent research on the role of 5-HT2B in disease progression, as well as the development of drug-like and receptor subtype-selective 5-HT2B antagonists. Such agents represent a promising class for the treatment of PAH and VHD, but their utility has been historically understudied due to the clinical disasters associated with 5-HT2B. Herein, it is our aim to examine the current state of 5-HT2B drug discovery, with an emphasis on the receptor's role in the central nervous system (CNS) versus the periphery.
Collapse
Affiliation(s)
- Aaron M Bender
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lauren C Parr
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - William B Livingston
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37240, United States
| |
Collapse
|
10
|
Xi NM, Hsu YY, Dang Q, Huang DP. Statistical learning in preclinical drug proarrhythmic assessment. J Biopharm Stat 2022; 32:450-473. [PMID: 35771997 DOI: 10.1080/10543406.2022.2065505] [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: 10/17/2022]
Abstract
Torsades de pointes (TdP) is an irregular heart rhythm characterized by faster beat rates and potentially could lead to sudden cardiac death. Much effort has been invested in understanding the drug-induced TdP in preclinical studies. However, a comprehensive statistical learning framework that can accurately predict the drug-induced TdP risk from preclinical data is still lacking. We proposed ordinal logistic regression and ordinal random forest models to predict low-, intermediate-, and high-risk drugs based on datasets generated from two experimental protocols. Leave-one-drug-out cross-validation, stratified bootstrap, and permutation predictor importance were applied to estimate and interpret the model performance under uncertainty. The potential outlier drugs identified by our models are consistent with their descriptions in the literature. Our method is accurate, interpretable, and thus useable as supplemental evidence in the drug safety assessment.
Collapse
Affiliation(s)
- Nan Miles Xi
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, Illinois, USA
| | - Yu-Yi Hsu
- Office of Biostatistics, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Qianyu Dang
- Office of Biostatistics, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Dalong Patrick Huang
- Office of Biostatistics, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
11
|
The grapefruit polyphenol naringenin inhibits multiple cardiac ion channels. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:735-740. [PMID: 35412073 DOI: 10.1007/s00210-022-02240-4] [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] [Received: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 01/08/2023]
Abstract
Drinking fresh grapefruit juice is associated with a significant prolongation of the QT segment on the electrocardiogram (ECG) in healthy volunteers. Among the prominent polyphenols contained in citrus fruits and primarily in grapefruit, the flavonoid naringenin is known to be a blocker of the human ether-a-go-go related gene (hERG) potassium channel. Here we hypothesized that naringenin could interfere with other major ion channels shaping the cardiac ventricular action potential (AP). To test this hypothesis, we examined the effects of naringenin on the seven channels comprising the Comprehensive in vitro Pro-Arrhythmia (CiPA) ion channel panel for early arrhythmogenic risk assessment in drug discovery and development. We used automated population patch-clamp of human ion channels heterologously expressed in mammalian cells to evaluate half-maximal inhibitory concentrations (IC50). Naringenin blocked all CiPA ion channels tested with IC50 values in the 30-100 µM concentration-range. The rank-order of channel sensitivity was the following: hERG > Kir2.1 > NaV1.5 (late current) > NaV1.5 (peak current) > KV7.1 > KV4.3 > CaV1.2. This multichannel inhibitory profile of naringenin suggests exercising caution when large amounts of grapefruit juice or other citrus juices enriched in this flavonoid polyphenol are drunk in conjunction with QT prolonging drugs or by carriers of congenital long-QT syndromes.
Collapse
|
12
|
Rodríguez-Lavado J, Alarcón-Espósito J, Mallea M, Lorente A. A new paradigm shift in antidepressant therapy? From dual-action to multitarget-directed ligands. Curr Med Chem 2022; 29:4896-4922. [PMID: 35301942 DOI: 10.2174/0929867329666220317121551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/10/2022] [Accepted: 01/15/2022] [Indexed: 11/22/2022]
Abstract
Major Depressive Disorder is a chronic, recurring, and potentially fatal disease affecting up to 20% of the global population. Since the monoamine hypothesis was proposed more than 60 years ago, only a few relevant advances have been achieved, with very little disease course changing, from a pharmacological perspective. Moreover, since negative efficacy studies with novel molecules are frequent, many pharmaceutical companies have put new studies on hold. Fortunately, relevant clinical studies are currently being performed, and extensive striving is being developed by universities, research centers, and other public and private institutions. Depression is no longer considered a simple disease but a multifactorial one. New research fields are emerging in what could be a paradigm shift: the multitarget approach beyond monoamines. In this review, we summarize the present and the past of antidepressant drug discovery, with the aim to shed some light on the current state of the art in clinical and preclinical advances to face this increasingly devastating disease.
Collapse
Affiliation(s)
- Julio Rodríguez-Lavado
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Química y Ciencias Farmacéuticas, Universidad de Chile, Casilla 233, Santiago, Chile
| | - Jazmín Alarcón-Espósito
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Química y Ciencias Farmacéuticas, Universidad de Chile, Casilla 233, Santiago, Chile
| | - Michael Mallea
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Química y Ciencias Farmacéuticas, Universidad de Chile, Casilla 233, Santiago, Chile
| | - Alejandro Lorente
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Química y Ciencias Farmacéuticas, Universidad de Chile, Casilla 233, Santiago, Chile
| |
Collapse
|
13
|
Ballet V, Bohme GA, Brohan E, Boukaiba R, Chambard JM, Angouillant-Boniface O, Carriot T, Chantoiseau C, Fouconnier S, Houtmann S, Prévost C, Schombert B, Schio L, Partiseti M. In vitro ion channel profile and ex vivo cardiac electrophysiology properties of the R(-) and S(+) enantiomers of hydroxychloroquine. Eur J Pharmacol 2022; 915:174670. [PMID: 34863995 PMCID: PMC9749463 DOI: 10.1016/j.ejphar.2021.174670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/03/2022]
Abstract
Hydroxychloroquine (HCQ) is a derivative of the antimalaria drug chloroquine primarily prescribed for autoimmune diseases. Recent attempts to repurpose HCQ in the treatment of corona virus disease 2019 has raised concerns because of its propensity to prolong the QT-segment on the electrocardiogram, an effect associated with increased pro-arrhythmic risk. Since chirality can affect drug pharmacological properties, we have evaluated the functional effects of the R(-) and S(+) enantiomers of HCQ on six ion channels contributing to the cardiac action potential and on electrophysiological parameters of isolated Purkinje fibers. We found that R(-)HCQ and S(+)HCQ block human Kir2.1 and hERG potassium channels in the 1 μM-100 μM range with a 2-4 fold enantiomeric separation. NaV1.5 sodium currents and CaV1.2 calcium currents, as well as KV4.3 and KV7.1 potassium currents remained unaffected at up to 90 μM. In rabbit Purkinje fibers, R(-)HCQ prominently depolarized the membrane resting potential, inducing autogenic activity at 10 μM and 30 μM, while S(+)HCQ primarily increased the action potential duration, inducing occasional early afterdepolarization at these concentrations. These data suggest that both enantiomers of HCQ can alter cardiac tissue electrophysiology at concentrations above their plasmatic levels at therapeutic doses, and that chirality does not substantially influence their arrhythmogenic potential in vitro.
Collapse
Affiliation(s)
- Véronique Ballet
- Preclinical Safety Investigative Toxicology, Sanofi-Aventis R&D, Chilly-Mazarin, France
| | - G Andrees Bohme
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France.
| | - Eric Brohan
- Early Development, Advanced Preparative Chromatography, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Rachid Boukaiba
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Jean-Marie Chambard
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | | | - Thierry Carriot
- Preclinical Safety Investigative Toxicology, Sanofi-Aventis R&D, Chilly-Mazarin, France
| | - Céline Chantoiseau
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Sophie Fouconnier
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Sylvie Houtmann
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Céline Prévost
- Early Development, Advanced Preparative Chromatography, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Brigitte Schombert
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Laurent Schio
- Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| | - Michel Partiseti
- High Content Biology, Integrated Drug Discovery, Sanofi-Aventis R&D, Vitry-sur-Seine, France
| |
Collapse
|
14
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
15
|
Whiting RL, Choppin A, Luehr G, Jasper JR. Preclinical Evaluation of the Effects of Trazpiroben (TAK-906), a Novel, Potent Dopamine D 2/D 3 Receptor Antagonist for the Management of Gastroparesis. J Pharmacol Exp Ther 2021; 379:85-95. [PMID: 34253646 DOI: 10.1124/jpet.121.000698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/01/2021] [Indexed: 11/22/2022] Open
Abstract
Current therapies for gastroparesis metoclopramide and domperidone carry risks of extrapyramidal symptoms and life-threatening cardiac arrhythmias. Trazpiroben, a novel, potent dopamine D2/D3 receptor antagonist, has low brain permeation and very low affinity for human ether-à-go-go-related gene (hERG) channel inhibition, potentially improving on safety profiles of existing therapies. Trazpiroben demonstrated the following receptor affinities: high for D2 and D3, moderate for D4, and minimal for D1 and D5 It demonstrated moderate affinity for adrenergic α 1B (α 1B) and 5-hydroxytryptamine (5HT) 2A receptors and low potential for off-target adverse events (AEs). Trazpiroben potently inhibited dopamine-activated D2L receptor activation of cognate G-proteins in human embryonic kidney 293 cell membranes and was a neutral D2L receptor antagonist. In vivo, trazpiroben dose-dependently increased prolactin release in orally dosed rat (0.1-1 mg/kg). Additionally, multiple oral doses in the rat (100 mg/kg) and dog (50 mg/kg) for 3 days produced robust plasma exposures and prolactin increases in both species. Trazpiroben inhibited retching/vomiting in the dog with apomorphine-induced emesis with a potency (0.1-1 mg/kg) like that of trazpiroben-mediated prolactin increases in rat. Oral trazpiroben (1, 10, and 30 mg/kg) did not affect rat rotarod performance, suggesting low brain penetration. Trazpiroben concentrations were low in cerebrospinal fluid versus plasma after multiple oral doses for 4 days in rat and dog. Trazpiroben weakly inhibited the hERG channel current (concentration causing half-maximal inhibition of control-specific binding of 15.6 µM), indicating little potential for disrupting cardiac rhythm. Overall, trazpiroben is a potent D2/D3 receptor antagonist designed to avoid the serious potential AEs associated with current gastroparesis therapies. SIGNIFICANCE STATEMENT: Trazpiroben is a novel, potent dopamine D2/D3 selective receptor antagonist designed to avoid adverse effects associated with the current pharmacological therapies metoclopramide and domperidone. Preclinical studies have demonstrated low brain penetration and weak affinity for the hERG channel, indicating that trazpiroben is not expected to be associated with central nervous system or cardiovascular safety issues. With these pharmacological properties, trazpiroben may represent a viable new treatment option for gastroparesis because of a potentially improved safety profile relative to existing therapies.
Collapse
Affiliation(s)
- Roger L Whiting
- Altos Therapeutics LLC, Los Altos, California (R.L.W., J.R.J.), and ARYx Therapeutics Inc., Fremont, California (A.C., G.L.)
| | - Agnes Choppin
- Altos Therapeutics LLC, Los Altos, California (R.L.W., J.R.J.), and ARYx Therapeutics Inc., Fremont, California (A.C., G.L.)
| | - Gary Luehr
- Altos Therapeutics LLC, Los Altos, California (R.L.W., J.R.J.), and ARYx Therapeutics Inc., Fremont, California (A.C., G.L.)
| | - Jeffrey R Jasper
- Altos Therapeutics LLC, Los Altos, California (R.L.W., J.R.J.), and ARYx Therapeutics Inc., Fremont, California (A.C., G.L.)
| |
Collapse
|
16
|
Dodson A, Mi K, Russo DP, Scott C, Saulnier M, Snyder K, Racz R. Aggregation and analysis of secondary pharmacology data from investigational new drug submissions at the US Food and Drug Administration. J Pharmacol Toxicol Methods 2021; 111:107098. [PMID: 34229067 DOI: 10.1016/j.vascn.2021.107098] [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: 03/29/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 01/23/2023]
Abstract
Secondary pharmacology studies are utilized by the pharmaceutical industry as a cost-efficient tool to identify potential safety liabilities of drugs before entering Phase 1 clinical trials. These studies are recommended by the Food and Drug Administration (FDA) as a part of the Investigational New Drug (IND) application. However, despite the utility of these assays, there is little guidance on which targets should be screened and which format should be used. Here, we evaluated 226 secondary pharmacology profiles obtained from close to 90 unique sponsors. The results indicated that the most tested target in our set was the GABA benzodiazepine receptor (tested 168 times), the most hit target was adenosine 3 (hit 24 times), and the target with the highest hit percentage was the quinone reductase 2 (NQO2) receptor (hit 29% of the time). The overall results were largely consistent with those observed in previous publications. However, this study also identified the need for improvement in the submission process of secondary pharmacology studies by industry, which could enhance their utility for regulatory purpose. FDA-industry collaborative working groups will utilize this data to determine the best methods for regulatory submission of these studies and evaluate the need for a standard target panel.
Collapse
Affiliation(s)
- Andrew Dodson
- Division of Applied Regulatory Science, Food and Drug Administration, Silver Spring, MD 20993, United States of America; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States of America
| | - Kevin Mi
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Daniel P Russo
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States of America; Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Christina Scott
- Division of Applied Regulatory Science, Food and Drug Administration, Silver Spring, MD 20993, United States of America; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States of America
| | - Muriel Saulnier
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Kevin Snyder
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Rebecca Racz
- Division of Applied Regulatory Science, Food and Drug Administration, Silver Spring, MD 20993, United States of America.
| |
Collapse
|
17
|
Cinnamic Acid Derivatives as Cardioprotective Agents against Oxidative and Structural Damage Induced by Doxorubicin. Int J Mol Sci 2021; 22:ijms22126217. [PMID: 34207549 PMCID: PMC8227863 DOI: 10.3390/ijms22126217] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Doxorubicin (DOX) is a widely used anticancer drug. However, its clinical use is severely limited due to drug-induced cumulative cardiotoxicity, which leads to progressive cardiomyocyte dysfunction and heart failure. Enormous efforts have been made to identify potential strategies to alleviate DOX-induced cardiotoxicity; however, to date, no universal and highly effective therapy has been introduced. Here we reported that cinnamic acid (CA) derivatives exert a multitarget protective effect against DOX-induced cardiotoxicity. The experiments were performed on rat cardiomyocytes (H9c2) and human induced-pluripotent-stem-cell-derived cardiomyocytes (hiPSC-CMs) as a well-established model for cardiac toxicity assessment. CA derivatives protected cardiomyocytes by ameliorating DOX-induced oxidative stress and viability reduction. Our data indicated that they attenuated the chemotherapeutic’s toxicity by downregulating levels of caspase-3 and -7. Pre-incubation of cardiomyocytes with CA derivatives prevented DOX-induced motility inhibition in a wound-healing assay and limited cytoskeleton rearrangement. Detailed safety analyses—including hepatotoxicity, mutagenic potential, and interaction with the hERG channel—were performed for the most promising compounds. We concluded that CA derivatives show a multidirectional protective effect against DOX-induced cardiotoxicity. The results should encourage further research to elucidate the exact molecular mechanism of the compounds’ activity. The lead structure of the analyzed CA derivatives may serve as a starting point for the development of novel therapeutics to support patients undergoing DOX therapy.
Collapse
|
18
|
Yang PC, Giles WR, Belardinelli L, Clancy CE. Mechanisms of flecainide induced negative inotropy: An in silico study. J Mol Cell Cardiol 2021; 158:26-37. [PMID: 34004185 PMCID: PMC8772296 DOI: 10.1016/j.yjmcc.2021.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/27/2022]
Abstract
It is imperative to develop better approaches to predict how antiarrhythmic drugs with multiple interactions and targets may alter the overall electrical and/or mechanical function of the heart. Safety Pharmacology studies have provided new insights into the multi-target effects of many different classes of drugs and have been aided by the addition of robust new in vitro and in silico technology. The primary focus of Safety Pharmacology studies has been to determine the risk profile of drugs and drug candidates by assessing their effects on repolarization of the cardiac action potential. However, for decades experimental and clinical studies have described substantial and potentially detrimental effects of Na+ channel blockers in addition to their well-known conduction slowing effects. One such side effect, associated with administration of some Na+ channel blocking drugs is negative inotropy. This reduces the pumping function of the heart, thereby resulting in hypotension. Flecainide is a well-known example of a Na+ channel blocking drug, that exhibits strong rate-dependent block of INa and may cause negative cardiac inotropy. While the phenomenon of Na+ channel suppression and resulting negative inotropy is well described, the mechanism(s) underlying this effect are not. Here, we set out to use a modeling and simulation approach to reveal plausible mechanisms that could explain the negative inotropic effect of flecainide. We utilized the Grandi-Bers model [1] of the cardiac ventricular myocyte because of its robust descriptions of ion homeostasis in order to characterize and resolve the relative effects of QRS widening, flecainide off-target effects and changes in intracellular Ca2+ and Na+ homeostasis. The results of our investigations and predictions reconcile multiple data sets and illustrate how multiple mechanisms may play a contributing role in the flecainide induced negative cardiac inotropic effect.
Collapse
Affiliation(s)
- Pei-Chi Yang
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, United States of America
| | - Wayne R Giles
- Department of Physiology & Pharmacology, University of Calgary, Canada
| | | | - Colleen E Clancy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, United States of America.
| |
Collapse
|
19
|
Bell DC, Fermini B. Use of automated patch clamp in cardiac safety assessment: past, present and future perspectives. J Pharmacol Toxicol Methods 2021; 110:107072. [PMID: 33962018 DOI: 10.1016/j.vascn.2021.107072] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/23/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022]
Abstract
There is no doubt that automated patch clamp (APC) technology has revolutionized research in biomedical science. High throughput ion channel screening is now an integral part of the development and safety profiling of the majority of new chemical entities currently developed to address unmet medical needs. The increased throughput it provides has significantly improved the ability to overcome the time-consuming, low throughput bottlenecks resulting from the more conventional manual patch clamp method, considered the 'gold standard', for studying ion channel function and pharmacology. While systems offering the luxury of automation have only been commercially available for two decades, the road leading to this new technology is long and rich in seminal, hands-on, studies dating back as far as the 18th century. So where does this technology currently stand, and what will it look like in the future? In the current article, we review the scientific history leading to the development of APC systems, examine key drivers in the rapid development of this technology (such as failed ion channel programmes and the issue of drug-induced hERG inhibition and QT interval prolongation), highlight key capabilities and finally provide some perspective on the current and future impact of the technology on cardiac safety assessment and biomedical science.
Collapse
|
20
|
Su S, Sun J, Wang Y, Xu Y. Cardiac hERG K + Channel as Safety and Pharmacological Target. Handb Exp Pharmacol 2021; 267:139-166. [PMID: 33829343 DOI: 10.1007/164_2021_455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The human ether-á-go-go related gene (hERG, KCNH2) encodes the pore-forming subunit of the potassium channel responsible for a fast component of the cardiac delayed rectifier potassium current (IKr). Outward IKr is an important determinant of cardiac action potential (AP) repolarization and effectively controls the duration of the QT interval in humans. Dysfunction of hERG channel can cause severe ventricular arrhythmias and thus modulators of the channel, including hERG inhibitors and activators, continue to attract intense pharmacological interest. Certain inhibitors of hERG channel prolong the action potential duration (APD) and effective refractory period (ERP) to suppress premature ventricular contraction and are used as class III antiarrhythmic agents. However, a reduction of the hERG/IKr current has been recognized as a predominant mechanism responsible for the drug-induced delayed repolarization known as acquired long QT syndromes (LQTS), which is linked to an increased risk for "torsades de pointes" (TdP) ventricular arrhythmias and sudden cardiac death. Many drugs of different classes and structures have been identified to carry TdP risk. Hence, assessing hERG/IKr blockade of new drug candidates is mandatory in the drug development process according to the regulatory agencies. In contrast, several hERG channel activators have been shown to enhance IKr and shorten the APD and thus might have potential antiarrhythmic effects against pathological LQTS. However, these activators may also be proarrhythmic due to excessive shortening of APD and the ERP.
Collapse
Affiliation(s)
- Shi Su
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Jinglei Sun
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Yi Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China.
| |
Collapse
|
21
|
Hrabinova M, Pejchal J, Kucera T, Jun D, Schmidt M, Soukup O. Is It the Twilight of BACE1 Inhibitors? Curr Neuropharmacol 2021; 19:61-77. [PMID: 32359337 PMCID: PMC7903497 DOI: 10.2174/1570159x18666200503023323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
β-secretase (BACE1) has been regarded as a prime target for the development of amyloid beta (Aβ) lowering drugs in the therapy of Alzheimer´s disease (AD). Although the enzyme was discovered in 1991 and helped to formulate the Aβ hypothesis as one of the very important features of AD etiopathogenesis, progress in AD treatment utilizing BACE1 inhibitors has remained limited. Moreover, in the last years, major pharmaceutical companies have discontinued clinical trials of five BACE1 inhibitors that had been strongly perceived as prospective. In our review, the Aβ hypothesis, the enzyme, its functions, and selected substrates are described. BACE1 inhibitors are classified into four generations. Those that underwent clinical trials displayed adverse effects, including weight loss, skin rashes, worsening of neuropsychiatric symptoms, etc. Some inhibitors could not establish a statistically significant risk-benefit ratio, or even scored worse than placebo. We still believe that drugs targeting BACE1 may still hide some potential, but a different approach to BACE1 inhibition or a shift of focus to modulation of its trafficking and/or post-translational modification should now be followed.
Collapse
Affiliation(s)
| | - Jaroslav Pejchal
- Address correspondence to this author at the Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence in Brno, Hradec Kralove, Czech Republic;E-mail:
| | | | | | | | | |
Collapse
|
22
|
Vargas HM, Rolf MG, Wisialowski TA, Achanzar W, Bahinski A, Bass A, Benson CT, Chaudhary KW, Couvreur N, Dota C, Engwall MJ, Michael Foley C, Gallacher D, Greiter-Wilke A, Guillon JM, Guth B, Himmel HM, Hegele-Hartung C, Ito M, Jenkinson S, Chiba K, Lagrutta A, Levesque P, Martel E, Okai Y, Peri R, Pointon A, Qu Y, Teisman A, Traebert M, Yoshinaga T, Gintant GA, Leishman DJ, Valentin JP. Time for a Fully Integrated Nonclinical-Clinical Risk Assessment to Streamline QT Prolongation Liability Determinations: A Pharma Industry Perspective. Clin Pharmacol Ther 2020; 109:310-318. [PMID: 32866317 PMCID: PMC7891594 DOI: 10.1002/cpt.2029] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/13/2020] [Indexed: 02/03/2023]
Abstract
Defining an appropriate and efficient assessment of drug‐induced corrected QT interval (QTc) prolongation (a surrogate marker of torsades de pointes arrhythmia) remains a concern of drug developers and regulators worldwide. In use for over 15 years, the nonclinical International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) S7B and clinical ICH E14 guidances describe three core assays (S7B: in vitro hERG current & in vivo QTc studies; E14: thorough QT study) that are used to assess the potential of drugs to cause delayed ventricular repolarization. Incorporating these assays during nonclinical or human testing of novel compounds has led to a low prevalence of QTc‐prolonging drugs in clinical trials and no new drugs having been removed from the marketplace due to unexpected QTc prolongation. Despite this success, nonclinical evaluations of delayed repolarization still minimally influence ICH E14‐based strategies for assessing clinical QTc prolongation and defining proarrhythmic risk. In particular, the value of ICH S7B‐based “double‐negative” nonclinical findings (low risk for hERG block and in vivo QTc prolongation at relevant clinical exposures) is underappreciated. These nonclinical data have additional value in assessing the risk of clinical QTc prolongation when clinical evaluations are limited by heart rate changes, low drug exposures, or high‐dose safety considerations. The time has come to meaningfully merge nonclinical and clinical data to enable a more comprehensive, but flexible, clinical risk assessment strategy for QTc monitoring discussed in updated ICH E14 Questions and Answers. Implementing a fully integrated nonclinical/clinical risk assessment for compounds with double‐negative nonclinical findings in the context of a low prevalence of clinical QTc prolongation would relieve the burden of unnecessary clinical QTc studies and streamline drug development.
Collapse
Affiliation(s)
- Hugo M Vargas
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| | - Michael G Rolf
- Research & Development, Clinical Pharmacology & Safety Sciences, AstraZeneca, Gothenburg, Sweden
| | - Todd A Wisialowski
- Global Safety Pharmacology, Pfizer Global Research and Development, Groton, Connecticut, USA
| | | | | | - Alan Bass
- Merck & Co., Inc., Boston, Massachusetts, USA
| | | | | | - Nicolas Couvreur
- Safety Pharmacology, Institute de Recherches Servier, Suresnes, France
| | - Corina Dota
- Research & Development, Chief Medical Officer Organization, AstraZeneca, Gothenburg, Sweden
| | - Michael J Engwall
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| | - C Michael Foley
- Integrative Pharmacology, Abbvie, Inc, North Chicago, Illinois, USA
| | - David Gallacher
- Global Safety Pharmacology, Janssen Research & Development, Beerse, Belgium
| | | | | | - Brian Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | | | - Maki Ito
- Japan Pharmaceutical Manufacturers Association, Tokyo, Japan
| | - Stephen Jenkinson
- Global Safety Pharmacology, Pfizer Global Research and Development, San Diego, California, USA
| | - Katsuyoshi Chiba
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Paul Levesque
- BMS Bristol-Myers Squibb Company, Princeton, New Jersey, USA
| | - Eric Martel
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Yoshiko Okai
- Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Ravikumar Peri
- Takeda Pharmaceutical Company Ltd., Cambridge, Massachusetts, USA
| | - Amy Pointon
- Research & Development, Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, UK
| | - Yusheng Qu
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, USA
| | - Ard Teisman
- Global Safety Pharmacology, Janssen Research & Development, Beerse, Belgium
| | - Martin Traebert
- Safety Pharmacology, Novartis Institute of Biomedical Research, Basel, Switzerland
| | | | - Gary A Gintant
- Integrative Pharmacology, Abbvie, Inc, North Chicago, Illinois, USA
| | | | | |
Collapse
|
23
|
Crawford JJ, Lee W, Johnson AR, Delatorre KJ, Chen J, Eigenbrot C, Heidmann J, Kakiuchi-Kiyota S, Katewa A, Kiefer JR, Liu L, Lubach JW, Misner D, Purkey H, Reif K, Vogt J, Wong H, Yu C, Young WB. Stereochemical Differences in Fluorocyclopropyl Amides Enable Tuning of Btk Inhibition and Off-Target Activity. ACS Med Chem Lett 2020; 11:1588-1597. [PMID: 32832028 DOI: 10.1021/acsmedchemlett.0c00249] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Bruton's tyrosine kinase (Btk) is thought to play a pathogenic role in chronic immune diseases such as rheumatoid arthritis and lupus. While covalent, irreversible Btk inhibitors are approved for treatment of hematologic malignancies, they are not approved for autoimmune indications. In efforts to develop additional series of reversible Btk inhibitors for chronic immune diseases, we sought to differentiate from our clinical stage inhibitor fenebrutinib using cyclopropyl amide isosteres of the 2-aminopyridyl group to occupy the flat, lipophilic H2 pocket. While drug-like properties were retained-and in some cases improved-a safety liability in the form of hERG inhibition was observed. When a fluorocyclopropyl amide was incorporated, Btk and off-target activity was found to be stereodependent and a lead compound was identified in the form of the (R,R)- stereoisomer.
Collapse
Affiliation(s)
- James J. Crawford
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Wendy Lee
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Adam R. Johnson
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Kelly J. Delatorre
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jacob Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Charles Eigenbrot
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Julia Heidmann
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Arna Katewa
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - James R. Kiefer
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lichuan Liu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joseph W. Lubach
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dinah Misner
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hans Purkey
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Karin Reif
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jennifer Vogt
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Harvey Wong
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Christine Yu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Wendy B. Young
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
24
|
Dynamic features of cardiac vector as alternative markers of drug-induced spatial dispersion. J Pharmacol Toxicol Methods 2020; 104:106894. [PMID: 32645483 DOI: 10.1016/j.vascn.2020.106894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 05/24/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The abnormal amplification of ventricular repolarization dispersion (VRD) has long been linked to proarrhythmia risk. Recently, the measure of VRD through electrocardiogram intervals has been strongly questioned. The search for an efficient and non-invasive surrogate marker of drug-induced dispersion effects constitute an urgent research challenge. METHODS Herein, drug-induced ventricular dispersion is generated by d-Sotalol supply in an In-vitro rabbit heart model. A cilindrical chamber simulates the thorax and a multi-electrode net is used to obtain spatial electrocardiographic signals. Cardiac vector dynamics is captured by novel velocity cardiomarkers obtained by quaternion methods. Through statistical analysis and machine learning technics, we compute potential dispersion markers that could define proarrhythmic risk. RESULTS The cardiomarkers with the greatest statistical significance, both obtained from the electrical cardiac vector, were: the QTω, which is the difference between first and last maxima of angular velocity and λ21vT, the roundness of linear velocity. When comparing with the performance of the current standards (89%), this pair was able to correctly separate 21 out of 22 experiments achieving a performance of 95%. Moreover, the QTω computes in a much more robust basis the QT interval, the current index for drug regulation. DISCUSSION These velocity markers circumvent the problems of accuratelly finding the fiducial points such as the always tricky T-wave end. Given the high performance they achieved, it is provided a promising outcome for future applications to the detection of anomalous changes of heterogeneity that may be useful for the purposes of torsadogenic toxicity studies.
Collapse
|
25
|
Avgoustou P, Jailani ABA, Zirimwabagabo JO, Tozer MJ, Gibson KR, Glossop PA, Mills JEJ, Porter RA, Blaney P, Bungay PJ, Wang N, Shaw AP, Bigos KJA, Holmes JL, Warrington JI, Skerry TM, Harrity JPA, Richards GO. Discovery of a First-in-Class Potent Small Molecule Antagonist against the Adrenomedullin-2 Receptor. ACS Pharmacol Transl Sci 2020; 3:706-719. [PMID: 32832872 DOI: 10.1021/acsptsci.0c00032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 01/10/2023]
Abstract
The hormone adrenomedullin has both physiological and pathological roles in biology. As a potent vasodilator, adrenomedullin is critically important in the regulation of blood pressure, but it also has several roles in disease, of which its actions in cancer are becoming recognized to have clinical importance. Reduced circulating adrenomedullin causes increased blood pressure but also reduces tumor progression, so drugs blocking all effects of adrenomedullin would be unacceptable clinically. However, there are two distinct receptors for adrenomedullin, each comprising the same G protein-coupled receptor (GPCR), the calcitonin receptor-like receptor (CLR), together with a different accessory protein known as a receptor activity-modifying protein (RAMP). The CLR with RAMP2 forms an adrenomedullin-1 receptor, and the CLR with RAMP3 forms an adrenomedullin-2 receptor. Recent research suggests that a selective blockade of adrenomedullin-2 receptors would be therapeutically valuable. Here we describe the design, synthesis, and characterization of potent small-molecule adrenomedullin-2 receptor antagonists with 1000-fold selectivity over the adrenomedullin-1 receptor, although retaining activity against the CGRP receptor. These molecules have clear effects on markers of pancreatic cancer progression in vitro, drug-like pharmacokinetic properties, and inhibit xenograft tumor growth and extend life in a mouse model of pancreatic cancer. Taken together, our data support the promise of a new class of anticancer therapeutics as well as improved understanding of the pharmacology of the adrenomedullin receptors and other GPCR/RAMP heteromers.
Collapse
Affiliation(s)
- Paris Avgoustou
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Ameera B A Jailani
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | | | | | - Karl R Gibson
- Sandexis Medicinal Chemistry Ltd, Sandwich, Kent CT13 9ND, U.K
| | - Paul A Glossop
- Sandexis Medicinal Chemistry Ltd, Sandwich, Kent CT13 9ND, U.K
| | - James E J Mills
- Sandexis Medicinal Chemistry Ltd, Sandwich, Kent CT13 9ND, U.K
| | | | - Paul Blaney
- Concept Life Sciences, High Peak, SK23 0PG, U.K
| | - Peter J Bungay
- Sympetrus Ltd., Bishop's Stortford, Hertfordshire CM23 3BT, U.K
| | - Ning Wang
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Alice P Shaw
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Kamilla J A Bigos
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Joseph L Holmes
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Jessica I Warrington
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Timothy M Skerry
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Joseph P A Harrity
- Department of Chemistry, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Gareth O Richards
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| |
Collapse
|
26
|
Kalisch-Smith JI, Ved N, Sparrow DB. Environmental Risk Factors for Congenital Heart Disease. Cold Spring Harb Perspect Biol 2020; 12:a037234. [PMID: 31548181 PMCID: PMC7050589 DOI: 10.1101/cshperspect.a037234] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Congenital heart disease (CHD) has many forms and a wide range of causes. Clinically, it is important to understand the causes. This allows estimation of recurrence rate, guides treatment options, and may also be used to formulate public health advice to reduce the population prevalence of CHD. The recent advent of sophisticated genetic and genomic methods has led to the identification of more than 100 genes associated with CHD. However, despite these great strides, to date only one-third of CHD cases have been shown to have a simple genetic cause. This is because CHD can also be caused by oligogenic factors, environmental factors, and/or gene-environment interaction. Although solid evidence for environmental causes of CHD have been available for almost 80 years, it is only very recently that the molecular mechanisms for these risk factors have begun to be investigated. In this review, we describe the most important environmental CHD risk factors, and what is known about how they cause CHD.
Collapse
Affiliation(s)
| | - Nikita Ved
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, Oxfordshire OX1 3PT, United Kingdom
| | - Duncan Burnaby Sparrow
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, Oxfordshire OX1 3PT, United Kingdom
| |
Collapse
|
27
|
Samak M, Hinkel R. Stem Cells in Cardiovascular Medicine: Historical Overview and Future Prospects. Cells 2019; 8:cells8121530. [PMID: 31783680 PMCID: PMC6952821 DOI: 10.3390/cells8121530] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death in the developed world, accounting for more than 30% of all deaths. In a large proportion of these patients, acute myocardial infarction is usually the first manifestation, which might further progress to heart failure. In addition, the human heart displays a low regenerative capacity, leading to a loss of cardiomyocytes and persistent tissue scaring, which entails a morbid pathologic sequela. Novel therapeutic approaches are urgently needed. Stem cells, such as induced pluripotent stem cells or embryonic stem cells, exhibit great potential for cell-replacement therapy and an excellent tool for disease modeling, as well as pharmaceutical screening of novel drugs and their cardiac side effects. This review article covers not only the origin of stem cells but tries to summarize their translational potential, as well as potential risks and clinical translation.
Collapse
Affiliation(s)
- Mostafa Samak
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
28
|
Cavalluzzi MM, Imbrici P, Gualdani R, Stefanachi A, Mangiatordi GF, Lentini G, Nicolotti O. Human ether-à-go-go-related potassium channel: exploring SAR to improve drug design. Drug Discov Today 2019; 25:344-366. [PMID: 31756511 DOI: 10.1016/j.drudis.2019.11.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/22/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
hERG is best known as a primary anti-target, the inhibition of which is responsible for serious side effects. A renewed interest in hERG as a desired target, especially in oncology, was sparked because of its role in cellular proliferation and apoptosis. In this study, we survey the most recent advances regarding hERG by focusing on SAR in the attempt to elucidate, at a molecular level, off-target and on-target actions of potential hERG binders, which are highly promiscuous and largely varying in structure. Understanding the rationale behind hERG interactions and the molecular determinants of hERG activity is a real challenge and comprehension of this is of the utmost importance to prioritize compounds in early stages of drug discovery and to minimize cardiotoxicity attrition in preclinical and clinical studies.
Collapse
Affiliation(s)
- Maria Maddalena Cavalluzzi
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari 'Aldo Moro', Via E. Orabona, 4, 70126 Bari, Italy
| | - Paola Imbrici
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari 'Aldo Moro', Via E. Orabona, 4, 70126 Bari, Italy
| | - Roberta Gualdani
- Laboratory of Cell Physiology, Institute of Neuroscience, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Angela Stefanachi
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari 'Aldo Moro', Via E. Orabona, 4, 70126 Bari, Italy
| | | | - Giovanni Lentini
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari 'Aldo Moro', Via E. Orabona, 4, 70126 Bari, Italy
| | - Orazio Nicolotti
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari 'Aldo Moro', Via E. Orabona, 4, 70126 Bari, Italy.
| |
Collapse
|
29
|
Li X, Payne DT, Ampolu B, Bland N, Brown JT, Dutton MJ, Fitton CA, Gulliver A, Hale L, Hamza D, Jones G, Lane R, Leach AG, Male L, Merisor EG, Morton MJ, Quy AS, Roberts R, Scarll R, Schulz-Utermoehl T, Stankovic T, Stevenson B, Fossey JS, Agathanggelou A. Derivatisation of parthenolide to address chemoresistant chronic lymphocytic leukaemia. MEDCHEMCOMM 2019; 10:1379-1390. [PMID: 32952998 PMCID: PMC7478165 DOI: 10.1039/c9md00297a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022]
Abstract
Parthenolide is a natural product that exhibits anti-leukaemic activity, however, its clinical use is limited by its poor bioavailability. It may be extracted from feverfew and protocols for growing, extracting and derivatising it are reported. A novel parthenolide derivative with good bioavailability and pharmacological properties was identified through a screening cascade based on in vitro anti-leukaemic activity and calculated "drug-likeness" properties, in vitro and in vivo pharmacokinetics studies and hERG liability testing. In vitro studies showed the most promising derivative to have comparable anti-leukaemic activity to DMAPT, a previously described parthenolide derivative. The newly identified compound was shown to have pro-oxidant activity and in silico molecular docking studies indicate a prodrug mode of action. A synthesis scheme is presented for the production of amine 7 used in the generation of 5f.
Collapse
Affiliation(s)
- Xingjian Li
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Daniel T Payne
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Badarinath Ampolu
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Nicholas Bland
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Jane T Brown
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Mark J Dutton
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Catherine A Fitton
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Abigail Gulliver
- Winterbourne Botanic Garden, University of Birmingham, 58 Edgbaston Park Road, Edgbaston, Birmingham, West Midlands B15 2RT, UK
| | - Lee Hale
- Winterbourne Botanic Garden, University of Birmingham, 58 Edgbaston Park Road, Edgbaston, Birmingham, West Midlands B15 2RT, UK
| | - Daniel Hamza
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Geraint Jones
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Rebecca Lane
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Andrew G Leach
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Louise Male
- X-Ray Crystallography Facility, School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK
| | - Elena G Merisor
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - Michael J Morton
- ApconiX Ltd, Alderly Park, Nether Alderly, Cheshire, SK10 4TG, UK
| | - Alex S Quy
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Ruth Roberts
- ApconiX Ltd, Alderly Park, Nether Alderly, Cheshire, SK10 4TG, UK
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK
| | - Rosanna Scarll
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | | | - Tatjana Stankovic
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Brett Stevenson
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, NG1 1GR, UK
| | - John S Fossey
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| | - Angelo Agathanggelou
- Institute for Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands B15 2TT, UK.
| |
Collapse
|
30
|
Sanson C, Schombert B, Filoche-Rommé B, Partiseti M, Bohme GA. Electrophysiological and Pharmacological Characterization of Human Inwardly Rectifying K ir2.1 Channels on an Automated Patch-Clamp Platform. Assay Drug Dev Technol 2019; 17:89-99. [PMID: 30835490 PMCID: PMC6479253 DOI: 10.1089/adt.2018.882] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inwardly rectifying IK1 potassium currents of the heart control the resting membrane potential of ventricular cardiomyocytes during diastole and contribute to their repolarization after each action potential. Mutations in the gene encoding Kir2.1 channels, which primarily conduct ventricular IK1, are associated with inheritable forms of arrhythmias and sudden cardiac death. Therefore, potential iatrogenic inhibition of Kir2.1-mediated IK1 currents is a cardiosafety concern during new drug discovery and development. Kir2.1 channels are part of the panel of cardiac ion channels currently considered for refined early compound risk assessment within the Comprehensive in vitro Proarrhythmia Assay initiative. In this study, we have validated a cell-based assay allowing functional quantification of Kir2.1 inhibitors using whole-cell recordings of Chinese hamster ovary cells stably expressing human Kir2.1 channels. We reproduced key electrophysiological and pharmacological features known for native IK1, including current enhancement by external potassium and voltage- and concentration-dependent blockade by external barium. Furthermore, the Kir inhibitors ML133, PA-6, and chloroquine, as well as the multichannel inhibitors chloroethylclonidine, chlorpromazine, SKF-96365, and the class III antiarrhythmic agent terikalant demonstrated slowly developing inhibitory activity in the low micromolar range. The robustness of this assay authorizes medium throughput screening for cardiosafety purposes and could help to enrich the currently limited Kir2.1 pharmacology.
Collapse
Affiliation(s)
- Camille Sanson
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Brigitte Schombert
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Bruno Filoche-Rommé
- 2 Integrated Drug Discovery, Medicinal Chemistry, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Michel Partiseti
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - G Andrees Bohme
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| |
Collapse
|
31
|
Shah RR, Stonier PD. Repurposing old drugs in oncology: Opportunities with clinical and regulatory challenges ahead. J Clin Pharm Ther 2018; 44:6-22. [PMID: 30218625 DOI: 10.1111/jcpt.12759] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/08/2018] [Accepted: 08/19/2018] [Indexed: 12/11/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE In order to expedite the availability of drugs to treat cancers in a cost-effective manner, repurposing of old drugs for oncological indications is gathering momentum. Revolutionary advances in pharmacology and genomics have demonstrated many old drugs to have activity at novel antioncogenic pharmacological targets. We decided to investigate whether prospective studies support the promises of nonclinical and retrospective clinical studies on repurposing three old drugs, namely metformin, valproate and astemizole. METHODS We conducted an extensive literature search through PubMed to gather representative nonclinical and retrospective clinical studies that investigated the potential repurposing of these three drugs for oncological indications. We then searched for prospective studies aimed at confirming the promises of retrospective data. RESULTS AND DISCUSSION While evidence from nonclinical and retrospective clinical studies with these drugs appears highly promising, large scale prospective studies are either lacking or have failed to substantiate this promise. We provide a brief discussion of some of the challenges in repurposing. Principal challenges and obstacles relate to heterogeneity of cancers studied without considering their molecular signatures, trials with small sample size and short duration, failure consider issues of ethnicity of study population and effective antioncogenic doses of the drug studied. WHAT IS NEW AND CONCLUSION Well-designed prospective studies demonstrating efficacy are required for repurposing old drugs for oncology indications, just as they are for new chemical entities for any indication. Early and ongoing interactions with regulatory authorities are invaluable. We outline a tentative framework for a structured approach to repurposing old drugs for novel indications in oncology.
Collapse
Affiliation(s)
- Rashmi R Shah
- Pharmaceutical Consultant, Gerrards Cross, Buckinghamshire, UK
| | - Peter D Stonier
- Department of Pharmaceutical Medicine, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College, London, UK
| |
Collapse
|
32
|
Microenvironmental Modulation of Calcium Wave Propagation Velocity in Engineered Cardiac Tissues. Cell Mol Bioeng 2018; 11:337-352. [PMID: 31719889 DOI: 10.1007/s12195-018-0522-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/09/2018] [Indexed: 10/17/2022] Open
Abstract
Introduction In the myocardium, rapid propagation of action potentials and subsequent calcium waves is critical for synchronizing the contraction of cardiac myocytes and maximizing cardiac output. In many pathological settings, diverse remodeling of the tissue microenvironment is correlated with arrhythmias and decreased cardiac output, but the precise impact of tissue remodeling on propagation is not completely understood. Our objective was to delineate how multiple features within the cardiac tissue microenvironment modulate propagation velocity. Methods To recapitulate diverse myocardial tissue microenvironments, we engineered substrates with tunable elasticity, patterning, composition, and topography using two formulations of polydimethylsiloxane (PDMS) micropatterned with fibronectin and gelatin hydrogels with flat or micromolded features. We cultured neonatal rat ventricular myocytes on these substrates and quantified cell density, tissue alignment, and cell shape. We used a fluorescent calcium indicator, high-speed microscopy, and newly-developed analysis software to record and quantify calcium wave propagation velocity (CPV). Results For all substrates, tissue alignment and cell aspect ratio were higher in aligned compared to isotropic tissues. Isotropic CPV and longitudinal CPV were similar across conditions, but transverse CPV was lower on micromolded gelatin hydrogels compared to micropatterned soft and stiff PDMS. In aligned tissues, the anisotropy ratio of CPV (longitudinal CPV/transverse CPV) was lower on micropatterned soft PDMS compared to micropatterned stiff PDMS and micromolded gelatin hydrogels. Conclusion Propagation velocity in engineered cardiac tissues is sensitive to features in the tissue microenvironment, such as alignment, matrix elasticity, and matrix topography, which may underlie arrhythmias in conditions with pathological tissue remodeling.
Collapse
|
33
|
Park E, Gintant GA, Bi D, Kozeli D, Pettit SD, Pierson JB, Skinner M, Willard J, Wisialowski T, Koerner J, Valentin JP. Can non-clinical repolarization assays predict the results of clinical thorough QT studies? Results from a research consortium. Br J Pharmacol 2018; 175:606-617. [PMID: 29181850 PMCID: PMC5786459 DOI: 10.1111/bph.14101] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/12/2017] [Accepted: 11/13/2017] [Indexed: 01/06/2023] Open
Abstract
Background and Purpose Translation of non‐clinical markers of delayed ventricular repolarization to clinical prolongation of the QT interval corrected for heart rate (QTc) (a biomarker for torsades de pointes proarrhythmia) remains an issue in drug discovery and regulatory evaluations. We retrospectively analysed 150 drug applications in a US Food and Drug Administration database to determine the utility of established non‐clinical in vitro IKr current human ether‐à‐go‐go‐related gene (hERG), action potential duration (APD) and in vivo (QTc) repolarization assays to detect and predict clinical QTc prolongation. Experimental Approach The predictive performance of three non‐clinical assays was compared with clinical thorough QT study outcomes based on free clinical plasma drug concentrations using sensitivity and specificity, receiver operating characteristic (ROC) curves, positive (PPVs) and negative predictive values (NPVs) and likelihood ratios (LRs). Key Results Non‐clinical assays demonstrated robust specificity (high true negative rate) but poor sensitivity (low true positive rate) for clinical QTc prolongation at low‐intermediate (1×–30×) clinical exposure multiples. The QTc assay provided the most robust PPVs and NPVs (ability to predict clinical QTc prolongation). ROC curves (overall test accuracy) and LRs (ability to influence post‐test probabilities) demonstrated overall marginal performance for hERG and QTc assays (best at 30× exposures), while the APD assay demonstrated minimal value. Conclusions and Implications The predictive value of hERG, APD and QTc assays varies, with drug concentrations strongly affecting translational performance. While useful in guiding preclinical candidates without clinical QT prolongation, hERG and QTc repolarization assays provide greater value compared with the APD assay.
Collapse
Affiliation(s)
- Eunjung Park
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | - Gary A Gintant
- Department of Integrative Pharmacology, AbbVie, North Chicago, IL, USA
| | - Daoqin Bi
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | - Devi Kozeli
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | | | | | - Matthew Skinner
- Drug Safety and Metabolism, AstraZeneca, Macclesfield, Cheshire, UK
| | - James Willard
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | | | - John Koerner
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | | |
Collapse
|
34
|
Sethi TK, Basdag B, Bhatia N, Moslehi J, Reddy NM. Beyond Anthracyclines: Preemptive Management of Cardiovascular Toxicity in the Era of Targeted Agents for Hematologic Malignancies. Curr Hematol Malig Rep 2017; 12:257-267. [PMID: 28233150 DOI: 10.1007/s11899-017-0369-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advances in drug discovery have led to the use of effective targeted agents in the treatment of hematologic malignancies. Drugs such as proteasome inhibitors in multiple myeloma and tyrosine kinase inhibitors in chronic myeloid leukemia and non-Hodgkin lymphoma have changed the face of treatment of hematologic malignancies. There are several reports of cardiovascular adverse events related to these newer agents. Both "on-target" and "off-target" effects of these agents can cause organ-specific toxicity. The need for long-term administration for most of these agents requires continued monitoring of toxicity. Moreover, the patient population is older, often over 50 years of age, making them more susceptible to cardiovascular side effects. Additional factors such as prior exposure to anthracyclines often add to this toxicity. In light of their success and widespread use, it is important to recognize and manage the unique side effect profile of targeted agents used in hematologic malignancies. In this article, we review the current data for the incidence of cardiovascular side effects of targeted agents in hematologic malignancies and discuss a preemptive approach towards managing these toxicities.
Collapse
Affiliation(s)
- Tarsheen K Sethi
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, 3927 The Vanderbilt Clinic, Nashville, TN, USA
| | - Basak Basdag
- Division of Internal Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nirmanmoh Bhatia
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid Moslehi
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nishitha M Reddy
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, 3927 The Vanderbilt Clinic, Nashville, TN, USA.
| |
Collapse
|
35
|
Kratz JM, Grienke U, Scheel O, Mann SA, Rollinger JM. Natural products modulating the hERG channel: heartaches and hope. Nat Prod Rep 2017; 34:957-980. [PMID: 28497823 PMCID: PMC5708533 DOI: 10.1039/c7np00014f] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This review covers natural products modulating the hERG potassium channel. Risk assessment strategies, structural features of blockers, and the duality target/antitarget are discussed.
Covering: 1996–December 2016 The human Ether-à-go-go Related Gene (hERG) channel is a voltage-gated potassium channel playing an essential role in the normal electrical activity in the heart. It is involved in the repolarization and termination of action potentials in excitable cardiac cells. Mutations in the hERG gene and hERG channel blockage by small molecules are associated with increased risk of fatal arrhythmias. Several drugs have been withdrawn from the market due to hERG channel-related cardiotoxicity. Moreover, as a result of its notorious ligand promiscuity, this ion channel has emerged as an important antitarget in early drug discovery and development. Surprisingly, the hERG channel blocking profile of natural compounds present in frequently consumed botanicals (i.e. dietary supplements, spices, and herbal medicinal products) is not routinely assessed. This comprehensive review will address these issues and provide a critical compilation of hERG channel data for isolated natural products and extracts over the past two decades (1996–2016). In addition, the review will provide (i) a solid basis for the molecular understanding of the physiological functions of the hERG channel, (ii) the translational potential of in vitro/in vivo results to cardiotoxicity in humans, (iii) approaches for the identification of hERG channel blockers from natural sources, (iv) future perspectives for cardiac safety guidelines and their applications within phytopharmaceuticals and dietary supplements, and (v) novel applications of hERG channel modulation (e.g. as a drug target).
Collapse
Affiliation(s)
- Jadel M Kratz
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria.
| | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- Yoshinori Yoshida
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| | - Shinya Yamanaka
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| |
Collapse
|
37
|
Hazell L, Raschi E, De Ponti F, Thomas SHL, Salvo F, Ahlberg Helgee E, Boyer S, Sturkenboom M, Shakir S. Evidence for the hERG Liability of Antihistamines, Antipsychotics, and Anti-Infective Agents: A Systematic Literature Review From the ARITMO Project. J Clin Pharmacol 2017; 57:558-572. [PMID: 28019033 DOI: 10.1002/jcph.838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/08/2016] [Indexed: 04/21/2025]
Abstract
A systematic review was performed to categorize the hERG (human ether-a-go-go-related gene) liability of antihistamines, antipsychotics, and anti-infectives and to compare it with current clinical risk of torsade de pointes (TdP). Eligible studies were hERG assays reporting half-minimal inhibitory concentrations (IC50). A "hERG safety margin" was calculated from the IC50 divided by the peak human plasma concentration (free Cmax ). A margin below 30 defined hERG liability. Each drug was assigned an "uncertainty score" based on volume, consistency, precision, and internal and external validity of evidence. The hERG liability was compared to existing knowledge on TdP risk (www.credibledrugs.org). Of 1828 studies, 82 were eligible, allowing calculation of safety margins for 61 drugs. Thirty-one drugs (51%) had evidence of hERG liability including 6 with no previous mention of TdP risk (eg, desloratadine, lopinavir). Conversely, 16 drugs (26%) had no evidence of hERG liability including 6 with known, or at least conditional or possible, TdP risk (eg, chlorpromazine, sulpiride). The main sources of uncertainty were the validity of the experimental conditions used (antihistamines and antipsychotics) and nonuse of reference compounds (anti-infectives). In summary, hERG liability was categorized for 3 widely used drug classes, incorporating a qualitative assessment of the strength of available evidence. Some concordance with TdP risk was observed, although several drugs had hERG liability without evidence of clinical risk and vice versa. This may be due to gaps in clinical evidence, limitations of hERG/Cmax data, or other patient/drug-specific factors that contribute to real-life TdP risk.
Collapse
Affiliation(s)
- Lorna Hazell
- Drug Safety Research Unit, Southampton, United Kingdom
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio De Ponti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simon H L Thomas
- Institute of Cellular Medicine, Faculty of Medicine, Newcastle University, Newcastle, United Kingdom
| | - Francesco Salvo
- University of Bordeaux U657, CHU de Bordeaux, Bordeaux, France
| | - Ernst Ahlberg Helgee
- Drug Safety and Metabolism, AstraZeneca Innovative Medicines and Early Development, Mölndal, Sweden
| | - Scott Boyer
- Computational Toxicology, Swedish Toxicology Sciences Research Center, Södertälje, Sweden
| | | | - Saad Shakir
- Drug Safety Research Unit, Southampton, United Kingdom
| |
Collapse
|
38
|
Van Vleet TR, Liu H, Lee A, Blomme EAG. Acyl glucuronide metabolites: Implications for drug safety assessment. Toxicol Lett 2017; 272:1-7. [PMID: 28286018 DOI: 10.1016/j.toxlet.2017.03.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/17/2017] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
Acyl glucuronides are important metabolites of compounds with carboxylic acid moieties and have unique properties that distinguish them from other phase 2 metabolites. In particular, in addition to being often unstable, acyl glucuronide metabolites can be chemically reactive leading to covalent binding with macromolecules and toxicity. While there is circumstantial evidence that drugs forming acyl glucuronide metabolites can be associated with rare, but severe idiosyncratic toxic reactions, many widely prescribed drugs with good safety records are also metabolized through acyl glucuronidation. Therefore, there is a need to understand the various factors that can affect the safety of acyl glucuronide-producing drugs including the rate of acyl glucuronide formation, the relative reactivity of the acyl glucuronide metabolite formed, the rate of elimination, potential proteins being targeted, and the rate of aglucuronidation. In this review, these factors are discussed and various approaches to de-risk the safety liabilities of acyl glucuronide metabolites are evaluated.
Collapse
Affiliation(s)
- Terry R Van Vleet
- Abbvie, Development Sciences, Department of Preclinical Safety, United States.
| | - Hong Liu
- Abbvie, Development Sciences, Biomeasure and Metabolism, United States
| | - Anthony Lee
- Abbvie, Development Sciences, Biomeasure and Metabolism, United States
| | - Eric A G Blomme
- Abbvie, Development Sciences, Department of Preclinical Safety, United States
| |
Collapse
|
39
|
Interaction among hERG channel blockers is a potential mechanism of death in caffeine overdose. Eur J Pharmacol 2017; 800:23-33. [PMID: 28216052 DOI: 10.1016/j.ejphar.2017.02.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 11/22/2022]
Abstract
Caffeine overdose death is due to cardiac arrest, but its mechanism has not been explored in detail. In this study, our data showed that caffeine significantly prolonged the heart rate-corrected QT interval (QTc) of rabbits in vivo (P<0.05; n=7). Caffeine was also found to be a hERG channel blocker with an IC50 of 5.04mM (n=5). Although these two findings likely link caffeine overdose death with hERG channel blockade, the amount of caffeine consumption needed to reach the IC50 is very high. Further study demonstrated that addition another hERG blocker could lower the consumption of caffeine significantly, no matter whether two hERG blockers share the same binding sites. Our data does not rule out other possibility, however, it suggests that there is a potential causal relationship between caffeine overdose death with hERG channel and the interaction among these hERG blockers.
Collapse
|
40
|
Houtmann S, Schombert B, Sanson C, Partiseti M, Bohme GA. Automated Patch-Clamp Methods for the hERG Cardiac Potassium Channel. Methods Mol Biol 2017; 1641:187-199. [PMID: 28748465 DOI: 10.1007/978-1-4939-7172-5_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The human Ether-a-go-go Related Gene (hERG) product has been identified as a central ion channel underlying both familial forms of elongated QT interval on the electrocardiogram and drug-induced elongation of the same QT segment. Indeed, reduced function of this potassium channel involved in the repolarization of the cardiac action potential can produce a type of life-threatening cardiac ventricular arrhythmias called Torsades de Pointes (TdP). Therefore, hERG inhibitory activity of newly synthetized molecules is a relevant structure-activity metric for compound prioritization and optimization in medicinal chemistry phases of drug discovery. Electrophysiology remains the gold standard for the functional assessment of ion channel pharmacology. The recent years have witnessed automatization and parallelization of the manual patch-clamp technique, allowing higher throughput screening on recombinant hERG channels. However, the multi-well plate format of automatized patch-clamp does not allow visual detection of potential micro-precipitation of poorly soluble compounds. In this chapter we describe bench procedures for the culture and preparation of hERG-expressing CHO cells for recording on an automated patch-clamp workstation. We also show that the sensitivity of the assay can be improved by adding a surfactant to the extracellular medium.
Collapse
Affiliation(s)
- Sylvie Houtmann
- Integrated Drug Discovery, Sanofi R&D, 13 Quai Jules Guesde, F-94403, Vitry-sur-Seine, France
| | - Brigitte Schombert
- Integrated Drug Discovery, Sanofi R&D, 13 Quai Jules Guesde, F-94403, Vitry-sur-Seine, France
| | - Camille Sanson
- Integrated Drug Discovery, Sanofi R&D, 13 Quai Jules Guesde, F-94403, Vitry-sur-Seine, France
| | - Michel Partiseti
- Integrated Drug Discovery, Sanofi R&D, 13 Quai Jules Guesde, F-94403, Vitry-sur-Seine, France
| | - G Andrees Bohme
- Integrated Drug Discovery, Sanofi R&D, 13 Quai Jules Guesde, F-94403, Vitry-sur-Seine, France.
| |
Collapse
|
41
|
Kang J, Luo Y, Searles M, Rampe D. Observations on conducting whole-cell patch clamping of the hERG cardiac K + channel in pure human serum. J Appl Toxicol 2016; 37:445-453. [PMID: 27553911 DOI: 10.1002/jat.3377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/06/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023]
Abstract
Inhibition of the human ether-a-go-go-related gene (hERG) K+ channel by drugs leads to QT prolongation on the electrocardiogram and can result in serious cardiac arrhythmia. For this reason, screening of drugs on hERG is mandatory during the drug development process. Patch clamp electrophysiology in a defined physiological saline solution (PSS) represents the standard method for assaying drug effects on the channel. To make the assay more translatable to clinical studies, we have conducted whole-cell patch clamping of hERG using pure human serum as the extracellular medium. Pure human serum had little effect on the hERG channel waveform or the current-voltage relationship when compared to PSS. hERG current recordings were highly stable in serum at room temperature, but prolonged recordings at the physiological temperature required prior heat inactivation of the serum. Compared to PSS, the IC50 values, conducted at room temperature, of the classic hERG blocking drugs cisapride, moxifloxacin, and terfenadine were shifted to the right by an extent predicted by their known plasma protein binding, but we did not detect any differences in IC50 s between male and female serum. Total plasma levels of these drugs associated with clinical QT prolongation corresponded to small (<15%) inhibition of hERG current in pure serum suggesting that minor inhibition of the channel leads to observable pharmacodynamic effects. Conducting whole-cell patch clamping of hERG in human serum has the potential to make the assay more translatable to clinical studies and improve its predictive value for safety testing. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jiesheng Kang
- Departments of Disposition, Safety, and Animal Research Sanofi, Inc., Waltham, Massachusetts, USA
| | - Yongyi Luo
- Departments of Disposition, Safety, and Animal Research Sanofi, Inc., Waltham, Massachusetts, USA
| | - Michelle Searles
- Departments of Disposition, Safety, and Animal Research Sanofi, Inc., Framingham, Massachusetts, USA
| | - David Rampe
- Departments of Disposition, Safety, and Animal Research Sanofi, Inc., Bridgewater, New Jersey, USA
| |
Collapse
|
42
|
Proarrhythmic mechanisms of the common anti-diarrheal medication loperamide: revelations from the opioid abuse epidemic. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1133-7. [DOI: 10.1007/s00210-016-1286-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/09/2016] [Indexed: 11/26/2022]
|
43
|
Pugsley MK, Authier S, Hayes ES, Hamlin RL, Accardi MV, Curtis MJ. Recalibration of nonclinical safety pharmacology assessment to anticipate evolving regulatory expectations. J Pharmacol Toxicol Methods 2016; 81:1-8. [PMID: 27343819 DOI: 10.1016/j.vascn.2016.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Safety pharmacology (SP) has evolved in terms of architecture and content since the inception of the SP Society (SPS). SP was initially focused on the issue of drug-induced QT prolongation, but has now become a broad spectrum discipline with expanding expectations for evaluation of drug adverse effect liability in all organ systems, not merely the narrow consideration of torsades de pointes (TdP) liability testing. An important part of the evolution of SP has been the elaboration of architecture for interrogation of non-clinical models in terms of model development, model validation and model implementation. While SP has been defined by mandatory cardiovascular, central nervous system (CNS) and respiratory system studies ever since the core battery was elaborated, it also involves evaluation of drug effects on other physiological systems. The current state of SP evolution is the incorporation of emerging new technologies in a wide range of non-clinical drug safety testing models. This will refine the SP process, while potentially expanding the core battery. The continued refinement of automated technologies (e.g., automated patch clamp systems) is enhancing the scope for detection of adverse effect liability (i.e., for more than just IKr blockade), while introducing a potential for speed and accuracy in cardiovascular and CNS SP by providing rapid, high throughput ion channel screening methods for implementation in early drug development. A variety of CNS liability assays, which exploit isolated brain tissue, and in vitro electrophysiological techniques, have provided an additional level of complimentary preclinical safety screens aimed at establishing the seizurogenic potential and risk for memory dysfunction of new chemical entities (NCEs). As with previous editorials that preface the annual themed issue on SP methods published in the Journal of Pharmacological and Toxicological Methods (JPTM), we highlight here the content derived from the most recent (2015) SPS meeting held in Prague, Czech Republic. This issue of JPTM continues the tradition of providing a publication summary of articles primarily presented at the SPS meeting with direct bearing on the discipline of SP. Novel method development and refinement in all areas of the discipline are reflected in the content.
Collapse
Affiliation(s)
- Michael K Pugsley
- Department of Toxicology & PKDM, Purdue Pharma LP., 6 Cedar Brook Dr., Cranbury, NJ 08512, U.S.A..
| | - Simon Authier
- CiToxLAB Research Inc., 445 Armand Frappier, Laval, QC H7V 4B3, Canada
| | | | | | - Michael V Accardi
- CiToxLAB Research Inc., 445 Armand Frappier, Laval, QC H7V 4B3, Canada
| | - Michael J Curtis
- Cardiovascular Division, Rayne Institute, St Thomas' Hospital, London, SE17EH, UK
| |
Collapse
|
44
|
Colatsky T, Fermini B, Gintant G, Pierson JB, Sager P, Sekino Y, Strauss DG, Stockbridge N. The Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative - Update on progress. J Pharmacol Toxicol Methods 2016; 81:15-20. [PMID: 27282641 DOI: 10.1016/j.vascn.2016.06.002] [Citation(s) in RCA: 303] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/01/2016] [Accepted: 06/04/2016] [Indexed: 11/17/2022]
Abstract
The implementation of the ICH S7B and E14 guidelines has been successful in preventing the introduction of potentially torsadogenic drugs to the market, but it has also unduly constrained drug development by focusing on hERG block and QT prolongation as essential determinants of proarrhythmia risk. The Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative was established to develop a new paradigm for assessing proarrhythmic risk, building on the emergence of new technologies and an expanded understanding of torsadogenic mechanisms beyond hERG block. An international multi-disciplinary team of regulatory, industry and academic scientists are working together to develop and validate a set of predominantly nonclinical assays and methods that eliminate the need for the thorough-QT study and enable a more precise prediction of clinical proarrhythmia risk. The CiPA effort is led by a Steering Team that provides guidance, expertise and oversight to the various working groups and includes partners from US FDA, HESI, CSRC, SPS, EMA, Health Canada, Japan NIHS, and PMDA. The working groups address the three pillars of CiPA that evaluate drug effects on: 1) human ventricular ionic channel currents in heterologous expression systems, 2) in silico integration of cellular electrophysiologic effects based on ionic current effects, the ion channel effects, and 3) fully integrated biological systems (stem-cell-derived cardiac myocytes and the human ECG). This article provides an update on the progress of the initiative towards its target date of December 2017 for completing validation.
Collapse
Affiliation(s)
- Thomas Colatsky
- US FDA, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States.
| | - Bernard Fermini
- Pfizer, Eastern Point Road MS 4083, Groton, CT 06340, United States.
| | - Gary Gintant
- AbbVie, R46R AP-9, 1 North Waukegan Rd, North Chicago, IL 60064-6118, United States.
| | - Jennifer B Pierson
- ILSI-Health and Environmental Sciences Institute, 1156 15th Street NW, Suite 200, Washington, DC 20005, United States.
| | - Philip Sager
- Stanford University, 719 Carolina St., San Francisco, CA 94107, United States.
| | - Yuko Sekino
- NIHS Japan, Kamiyoga 1-18-1, Setagaya-ku, Tokyo 158-8501, Japan.
| | - David G Strauss
- US FDA, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States.
| | - Norman Stockbridge
- US FDA, 10903 New Hampshire Ave, Silver Spring, MD 20993, United States.
| |
Collapse
|
45
|
Spence S, Deurinck M, Ju H, Traebert M, McLean L, Marlowe J, Emotte C, Tritto E, Tseng M, Shultz M, Friedrichs GS. Histone Deacetylase Inhibitors Prolong Cardiac Repolarization through Transcriptional Mechanisms. Toxicol Sci 2016; 153:39-54. [PMID: 27255383 DOI: 10.1093/toxsci/kfw104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are an emerging class of anticancer agents that modify gene expression by altering the acetylation status of lysine residues of histone proteins, thereby inducing transcription, cell cycle arrest, differentiation, and cell death or apoptosis of cancer cells. In the clinical setting, treatment with HDAC inhibitors has been associated with delayed cardiac repolarization and in rare instances a lethal ventricular tachyarrhythmia known as torsades de pointes. The mechanism(s) of HDAC inhibitor-induced effects on cardiac repolarization is unknown. We demonstrate that administration of structurally diverse HDAC inhibitors to dogs causes delayed but persistent increases in the heart rate corrected QT interval (QTc), an in vivo measure of cardiac repolarization, at timepoints far removed from the Tmax for parent drug and metabolites. Transcriptional profiling of ventricular myocardium from dogs treated with various HDAC inhibitors demonstrated effects on genes involved in protein trafficking, scaffolding and insertion of various ion channels into the cell membrane as well as genes for specific ion channel subunits involved in cardiac repolarization. Extensive in vitro ion channel profiling of various structural classes of HDAC inhibitors (and their major metabolites) by binding and acute patch clamp assays failed to show any consistent correlations with direct ion channel blockade. Drug-induced rescue of an intracellular trafficking-deficient mutant potassium ion channel, hERG (G601S), and decreased maturation (glycosylation) of wild-type hERG expressed by CHO cells in vitro correlated with prolongation of QTc intervals observed in vivo The results suggest that HDAC inhibitor-induced prolongation of cardiac repolarization may be mediated in part by transcriptional changes of genes required for ion channel trafficking and localization to the sarcolemma. These data have broad implications for the development of these drug classes and suggest that the optimal time to assess potentially transcriptionally mediated physiologic effects will be delayed relative to an epigenetic drug's Tmax/Cmax.
Collapse
Affiliation(s)
- Stan Spence
- *Preclinical Safety, Novartis Institutes of Biomedical Research, 100 Technology Square, Cambridge, MA 02139
| | - Mark Deurinck
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - Haisong Ju
- Preclinical Safety, Novartis Institutes of Biomedical Research, One Health Plaza, East Hanover, NJ 07936-1080
| | - Martin Traebert
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - LeeAnne McLean
- Oncology Global Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ 07936-1080
| | - Jennifer Marlowe
- *Preclinical Safety, Novartis Institutes of Biomedical Research, 100 Technology Square, Cambridge, MA 02139
| | - Corinne Emotte
- Drug Metabolism and Pharmacokinetics, Novartis Pharmaceuticals Corporation, Fabrikstrasse 14-3.02.08, Basel, CH
| | - Elaine Tritto
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - Min Tseng
- Development Sciences, Safety Assessment, Investigative Toxicology, Genentech Inc, 550 Grandview Drive, South San Francisco, CA 94080
| | - Michael Shultz
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research Inc, 250 Massachusetts Avenue, Cambridge, MA 02139
| | - Gregory S Friedrichs
- Preclinical Safety, Novartis Institutes of Biomedical Research, One Health Plaza, East Hanover, NJ 07936-1080
| |
Collapse
|
46
|
|
47
|
Dambach DM, Misner D, Brock M, Fullerton A, Proctor W, Maher J, Lee D, Ford K, Diaz D. Safety Lead Optimization and Candidate Identification: Integrating New Technologies into Decision-Making. Chem Res Toxicol 2015; 29:452-72. [DOI: 10.1021/acs.chemrestox.5b00396] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Donna M. Dambach
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dinah Misner
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Aaron Fullerton
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - William Proctor
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Jonathan Maher
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dong Lee
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Kevin Ford
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dolores Diaz
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| |
Collapse
|
48
|
Abstract
Attrition due to nonclinical safety represents a major issue for the productivity of pharmaceutical research and development (R&D) organizations, especially during the compound optimization stages of drug discovery and the early stages of clinical development. Focusing on decreasing nonclinical safety-related attrition is not a new concept, and various approaches have been experimented with over the last two decades. Front-loading testing funnels in Discovery with in vitro toxicity assays designed to rapidly identify unfavorable molecules was the approach adopted by most pharmaceutical R&D organizations a few years ago. However, this approach has also a non-negligible opportunity cost. Hence, significant refinements to the "fail early, fail often" paradigm have been proposed recently to reflect the complexity of accurately categorizing compounds with early data points without taking into account other important contextual aspects, in particular efficacious systemic and tissue exposures. This review provides an overview of toxicology approaches and models that can be used in pharmaceutical Discovery at the series/lead identification and lead optimization stages to guide and inform chemistry efforts, as well as a personal view on how to best use them to meet nonclinical safety-related attrition objectives consistent with a sustainable pharmaceutical R&D model. The scope of this review is limited to small molecules, as large molecules are associated with challenges that are quite different. Finally, a perspective on how several emerging technologies may impact toxicity evaluation is also provided.
Collapse
Affiliation(s)
- Eric A G Blomme
- Global Preclinical Safety, AbbVie Inc. , 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Yvonne Will
- Drug Safety Research and Development, Pfizer , Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
49
|
Obejero-Paz CA, Bruening-Wright A, Kramer J, Hawryluk P, Tatalovic M, Dittrich HC, Brown AM. Quantitative Profiling of the Effects of Vanoxerine on Human Cardiac Ion Channels and its Application to Cardiac Risk. Sci Rep 2015; 5:17623. [PMID: 26616666 PMCID: PMC4663487 DOI: 10.1038/srep17623] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/03/2015] [Indexed: 12/19/2022] Open
Abstract
Vanoxerine has been in clinical trials for Parkinsonism, depression and cocaine addiction but lacked efficacy. Although a potent blocker of hERG, it produced no serious adverse events. We attributed the unexpected result to offsetting Multiple Ion Channel Effects (MICE). Vanoxerine’s effects were strongly frequency-dependent and we repositioned it for treatment of atrial fibrillation and flutter. Vanoxerine terminated AF/AFL in an animal model and a dose-ranging clinical trial. Reversion to normal rhythm was associated with QT prolongation yet absent proarrhythmia markers for Torsade de Pointes (TdP). To understand the QT/TdP discordance, we used quantitative profiling and compared vanoxerine with dofetilide, a selective hERG-blocking torsadogen used for intractable AF, verapamil, a non-torsadogenic MICE comparator and bepridil, a torsadogenic MICE comparator. At clinically relevant concentrations, verapamil blocked hCav1.2 and hERG, as did vanoxerine and bepridil both of which also blocked hNav1.5. In acute experiments and simulations, dofetilide produced early after depolarizations (EADs) and arrhythmias, whereas verapamil, vanoxerine and bepridil produced no proarrhythmia markers. Of the MICE drugs only bepridil inhibited hERG trafficking following overnight exposure. The results are consistent with the emphasis on MICE of the CiPA assay. Additionally we propose that trafficking inhibition of hERG be added to CiPA.
Collapse
Affiliation(s)
- Carlos A Obejero-Paz
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Andrew Bruening-Wright
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - James Kramer
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Peter Hawryluk
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Milos Tatalovic
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Howard C Dittrich
- Laguna Pharmaceuticals, 4225 Executive Square, Suite 960, La Jolla, CA 92037, USA
| | - Arthur M Brown
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA.,Laguna Pharmaceuticals, 4225 Executive Square, Suite 960, La Jolla, CA 92037, USA
| |
Collapse
|
50
|
Rampe D, Gopalakrishnan M. On Becoming a Pharmacologist: Channeling David Triggle. Biochem Pharmacol 2015; 98:292-8. [DOI: 10.1016/j.bcp.2015.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 01/21/2023]
|