Summary
Drug safety initiatives have endorsed human iPSC-derived cardiomyocytes (hiPSC-CMs) as an in vitro model for predicting drug-induced cardiac arrhythmia. However, the extent to which human-defined features of in vitro arrhythmia predict actual clinical risk has been much debated. Here, we trained a convolutional neural network classifier (CNN) to learn features of in vitro action potential recordings of hiPSC-CMs that are associated with lethal Torsade de Pointes arrhythmia. The CNN classifier accurately predicted the risk of drug-induced arrhythmia in people. The risk profile of the test drugs was similar across hiPSC-CMs derived from different healthy donors. In contrast, pathogenic mutations that cause arrhythmogenic cardiomyopathies in patients significantly increased the proarrhythmic propensity to certain intermediate and high-risk drugs in the hiPSC-CMs. Thus, deep learning can identify in vitro arrhythmic features that correlate with clinical arrhythmia and discern the influence of patient genetics on the risk of drug-induced arrhythmia.
Keywords: induced pluripotent stem cells, iPSC, cardiomyocytes, artificial intelligence, AI, deep learning, neural network, drug screening, drug-induced arrhythmia, CiPA, safety pharmacology
Graphical Abstract
ETOC:
Serrano et al. used human iPSC-derived cardiomyocytes and deep learning data analysis to establish an In vitro safety margin that predicts clinical proarrhythmic effects of drugs. Their platform shows high accuracy in identifying risky drugs as well as genotypes associated with increased risk of arrhythmia in people.
Introduction
Drug-induced arrhythmias are a common cause of drug attrition during development and for restricted use or withdrawal from the market1–3. As people vary in their predisposition to drug-induced arrhythmia 4–6, there is a widely accepted need to assess risk in susceptible populations 7,8. For ethical reasons and practical limitations, susceptible individuals, including carriers of rare predisposing gene variants, are not generally included in clinical trials 9. hiPSC-CMs retain an individual’s genetic makeup and enable scalable production of cardiac cells for in vitro testing and, therefore, are a breakthrough technology for risk assessment 10,11. This notion is supported by the findings that several mutations that cause electrophysiological or myopathic heart disease predispose cardiomyocytes to drug-induced arrhythmia 12.
Cell-based assays assess arrhythmia risk by quantifying waveform features in the cells’ action potential. Typically, these features are quantified using human-defined metrics such as the action potential duration at 90% amplitude (APD90) or incidence of after depolarizations 6,13,14. However, these human-defined features do not accurately predict clinical arrhythmia 15–17. Altogether, the complex manifestations of arrhythmia, the uncertain correspondence between in vitro action potential waveform features and actual clinical arrhythmia, and the influence of disease susceptibility loci present significant challenges for risk prediction.
To address this problem, we developed a deep learning approach to discriminate the in vitro electrophysiological features induced in hiPSC-CMs by reference drugs with well-established (high to low) risk of eliciting the life-threatening ventricular tachyarrhythmia (Torsades de Pointes, TdP). Deep learning is a type of artificial intelligence (AI) that uses multiple computational layers in a deep neural network (DNN). DNNs extract features relevant to discriminating input classes in a systematic and unbiased manner, effectively removing the need for human-defined metrics 18. Among the different types of DNNs, convolutional neural networks (CNNs), which learn complex features from input data by assigning weights to the result of convolutional operations, are showing tremendous success in various biomedical applications such as medical image analysis 19 and physiological signal analysis 20. Recently, CNNs have been used to automate the detection and classification of arrhythmias both in vitro21 and in vivo 22.
We trained a CNN to discriminate high versus low-risk drugs based on intrinsic drug-induced electrophysiological waveforms in hiPSC-CMs rather than human expectations. The CNN more accurately classified actual drug risk in patients than did human-defined metrics. Moreover, the trained CNN successfully quantified the increase risk of drug induced arrhythmia caused by cardiomyopathic gene variants, which pose a clinically significant risk factor that has been challenging to quantify 23. In summary, deep learning out-performed human-defined methods for drug risk assessment and detected an influence of patient genetics on susceptibility to drug-induced arrhythmia.
Results
High throughput screening of electrophysiological effects of drugs in healthy and disease hiPSC-CMs models
This study aimed to develop a new paradigm to accurately predict the risk of drugs and genetics on TdP arrhythmia without imposing human bias (Fig. 1). Genotype-phenotype relationships were modeled in a cohort of hiPSC-CMs generated from 8 hiPSC lines. Three (3) lines were derived from healthy donors (HD.113, HD.273, and HD.15S1), while five (5) lines harbored pathogenic mutations that cause HCM [HCM.MYBPC3 p.R943X 24], left ventricular noncompaction (LVNC), and HCM [HCM.TPM1 p.K37E 25] and DCM [DCM.PLN p.R14del 26, DCM.RBM20 p.R634Q 27, DCM.TNNT2 p.R183W 28]. These mutations were introduced in the same genetic background (HD.15S1 iPSCs) by CRISPR-mediated genome editing, minimizing the potentially confounding effects of the genetic background. All these mutations, except for the TNNT2 p.R183W mutation, are associated with elevated arrhythmic risk in patients, including ventricular arrhythmias and sudden cardiac death.29–34
Figure 1. Strategy to determine the influence of myopathic gene variants on the proarrhythmic effect of drugs.
hiPSC-CMs were generated from three donor patients without risk-associated genetics. DCM and HCM causing mutations were introduced to one of the healthy background and multiple batches of hiPSC-CMs were generated. The hiPSC-CMs were treated with 37 drugs of characterized high, intermediate and low/no arrhythmic risk. Each drug was tested at 8 different concentrations and a voltage sensitive dye was used to obtain membrane potential recordings. A CNN was trained to classify voltage traces based on the drug’s risk of inducing arrhythmia in patients. Class probabilities from the CNN were used to rank the proarrhythmic effects of drugs and evaluate the influence of myopathic gene variants.
Experiments were carried out using a panel of drugs with well-characterized proarrhythmia risk profiles, including the 28 drugs proposed by Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative 35 and 9 additional drugs to facilitate generalization across studies. For training and evaluation purposes, we used the CiPA collection categorized as high, intermediate, and low/no risk for developing TdP arrhythmia at clinical exposures based on published reports, the FDA adverse effect database, and expert opinion 36.
The uniformity and quality of the cardiomyocyte batches were assessed based on the baseline morphology of the action potential traces from 28 differentiation batches from 8 hiPSC lines displayed typical electrophysiological characteristics at baseline (see Methods). Differentiation batches that presented with abnormal baseline characteristics, such as baseline arrhythmias and cessation in certain wells, were excluded from further analysis (Supplementary Fig. S2).
Deep learning features from voltage traces
A CNN was designed to classify voltage traces as non-arrhythmic, arrhythmic, or asystolic (Fig. 2A, see Supplementary Fig. S1A and Methods for details). For training and testing the CNN, sets of annotated traces are required. First, traces with no spontaneous action potential activity were manually labeled as asystolic (Fig. 2B). To remove human bias regarding the classification of non-arrhythmic versus arrhythmic traces, we employed the following criteria: 1) Traces from wells treated with drugs carrying high risk of inducing TdP arrhythmia [classified according to CiPA 35] and treated at a concentration greater or equal than maximum free plasma concentration (free Cmax) were annotated as arrhythmic. 2) Traces from wells treated with drugs with a no or low-risk CiPA classification and the concentration was less or equal to free Cmax were annotated as non-arrhythmic. Traces that did not follow any of these criteria – i.e., wells treated with drugs of intermediate CiPA risk, wells treated with low doses of high-risk compounds, or high doses of low-risk compounds – were not included in either training or test datasets.
Figure 2. A deep learning neural network for classification of voltage traces.
A) Schematic of CNN classifier for voltage traces into the classes: non-arrhythmic, asystolic, and arrhythmic.
B) Steps for trace annotation
C) Splitting data for training and test datasets.
D) Examples of each trace category and values of class probabilities outputted by the CNN.
E) Confusion matrices for the training and test datasets.
The training dataset comprised traces from healthy donor lines treated with the training compound library proposed by CiPA (Fig. 2C, Supplementary Fig. S1B). The training dataset was used to refine the convolutional layers’ number and size of filters and dropout percentage until an accuracy of 88.6% was achieved. The confusion matrix of the training set (Fig. 2E) revealed that most errors involved traces classified as ‘non-arrhythmic’ by the CNN but were derived from drugs annotated as ‘arrhythmic.’ Further analysis showed that 127 of these 142 misclassified traces were the result of treatment with bepridil, for which previous studies had shown consistently classified as producing non-arrhythmic responses in hiPSC-derived cardiomyocytes despite being tested at concentrations exceeding 100 times the free Cmax 13,14,37.
The accuracy of the trace classifier was verified against different sets of unseen data (Fig 2C, E). The test set 1 contained data from compounds that differed from the training library but in the same cell lines used for training (yielding an accuracy of 98.5%). The test set 2 contained data from different cell lines than those used for training but the same compounds (yielding an accuracy of 86.2%). Lastly, test set 3 contained data from both cell lines and compounds that were not used for training (yielding an accuracy of 97.5%). Note that test set 2 contained unseen data from bepridil, which explains the lower accuracy relative to test sets 1 and 3.
Representative traces and the CNN class probabilities output are shown in Fig. 2D. Briefly, the CNN interpreted whole voltage waveforms from non-arrhythmic, cessation, and arrhythmic classes and generated their respective probabilities (Fig. 2D). These probabilities can be used as a unified set of metrics applicable for all action potential waveforms, overcoming the problems that human-defined metrics suffer when quantifying specific phenotypes (e.g., in asystole APD90 loses its meaning as no action potential exists; or the detection of early after depolarizations (EADs) or delayed after depolarizations (DADs) which often requires human inspection of the trace). Most importantly, the training trace annotations were based on the risk of clinical arrhythmia. This approach allowed the CNN to learn features of the in vitro traces that corresponded to the effect of clinically risky drugs, circumventing the concern that human intuition might not recognize the in vitro features that are predictive of clinical arrhythmia.
Use of the CNN to determine the probability of drug-induced arrhythmia
The trained CNN reflected the dose-dependent proarrhythmic effects in the voltage waveforms of cardiomyocytes treated with high-risk drugs as increases in the probability of arrhythmic or asystolic phenotypes. To illustrate this result, Fig. 3A shows traces from ibutilide treated hiPSC-CMs showing progressively arrhythmic traces with increasing dose [manifested by action potential prolongation and EADs]. The trained CNN generated a continuous, dose-dependent increase in arrhythmia probability based on the waveform input (Fig. 3B). Note that the calculated 50% arrhythmia class probability (EC50, triangle) was lower than the free Cmax value (dashed line), indicating that arrhythmia is detected at therapeutic concentrations of the drug. Consistent with the dependence of contractility on intracellular [Ca2+], the calcium channel blocker nifedipine showed a dose-dependent induction of asystole (Fig. 3C) that corresponded with continuous increase in asystole probability determined by the CNN (Fig. 3D). Similarly, dose-response curves can be calculated for any compound regardless of whether arrhythmic risk or even pharmacokinetic data are known.
Figure 3. Development of the Torsadogenic Safety Margin.
A) Representative traces of different concentrations of the torsadogenic drug ibutilide.
B) Proarrhythmic dose-response curve (solid line) plotting the probability of arrhythmic class from the traces shown in A. 50% probability of arrhythmic value EC50 (triangle). Clinical maximum free plasma concentration, Cmax (dashed line).
C) Representative traces of different concentrations of the calcium channel blocker nifedipine.
D) Proarrhythmic dose-response curve (solid line) plotting the probability of asystole class from the traces shown in A. 50% probability of asystole value EC50 (triangle). Clinical maximum free plasma concentration, Cmax (dashed line). Average of 3 differentiation batches per cell line with 3 technical repeats per dose in each batch.
E) Dose-response curves of for other drugs and their CiPA risk classification.
F) Color-coded dose-response curves where probability of arrhythmic 0-1 is encoded as green-red and the dose is normalized by Cmax.
G) Torsadogenic safety margin for each drug and healthy donor line of the screen. Drug names are color-coded based on CiPA classification.
H) ROC curves for using the Torsadogenic Safety Margin as predictor for a model to identify high-intermediate risk drugs vs. no-low, high vs. no-low, and intermediate vs. no-low.
Quantitative metrics of drug safety
The preceding analysis was applied to the entire dataset (37 drugs, 8 hiPSC lines). Examples of CiPA-classified high (ibutilide), intermediate (ondansetron), and low (verapamil) arrhythmic risk compounds are shown with 3 drugs (flecainide, citalopram, and aspirin) that are not among the CiPA set but showed effects consistent with published clinical data (Fig. 3E, Supplementary Table S1, the entire dataset is shown in Supplementary Data. S1). In general, compounds classified by CiPA as having higher TdP risk displayed lower EC50 values when normalized by Cmax (Fig. 3F). To quantify this observation, we elaborated the Torsadogenic Safety Margin, defined as the log10(EC50/Cmax).
To visualize the relationship between the Torsadogenic safety margin and clinical risk, we rank-ordered the compounds by their Torsadogenic safety margin (Fig. 3G). 8 out of the top 9 places were occupied by drugs classified by CiPA as carrying high risk (Supplementary Table S1). The safety margin generally correlated with the CiPA-assigned clinical risk, although some exceptions were noted. Droperidol, classified as having intermediate risk by CiPA, ranked 7th highest, whereas bepridil, classified as high-risk by CiPA, was ranked 17th, placing it among intermediate-risk drugs. Mexiletine and ranolazine showed safety margin values comparable to intermediate-class drugs, as reported in other hiPSC-CMs studies 13,14,38, despite being considered a low-no risk by CiPA classification.
The Torsadogenic safety margin to classify risk can be used as a predictor in a logistic regression model to evaluate discretized CiPA risk. To compare to existing literature, we created a model to assign drugs to high-intermediate and low-no-risk discrete categories. This model resulted in an AUC value of 0.95 (Fig. 3H), higher than previously proposed models that use human-defined predictors applied to our dataset (Supplementary Fig. S3) 13. In addition, we trained models to distinguish high and low-risk compounds (AUC = 0.97) and intermediate and low-risk (AUC = 0.8) (Fig. 3H).
We computed the Torsadogenic safety margin of 9 additional drugs not contained in the CiPA collection (amiodarone, amitriptyline, aspirin, citalopram, digoxin, epinephrine, erythromycin, flecainide, and fluoxetine). We determined their risk by comparing their rank position against the CiPA reference compounds (Fig. 3G). Flecainide ranked among high-risk drugs, whereas citalopram, fluoxetine, and amiodarone ranked as intermediate-risk, and erythromycin was at the border between high and intermediate-risk. The probability of arrhythmia EC50 values could not be determined for amitriptyline, aspirin, digoxin, and epinephrine, indicating that they are no or low-risk drugs in agreement with the clinical literature on these drugs (Supplementary Table 1). We conclude that the continuous nature of the Torsadogenic safety margin allows it to stratify the risk of drugs at a finer scale than discrete categorization, in which all drugs within the same category (e.g., high risk) are considered equal.
DCM and HCM mutations influence responses to proarrhythmic drugs
Next, we asked whether the CNN could discern the influence of patient genetics on drug-induced arrhythmia. To test this idea, we focused on variants that cause familial DCM and HCM. Certain DCM and HCM-causing variants place patients at risk for ventricular arrhythmias, and current treatment guidelines call for caution in treating familial DCM and HCM patients with Torsadogenic drugs 39,40. The mechanisms responsible for arrhythmia in familial DCM and HCM involve altered Ca2+ and Na+ flux as well as tissue remodeling (e.g., elevated fibrosis) in addition to K+ current inhibition 41–43 that is responsible for most drug-induced TdP 44. Despite these clinical and mechanistic associations, the contribution of DCM and HCM to drug-induced TdP risk has not been addressed quantitatively. Furthermore, an in vitro system to quantify TdP probability would aid in elucidating risk mechanisms.
We used the trained CNN to determine if DCM and HCM mutations increase TdP probabilities in response to drug treatment. To remove the influence of background genetics, we created a panel of isogenic DCM and HCM lines by introducing disease-causing mutations into the healthy donor hiPSC line HD.15S1 (Fig. 4A). The DCM-causing RBM20 p.R634Q and PLN p.R14del variants and the HCM-causing MYBPC3 p.R943X and TPM1 p.K37E variants are recognized as “at-risk” for fatal arrhythmia 29,30,32,33. In contrast, the DCM TNNT2 p.R183W is reported to confer less arrhythmic risk 34.
Figure 4. Influence of DCM and HCM gene variants.
A) Myopathic gene variants were introduced by CRISPR/Cas9 gene editing onto a common healthy donor hiPSC line.
B) Torsadogenic safety margin for each drug and healthy donor line of the screen for the HCM and DCM cell lines and the healthy donor isogenic line. Drug names are color-coded based on CiPA classification.
C) Asystolic safety margin for each drug and healthy donor line of the screen for the HCM and DCM cell lines and the healthy donor isogenic line. Drug names are color-coded based on CiPA classification.
D) Hypothesized model for increased sensitivity of arrhythmogenic cell lines to TdP and asystole (see Discussion).
E) Dose-response curves of probability of arrhythmic in HCM and DCM lines and the isogenic control cell lines, treated with ibutilide, droperidol, vandetanib, nifedipine. Point markers indicate EC50 with error bars at a 95% confidence interval. Shaded region signifies all traces were asystolic at that concentration range. Average of 3 differentiation batches per cell line with 3 technical repeats per dose in each batch.
F) Dose-response curves of probability of asystolic in HCM and DCM lines and the isogenic control cell lines, treated with ibutilide, droperidol, vandetanib, nifedipine. Point markers indicate EC50 with error bars at a 95% confidence interval. Average of 3 differentiation batches per cell line with 3 technical repeats per dose in each batch.
Action potential waveforms in hiPSC-CMs derived from each line were recorded at baseline and upon dose-escalation treatment with the 37 drugs. Results in the mutant lines were compared to the isogenic control to determine the contribution of the gene variants. Most drugs showed similar Torsadogenic safety margins in the DCM and HCM hiPSC-CMs as for the isogenic control, although some trended towards increased risk (e.g., ibutilide, droperidol) (Fig. 4B, E). In contrast, the asystole safety margin (calculated analogously using the EC50 values of the probability of asystole) revealed a strong influence of DCM and HCM genotype on drug effects (Fig 4C). For example, ibutilide, dofetilide, and droperidol showed a heightened propensity to cause asystole in the DCM variants RBM20 p.R634Q and PLN p.R14del, and the HCM causal variants MYBPC3 p.R943X, and TPM1 p.K37E relative to the isogenic control hiPSC-CMs (Fig. 4C, F). Interestingly, hiPSC-CMs carrying the DCM TNNT2 p.R183W variant (that is less arrhythmogenic in patients) had a similar profile to the isogenic healthy donor control (Fig. 4C, F).
Discussion
Current in vitro proarrhythmia assays rely on the measurement of human-defined features of cardiac electrophysiology, such as APD and beat rate 6,13,45. However, arrhythmias present complex geometries, such as EADs, that are challenging to quantify by conventional metrics. In practice, arrhythmic phenotypes are typically classified based on categorical descriptors (e.g., sustained ventricular tachycardia or after depolarizations) or binary variables (e.g., presence or absence of EADs) that require human interpretation of the action potential 13,14,38. Treating arrhythmic phenotypes as yes/no parameters imposes an artificial threshold for an arrhythmia that depends on human intuition and cannot quantify progression from normal to arrhythmic waveforms as a function of drug dose, chemical modification, or hiPSC genetics. More fundamentally, human intuition is not based on an established ground truth regarding the in vitro effect of drugs that cause arrhythmia in patients. Thus, the motivation for this study was that human-defined metrics might not capture features of the in vitro waveforms that correspond with the actual arrhythmic risk of the drugs in people.
We developed our deep learning algorithm to overcome the limitations of human-defined metrics by recognizing features uniquely associated with drug-induced arrhythmia in hiPSC-CMs. Training a CNN to discriminate features was based on a dataset of 3 classes of action potential traces: 1) traces generated by treating hiPSC-CMs with high doses of drugs that cause ventricular arrhythmia in people (class: arrhythmic), 2) traces generated with low doses of safe compounds (class: non-arrhythmic), and 3) traces in which drugs induced asystole, which is characteristic of high doses of Ca2+ channel blockers (class: asystole) but also resulted from treatment with very high doses of proarrhythmic drugs (Fig. 2). The probability of classification as arrhythmic and asystolic was a continuous dose-dependent metric that quantified the behavior of a drug. Relating the EC50 values for these probabilities to the free plasma concentration of a drug used in clinical practice (free Cmax) defined a Torsadogenic safety margin for each drug (Fig. 3). In hiPSC-CMs from healthy donors, the Torsadogenic safety margin accurately predicted the clinical risk of drugs with an AUC of 0.95 (Fig. 3H), representing an improvement over previously published multiparametric methods based on human classification (Supplementary Fig. 3).
Patients with structural heart diseases such as DCM and HCM are at risk for drug-induced arrhythmia. They should be carefully monitored using electrocardiographic and other modalities when treated with drugs at risk for inducing arrhythmia 40. We applied the Torsadogenic and asystolic safety margins to traces generated by treating isogenic hiPSC-CMs carrying gene variants that cause DCM and HCM in patients. The CNN probabilities and the calculated safety margins revealed that pathological gene variants associated with arrhythmic cardiomyopathies in patients sensitized hiPSC-CMs to adverse proarrhythmic and asystolic effects of high-risk drugs. In particular, hiPSC-CMs carrying pathogenic variants in PLN, RBM20, TPM1, and MYBPC3 associated with increased arrhythmic risk in patients 29,30,32,33 trended towards increased probabilities of being classified as arrhythmic (Fig. 4B, D) and showed highly significant increases in the probabilities of asystole (Fig. 4C, E) compared to isogenic controls.
The finding that arrhythmogenic DCM and HCM gene variants increase the risk of Torsadogenic drugs has implications for understanding the electrophysiological substrates for arrhythmia. The genetic lesions examined here affect sarcomeric, RNA splicing, and Ca2+ handling proteins. Electrophysiological remodeling in familial DCM and HCM includes reduced repolarizing K+ currents (IK) and increased intracellular diastolic Ca2+ and late Na+ (INaL) current, as reviewed 42,43. For example, these current changes have been reported in isolated cardiomyocytes from mice, human myectomy samples, and hiPSCs carrying MYBPC3 mutations that are functionally equivalent to the p.R943X truncation mutation used in this study 41,46–48. A downstream circuit involving CaMKII sustains electrophysiological remodeling 49,50 and decreases repolarizing IK 51. Decreased IK in the hiPSC-CMs, which are more depolarized relative to adult cardiomyocytes (at least in monolayer culture) 52,53, would enhance the effect of Torsadogenic drugs 44 and is a possible mechanism for their asystolic effect in hiPSC-CMs carrying the DCM and HCM mutations (Fig. 4D).
In conclusion, the deep learning algorithm recognized in vitro arrhythmic features in the hiPSC-CMs based on drug effects. It did not rely on human adjudication nor human-defined in vitro hiPSC-CM arrhythmic phenotypes; instead, it focused the CNN on recognizing hiPSC-CM phenotypes associated with patient drug responses. We derived safety margins from the relationship between the machine-generated class probabilities and the free plasma concentrations of each drug. Unlike categorical descriptors of arrhythmia (such as EADs, action potential prolongation, and triangulation), the CNN class probabilities map each trace to a continuous spectrum spanning non-arrhythmic to arrhythmic and asystolic phenotypes. Overall, the calculated safety margins more accurately discriminated high, intermediate, and low-risk drugs than prior methods based on human-defined features, and revealed arrhythmogenic HCM and DCM variants increase sensitivity to drug-induced arrhythmia. Thus, the recognition of hiPSC-CM features of arrhythmia by deep learning should improve the detection of risky compounds during development as well as assign risk to gene variants of unknown significance.
Limitations of the study
Deep learning has the inherent limitation that the machine-learned features typically lack human-intelligible meaning (the so-called ‘black box’ problem). In other words, the mathematical operations that the machine has optimized to calculate the probability of arrhythmia are too abstract to be interpretable by humans and cannot be ascribed to action potential features. Although we cannot query the CNN to understand the basis for classification, visual inspection of the traces revealed that the dose-dependent increases in arrhythmia probability corresponded to AP prolongation, EADs, and alternans. The CiPA reference collection causes a limited range of arrhythmogenic phenotypes (typically AP prolongation and EADs). We have found that the CNN (RS, unpublished data) can be applied to a broader range of phenotypes [e.g., DADs, sustained and non-sustained ventricular tachycardia [Sustained VT (SVT), Non-sustained VT (NSVT)] associated with cardiac electrophysiological disorders 54–56] by increasing the number of possible classifications (i.e., the number nodes in the last layer of the CNN).
Certain drugs in this study (droperidol, ranolazine and mexiletine) presented as “riskier” than in actual clinical practice while bepridil appeared less “risky” as discussed above. Previous studies using hiPSC-CMs similarly misclassified these drugs 13,37. This might reflect a limitation in using hiPS-CM datasets to classify these drugs. Concerning the deep learning methodology, we speculate that the features needed to accurately classify these drugs might have been masked by variability in the waveforms in the current dataset, and that a larger dataset might be adequately powered to train the neural network to correctly define the risk of these drugs.
STAR METHODS
RESOURCE AVAILABILITY
Lead Contact
Further information and request for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Mark Mercola (mmercola@stanford.edu).
Materials Availability
All unique reagents generated in this study are available from the Lead Contact with a completed Materials Transfer Agreement.
Data and Code Availability
The datasets and codes generated during this study are available in the Supplementary Information and publicly available from GitHub (https://github.com/rikserrano/DeepLearning_TdP_Risk).
EXPERIMENTAL MODEL AND SUBJECT DETAILS
hiPSCs culture and differentiation
The protocols used in this study were approved by the Stanford University Institutional Review Board. Sendai viral vector was used for the reprogramming of human PBMCs. The de-identified healthy donor hiPSC lines HD.113 (male), HD.15S1 (male), and HD.273 (female) were obtained from the Stanford Cardiovascular Institute Biobank. The hiPSC clones were cultured in E8 cell culture media (Thermo Fisher Scientific, A1517001) in plates coated with growth factor-reduced Matrigel (Corning, 356231). The hiPSC were differentiated into CMs utilizing a chemically defined cardiomyocyte differentiation protocol 57 with modifications. Briefly, hiPSCs were treated with CHIR99021 (Tocris, 4423) for 3 days in RPMI 1640 (Thermo Fisher Scientific, 11875119) with B27-insulin (Thermo Fisher Scientific, A1895601). Subsequently, the cells were treated with the Wnt inhibitor C59 (Tocris, 5148) in RPMI supplemented with B27-insulin (RPMI/B27-) for another 2 days. Between 5–11 days of differentiation, RPMI/B27-media refreshed every other day and switched to RPMI supplemented with 1X B27(RPMI/B27+) after beating was observed. On day 11, cells were cultured in RPMI/B27+ without glucose, supplemented with 5 mM sodium L-lactate (Sigma Aldrich, 71718) for 3 days, to improve the CMs purity. On day 14, the cells were cultured on RPMI/B27+. On day 15, the hiPSC-CMs were dissociated with TrypLE 10x (Thermo Fisher Scientific, A1217703) and seeded in 6-well Matrigel-coated plates at a density of 3x106 per well in RPMI/B27+ containing 10% KOSR and ROCK inhibitor Y-27632 (Tocris, 1253) (replating media). After 2 days, cells were cultured in RPMI/B27+ without glucose for 3 days and then switched to maturation media 58 and cultured for an additional 4-5 weeks prior to replating in 384-well plates for high throughput analysis.
Genome Editing in hiPSCs
The MYBPC3 p. R943X, TPM1 p. K37E, PLN p.R14del, RBM20 p. R634Q, and TNNT2 p.R183W mutation were introduced to the HD.15s1 iPSC line as previously described 24,25,26,27,28 Briefly, CRISPR guide RNA (gRNA) sequences were designed using an online tool (http://crispr.mit.edu/), and we selected the gRNAs with the highest score (specificity) for cloning into into the BbsI site of the pSpCas9(BB)-2A-GFP (PX458; a gift from Feng Zhang; Addgene plasmid #48138) by reverse complementary guide DNA oligos. Briefly, oligos were annealed in T4 ligation buffer (NEB) and phosphorylated with T4 PNK (NEB). The annealed and phosphorylated oligos were cloned into the BbsI sites of the pSpCas9(BB)-2A-GFP plasmid 59 and transformed in STBL3 E. coli cells. The Cas9-gRNA vector (1.0μg) was co-transfected with single-stranded oligodeoxynucleotide (ssODN, 4.0μg) into hiPSCs cultured on 6-well plate using 10 μL Lipofectamine Stemfect (ThermoFisher Scientific). At 24 hours after transfection, the cells were dissociated with TrypLE Express (ThermoFisher Scientific) and GFP+ cells were sorted and plated in 6-well plates at a density of 2,000 cells per well in E8 media supplemented with Rock inhibitor (Y-27632, 5 μM). Single cell clones were manually picked and expanded. To verify the on-target HDR, the clones were genotyped by direct PCR and Sanger sequencing.
METHOD DETAILS
High throughout optical voltage assay
hiPSC-CMs were plated on Matrigel at 25,000 cells per well of a 384-well plate (Greiner Bio-One) in 50 μl replating media. The subsequent day an additional 50 μl maturation media was added and cells were grown for a minimum of 4 days prior to analysis. For the analysis, hiPSC-CMs were removed from a 37°C 5% CO2 incubator and placed immediately into a tissue culture cabinet on a dry heat block set to 37°C to prevent temperature fluctuation during the subsequent washing and dye loading steps. Cells were washed by removing 50 μl media and replacing it with 50 μl FluoroBrite DMEM (Gibco) solution five times. After the last wash step, 50 μL of the 2x VF2.1.Cl dye with Hoechst 33258 (Life Technologies) in FluoroBrite was added to each well 45. The plate was returned to the 37°C 5% CO2 incubator for 50 min. After incubation, cells were washed four times in FluoroBrite as described above. After a 20min recovery in the incubator, 2x reference drug was added to the respective well and incubated at 37°C and 5% CO2 for 15 min before image acquisition. Time series images were acquired automatically using the IC200 KIC instrument (Vala Sciences) at an acquisition frequency of 33 Hz for a duration of 10 seconds. The voltage image analysis and physiological parameter calculation were conducted using commercially available Cyteseer (Vala Sciences) as previously described 45.
Convolutional Neural Network Classifier
The convolutional neural network classifier was implemented using R interface to Keras 60 and RStudio (PBC, Boston, MA http://www.rstudio.com/). A schematic view of our architecture is shown in Supplementary Fig. S1. Briefly, an input layer with 330 nodes (one per data point of the voltage trace) receives each normalized voltage trace. The following formula was used to normalize the traces , where and are the maximum and minimum fluorescence intensity signal of all traces recorded from wells within a multiwell plate. Then two convolutional layers with 33 filters and kernel size five. Next, a max pool 1D layer with a pool size of three. After, two more convolutional layers with 66 filters and kernel size of ten. Later, a global average pooling. A dropout of 20% was added to decrease overfitting. Finally, a dense layer with 3 nodes and softmax activation for the output of the classifier. The experimental design, deep neural network classifier architecture and representative traces are described in Fig. 2A–C.
The network was trained on a training set comprising 645 “Non-arrhythmic”, 550 “Arrhythmic”, and 150 “Asystolic” traces (see examples in Fig. 2D), from the three healthy donor lines. The traces labelled as arrhythmic were from wells treated with CiPA high risk drugs at doses higher or equal than maximum free plasma concentration Cmax. The traces labelled as non-arrhythmic were from wells treated with CiPA no-low risk drugs at doses lower or equal than maximum free plasma concentration Cmax. The asystolic traces were manually identified in the dataset. The hyperparameters of the network were manually tuned to achieve 88.6% accuracy using Adam optimizer with categorical cross-entropy as loss function. After hyperparameter tuning, the trained network was tested on three different datasets to check performance against effects of unseen drugs in the same cell lines (test set 1), the effects of training drugs in the HCM/DCM lines (test set 2), and the effects of unseen drugs in HCM/DCM lines (test set 3). Accuracies and confusion matrices are shown in Fig. 2E.
QUANTIFICATION AND STATISTICAL ANALYSIS
28 independent differentiation batches of hiPSC were utilized in this study, some of them were rejected during quality control (Supplementary Fig. S1). Cell line name: included batches-rejected batches. HD.113: 3-1; rejected; HD.15S1: 3-0; HD.273: 5-0; HCM.TPM1: 4-2; HCM.MYBPC3: 3-2; DCM.TNNT2: 3-1; DCM.RBM20: 3-1; DCM.PLN: 3-0. All compounds of the library were tested in an 8-dose response curve with 3 technical replicates.
Probability of arrhythmic and asystolic dose-response curves
Dose-response curves were fitted using the binomial fit from the generalized linear model function in R ‘glm’. The logarithm of the dose was provided as ‘x-data’ and the probability of arrhythmic from the CNN classifier was provided as ‘y-data’.
Calculation of AUC of discretized TdP safety margin
The accuracy of using the Torsadogenic safety margin (Fig. 3G) against the discretized proarrhythmic classification of drugs was reported as the area under the curve of a binomial model with the Torsadogenic safety margin as its sole predictor.
Logit(P) = (Torsadogenic safety margin) ,where and P is the probability of a drug of being High-intermediate, High, or Intermediate (Fig. 3H). The models were trained following k-fold cross-validation with k=10, in R using the ‘glm.fit’ function. Receiver Operating Characteristic (ROC) curves were generated in R using the ‘plotROC’ package.
Supplementary Material
KEY RESOURCES TABLE
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Chemicals, Peptides, and Recombinant Proteins | ||
CHIR99021 | Tocris | Cat#4423 |
Wnt-C59 | Tocris | Cat#5148 |
Y-27632 dihydrochloride | Tocris | Cat#1254 |
Amiodarone | Tocris | Cat#4095 |
Amitriptyline | Tocris | Cat#4456 |
Aspirin | Sigma-Aldrich | Cat#A5376-100G |
Astemizole | Santa Cruz Biotechnology | Cat#sc-201088 |
Azimilide | Fisher Scientific | Cat#501151950 |
Bepridil | Santa Cruz Biotechnology | Cat#sc-202974 |
Chlorpromazine | Santa Cruz Biotechnology | Cat#sc-202537 |
Cisapride | Abcam | Cat#ab120548 |
Citalopram | Tocris | Cat#1427 |
Clarithromycin | Abcam | Cat#ab141202 |
Clozapine | Santa Cruz Biotechnology | Cat#sc-200402 |
Digoxin | Tocris | Cat#4583 |
Diltiazem | Santa Cruz Biotechnology | Cat#sc-200199 |
Disopyramide | Sigma-Aldrich | Cat#D6035-1G |
Dofetilide | Tocris | Cat#3757 |
Domperidone | Santa Cruz Biotechnology | Cat#sc-203032 |
Droperidol | Fisher Scientific | Cat#585850 |
Epinephrine | Sigma-Aldrich | Cat#E4250-1G |
Erythromycin | Sigma-Aldrich | Cat#E5389-1G |
Flecainide | Tocris | Cat#1470 |
Fluoxetine | Tocris | Cat#0927 |
Ibutilide | Fisher Scientific | Cat#501362076 |
Loratadine | Fisher Scientific | Cat#501361172 |
Metoprolol | Abcam | Cat#Ab120711 |
Mexiletine | Tocris | Cat#2596 |
Nifedipine | Abcam | Cat#ab120135 |
Nitrendipine | Fisher Scientific | Cat#507407 |
Ondansetron | Fisher Scientific | Cat#501361204 |
Pimozide | Santa Cruz Biotechnology | Cat#sc-203662 |
Quinidine | Fisher Scientific | Cat#AAJ6042606 |
Ranolazine | Tocris | Cat#3118 |
Risperidone | Fisher Scientific | Cat#505885 |
Sotalol | Tocris | Cat#0952 |
Tamoxifen | Abcam | Cat#Ab120656 |
Terfenadine | Fisher Scientific | Cat#394850 |
Vandetanib | Selleckchem | Cat#S1046 |
Verapamil | Tocris | Cat#0654 |
Experimental Models: Cell Lines | ||
Human iPSC line (healthy donor) | SCVI Biobank | SCVI-273 |
Human iPSC line (healthy donor) | SCVI Biobank | SCVI-15S1 |
Human iPSC line (healthy donor) | SCVI Biobank | SCVI-113 |
Human iPSC line DCM.TNNT2 | (Perea-Gil et al., 2022) | TNNT2 R173W |
Human iPSC line DCM.RBM20 | (Briganti et al., 2020) | RBM20 R634Q |
Human iPSC line DCM.PLN | (Feyen et al., 2021) | PLN R14del C9 |
Human iPSC line HCM.TPM1 | (Perea-Gil et al. 2020) | TPM1 K37E 8 |
Human iPSC line HCM.MYBPC3 | (Seeger et al., 2019) | MYBPC3 R943x |
Oligonucleotides | ||
Single guide RNA sequence for DCM.TNNT2: GGACAAAGCCTTCTTCTTCC |
Integrated DNA Technologies | |
Single guide RNA sequence for HCM.TPM1: GGCGGAAGACAGGAGCAAGC |
Integrated DNA Technologies | |
HDR sequence DCM.TNNT2: TAATTTGCTTTCTTCCTCCCTGCTGTAAATCAG GAAGAGAGGGCTCGACGAGAGGAGGAGGAGA ACAGGAGGAAGGCTGAGGATGAGGCATGGAA GAAGAAGGCTTTGTCCAACATGATGCATTTTG |
ThermoFisher Scientific | |
HDR sequence HCM.TPM1: CCGAGTCCTCGGGGTGGGGATCCAGCCGGGG GTGCCAGGCTCGAGTCCCGGCGCTCTGGGCG CGGGCGCAGGGCCGGGGAGGCGCAGACCTG CTCGCTCCTGTCTTCCGCCGCCTTCTTGTCGG CCT |
ThermoFisher Scientific | |
Recombinant DNA | ||
Plasmid: pSpCas9(BB)-2A-GFP | Addgene | RRID:Addgene_48138 |
Software and Algorithms | ||
CyteSeer | Vala Sciences | RRID:SCR_012232 |
RStudio | PBC | RRID:SCR_000432 |
Codes and Datasets | This study and Github | http://doi.org/10.5281/zenodo.7348974 |
Highlights:
Deep learning detects in vitro features that correlate with clinical arrhythmia
Drug safety margin from AI identifies drug risk with AUC = 0.95
Cardiomyopathic genotypes increase sensitivity to drug proarrhythmia risk
Acknowledgments
This research was made possible by grants from the National Institutes of Health (R01HL130840, R01HL138539, R01HL141358, 1R01HL152055, 1R42HL158510, and Fondation Leducq 18CVD01 to MM, P01HL141084 to MM and JCW, American Heart Association 17MERIT33610009 and Fondation Leducq 18CVD05 to JCW and R01HL150414 and R01HL139679 to IK) and the Joan and Sanford I. Weill Scholars Endowment. DAMF was funded by the European Union’s Horizon 2020 Research and Innovation Programme under the Marie Sklodowska-Curie grant agreement No. 708459.Graphical abstract was created with BioRender.com.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of Interests
RR is a paid consultant of Vala Sciences, which manufactures a high content instrument used in these studies. MM serves on the scientific advisory board of Vala Sciences. JCW is co-founder and scientific advisory board of Greenstone Biosciences. The other authors declare no competing financial interests.
References
- 1.Valentin J-P, and Delaunois A (2018). Developing solutions to detect and avoid cardiovascular toxicity in the clinic. Toxicology Letters 295, S48–S49. [Google Scholar]
- 2.Onakpoya IJ, Heneghan CJ, and Aronson JK (2016). Post-marketing withdrawal of 462 medicinal products because of adverse drug reactions: a systematic review of the world literature. BMC Med 14, 10. 10.1186/s12916-016-0553-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, Pairaudeau G, Pennie WD, Pickett SD, Wang J, et al. (2015). An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat Rev Drug Discov 14, 475–486. 10.1038/nrd4609. [DOI] [PubMed] [Google Scholar]
- 4.Baguley WA, Hay WT, Mackie KP, Cheney FW, and Cullen BF (1997). Cardiac dysrhythmias associated with the intravenous administration of ondansetron and metoclopramide. Anesth Analg 84, 1380–1381. 10.1097/00000539-199706000-00038. [DOI] [PubMed] [Google Scholar]
- 5.Frommeyer G, and Eckardt L (2016). Drug-induced proarrhythmia: risk factors and electrophysiological mechanisms. Nat Rev Cardiol 13, 36–47. 10.1038/nrcardio.2015.110. [DOI] [PubMed] [Google Scholar]
- 6.Liang P, Lan F, Lee AS, Gong T, Sanchez-Freire V, Wang Y, Diecke S, Sallam K, Knowles JW, Wang PJ, et al. (2013). Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation 127, 1677–1691. 10.1161/CIRCULATIONAHA.113.001883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Laverty H, Benson C, Cartwright E, Cross M, Garland C, Hammond T, Holloway C, McMahon N, Milligan J, Park B, et al. (2011). How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines? Br J Pharmacol 163, 675–693. 10.1111/j.1476-5381.2011.01255.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gintant G, Sager PT, and Stockbridge N (2016). Evolution of strategies to improve preclinical cardiac safety testing. Nat Rev Drug Discov 15, 457–471. 10.1038/nrd.2015.34. [DOI] [PubMed] [Google Scholar]
- 9.Heist EK, and Ruskin JN (2010). Drug-induced arrhythmia. Circulation 122, 1426–1435. 10.1161/CIRCULATIONAHA.109.894725. [DOI] [PubMed] [Google Scholar]
- 10.Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, and Radisic M (2019). Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 198, 3–26. 10.1016/j.biomaterials.2018.09.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Paik DT, Chandy M, and Wu JC (2020). Patient and Disease-Specific Induced Pluripotent Stem Cells for Discovery of Personalized Cardiovascular Drugs and Therapeutics. Pharmacol Rev 72, 320–342. 10.1124/pr.116.013003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hnatiuk AP, Briganti F, Staudt D, and Mercola M (2021). Human iPSC Modeling of Heart Disease for Drug Development. Cell Chem Biol in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Blinova K, Dang Q, Millard D, Smith G, Pierson J, Guo L, Brock M, Lu HR, Kraushaar U, Zeng H, et al. (2018). International Multisite Study of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Drug Proarrhythmic Potential Assessment. Cell Rep 24, 3582–3592. 10.1016/j.celrep.2018.08.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pfeiffer ER, Vega R, McDonough PM, Price JH, and Whittaker R (2016). Specific prediction of clinical QT prolongation by kinetic image cytometry in human stem cell derived cardiomyocytes. J Pharmacol Toxicol Methods 81, 263–273. 10.1016/j.vascn.2016.04.007. [DOI] [PubMed] [Google Scholar]
- 15.Antzelevitch C (2004). Arrhythmogenic mechanisms of QT prolonging drugs: is QT prolongation really the problem? J Electrocardiol 37 Suppl, 15–24. [DOI] [PubMed] [Google Scholar]
- 16.Vicente J, Stockbridge N, and Strauss DG (2016). Evolving regulatory paradigm for proarrhythmic risk assessment for new drugs. J Electrocardiol 49, 837–842. 10.1016/j.jelectrocard.2016.07.017. [DOI] [PubMed] [Google Scholar]
- 17.Bhuiyan TA, Graff C, Kanters JK, Melgaard J, Toft E, Kaab S, and Struijk JJ (2018). A History of Drug-Induced Torsades de Pointes Is Associated With T-wave Morphological Abnormalities. Clin Pharmacol Ther 103, 1100–1106. 10.1002/cpt.886. [DOI] [PubMed] [Google Scholar]
- 18.LeCun Y, Bengio Y, and Hinton G (2015). Deep learning. Nature 521, 436–444. 10.1038/nature14539. [DOI] [PubMed] [Google Scholar]
- 19.Ker J, Wang L, Rao J, and Lim T (2018). Deep Learning Applications in Medical Image Analysis. IEEE Access 6, 9375–9389. 10.1109/ACCESS.2017.2788044. [DOI] [Google Scholar]
- 20.Faust O, Hagiwara Y, Hong TJ, Lih OS, and Acharya UR (2018). Deep learning for healthcare applications based on physiological signals: A review. Comput Methods Programs Biomed 161, 1–13. 10.1016/j.cmpb.2018.04.005. [DOI] [PubMed] [Google Scholar]
- 21.Golgooni Z, Mirsadeghi S, Baghshah MS, Ataee P, Baharvand H, Pahlavan S, and Rabiee HR (2019). Deep Learning-Based Proarrhythmia Analysis Using Field Potentials Recorded From Human Pluripotent Stem Cells Derived Cardiomyocytes. IEEE Journal of Translational Engineering in Health and Medicine 7, 1–9. 10.1109/JTEHM.2019.2907945. [DOI] [Google Scholar]
- 22.Hannun AY, Rajpurkar P, Haghpanahi M, Tison GH, Bourn C, Turakhia MP, and Ng AY (2019). Cardiologist-level arrhythmia detection and classification in ambulatory electrocardiograms using a deep neural network. Nat Med 25, 65–69. 10.1038/s41591-018-0268-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Petropoulou E, Jamshidi Y, and Behr ER (2014). The genetics of pro-arrhythmic adverse drug reactions. Br J Clin Pharmacol 77, 618–625. 10.1111/bcp.12208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Seeger T, Shrestha R, Lam CK, Chen C, McKeithan WL, Lau E, Wnorowski A, McMullen G, Greenhaw M, Lee J, et al. (2019). A Premature Termination Codon Mutation in MYBPC3 Causes Hypertrophic Cardiomyopathy via Chronic Activation of Nonsense-Mediated Decay. Circulation 139, 799–811. 10.1161/CIRCULATIONAHA.118.034624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gil IP, Bellbachir N, Gavidia AA, Arthur J, Zhang Y, Vadgama N, Oikonomopoulos A, Roura S, Wu JC, Bayes-Genis A, and Karakikes I (2020). Activation of CaMKII Signaling Pathway Contributes to the Pathogenesis of Genetic Hypertrophic Cardiomyopathy. Circ Res 127. 10.1161/res.127.suppl_1.274. [DOI] [Google Scholar]
- 26.Feyen DAM, Perea-Gil I, Maas RGC, Harakalova M, Gavidia AA, Arthur Ataam J, Wu TH, Vink A, Pei J, Vadgama N, et al. (2021). Unfolded Protein Response as a Compensatory Mechanism and Potential Therapeutic Target in PLN R14del Cardiomyopathy. Circulation 144, 382–392. 10.1161/CIRCULATIONAHA.120.049844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Briganti F, Sun H, Wei W, Wu J, Zhu C, Liss M, Karakikes I, Rego S, Cipriano A, Snyder M, et al. (2020). iPSC Modeling of RBM20-Deficient DCM Identifies Upregulation of RBM20 as a Therapeutic Strategy. Cell Rep 32, 108117. 10.1016/j.celrep.2020.108117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Perea-Gil I, Seeger T, Bruyneel AAN, Termglinchan V, Monte E, Lim EW, Vadgama N, Furihata T, Gavidia AA, Arthur Ataam J, et al. (2022). Serine biosynthesis as a novel therapeutic target for dilated cardiomyopathy. Eur Heart J 43, 3477–3489. 10.1093/eurheartj/ehac305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Niimura H, Bachinski LL, Sangwatanaroj S, Watkins H, Chudley AE, McKenna W, Kristinsson A, Roberts R, Sole M, Maron BJ, et al. (1998). Mutations in the gene for cardiac myosin-binding protein C and late-onset familial hypertrophic cardiomyopathy. N Engl J Med 338, 1248–1257. [DOI] [PubMed] [Google Scholar]
- 30.Chang B, Nishizawa T, Furutani M, Fujiki A, Tani M, Kawaguchi M, Ibuki K, Hirono K, Taneichi H, Uese K, et al. (2011). Identification of a novel TPM1 mutation in a family with left ventricular noncompaction and sudden death. Mol Genet Metab 102, 200–206. 10.1016/j.ymgme.2010.09.009. [DOI] [PubMed] [Google Scholar]
- 31.Haghighi K, Kolokathis F, Gramolini AO, Waggoner JR, Pater L, Lynch RA, Fan GC, Tsiapras D, Parekh RR, Dorn GW 2nd, et al. (2006). A mutation in the human phospholamban gene, deleting arginine 14, results in lethal, hereditary cardiomyopathy. Proc Natl Acad Sci U S A 103, 1388–1393. 10.1073/pnas.0510519103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.van der Zwaag PA, van Rijsingen IA, Asimaki A, Jongbloed JD, van Veldhuisen DJ, Wiesfeld AC, Cox MG, van Lochem LT, de Boer RA, Hofstra RM, et al. (2012). Phospholamban R14del mutation in patients diagnosed with dilated cardiomyopathy or arrhythmogenic right ventricular cardiomyopathy: evidence supporting the concept of arrhythmogenic cardiomyopathy. Eur J Heart Fail 14, 1199–1207. 10.1093/eurjhf/hfs119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Parikh VN, Caleshu C, Reuter C, Lazzeroni LC, Ingles J, Garcia J, McCaleb K, Adesiyun T, Sedaghat-Hamedani F, Kumar S, et al. (2019). Regional Variation in RBM20 Causes a Highly Penetrant Arrhythmogenic Cardiomyopathy. Circ Heart Fail 12, e005371. 10.1161/CIRCHEARTFAILURE.118.005371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Campbell N, Sinagra G, Jones KL, Slavov D, Gowan K, Merlo M, Carniel E, Fain PR, Aragona P, Di Lenarda A, et al. (2013). Whole exome sequencing identifies a troponin T mutation hot spot in familial dilated cardiomyopathy. PLoS One 8, e78104. 10.1371/journal.pone.0078104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sager PT, Gintant G, Turner JR, Pettit S, and Stockbridge N (2014). Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. Am Heart J 167, 292–300. 10.1016/j.ahj.2013.11.004. [DOI] [PubMed] [Google Scholar]
- 36.Colatsky T, Fermini B, Gintant G, Pierson JB, Sager P, Sekino Y, Strauss DG, and Stockbridge N (2016). The Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative - Update on progress. J Pharmacol Toxicol Methods 81, 15–20. 10.1016/j.vascn.2016.06.002. [DOI] [PubMed] [Google Scholar]
- 37.Ando H, Yoshinaga T, Yamamoto W, Asakura K, Uda T, Taniguchi T, Ojima A, Shinkyo R, Kikuchi K, Osada T, et al. (2017). A new paradigm for drug-induced torsadogenic risk assessment using human iPS cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 84, 111–127. 10.1016/j.vascn.2016.12.003. [DOI] [PubMed] [Google Scholar]
- 38.Kitaguchi T, Moriyama Y, Taniguchi T, Ojima A, Ando H, Uda T, Otabe K, Oguchi M, Shimizu S, Saito H, et al. (2016). CSAHi study: Evaluation of multi-electrode array in combination with human iPS cell-derived cardiomyocytes to predict drug-induced QT prolongation and arrhythmia--effects of 7 reference compounds at 10 facilities. J Pharmacol Toxicol Methods 78, 93–102. 10.1016/j.vascn.2015.12.002. [DOI] [PubMed] [Google Scholar]
- 39.Maron BJ, Rowin EJ, and Maron MS (2022). Hypertrophic Cardiomyopathy: New Concepts and Therapies. Annu Rev Med 73, 363–375. 10.1146/annurev-med-042220-021539. [DOI] [PubMed] [Google Scholar]
- 40.Tisdale JE, Chung MK, Campbell KB, Hammadah M, Joglar JA, Leclerc J, Rajagopalan B, American Heart Association Clinical Pharmacology Committee of the Council on Clinical, C., Council on, C., and Stroke, N. (2020). Drug-Induced Arrhythmias: A Scientific Statement From the American Heart Association. Circulation 142, e214–e233. 10.1161/CIR.0000000000000905. [DOI] [PubMed] [Google Scholar]
- 41.Toib A, Zhang C, Borghetti G, Zhang X, Wallner M, Yang Y, Troupes CD, Kubo H, Sharp TE, Feldsott E, et al. (2017). Remodeling of repolarization and arrhythmia susceptibility in a myosin-binding protein C knockout mouse model. Am J Physiol Heart Circ Physiol 313, H620–H630. 10.1152/ajpheart.00167.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Santini L, Coppini R, and Cerbai E (2021). Ion Channel Impairment and Myofilament Ca(2+) Sensitization: Two Parallel Mechanisms Underlying Arrhythmogenesis in Hypertrophic Cardiomyopathy. Cells 10. 10.3390/cells10102789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Maron BJ, Rowin EJ, and Maron MS (2019). Paradigm of Sudden Death Prevention in Hypertrophic Cardiomyopathy. Circ Res 125, 370–378. 10.1161/CIRCRESAHA.119.315159. [DOI] [PubMed] [Google Scholar]
- 44.Roden DM (2008). Cellular basis of drug-induced torsades de pointes. Br J Pharmacol 154, 1502–1507. 10.1038/bjp.2008.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.McKeithan WL, Savchenko A, Yu MS, Cerignoli F, Bruyneel AAN, Price JH, Colas AR, Miller EW, Cashman JR, and Mercola M (2017). An Automated Platform for Assessment of Congenital and Drug-Induced Arrhythmia with hiPSC-Derived Cardiomyocytes. Front Physiol 8, 766. 10.3389/fphys.2017.00766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Coppini R, Ferrantini C, Yao L, Fan P, Del Lungo M, Stillitano F, Sartiani L, Tosi B, Suffredini S, Tesi C, et al. (2013). Late sodium current inhibition reverses electromechanical dysfunction in human hypertrophic cardiomyopathy. Circulation 127, 575–584. 10.1161/CIRCULATIONAHA.112.134932. [DOI] [PubMed] [Google Scholar]
- 47.Flenner F, Jungen C, Kupker N, Ibel A, Kruse M, Koivumaki JT, Rinas A, Zech ATL, Rhoden A, Wijnker PJM, et al. (2021). Translational investigation of electrophysiology in hypertrophic cardiomyopathy. J Mol Cell Cardiol 157, 77–89. 10.1016/j.yjmcc.2021.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wu H, Yang H, Rhee JW, Zhang JZ, Lam CK, Sallam K, Chang ACY, Ma N, Lee J, Zhang H, et al. (2019). Modelling diastolic dysfunction in induced pluripotent stem cell-derived cardiomyocytes from hypertrophic cardiomyopathy patients. Eur Heart J 40, 3685–3695. 10.1093/eurheartj/ehz326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fischer TH, Herting J, Mason FE, Hartmann N, Watanabe S, Nikolaev VO, Sprenger JU, Fan P, Yao L, Popov AF, et al. (2015). Late INa increases diastolic SR-Ca2+-leak in atrial myocardium by activating PKA and CaMKII. Cardiovasc Res 107, 184–196. 10.1093/cvr/cvv153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Morotti S, Edwards AG, McCulloch AD, Bers DM, and Grandi E (2014). A novel computational model of mouse myocyte electrophysiology to assess the synergy between Na+ loading and CaMKII. J Physiol 592, 1181–1197. 10.1113/jphysiol.2013.266676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hegyi B, Borst JM, Bailey LRJ, Shen EY, Lucena AJ, Navedo MF, Bossuyt J, and Bers DM (2020). Hyperglycemia regulates cardiac K(+) channels via O-GlcNAc-CaMKII and NOX2-ROS-PKC pathways. Basic Res Cardiol 115, 71. 10.1007/s00395-020-00834-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ, Kolaja KL, Swanson BJ, and January CT (2011). High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. American journal of physiology. Heart and circulatory physiology 301, H2006–2017. 10.1152/ajpheart.00694.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Horvath A, Lemoine MD, Loser A, Mannhardt I, Flenner F, Uzun AU, Neuber C, Breckwoldt K, Hansen A, Girdauskas E, et al. (2018). Low Resting Membrane Potential and Low Inward Rectifier Potassium Currents Are Not Inherent Features of hiPSC-Derived Cardiomyocytes. Stem Cell Reports 10, 822–833. 10.1016/j.stemcr.2018.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.McKeithan WL, Feyen DAM, Bruyneel AAN, Okolotowicz KJ, Ryan DA, Sampson KJ, Potet F, Savchenko A, Gomez-Galeno J, Vu M, et al. (2020). Reengineering an Antiarrhythmic Drug Using Patient hiPSC Cardiomyocytes to Improve Therapeutic Potential and Reduce Toxicity. Cell Stem Cell 27, 813–821 e816. 10.1016/j.stem.2020.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, Feldman O, Gepstein A, Arbel G, Hammerman H, et al. (2011). Modelling the long QT syndrome with induced pluripotent stem cells. Nature 471, 225–229. 10.1038/nature09747. [DOI] [PubMed] [Google Scholar]
- 56.Prajapati C, Ojala M, and Aalto-Setala K (2018). Divergent effects of adrenaline in human induced pluripotent stem cell-derived cardiomyocytes obtained from hypertrophic cardiomyopathy. Dis Model Mech 11. 10.1242/dmm.032896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp TJ, and Palecek SP (2012). Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci U S A 109, E1848–1857. 10.1073/pnas.1200250109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Feyen DAM, McKeithan WL, Bruyneel AAN, Spiering S, Hormann L, Ulmer B, Zhang H, Briganti F, Schweizer M, Hegyi B, et al. (2020). Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep 32, 107925. 10.1016/j.celrep.2020.107925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, and Zhang F (2013). Genome engineering using the CRISPR-Cas9 system. Nat Protoc 8, 2281–2308. 10.1038/nprot.2013.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Allaire JJ, and Chollet F (2019). keras: R Interface to Keras. https://github.com/rstudio/keras.
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The datasets and codes generated during this study are available in the Supplementary Information and publicly available from GitHub (https://github.com/rikserrano/DeepLearning_TdP_Risk).