1
|
Yu MS, Lee J, Lee Y, Cho D, Oh KS, Jang J, Nong NT, Lee HM, Na D. hERGBoost: A gradient boosting model for quantitative IC 50 prediction of hERG channel blockers. Comput Biol Med 2025; 184:109416. [PMID: 39550914 DOI: 10.1016/j.compbiomed.2024.109416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024]
Abstract
The human ether-a-go-go-related gene (hERG) potassium channel is pivotal in drug discovery due to its susceptibility to blockage by drug candidate molecules, which can cause severe cardiotoxic effects. Consequently, identifying and excluding potential hERG channel blockers at the earliest stages of drug development is crucial. Most traditional machine learning models predict a molecule's cardiotoxicity or non-cardiotoxicity typically at 10 μM, which doesn't account for compounds with low IC50 values that are non-toxic at therapeutic levels due to their high effectiveness at lower concentrations. To address the need for more precise, quantitative predictions, we developed hERGBoost, a cutting-edge machine learning model employing a gradient-boosting algorithm. This model demonstrates superior accuracy in predicting the IC50 of drug candidates. Trained on a specially curated dataset for this study, hERGBoost not only exhibited excellent performance in external validation, achieving an R2 score of 0.394 and a low root mean square error of 0.616 but also significantly outstripped previous models in both qualitative and quantitative assessments. Representing a notable leap forward in the prediction of hERG channel blockers, the hERGBoost model and its datasets are freely available to the drug discovery community on our web server at. http://ssbio.cau.ac.kr/software/hergboost This resource promises to be invaluable in advancing safer pharmaceutical development.
Collapse
Affiliation(s)
- Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jingyu Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Yunhyeok Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Daeahn Cho
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kwang-Seok Oh
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, Daejeon, 34129, Republic of Korea
| | - Jidon Jang
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Nuong Thi Nong
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Lu HR, Damiano BP, Kreir M, Rohrbacher J, van der Linde H, Saidov T, Teisman A, Gallacher DJ. The Potential Mechanisms behind Loperamide-Induced Cardiac Arrhythmias Associated with Human Abuse and Extreme Overdose. Biomolecules 2023; 13:1355. [PMID: 37759755 PMCID: PMC10527387 DOI: 10.3390/biom13091355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Loperamide has been a safe and effective treatment for diarrhea for many years. However, many cases of cardiotoxicity with intentional abuse of loperamide ingestion have recently been reported. We evaluated loperamide in in vitro and in vivo cardiac safety models to understand the mechanisms for this cardiotoxicity. Loperamide slowed conduction (QRS-duration) starting at 0.3 µM [~1200-fold (×) its human Free Therapeutic Plasma Concentration; FTPC] and reduced the QT-interval and caused cardiac arrhythmias starting at 3 µM (~12,000× FTPC) in an isolated rabbit ventricular-wedge model. Loperamide also slowed conduction and elicited Type II/III A-V block in anesthetized guinea pigs at overdose exposures of 879× and 3802× FTPC. In ion-channel studies, loperamide inhibited hERG (IKr), INa, and ICa currents with IC50 values of 0.390 µM, 0.526 µM, and 4.091 µM, respectively (i.e., >1560× FTPC). Additionally, in silico trials in human ventricular action potential models based on these IC50s confirmed that loperamide has large safety margins at therapeutic exposures (≤600× FTPC) and confirmed repolarization abnormalities in the case of extreme doses of loperamide. The studies confirmed the large safety margin for the therapeutic use of loperamide but revealed that at the extreme exposure levels observed in human overdose, loperamide can cause a combination of conduction slowing and alterations in repolarization time, resulting in cardiac proarrhythmia. Loperamide's inhibition of the INa channel and hERG-mediated IKr are the most likely basis for this cardiac electrophysiological toxicity at overdose exposures. The cardiac toxic effects of loperamide at the overdoses could be aggravated by co-medication with other drug(s) causing ion channel inhibition.
Collapse
Affiliation(s)
- Hua Rong Lu
- Global Safety Pharmacology, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium; (B.P.D.); (J.R.); (H.v.d.L.); (T.S.); (A.T.); (D.J.G.)
| | | | - Mohamed Kreir
- Global Safety Pharmacology, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium; (B.P.D.); (J.R.); (H.v.d.L.); (T.S.); (A.T.); (D.J.G.)
| | | | | | | | | | | |
Collapse
|
3
|
El Harchi A, Hancox JC. hERG agonists pose challenges to web-based machine learning methods for prediction of drug-hERG channel interaction. J Pharmacol Toxicol Methods 2023; 123:107293. [PMID: 37468081 DOI: 10.1016/j.vascn.2023.107293] [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: 02/07/2023] [Revised: 05/23/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Pharmacological blockade of the IKr channel (hERG) by diverse drugs in clinical use is associated with the Long QT Syndrome that can lead to life threatening arrhythmia. Various computational tools including machine learning models (MLM) for the prediction of hERG inhibition have been developed to facilitate the throughput screening of drugs in development and optimise thus the prediction of hERG liabilities. The use of MLM relies on large libraries of training compounds for the quantitative structure-activity relationship (QSAR) modelling of hERG inhibition. The focus on inhibition omits potential effects of hERG channel agonist molecules and their associated QT shortening risk. It is instructive, therefore, to consider how known hERG agonists are handled by MLM. Here, two highly developed online computational tools for the prediction of hERG liability, Pred-hERG and HergSPred were probed for their ability to detect hERG activator drug molecules as hERG interactors. In total, 73 hERG blockers were tested with both computational tools giving overall good predictions for hERG blockers with reported IC50s below Pred-hERG and HergSPred cut-off threshold for hERG inhibition. However, for compounds with reported IC50s above this threshold such as disopyramide or sotalol discrepancies were observed. HergSPred identified all 20 hERG agonists selected as interacting with the hERG channel. Further studies are warranted to improve online MLM prediction of hERG related cardiotoxicity, by explicitly taking into account channel agonism as well as inhibition.
Collapse
Affiliation(s)
- Aziza El Harchi
- School of Physiology and Pharmacology and Neuroscience, Biomedical Sciences Building, The University of Bristol, University Walk, Bristol BS8 1TD, UK.
| | - Jules C Hancox
- School of Physiology and Pharmacology and Neuroscience, Biomedical Sciences Building, The University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
4
|
Haq KT, Cooper BL, Berk F, Posnack NG. The effect of sex and age on ex vivo cardiac electrophysiology: insight from a guinea pig model. Am J Physiol Heart Circ Physiol 2023; 324:H141-H154. [PMID: 36487188 PMCID: PMC9829463 DOI: 10.1152/ajpheart.00497.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Highlighting the importance of sex as a biological variable, we recently reported sex differences in guinea pig in vivo electrocardiogram (ECG) measurements. However, substantial inconsistencies exist in this animal model, with conflicting reports of sex-specific differences in cardiac electrophysiology observed in vivo and in vitro. Herein, we evaluated whether sexual dimorphism persists in ex vivo preparations, using an isolated intact heart preparation. Pseudo-ECG recordings were collected in conjunction with dual optical mapping of transmembrane voltage and intracellular calcium from Langendorff-perfused hearts. In contrast to our in vivo results, we did not observe sex-specific differences in ECG parameters collected from isolated hearts. Furthermore, we observed significant age-specific differences in action potential duration (APD) and Ca2+ transient duration (CaD) during both normal sinus rhythm (NSR) and in response to dynamic pacing but only a modest sex-specific difference in CaD30. Similarly, the alternans fluctuation coefficient, conduction velocity during sinus rhythm or in response to pacing, and electrophysiology parameters (atrioventricular nodal effective refractory period, Wenckebach cycle length) were comparable between males and females. Results of our study suggest that the observed sex-specific differences in in vivo ECG parameters from guinea pigs are diminished in ex vivo isolated heart preparations, although age-specific patterns are prevalent. To assess sex as a biological variable in cardiac electrophysiology, a comprehensive approach may be necessary using both in vitro measurements from cardiomyocyte or intact heart preparations with secondary follow-up in vivo studies.NEW & NOTEWORTHY We evaluated whether the guinea pig heart has intrinsic sex-specific differences in cardiac electrophysiology. Although we observed sex-specific differences in in vivo ECGs, these differences did not persist ex vivo. Using a whole heart model, we observed similar APD, CaD, conduction velocity, and alternans susceptibility in males and females. We conclude that sex-specific differences in guinea pig cardiac electrophysiology are likely influenced by the in vivo environment and less dependent on the intrinsic electrical properties of the heart.
Collapse
Affiliation(s)
- Kazi T Haq
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia
| | - Blake L Cooper
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
| | - Fiona Berk
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
- Department of Pediatrics, The George Washington University, Washington, District of Columbia
| |
Collapse
|
5
|
Skinner M, Hale E, Ceuppens P, Pollard C. Differentiating multichannel block on the guinea pig ECG: Use of T peak-T end and J-T peak. J Pharmacol Toxicol Methods 2021; 111:107085. [PMID: 34182121 DOI: 10.1016/j.vascn.2021.107085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The anaesthetised guinea pig is a well characterised assay for early assessment of drug effects on ventricular repolarisation and risk of Torsade de Pointes (TdP). We assessed whether a selective hERG blocker with known TdP risk could be differentiated from lower risk, balanced ion channel blockers in the guinea pig, using corrected QT (QTc) interval alongside novel electrocardiogram (ECG) biomarkers J-Tpeakc and Tpeak-Tend. Effects were compared with previous clinical investigations at similar plasma concentrations and with another index of TdP risk, the electromechanical window (EMW). METHODS Twenty-two Dunkin Hartley guinea pigs anaesthetised with sodium pentobarbitone were instrumented for haemodynamic measurement and ECG recording. Three ascending doses of vehicle (n = 6), dofetilide (2, 6 or 20 μg/kg; n = 7), ranolazine (2, 6 or 20 mg/kg; n = 5) or verapamil (0.1, 0.3 or 1.0 mg/kg; n = 4) were administered intravenously. RESULTS As reported in previous clinical studies, dofetilide induced dose-dependent increases in QTc interval, with increases in both J-TpeakC or Tpeak-Tend, while verapamil caused no significant increase in QTc interval, J-TpeakC or Tpeak-Tend. Ranolazine caused dose-dependent increases in QTc interval and corrected J-Tpeakc, but had no effect on Tpeak-Tend, which is in contrast to the effects reported in humans at similar concentrations. Only dofetilide caused a clear, dose-related decrease in the EMW. DISCUSSION These findings suggest that measurements of J-Tpeakc and Tpeak-Tend in addition to QT interval, may help differentiate pure hERG channel blockers with high risk of TdP from lower risk, multichannel blockers.
Collapse
Affiliation(s)
- Matt Skinner
- Vivonics Preclinical Ltd, BioCity Nottingham, Pennyfoot Street, Nottingham NG1 1GF, UK.
| | - Ed Hale
- Vivonics Preclinical Ltd, BioCity Nottingham, Pennyfoot Street, Nottingham NG1 1GF, UK.
| | - Peter Ceuppens
- Inferstats Consulting Ltd, Biohub at Alderley Park, Cheshire SK10 4TG, UK.
| | - Chris Pollard
- Vivonics Preclinical Ltd, BioCity Nottingham, Pennyfoot Street, Nottingham NG1 1GF, UK.
| |
Collapse
|
6
|
Aslostovar L, Boyd AL, Benoit YD, Di Lu J, Garcia Rodriguez JL, Nakanishi M, Porras DP, Reid JC, Mitchell RR, Leber B, Xenocostas A, Foley R, Bhatia M. Abnormal dopamine receptor signaling allows selective therapeutic targeting of neoplastic progenitors in AML patients. CELL REPORTS MEDICINE 2021; 2:100202. [PMID: 33665638 PMCID: PMC7897800 DOI: 10.1016/j.xcrm.2021.100202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/02/2020] [Accepted: 01/19/2021] [Indexed: 12/17/2022]
Abstract
The aberrant expression of dopamine receptors (DRDs) in acute myeloid leukemia (AML) cells has encouraged the repurposing of DRD antagonists such as thioridazine (TDZ) as anti-leukemic agents. Here, we access patient cells from a Phase I dose escalation trial to resolve the cellular and molecular bases of response to TDZ, and we extend these findings to an additional independent cohort of AML patient samples tested preclinically. We reveal that in DRD2+ AML patients, DRD signaling in leukemic progenitors provides leukemia-exclusive networks of sensitivity that spare healthy hematopoiesis. AML progenitor cell suppression can be increased by the isolation of the positive enantiomer from the racemic TDZ mixture (TDZ+), and this is accompanied by reduced cardiac liability. Our study indicates that the development of DRD-directed therapies provides a targeting strategy for a subset of AML patients and potentially other cancers that acquire DRD expression upon transformation from healthy tissue. Leukemic progenitors are a critical cellular target of DRD2 antagonist TDZ DRD2 protein expression is a reliable biomarker of TDZ response DRD2 antagonism selectively triggers leukemic maturation programs via cyclic AMP An enantiomer of TDZ displays a superior efficacy:risk ratio relative to racemic TDZ
Collapse
Affiliation(s)
- Lili Aslostovar
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Allison L Boyd
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Yannick D Benoit
- Department of Cellular and Molecular Medicine, Ottawa University, Ottawa, ON, Canada
| | - Justin Di Lu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | | | - Mio Nakanishi
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Deanna P Porras
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Jennifer C Reid
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Ryan R Mitchell
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Brian Leber
- Department of Medicine, McMaster University, Juravinski Hospital, Hamilton, ON, Canada
| | - Anargyros Xenocostas
- Division of Hematology, Department of Medicine, University of Western Ontario, London Health Sciences Centre, London, ON, Canada
| | - Ronan Foley
- Department of Pathology and Molecular Medicine, McMaster University, Juravinski Hospital, Hamilton, ON, Canada
| | - Mickie Bhatia
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
7
|
Thompson AM, O'Connor PD, Marshall AJ, Yardley V, Maes L, Gupta S, Launay D, Braillard S, Chatelain E, Wan B, Franzblau SG, Ma Z, Cooper CB, Denny WA. Heteroaryl ether analogues of an antileishmanial 7-substituted 2-nitroimidazooxazine lead afford attenuated hERG risk: In vitro and in vivo appraisal. Eur J Med Chem 2020; 209:112914. [PMID: 33268145 DOI: 10.1016/j.ejmech.2020.112914] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/11/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022]
Abstract
Previous investigation of the potent antileishmanial properties of antitubercular 7-substituted 2-nitroimidazo[2,1-b][1,3]oxazines with biaryl side chains led to our development of a new clinical candidate for visceral leishmaniasis (DNDI-0690). Within a collaborative backup program, a racemic monoaryl lead (3) possessing comparable activity in mice but a greater hERG liability formed the starting point for our pursuit of efficacious second generation analogues having good solubility and safety. Asymmetric synthesis and appraisal of its enantiomers first established that chiral preferences for in vivo efficacy were species dependent and that neither form afforded a reduced hERG risk. However, in line with our findings in a structurally related series, less lipophilic heteroaryl ethers provided significant solubility enhancements (up to 16-fold) and concomitantly attenuated hERG inhibition. One promising pyridine derivative (49) displayed 100% oral bioavailability in mice and delivered a 96% parasite burden reduction when dosed at 50 mg/kg in a Leishmania donovani mouse model of visceral leishmaniasis.
Collapse
Affiliation(s)
- Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Andrew J Marshall
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Vanessa Yardley
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Suman Gupta
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Delphine Launay
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202, Geneva, Switzerland
| | - Stephanie Braillard
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202, Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202, Geneva, Switzerland
| | - Baojie Wan
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Zhenkun Ma
- Global Alliance for TB Drug Development, 40 Wall Street, New York, 10005, USA
| | | | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| |
Collapse
|
8
|
Van Brantegem P, Chatterjee S, De Bruyn T, Annaert P, Deferm N. Drug-induced cholestasis assay in primary hepatocytes. MethodsX 2020; 7:101080. [PMID: 33088729 PMCID: PMC7559231 DOI: 10.1016/j.mex.2020.101080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
Abstract
Drug-induced cholestasis (DIC) is a major cause of clinical failure of drug candidates. Numerous patients worldwide are affected when exposed to marketed drugs exhibiting a DIC signature. Prospective identification of DIC during early compound development remains challenging. Here we describe the optimized in vitro procedure for early assessment and prediction of an increased DIC risk. Our method is based on three principles:•Exposure of primary human hepatocyte cultures to test compounds in the absence and presence of a physiologically relevant mixture of endogenous bile salts.•Rapid and quantitative assessment of the influence of concomitant bile salt exposure on hepatocyte functionality and integrity after 24 h or 48 h of incubation.•Translation of the in vitro result, expressed as a DIC index (DICI) value, into an in vivo safety margin.Using our historical control data, a new (data driven) DICI cut-off value of 0.78 was established for discerning cholestatic and non-cholestatic compounds. Our DIC assay protocol was further improved by now relying on the principle of the no observable adverse effect level (NOAEL) for determining the highest test compound concentration corresponding to a DICI ≥ 0.78. Predicted safety margin values were subsequently calculated for compounds displaying hepatotoxic and/or cholestatic effects in patients, thus enabling evaluation of the performance of our DIC assay. Of note, this assay can be extended to explore the role of drug metabolites in precipitating DIC.
Collapse
Affiliation(s)
- Pieter Van Brantegem
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Sagnik Chatterjee
- Pharmaceutical Candidate Optimization, Biocon, Bristol-Myers Squibb R& D Center (BBRC), Syngene International Ltd., Bangalore, India
| | - Tom De Bruyn
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc, South San Francisco, CA, USA
| | - Pieter Annaert
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Neel Deferm
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| |
Collapse
|
9
|
Corpas-López V, Tabraue-Chávez M, Sixto-López Y, Panadero-Fajardo S, Alves de Lima Franco F, Domínguez-Seglar JF, Morillas-Márquez F, Franco-Montalbán F, Díaz-Gavilán M, Correa-Basurto J, López-Viota J, López-Viota M, Pérez del Palacio J, de la Cruz M, de Pedro N, Martín-Sánchez J, Gómez-Vidal JA. O-Alkyl Hydroxamates Display Potent and Selective Antileishmanial Activity. J Med Chem 2020; 63:5734-5751. [DOI: 10.1021/acs.jmedchem.9b02016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Victoriano Corpas-López
- Departamento de Parasitologı́a, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Mavys Tabraue-Chávez
- Departamento de Quı́mica Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Yudibeth Sixto-López
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos y Productos Biotecnológicos, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 México City, México
| | - Sonia Panadero-Fajardo
- Departamento de Quı́mica Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Fernando Alves de Lima Franco
- Departamento de Parasitologı́a, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - José F. Domínguez-Seglar
- Departamento de Quı́mica Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Francisco Morillas-Márquez
- Departamento de Parasitologı́a, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Francisco Franco-Montalbán
- Departamento de Quı́mica Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Mónica Díaz-Gavilán
- Departamento de Quı́mica Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos y Productos Biotecnológicos, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 México City, México
| | - Julián López-Viota
- Departamento de Farmacia y Tecnologı́a Farmacéutica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - Margarita López-Viota
- Departamento de Farmacia y Tecnologı́a Farmacéutica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | | | | | - Nuria de Pedro
- Fundación MEDINA, Parque Tecnológico de la Salud, 18016 Granada, Spain
| | - Joaquina Martín-Sánchez
- Departamento de Parasitologı́a, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| | - José A. Gómez-Vidal
- Departamento de Quı́mica Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Campus de Cartuja, 18071 Granada, Spain
| |
Collapse
|
10
|
Wang Y, Dai Y, Wu X, Li F, Liu B, Li C, Liu Q, Zhou Y, Wang B, Zhu M, Cui R, Tan X, Xiong Z, Liu J, Tan M, Xu Y, Geng M, Jiang H, Liu H, Ai J, Zheng M. Discovery and Development of a Series of Pyrazolo[3,4-d]pyridazinone Compounds as the Novel Covalent Fibroblast Growth Factor Receptor Inhibitors by the Rational Drug Design. J Med Chem 2019; 62:7473-7488. [DOI: 10.1021/acs.jmedchem.9b00510] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | - Fei Li
- School of Chemistry, Shanghai University, 99 ShangDa Road, Shanghai 200444, China
| | | | | | | | - Yuanyang Zhou
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Bao Wang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 200031, China
| | | | | | - Xiaoqin Tan
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Zhaoping Xiong
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 200031, China
| | | | | | | | | | - Hualiang Jiang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 200031, China
| | | | - Jing Ai
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | | |
Collapse
|
11
|
McAleer CW, Pointon A, Long CJ, Brighton RL, Wilkin BD, Bridges LR, Narasimhan Sriram N, Fabre K, McDougall R, Muse VP, Mettetal JT, Srivastava A, Williams D, Schnepper MT, Roles JL, Shuler ML, Hickman JJ, Ewart L. On the potential of in vitro organ-chip models to define temporal pharmacokinetic-pharmacodynamic relationships. Sci Rep 2019; 9:9619. [PMID: 31270362 PMCID: PMC6610665 DOI: 10.1038/s41598-019-45656-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Functional human-on-a-chip systems hold great promise to enable quantitative translation to in vivo outcomes. Here, we explored this concept using a pumpless heart only and heart:liver system to evaluate the temporal pharmacokinetic/pharmacodynamic (PKPD) relationship for terfenadine. There was a time dependent drug-induced increase in field potential duration in the cardiac compartment in response to terfenadine and that response was modulated using a metabolically competent liver module that converted terfenadine to fexofenadine. Using this data, a mathematical model was developed to predict the effect of terfenadine in preclinical species. Developing confidence that microphysiological models could have a transformative effect on drug discovery, we also tested a previously discovered proprietary AstraZeneca small molecule and correctly determined the cardiotoxic response to its metabolite in the heart:liver system. Overall our findings serve as a guiding principle to future investigations of temporal concentration response relationships in these innovative in vitro models, especially, if validated across multiple time frames, with additional pharmacological mechanisms and molecules representing a broad chemical diversity.
Collapse
Affiliation(s)
| | - Amy Pointon
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Christopher J Long
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Rocky L Brighton
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Benjamin D Wilkin
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - L Richard Bridges
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | | | - Kristin Fabre
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Robin McDougall
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Victorine P Muse
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Jerome T Mettetal
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | | | - Dominic Williams
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Mark T Schnepper
- NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Jeff L Roles
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Michael L Shuler
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - James J Hickman
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA.
- NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| | - Lorna Ewart
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
12
|
Patel N, Hatley O, Berg A, Romero K, Wisniowska B, Hanna D, Hermann D, Polak S. Towards Bridging Translational Gap in Cardiotoxicity Prediction: an Application of Progressive Cardiac Risk Assessment Strategy in TdP Risk Assessment of Moxifloxacin. AAPS JOURNAL 2018. [DOI: 10.1208/s12248-018-0199-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
13
|
Thompson AM, O'Connor PD, Marshall AJ, Blaser A, Yardley V, Maes L, Gupta S, Launay D, Braillard S, Chatelain E, Wan B, Franzblau SG, Ma Z, Cooper CB, Denny WA. Development of (6 R)-2-Nitro-6-[4-(trifluoromethoxy)phenoxy]-6,7-dihydro-5 H-imidazo[2,1- b][1,3]oxazine (DNDI-8219): A New Lead for Visceral Leishmaniasis. J Med Chem 2018; 61:2329-2352. [PMID: 29461823 PMCID: PMC5867678 DOI: 10.1021/acs.jmedchem.7b01581] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
Discovery
of the potent antileishmanial effects of antitubercular
6-nitro-2,3-dihydroimidazo[2,1-b][1,3]oxazoles and
7-substituted 2-nitro-5,6-dihydroimidazo[2,1-b][1,3]oxazines
stimulated the examination of further scaffolds (e.g., 2-nitro-5,6,7,8-tetrahydroimidazo[2,1-b][1,3]oxazepines), but the results for these seemed less
attractive. Following the screening of a 900-compound pretomanid analogue
library, several hits with more suitable potency, solubility, and
microsomal stability were identified, and the superior efficacy of
newly synthesized 6R enantiomers with phenylpyridine-based
side chains was established through head-to-head assessments in a Leishmania donovani mouse model. Two such leads (R-84 and R-89) displayed promising activity in the more stringent Leishmania
infantum hamster model but were unexpectedly found to be
potent inhibitors of hERG. An extensive structure–activity
relationship investigation pinpointed two compounds (R-6 and pyridine R-136)
with better solubility and pharmacokinetic properties that also provided
excellent oral efficacy in the same hamster model (>97% parasite
clearance
at 25 mg/kg, twice daily) and exhibited minimal hERG inhibition. Additional
profiling earmarked R-6 as the favored
backup development candidate.
Collapse
Affiliation(s)
- Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Andrew J Marshall
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Adrian Blaser
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| | - Vanessa Yardley
- Faculty of Infectious & Tropical Diseases , London School of Hygiene & Tropical Medicine , Keppel Street , London WC1E 7HT , United Kingdom
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences , University of Antwerp , Universiteitsplein 1 , B-2610 Antwerp , Belgium
| | - Suman Gupta
- Division of Parasitology , CSIR-Central Drug Research Institute , Lucknow 226031 , India
| | - Delphine Launay
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant , 1202 Geneva , Switzerland
| | - Stephanie Braillard
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant , 1202 Geneva , Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant , 1202 Geneva , Switzerland
| | - Baojie Wan
- Institute for Tuberculosis Research, College of Pharmacy , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Zhenkun Ma
- Global Alliance for TB Drug Development , 40 Wall Street , New York , New York 10005 , United States
| | - Christopher B Cooper
- Global Alliance for TB Drug Development , 40 Wall Street , New York , New York 10005 , United States
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences , The University of Auckland , Private Bag 92019, Auckland 1142 , New Zealand
| |
Collapse
|
14
|
Rubi L, Kovar M, Zebedin-Brandl E, Koenig X, Dominguez-Rodriguez M, Todt H, Kubista H, Boehm S, Hilber K. Modulation of the heart's electrical properties by the anticonvulsant drug retigabine. Toxicol Appl Pharmacol 2017. [PMID: 28641963 DOI: 10.1016/j.taap.2017.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Retigabine, currently used as antiepileptic drug, has a wide range of potential medical uses. Administration of the drug in patients can lead to QT interval prolongation in the electrocardiogram and to cardiac arrhythmias in rare cases. This suggests that the drug may perturb the electrical properties of the heart, and the underlying mechanisms were investigated here. Effects of retigabine on currents through human cardiac ion channels, heterologously expressed in tsA-201 cells, were studied in whole-cell patch-clamp experiments. In addition, the drug's impact on the cardiac action potential was tested. This was done using ventricular cardiomyocytes isolated from Langendorff-perfused guinea pig hearts and cardiomyocytes derived from human induced pluripotent stem cells. Further, to unravel potential indirect effects of retigabine on the heart which might involve the autonomic nervous system, membrane potential and noradrenaline release from sympathetic ganglionic neurons were measured in the absence and presence of the drug. Retigabine significantly inhibited currents through hKv11.1 potassium, hNav1.5 sodium, as well as hCav1.2 calcium channels, but only in supra-therapeutic concentrations. In a similar concentration range, the drug shortened the action potential in both guinea pig and human cardiomyocytes. Therapeutic concentrations of retigabine, on the other hand, were sufficient to inhibit the activity of sympathetic ganglionic neurons. We conclude that retigabine- induced QT interval prolongation, and the reported cases of cardiac arrhythmias after application of the drug in a typical daily dose range, cannot be explained by a direct modulatory effect on cardiac ion channels. They are rather mediated by indirect actions at the level of the autonomic nervous system.
Collapse
Affiliation(s)
- Lena Rubi
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Kovar
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Eva Zebedin-Brandl
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Manuel Dominguez-Rodriguez
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannes Todt
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | - Karlheinz Hilber
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
15
|
Thompson AM, O'Connor PD, Marshall AJ, Yardley V, Maes L, Gupta S, Launay D, Braillard S, Chatelain E, Franzblau SG, Wan B, Wang Y, Ma Z, Cooper CB, Denny WA. 7-Substituted 2-Nitro-5,6-dihydroimidazo[2,1-b][1,3]oxazines: Novel Antitubercular Agents Lead to a New Preclinical Candidate for Visceral Leishmaniasis. J Med Chem 2017; 60:4212-4233. [PMID: 28459575 PMCID: PMC7722354 DOI: 10.1021/acs.jmedchem.7b00034] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Within a backup program for the clinical investigational agent pretomanid (PA-824), scaffold hopping from delamanid inspired the discovery of a novel class of potent antitubercular agents that unexpectedly possessed notable utility against the kinetoplastid disease visceral leishmaniasis (VL). Following the identification of delamanid analogue DNDI-VL-2098 as a VL preclinical candidate, this structurally related 7-substituted 2-nitro-5,6-dihydroimidazo[2,1-b][1,3]oxazine class was further explored, seeking efficacious backup compounds with improved solubility and safety. Commencing with a biphenyl lead, bioisosteres formed by replacing one phenyl by pyridine or pyrimidine showed improved solubility and potency, whereas more hydrophilic side chains reduced VL activity. In a Leishmania donovani mouse model, two racemic phenylpyridines (71 and 93) were superior, with the former providing >99% inhibition at 12.5 mg/kg (b.i.d., orally) in the Leishmania infantum hamster model. Overall, the 7R enantiomer of 71 (79) displayed more optimal efficacy, pharmacokinetics, and safety, leading to its selection as the preferred development candidate.
![]()
Collapse
Affiliation(s)
- Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Andrew J Marshall
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Vanessa Yardley
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine , Keppel Street, London WC1E 7HT, United Kingdom
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp , Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Suman Gupta
- Division of Parasitology, CSIR-Central Drug Research Institute , Lucknow 226031, India
| | - Delphine Launay
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Stephanie Braillard
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Baojie Wan
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Zhenkun Ma
- Global Alliance for TB Drug Development , 40 Wall Street, New York 10005, United States
| | - Christopher B Cooper
- Global Alliance for TB Drug Development , 40 Wall Street, New York 10005, United States
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
16
|
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
|
17
|
Panchaud P, Bruyère T, Blumstein AC, Bur D, Chambovey A, Ertel EA, Gude M, Hubschwerlen C, Jacob L, Kimmerlin T, Pfeifer T, Prade L, Seiler P, Ritz D, Rueedi G. Discovery and Optimization of Isoquinoline Ethyl Ureas as Antibacterial Agents. J Med Chem 2017; 60:3755-3775. [DOI: 10.1021/acs.jmedchem.6b01834] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Philippe Panchaud
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Thierry Bruyère
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | | - Daniel Bur
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Alain Chambovey
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Eric A. Ertel
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Markus Gude
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | | - Loïc Jacob
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Thierry Kimmerlin
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Thomas Pfeifer
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Lars Prade
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Peter Seiler
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Daniel Ritz
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Georg Rueedi
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| |
Collapse
|
18
|
Ando H, Yoshinaga T, Yamamoto W, Asakura K, Uda T, Taniguchi T, Ojima A, Shinkyo R, Kikuchi K, Osada T, Hayashi S, Kasai C, Miyamoto N, Tashibu H, Yamazaki D, Sugiyama A, Kanda Y, Sawada K, Sekino Y. A new paradigm for drug-induced torsadogenic risk assessment using human iPS cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 2017; 84:111-127. [DOI: 10.1016/j.vascn.2016.12.003] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 01/05/2023]
|
19
|
Lester RM, Olbertz J. Early drug development: assessment of proarrhythmic risk and cardiovascular safety. Expert Rev Clin Pharmacol 2016; 9:1611-1618. [PMID: 27718759 DOI: 10.1080/17512433.2016.1245142] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION hERG assays and thorough ECG trials have been mandated since 2005 to evaluate the QT interval and potential proarrhythmic risk of new chemical entities. The high cost of these studies and the shortcomings inherent in these binary and limited approaches to drug evaluation have prompted regulators to search for more cost effective and mechanistic paradigms to assess drug liability as exemplified by the CiPA initiative and the exposure response ICH E14(R3) guidance document. Areas covered: This review profiles the changing regulatory landscape as it pertains to early drug development and outlines the analyses that can be performed to characterize preclinical and early clinical cardiovascular risk. Expert commentary: It is further acknowledged that the narrow focus on the QT interval needs to be expanded to include a more comprehensive evaluation of cardiovascular risk since unanticipated off target effects have led to the withdrawal of multiple drugs after they had been approved and marketed.
Collapse
Affiliation(s)
- Robert M Lester
- a Cardiovascular Safety Services , Celerion Inc. , Tempe , AZ , USA
| | - Joy Olbertz
- a Cardiovascular Safety Services , Celerion Inc. , Tempe , AZ , USA
| |
Collapse
|
20
|
Oorts M, Baze A, Bachellier P, Heyd B, Zacharias T, Annaert P, Richert L. Drug-induced cholestasis risk assessment in sandwich-cultured human hepatocytes. Toxicol In Vitro 2016; 34:179-186. [DOI: 10.1016/j.tiv.2016.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/09/2016] [Accepted: 03/11/2016] [Indexed: 12/19/2022]
|
21
|
Kui P, Orosz S, Takács H, Sarusi A, Csík N, Rárosi F, Csekő C, Varró A, Papp JG, Forster T, Farkas AS, Farkas A. New in vitro model for proarrhythmia safety screening: IKs inhibition potentiates the QTc prolonging effect of IKr inhibitors in isolated guinea pig hearts. J Pharmacol Toxicol Methods 2016; 80:26-34. [DOI: 10.1016/j.vascn.2016.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/25/2023]
|
22
|
Salvo F, Pariente A, Shakir S, Robinson P, Arnaud M, Thomas SHL, Raschi E, Fourrier-Réglat A, Moore N, Sturkenboom M, Hazell on behalf of Investigators o L. Sudden cardiac and sudden unexpected death related to antipsychotics: A meta-analysis of observational studies. Clin Pharmacol Ther 2015; 99:306-14. [DOI: 10.1002/cpt.250] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/09/2015] [Indexed: 01/11/2023]
Affiliation(s)
- F Salvo
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
| | - A Pariente
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
- CIC Bordeaux CIC1401; Bordeaux France
| | - S Shakir
- Drug Safety Research Unit; Southampton Hampshire UK
| | - P Robinson
- CIC Bordeaux CIC1401; Bordeaux France
- ADERA; Pessac France
| | - M Arnaud
- University of Bordeaux, INSERM U657; Bordeaux France
| | - SHL Thomas
- Medical Toxicology Centre, Institute of Cellular Medicine; Newcastle University; Newcastle UK
| | - E Raschi
- Department of Medical and Surgical Sciences; University of Bologna; Bologna Italy
| | - A Fourrier-Réglat
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
| | - N Moore
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
- CIC Bordeaux CIC1401; Bordeaux France
| | - M Sturkenboom
- Department of Medical Informatics; Erasmus University Medical Centre; Rotterdam Netherlands
| | | | | |
Collapse
|
23
|
Morissette P, Regan HK, Fitzgerald K, Bernasconi S, Gerenser P, Travis J, Fanelli P, Sannajust F, Regan CP. QT interval correction assessment in the anesthetized guinea pig. J Pharmacol Toxicol Methods 2015; 75:52-61. [DOI: 10.1016/j.vascn.2015.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/10/2015] [Accepted: 05/13/2015] [Indexed: 01/08/2023]
|
24
|
Clements M, Millar V, Williams AS, Kalinka S. Bridging Functional and Structural Cardiotoxicity Assays Using Human Embryonic Stem Cell-Derived Cardiomyocytes for a More Comprehensive Risk Assessment. Toxicol Sci 2015; 148:241-60. [DOI: 10.1093/toxsci/kfv180] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
25
|
Pergola C, Gerstmeier J, Mönch B, Çalışkan B, Luderer S, Weinigel C, Barz D, Maczewsky J, Pace S, Rossi A, Sautebin L, Banoglu E, Werz O. The novel benzimidazole derivative BRP-7 inhibits leukotriene biosynthesis in vitro and in vivo by targeting 5-lipoxygenase-activating protein (FLAP). Br J Pharmacol 2015; 171:3051-64. [PMID: 24641614 DOI: 10.1111/bph.12625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/25/2014] [Accepted: 02/03/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Leukotrienes (LTs) are inflammatory mediators produced via the 5-lipoxygenase (5-LOX) pathway and are linked to diverse disorders, including asthma, allergic rhinitis and cardiovascular diseases. We recently identified the benzimidazole derivative BRP-7 as chemotype for anti-LT agents by virtual screening targeting 5-LOX-activating protein (FLAP). Here, we aimed to reveal the in vitro and in vivo pharmacology of BRP-7 as an inhibitor of LT biosynthesis. EXPERIMENTAL APPROACH We analysed LT formation and performed mechanistic studies in human neutrophils and monocytes, in human whole blood (HWB) and in cell-free assays. The effectiveness of BRP-7 in vivo was evaluated in rat carrageenan-induced pleurisy and mouse zymosan-induced peritonitis. KEY RESULTS BRP-7 potently suppressed LT formation in neutrophils and monocytes and this was accompanied by impaired 5-LOX co-localization with FLAP. Neither the cellular viability nor the activity of 5-LOX in cell-free assays was affected by BRP-7, indicating that a functional FLAP is needed for BRP-7 to inhibit LTs, and FLAP bound to BRP-7 linked to a solid matrix. Compared with the FLAP inhibitor MK-886, BRP-7 did not significantly inhibit COX-1 or microsomal prostaglandin E2 synthase-1, implying the selectivity of BRP-7 for FLAP. Finally, BRP-7 was effective in HWB and impaired inflammation in vivo, in rat pleurisy and mouse peritonitis, along with reducing LT levels. CONCLUSIONS AND IMPLICATIONS BRP-7 potently suppresses LT biosynthesis by interacting with FLAP and exhibits anti-inflammatory effectiveness in vivo, with promising potential for further development.
Collapse
Affiliation(s)
- C Pergola
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wiśniowska B, Mendyk A, Szlęk J, Kołaczkowski M, Polak S. Enhanced QSAR models for drug-triggered inhibition of the main cardiac ion currents. J Appl Toxicol 2015; 35:1030-9. [DOI: 10.1002/jat.3095] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/27/2014] [Accepted: 10/31/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College; Jagiellonian University; Krakow Poland
| | - Aleksander Mendyk
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical College; Jagiellonian University; Krakow Poland
| | - Jakub Szlęk
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical College; Jagiellonian University; Krakow Poland
| | - Michał Kołaczkowski
- Building and Structure Physics Division, Institute of Building Materials and Structures, Faculty of Civil Engineering; Cracow University of Technology; Krakow Poland
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College; Jagiellonian University; Krakow Poland
| |
Collapse
|
27
|
Abstract
Nonclinical safety pharmacology and toxicology testing of drug candidates assess the potential adverse effects caused by the drug in relation to its intended use in humans. Hazards related to a drug have to be identified and the potential risks at the intended exposure have to be evaluated in comparison to the potential benefit of the drug. Preclinical safety is thus an integral part of drug discovery and drug development. It still causes significant attrition during drug development.Therefore, there is a need for smart selection of drug candidates in drug discovery including screening of important safety endpoints. In the recent years,there was significant progress in computational and in vitro technology allowing in silico assessment as well as high-throughput screening of some endpoints at very early stages of discovery. Despite all this progress, in vivo evaluation of drug candidates is still an important part to safety testing. The chapter provides an overview on the most important areas of nonclinical safety screening during drug discovery of small molecules.
Collapse
|
28
|
Kojima A, Ito Y, Kitagawa H, Matsuura H. Ionic mechanisms underlying the negative chronotropic action of propofol on sinoatrial node automaticity in guinea pig heart. Br J Pharmacol 2014; 172:799-814. [PMID: 25220338 DOI: 10.1111/bph.12936] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/16/2014] [Accepted: 09/09/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Propofol is a widely used intravenous anaesthetic agent, but has undesirable cardiac side effects, including bradyarrhythmia and its severe form asystole. This study examined the ionic and cellular mechanisms underlying propofol-induced bradycardia. EXPERIMENTAL APPROACH Sinoatrial node cells, isolated from guinea pig hearts, were current- and voltage-clamped to record action potentials and major ionic currents involved in their spontaneous activity, such as the hyperpolarization-activated cation current (If ), T-type and L-type Ca(2+) currents (ICa,T and ICa,L , respectively) and the rapidly and slowly activating delayed rectifier K(+) currents (IKr and IK s , respectively). ECGs were recorded from Langendorff-perfused, isolated guinea pig hearts. KEY RESULTS Propofol (≥5 μM) reversibly decreased the firing rate of spontaneous action potentials and their diastolic depolarization rate. Propofol impaired If activation by shifting the voltage-dependent activation to more hyperpolarized potentials (≥1 μM), slowing the activation kinetics (≥3 μM) and decreasing the maximal conductance (≥10 μM). Propofol decreased ICa,T (≥3 μM) and ICa,L (≥1 μM). Propofol suppressed IKs (≥3 μM), but had a minimal effect on IKr . Furthermore, propofol (≥5 μM) decreased heart rates in Langendorff-perfused hearts. The sinoatrial node cell model reasonably well reproduced the negative chronotropic action of propofol. CONCLUSIONS AND IMPLICATIONS Micromolar concentrations of propofol suppressed the slow diastolic depolarization and firing rate of sinoatrial node action potentials by impairing If activation and reducing ICa,T , ICa,L and IKs . These observations suggest that the direct inhibitory effect of propofol on sinoatrial node automaticity, mediated via multiple channel inhibition, underlies the propofol-induced bradycardia observed in clinical settings.
Collapse
Affiliation(s)
- Akiko Kojima
- Department of Anesthesiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | | | | |
Collapse
|
29
|
Kauthale RR, Dadarkar SS, Husain R, Karande VV, Gatne MM. Assessment of temperature-induced hERG channel blockade variation by drugs. J Appl Toxicol 2014; 35:799-805. [PMID: 25348819 DOI: 10.1002/jat.3074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 08/20/2014] [Accepted: 08/20/2014] [Indexed: 12/21/2022]
Abstract
Drug-induced QT prolongation has been reported in humans and animals. This potentially lethal effect can be induced by drugs interacting with a cardiac potassium channel, namely hERG (human ether-a go-go-related gene) leading to arrhythmia or torsade de pointes (TdP). Hence, in vitro evaluation of therapeutics for their effects on the rapid delayed rectifier current (IKr) mediated by the K(+) ion channel encoded by hERG is a valuable tool for identifying potential arrhythmic side effects during drug safety testing. Our objective was to evaluate the temperature-induced hERG channel blockade variation by human and veterinary drugs using the IonFlux 16 system. A panel of eight drugs was tested for IKr inhibition at both ambient (23 °C) and physiological (37 °C) temperatures at various concentrations using IonFlux 16, an automated patch clamp system. Our results established that both amiodarone (IC(50) = 0.56 μM at 23 °C and 0.30 μM at 37 °C) and β-estradiol (IC(50) = 24.72 μM at 23 °C and 8.17 μM at 37 °C) showed a dose-dependent IKr blockade with a higher blockade at 37 °C. Whereas, blockade of IKr by both ivermectin (IC(50) = 12.52 μM at 23 °C and 24.41 μM at 37 °C) and frusemide (IC(50) = 12.58 μM at 23 °C and 25.55 μM at 37 °C) showed a dose-dependent IKr blockade with a lower blockade at 37 °C. Gentamicin, enrofloxacin, xylazine and albendazole did not block IKr at both the assessed temperatures. Collectively, these results demonstrate that the effect of temperature variation should be taken into consideration during the evaluation of test drugs for their hERG channel blockade potential.
Collapse
Affiliation(s)
- Rahul R Kauthale
- Department of Pharmacology and Toxicology, Bombay Veterinary College, Mumbai, 400012, Maharashtra, India
| | - Shruta S Dadarkar
- Department of Toxicology, Piramal Enterprises Limited, Mumbai, 400063, Maharashtra, India
| | - Raghib Husain
- Department of Toxicology, Piramal Enterprises Limited, Mumbai, 400063, Maharashtra, India
| | - Vikas V Karande
- Department of Pharmacology and Toxicology, Bombay Veterinary College, Mumbai, 400012, Maharashtra, India
| | - Madhumanjiri M Gatne
- Department of Pharmacology and Toxicology, Bombay Veterinary College, Mumbai, 400012, Maharashtra, India
| |
Collapse
|
30
|
Soragni E, Miao W, Iudicello M, Jacoby D, De Mercanti S, Clerico M, Longo F, Piga A, Ku S, Campau E, Du J, Penalver P, Rai M, Madara JC, Nazor K, O'Connor M, Maximov A, Loring JF, Pandolfo M, Durelli L, Gottesfeld JM, Rusche JR. Epigenetic therapy for Friedreich ataxia. Ann Neurol 2014; 76:489-508. [PMID: 25159818 PMCID: PMC4361037 DOI: 10.1002/ana.24260] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate whether a histone deacetylase inhibitor (HDACi) would be effective in an in vitro model for the neurodegenerative disease Friedreich ataxia (FRDA) and to evaluate safety and surrogate markers of efficacy in a phase I clinical trial in patients. METHODS We used a human FRDA neuronal cell model, derived from patient induced pluripotent stem cells, to determine the efficacy of a 2-aminobenzamide HDACi (109) as a modulator of FXN gene expression and chromatin histone modifications. FRDA patients were dosed in 4 cohorts, ranging from 30mg/day to 240mg/day of the formulated drug product of HDACi 109, RG2833. Patients were monitored for adverse effects as well as for increases in FXN mRNA, frataxin protein, and chromatin modification in blood cells. RESULTS In the neuronal cell model, HDACi 109/RG2833 increases FXN mRNA levels and frataxin protein, with concomitant changes in the epigenetic state of the gene. Chromatin signatures indicate that histone H3 lysine 9 is a key residue for gene silencing through methylation and reactivation through acetylation, mediated by the HDACi. Drug treatment in FRDA patients demonstrated increased FXN mRNA and H3 lysine 9 acetylation in peripheral blood mononuclear cells. No safety issues were encountered. INTERPRETATION Drug exposure inducing epigenetic changes in neurons in vitro is comparable to the exposure required in patients to see epigenetic changes in circulating lymphoid cells and increases in gene expression. These findings provide a proof of concept for the development of an epigenetic therapy for this fatal neurological disease.
Collapse
Affiliation(s)
- Elisabetta Soragni
- Departments of Cell and Molecular Biology, Scripps Research Institute, La Jolla, CA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee YSE, Chuang SH, Huang LYL, Lai CL, Lin YH, Yang JY, Liu CW, Yang SC, Lin HS, Chang CC, Lai JY, Jian PS, Lam K, Chang JM, Lau JYN, Huang JJ. Discovery of 4-aryl-N-arylcarbonyl-2-aminothiazoles as Hec1/Nek2 inhibitors. Part I: optimization of in vitro potencies and pharmacokinetic properties. J Med Chem 2014; 57:4098-110. [PMID: 24773549 DOI: 10.1021/jm401990s] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A series of 4-aryl-N-arylcarbonyl-2-aminothiazoles of scaffold 4 was designed and synthesized as Hec1/Nek2 inhibitors. Structural optimization of 4 led to compound 32 bearing C-4' 4-methoxyphenoxy and 4-(o-fluoropyridyl)carbonyl groups that showed low nanomolar in vitro antiproliferative activity (IC50: 16.3-42.7 nM), high intravenous AUC (64.9 μM·h, 2.0 mg/kg) in SD rats, and significant in vivo antitumor activity (T/C = 32%, 20 mg/kg, IV) in mice bearing human MDA-MB-231 xenografts. Cell responses resulting from Hec1/Nek2 inhibition were observed in cells treated with 32, including a reduced level of Hec1 coimmunoprecipitated with Nek2, degradation of Nek2, mitotic abnormalities, and apoptosis. Compound 32 showed selectivity toward cancer cells over normal phenotype cells and was inactive in a [(3)H]astemizole competitive binding assay for hERG liability screening. Therefore, 32 is as a good lead toward the discovery of a preclinical candidate targeting Hec1/Nek2 interaction.
Collapse
Affiliation(s)
- Ying-Shuan E Lee
- Development Center for Biotechnology , No. 101, Lane 169, Kangning Street, Xizhi District, New Taipei City 22180, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hepatocyte-based in vitro model for assessment of drug-induced cholestasis. Toxicol Appl Pharmacol 2014; 274:124-36. [DOI: 10.1016/j.taap.2013.10.032] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/25/2013] [Accepted: 10/29/2013] [Indexed: 11/20/2022]
|
33
|
Zhang J, Huan J. Predicting Drug-Induced QT Prolongation Effects Using Multi-View Learning. IEEE Trans Nanobioscience 2013; 12:206-13. [DOI: 10.1109/tnb.2013.2263511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Chain ASY, Sturkenboom MCJM, Danhof M, Della Pasqua OE. Establishing in vitro to clinical correlations in the evaluation of cardiovascular safety pharmacology. DRUG DISCOVERY TODAY. TECHNOLOGIES 2013; 10:e373-e383. [PMID: 24050134 DOI: 10.1016/j.ddtec.2012.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Preclinical studies are vital in establishing the efficacy and safety of a new chemical entity (NCE) in humans. To deliver meaningful information, experiments have to be well defined and provide outcome that is relevant and translatable to humans. This review briefly surveys the various preclinical experiments that are frequently conducted to assess drug effects on cardiac conductivity in early drug development. We examine the different approaches used to establish correlations between non-clinical and clinical settings and discuss their value in the evaluation of cardiovascular risk.
Collapse
|
35
|
Morissette P, Nishida M, Trepakova E, Imredy J, Lagrutta A, Chaves A, Hoagland K, Hoe CML, Zrada MM, Travis JJ, Zingaro GJ, Gerenser P, Friedrichs G, Salata JJ. The anesthetized guinea pig: An effective early cardiovascular derisking and lead optimization model. J Pharmacol Toxicol Methods 2013; 68:137-49. [DOI: 10.1016/j.vascn.2013.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 04/19/2013] [Accepted: 04/27/2013] [Indexed: 10/26/2022]
|
36
|
Koenig X, Kovar M, Rubi L, Mike AK, Lukacs P, Gawali VS, Todt H, Hilber K, Sandtner W. Anti-addiction drug ibogaine inhibits voltage-gated ionic currents: a study to assess the drug's cardiac ion channel profile. Toxicol Appl Pharmacol 2013; 273:259-68. [PMID: 23707769 PMCID: PMC3853361 DOI: 10.1016/j.taap.2013.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 05/02/2013] [Accepted: 05/04/2013] [Indexed: 01/15/2023]
Abstract
The plant alkaloid ibogaine has promising anti-addictive properties. Albeit not licensed as a therapeutic drug, and despite hints that ibogaine may perturb the heart rhythm, this alkaloid is used to treat drug addicts. We have recently reported that ibogaine inhibits human ERG (hERG) potassium channels at concentrations similar to the drugs affinity for several of its known brain targets. Thereby the drug may disturb the heart's electrophysiology. Here, to assess the drug's cardiac ion channel profile in more detail, we studied the effects of ibogaine and its congener 18-Methoxycoronaridine (18-MC) on various cardiac voltage-gated ion channels. We confirmed that heterologously expressed hERG currents are reduced by ibogaine in low micromolar concentrations. Moreover, at higher concentrations, the drug also reduced human Nav1.5 sodium and Cav1.2 calcium currents. Ion currents were as well reduced by 18-MC, yet with diminished potency. Unexpectedly, although blocking hERG channels, ibogaine did not prolong the action potential (AP) in guinea pig cardiomyocytes at low micromolar concentrations. Higher concentrations (≥ 10 μM) even shortened the AP. These findings can be explained by the drug's calcium channel inhibition, which counteracts the AP-prolonging effect generated by hERG blockade. Implementation of ibogaine's inhibitory effects on human ion channels in a computer model of a ventricular cardiomyocyte, on the other hand, suggested that ibogaine does prolong the AP in the human heart. We conclude that therapeutic concentrations of ibogaine have the propensity to prolong the QT interval of the electrocardiogram in humans. In some cases this may lead to cardiac arrhythmias.
Collapse
Affiliation(s)
- Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and -pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fijorek K, Patel N, Klima Ł, Stolarz-Skrzypek K, Kawecka-Jaszcz K, Polak S. Age and gender dependent heart rate circadian model development and performance verification on the proarrhythmic drug case study. Theor Biol Med Model 2013; 10:7. [PMID: 23394137 PMCID: PMC3598978 DOI: 10.1186/1742-4682-10-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 02/06/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are two main reasons for drug withdrawals at the various levels of the development path - hepatic and cardiac toxicity. The latter one is mainly connected with the proarrhythmic potency and according to the present practice is supposed to be recognized at the pre-clinical (in vitro and animal in vivo) or clinical level (human in vivo studies). There are, although, some limitations to all the above mentioned methods which have led to novel in vitro - in vivo extrapolation methods being introduced. With the use of in silico implemented mathematical and statistical modelling it is possible to translate the in vitro findings into the human in vivo situation at the population level. Human physiology is influenced by many parameters and one of them which needs to be properly accounted for is a heart rate which follows the circadian rhythm. We described such phenomenon statistically which enabled the improved assessment of the drug proarrhythmic potency. METHODS A publicly available data set describing the circadian changes of the heart rate of 18 healthy subjects, 5 males (average age 36, range 26-45) and 13 females (average age 34, range 20-50) was used for the heart rate model development. External validation was done with the use of a clinical research database containing heart rate measurements derived from 67 healthy subjects, 34 males and 33 females (average age 33, range 17-72). The developed heart rate model was then incorporated into the ToxComp platform to simulate the impact of circadian variation in the heart rate on QTc interval. The usability of the combined models was assessed with moxifloxacin (MOXI) as a model drug. RESULTS The developed heart rate model fitted well, both to the training data set (RMSE = 128 ms and MAPE = 12.3%) and the validation data set (RMSE = 165 ms and MAPE = 17.1%). Simulations performed at the population level proved that the combination of the IVIVE platform and the population variability description allows for the precise prediction of the circadian variation of drugs proarrhythmic effect. CONCLUSIONS It can be concluded that a flexible and practically useful model describing the heart rate circadian variation has been developed and its performance was verified.
Collapse
Affiliation(s)
- Kamil Fijorek
- Department of Statistics, Cracow University of Economics, Krakow, Poland.
| | | | | | | | | | | |
Collapse
|
38
|
Gillie DJ, Novick SJ, Donovan BT, Payne LA, Townsend C. Development of a high-throughput electrophysiological assay for the human ether-à-go-go related potassium channel hERG. J Pharmacol Toxicol Methods 2013; 67:33-44. [DOI: 10.1016/j.vascn.2012.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 10/02/2012] [Accepted: 10/18/2012] [Indexed: 01/03/2023]
|
39
|
Park MJ, Lee KR, Shin DS, Chun HS, Kim CH, Ahn SH, Bae MA. Predicted drug-induced bradycardia related cardio toxicity using a zebrafish in vivo model is highly correlated with results from in vitro tests. Toxicol Lett 2013; 216:9-15. [DOI: 10.1016/j.toxlet.2012.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 10/26/2012] [Accepted: 10/29/2012] [Indexed: 11/30/2022]
|
40
|
Glinka A, Polak S. Wild type and K897T polymorphisms of the hERG gene: modeling the APD in Caucasians. Bioinformation 2012; 8:1062-5. [PMID: 23251039 PMCID: PMC3523219 DOI: 10.6026/97320630081062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/01/2012] [Indexed: 11/25/2022] Open
Abstract
The presented study aims to assess the possibility of simulating changes in cardiac cell electrophysiology due to K897T polymorphism in the Caucasian population. In the first part of the experiment, the parameters of the equations describing channel gating were fitted to the experimental data. Then, the action potentials of midmyocardial cells of 100 individuals were simulated in the in vitro - in vivo extrapolation system - ToxComp. Mean APD90 for the entire simulated population is 352.05 ms (SD = 21.69 ms). Mean APD90 for the 80 individuals with the WT version of the hERG gene and for the 20 K897T homo- and heterozygotes is respectively 349.08 ms (SD = 21.09 ms) and 363.95ms (SD = 20.41 ms). The ToxComp system can be useful in predicting the impact of genetic variability on drug triggered cardiac cell electrophysiology interference.
Collapse
Affiliation(s)
- Anna Glinka
- Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian, University Medical College, Cracow, Poland, Medyczna 9 Str, 30-688 Kraków, Poland
| | - Sebastian Polak
- Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian, University Medical College, Cracow, Poland, Medyczna 9 Str, 30-688 Kraków, Poland
| |
Collapse
|
41
|
Guns PJ, Johnson DM, Van Op den Bosch J, Weltens E, Lissens J. The electro-mechanical window in anaesthetized guinea pigs: a new marker in screening for Torsade de Pointes risk. Br J Pharmacol 2012; 166:689-701. [PMID: 22122450 DOI: 10.1111/j.1476-5381.2011.01795.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE QT prolongation is commonly used as a surrogate marker for Torsade de Pointes (TdP) risk of non-cardiovascular drugs. However, use of this indirect marker often leads to misinterpretation of the realistic TdP risk, as tested compounds may cause QT prolongation without evoking TdP in humans. A negative electro-mechanical (E-M) window has recently been proposed as an alternative risk marker for TdP in a canine LQT1 model. Here, we evaluated the E-M window in anaesthetized guinea pigs as a screening marker for TdP in humans. EXPERIMENTAL APPROACH The effects of various reference drugs and changes in body temperature on the E-M window were assessed in instrumented guinea pigs. The E-M window was defined as the delay between the duration of the electrical (QT interval) and mechanical (QLVP(end) ) systole. KEY RESULTS Drugs with known TdP liability (quinidine, haloperidol, domperidone, terfenadine, thioridazine and dofetilide), but not those with no TdP risk in humans (salbutamol and diltiazem) consistently decreased the E-M window. Interestingly, drugs with known clinical QT prolongation, but with low risk for TdP (amiodarone, moxifloxacin and ciprofloxacin) did not decrease the E-M window. Furthermore, the E-M window was minimally affected by changes in heart rate or body temperature. CONCLUSIONS AND IMPLICATIONS A decreased E-M window was consistently observed with drugs already known to have high TdP risk, but not with drugs with low or no TdP risk. These results suggest that the E-M window in anaesthetized guinea pigs is a risk marker for TdP in humans.
Collapse
Affiliation(s)
- P-J Guns
- Bio-Plus Safety Pharmacology, Bio-Plus Services, Mol, Belgium.
| | | | | | | | | |
Collapse
|
42
|
Marks L, Borland S, Philp K, Ewart L, Lainée P, Skinner M, Kirk S, Valentin JP. The role of the anaesthetised guinea-pig in the preclinical cardiac safety evaluation of drug candidate compounds. Toxicol Appl Pharmacol 2012; 263:171-83. [DOI: 10.1016/j.taap.2012.06.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 06/07/2012] [Accepted: 06/11/2012] [Indexed: 11/30/2022]
|
43
|
Negative electro-mechanical windows are required for drug-induced Torsades de Pointes in the anesthetized guinea pig. J Pharmacol Toxicol Methods 2012; 66:125-34. [DOI: 10.1016/j.vascn.2012.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 03/13/2012] [Accepted: 03/29/2012] [Indexed: 11/17/2022]
|
44
|
Johnson DM, Geys R, Lissens J, Guns PJ. Drug-induced effects on cardiovascular function in pentobarbital anesthetized guinea-pigs: invasive LVP measurements versus the QA interval. J Pharmacol Toxicol Methods 2012; 66:152-9. [PMID: 22813982 DOI: 10.1016/j.vascn.2012.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 06/13/2012] [Accepted: 07/05/2012] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Evaluation of drug-related effects on cardiovascular function is part of the core battery described in the ICH S7A guideline. Anesthetized guinea-pigs are excellent models for the evaluation of drug-induced prolongation of ventricular repolarization; however less information is available regarding other cardio-hemodynamic parameters in this model. The current study aimed to document cardio-hemodynamic responses in anesthetized guinea-pigs after administration of a number of reference drugs with known pharmacological actions. METHODS Experiments were carried out in closed chest pentobarbital anesthetized female guinea-pigs. Compounds were administered intravenously while arterial blood pressure, left ventricular pressure (LVP) and the electrocardiogram were measured continuously. The rate of LVP contraction (LV dP/dt(max)) was used to evaluate cardiac performance; and was compared to the QA interval; which has previously been proposed as an indirect measurement of cardiac function. RESULTS Baseline values for heart rate and blood pressure were lower in anesthetized animals compared to literature data of conscious guinea-pigs. Heart rate increased after administration of adrenaline, isoprenaline and salbutamol, but not after L-phenylephrine. Verapamil and amiodarone decreased heart rate and blood pressure. Zatebradine infusion led to a decrease in heart rate with minimal effects on blood pressure. Sodium nitroprusside (SNP) caused a reduction in mean blood pressure at higher doses followed by reflex tachycardia. Both adrenaline and L-phenylephrine increased arterial blood pressure. Furthermore, adrenaline, isoprenaline and salbutamol increased LV dP/dt(max) and decreased the QA interval. L-phenylephrine increased LV dP/dt(max), but transiently prolonged the QA interval. Both verapamil and amiodarone decreased LV dP/dt(max) and prolonged the QA interval, whereas zatebradine did not affect this parameter. DISCUSSION In addition to its utility for the assessment of test compounds on ventricular repolarization the pentobarbital anesthetized guinea-pig model shows promise for early stage cardio-hemodynamic screening. Furthermore, the QA interval shows potential for prediction of adverse effects on cardiac contractility.
Collapse
Affiliation(s)
- D M Johnson
- Bio-Plus Safety Pharmacology, Vlasmeer 5/0003, B-2400 Mol, Belgium.
| | | | | | | |
Collapse
|
45
|
Wen D, Liu A, Chen F, Yang J, Dai R. Validation of visualized transgenic zebrafish as a high throughput model to assay bradycardia related cardio toxicity risk candidates. J Appl Toxicol 2012; 32:834-42. [PMID: 22744888 DOI: 10.1002/jat.2755] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/27/2012] [Accepted: 02/27/2012] [Indexed: 11/09/2022]
Abstract
Drug-induced QT prolongation usually leads to torsade de pointes (TdP), thus for drugs in the early phase of development this risk should be evaluated. In the present study, we demonstrated a visualized transgenic zebrafish as an in vivo high-throughput model to assay the risk of drug-induced QT prolongation. Zebrafish larvae 48 h post-fertilization expressing green fluorescent protein in myocardium were incubated with compounds reported to induce QT prolongation or block the human ether-a-go-go-related gene (hERG) K⁺ current. The compounds sotalol, indapaminde, erythromycin, ofoxacin, levofloxacin, sparfloxacin and roxithromycin were additionally administrated by microinjection into the larvae yolk sac. The ventricle heart rate was recorded using the automatic monitoring system after incubation or microinjection. As a result, 14 out of 16 compounds inducing dog QT prolongation caused bradycardia in zebrafish. A similar result was observed with 21 out of 26 compounds which block hERG current. Among the 30 compounds which induced human QT prolongation, 25 caused bradycardia in this model. Thus, the risk of compounds causing bradycardia in this transgenic zebrafish correlated with that causing QT prolongation and hERG K⁺ current blockage in established models. The tendency that high logP values lead to high risk of QT prolongation in this model was indicated, and non-sensitivity of this model to antibacterial agents was revealed. These data suggest application of this transgenic zebrafish as a high-throughput model to screen QT prolongation-related cardio toxicity of the drug candidates.
Collapse
Affiliation(s)
- Dingsheng Wen
- South China University of Technology, Guangzhou 510641, China
| | | | | | | | | |
Collapse
|
46
|
Mooney L, Marks L, Philp KL, Skinner M, Coker SJ, Currie S. Optimising conditions for studying the acute effects of drugs on indices of cardiac contractility and on haemodynamics in anaesthetized guinea pigs. J Pharmacol Toxicol Methods 2012; 66:43-51. [PMID: 22659057 DOI: 10.1016/j.vascn.2012.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/19/2012] [Accepted: 05/23/2012] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Detecting adverse effects of drugs on cardiac contractility is becoming a priority in pre-clinical safety pharmacology. The aim of this work was to optimise conditions and explore the potential of using the anaesthetized guinea pig as an in vivo model. METHODS Guinea pigs were anaesthetized with Hypnorm/Hypnovel, isoflurane, pentobarbital or fentanyl/pentobarbital. The electrocardiogram (ECG), heart rate, arterial blood pressure and indices of cardiac contractility were recorded. In further experiments in fentanyl/pentobarbital anaesthetized guinea pigs the influence of bilateral versus unilateral carotid artery occlusion on haemodynamic responses was investigated and the effects of inotropic drugs on left ventricular (LV) dP/dt(max) and the QA interval were determined. RESULTS Pentobarbital, given alone or after fentanyl, provided suitable anaesthesia for these experiments. Bilateral carotid artery occlusion did not alter heart rate or arterial blood pressure responses to isoprenaline or angiotensin II. Isoprenaline and ouabain increased LVdP/dt(max) and decreased the QA interval whereas verapamil had opposite effects and strong inverse correlations between LVdP/dt(max) and the QA interval were found. DISCUSSION Conditions can be optimised to allow the pentobarbital-anaesthetized guinea pig to be used for simultaneous measurement of the effects of drugs on the ECG, haemodynamics and indices of cardiac contractility. The use of this small animal model in early pre-clinical safety pharmacology should contribute to improvements in detecting unwanted actions on the heart during the drug development process.
Collapse
Affiliation(s)
- Laura Mooney
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | | | | | | | | | | |
Collapse
|
47
|
Satin LZ, Durham TA, Turner JR. Assessing a Drug’s Proarrhythmic Liability: An Overview of Computer Simulation Modeling, Nonclinical Assays, and the Thorough QT/QTc Study. ACTA ACUST UNITED AC 2011. [DOI: 10.1177/009286151104500315] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
48
|
Supplemental Studies for Cardiovascular Risk Assessment in Safety Pharmacology: A Critical Overview. Cardiovasc Toxicol 2011; 11:285-307. [DOI: 10.1007/s12012-011-9133-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
49
|
Green JR, Diaz GJ, Limberis JT, Houseman KA, Su Z, Martin RL, Cox BF, Kantor S, Gintant GA. Ventricular rate adaptation: A novel, rapid, cellular-based in-vitro assay to identify proarrhythmic and torsadogenic compounds. J Pharmacol Toxicol Methods 2011; 64:68-73. [DOI: 10.1016/j.vascn.2011.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 03/17/2011] [Accepted: 03/20/2011] [Indexed: 10/18/2022]
|
50
|
Kazmierski WM, Anderson DL, Aquino C, Chauder BA, Duan M, Ferris R, Kenakin T, Koble CS, Lang DG, McIntyre MS, Peckham J, Watson C, Wheelan P, Spaltenstein A, Wire MB, Svolto A, Youngman M. Novel 4,4-disubstituted piperidine-based C-C chemokine receptor-5 inhibitors with high potency against human immunodeficiency virus-1 and an improved human ether-a-go-go related gene (hERG) profile. J Med Chem 2011; 54:3756-67. [PMID: 21539377 DOI: 10.1021/jm200279v] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We recently described ( J. Med. Chem. 2008 , 51 , 6538 - 6546 ) a novel class of CCR5 antagonists with strong anti-HIV potency. Herein, we detail SAR converting leads 1 and 2 to druglike molecules. The pivotal structural motif enabling this transition was the secondary sulfonamide substituent. Further fine-tuning of the substituent pattern in the sulfonamide paved the way to enhancing potency and bioavailability and minimizing hERG inhibition, resulting in discovery of clinical compound 122 (GSK163929).
Collapse
|