1
|
Cohen JD, You D, Sharma AK, Takai T, Hara H, Sales VT, Yukawa T, Cai B. In vitro human ion channel assays predictive of drug-induced seizure. Toxicol Sci 2025; 203:253-268. [PMID: 39661496 PMCID: PMC11775423 DOI: 10.1093/toxsci/kfae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Seizure is among the most severe FDA black box warnings of neurotoxicity reported on drug labels. Gaining a better mechanistic understanding of off-targets causative of seizure will improve the identification of potential seizure risks preclinically. In the present study, we evaluated an in vitro panel of 9 investigational (Cav2.1, Cav3.2, GlyRA1, AMPA, HCN1, Kv1.1, Kv7.2/7.3, NaV1.1, Nav1.2) and 2 standard (GABA-A, NMDA) ion channel targets with strong correlative links to seizure, using automated electrophysiology. Each target was assessed with a library of 34 preclinical compounds and 10 approved drugs with known effects of convulsion in vivo and/or in patients. Cav2.1 had the highest frequency of positive hits, 20 compounds with an EC30 or IC30 ≤ 30 µM, and the highest importance score relative to the 11 targets. An additional 35 approved drugs, with categorized low to frequent seizure risk in patients, were evaluated in the Cav2.1 assay. The Cav2.1 assay predicted preclinical compounds to cause convulsion in nonclinical species with a sensitivity of 52% and specificity of 78%, and approved drugs to cause seizure in nonclinical species or in patients with a sensitivity of 48% or 54% and specificity of 71% or 78%, respectively. The integrated panel of 11 ion channel targets predicted preclinical compounds to cause convulsion in nonclinical species with a sensitivity of 68%, specificity of 56%, and accuracy of 65%. This study highlights the utility of expanding the in vitro panel of targets evaluated for seizurogenic activity, in order to reduce compound attrition early on in drug discovery.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Dahea You
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Ashok K Sharma
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Takafumi Takai
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Hideto Hara
- Drug Safety Research & Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Vicencia T Sales
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Tomoya Yukawa
- Drug Safety Research & Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Beibei Cai
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| |
Collapse
|
2
|
Chen LC, Lee TW, Chang CH. Neurobehavioral assessment of BMEDA by modified Irwin test in Sprague-Dawley rats. Regul Toxicol Pharmacol 2024; 153:105703. [PMID: 39299678 DOI: 10.1016/j.yrtph.2024.105703] [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: 06/12/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
The neurobehavioral assessment of N,N-bis(2-mercapatoethly)-N',N'-diethylenediamine (BMEDA), which can form a chelate with rhenium-188 (188Re) to produce the 188Re-BMEDA-liposome, was evaluated. The purpose of this study was to evaluate the potential neurobehavioral changes by using the functional observational battery observation procedures when intravenous injection of BMEDA to Sprague-Dawley rats. Rats were administered BMEDA at dose levels of 1, 2, and 5 mg/kg. No mortalities were observed. There are some observations related to BMEDA treatment found in the 5 mg/kg dose group at 10 min post-dose. Tremor was observed in one male rat and seven female rats. The increased respiration, vocalization, not easy to handle and/or loss of tone in the limb were observed in both males and females, and increased body temperature was observed in male animals. Based on the results, a single intravenous dose of BMEDA administered to rats resulted in increased respiration, vocalization, not easy to handle and/or loss of tone in the limb increasing at the dose level of 5 mg/kg. No neurobehavioral effects were noted after BMEDA administration up to the dose level of 2 mg/kg. The information of this study will provides a point of reference to design appropriately therapeutic studies for future human use.
Collapse
Affiliation(s)
- Liang-Cheng Chen
- Department of Isotope Application Research, National Atomic Research Institute, 1000 Wenhua Road, Jiaan Village, Longtan District, Taoyuan, 325, Taiwan, ROC
| | - Te-Wei Lee
- Department of Isotope Application Research, National Atomic Research Institute, 1000 Wenhua Road, Jiaan Village, Longtan District, Taoyuan, 325, Taiwan, ROC
| | - Chih-Hsien Chang
- Department of Isotope Application Research, National Atomic Research Institute, 1000 Wenhua Road, Jiaan Village, Longtan District, Taoyuan, 325, Taiwan, ROC.
| |
Collapse
|
3
|
Bällgren F, Hu Y, Li S, van de Beek L, Hammarlund-Udenaes M, Loryan I. Region-independent active CNS net uptake of marketed H +/OC antiporter system substrates. Front Cell Neurosci 2024; 18:1493644. [PMID: 39534684 PMCID: PMC11554538 DOI: 10.3389/fncel.2024.1493644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The pyrilamine-sensitive proton-coupled organic cation (H+/OC) antiporter system facilitates the active net uptake of several marketed organic cationic drugs across the blood-brain barrier (BBB). This rare phenomenon has garnered interest in the H+/OC antiporter system as a potential target for CNS drug delivery. However, analysis of pharmacovigilance data has uncovered a significant association between substrates of the H+/OC antiporter and neurotoxicity, particularly drug-induced seizures (DIS) and mood- and cognitive-related adverse events (MCAEs). This preclinical study aimed to elucidate the CNS regional disposition of H+/OC antiporter substrates at therapeutically relevant plasma concentrations to uncover potential pharmacokinetic mechanisms underlying DIS and MCAEs. Here, we investigated the neuropharmacokinetics of pyrilamine, diphenhydramine, bupropion, tramadol, oxycodone, and memantine. Using the Combinatory Mapping Approach for Regions of Interest (CMA-ROI), we characterized the transport of unbound drugs across the BBB in specific CNS regions, as well as the blood-spinal cord barrier (BSCB) and the blood-cerebrospinal fluid barrier (BCSFB). Our findings demonstrated active net uptake across the BBB and BSCB, with unbound ROI-to-plasma concentration ratio, Kp,uu,ROI, values consistently exceeding unity in all assessed regions. Despite minor regional differences, no significant distinctions were found when comparing the whole brain to investigated regions of interest, indicating region-independent active transport. Furthermore, we observed intracellular accumulation via lysosomal trapping for all studied drugs. These results provide new insights into the CNS regional neuropharmacokinetics of these drugs, suggesting that while the brain uptake is region-independent, the active transport mechanism enables high extracellular and intracellular drug concentrations, potentially contributing to neurotoxicity. This finding emphasizes the necessity of thorough neuropharmacokinetic evaluation and neurotoxicity profiling in the development of drugs that utilize this transport pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Irena Loryan
- Department of Pharmacy, Faculty of Pharmacy, Translational Pharmacokinetics-Pharmacodynamics Group, Translational Pharmacokinetics Pharmacodynamics (tPKPD), Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Kreir M, Putri D, Tekle F, Pibiri F, d’Ydewalle C, Van Ammel K, Geys H, Teisman A, Gallacher DJ, Lu HR. Development of a new hazard scoring system in primary neuronal cell cultures for drug-induced acute neuronal toxicity identification in early drug discovery. Front Pharmacol 2024; 15:1308547. [PMID: 38873414 PMCID: PMC11170107 DOI: 10.3389/fphar.2024.1308547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 05/03/2024] [Indexed: 06/15/2024] Open
Abstract
We investigated drug-induced acute neuronal electrophysiological changes using Micro-Electrode arrays (MEA) to rat primary neuronal cell cultures. Data based on 6-key MEA parameters were analyzed for plate-to-plate vehicle variability, effects of positive and negative controls, as well as data from over 100 reference drugs, mostly known to have pharmacological phenotypic and clinical outcomes. A Least Absolute Shrinkage and Selection Operator (LASSO) regression, coupled with expert evaluation helped to identify the 6-key parameters from many other MEA parameters to evaluate the drug-induced acute neuronal changes. Calculating the statistical tolerance intervals for negative-positive control effects on those 4-key parameters helped us to develop a new weighted hazard scoring system on drug-induced potential central nervous system (CNS) adverse effects (AEs). The weighted total score, integrating the effects of a drug candidate on the identified six-pivotal parameters, simply determines if the testing compound/concentration induces potential CNS AEs. Hereto, it uses four different categories of hazard scores: non-neuroactive, neuroactive, hazard, or high hazard categories. This new scoring system was successfully applied to differentiate the new compounds with or without CNS AEs, and the results were correlated with the outcome of in vivo studies in mice for one internal program. Furthermore, the Random Forest classification method was used to obtain the probability that the effect of a compound is either inhibitory or excitatory. In conclusion, this new neuronal scoring system on the cell assay is actively applied in the early de-risking of drug development and reduces the use of animals and associated costs.
Collapse
Affiliation(s)
- Mohamed Kreir
- Global Toxicology and Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen Research and Development, Beerse, Belgium
| | - Dea Putri
- Statistics and Decision Sciences, Global Development, Janssen Research and Development, Beerse, Belgium
| | - Fetene Tekle
- Statistics and Decision Sciences, Global Development, Janssen Research and Development, Beerse, Belgium
| | - Francesca Pibiri
- Global Toxicology and Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen Research and Development, Beerse, Belgium
| | | | - Karel Van Ammel
- Global Toxicology and Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen Research and Development, Beerse, Belgium
| | - Helena Geys
- Statistics and Decision Sciences, Global Development, Janssen Research and Development, Beerse, Belgium
| | - Ard Teisman
- Global Toxicology and Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen Research and Development, Beerse, Belgium
| | - David J. Gallacher
- Global Toxicology and Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen Research and Development, Beerse, Belgium
| | - Hua Rong Lu
- Global Toxicology and Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen Research and Development, Beerse, Belgium
| |
Collapse
|
5
|
Orciani C, Ballesteros C, Troncy E, Berthome C, Bujold K, Bennamoune N, Sparapani S, Pugsley MK, Paquette D, Boulay E, Authier S. The Spontaneous Incidence of Neurological Clinical Signs in Preclinical Species Using Cage-side Observations or High-definition Video Monitoring: A Retrospective Analysis. Int J Toxicol 2024; 43:123-133. [PMID: 38063479 DOI: 10.1177/10915818231218984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
When conducting toxicology studies, the interpretation of drug-related neurological clinical signs such as convulsions, myoclonus/myoclonic jerks, tremors, ataxia, and salivation requires an understanding of the spontaneous incidence of those observations in commonly used laboratory animal species. The spontaneous incidence of central nervous system clinical signs in control animals from a single facility using cage-side observations or high definition video monitoring was retrospectively analyzed. Spontaneous convulsions were observed at low incidence in Beagle dogs and Sprague-Dawley rats but were not identified in cynomolgus monkeys and Göttingen minipigs. Spontaneous myoclonic jerks and muscle twitches were observed at low incidence in Beagle dogs, cynomolgus monkeys, and Sprague-Dawley rats but were not seen in Göttingen minipigs. Spontaneous ataxia/incoordination was identified in all species and generally with a higher incidence when using video monitoring. Salivation and tremors were the two most frequent spontaneous clinical signs and both were observed in all species. Data from the current study unveil potential limitations when using control data obtained from a single study for toxicology interpretation related to low incidence neurological clinical signs while providing historical control data from Beagle dogs, cynomolgus monkeys, Sprague-Dawley rats, and Göttingen minipigs.
Collapse
Affiliation(s)
| | | | - Eric Troncy
- GREPAQ, Faculté de Médecine Vétérinaire, Universite de Montreal, Saint Hyacinthe, QC, Canada
| | | | | | | | | | | | - Dominique Paquette
- GREPAQ, Faculté de Médecine Vétérinaire, Universite de Montreal, Saint Hyacinthe, QC, Canada
| | - Emmanuel Boulay
- Charles River, Laval, QC, Canada
- GREPAQ, Faculté de Médecine Vétérinaire, Universite de Montreal, Saint Hyacinthe, QC, Canada
| | - Simon Authier
- Charles River, Laval, QC, Canada
- GREPAQ, Faculté de Médecine Vétérinaire, Universite de Montreal, Saint Hyacinthe, QC, Canada
| |
Collapse
|
6
|
Rockley K, Roberts R, Jennings H, Jones K, Davis M, Levesque P, Morton M. An integrated approach for early in vitro seizure prediction utilizing hiPSC neurons and human ion channel assays. Toxicol Sci 2023; 196:126-140. [PMID: 37632788 DOI: 10.1093/toxsci/kfad087] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2023] Open
Abstract
Seizure liability remains a significant cause of attrition throughout drug development. Advances in stem cell biology coupled with an increased understanding of the role of ion channels in seizure offer an opportunity for a new paradigm in screening. We assessed the activity of 15 pro-seizurogenic compounds (7 CNS active therapies, 4 GABA receptor antagonists, and 4 other reported seizurogenic compounds) using automated electrophysiology against a panel of 14 ion channels (Nav1.1, Nav1.2, Nav1.6, Kv7.2/7.3, Kv7.3/7.5, Kv1.1, Kv4.2, KCa4.1, Kv2.1, Kv3.1, KCa1.1, GABA α1β2γ2, nicotinic α4β2, NMDA 1/2A). These were selected based on linkage to seizure in genetic/pharmacological studies. Fourteen compounds demonstrated at least one "hit" against the seizure panel and 11 compounds inhibited 2 or more ion channels. Next, we assessed the impact of the 15 compounds on electrical signaling using human-induced pluripotent stem cell neurons in microelectrode array (MEA). The CNS active therapies (amoxapine, bupropion, chlorpromazine, clozapine, diphenhydramine, paroxetine, quetiapine) all caused characteristic changes to electrical activity in key parameters indicative of seizure such as network burst frequency and duration. The GABA antagonist picrotoxin increased all parameters, but the antibiotics amoxicillin and enoxacin only showed minimal changes. Acetaminophen, included as a negative control, caused no changes in any of the parameters assessed. Overall, pro-seizurogenic compounds showed a distinct fingerprint in the ion channel/MEA panel. These studies highlight the potential utility of an integrated in vitro approach for early seizure prediction to provide mechanistic information and to support optimal drug design in early development, saving time and resources.
Collapse
Affiliation(s)
| | - Ruth Roberts
- ApconiX, Macclesfield SK10 4TG, UK
- Department of Biosciences, University of Birmingham, Edgbaston B15 1TT, UK
| | | | | | - Myrtle Davis
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | | |
Collapse
|
7
|
Zhai J, Traebert M, Zimmermann K, Delaunois A, Royer L, Salvagiotto G, Carlson C, Lagrutta A. Comparative study for the IMI2-NeuroDeRisk project on microelectrode arrays to derisk drug-induced seizure liability. J Pharmacol Toxicol Methods 2023; 123:107297. [PMID: 37499956 DOI: 10.1016/j.vascn.2023.107297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/01/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION In the framework of the IMI2-NeuroDeRisk consortium, three in vitro electrophysiology assays were compared to improve preclinical prediction of seizure-inducing liabilities. METHODS Two cell models, primary rat cortical neurons and human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons co-cultured with hiPSC-derived astrocytes were tested on two different microelectrode array (MEA) platforms, Maestro Pro (Axion Biosystems) and Multiwell-MEA-System (Multi Channel Systems), in three separate laboratories. Pentylenetetrazole (PTZ) and/or picrotoxin (PTX) were included in each plate as positive (n = 3-6 wells) and ≤0.2% DMSO was used as negative controls (n = 3-12 wells). In general, concentrations in a range of 0.1-30 μM were tested, anchored, when possible, on clinically relevant exposures (unbound Cmax) were tested. Activity thresholds for drug-induced changes were set at 20%. To evaluate sensitivity, specificity and predictivity of the cell models, seizurogenic responses were defined as changes in 4 or more endpoints. Concentration dependence trends were also considered. RESULTS Neuronal activity of 33 compounds categorized as positive tool drugs, seizure-positive or seizure-negative compounds was evaluated. Acute drug effects (<60 min) were compared to baseline recordings. Time points < 15 min exhibited stronger, less variable responses to many of the test agents. For many compounds a reduction and cessation of neuronal activity was detected at higher test concentrations. There was not a single pattern of seizurogenic activity detected, even among tool compounds, likely due to different mechanisms of actions and/or off-target profiles. A post-hoc analysis focusing on changes indicative of neuronal excitation is presented. CONCLUSION All cell models showed good sensitivity, ranging from 70 to 86%. Specificity ranged from 40 to 70%. Compared to more conventional measurements of evoked activity in hippocampal slices, these plate-based models provide higher throughput and the potential to study subacute responses. Yet, they may be limited by the random, spontaneous nature of their network activity.
Collapse
Affiliation(s)
- Jin Zhai
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | | | | | - Coby Carlson
- Fujifilm Cellular Dynamics, Inc., Madison, WI, USA
| | | |
Collapse
|
8
|
Utsumi Y, Taketoshi M, Miwa M, Tominaga Y, Tominaga T. Assessing seizure liability in vitro with voltage-sensitive dye imaging in mouse hippocampal slices. Front Cell Neurosci 2023; 17:1217368. [PMID: 37680865 PMCID: PMC10481167 DOI: 10.3389/fncel.2023.1217368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Non-clinical toxicology is a major cause of drug candidate attrition during development. In particular, drug-induced seizures are the most common finding in central nervous system (CNS) toxicity. Current safety pharmacology tests for assessing CNS functions are often inadequate in detecting seizure-inducing compounds early in drug development, leading to significant delays. This paper presents an in vitro seizure liability assay using voltage-sensitive dye (VSD) imaging techniques in hippocampal brain slices, offering a powerful alternative to traditional electrophysiological methods. Hippocampal slices were isolated from mice, and VSD optical responses evoked by stimulating the Schaffer collateral pathway were recorded and analyzed in the stratum radiatum (SR) and stratum pyramidale (SP). VSDs allow for the comprehensive visualization of neuronal action potentials and postsynaptic potentials on a millisecond timescale. By employing this approach, we investigated the in vitro drug-induced seizure liability of representative pro-convulsant compounds. Picrotoxin (PiTX; 1-100 μM), gabazine (GZ; 0.1-10 μM), and 4-aminopyridine (4AP; 10-100 μM) exhibited seizure-like responses in the hippocampus, but pilocarpine hydrochloride (Pilo; 10-100 μM) did not. Our findings demonstrate the potential of VSD-based assays in identifying seizurogenic compounds during early drug discovery, thereby reducing delays in drug development and providing insights into the mechanisms underlying seizure induction and the associated risks of pro-convulsant compounds.
Collapse
Affiliation(s)
- Yuichi Utsumi
- Graduate School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Japan
- Institute of Neuroscience, Tokushima Bunri University, Sanuki, Japan
| | - Makiko Taketoshi
- Institute of Neuroscience, Tokushima Bunri University, Sanuki, Japan
| | - Michiko Miwa
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Japan
| | - Yoko Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki, Japan
| | - Takashi Tominaga
- Graduate School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Japan
- Institute of Neuroscience, Tokushima Bunri University, Sanuki, Japan
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Japan
| |
Collapse
|
9
|
Lu HR, Seo M, Kreir M, Tanaka T, Yamoto R, Altrocchi C, van Ammel K, Tekle F, Pham L, Yao X, Teisman A, Gallacher DJ. High-Throughput Screening Assay for Detecting Drug-Induced Changes in Synchronized Neuronal Oscillations and Potential Seizure Risk Based on Ca 2+ Fluorescence Measurements in Human Induced Pluripotent Stem Cell (hiPSC)-Derived Neuronal 2D and 3D Cultures. Cells 2023; 12:cells12060958. [PMID: 36980298 PMCID: PMC10046961 DOI: 10.3390/cells12060958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Drug-induced seizure liability is a significant safety issue and the basis for attrition in drug development. Occurrence in late development results in increased costs, human risk, and delayed market availability of novel therapeutics. Therefore, there is an urgent need for biologically relevant, in vitro high-throughput screening assays (HTS) to predict potential risks for drug-induced seizure early in drug discovery. We investigated drug-induced changes in neural Ca2+ oscillations, using fluorescent dyes as a potential indicator of seizure risk, in hiPSC-derived neurons co-cultured with human primary astrocytes in both 2D and 3D forms. The dynamics of synchronized neuronal calcium oscillations were measured with an FDSS kinetics reader. Drug responses in synchronized Ca2+ oscillations were recorded in both 2D and 3D hiPSC-derived neuron/primary astrocyte co-cultures using positive controls (4-aminopyridine and kainic acid) and negative control (acetaminophen). Subsequently, blinded tests were carried out for 25 drugs with known clinical seizure incidence. Positive predictive value (accuracy) based on significant changes in the peak number of Ca2+ oscillations among 25 reference drugs was 91% in 2D vs. 45% in 3D hiPSC-neuron/primary astrocyte co-cultures. These data suggest that drugs that alter neuronal activity and may have potential risk for seizures can be identified with high accuracy using an HTS approach using the measurements of Ca2+ oscillations in hiPSC-derived neurons co-cultured with primary astrocytes in 2D.
Collapse
Affiliation(s)
- Hua-Rong Lu
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Manabu Seo
- Elixirgen Scientific, Incorporated, Baltimore, MD 21205, USA
| | - Mohamed Kreir
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Tetsuya Tanaka
- Elixirgen Scientific, Incorporated, Baltimore, MD 21205, USA
| | - Rie Yamoto
- Healthcare Business Group, Drug Discovery Business Department, Ricoh Company Ltd., Tokyo 143-8555, Japan
| | - Cristina Altrocchi
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Karel van Ammel
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Fetene Tekle
- Statistics and Decision Sciences, Global Development, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Ly Pham
- Computational Biology & Toxicology, Preclinical Sciences and Translational Safety, A Division of Janssen Pharmaceutica NV, San Diego, CA 921921, USA
| | - Xiang Yao
- Computational Biology & Toxicology, Preclinical Sciences and Translational Safety, A Division of Janssen Pharmaceutica NV, San Diego, CA 921921, USA
| | - Ard Teisman
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - David J Gallacher
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| |
Collapse
|
10
|
Enterovirus D-68 Infection of Primary Rat Cortical Neurons: Entry, Replication, and Functional Consequences. mBio 2023; 14:e0024523. [PMID: 36877033 PMCID: PMC10127580 DOI: 10.1128/mbio.00245-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Enterovirus D68 (EV-D68) is an emerging pathogen associated with mild to severe respiratory disease. Since 2014, EV-D68 is also linked to acute flaccid myelitis (AFM), causing paralysis and muscle weakness in children. However, it remains unclear whether this is due to an increased pathogenicity of contemporary EV-D68 clades or increased awareness and detection of this virus. Here, we describe an infection model of primary rat cortical neurons to study the entry, replication, and functional consequences of different EV-D68 strains, including historical and contemporary strains. We demonstrate that sialic acids are important (co)receptors for infection of both neurons and respiratory epithelial cells. Using a collection of glycoengineered isogenic HEK293 cell lines, we show that sialic acids on either N-glycans or glycosphingolipids can be used for infection. Additionally, we show that both excitatory glutamatergic and inhibitory GABA-ergic neurons are susceptible and permissive to historical and contemporary EV-D68 strains. EV-D68 infection of neurons leads to the reorganization of the Golgi-endomembranes forming replication organelles, first in the soma and later in the processes. Finally, we demonstrate that the spontaneous neuronal activity of EV-D68-infected neuronal network cultured on microelectrode arrays (MEA) is decreased, independent of the virus strain. Collectively, our findings provide novel insights into neurotropism and -pathology of different EV-D68 strains, and argue that it is unlikely that increased neurotropism is a recently acquired phenotype of a specific genetic lineage. IMPORTANCE Acute flaccid myelitis (AFM) is a serious neurological illness characterized by muscle weakness and paralysis in children. Since 2014, outbreaks of AFM have emerged worldwide, and they appear to be caused by nonpolio enteroviruses, particularly enterovirus-D68 (EV-D68), an unusual enterovirus that is known to mainly cause respiratory disease. It is unknown whether these outbreaks reflect a change of EV-D68 pathogenicity or are due to increased detection and awareness of this virus in recent years. To gain more insight herein, it is crucial to define how historical and circulating EV-D68 strains infect and replicate in neurons and how they affect their physiology. This study compares the entry and replication in neurons and the functional consequences on the neural network upon infection with an old "historical" strain and contemporary "circulating" strains of EV-D68.
Collapse
|
11
|
Pressman P, Clemens R, Hayes AW. Significant shifts in preclinical and clinical neurotoxicology: a review and commentary. Toxicol Mech Methods 2023; 33:173-182. [PMID: 35920262 DOI: 10.1080/15376516.2022.2109228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The ever-expanding prevalence of adverse neurotoxic reactions of the brain in response to therapeutic and recreational drugs, dietary supplements, environmental hazards, cosmetic ingredients, a spectrum of herbals, health status, and environmental stressors continues to prompt the development of novel cell-based assays to better determine neurotoxic hazard. Neurotoxicants may cause direct and epigenetic damage to the nervous tissue and alter the chemistry, structure, or normal activity of the nervous system. In severe neurotoxicity due to exposure to physical or psychosocial toxicants, neurons are disrupted or killed, and a consistent pattern of clinical neural dysfunction appears. In utero exposure to neurotoxicants can lead to altered development of the nervous system [developmental neurotoxicity (DNT)]. Patients with certain disorders and certain genomic makeup may be particularly susceptible to neurotoxicants. Traditional cytotoxicity measurements, like cell death, are easy to measure, but insufficient at identifying current routine biomarkers of toxicity including functional impairment in cell communication, which often occurs before or even in the absence of cell death. The present paper examines some of the limitations of existing neurotoxicology in light of the increasing need to develop tools to meet the challenges of achieving greater sensitivity in detection and developing and standardizing methods for exploring the toxicologic risk of such neurotoxic entities as engineered nanomaterials and even variables associated with poverty.
Collapse
Affiliation(s)
- Peter Pressman
- Clinical Medicine, Saba University School of Medicine, The Bottom, Caribbean, The Netherlands
| | - Roger Clemens
- School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - A Wallace Hayes
- College of Public Health, University of South Florida, Tampa, FL, USA
| |
Collapse
|
12
|
Lipponen A, Kajevu N, Natunen T, Ciszek R, Puhakka N, Hiltunen M, Pitkänen A. Gene Expression Profile as a Predictor of Seizure Liability. Int J Mol Sci 2023; 24:ijms24044116. [PMID: 36835526 PMCID: PMC9963992 DOI: 10.3390/ijms24044116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Analysis platforms to predict drug-induced seizure liability at an early phase of drug development would improve safety and reduce attrition and the high cost of drug development. We hypothesized that a drug-induced in vitro transcriptomics signature predicts its ictogenicity. We exposed rat cortical neuronal cultures to non-toxic concentrations of 34 compounds for 24 h; 11 were known to be ictogenic (tool compounds), 13 were associated with a high number of seizure-related adverse event reports in the clinical FDA Adverse Event Reporting System (FAERS) database and systematic literature search (FAERS-positive compounds), and 10 were known to be non-ictogenic (FAERS-negative compounds). The drug-induced gene expression profile was assessed from RNA-sequencing data. Transcriptomics profiles induced by the tool, FAERS-positive and FAERS-negative compounds, were compared using bioinformatics and machine learning. Of the 13 FAERS-positive compounds, 11 induced significant differential gene expression; 10 of the 11 showed an overall high similarity to the profile of at least one tool compound, correctly predicting the ictogenicity. Alikeness-% based on the number of the same differentially expressed genes correctly categorized 85%, the Gene Set Enrichment Analysis score correctly categorized 73%, and the machine-learning approach correctly categorized 91% of the FAERS-positive compounds with reported seizure liability currently in clinical use. Our data suggest that the drug-induced gene expression profile could be used as a predictive biomarker for seizure liability.
Collapse
Affiliation(s)
- Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, P.O. Box 95, FIN-70701 Kuopio, Finland
| | - Natallie Kajevu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Robert Ciszek
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Noora Puhakka
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
- Correspondence: ; Tel.: +358-50-517-2091; Fax: +358-17-16-3030
| |
Collapse
|
13
|
de Leeuw VC, van Oostrom CTM, Wackers PFK, Pennings JLA, Hodemaekers HM, Piersma AH, Hessel EVS. Neuronal differentiation pathways and compound-induced developmental neurotoxicity in the human neural progenitor cell test (hNPT) revealed by RNA-seq. CHEMOSPHERE 2022; 304:135298. [PMID: 35700809 PMCID: PMC9247748 DOI: 10.1016/j.chemosphere.2022.135298] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 05/27/2023]
Abstract
There is an increased awareness that the use of animals for compound-induced developmental neurotoxicity (DNT) testing has limitations. Animal-free innovations, especially the ones based on human stem cell-based models are pivotal in studying DNT since they can mimic processes relevant to human brain development. Here we present the human neural progenitor test (hNPT), a 10-day protocol in which neural progenitor cells differentiate into a neuron-astrocyte co-culture. The study aimed to characterise differentiation over time and to find neurodevelopmental processes sensitive to compound exposure using transcriptomics. 3992 genes regulated in unexposed control cultures (p ≤ 0.001, log2FC ≥ 1) showed Gene Ontology (GO-) term enrichment for neuronal and glial differentiation, neurite extension, synaptogenesis, and synaptic transmission. Exposure to known or suspected DNT compounds (acrylamide, chlorpyrifos, fluoxetine, methyl mercury, or valproic acid) at concentrations resulting in 95% cell viability each regulated unique combinations of GO-terms relating to neural progenitor proliferation, neuronal and glial differentiation, axon development, synaptogenesis, synaptic transmission, and apoptosis. Investigation of the GO-terms 'neuron apoptotic process' and 'axon development' revealed common genes that were responsive across compounds, and might be used as biomarkers for DNT. The GO-term 'synaptic signalling', on the contrary, whilst also responsive to all compounds tested, showed little overlap in gene expression regulation patterns between the conditions. This GO-term may articulate compound-specific effects that may be relevant for revealing differences in mechanism of toxicity. Given its focus on neural progenitor cell to mature multilineage neuronal cell maturation and its detailed molecular readout based on gene expression analysis, hNPT might have added value as a tool for neurodevelopmental toxicity testing in vitro. Further assessment of DNT-specific biomarkers that represent these processes needs further studies.
Collapse
Affiliation(s)
- Victoria C de Leeuw
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | - Conny T M van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Paul F K Wackers
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Hennie M Hodemaekers
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Ellen V S Hessel
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
14
|
Crofton KM, Bassan A, Behl M, Chushak YG, Fritsche E, Gearhart JM, Marty MS, Mumtaz M, Pavan M, Ruiz P, Sachana M, Selvam R, Shafer TJ, Stavitskaya L, Szabo DT, Szabo ST, Tice RR, Wilson D, Woolley D, Myatt GJ. Current status and future directions for a neurotoxicity hazard assessment framework that integrates in silico approaches. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 22:100223. [PMID: 35844258 PMCID: PMC9281386 DOI: 10.1016/j.comtox.2022.100223] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Neurotoxicology is the study of adverse effects on the structure or function of the developing or mature adult nervous system following exposure to chemical, biological, or physical agents. The development of more informative alternative methods to assess developmental (DNT) and adult (NT) neurotoxicity induced by xenobiotics is critically needed. The use of such alternative methods including in silico approaches that predict DNT or NT from chemical structure (e.g., statistical-based and expert rule-based systems) is ideally based on a comprehensive understanding of the relevant biological mechanisms. This paper discusses known mechanisms alongside the current state of the art in DNT/NT testing. In silico approaches available today that support the assessment of neurotoxicity based on knowledge of chemical structure are reviewed, and a conceptual framework for the integration of in silico methods with experimental information is presented. Establishing this framework is essential for the development of protocols, namely standardized approaches, to ensure that assessments of NT and DNT based on chemical structures are generated in a transparent, consistent, and defendable manner.
Collapse
Affiliation(s)
| | - Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Mamta Behl
- Division of the National Toxicology Program, National
Institutes of Environmental Health Sciences, Durham, NC 27709, USA
| | - Yaroslav G. Chushak
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental
Medicine & Medical Faculty Heinrich-Heine-University, Düsseldorf,
Germany
| | - Jeffery M. Gearhart
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | | | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Patricia Ruiz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Magdalini Sachana
- Environment Health and Safety Division, Environment
Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775
Paris Cedex 16, France
| | - Rajamani Selvam
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | - Timothy J. Shafer
- Biomolecular and Computational Toxicology Division, Center
for Computational Toxicology and Exposure, US EPA, Research Triangle Park, NC,
USA
| | - Lidiya Stavitskaya
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | | | | | | | - Dan Wilson
- The Dow Chemical Company, Midland, MI 48667, USA
| | | | - Glenn J. Myatt
- Instem, Columbus, OH 43215, USA
- Corresponding author.
(G.J. Myatt)
| |
Collapse
|
15
|
Matsuda N, Odawara A, Kinoshita K, Okamura A, Shirakawa T, Suzuki I. Raster plots machine learning to predict the seizure liability of drugs and to identify drugs. Sci Rep 2022; 12:2281. [PMID: 35145132 PMCID: PMC8831568 DOI: 10.1038/s41598-022-05697-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 01/03/2022] [Indexed: 11/17/2022] Open
Abstract
In vitro microelectrode array (MEA) assessment using human induced pluripotent stem cell (iPSC)-derived neurons holds promise as a method of seizure and toxicity evaluation. However, there are still issues surrounding the analysis methods used to predict seizure and toxicity liability as well as drug mechanisms of action. In the present study, we developed an artificial intelligence (AI) capable of predicting the seizure liability of drugs and identifying drugs using deep learning based on raster plots of neural network activity. The seizure liability prediction AI had a prediction accuracy of 98.4% for the drugs used to train it, classifying them correctly based on their responses as either seizure-causing compounds or seizure-free compounds. The AI also made concentration-dependent judgments of the seizure liability of drugs that it was not trained on. In addition, the drug identification AI implemented using the leave-one-sample-out scheme could distinguish among 13 seizure-causing compounds as well as seizure-free compound responses, with a mean accuracy of 99.9 ± 0.1% for all drugs. These AI prediction models are able to identify seizure liability concentration-dependence, rank the level of seizure liability based on the seizure liability probability, and identify the mechanism of the action of compounds. This holds promise for the future of in vitro MEA assessment as a powerful, high-accuracy new seizure liability prediction method.
Collapse
Affiliation(s)
- N Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - A Odawara
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - K Kinoshita
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - A Okamura
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - T Shirakawa
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - I Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan.
| |
Collapse
|
16
|
A kainic acid-induced seizure model in human pluripotent stem cell-derived cortical neurons for studying the role of IL-6 in the functional activity. Stem Cell Res 2022; 60:102665. [DOI: 10.1016/j.scr.2022.102665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/20/2022] Open
|
17
|
Any behavioral change may have physiological significance: Benign neglect in tier I neurotoxicity testing. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Zhai J, Zhou YY, Lagrutta A. Sensitivity, specificity and limitation of in vitro hippocampal slice and neuron-based assays for assessment of drug-induced seizure liability. Toxicol Appl Pharmacol 2021; 430:115725. [PMID: 34536444 DOI: 10.1016/j.taap.2021.115725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
An effective in vitro screening assay to detect seizure liability in preclinical development can contribute to better lead molecule optimization prior to candidate selection, providing higher throughput and overcoming potential brain exposure limitations in animal studies. This study explored effects of 26 positive and 14 negative reference pharmacological agents acting through different mechanisms, including 18 reference agents acting on glutamate signaling pathways, in a brain slice assay (BSA) of adult rat to define the assay's sensitivity, specificity, and limitations. Evoked population spikes (PS) were recorded from CA1 pyramidal neurons of hippocampus (HPC) in the BSA. Endpoints for analysis were PS area and PS number. Most positive references (24/26) elicited a concentration-dependent increase in PS area and/or PS number. The negative references (14/14) had little effect on the PS. Moreover, we studied the effects of 15 reference agents testing positive in the BSA on spontaneous activity in E18 rat HPC neurons monitored with microelectrode arrays (MEA), and compared these effects to the BSA results. From these in vitro studies we conclude that the BSA provides 93% sensitivity and 100% specificity in prediction of drug-induced seizure liability, including detecting seizurogenicity by 3 groups of metabotropic glutamate receptor (mGluR) ligands. The MEA results seemed more variable, both quantitatively and directionally, particularly for endpoints capturing synchronized electrical activity. We discuss these results from the two models, comparing each with published results, and provide potential explanations for differences and future directions.
Collapse
Affiliation(s)
- Jin Zhai
- Department of Genetic Toxicology and In Vitro Cellular Toxicity, Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA 19486, USA.
| | - Ying-Ying Zhou
- Program Discovery and Development, Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA 19486, USA
| | - Armando Lagrutta
- Program Discovery and Development, Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA 19486, USA
| |
Collapse
|
19
|
Authier S, Brock WJ, Halpern W, Harris SN, Jones D, McGovern T, McGovern PD, Pugsley MK. Current Trends of Practices in Nonclinical Toxicology: An Industry Survey. Int J Toxicol 2021; 40:487-505. [PMID: 34569357 DOI: 10.1177/10915818211043435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The growth in drug development over the past years reflects significant advancements in basic sciences and a greater understanding of molecular pathways of disease. Benchmarking industry practices has been important to enable a critical reflection on the path to evolve pharmaceutical testing, and the outcome of past industry surveys has had some impact on best practices in testing. A survey was provided to members of SPS, ACT, and STP. The survey consisted of 37 questions and was provided to 2550 participants with a response rate of 24%. Most respondents (∼75%) came from the US and Europe. The survey encompassed multiple topics encountered in nonclinical testing of pharmaceuticals. The most frequent target indications were oncology (69%), inflammation (55%), neurology/psychiatry/pain (46%), cardiovascular (44%), and metabolic diseases (39%). The most frequent drug-induced toxicology issues confronted were hepatic, hematopoietic, and gastrointestinal. Toxicological effects that impacted the no observed adverse effect level (NOAEL) were most frequently based on histopathology findings. The survey comprised topics encountered in the use of biomarkers in nonclinical safety assessment, most commonly those used to assess inflammation, cardiac/vascular, renal, and hepatic toxicity as well as common practices related to the assessment of endocrine effects, carcinogenicity, genotoxicity, juvenile and male-mediated developmental and female reproductive toxicity. The survey explored the impact of regulatory meetings on program design, application of the 3 Rs, and reasons for program delays. Overall, the survey results provide a broad perspective of current practices based on the experience of the scientific community engaged in nonclinical safety assessment.
Collapse
Affiliation(s)
| | - William J Brock
- Brock Scientific Consulting LLC, Montgomery Village, MD, USA
| | - Wendy Halpern
- Genentech, Safety Assessment Pathology, South San Francisco CA, US
| | | | - David Jones
- 9059Medicines and Healthcare products Regulatory Agency (MHRA), London, UK
| | | | - Pamela D McGovern
- 17136USDA National Agricultural Statistics Service (NASS), Washington, DC, US
| | | |
Collapse
|
20
|
Larson EA, Accardi MV, Zhong Y, Paquette D, Authier S. Drug-Induced Seizures: Considerations for Underlying Molecular Mechanisms. Int J Toxicol 2021; 40:403-412. [PMID: 34514888 DOI: 10.1177/10915818211040483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A broad spectrum of chemical entities have been associated with drug-induced seizure (DIS), emphasizing the importance of this potential liability across various drug classes (e.g., antidepressants, antipsychotics, antibiotics, and analgesics among others). Despite its importance within drug safety testing, an understanding of the molecular mechanisms associated with DIS is often lacking. The etiology of DIS is understood to be a result of either a deficit in inhibitory (e.g., gamma aminobutyric acid) or an elevated excitatory (e.g., glutamate) signaling, leading to synchronous neuronal depolarization affecting various brain regions and impairing normal neurological functions. How this altered neuronal signaling occurs and how these changes interact with other non-brain receptor driven DIS-associated changes such as metabolic disturbances, electrolyte imbalances, altered drug metabolism, and withdrawal effects are poorly understood. Herein, we discuss important molecular mechanisms identified in DIS for several drugs and/or drug classes. With a better understanding of the molecular mechanisms associated with DIS, in vivo or in vitro models may be applied to characterize and mitigate DIS risk during drug development. Susceptibility stratification for DIS presents species differences in the following order beagle dogs > rodents and cynomolgus monkeys > Göttingen minipigs with a more than 2-fold difference between canines and minipigs, which is important to consider during non-clinical species selection. While clinical signs such as myoclonus, severe muscle jerks, or convulsions are often associated with abnormal epileptiform EEG activity, tremors are most of the time physiological and rarely observed with concurrent epileptiform EEG activity which need to be considered during DIS risk evaluation.
Collapse
Affiliation(s)
| | | | - Yifei Zhong
- Charles River Laboratories, Laval, Quebec, Canada
| | | | - Simon Authier
- Charles River Laboratories, Laval, Quebec, Canada.,Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| |
Collapse
|
21
|
Matsuda N, Kinoshita K, Okamura A, Shirakawa T, Suzuki I. Histograms of Frequency-Intensity Distribution Deep Learning to Predict the Seizure Liability of Drugs in Electroencephalography. Toxicol Sci 2021; 182:229-242. [PMID: 34021344 DOI: 10.1093/toxsci/kfab061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Detection of seizures as well as that of seizure auras is effective in improving the predictive accuracy of seizure liability of drugs. Whereas electroencephalography has been known to be effective for the detection of seizure liability, no established methods are available for the detection of seizure auras. We developed a method for detecting seizure auras through machine learning using frequency-characteristic images of electroencephalograms. Histograms of frequency-intensity distribution prepared from electroencephalograms of rats analyzed during seizures induced with 4-aminopyridine (6 mg/kg), strychnine (3 mg/kg), and pilocarpine (400 mg/kg), were used to create an artificial intelligence (AI) system that learned the features of frequency-characteristic images during seizures. The AI system detected seizure states learned in advance with 100% accuracy induced even by convulsants acting through different mechanisms, and the risk of seizure before a seizure was detected in general observation. The developed AI system determined that the unlearned convulsant Tramadol (150 mg/kg) was the risk of seizure and the negative compounds aspirin and vehicle were negative. Moreover, the AI system detected seizure liability even in electroencephalography data associated with the use of 4-aminopyridine (3 mg/kg), strychnine (1 mg/kg), and pilocarpine (150 mg/kg), which did not induce seizures detectable in general observation. These results suggest that the AI system developed herein is an effective means for electroencephalographic detection of seizure auras, raising expectations for its practical use as a new analytical method that allows for the sensitive detection of seizure liability of drugs that has been overlooked previously in preclinical studies.
Collapse
Affiliation(s)
- Naoki Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai, Miyagi 982-8577, Japan
| | - Kenichi Kinoshita
- Drug Safety Research Labs, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Ai Okamura
- Drug Safety Research Labs, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Takafumi Shirakawa
- Drug Safety Research Labs, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai, Miyagi 982-8577, Japan
| |
Collapse
|
22
|
Tukker AM, Westerink RHS. Novel test strategies for in vitro seizure liability assessment. Expert Opin Drug Metab Toxicol 2021; 17:923-936. [PMID: 33595380 PMCID: PMC8367052 DOI: 10.1080/17425255.2021.1876026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/11/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The increasing incidence of mental illnesses and neurodegenerative diseases results in a high demand for drugs targeting the central nervous system (CNS). These drugs easily reach the CNS, have a high affinity for CNS targets, and are prone to cause seizures as an adverse drug reaction. Current seizure liability assessment heavily depends on in vivo or ex vivo animal models and is therefore ethically debated, labor intensive, expensive, and not always predictive for human risk. AREAS COVERED The demand for CNS drugs urges the development of alternative safety assessment strategies. Yet, the complexity of the CNS hampers reliable detection of compound-induced seizures. This review provides an overview of the requirements of in vitro seizure liability assays and highlights recent advances, including micro-electrode array (MEA) recordings using rodent and human cell models. EXPERT OPINION Successful and cost-effective replacement of in vivo and ex vivo models for seizure liability screening can reduce animal use for drug development, while increasing the predictive value of the assays, particularly if human cell models are used. However, these novel test strategies require further validation and standardization as well as additional refinements to better mimic the human in vivo situation and increase their predictive value.
Collapse
Affiliation(s)
- Anke M. Tukker
- School of Health Sciences, Purdue University, Hall for Discovery and Learning Research (DLR 339), INUSA
| | - Remco H. S. Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, TD Utrecht, The Netherlands
| |
Collapse
|
23
|
Tsuji K, Satsuka A, Kanda Y. [Current challenges and future perspectives of pharmacological testing using new approach methodologies]. Nihon Yakurigaku Zasshi 2021; 156:208-213. [PMID: 34193697 DOI: 10.1254/fpj.21020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The mission of regulatory science is to promote human longevity by providing safer and more effective drugs and ensuring human health. At present, various in vitro and in vivo evaluation methods are used for drug development, and no major problems have been observed. However, there is still room for improvement in terms of risk prediction in humans. Thus, new approaches and methodologies (NAMs) have recently been developed to predict adverse events in humans more accurately. Based on the animal alternative methods and the current COVID-19 pandemic, in vitro methods, such as human iPS cells, and computational approach are accelerated to improve the efficiency of drug development, ensure the patients' safety and speed up the review process. In this review, we would like to summarize the current status and future perspectives of pharmacological assay system using NAM in drug development.
Collapse
Affiliation(s)
- Kayoko Tsuji
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| | - Ayano Satsuka
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| |
Collapse
|
24
|
Gerber LS, van Melis LVJ, van Kleef RGDM, de Groot A, Westerink RHS. Culture of Rat Primary Cortical Cells for Microelectrode Array (MEA) Recordings to Screen for Acute and Developmental Neurotoxicity. Curr Protoc 2021; 1:e158. [PMID: 34152700 DOI: 10.1002/cpz1.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neurotoxicity testing of chemicals, drug candidates, and environmental pollutants still relies on extensive in vivo studies that are very costly, time-consuming, and ethically debated due to the large number of animals typically used. Currently, rat primary cortical cultures are widely used for in vitro neurotoxicity studies, as they closely resemble the in vitro brain with respect to the diversity of cell types, their physiological functions, and the pathological processes that they undergo. Common in vitro assays for neurotoxicity screening often focus on very target-specific endpoints such as morphological, biochemical, or electrophysiological changes, and such narrow focus can hamper translation and interpretation. Microelectrode array (MEA) recordings provide a non-invasive platform for extracellular recording of electrical activity of cultured neuronal cells, thereby enabling the evaluation of changes in neuronal (network) function as a sensitive and integrated endpoint for neurotoxicity screening. Here, we describe an in vitro approach for assessing changes in neuronal network function as a measure for neurotoxicity, using rat primary cortical cultures grown on MEAs. We provide a detailed protocol for the culture of rat primary cortical cells, and describe several experimental procedures to address acute, subchronic, and chronic exposure scenarios. We additionally describe the steps for processing and analyzing MEA and cell viability data. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation and culture of rat primary cortical cells on 48-well MEA plates Support Protocol 1: Pretreatment and washing of 48-well MEA plates before first use or for re-use Support Protocol 2: Coating of 48-well MEA plates with 0.1% PEI solution Basic Protocol 2: MEA measurements during acute exposure Alternate Protocol 1: MEA measurements during subchronic exposure Alternate Protocol 2: MEA measurements during chronic exposure Support Protocol 3: Determination of cell viability after MEA experiments Basic Protocol 3: MEA data processing Basic Protocol 4: Analyzing MEA experiments after acute and subchronic exposure Alternate Protocol 3: Analyzing MEA experiments after chronic exposure.
Collapse
Affiliation(s)
- Lora-Sophie Gerber
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Lennart V J van Melis
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
25
|
Deguchi Y. [Current state of and challenges in in vivo central nervous system evaluation in safety pharmacology studies-Topic overview and initiatives of the Japanese Safety Pharmacology Society]. Nihon Yakurigaku Zasshi 2021; 156:171-177. [PMID: 33952847 DOI: 10.1254/fpj.20102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The modified Irwin's method and functional observational battery (FOB)used in non-clinical studies for predicting side effects that may appear in the central nervous system (CNS)in clinical studies consist of mainly macroscopic observation and largely depend on the observer's ability. Therefore, appropriate training for the observer and consistency of findings are extremely important, making it necessary for methods and judgment criteria to be standardized. In addition, because of concern for animal welfare as well as an increase in biopharmaceutical and anticancer drug development, there is increasing opportunity to incorporate safety pharmacological evaluation into general toxicity studies. While CNS evaluation can be incorporated into general toxicity studies relatively easily, studies need to be designed in such a way that reliable data can be obtained without reducing the ability to detect neurobehavioral abnormalities. It is therefore important to improve CNS evaluation techniques and to share these techniques with new observers in order to reliably detect the effects on the CNS during drug development.
Collapse
|
26
|
Tsunemoto K, Yamada S, Kanda Y. [Current challenges and future perspectives of iPSC-based neurotoxicity testing]. Nihon Yakurigaku Zasshi 2021; 156:107-113. [PMID: 33642528 DOI: 10.1254/fpj.20097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Predicting drug-induced side effects in central nervous system is important because they can lead to the discontinuation of new drugs/candidates or the withdrawal of marketed drugs. Although many efforts are made, evaluation system using animals have not been highly predictive in humans. In addition, animal experiments are time-consuming and costly. To address these issues, in vitro evaluation methods, such as the use of New Approach Methodologies (NAM) have been explored. Human iPS cell technology has already been applied to assess drug-induced cardiotoxicity. In addition, the use of human iPS cell technology and in silico has been promoted for neurotoxicity assessment during the developmental neurotoxicity in terms of chemical safety issues. Organization for Economic Cooperation and Development (OECD) guidance regarding developmental neurotoxicity is under preparation. In this review, we will review the current trends in safety assessment methods for the central nervous system in light of these international trends.
Collapse
Affiliation(s)
| | - Shigeru Yamada
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| |
Collapse
|
27
|
Roberts R, Authier S, Mellon RD, Morton M, Suzuki I, Tjalkens RB, Valentin JP, Pierson JB. Can We Panelize Seizure? Toxicol Sci 2021; 179:3-13. [PMID: 33165543 DOI: 10.1093/toxsci/kfaa167] [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] [Indexed: 11/13/2022] Open
Abstract
Seizure liability remains a significant cause of attrition in drug discovery and development, leading to loss of competitiveness, delays, and increased costs. Current detection methods rely on observations made in in vivo studies intended to support clinical trials, such as tremors or other abnormal movements. These signs could be missed or misinterpreted; thus, definitive confirmation of drug-induced seizure requires a follow-up electroencephalogram study. There has been progress in in vivo detection of seizure using automated video systems that record and analyze animal movements. Nonetheless, it would be preferable to have earlier prediction of seizurogenic risk that could be used to eliminate liabilities early in discovery while there are options for medicinal chemists making potential new drugs. Attrition due to cardiac adverse events has benefited from routine early screening; could we reduce attrition due to seizure using a similar approach? Specifically, microelectrode arrays could be used to detect potential seizurogenic signals in stem-cell-derived neurons. In addition, there is clear evidence implicating neuronal voltage-gated and ligand-gated ion channels, GPCRs and transporters in seizure. Interactions with surrounding glial cells during states of stress or inflammation can also modulate ion channel function in neurons, adding to the challenge of seizure prediction. It is timely to evaluate the opportunity to develop an in vitro assessment of seizure linked to a panel of ion channel assays that predict seizure, with the aim of influencing structure-activity relationship at the design stage and eliminating compounds predicted to be associated with pro-seizurogenic state.
Collapse
Affiliation(s)
- Ruth Roberts
- ApconiX, Alderley Park, SK10 4TG, UK.,University of Birmingham, B15 2SD, UK
| | | | - R Daniel Mellon
- US Food and Drug Administration, Silver Spring, Maryland 20993
| | | | - Ikuro Suzuki
- Tohoku Institute of Technology, Sendai, 980-8577, Japan
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Early Solutions, Development Science, Investigative Toxicology, Chemin du Foriest, B-1420, Braine-l'Alleud, Belgium
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, District of Columbia 20005
| |
Collapse
|
28
|
Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Applicability of hiPSC-Derived Neuronal Cocultures and Rodent Primary Cortical Cultures for In Vitro Seizure Liability Assessment. Toxicol Sci 2020; 178:71-87. [PMID: 32866265 PMCID: PMC7657345 DOI: 10.1093/toxsci/kfaa136] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Seizures are life-threatening adverse drug reactions which are investigated late in drug development using rodent models. Consequently, if seizures are detected, a lot of time, money and animals have been used. Thus, there is a need for in vitro screening models using human cells to circumvent interspecies translation. We assessed the suitability of cocultures of human-induced pluripotent stem cell (hiPSC)-derived neurons and astrocytes compared with rodent primary cortical cultures for in vitro seizure liability assessment using microelectrode arrays. hiPSC-derived and rodent primary cortical neuronal cocultures were exposed to 9 known (non)seizurogenic compounds (pentylenetetrazole, amoxapine, enoxacin, amoxicillin, linopirdine, pilocarpine, chlorpromazine, phenytoin, and acetaminophen) to assess effects on neuronal network activity using microelectrode array recordings. All compounds affect activity in hiPSC-derived cocultures. In rodent primary cultures all compounds, except amoxicillin changed activity. Changes in activity patterns for both cell models differ for different classes of compounds. Both models had a comparable sensitivity for exposure to amoxapine (lowest observed effect concentration [LOEC] 0.03 µM), linopirdine (LOEC 1 µM), and pilocarpine (LOEC 0.3 µM). However, hiPSC-derived cultures were about 3 times more sensitive for exposure to pentylenetetrazole (LOEC 30 µM) than rodent primary cortical cultures (LOEC 100 µM). Sensitivity of hiPSC-derived cultures for chlorpromazine, phenytoin, and enoxacin was 10-30 times higher (LOECs 0.1, 0.3, and 0.1 µM, respectively) than in rodent cultures (LOECs 10, 3, and 3 µM, respectively). Our data indicate that hiPSC-derived neuronal cocultures may outperform rodent primary cortical cultures with respect to detecting seizures, thereby paving the way towards animal-free seizure assessment.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Fiona M J Wijnolts
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| |
Collapse
|
29
|
Matsui T, Miyamoto N, Saito F, Shinozawa T. Molecular Profiling of Human Induced Pluripotent Stem Cell-Derived Cells and their Application for Drug Safety Study. Curr Pharm Biotechnol 2020; 21:807-828. [PMID: 32321398 DOI: 10.2174/1389201021666200422090952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/10/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Drug-induced toxicity remains one of the leading causes of discontinuation of the drug candidate and post-marketing withdrawal. Thus, early identification of the drug candidates with the potential for toxicity is crucial in the drug development process. With the recent discovery of human- Induced Pluripotent Stem Cells (iPSC) and the establishment of the differentiation protocol of human iPSC into the cell types of interest, the differentiated cells from human iPSC have garnered much attention because of their potential applicability in toxicity evaluation as well as drug screening, disease modeling and cell therapy. In this review, we expanded on current information regarding the feasibility of human iPSC-derived cells for the evaluation of drug-induced toxicity with a focus on human iPSCderived hepatocyte (iPSC-Hep), cardiomyocyte (iPSC-CMs) and neurons (iPSC-Neurons). Further, we CSAHi, Consortium for Safety Assessment using Human iPS Cells, reported our gene expression profiling data with DNA microarray using commercially available human iPSC-derived cells (iPSC-Hep, iPSC-CMs, iPSC-Neurons), their relevant human tissues and primary cultured human cells to discuss the future direction of the three types of human iPSC-derived cells.
Collapse
Affiliation(s)
- Toshikatsu Matsui
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Norimasa Miyamoto
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Fumiyo Saito
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | | |
Collapse
|
30
|
Shirakawa T, Suzuki I. [Evaluation methods for drug-induced seizure by microelectrode array recording using human iPS cell-derived neurons]. Nihon Yakurigaku Zasshi 2020; 155:284-288. [PMID: 32879165 DOI: 10.1254/fpj.20028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In the drug development in pharmaceuticals, development of drugs may be discontinued due to the toxicity and clinical side effect, therefore, safety assessment is one of the important factors in drug development. Consortium for Safety Assessment using Human Cells (CSAHi) has been launched for developing and standardizing a toxicity evaluation system for development of drug using human iPS cell differentiated cells. CSAHi focuses on hepato-, cardio-, and neuro-toxicities as important toxicity organs which are attributed to the causes of discontinuation of drug development. In neurotoxicity, seizure is an important finding because of high frequency expression in nonclinical. Multi-electrode array (MEA) systems have recently attracted attention as useful for evaluating seizure risk because they can non-invasively measure the electrophysiological activities of neural networks. We are evaluating the electrophysiological responses to several seizure compounds using MEA in cultured hiPSC-derived neurons. It is important to establish an analytical method to detecting seizure-like activities. We have focused the establish of the effective analysis parameters for detecting seizure risk. We identify to be separate the responses between seizure-positive and seizure-negative compounds using principal component analysis of 10 analysis parameters. In addition, we could separate the mechanism of action of the seizure-positive compounds by principal component analysis and cluster analysis using 10 parameters. It is considered that principal component analysis or cluster analysis could not only assess the seizure risk but also classify mechanism of action by in vitro MEA system using human iPS cell-derived neurons.
Collapse
Affiliation(s)
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology
| |
Collapse
|
31
|
Miraucourt LS, Accardi MV, Asin KE, Pugsley MK, Curtis MJ, Authier S. The application of electrophysiological methods to characterize AMPA receptors in dissociated adult rat and non-human primate cerebellar neurons for use in neuronal safety pharmacology assessments of the central nervous system. J Pharmacol Toxicol Methods 2020; 105:106883. [DOI: 10.1016/j.vascn.2020.106883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/15/2019] [Accepted: 05/21/2020] [Indexed: 11/25/2022]
|
32
|
Unraveling the mechanism and the risk behind seizure liability of lead compounds in a neuroscience project. J Pharmacol Toxicol Methods 2020; 104:106874. [DOI: 10.1016/j.vascn.2020.106874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/20/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022]
|
33
|
Shirakawa T, Suzuki I. Approach to Neurotoxicity using Human iPSC Neurons: Consortium for Safety Assessment using Human iPS Cells. Curr Pharm Biotechnol 2020; 21:780-786. [DOI: 10.2174/1389201020666191129103730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/27/2019] [Accepted: 11/03/2019] [Indexed: 01/05/2023]
Abstract
Neurotoxicity, as well as cardiotoxicity and hepatotoxicity, resulting from administration of
a test article is considered a major adverse effect both pre-clinically and clinically. Among the different
types of neurotoxicity occurring during the drug development process, seizure is one of the most serious
one. Seizure occurrence is usually assessed using in vivo animal models, the Functional Observational
Battery, the Irwin test or electroencephalograms. In in vitro studies, a number of assessments can
be performed using animal organs/cells. Interestingly, recent developments in stem cell biology, especially
the development of Human-Induced Pluripotent Stem (iPS) cells, are enabling the assessment of
neurotoxicity in human iPS cell-derived neurons. Further, a Multi-Electrode Array (MEA) using rodent
neurons is a useful tool for identifying seizure-inducing compounds. The Consortium for Safety Assessment
using Human iPS Cells (CSAHi; http://csahi.org/en/) was established in 2013 by the Japan
Pharmaceutical Manufacturers Association (JPMA) to verify the application of human iPS cell-derived
neuronal cells to drug safety evaluation. The Neuro Team of CSAHi has been attempting to evaluate the
seizure risk of compounds using the MEA platform. Here, we review the current status of neurotoxicity
and recent work, including problems related to the use of the MEA assay with human iPS neuronal
cell-derived neurons, and future developments.
Collapse
Affiliation(s)
- Takafumi Shirakawa
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Neuro Team, Japan
| | - Ikuro Suzuki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Neuro Team, Japan
| |
Collapse
|
34
|
Authier S, Abernathy MM, Correll K, Chui RW, Dalton J, Foley CM, Friedrichs GS, Koerner JE, Kallman MJ, Pannirselvam M, Redfern WS, Urmaliya V, Valentin JP, Wisialowski T, Zabka TS, Pugsley MK. An Industry Survey With Focus on Cardiovascular Safety Pharmacology Study Design and Data Interpretation. Int J Toxicol 2020; 39:274-293. [DOI: 10.1177/1091581820921338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction: The Safety Pharmacology Society (SPS) conducted a membership survey to examine industry practices related mainly to cardiovascular (CV) safety pharmacology (SP). Methods: Questions addressed nonclinical study design, data analysis methods, drug-induced effects, and conventional and novel CV assays. Results: The most frequent therapeutic area targeted by drugs developed by the companies/institutions that employ survey responders was oncology. The most frequently observed drug-mediated effects included an increased heart rate, increased arterial blood pressure, hERG (IKr) block, decreased arterial blood pressure, decreased heart rate, QTc prolongation, and changes in body temperature. Broadly implemented study practices included Latin square crossover study design with n = 4 for nonrodent CV studies, statistical analysis of data (eg, analysis of variance), use of arrhythmia detection software, and the inclusion of data from all study animals when integrating SP studies into toxicology studies. Most responders frequently used individual animal housing conditions. Responders commonly evaluated drug effects on multiple ion channels, but in silico modeling methods were used much less frequently. Most responders rarely measured the J-Tpeak interval in CV studies. Uncertainties relative to Standard for Exchange of Nonclinical Data applications for data derived from CV SP studies were common. Although available, the use of human induced pluripotent stem cell cardiomyocytes remains rare. The respiratory SP study was rarely involved with identifying drug-induced functional issues. Responders indicated that the study-derived no observed effect level was more frequently determined than the no observed adverse effect level in CV SP studies; however, a large proportion of survey responders used neither.
Collapse
Affiliation(s)
| | | | | | - Ray W. Chui
- Amgen Research, Safety Pharmacology & Animal Research Center, Amgen, Inc, Thousand Oaks, CA, USA
| | | | - C. Michael Foley
- Department of Safety Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, USA
| | | | - John E. Koerner
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | | | | | - Vijay Urmaliya
- Global Safety Pharmacology, Janssen Research & Development, Beerse, Belgium
| | | | | | - Tanja S. Zabka
- Development Sciences Safety Assessment, Genentech, South San Francisco, CA, USA
| | | |
Collapse
|
35
|
González-Cano R, Montilla-García Á, Ruiz-Cantero MC, Bravo-Caparrós I, Tejada MÁ, Nieto FR, Cobos EJ. The search for translational pain outcomes to refine analgesic development: Where did we come from and where are we going? Neurosci Biobehav Rev 2020; 113:238-261. [PMID: 32147529 DOI: 10.1016/j.neubiorev.2020.03.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/06/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Pain measures traditionally used in rodents record mere reflexes evoked by sensory stimuli; the results thus may not fully reflect the human pain phenotype. Alterations in physical and emotional functioning, pain-depressed behaviors and facial pain expressions were recently proposed as additional pain outcomes to provide a more accurate measure of clinical pain in rodents, and hence to potentially enhance analgesic drug development. We aimed to review how preclinical pain assessment has evolved since the development of the tail flick test in 1941, with a particular focus on a critical analysis of some nonstandard pain outcomes, and a consideration of how sex differences may affect the performance of these pain surrogates. We tracked original research articles in Medline for the following periods: 1973-1977, 1983-1987, 1993-1997, 2003-2007, and 2014-2018. We identified 606 research articles about alternative surrogate pain measures, 473 of which were published between 2014 and 2018. This indicates that preclinical pain assessment is moving toward the use of these measures, which may soon become standard procedures in preclinical pain laboratories.
Collapse
Affiliation(s)
- Rafael González-Cano
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.
| | - Francisco R Nieto
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Teófilo Hernando Institute for Drug Discovery, Madrid, Spain.
| |
Collapse
|
36
|
Ballesteros C, Pouliot M, Froment R, Maghezzi MS, St-Jean C, Li C, Paquette D, Authier S. Cerebrospinal Fluid Characterization in Cynomolgus Monkeys, Beagle Dogs, and Göttingen Minipigs. Int J Toxicol 2020; 39:124-130. [PMID: 32066300 PMCID: PMC7079291 DOI: 10.1177/1091581820905092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intrathecal administration is an important route for drug delivery, and in pharmacology and toxicology studies, cerebrospinal fluid (CSF) collection and analysis is required for evaluating blood–brain barrier penetration and central nervous system exposure. The characteristics of CSF in commonly used nonrodent models are lacking. The purpose of this study is to evaluate and provide some insights into normal cellular and biochemical composition of CSF as well as diffusion potential following intrathecal injection across several nonrodent species. Cerebrospinal fluid samples were collected from the cerebellomedullary cistern of beagle dogs, cynomolgus monkeys, and Göttingen minipigs and analyzed for clinical chemistry and cytological evaluation. Diffusion into the intrathecal space following intrathecal injection was assessed following administration of a contrast agent using fluoroscopy. The predominant cell types identified in CSF samples were lymphocytes and monocytoid cells; however, lymphocytes were represented in a higher percentage in dogs and monkeys as opposed to monocytoid cells in minipigs. Clinical chemistry parameters in CSF revealed higher Cl− concentrations than plasma, but lower K+, Ca2+, phosphorus, glucose, creatinine, and total protein levels consistent across all 3 species. Diffusion rates following intrathecal injection of iodixanol showed some variability with dogs, showing the greatest diffusion distance; however, the longest diffusion time through the intervertebral space, followed by monkeys and minipigs. Minimal diffusion was observed in minipigs, which could have been attributed to anatomical spinal constraints that have been previously identified in this species.
Collapse
Affiliation(s)
| | | | - Rémi Froment
- Faculty of Veterinary Medicine, University of Montreal, Quebec, Canada
| | | | - Camille St-Jean
- Faculty of Veterinary Medicine, University of Montreal, Quebec, Canada
| | - Christian Li
- Charles River Laboratories Laval, Quebec, Canada
| | | | - Simon Authier
- Charles River Laboratories Laval, Quebec, Canada.,Faculty of Veterinary Medicine, University of Montreal, Quebec, Canada
| |
Collapse
|
37
|
Rockley KL, Roberts RA, Morton MJ. Innovative models for in vitro detection of seizure. Toxicol Res (Camb) 2019; 8:784-788. [PMID: 32206299 DOI: 10.1039/c9tx00210c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022] Open
Abstract
Data show that toxicity to the central nervous system (CNS) is the most frequent cause of safety failures during the clinical phase of drug development. CNS endpoints such as seizure pose a safety risk to patients and volunteers and can lead to a loss of competitiveness, delays, and increased costs. Current methods rely on detection in the nonclinical rodent and non-rodent studies required to support clinical trials. There are two main issues with this approach; seizure may be missed in the animal studies and, even if seizure is detected, significant resource has already been invested in the project by this stage. Thus, there is a need to develop improved screening methods that can be used earlier in drug discovery to predict seizure. Advances in stem cell biology coupled with an increased understanding of the role of ion channels in seizure offer an opportunity for a new paradigm in screening. Human derived induced pluripotent stem cells (hiPSCs) representative of almost all cellular subtypes present in the brain can be incorporated into physiologically relevant in vitro models that can be used to determine seizure risk using high-throughput methods. Akin to the success of screening against a panel of ion channels such as hERG to reduce cardiovascular safety liability, the involvement of ion channels in seizure suggests that a similar approach to early seizure detection is valid. Profiling of the ion channels expressed in hiPSC models showing the seizurogenic phenotype coupled with electrophysiological assessment of ion channel function could translate into an ion channel seizure panel for rapid and reliable in vitro detection of seizure. The mechanistic information gathered would support optimal drug design early in development before resources, animals and time have been wasted.
Collapse
Affiliation(s)
- Kimberly L Rockley
- ApconiX , Alderley Park , Alderley Edge , SK10 4TG , UK . ; Tel: +44 (0)77 33 01 43 96
| | - Ruth A Roberts
- ApconiX , Alderley Park , Alderley Edge , SK10 4TG , UK . ; Tel: +44 (0)77 33 01 43 96.,School of Biosciences , University of Birmingham , B15 2TT , UK
| | - Michael J Morton
- ApconiX , Alderley Park , Alderley Edge , SK10 4TG , UK . ; Tel: +44 (0)77 33 01 43 96
| |
Collapse
|
38
|
Jackson SJ, Authier S, Brohmann H, Goody SM, Jones D, Prior H, Rosch A, Traebert M, Tse K, Valentin JP, Milne A. Neurofunctional test batteries in safety pharmacology – Current and emerging considerations for the drug development process. J Pharmacol Toxicol Methods 2019; 100:106602. [DOI: 10.1016/j.vascn.2019.106602] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022]
|
39
|
Abt M, Dinklo T, Rothfuss A, Husar E, Dannecker R, Kallivroussis K, Peck R, Doessegger L, Wandel C. A Framework Proposal to Follow-Up on Preclinical Convulsive Signals of a New Molecular Entity in First-in-Human Studies Using Electroencephalographic Monitoring. Clin Pharmacol Ther 2019; 106:968-980. [PMID: 30993670 PMCID: PMC6851537 DOI: 10.1002/cpt.1455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/18/2019] [Indexed: 01/11/2023]
Abstract
Traditionally, in dose-escalating first-in-human (FiH) studies, a dose cap with a 10-fold safety margin to the no observed effect level in animals is implemented if convulsive events are observed in animals. However, the convulsive risk seen in animals does not generally translate to humans. Several lines of evidence are summarized indicating that in a dose-escalating setting, electroencephalographic epileptiform abnormalities occur at lower doses than clinical convulsive events. Therefore, we propose to consider the occurrence of epileptiform abnormalities in toxicology studies as premonitory signals for convulsions in dose-escalating FiH studies. Compared with the traditional dose-cap approach, this may allow the exploration of higher doses in FiH and, subsequently, phase II studies without compromising human safety. Similarly, the presence or absence of electroencephalographic epileptiform abnormalities may also aid the assessment of proconvulsive risk in situations of increased perpetrator burden as potentially present in pharmacokinetic and/or pharmacodynamic drug-drug interactions.
Collapse
Affiliation(s)
- Markus Abt
- Department of BiometricsF. Hoffmann‐La Roche AGBaselSwitzerland
| | - Theo Dinklo
- Roche Pharma Research and Early DevelopmentRoche Innovation Center BaselF. Hoffmann‐La Roche AGBaselSwitzerland
| | - Andreas Rothfuss
- Roche Pharma Research and Early DevelopmentRoche Innovation Center BaselF. Hoffmann‐La Roche AGBaselSwitzerland
| | - Elisabeth Husar
- Roche Pharma Research and Early DevelopmentRoche Innovation Center BaselF. Hoffmann‐La Roche AGBaselSwitzerland
| | | | | | - Richard Peck
- Roche Pharma Research and Early DevelopmentRoche Innovation Center BaselF. Hoffmann‐La Roche AGBaselSwitzerland
| | | | - Christoph Wandel
- Department of Safety & Risk ManagementF. Hoffmann‐La Roche AGBaselSwitzerland
| |
Collapse
|
40
|
Authier S, Arezzo J, Pouliot M, Accardi MV, Boulay E, Troncy E, Dubuc Mageau M, Tan W, Sanfacon A, Mignault Goulet S, Paquette D. Reprint of "EEG: Characteristics of drug-induced seizures in rats, dogs and non-human primates". J Pharmacol Toxicol Methods 2019; 99:106611. [DOI: 10.1016/j.vascn.2019.106611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 10/26/2022]
|
41
|
Redfern WS, Dymond A, Strang I, Storey S, Grant C, Marks L, Barnard C, Heys C, Moyser K, Greenwood K, Cobey D, Moore N, Karp NA, Prior H. The functional observational battery and modified Irwin test as global neurobehavioral assessments in the rat: Pharmacological validation data and a comparison of methods. J Pharmacol Toxicol Methods 2019; 98:106591. [DOI: 10.1016/j.vascn.2019.106591] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/15/2022]
|
42
|
Steidl E, Gleyzes M, Maddalena F, Debanne D, Buisson B. Neuroservice proconvulsive (NS-PC) set: A new platform of electrophysiology-based assays to determine the proconvulsive potential of lead compounds. J Pharmacol Toxicol Methods 2019; 99:106587. [PMID: 31207287 DOI: 10.1016/j.vascn.2019.106587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Failures in drug development often result from the emergence of unexpected adverse drug reactions. It is clear that adverse drug reactions, including seizure liability, should be assessed earlier. The goal of the present work was to develop a new platform of in vitro assays, NS-PC set (for Neuroservice proconvulsive set), to determine the proconvulsive potential of compounds earlier in preclinical development. METHODS Assays were based on electrophysiological recordings in acute hippocampal slices performed with multielectrode arrays. 4 reference proconvulsive/seizurogenic compounds (4-aminopyridine, bicuculline, kainate and carbachol) and 4 anti-epileptic drugs (AEDs; phenobarbital, carbamazepine, clonazepam and valproic acid) were evaluated on electrophysiological endpoints involved in seizure risk (neuronal excitability, balance of excitatory/inhibitory synaptic transmission, occurrence of neuronal synchronization mechanisms materialized by epileptiform discharges). RESULTS The reference compounds increased the number and area under the curve of population spikes, triggered epileptiform discharges and enhanced the firing rate of CA1 neurons. The effects of the 4 antiepileptic drugs were assessed on these 3 parameters. They were able to partially of completely reverse the effects of proconvulsive compounds. DISCUSSION The use of reference proconvulsive compounds and AEDs validated the electrophysiological parameters to detect proconvulsive risk. Systematic evaluation of compounds with the 3 complementary endpoints increase the probability to detect seizure liability in vitro. Depending on the compound mechanism of action, only one or two of the identified parameters might be modified.
Collapse
Affiliation(s)
- Esther Steidl
- Neuroservice SARL, 595 rue Pierre Berthier, 13593 Aix-en-Provence, France.
| | - Melanie Gleyzes
- Neuroservice SARL, 595 rue Pierre Berthier, 13593 Aix-en-Provence, France
| | - Fabien Maddalena
- Neuroservice SARL, 595 rue Pierre Berthier, 13593 Aix-en-Provence, France
| | - Dominique Debanne
- UNIS, UMR1072 INSERM - Aix-Marseille Université, 53 Bvd Pierre Dramard, 13015 Marseille, France
| | - Bruno Buisson
- Neuroservice SARL, 595 rue Pierre Berthier, 13593 Aix-en-Provence, France
| |
Collapse
|
43
|
EEG: Characteristics of drug-induced seizures in rats, dogs and non-human primates. J Pharmacol Toxicol Methods 2019; 97:52-58. [DOI: 10.1016/j.vascn.2019.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 11/21/2022]
|
44
|
Bradley JA, Luithardt HH, Metea MR, Strock CJ. In Vitro Screening for Seizure Liability Using Microelectrode Array Technology. Toxicol Sci 2019; 163:240-253. [PMID: 29432603 DOI: 10.1093/toxsci/kfy029] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug-induced seizure liabilities produce significant compound attrition during drug discovery. Currently available in vitro cytotoxicity assays cannot predict all toxicity mechanisms due to the failure of these assays to predict sublethal target-specific electrophysiological liabilities. Identification of seizurogenic and other electrophysiological effects at early stages of the drug development process is important to ensure that safe candidate compounds can be developed while chemical design is taking place, long before these liabilities are discovered in costly preclinical in vivo studies. The development of a high throughput and reliable in vitro assay to screen compounds for seizure liabilities would de-risk compounds significantly earlier in the drug discovery process and with greater dependability. Here we describe a method for screening compounds that utilizes rat cortical neurons plated onto multiwell microelectrode array plates to identify compounds that cause neurophysiological disruptions. Changes in 12 electrophysiological parameters (spike train descriptors) were measured after application of known seizurogenic compounds and the response pattern was mapped relative to negative controls, vehicle control and neurotoxic controls. Twenty chemicals with a variety of therapeutic indications and targets, including GABAA antagonists, glycine receptor antagonists, ion channel blockers, muscarinic agonist, δ-opioid receptor agonist, dopaminergic D2/adrenergic receptor blocker and nonsteroidal anti-inflammatory drugs, were tested to assess this system. Sixteen of the seventeen seizurogenic/neurotoxic compounds tested positive for seizure liability or neurotoxicity, moreover, different endpoint response patterns for firing rate, burst characteristics and synchrony that distinguished the chemicals into groups relating to target and seizurogenic response emerged from the data. The negative and vehicle control compounds had no effect on neural activity. In conclusion, the multiwell microelectrode array platform using cryopreserved rat cortical neurons is a highly effective high throughput method for reliably screening seizure liabilities within an early de-risking drug development paradigm.
Collapse
Affiliation(s)
| | | | - Monica R Metea
- Cyprotex US, LLC, An Evotec Company, Watertown, Massachusetts
| | | |
Collapse
|
45
|
Bradley JA, Strock CJ. Screening for Neurotoxicity with Microelectrode Array. ACTA ACUST UNITED AC 2018; 79:e67. [DOI: 10.1002/cptx.67] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
46
|
Grainger AI, King MC, Nagel DA, Parri HR, Coleman MD, Hill EJ. In vitro Models for Seizure-Liability Testing Using Induced Pluripotent Stem Cells. Front Neurosci 2018; 12:590. [PMID: 30233290 PMCID: PMC6127295 DOI: 10.3389/fnins.2018.00590] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
The brain is the most complex organ in the body, controlling our highest functions, as well as regulating myriad processes which incorporate the entire physiological system. The effects of prospective therapeutic entities on the brain and central nervous system (CNS) may potentially cause significant injury, hence, CNS toxicity testing forms part of the “core battery” of safety pharmacology studies. Drug-induced seizure is a major reason for compound attrition during drug development. Currently, the rat ex vivo hippocampal slice assay is the standard option for seizure-liability studies, followed by primary rodent cultures. These models can respond to diverse agents and predict seizure outcome, yet controversy over the relevance, efficacy, and cost of these animal-based methods has led to interest in the development of human-derived models. Existing platforms often utilize rodents, and so lack human receptors and other drug targets, which may produce misleading data, with difficulties in inter-species extrapolation. Current electrophysiological approaches are typically used in a low-throughput capacity and network function may be overlooked. Human-derived induced pluripotent stem cells (iPSCs) are a promising avenue for neurotoxicity testing, increasingly utilized in drug screening and disease modeling. Furthermore, the combination of iPSC-derived models with functional techniques such as multi-electrode array (MEA) analysis can provide information on neuronal network function, with increased sensitivity to neurotoxic effects which disrupt different pathways. The use of an in vitro human iPSC-derived neural model for neurotoxicity studies, combined with high-throughput techniques such as MEA recordings, could be a suitable addition to existing pre-clinical seizure-liability testing strategies.
Collapse
Affiliation(s)
| | - Marianne C King
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - David A Nagel
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - H Rheinallt Parri
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Michael D Coleman
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Eric J Hill
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
47
|
Ojima A, Miyamoto N. Method for MEA Data Analysis of Drug-treated Rat Primary Neurons and Human iPSC-derived Neurons to Evaluate the Risk of Drug-induced Seizures. YAKUGAKU ZASSHI 2018; 138:823-828. [DOI: 10.1248/yakushi.17-00213-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Do in vitro assays in rat primary neurons predict drug-induced seizure liability in humans? Toxicol Appl Pharmacol 2018; 346:45-57. [PMID: 29596924 DOI: 10.1016/j.taap.2018.03.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 11/22/2022]
Abstract
Drug-induced seizures contribute to the high attrition rate of pharmaceutical compounds in development. The assessment of drug-induced seizure liability generally occurs in later phases of development using low throughput and intensive in vivo assays. In the present study, we evaluated the potential of an in vitro assay for detecting drug-induced seizure risk compared to evaluation in rats in vivo. We investigated the effects of 8 reference drugs with a known seizurogenic risk using micro-electrode array (MEA) recordings from freshly-dissociated rat primary neurons cultured on 48-well dishes for 28 days, compared to their effects on the EEG in anesthetized rats. In addition, we evaluated functional responses and mRNA expression levels of different receptors in vitro to understand the potential mechanisms of drug-induced seizure risk. Combining the functional MEA in vitro data with concomitant gene expression allowed us to identify several potential molecular targets that might explain the drug-induced seizures occurring in both rats and humans. Our data 1) demonstrate the utility of a group of MEA parameters for detecting potential drug-induced seizure risk in vitro; 2) suggest that an in vitro MEA assay with rat primary neurons may have advantages over an in vivo rat model; and 3) identify potential mechanisms for the discordance between rat assays and human seizure risk for certain seizurogenic drugs.
Collapse
|
49
|
Seizure liability assessments using the hippocampal tissue slice: Comparison of non-clinical species. J Pharmacol Toxicol Methods 2017; 93:59-68. [PMID: 29155282 DOI: 10.1016/j.vascn.2017.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/29/2017] [Accepted: 11/13/2017] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Traditionally, rat hippocampal tissue slice models are used as an in vitro electrophysiology assay to assess seizurogenic potential in early drug development despite non-clinical species-specific differences noted during in vivo seizure studies. METHODS Hippocampal tissue slices were acutely isolated from rats, minipigs, dogs and nonhuman primates (NHP). Population spikes (PS) were evoked through stimulation of the CA3 Schaffer collateral pathway and recorded using in vitro electrophysiological techniques via an extracellular electrode placed within the CA1 stratum pyramidale cell body layer. RESULTS Hippocampal slices, across all species, displayed a concentration-dependent increase in PS area and number with the pro-convulsant pentylenetetrazol (PTZ; 0.1-10mM). Beagle dogs exhibited higher sensitivities to PTZ-induced changes in PS area and number compared to both rats and NHPs which presented nuanced differences in their responsiveness to PTZ modulation. Minipigs were comparatively resistant to PTZ-induced changes in both PS area and number. Rat and NHP hippocampal tissues were further characterized with the pro-convulsant agents 4-aminopyradine (4-AP; 1-100μM) and cefazolin (0.001-10mM). Rats possessed higher sensitivities to 4-AP- and cefazolin-induced changes to both PS area and number whereas NHP displayed greater modulation in PS duration. The anti-convulsant agents, diazepam (10-500μM) and lidocaine (1-500μM), were also tested on either rat and/or NHP tissue with both drugs repressing PS activation at high concentrations. DISCUSSION Hippocampal tissue slices, across all species, possessed distinct sensitivities to pro- and anti-convulsant agents which may benefit the design of non-clinical seizure liability studies and their associated data interpretation.
Collapse
|
50
|
Authier S, Pugsley MK, Koerner JE, Fermini B, Redfern WS, Valentin JP, Vargas HM, Leishman DJ, Correll K, Curtis MJ. Proarrhythmia liability assessment and the comprehensive in vitro Proarrhythmia Assay (CiPA): An industry survey on current practice. J Pharmacol Toxicol Methods 2017; 86:34-43. [PMID: 28223123 DOI: 10.1016/j.vascn.2017.02.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The Safety Pharmacology Society (SPS) has conducted a survey of its membership to identify industry practices related to testing considered in the Comprehensive In vitro Proarrhythmia Assay (CiPA). METHODS Survey topics included nonclinical approaches to address proarrhythmia issues, conduct of in silico studies, in vitro ion channel testing methods used, drugs used as positive controls during the conduct of cardiac ion channel studies, types of arrhythmias observed in non-clinical studies and use of the anticipated CiPA ion channel assay. RESULTS In silico studies were used to evaluate effects on ventricular action potentials by only 15% of responders. In vitro assays were used mostly to assess QT prolongation (95%), cardiac Ca2+ and Na+ channel blockade (82%) and QT shortening or QRS prolongation (53%). For de-risking of candidate drugs for proarrhythmia, those assays most relevant to CiPA including cell lines stably expressing ion channels used to determine potency of drug block were most frequently used (89%) and human stem cell-derived or induced pluripotent stem cell cardiomyocytes (46%). Those in vivo assays related to general proarrhythmia derisking include ECG recording using implanted telemetry technology (88%), jacketed external telemetry (62%) and anesthetized animal models (53%). While the CiPA initiative was supported by 92% of responders, there may be some disconnect between current practice and future expectations, as explained. DISCUSSION Proarrhythmia liability assessment in drug development presently includes study types consistent with CiPA. It is anticipated that CiPA will develop into a workable solution to the concern that proarrhythmia liability testing remains suboptimal.
Collapse
Affiliation(s)
- Simon Authier
- CiToxLAB North America, 445, Armand-Frappier Boul, Laval, QC H7V 4B3, Canada.
| | - Michael K Pugsley
- Department of Toxicology & PKDM, Purdue Pharma L.P., Cranbury, NJ 08512, USA
| | - John E Koerner
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Bernard Fermini
- Safety & Toxicology Assessment, Coyne Scientific, Atlanta, GA 30303, USA
| | | | | | - Hugo M Vargas
- Integrated Discovery and Safety Pharmacology, Amgen, Inc., Thousand Oaks, CA, USA
| | | | | | - Michael J Curtis
- Cardiovascular Division, Faculty of Life Sciences & Medicine, King's College London, Rayne Institute, St Thomas' Hospital, London SE17EH, UK
| |
Collapse
|