97 results on '"Gintant G"'
Search Results
2. The Role of Local Anesthetic Effects in the Actions of Antiarrhythmic Drugs
- Author
-
Gintant, G. A., Hoffman, B. F., and Strichartz, Gary R., editor
- Published
- 1987
- Full Text
- View/download PDF
3. The Use of Ratiometric Fluorescence Measurements of the Voltage Sensitive Dye Di-4-ANEPPS to Examine Action Potential Characteristics and Drug Effects on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes
- Author
-
Hortigon-Vinagre, M. P., primary, Zamora, V., additional, Burton, F. L., additional, Green, J., additional, Gintant, G. A., additional, and Smith, G. L., additional
- Published
- 2016
- Full Text
- View/download PDF
4. Sphingosine-1-Phosphate (S1P) Lyase Inhibition Causes Increased Cardiac S1P Levels and Bradycardia in Rats
- Author
-
Harris, C. M., primary, Mittelstadt, S., additional, Banfor, P., additional, Bousquet, P., additional, Duignan, D. B., additional, Gintant, G., additional, Hart, M., additional, Kim, Y., additional, and Segreti, J., additional
- Published
- 2016
- Full Text
- View/download PDF
5. Versatile temperature controlled tissue bath for studies of isolated cells using an inverted microscope
- Author
-
Datyner, N. B., Gintant, G. A., and Cohen, I. S.
- Published
- 1985
- Full Text
- View/download PDF
6. Microprocessor controlled trituration device for the dissociation of cardiac and other tissues
- Author
-
Datyner, N. B., Gintant, G. A., and Cohen, I. S.
- Published
- 1985
- Full Text
- View/download PDF
7. The Role of Local Anesthetic Effects in the Actions of Antiarrhythmic Drugs
- Author
-
Gintant, G. A., primary and Hoffman, B. F., additional
- Published
- 1987
- Full Text
- View/download PDF
8. A novel method for Poincaré Plot shape quantification demonstrates cardiac tissue repolarization inhomogeneities induced by drugs
- Author
-
Mensing, S., primary, Limberis, J., additional, Gintant, G., additional, and Safer, A., additional
- Published
- 2008
- Full Text
- View/download PDF
9. A modification of the in vitro canine Purkinje fiber repolarization assay using variable frequency pacing.
- Author
-
Safer, A. and Gintant, G.
- Published
- 2003
- Full Text
- View/download PDF
10. Regional differences in IK density in canine left ventricle: role of IK,s in electrical heterogeneity
- Author
-
Gintant, G. A., primary
- Published
- 1995
- Full Text
- View/download PDF
11. Ionic bases for electrophysiological distinctions among epicardial, midmyocardial, and endocardial myocytes from the free wall of the canine left ventricle.
- Author
-
Liu, D W, primary, Gintant, G A, additional, and Antzelevitch, C, additional
- Published
- 1993
- Full Text
- View/download PDF
12. Beta-adrenergic modulation of fast inward sodium current in canine myocardium. Syncytial preparations versus isolated myocytes.
- Author
-
Gintant, G A, primary and Liu, D W, additional
- Published
- 1992
- Full Text
- View/download PDF
13. Heterogeneity within the ventricular wall. Electrophysiology and pharmacology of epicardial, endocardial, and M cells.
- Author
-
Antzelevitch, C, primary, Sicouri, S, additional, Litovsky, S H, additional, Lukas, A, additional, Krishnan, S C, additional, Di Diego, J M, additional, Gintant, G A, additional, and Liu, D W, additional
- Published
- 1991
- Full Text
- View/download PDF
14. Advances in Cardiac Cellular Electrophysiology: Implications for Automaticity and Therapeutics.
- Author
-
Gintant, G A and Cohen, I S
- Published
- 1988
- Full Text
- View/download PDF
15. A Note on the Relation of Maximum Upstroke Velocity to Peak Inward Current Recorded by the Voltage Clamp.
- Author
-
Cohen, I. S., Datyner, N. B., Gintant, G. A., Mulrine, N. K., and Pennefather, P.
- Published
- 1985
16. Saturation of the internal sodium site of the sodium pump can distort estimates of potassium affinity
- Author
-
Cohen, I., Falk, R., and Gintant, G.
- Published
- 1984
- Full Text
- View/download PDF
17. Properties of an electrogenic sodium‐potassium pump in isolated canine Purkinje myocytes.
- Author
-
Cohen, I S, Datyner, N B, Gintant, G A, Mulrine, N K, and Pennefather, P
- Abstract
1. Purkinje myocytes were isolated from canine Purkinje strands by collagenase exposure and gentle trituration. The myocytes were studied by a switched single‐micro‐electrode voltage‐clamp technique at 37 degrees C in Tyrode solution containing 8 mM‐K+ and 2 mM‐Ca2+. 2. The dose‐response relation for the cardiotonic steroid dihydroouabain (DHO) was obtained by measuring the change in membrane current caused by application of concentrations of 1‐100 microM. The KD obtained in fourteen experiments was 3.7 +/‐ 1.1 microM (mean +/‐ S.E. of mean). 3. We employed 100 microM‐DHO (a concentration more than 25‐fold greater than the KD) to estimate the resting pump current (Ip) in the isolated myocytes. A value of 0.27 +/‐ 0.02 microA microF‐1 (mean +/‐ S.E. of mean, n = 32) was obtained. 4. Myocytes were also exposed to K+‐free solution for a period of 200 s. On return to K+‐containing Tyrode solution there was a slowly decaying outward current. The time constant of decay of this pump current transient was 87 +/‐ 8 s (mean +/‐ S.E. of mean, n = 8). The integral beneath this transient was used to obtain a second estimate of the resting pump current. In four preparations where exposures in DHO and in K+‐free solutions were employed the ratio Ip, DHO/Ip, K‐free was 1.76 +/‐ 0.15 (mean +/‐ S.E. of mean). 5. From the magnitude of resting pump current, in the presence of total pump blockade the Na+ activity should rise at a rate of 1.3 mM min‐1. 6. Reducing [K+]o from 8 to 1 mM reduced Ip by more than 40% initially. Ip then slowly increased over the next 30 min. These results suggest that the steady‐state inward background current is not greatly altered by changes in [K+]o, and that [Na+]i rises to a new level. The changes in Ip obtained at early times following reduction of [K+]o to 1 or 0.5 mM (t less than 1.75 min) were used to estimate the Km for external K+; a value of 0.8 mM was obtained. 7. The results suggest that the properties of the Na+‐K+ pump in isolated canine Purkinje myocytes are similar to those in canine Purkinje strands. This argues against major distortions of measured pump properties in the canine Purkinje strand and for the physiological state of the Na+‐K+ pump in the isolated Purkinje myocyte.
- Published
- 1987
- Full Text
- View/download PDF
18. A novel Na+-dependent slow current component in canine cardiac Purkinje fibers
- Author
-
GINTANT, G, primary
- Published
- 1989
- Full Text
- View/download PDF
19. The influence of molecular form of local anesthetic-type antiarrhythmic agents on reduction of the maximum upstroke velocity of canine cardiac Purkinje fibers.
- Author
-
Gintant, G A, primary, Hoffman, B F, additional, and Naylor, R E, additional
- Published
- 1983
- Full Text
- View/download PDF
20. Assessing the fidelity of translation of non-clinical assays: a Pharma perspective.
- Author
-
Gintant G
- Subjects
- Humans, Predictive Value of Tests, Sensitivity and Specificity, ROC Curve
- Abstract
Advances in non-clinical in vitro models, higher throughput approaches and the promise of human-derived preparations require methods to reliably assess the fidelity of translation of such assays, compared with in vivo models and clinical studies. This review discusses general principles and parameters useful to evaluate the value of non-clinical assays typically used to guide compound progression. I first consider the biological characteristics (including sensitivity and ability to replicate relevant responses) of models that form the foundation of an assay based on the questions posed. I then discuss the quantitative assessment of diagnostic performance and assay utility, including sensitivity and specificity, receiver operating characteristic curves, positive and negative predictive values, likelihood ratios, along with advantages of combining two independent assays. Understanding the strengths and limitations of the biological model employed along with assay performance and context of use is essential to selecting the best assays supporting the best drug candidates., (© 2022 The British Pharmacological Society.)
- Published
- 2022
- Full Text
- View/download PDF
21. Discovery of (R)-(3-fluoropyrrolidin-1-yl)(6-((5-(trifluoromethyl)pyridin-2-yl)oxy)quinolin-2-yl)methanone (ABBV-318) and analogs as small molecule Na v 1.7/ Nav1.8 blockers for the treatment of pain.
- Author
-
Patel MV, Peltier HM, Matulenko MA, Koenig JR, C Scanio MJ, Gum RJ, El-Kouhen OF, Fricano MM, Lundgaard GL, Neelands T, Zhang XF, Zhan C, Pai M, Ghoreishi-Haack N, Hudzik T, Gintant G, Martin R, McGaraughty S, Xu J, Bow D, Kalvass JC, Kym PR, DeGoey DA, and Kort ME
- Subjects
- Humans, Pain Management, Protein Isoforms, Structure-Activity Relationship, Pain drug therapy, Sodium Channels metabolism
- Abstract
The voltage-gated sodium channel Na
v 1.7 is an attractive target for the treatment of pain based on the high level of target validation with genetic evidence linking Nav 1.7 to pain in humans. Our effort to identify selective, CNS-penetrant Nav 1.7 blockers with oral activity, improved selectivity, good drug-like properties, and safety led to the discovery of 2-substituted quinolines and quinolones as potent small molecule Nav 1.7 blockers. The design of these molecules focused on maintaining potency at Nav 1.7, improving selectivity over the hERG channel, and overcoming phospholipidosis observed with the initial leads. The structure-activity relationship (SAR) studies leading to the discovery of (R)-(3-fluoropyrrolidin-1-yl)(6-((5-(trifluoromethyl)pyridin-2-yl)oxy)quinolin-2-yl)methanone (ABBV-318) are described herein. ABBV-318 displayed robust in vivo efficacy in both inflammatory and neuropathic rodent models of pain. ABBV-318 also inhibited Nav 1.8, another sodium channel isoform that is an active target for the development of new pain treatments., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
22. The Challenges of Predicting Drug-Induced QTc Prolongation in Humans.
- Author
-
Valentin JP, Hoffmann P, Ortemann-Renon C, Koerner J, Pierson J, Gintant G, Willard J, Garnett C, Skinner M, Vargas HM, Wisialowski T, and Pugsley MK
- Subjects
- Action Potentials, Animals, Electrocardiography, Heart, Humans, Retrospective Studies, Long QT Syndrome chemically induced, Torsades de Pointes chemically induced
- Abstract
The content of this article derives from a Health and Environmental Sciences Institute (HESI) consortium with a focus to improve cardiac safety during drug development. A detailed literature review was conducted to evaluate the concordance between nonclinical repolarization assays and the clinical thorough QT (TQT) study. Food and Drug Administration and HESI developed a joint database of nonclinical and clinical data, and a retrospective analysis of 150 anonymized drug candidates was reviewed to compare the performance of 3 standard nonclinical assays with clinical TQT study findings as well as investigate mechanism(s) potentially responsible for apparent discrepancies identified. The nonclinical assays were functional (IKr) current block (Human ether-a-go-go related gene), action potential duration, and corrected QT interval in animals (in vivo corrected QT). Although these nonclinical assays demonstrated good specificity for predicting negative clinical QT prolongation, they had relatively poor sensitivity for predicting positive clinical QT prolongation. After review, 28 discordant TQT-positive drugs were identified. This article provides an overview of direct and indirect mechanisms responsible for QT prolongation and theoretical reasons for lack of concordance between clinical TQT studies and nonclinical assays. We examine 6 specific and discordant TQT-positive drugs as case examples. These were derived from the unique HESI/Food and Drug Administration database. We would like to emphasize some reasons for discordant data including, insufficient or inadequate nonclinical data, effects of the drug on other cardiac ion channels, and indirect and/or nonelectrophysiological effects of drugs, including altered heart rate. We also outline best practices that were developed based upon our evaluation., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2022
- Full Text
- View/download PDF
23. Cardiovascular microphysiological systems (CVMPS) for safety studies - a pharma perspective.
- Author
-
Pointon A, Maher J, Davis M, Baker T, Cichocki J, Ramsden D, Hale C, Kolaja KL, Levesque P, Sura R, Stresser DM, and Gintant G
- Subjects
- Models, Cardiovascular, Myocytes, Cardiac, Reproducibility of Results, Endothelial Cells, Induced Pluripotent Stem Cells
- Abstract
The integrative responses of the cardiovascular (CV) system are essential for maintaining blood flow to provide oxygenation, nutrients, and waste removal for the entire body. Progress has been made in independently developing simple in vitro models of two primary components of the CV system, namely the heart (using induced pluripotent stem-cell derived cardiomyocytes) and the vasculature (using endothelial cells and smooth muscle cells). These two in vitro biomimics are often described as immature and simplistic, and typically lack the structural complexity of native tissues. Despite these limitations, they have proven useful for specific "fit for purpose" applications, including early safety screening. More complex in vitro models offer the tantalizing prospect of greater refinement in risk assessments. To this end, efforts to physically link cardiac and vascular components to mimic a true CV microphysiological system (CVMPS) are ongoing, with the goal of providing a more holistic and integrated CV response model. The challenges of building and implementing CVMPS in future pharmacological safety studies are many, and include a) the need for more complex (and hence mature) cell types and tissues, b) the need for more realistic vasculature (within and across co-modeled tissues), and c) the need to meaningfully couple these two components to allow for integrated CV responses. Initial success will likely come with simple, bioengineered tissue models coupled with fluidics intended to mirror a vascular component. While the development of more complex integrated CVMPS models that are capable of differentiating safe compounds and providing mechanistic evaluations of CV liabilities may be feasible, adoption by pharma will ultimately hinge on model efficiency, experimental reproducibility, and added value above current strategies.
- Published
- 2021
- Full Text
- View/download PDF
24. Repolarization studies using human stem cell-derived cardiomyocytes: Validation studies and best practice recommendations.
- Author
-
Gintant G, Kaushik EP, Feaster T, Stoelzle-Feix S, Kanda Y, Osada T, Smith G, Czysz K, Kettenhofen R, Lu HR, Cai B, Shi H, Herron TJ, Dang Q, Burton F, Pang L, Traebert M, Abassi Y, Pierson JB, and Blinova K
- Subjects
- Adult Stem Cells pathology, Adult Stem Cells physiology, Arrhythmias, Cardiac pathology, Arrhythmias, Cardiac physiopathology, Humans, Membrane Potentials drug effects, Membrane Potentials physiology, Myocytes, Cardiac pathology, Adult Stem Cells drug effects, Arrhythmias, Cardiac chemically induced, Cardiotoxins toxicity, Myocytes, Cardiac drug effects, Practice Guidelines as Topic standards, Validation Studies as Topic
- Abstract
Human stem cell-derived cardiomyocytes (hSC-CMs) hold great promise as in vitro models to study the electrophysiological effects of novel drug candidates on human ventricular repolarization. Two recent large validation studies have demonstrated the ability of hSC-CMs to detect drug-induced delayed repolarization and "cellrhythmias" (interrupted repolarization or irregular spontaneous beating of myocytes) linked to Torsade-de-Pointes proarrhythmic risk. These (and other) studies have also revealed variability of electrophysiological responses attributable to differences in experimental approaches and experimenter, protocols, technology platforms used, and pharmacologic sensitivity of different human-derived models. Thus, when evaluating drug-induced repolarization effects, there is a need to consider 1) the advantages and disadvantages of different approaches, 2) the need for robust functional characterization of hSC-CM preparations to define "fit for purpose" applications, and 3) adopting standardized best practices to guide future studies with evolving hSC-CM preparations. Examples provided and suggested best practices are instructional in defining consistent, reproducible, and interpretable "fit for purpose" hSC-CM-based applications. Implementation of best practices should enhance the clinical translation of hSC-CM-based cell and tissue preparations in drug safety evaluations and support their growing role in regulatory filings., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
25. Identification of Drug-Induced Multichannel Block and Proarrhythmic Risk in Humans Using Continuous T Vector Velocity Effect Profiles Derived From Surface Electrocardiograms.
- Author
-
Bystricky W, Maier C, Gintant G, Bergau D, and Carter D
- Abstract
We present continuous T vector velocity (TVV) effect profiles as a new method for identifying drug effects on cardiac ventricular repolarization. TVV measures the temporal change in the myocardial action potential distribution during repolarization. The T vector dynamics were measured as the time required to reach p percent of the total T vector trajectory length, denoted as Tr(p), with p in {1, …, 100%}. The Tr(p) values were individually corrected for heart rate at each trajectory length percentage p. Drug effects were measured by evaluating the placebo corrected changes from baseline of Tr(p)c jointly for all p using functional mixed effects models. The p-dependent model parameters were implemented as cubic splines, providing continuous drug effect profiles along the entire ventricular repolarization process. The effect profile distributions were approximated by bootstrap simulations. We applied this TVV-based analysis approach to ECGs available from three published studies that were conducted in the CiPA context. These studies assessed the effect of 10 drugs and drug combinations with different ion channel blocking properties on myocardial repolarization in a total of 104 healthy volunteers. TVV analysis revealed that blockade of outward potassium currents alone presents an effect profile signature of continuous accumulation of delay throughout the entire repolarization interval. In contrast, block of inward sodium or calcium currents involves acceleration, which accumulates during early repolarization. The balance of blocking inward versus outward currents was reflected in the percentage p
zero of the T vector trajectory length where accelerated repolarization transitioned to delayed repolarization. Binary classification using a threshold pzero = 43% separated predominant hERG channel blocking drugs with potentially higher proarrhythmic risk (moxifloxacin, dofetilide, quinidine, chloroquine) from multichannel blocking drugs with low proarrhythmic risk (ranolazine, verapamil, lopinavir/ritonavir) with sensitivity 0.99 and specificity 0.97. The TVV-based effect profile provides a detailed view of drug effects throughout the entire ventricular repolarization interval. It enables the evaluation of drug-induced blocks of multiple cardiac repolarization currents from clinical ECGs. The proposed pzero parameter enhances identification of the proarrhythmic risk of a drug beyond QT prolongation, and therefore constitutes an important tool for cardiac arrhythmia risk assessment., (Copyright © 2020 Bystricky, Maier, Gintant, Bergau and Carter.)- Published
- 2020
- Full Text
- View/download PDF
26. Publisher Correction: Cross-site and cross-platform variability of automated patch clamp assessments of drug effects on human cardiac currents in recombinant cells.
- Author
-
Kramer J, Himmel HM, Lindqvist A, Stoelzle-Feix S, Chaudhary KW, Li D, Bohme GA, Bridgland-Taylor M, Hebeisen S, Fan J, Renganathan M, Imredy J, Humphries ESA, Brinkwirth N, Strassmaier T, Ohtsuki A, Danker T, Vanoye C, Polonchuk L, Fermini B, Pierson JB, and Gintant G
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
27. The roles of human induced pluripotent stem cell-derived cardiomyocytes in drug discovery: managing in vitro safety study expectations.
- Author
-
Gintant G and Traebert M
- Subjects
- Adult, Animals, Cardiotoxicity etiology, Drug Discovery methods, Humans, Induced Pluripotent Stem Cells cytology, Myocytes, Cardiac pathology, Reproducibility of Results, Cardiotoxicity physiopathology, Drug Development methods, Myocytes, Cardiac drug effects
- Abstract
Introduction: Human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) preparations are increasingly employed in in vitro cardiac safety studies to support candidate drug selection and regulatory submissions. The value of hiPSC-CM-based approaches depends on their ability to recapitulate the cellular mechanisms responsible for cardiotoxicity as well as overall assay characteristics (thus defining model performance). Different expectations at different drug development stages define the utility of these human-derived models., Areas Covered: Herein, the authors review the importance of understanding the functional characteristics of the evolving spectrum of simpler (2D) and more complex (co-cultures, 3D constructs, and engineered tissues) human-derived cardiac preparations, and how their performance may be evaluated based on analytical sensitivity, variability, and reproducibility in order to correctly match preparations with expectations of different safety assays. The need for consensus clinical examples of electrophysiologic, contractile, and structural cardiotoxicities essential for benchmarking human-derived models is also discussed., Expert Opinion: It is helpful (but not essential) that hiPSC-CMs preparations fully recapitulate pharmacological responses of native adult human ventricular myocytes when evaluating cardiotoxicity in vitro . Further calibration and model standardization (aligning concordance with clinical findings) are necessary to understand the role of hiPSC-CMs in guiding cardiotoxicity assessments in early drug discovery efforts.
- Published
- 2020
- Full Text
- View/download PDF
28. Cross-site and cross-platform variability of automated patch clamp assessments of drug effects on human cardiac currents in recombinant cells.
- Author
-
Kramer J, Himmel HM, Lindqvist A, Stoelzle-Feix S, Chaudhary KW, Li D, Bohme GA, Bridgland-Taylor M, Hebeisen S, Fan J, Renganathan M, Imredy J, Humphries ESA, Brinkwirth N, Strassmaier T, Ohtsuki A, Danker T, Vanoye C, Polonchuk L, Fermini B, Pierson JB, and Gintant G
- Abstract
Automated patch clamp (APC) instruments enable efficient evaluation of electrophysiologic effects of drugs on human cardiac currents in heterologous expression systems. Differences in experimental protocols, instruments, and dissimilar site procedures affect the variability of IC
50 values characterizing drug block potency. This impacts the utility of APC platforms for assessing a drug's cardiac safety margin. We determined variability of APC data from multiple sites that measured blocking potency of 12 blinded drugs (with different levels of proarrhythmic risk) against four human cardiac currents (hERG [IKr ], hCav1.2 [L-Type ICa ], peak hNav1.5, [Peak INa ], late hNav1.5 [Late INa ]) with recommended protocols (to minimize variance) using five APC platforms across 17 sites. IC50 variability (25/75 percentiles) differed for drugs and currents (e.g., 10.4-fold for dofetilide block of hERG current and 4-fold for mexiletine block of hNav1.5 current). Within-platform variance predominated for 4 of 12 hERG blocking drugs and 4 of 6 hNav1.5 blocking drugs. hERG and hNav1.5 block. Bland-Altman plots depicted varying agreement across APC platforms. A follow-up survey suggested multiple sources of experimental variability that could be further minimized by stricter adherence to standard protocols. Adoption of best practices would ensure less variable APC datasets and improved safety margins and proarrhythmic risk assessments.- Published
- 2020
- Full Text
- View/download PDF
29. Corrigendum to "Assessing cardiac safety in oncology drug development" [Am Heart J 214 (2019) 125-133].
- Author
-
Seltzer JH, Gintant G, Amiri-Kordestani L, Singer J, Koplowitz LP, Moslehi JJ, Barac A, and Yu AF
- Published
- 2019
- Full Text
- View/download PDF
30. Use of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes in Preclinical Cancer Drug Cardiotoxicity Testing: A Scientific Statement From the American Heart Association.
- Author
-
Gintant G, Burridge P, Gepstein L, Harding S, Herron T, Hong C, Jalife J, and Wu JC
- Subjects
- Animals, Cardiotoxicity metabolism, Cardiotoxicity pathology, Drug Evaluation, Preclinical methods, Humans, Induced Pluripotent Stem Cells pathology, Induced Pluripotent Stem Cells physiology, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, United States epidemiology, American Heart Association, Antineoplastic Agents toxicity, Cardiotoxicity genetics, Induced Pluripotent Stem Cells drug effects, Myocytes, Cardiac drug effects
- Abstract
It is now well recognized that many lifesaving oncology drugs may adversely affect the heart and cardiovascular system, including causing irreversible cardiac injury that can result in reduced quality of life. These effects, which may manifest in the short term or long term, are mechanistically not well understood. Research is hampered by the reliance on whole-animal models of cardiotoxicity that may fail to reflect the fundamental biology or cardiotoxic responses of the human myocardium. The emergence of human induced pluripotent stem cell-derived cardiomyocytes as an in vitro research tool holds great promise for understanding drug-induced cardiotoxicity of oncological drugs that may manifest as contractile and electrophysiological dysfunction, as well as structural abnormalities, making it possible to deliver novel drugs free from cardiac liabilities and guide personalized therapy. This article briefly reviews the challenges of cardio-oncology, the strengths and limitations of using human induced pluripotent stem cell-derived cardiomyocytes to represent clinical findings in the nonclinical research space, and future directions for their further use.
- Published
- 2019
- Full Text
- View/download PDF
31. Considerations for an In Vitro , Cell-Based Testing Platform for Detection of Drug-Induced Inotropic Effects in Early Drug Development. Part 2: Designing and Fabricating Microsystems for Assaying Cardiac Contractility With Physiological Relevance Using Human iPSC-Cardiomyocytes.
- Author
-
Ribeiro AJS, Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Brock M, Chaudhary KW, Kanda Y, and Berridge B
- Abstract
Contractility of the myocardium engines the pumping function of the heart and is enabled by the collective contractile activity of its muscle cells: cardiomyocytes. The effects of drugs on the contractility of human cardiomyocytes in vitro can provide mechanistic insight that can support the prediction of clinical cardiac drug effects early in drug development. Cardiomyocytes differentiated from human-induced pluripotent stem cells have high potential for overcoming the current limitations of contractility assays because they attach easily to extracellular materials and last long in culture, while having human- and patient-specific properties. Under these conditions, contractility measurements can be non-destructive and minimally invasive, which allow assaying sub-chronic effects of drugs. For this purpose, the function of cardiomyocytes in vitro must reflect physiological settings, which is not observed in cultured cardiomyocytes derived from induced pluripotent stem cells because of the fetal-like properties of their contractile machinery. Primary cardiomyocytes or tissues of human origin fully represent physiological cellular properties, but are not easily available, do not last long in culture, and do not attach easily to force sensors or mechanical actuators. Microengineered cellular systems with a more mature contractile function have been developed in the last 5 years to overcome this limitation of stem cell-derived cardiomyocytes, while simultaneously measuring contractile endpoints with integrated force sensors/actuators and image-based techniques. Known effects of engineered microenvironments on the maturity of cardiomyocyte contractility have also been discovered in the development of these systems. Based on these discoveries, we review here design criteria of microengineered platforms of cardiomyocytes derived from pluripotent stem cells for measuring contractility with higher physiological relevance. These criteria involve the use of electromechanical, chemical and morphological cues, co-culture of different cell types, and three-dimensional cellular microenvironments. We further discuss the use and the current challenges for developing and improving these novel technologies for predicting clinical effects of drugs based on contractility measurements with cardiomyocytes differentiated from induced pluripotent stem cells. Future research should establish contexts of use in drug development for novel contractility assays with stem cell-derived cardiomyocytes.
- Published
- 2019
- Full Text
- View/download PDF
32. Considerations for an In Vitro , Cell-Based Testing Platform for Detection of Adverse Drug-Induced Inotropic Effects in Early Drug Development. Part 1: General Considerations for Development of Novel Testing Platforms.
- Author
-
Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Ribeiro AJS, Zabka T, Chaudhary KW, Kanda Y, and Berridge B
- Abstract
Drug-induced effects on cardiac contractility can be assessed through the measurement of the maximal rate of pressure increase in the left ventricle (LVdP/dt
max ) in conscious animals, and such studies are often conducted at the late stage of preclinical drug development. Detection of such effects earlier in drug research using simpler, in vitro test systems would be a valuable addition to our strategies for identifying the best possible drug development candidates. Thus, testing platforms with reasonably high throughput, and affordable costs would be helpful for early screening purposes. There may also be utility for testing platforms that provide mechanistic information about how a given drug affects cardiac contractility. Finally, there could be in vitro testing platforms that could ultimately contribute to the regulatory safety package of a new drug. The characteristics needed for a successful cell or tissue-based testing platform for cardiac contractility will be dictated by its intended use. In this article, general considerations are presented with the intent of guiding the development of new testing platforms that will find utility in drug research and development. In the following article (part 2), specific aspects of using human-induced stem cell-derived cardiomyocytes for this purpose are addressed.- Published
- 2019
- Full Text
- View/download PDF
33. Assessing cardiac safety in oncology drug development.
- Author
-
Seltzer JH, Gintant G, Amiri-Kordestani L, Singer J, Koplowitz LP, Moslehi JJ, Barac A, and Yu AF
- Subjects
- Antineoplastic Agents, Immunological adverse effects, Biomarkers blood, Cardiologists, Cell Line, Tumor, Clinical Trials as Topic, Data Collection, Drug Screening Assays, Antitumor, Humans, Immunotherapy adverse effects, Medical Oncology, Research Design, Trastuzumab adverse effects, Antineoplastic Agents adverse effects, Cardiovascular Diseases chemically induced, Drug Development, Heart drug effects
- Published
- 2019
- Full Text
- View/download PDF
34. T vector velocity: A new ECG biomarker for identifying drug effects on cardiac ventricular repolarization.
- Author
-
Bystricky W, Maier C, Gintant G, Bergau D, Kamradt K, Welsh P, and Carter D
- Subjects
- Adult, Databases, Factual, Double-Blind Method, Electrocardiography methods, Female, Humans, Lidocaine pharmacology, Male, Mexiletine pharmacology, Phenethylamines pharmacology, Ranolazine pharmacology, Sulfonamides pharmacology, Verapamil pharmacology, Young Adult, Action Potentials drug effects, Heart drug effects, Potassium Channel Blockers pharmacology, Sodium Channel Blockers pharmacology
- Abstract
Background: We present a new family of ECG biomarkers for assessing drug effects on ventricular repolarization. We show that drugs blocking inward (depolarizing) ion currents cause a relative increase of the T vector velocity (TVV) and accelerate repolarization, while drugs blocking outward ion currents cause a relative decrease of the TVV and delay repolarization. The results suggest a link between the TVV and the instantaneous change of the cellular action potentials that may contribute to bridge the gap between the surface ECG and myocardial cellular processes., Methods: We measure TVV as the time required to reach X% of the total Trajectory length of the T vector loop, denoted as TrX. Applied to data from two FDA funded studies (22+22 subjects, 5232+4208 ECGs) which target ECG effects of various ion-channel blocking drugs, the TrX effect profiles indicate increasingly delayed electrical activity over the entire repolarization process for drugs solely reducing outward potassium current (dofetilide, moxifloxacin). For drugs eliciting block of the inward sodium or calcium currents (mexiletine, lidocaine), the TrX effect profiles were consistent with accelerated electrical activity in the initial repolarization phase. For multichannel blocking drugs (ranolazine) or drug combinations blocking multiple ion currents (dofetilide + mexiletine, dofetilide + lidocaine), the overall TrX effect profiles indicate a superposition of the individual TrX effect profiles., Results: The parameter Tr40c differentiates pure potassium channel blocking drugs from multichannel blocking drugs with an area under the ROC curve (AUC) of 0.90, CI = [0.88 to 0.92]. This is significantly better than the performance of J-Tpeakc (0.81, CI = [0.78 to 0.84]) identified as the best parameter in the second FDA study. Combining the ten parameters Tr10c to Tr100c in a logistic regression model further improved the AUC to 0.94, CI = [0.92 to 0.96]., Conclusions: TVV analysis substantially improves assessment of drug effects on cardiac repolarization, providing a plausible and improved mechanistic link between drug effects on ionic currents and overall ventricular repolarization reflected in the body surface ECG. TVV contributes to an enhanced appraisal of the proarrhythmic risk of drugs beyond QTc prolongation and J-Tpeakc., Competing Interests: W declare the following competing interests statements: Authors WB CM are consultants for AbbVie, Inc. and authors GG DB KK PW DC are employees of AbbVie Inc. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2019
- Full Text
- View/download PDF
35. Comprehensive In Vitro Proarrhythmia Assay (CiPA) Update from a Cardiac Safety Research Consortium / Health and Environmental Sciences Institute / FDA Meeting.
- Author
-
Strauss DG, Gintant G, Li Z, Wu W, Blinova K, Vicente J, Turner JR, and Sager PT
- Subjects
- Biological Assay, Computer Simulation, Electrocardiography, Humans, Induced Pluripotent Stem Cells physiology, Ion Channels physiology, Myocytes, Cardiac, Arrhythmias, Cardiac chemically induced, Drug Evaluation, Preclinical, Drug-Related Side Effects and Adverse Reactions prevention & control
- Abstract
A Cardiac Safety Research Consortium / Health and Environmental Sciences Institute / FDA-sponsored Think Tank Meeting was convened in Washington, DC, on May 21, 2018, to bring together scientists, clinicians, and regulators from multiple geographic regions to evaluate progress to date in the Comprehensive In Vitro Proarrhythmia Assay (CiPA) Initiative, a new paradigm to evaluate proarrhythmic risk. Study reports from the 4 different components of the CiPA paradigm (ionic current studies, in silico modeling to generate a Torsade Metric Score, human induced pluripotent stem cell-derived ventricular cardiomyocytes, and clinical ECG assessments including J-Tpeakc) were presented and discussed. This paper provides a high-level summary of the CiPA data presented at the meeting.
- Published
- 2019
- Full Text
- View/download PDF
36. International Multisite Study of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Drug Proarrhythmic Potential Assessment.
- Author
-
Blinova K, Dang Q, Millard D, Smith G, Pierson J, Guo L, Brock M, Lu HR, Kraushaar U, Zeng H, Shi H, Zhang X, Sawada K, Osada T, Kanda Y, Sekino Y, Pang L, Feaster TK, Kettenhofen R, Stockbridge N, Strauss DG, and Gintant G
- Subjects
- Cardiotoxicity, Cell Line, Cellular Reprogramming, Drug Evaluation, Preclinical standards, Electrophysiology standards, Humans, Membrane Potentials drug effects, Myocytes, Cardiac cytology, Myocytes, Cardiac physiology, Drug Evaluation, Preclinical methods, Electrophysiology methods, Induced Pluripotent Stem Cells cytology, Myocytes, Cardiac drug effects, Torsades de Pointes chemically induced
- Abstract
To assess the utility of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as an in vitro proarrhythmia model, we evaluated the concentration dependence and sources of variability of electrophysiologic responses to 28 drugs linked to low, intermediate, and high torsades de pointes (TdP) risk categories using two commercial cell lines and standardized protocols in a blinded multisite study using multielectrode array or voltage-sensing optical approaches. Logistical and ordinal linear regression models were constructed using drug responses as predictors and TdP risk categories as outcomes. Three of seven predictors (drug-induced arrhythmia-like events and prolongation of repolarization at either maximum tested or maximal clinical exposures) categorized drugs with reasonable accuracy (area under the curve values of receiver operator curves ∼0.8). hiPSC-CM line, test site, and platform had minimal influence on drug categorization. These results demonstrate the utility of hiPSC-CMs to detect drug-induced proarrhythmic effects as part of the evolving Comprehensive In Vitro Proarrhythmia Assay paradigm., (Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
37. CiPA challenges and opportunities from a non-clinical, clinical and regulatory perspectives. An overview of the safety pharmacology scientific discussion.
- Author
-
Wallis R, Benson C, Darpo B, Gintant G, Kanda Y, Prasad K, Strauss DG, and Valentin JP
- Subjects
- Animals, Arrhythmias, Cardiac physiopathology, Arrhythmias, Cardiac prevention & control, Drug Evaluation, Preclinical methods, Drug Evaluation, Preclinical standards, ERG1 Potassium Channel agonists, ERG1 Potassium Channel antagonists & inhibitors, ERG1 Potassium Channel physiology, Electrocardiography standards, Humans, Long QT Syndrome chemically induced, Long QT Syndrome physiopathology, Long QT Syndrome prevention & control, Myocytes, Cardiac drug effects, Myocytes, Cardiac physiology, Arrhythmias, Cardiac chemically induced, Computer Simulation standards, Congresses as Topic standards, Electrocardiography drug effects, Societies, Pharmaceutical standards
- Abstract
The Safety Pharmacology Society organized a scientific session at its annual conference in 2017 to discuss the challenges and opportunities of the Comprehensive In-Vitro Proarrhythmia Assay (CiPA) paradigm. Our intention was to raise awareness of this initiative with its members and also to gauge the extent to which safety pharmacologists have incorporated the CiPA testing strategy within the pharmaceutical industry. CiPA offers many potential opportunities including 1) a focus on proarrhythmic risk (as opposed to QTc prolongation), 2) providing scientific rationale to support the continued development of compounds that may have a poor selectivity over hERG whilst also blocking other inward currents and 3) reducing the extent of ECG monitoring in clinical trials with a greater influence of the non-clinical studies. Such opportunities may speed drug development and reduce costs. However, there are also challenges for CiPA implementation. For example, the mixed ion channel paradigm does not easily lend itself to a prospective drug discovery strategy although testing for such effects can be achieved with assays with good throughput. However, it should also be recognized that compounds with a mixed ion channel profile might also have properties that are undesirable to treat non-life threatening indications. All components of CiPA (nonclinical and clinical) require validation, particularly as a composite package to impact drug development and evaluation. One of the significant discussion points was that the existing regulatory guidance supports the use of components of CiPA through follow-up studies. A survey of the conference audience showed that the level of awareness of CiPA is quite high and that companies are already conducting some testing against a wider panel of cardiac ion channels beyond hERG. However, the adoption of other technologies (stem cell derived cardiac myocytes and in silico modeling) is less well developed. Taken together, the session demonstrated the potential advantages of CiPA, but also some significant challenges., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
38. Drug-induced Proarrhythmia and Torsade de Pointes: A Primer for Students and Practitioners of Medicine and Pharmacy.
- Author
-
Turner JR, Rodriguez I, Mantovani E, Gintant G, Kowey PR, Klotzbaugh RJ, Prasad K, Sager PT, Stockbridge N, and Strnadova C
- Abstract
Multiple marketing withdrawals due to proarrhythmic concerns occurred in the United States, Canada, and the United Kingdom in the late 1980s to early 2000s. This primer reviews the clinical implications of a drug's identified proarrhythmic liability, the issues associated with these safety-related withdrawals, and the actions taken by the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) and by regulatory agencies in terms of changing drug development practices and introducing new nonclinical and clinical tests to asses proarrhythmic liability. ICH Guidelines S7B and E14 were released in 2005. Since then, they have been adopted by many regional regulatory authorities and have guided nonclinical and clinical proarrhythmic cardiac safety assessments during drug development. While this regulatory paradigm has been successful in preventing drugs with unanticipated potential for inducing the rare but potentially fatal polymorphic ventricular arrhythmia torsade de pointes from entering the market, it has led to the termination of drug development programs for other potentially useful medicines because of isolated results from studies with limited predictive value. Research efforts are now exploring alternative approaches to better predict potential proarrhythmic liabilities. For example, in the domain of human electrocardiographic assessments, concentration-response modeling conducted during phase 1 clinical development has recently become an accepted alternate primary methodology to the ICH E14 "thorough QT/QTc" study for defining a drug's corrected QT interval prolongation liability under certain conditions. When a drug's therapeutic benefit is considered important at a public health level but there is also an identified proarrhythmic liability that may result from administration of the single drug in certain individuals and/or drug-drug interactions, marketing approval will be accompanied by appropriate directions in the drug's prescribing information. Health-care professionals in the fields of medicine and pharmacy need to consider the prescribing information in conjunction with individual patients' clinical characteristics and concomitant medications when prescribing and dispensing such drugs., (© 2018, The American College of Clinical Pharmacology.)
- Published
- 2018
- Full Text
- View/download PDF
39. Cross-Site Reliability of Human Induced Pluripotent stem cell-derived Cardiomyocyte Based Safety Assays Using Microelectrode Arrays: Results from a Blinded CiPA Pilot Study.
- Author
-
Millard D, Dang Q, Shi H, Zhang X, Strock C, Kraushaar U, Zeng H, Levesque P, Lu HR, Guillon JM, Wu JC, Li Y, Luerman G, Anson B, Guo L, Clements M, Abassi YA, Ross J, Pierson J, and Gintant G
- Subjects
- Action Potentials drug effects, Cell Line, Drug Evaluation, Preclinical instrumentation, Electrophysiological Phenomena drug effects, Humans, Induced Pluripotent Stem Cells physiology, Myocytes, Cardiac physiology, Pilot Projects, Reproducibility of Results, Cardiovascular Agents pharmacology, Drug Evaluation, Preclinical methods, Induced Pluripotent Stem Cells drug effects, Microelectrodes, Myocytes, Cardiac drug effects
- Abstract
Recent in vitro cardiac safety studies demonstrate the ability of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to detect electrophysiologic effects of drugs. However, variability contributed by unique approaches, procedures, cell lines, and reagents across laboratories makes comparisons of results difficult, leading to uncertainty about the role of hiPSC-CMs in defining proarrhythmic risk in drug discovery and regulatory submissions. A blinded pilot study was conducted to evaluate the electrophysiologic effects of 8 well-characterized drugs on 4 cardiomyocyte lines using a standardized protocol across 3 microelectrode array platforms (18 individual studies). Drugs were selected to define assay sensitivity of prominent repolarizing currents (E-4031 for IKr, JNJ303 for IKs) and depolarizing currents (nifedipine for ICaL, mexiletine for INa) as well as drugs affecting multichannel block (flecainide, moxifloxacin, quinidine, and ranolazine). Inclusion criteria for final analysis was based on demonstrated sensitivity to IKr block (20% prolongation with E-4031) and L-type calcium current block (20% shortening with nifedipine). Despite differences in baseline characteristics across cardiomyocyte lines, multiple sites, and instrument platforms, 10 of 18 studies demonstrated adequate sensitivity to IKr block with E-4031 and ICaL block with nifedipine for inclusion in the final analysis. Concentration-dependent effects on repolarization were observed with this qualified data set consistent with known ionic mechanisms of single and multichannel blocking drugs. hiPSC-CMs can detect repolarization effects elicited by single and multichannel blocking drugs after defining pharmacologic sensitivity to IKr and ICaL block, supporting further validation efforts using hiPSC-CMs for cardiac safety studies.
- Published
- 2018
- Full Text
- View/download PDF
40. Drug-induced cardiac abnormalities in premature infants and neonates.
- Author
-
Pesco-Koplowitz L, Gintant G, Ward R, Heon D, Saulnier M, and Heilbraun J
- Subjects
- Animals, Humans, Infant, Infant, Newborn, Drug-Related Side Effects and Adverse Reactions complications, Heart Defects, Congenital chemically induced, Infant, Premature
- Abstract
The Cardiac Safety Research Consortium (CSRC) is a transparent, public-private partnership that was established in 2005 as a Critical Path Program and formalized in 2006 under a Memorandum of Understanding between the United States Food and Drug Administration and Duke University. Our continuing goal is to advance paradigms for more efficient regulatory science related to the cardiovascular safety of new therapeutics, both in the United States and globally, particularly where such safety questions add burden to innovative research and development. This White Paper provides a summary of discussions by a cardiovascular committee cosponsored by the CSRC and the US Food and Drug Administration (FDA) that initially met in December 2014, and periodically convened at FDA's White Oak headquarters from March 2015 to September 2016. The committee focused on the lack of information concerning the cardiac effects of medications in the premature infant and neonate population compared with that of the older pediatric and adult populations. Key objectives of this paper are as follows: Provide an overview of human developmental cardiac electrophysiology, as well as the electrophysiology of premature infants and neonates; summarize all published juvenile animal models relevant to drug-induced cardiac toxicity; provide a consolidated source for all reported drug-induced cardiac toxicities by therapeutic area as a resource for neonatologists; present drugs that have a known cardiac effect in an adult population, but no reported toxicity in the premature infant and neonate populations; and summarize what is not currently known about drug-induced cardiac toxicity in premature infants and neonates, and what could be done to address this lack of knowledge. This paper presents the views of the authors and should not be construed to represent the views or policies of the FDA or Health Canada., (Copyright © 2017. Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
41. The Evolving Roles of Human iPSC-Derived Cardiomyocytes in Drug Safety and Discovery.
- Author
-
Gintant G, Fermini B, Stockbridge N, and Strauss D
- Subjects
- Arrhythmias, Cardiac pathology, Humans, Induced Pluripotent Stem Cells pathology, Myocytes, Cardiac pathology, Arrhythmias, Cardiac drug therapy, Arrhythmias, Cardiac metabolism, Drug Evaluation, Preclinical methods, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac metabolism
- Abstract
Nonclinical studies of drug effects with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide new possibilities for evaluating drug safety and efficacy. The Comprehensive In Vitro Proarrhythmia Assay (CiPA) paradigm provides lessons from the cardiac field that also apply to drug studies with other stem cell-based assays., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
42. The Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative - Update on progress.
- Author
-
Colatsky T, Fermini B, Gintant G, Pierson JB, Sager P, Sekino Y, Strauss DG, and Stockbridge N
- Subjects
- Animals, Arrhythmias, Cardiac physiopathology, Computer Simulation, Drug Evaluation, Preclinical methods, Electrocardiography drug effects, Humans, Ion Channels drug effects, Myocytes, Cardiac drug effects, Stem Cells, Torsades de Pointes chemically induced, Torsades de Pointes physiopathology, Arrhythmias, Cardiac chemically induced
- Abstract
The implementation of the ICH S7B and E14 guidelines has been successful in preventing the introduction of potentially torsadogenic drugs to the market, but it has also unduly constrained drug development by focusing on hERG block and QT prolongation as essential determinants of proarrhythmia risk. The Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative was established to develop a new paradigm for assessing proarrhythmic risk, building on the emergence of new technologies and an expanded understanding of torsadogenic mechanisms beyond hERG block. An international multi-disciplinary team of regulatory, industry and academic scientists are working together to develop and validate a set of predominantly nonclinical assays and methods that eliminate the need for the thorough-QT study and enable a more precise prediction of clinical proarrhythmia risk. The CiPA effort is led by a Steering Team that provides guidance, expertise and oversight to the various working groups and includes partners from US FDA, HESI, CSRC, SPS, EMA, Health Canada, Japan NIHS, and PMDA. The working groups address the three pillars of CiPA that evaluate drug effects on: 1) human ventricular ionic channel currents in heterologous expression systems, 2) in silico integration of cellular electrophysiologic effects based on ionic current effects, the ion channel effects, and 3) fully integrated biological systems (stem-cell-derived cardiac myocytes and the human ECG). This article provides an update on the progress of the initiative towards its target date of December 2017 for completing validation., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. The Cardiac Safety Research Consortium enters its second decade: An invitation to participate.
- Author
-
Turner JR, Kowey PR, Rodriguez I, Cabell CH, Gintant G, Green CL, Kunz BL, Mortara J, Sager PT, Stockbridge N, Wright TJ, Finkle J, and Krucoff MW
- Subjects
- Humans, Patient Safety, United States, Biomedical Research, Cardiovascular Agents, Cardiovascular Surgical Procedures, Equipment Safety, Public-Private Sector Partnerships, United States Food and Drug Administration, Universities
- Abstract
The Cardiac Safety Research Consortium (CSRC), a transparent, public-private partnership established in 2005 as a Critical Path Program and formalized in 2006 under a Memorandum of Understanding between the United States Food and Drug Administration and Duke University, is entering its second decade. Our continuing goal is to advance paradigms for more efficient regulatory science related to the cardiovascular safety of new therapeutics, both in the United States and globally, particularly where such safety questions add burden to innovative research and development. Operationally, CSRC brings together a broad base of stakeholders from academia, industry, and government agencies in a collaborative forum focused on identifying barriers and then creating novel solutions through shared data, expertise, and collaborative research. This white paper provides a brief overview of the Consortium's activities in its first decade and a context for some of our current activities and future directions. The growth and success of the CSRC have been primarily driven by members' active participation and the development of goodwill and trust throughout our membership, which have facilitated novel collaborations across traditionally competitive or contentious stakeholder boundaries. The continued expansion of our base of participating academicians, industry experts, and regulators will define the Consortium's success in our second decade. It is our hope that sharing our endeavors to date will stimulate additional participation in the CSRC and also provide a model for other groups starting to develop similar collaborative forums., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. Evolution of strategies to improve preclinical cardiac safety testing.
- Author
-
Gintant G, Sager PT, and Stockbridge N
- Subjects
- Animals, Cardiotoxicity metabolism, Cells, Cultured, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells physiology, Myocytes, Cardiac drug effects, Myocytes, Cardiac physiology, Risk Assessment, Safety, Calcium Channel Blockers pharmacology, Cardiotoxicity diagnosis, Drug Evaluation, Preclinical, Induced Pluripotent Stem Cells cytology, Ion Channels antagonists & inhibitors, Myocytes, Cardiac cytology
- Abstract
The early and efficient assessment of cardiac safety liabilities is essential to confidently advance novel drug candidates. This article discusses evolving mechanistically based preclinical strategies for detecting drug-induced electrophysiological and structural cardiotoxicity using in vitro human ion channel assays, human-based in silico reconstructions and human stem cell-derived cardiomyocytes. These strategies represent a paradigm shift from current approaches, which rely on simplistic in vitro assays that measure blockade of the Kv11.1 current (also known as the hERG current or IKr) and on the use of non-human cells or tissues. These new strategies have the potential to improve sensitivity and specificity in the early detection of genuine cardiotoxicity risks, thereby reducing the likelihood of mistakenly discarding viable drug candidates and speeding the progression of worthy drugs into clinical trials.
- Published
- 2016
- Full Text
- View/download PDF
45. A New Perspective in the Field of Cardiac Safety Testing through the Comprehensive In Vitro Proarrhythmia Assay Paradigm.
- Author
-
Fermini B, Hancox JC, Abi-Gerges N, Bridgland-Taylor M, Chaudhary KW, Colatsky T, Correll K, Crumb W, Damiano B, Erdemli G, Gintant G, Imredy J, Koerner J, Kramer J, Levesque P, Li Z, Lindqvist A, Obejero-Paz CA, Rampe D, Sawada K, Strauss DG, and Vandenberg JI
- Subjects
- Animals, Humans, Long QT Syndrome chemically induced, Long QT Syndrome diagnosis, Torsades de Pointes chemically induced, Torsades de Pointes diagnosis, Arrhythmias, Cardiac chemically induced, Arrhythmias, Cardiac diagnosis, Drug-Related Side Effects and Adverse Reactions diagnosis, Drug-Related Side Effects and Adverse Reactions etiology, Heart drug effects
- Abstract
For the past decade, cardiac safety screening to evaluate the propensity of drugs to produce QT interval prolongation and Torsades de Pointes (TdP) arrhythmia has been conducted according to ICH S7B and ICH E14 guidelines. Central to the existing approach are hERG channel assays and in vivo QT measurements. Although effective, the present paradigm carries a risk of unnecessary compound attrition and high cost, especially when considering costly thorough QT (TQT) studies conducted later in drug development. The C: omprehensive I: n Vitro P: roarrhythmia A: ssay (CiPA) initiative is a public-private collaboration with the aim of updating the existing cardiac safety testing paradigm to better evaluate arrhythmia risk and remove the need for TQT studies. It is hoped that CiPA will produce a standardized ion channel assay approach, incorporating defined tests against major cardiac ion channels, the results of which then inform evaluation of proarrhythmic actions in silico, using human ventricular action potential reconstructions. Results are then to be confirmed using human (stem cell-derived) cardiomyocytes. This perspective article reviews the rationale, progress of, and challenges for the CiPA initiative, if this new paradigm is to replace existing practice and, in time, lead to improved and widely accepted cardiac safety testing guidelines., (© 2015 Society for Laboratory Automation and Screening.)
- Published
- 2016
- Full Text
- View/download PDF
46. Discovery of A-971432, An Orally Bioavailable Selective Sphingosine-1-Phosphate Receptor 5 (S1P5) Agonist for the Potential Treatment of Neurodegenerative Disorders.
- Author
-
Hobson AD, Harris CM, van der Kam EL, Turner SC, Abibi A, Aguirre AL, Bousquet P, Kebede T, Konopacki DB, Gintant G, Kim Y, Larson K, Maull JW, Moore NS, Shi D, Shrestha A, Tang X, Zhang P, and Sarris KK
- Subjects
- Administration, Oral, Animals, Azetidinecarboxylic Acid administration & dosage, Azetidinecarboxylic Acid chemistry, Azetidinecarboxylic Acid pharmacokinetics, Azetidinecarboxylic Acid pharmacology, Benzene Derivatives administration & dosage, Benzene Derivatives pharmacokinetics, Blood-Brain Barrier drug effects, Blood-Brain Barrier metabolism, Blood-Brain Barrier pathology, Cell Line, Cognitive Aging, Dogs, Female, Humans, Macaca fascicularis, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Rats, Rats, Sprague-Dawley, Receptors, Lysosphingolipid metabolism, Azetidinecarboxylic Acid analogs & derivatives, Benzene Derivatives chemistry, Benzene Derivatives pharmacology, Neurodegenerative Diseases drug therapy, Receptors, Lysosphingolipid agonists
- Abstract
S1P5 is one of 5 receptors for sphingosine-1-phosphate and is highly expressed on endothelial cells within the blood-brain barrier, where it maintains barrier integrity in in vitro models (J. Neuroinflamm. 2012, 9, 133). Little more is known about the effects of S1P5 modulation due to the absence of tool molecules with suitable selectivity and drug-like properties. We recently reported that molecule A-971432 (Harris, 2010) (29 in this paper) is highly efficacious in reversing lipid accumulation and age-related cognitive decline in rats (Van der Kam , , AAIC 2014). Herein we describe the development of a series of selective S1P5 agonists that led to the identification of compound 29, which is highly selective for S1P5 and has excellent plasma and CNS exposure after oral dosing in preclinical species. To further support its suitability for in vivo studies of S1P5 biology, we extensively characterized 29, including confirmation of its selectivity in pharmacodynamic assays of S1P1 and S1P3 function in rats. In addition, we found that 29 improves blood-brain barrier integrity in an in vitro model and reverses age-related cognitive decline in mice. These results suggest that S1P5 agonism is an innovative approach with potential benefit in neurodegenerative disorders involving lipid imbalance and/or compromised blood-brain barrier such as Alzheimer's disease or multiple sclerosis.
- Published
- 2015
- Full Text
- View/download PDF
47. In Vitro Early Safety Pharmacology Screening: Perspectives Related to Cardiovascular Safety.
- Author
-
Gintant G
- Subjects
- Calcium Channels, L-Type drug effects, Cardiotoxicity, Drug Discovery, Drug Industry, Humans, In Vitro Techniques, Risk Assessment, Safety, Sodium Channels drug effects, Drug Evaluation, Preclinical methods, Heart drug effects
- Abstract
In vitro screening for cardiovascular safety liabilities of novel drug candidates presents a challenge for the pharmaceutical industry. Such approaches rely on detecting pharmacologic effects on key components of complex integrated system early in drug discovery to define potential safety liabilities. Key to such studies are the concepts of hazard identification vs. risk assessment, drug specificity vs. selectivity, and an appreciation of the challenges faced when attempting to translate in vitro findings to preclinical in vivo as well as clinical effects. This chapter defines some key aspects of early safety pharmacology screening for cardiovascular liabilities, citing studies of two key depolarizing cardiac currents (fast sodium current and L-type calcium current) as examples linked to effects on cardiac conduction and repolarization.
- Published
- 2015
- Full Text
- View/download PDF
48. Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium.
- Author
-
Sager PT, Gintant G, Turner JR, Pettit S, and Stockbridge N
- Subjects
- Arrhythmias, Cardiac prevention & control, Drug-Related Side Effects and Adverse Reactions prevention & control, Humans, Long QT Syndrome chemically induced, Long QT Syndrome prevention & control, Risk Assessment, Torsades de Pointes chemically induced, Torsades de Pointes prevention & control, United States, Arrhythmias, Cardiac chemically induced, Drug Evaluation, Preclinical methods, Patch-Clamp Techniques methods
- Abstract
This white paper provides a summary of a scientific proposal presented at a Cardiac Safety Research Consortium/Health and Environmental Sciences Institute/Food and Drug Administration-sponsored Think Tank, held at Food and Drug Administration's White Oak facilities, Silver Spring, MD, on July 23, 2013, with the intention of moving toward consensus on defining a new paradigm in the field of cardiac safety in which proarrhythmic risk would be primarily assessed using nonclinical in vitro human models based on solid mechanistic considerations of torsades de pointes proarrhythmia. This new paradigm would shift the emphasis from the present approach that strongly relies on QTc prolongation (a surrogate marker of proarrhythmia) and could obviate the clinical Thorough QT study during later drug development. These discussions represent current thinking and suggestions for furthering our knowledge and understanding of the public health case for adopting a new, integrated nonclinical in vitro/in silico paradigm, the Comprehensive In Vitro Proarrhythmia Assay, for the assessment of a candidate drug's proarrhythmic liability, and for developing a public-private collaborative program to characterize the data content, quality, and approaches required to assess proarrhythmic risk in the absence of a Thorough QT study. This paper seeks to encourage multistakeholder input regarding this initiative and does not represent regulatory guidance., (Copyright © 2014 Mosby, Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
49. Assessment of drug-induced increases in blood pressure during drug development: report from the Cardiac Safety Research Consortium.
- Author
-
Sager P, Heilbraun J, Turner JR, Gintant G, Geiger MJ, Kowey PR, Mansoor GA, Mendzelevski B, Michelson EL, Stockbridge N, Weber MA, and White WB
- Subjects
- Blood Pressure Monitoring, Ambulatory, Cardiovascular Diseases chemically induced, Clinical Trials as Topic, Drug Discovery, Drug Evaluation, Preclinical, Humans, Patient Safety, Risk Assessment, Blood Pressure drug effects
- Abstract
This White Paper, prepared by members of the Cardiac Safety Research Consortium, discusses several important issues regarding the evaluation of blood pressure (BP) responses to drugs being developed for indications not of a direct cardiovascular (CV) nature. A wide range of drugs are associated with off-target BP increases, and both scientific attention and regulatory attention to this topic are increasing. The article provides a detailed summary of scientific discussions at a Cardiac Safety Research Consortium-sponsored Think Tank held on July 18, 2012, with the intention of moving toward consensus on how to most informatively collect and analyze BP data throughout clinical drug development to prospectively identify unacceptable CV risk and evaluate the benefit-risk relationship. The overall focus in on non-CV drugs, although many of the points also pertain to CV drugs. Brief consideration of how clinical assessment can be informed by nonclinical investigation is also outlined. These discussions present current thinking and suggestions for furthering our knowledge and understanding of off-target drug-induced BP increases and do not represent regulatory guidance., (Copyright © 2013 Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
50. Integrated and translational nonclinical in vivo cardiovascular risk assessment: gaps and opportunities.
- Author
-
Berridge BR, Hoffmann P, Turk JR, Sellke F, Gintant G, Hirkaler G, Dreher K, Schultze AE, Walker D, Edmunds N, Halpern W, Falls J, Sanders M, and Pettit SD
- Subjects
- Animals, Disease Models, Animal, Drug Industry methods, Endpoint Determination, Humans, MicroRNAs metabolism, Research Design, Risk Assessment methods, Species Specificity, Cardiovascular Diseases chemically induced, Drug Design, Drug-Related Side Effects and Adverse Reactions
- Abstract
Cardiovascular (CV) safety concerns are a significant source of drug development attrition in the pharmaceutical industry today. Though current nonclinical testing paradigms have largely prevented catastrophic CV events in Phase I studies, many challenges relating to the inability of current nonclinical safety testing strategies to model patient outcomes persist. Contemporary approaches include a spectrum of evaluations of CV structure and function in a variety of laboratory animal species. These approaches might be improved with a more holistic integration of these evaluations in repeat-dose studies, addition of novel endpoints with greater sensitivity and translational application, and use of more relevant animal models. Particular opportunities present with advances in imaging capabilities applicable to rodent and non-rodent species, technical capabilities for measuring CV function in repeat-dose animal studies, detection and quantitation of microRNAs and wider use of alternative animal models. Strategic application of these novel opportunities considering putative CV risk associated with the molecular drug target as well as inherent risks present in the target patient population could tailor or 'personalize' nonclinical safety assessment as a more translational evaluation. This paper is a call to action for the clinical and nonclinical drug safety communities to assess these opportunities to determine their utility in filling potential gaps in our current cardiovascular safety testing paradigms., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.