Both biomedical research and clinical practice rely on complex datasets for the physiological and genetic characterization of human hearts in health and disease. Given the complexity and variety of approaches and recordings, there is now growing recognition of the need to embed computational methods in cardiovascular medicine and science for analysis, integration and prediction. This paper describes a Workshop on Computational Cardiovascular Science that created an international, interdisciplinary and inter-sectorial forum to define the next steps for a human-based approach to disease supported by computational methodologies. The main ideas highlighted were (i) a shift towards human-based methodologies, spurred by advances in new in silico, in vivo, in vitro, and ex vivo techniques and the increasing acknowledgement of the limitations of animal models. (ii) Computational approaches complement, expand, bridge, and integrate in vitro, in vivo, and ex vivo experimental and clinical data and methods, and as such they are an integral part of human-based methodologies in pharmacology and medicine. (iii) The effective implementation of multi- and interdisciplinary approaches, teams, and training combining and integrating computational methods with experimental and clinical approaches across academia, industry, and healthcare settings is a priority. (iv) The human-based cross-disciplinary approach requires experts in specific methodologies and domains, who also have the capacity to communicate and collaborate across disciplines and cross-sector environments. (v) This new translational domain for human-based cardiology and pharmacology requires new partnerships supported financially and institutionally across sectors. Institutional, organizational, and social barriers must be identified, understood and overcome in each specific setting.

Motivation

Both biomedical research and clinical practice rely on complex datasets for the physiological and genetic characterization of human hearts in health and disease. The information shaping our knowledge of human hearts is obtained from a variety of techniques and models, including recordings obtained in vivo invasively and non-invasively, in ex vivo tissue and isolated human adult cardiomyocytes recordings, and more recently in vitro using human stem-cell-derived cardiomyocytes. Increasing evidence suggests that non-human animal models may have limited ability to predict human in vivo effects due to important species differences between humans, dogs, guinea pigs, and rabbits.13 Thus, methods firmly rooted in understanding physiology and pathophysiology in humans are clearly needed.

Advances in imaging technologies such as the multiple modalities of cardiac magnetic resonance (CMR) are combined with body surface or intra-cardiac electrophysiological recordings to evaluate in specific patients the in vivo structural and functional implications of cardiac disease. Recent progress in research using human cardiomyocytes derived from induced pluripotent stem cells promises exciting new developments as it allows the in vitro characterization of the phenotype of cardiomyocytes of specific patients, and therefore has the potential of introducing the flexibility of in vitro methodologies in personalized medicine. Furthermore, ex vivo tissue from biopsies or from donor human hearts provides measurements of tissue, cellular, and ionic properties of human adult cardiomyocytes in non-diseased and diseased conditions.

Each of these types of human-based assays and measurements provides us a single snapshot from one perspective of a complex set of variables through both time and spatial dimensions. In turn, this complex set of variables is able to define and explain the myriad of dynamic mechanisms and properties that underlie the activity of human hearts in health and disease. Given the complexity and variety of approaches and recordings, there is now growing recognition of the need to embed computational methods in cardiovascular medicine and science for analysis, integration, and prediction. Computational approaches in biomedicine range over a variety of techniques for signal, data, and image analysis, but also importantly multiscale modelling and simulation. Together, they provide a synergistic approach to organize and augment the information obtained from experimental and clinical recordings. The benefits gained include the quantitative analysis and organized reassembly of multiscale and multimodality datasets to probe, challenge and expand our knowledge of the complex and dynamic interactions in cardiac electrophysiology. Advances in computational cardiac electrophysiology were recently illustrated in two dedicated special issues of Europace 2014.4,5 Furthermore, new initiatives such as the Comprehensive in vitro Proarrhythmia Assay (CiPA) launched by the United States Food and Drug Administration (FDA) recognize the potential of human-based in silico and in vitro approaches as a new paradigm for drug safety assessment.6

On 17 September 2014, a Workshop on Computational Cardiovascular Science was hosted at the University of Oxford with the aim of creating an international, interdisciplinary, and inter-sectorial forum to discuss current trends in computational technologies to augment cardiovascular physiology, pharmacology, and medicine, and to propose solutions for the replacement, refinement and reduction of animal experimentation.7 Invited participants included experts in cardiology, computer science, physiology, pharmacology, philosophy, and biomedical engineering from academia and industry, from 11 Universities and 12 companies, from UK, several countries in Europe, USA, and Japan. In this paper, we aim at describing the main ideas discussed during the meeting, rather than providing a thorough review of the literature. Table 1 summarizes the forms of human-based in vivo, ex vivo, in vitro, and in silico experiments and techniques discussed, which are further illustrated in Figure 1.
Table 1

Summary of human-based in vivo, ex vivo, and in vitro techniques and in silico approaches

Human-basedData acquisition techniqueLimitations of the dataProgress of in silico approaches
In vivoElectrophysiological recordings during clinical procedures
[Taggart, Zhou]
  • – Limited data sets due to ethical and practical obstacles;

  • – Datasets derived from diseased hearts;

  • – Inter-patient variability;

  • – Limited experimentation

  • – Signal analysis and integration;

  • – Multiscale electrophysiological models and simulations;

  • – Population of models to mimic action potential variability

Multimodality imaging including magnetic resonance
[Ariga, Grau]
  • – Limited data sets;

  • – No experimentation

  • – Image analysis (tissue characterization);

  • – Ventricular shape analysis;

  • – Structural models and simulations

  • – Computer models for link between structure and diffusion;

  • – And link between micro structure and function

Body surface potentials, electrocardiogram
[Minchole, Lu]
  • – Captures global patterns of heart behaviour;

  • – Variability

  • – Automated quantification of ECG features for clinical diagnosis and identification of new bioamarkers (morphological QRS or T-wave based, iCEB);

  • – Electrocardiographic imaging;

  • – Multiscale human heart simulations from ion channel and microstructure to the electrocardiogram

mHealth recordings through mobile devices
[Oster]
  • – Very large quantities of data not amenable to manual analysis;

  • – Noisy and patchy data;

  • – Social, ethical and legal challenges

  • – Automated and semi-automated techniques for analysis, such as machine learning, implementable on mobile phones

Isolated human primary cells and non-clinical, real-world data from biopsies and medical histories
[Ghetti]
  • – Limited data sets;

  • – Social, ethical and legal challenges

  • – Computational models to integrate experimental data and to investigate multiscale mechanisms of disease and pharmacological interventions

Ex vivoMicroelectrode, optical mapping, patch clamp, protein, and mRNA expression
[Varro, Britton, Dutta]
  • – Limited availability, and mostly from diseased hearts;

  • – Variability;

  • – Difficulty of technique implementation (cell isolation; current separation);

  • – Change of properties due to cell isolation

  • – Data analysis and integration;

  • – Multiscale models for greater contextualization;

  • – Investigation of variability through approaches such as population of models

In vitroHuman cardiomyocytes derived from induced pluripotent stem cells (hiPSC-CMs)
[Daniels, Severi, Kopljar, Harmer]
  • – Inconsistent immaturity;

  • – Variability and associated difficulty of comparison

  • – Models to investigate variability, assist interpretation, and facilitate translation to in vivo cells

  • – Models to investigate gene mutations;

  • – Models for drug safety assessment

Cell cultures and high-speed optical imaging
[Burton]
  • – Limited cross-institution and cross-sector access to experiments

  • – Multiscale modelling to explain dynamics in heterogeneous preparations

Human-basedData acquisition techniqueLimitations of the dataProgress of in silico approaches
In vivoElectrophysiological recordings during clinical procedures
[Taggart, Zhou]
  • – Limited data sets due to ethical and practical obstacles;

  • – Datasets derived from diseased hearts;

  • – Inter-patient variability;

  • – Limited experimentation

  • – Signal analysis and integration;

  • – Multiscale electrophysiological models and simulations;

  • – Population of models to mimic action potential variability

Multimodality imaging including magnetic resonance
[Ariga, Grau]
  • – Limited data sets;

  • – No experimentation

  • – Image analysis (tissue characterization);

  • – Ventricular shape analysis;

  • – Structural models and simulations

  • – Computer models for link between structure and diffusion;

  • – And link between micro structure and function

Body surface potentials, electrocardiogram
[Minchole, Lu]
  • – Captures global patterns of heart behaviour;

  • – Variability

  • – Automated quantification of ECG features for clinical diagnosis and identification of new bioamarkers (morphological QRS or T-wave based, iCEB);

  • – Electrocardiographic imaging;

  • – Multiscale human heart simulations from ion channel and microstructure to the electrocardiogram

mHealth recordings through mobile devices
[Oster]
  • – Very large quantities of data not amenable to manual analysis;

  • – Noisy and patchy data;

  • – Social, ethical and legal challenges

  • – Automated and semi-automated techniques for analysis, such as machine learning, implementable on mobile phones

Isolated human primary cells and non-clinical, real-world data from biopsies and medical histories
[Ghetti]
  • – Limited data sets;

  • – Social, ethical and legal challenges

  • – Computational models to integrate experimental data and to investigate multiscale mechanisms of disease and pharmacological interventions

Ex vivoMicroelectrode, optical mapping, patch clamp, protein, and mRNA expression
[Varro, Britton, Dutta]
  • – Limited availability, and mostly from diseased hearts;

  • – Variability;

  • – Difficulty of technique implementation (cell isolation; current separation);

  • – Change of properties due to cell isolation

  • – Data analysis and integration;

  • – Multiscale models for greater contextualization;

  • – Investigation of variability through approaches such as population of models

In vitroHuman cardiomyocytes derived from induced pluripotent stem cells (hiPSC-CMs)
[Daniels, Severi, Kopljar, Harmer]
  • – Inconsistent immaturity;

  • – Variability and associated difficulty of comparison

  • – Models to investigate variability, assist interpretation, and facilitate translation to in vivo cells

  • – Models to investigate gene mutations;

  • – Models for drug safety assessment

Cell cultures and high-speed optical imaging
[Burton]
  • – Limited cross-institution and cross-sector access to experiments

  • – Multiscale modelling to explain dynamics in heterogeneous preparations

Table 1

Summary of human-based in vivo, ex vivo, and in vitro techniques and in silico approaches

Human-basedData acquisition techniqueLimitations of the dataProgress of in silico approaches
In vivoElectrophysiological recordings during clinical procedures
[Taggart, Zhou]
  • – Limited data sets due to ethical and practical obstacles;

  • – Datasets derived from diseased hearts;

  • – Inter-patient variability;

  • – Limited experimentation

  • – Signal analysis and integration;

  • – Multiscale electrophysiological models and simulations;

  • – Population of models to mimic action potential variability

Multimodality imaging including magnetic resonance
[Ariga, Grau]
  • – Limited data sets;

  • – No experimentation

  • – Image analysis (tissue characterization);

  • – Ventricular shape analysis;

  • – Structural models and simulations

  • – Computer models for link between structure and diffusion;

  • – And link between micro structure and function

Body surface potentials, electrocardiogram
[Minchole, Lu]
  • – Captures global patterns of heart behaviour;

  • – Variability

  • – Automated quantification of ECG features for clinical diagnosis and identification of new bioamarkers (morphological QRS or T-wave based, iCEB);

  • – Electrocardiographic imaging;

  • – Multiscale human heart simulations from ion channel and microstructure to the electrocardiogram

mHealth recordings through mobile devices
[Oster]
  • – Very large quantities of data not amenable to manual analysis;

  • – Noisy and patchy data;

  • – Social, ethical and legal challenges

  • – Automated and semi-automated techniques for analysis, such as machine learning, implementable on mobile phones

Isolated human primary cells and non-clinical, real-world data from biopsies and medical histories
[Ghetti]
  • – Limited data sets;

  • – Social, ethical and legal challenges

  • – Computational models to integrate experimental data and to investigate multiscale mechanisms of disease and pharmacological interventions

Ex vivoMicroelectrode, optical mapping, patch clamp, protein, and mRNA expression
[Varro, Britton, Dutta]
  • – Limited availability, and mostly from diseased hearts;

  • – Variability;

  • – Difficulty of technique implementation (cell isolation; current separation);

  • – Change of properties due to cell isolation

  • – Data analysis and integration;

  • – Multiscale models for greater contextualization;

  • – Investigation of variability through approaches such as population of models

In vitroHuman cardiomyocytes derived from induced pluripotent stem cells (hiPSC-CMs)
[Daniels, Severi, Kopljar, Harmer]
  • – Inconsistent immaturity;

  • – Variability and associated difficulty of comparison

  • – Models to investigate variability, assist interpretation, and facilitate translation to in vivo cells

  • – Models to investigate gene mutations;

  • – Models for drug safety assessment

Cell cultures and high-speed optical imaging
[Burton]
  • – Limited cross-institution and cross-sector access to experiments

  • – Multiscale modelling to explain dynamics in heterogeneous preparations

Human-basedData acquisition techniqueLimitations of the dataProgress of in silico approaches
In vivoElectrophysiological recordings during clinical procedures
[Taggart, Zhou]
  • – Limited data sets due to ethical and practical obstacles;

  • – Datasets derived from diseased hearts;

  • – Inter-patient variability;

  • – Limited experimentation

  • – Signal analysis and integration;

  • – Multiscale electrophysiological models and simulations;

  • – Population of models to mimic action potential variability

Multimodality imaging including magnetic resonance
[Ariga, Grau]
  • – Limited data sets;

  • – No experimentation

  • – Image analysis (tissue characterization);

  • – Ventricular shape analysis;

  • – Structural models and simulations

  • – Computer models for link between structure and diffusion;

  • – And link between micro structure and function

Body surface potentials, electrocardiogram
[Minchole, Lu]
  • – Captures global patterns of heart behaviour;

  • – Variability

  • – Automated quantification of ECG features for clinical diagnosis and identification of new bioamarkers (morphological QRS or T-wave based, iCEB);

  • – Electrocardiographic imaging;

  • – Multiscale human heart simulations from ion channel and microstructure to the electrocardiogram

mHealth recordings through mobile devices
[Oster]
  • – Very large quantities of data not amenable to manual analysis;

  • – Noisy and patchy data;

  • – Social, ethical and legal challenges

  • – Automated and semi-automated techniques for analysis, such as machine learning, implementable on mobile phones

Isolated human primary cells and non-clinical, real-world data from biopsies and medical histories
[Ghetti]
  • – Limited data sets;

  • – Social, ethical and legal challenges

  • – Computational models to integrate experimental data and to investigate multiscale mechanisms of disease and pharmacological interventions

Ex vivoMicroelectrode, optical mapping, patch clamp, protein, and mRNA expression
[Varro, Britton, Dutta]
  • – Limited availability, and mostly from diseased hearts;

  • – Variability;

  • – Difficulty of technique implementation (cell isolation; current separation);

  • – Change of properties due to cell isolation

  • – Data analysis and integration;

  • – Multiscale models for greater contextualization;

  • – Investigation of variability through approaches such as population of models

In vitroHuman cardiomyocytes derived from induced pluripotent stem cells (hiPSC-CMs)
[Daniels, Severi, Kopljar, Harmer]
  • – Inconsistent immaturity;

  • – Variability and associated difficulty of comparison

  • – Models to investigate variability, assist interpretation, and facilitate translation to in vivo cells

  • – Models to investigate gene mutations;

  • – Models for drug safety assessment

Cell cultures and high-speed optical imaging
[Burton]
  • – Limited cross-institution and cross-sector access to experiments

  • – Multiscale modelling to explain dynamics in heterogeneous preparations

Sources of experimental data integrated in computational models of human cardiac electrophysiology, and applications in physiology. Ionic current models are constructed mostly based on voltage/patch clamp data from ex vivo and in vitro preparations. The integration of ionic current models in single cell models, accounting for variability in protein expression and disease remodelling, allows for the simulation of the action potential and electrolyte concentrations in healthy and disease. Additionally, cardiac simulations at the whole organ and body levels require the construction of image-based anatomical models. When coupled to mathematical descriptions of electrical excitation through cardiac tissue, they allow for the high-resolution investigation of arrhythmia mechanisms based on clinical electrophysiology studies, as for the interpretation and identification of arrhythmic-risk biomarkers at the surface potential level. Transmural visualization of ventricular myofibre orientation, adapted from reference 8 with permission. ECG/whole body simulation, adapted from reference 9 with permission.
Figure 1

Sources of experimental data integrated in computational models of human cardiac electrophysiology, and applications in physiology. Ionic current models are constructed mostly based on voltage/patch clamp data from ex vivo and in vitro preparations. The integration of ionic current models in single cell models, accounting for variability in protein expression and disease remodelling, allows for the simulation of the action potential and electrolyte concentrations in healthy and disease. Additionally, cardiac simulations at the whole organ and body levels require the construction of image-based anatomical models. When coupled to mathematical descriptions of electrical excitation through cardiac tissue, they allow for the high-resolution investigation of arrhythmia mechanisms based on clinical electrophysiology studies, as for the interpretation and identification of arrhythmic-risk biomarkers at the surface potential level. Transmural visualization of ventricular myofibre orientation, adapted from reference 8 with permission. ECG/whole body simulation, adapted from reference 9 with permission.

The main ideas highlighted through the workshop were the following:

  • A shift towards human-based methodologies in pharmacology and medicine is occurring, spurred by advances in new in silico, in vivo, in vitro, and ex vivo techniques and the increasing acknowledgement of the limitations of animal models.

  • Computational approaches complement, expand, bridge, and integrate in vitro, in vivo, and ex vivo experimental and clinical data and methods, and as such they are an integral part of human-based methodologies in pharmacology and medicine.

  • The effective implementation of multi- and interdisciplinary approaches, teams, and training combining and integrating computational methods with experimental and clinical approaches across academia, industry, and healthcare settings is a priority.

  • The human-based cross-disciplinary approach requires experts in specific methodologies and domains, who also have the capacity to communicate and collaborate across disciplines and to work productively in interdisciplinary and cross-sector environments.

  • This new translational domain for human-based cardiology and pharmacology requires new partnerships supported financially and institutionally across pharma and biotech industry, contract research organizations (CRO), academia and research institutes, technology and service providers, non-profit and governmental organizations, and regulatory agencies. Institutional, organizational, and social barriers must be identified, understood, and overcome in each specific setting.

Description of the workshop

The workshop consisted of four sessions aiming at exploring different aspects of human-based cardiovascular science, and specifically the synergies with in silico approaches in the three main experimental settings, i.e. in vivo, in vitro, and ex vivo. Table 1 summarizes the forms of experiments and techniques discussed.

Human in vivo cardiovascular science

Prof. Peter Taggart examined the challenges involved in obtaining basic electrophysiological data from in vivo human subjects, during routine clinical procedures. The example discussed was the application of percutaneous transluminal coronary angioplasty (PTCA) to study in vivo the effect of myocardial ischaemia (a major cause of mortality) on human electrophysiology, by recording either monophasic action potentials or unipolar electrograms on the ventricular endocardium in the region served by the artery undergoing PTCA. In vivo electrophysiological recording techniques include the use of the multi-electrode sock (264 electrodes) over the human ventricles to study global patterns of activation such as ventricular fibrillation, action potential duration (APD) changes, and post-repolarization refractoriness during early ischaemia and the demonstration of mechano-electric feedback in humans.1020 Findings addressed the debate as to whether rotors or multiple wavelets drive ventricular fibrillation (VF) and showed that both coexist in human VF, and the very rapid time course of the early electrophysiological changes during early ischaemia in humans. In vivo electrophysiological recordings are critical for translational research (from basic to clinical) but they are limited due to considerations for patient comfort and safety. Therefore, they need to be complemented by and combined with alternative ways of probing the human heart, including non-invasive in vivo imaging as well as ex vivo investigations, as described below.

Dr Rina Ariga provided an overview of multimodality magnetic resonance imaging and its application in patients with hypertrophic cardiomyopathy (HCM). HCM is the most common genetic heart disease (affects 1 in 500)21 and the commonest cause of sudden cardiac death in the young.22 Transthoracic echocardiography (TTE) is routinely used to assess HCM, but is limited in patients with poor acoustic windows or poor visualization of some LV regions. CMR is now the gold standard in assessing mass, hypertrophy, volume, and function in HCM due to high spatial and temporal resolution.23 Unlike TTE, CMR also provides tissue characterization using late gadolinium enhancement (detects focal fibrosis which has been associated with ventricular arrhythmias and SCD)24 and T1 mapping (detects diffuse and focal fibrosis). CMR also offers insights into several hallmark features of HCM that are potential contributors of disease progression using novel techniques such as stress perfusion imaging (impaired perfusion),25 blood oxygen level dependent (BOLD) imaging at stress (deoxygenation at stress),26 phosphorus MR spectroscopy (abnormal myocardial energetics at rest with further deficit in exercise),27 and most recently, diffusion tensor imaging (to assess fibre disarray).28 CMR is not only a useful imaging adjunct in cases with limited TTE views, but also provides accurate disease characterization of subtle differences. Research using CMR is improving our understanding of this complex heterogeneous disease and is helping to guide risk stratification and treatment strategies.29

Prof. Vicente Grau spoke about methods to investigate myocardial microstructure, including quantification using imaging as well as determination of functional repercussions of microstructural changes, investigated using computational modelling and simulation. Recent developments in MRI technology, in particular using diffusion sensitive sequences, allow the quantification of microstructure, initially in fixed hearts8,30,31 or in hearts at different stages of contraction.32 Diffusion MRI uses an indirect measurement to estimate cardiac structure, and its relationship to cardiac microstructure is not fully understood. Two methods to improve this understanding were discussed. Histology offers direct insights into microstructure, but three-dimensional reconstruction from histological slices is challenging.33 Computational models, simulating water diffusion and MRI acquisition sequences, can be used to provide a direct link between structure and diffusion.34 The relationship between microstructural and functional changes is not fully understood, and here computational models can again provide a unique tool as shown for example in references.3537

In addition to imaging, clinical in vivo recordings to evaluate the human heart include the surface electrocardiograms (ECGs), a rapid and cost-effective method to acquire non-invasive recordings. A large body of research has been devoted to the quantification of ECG features for patient stratification in terms of arrhythmic risk and disease.38 Dr Ana Mincholé described computational approaches applied to the ECG for the detection of electrophysiological abnormalities and patient stratification in HCM. Using a database of Holter recordings obtained in HCM patients and volunteers, she described the quantification of standard ECG-based biomarkers such as QT T peak to T end, as well as new mathematical model-based morphological features of the QRS complex and T wave morphology.39,40 Dr Minchole also described how information about the electrophysiological and structural signature of disease from the ionic to the whole organ level can be integrated in multiscale human heart models and used in the development and understanding of ECG-based biomarkers.41 Multiscale simulations as illustrated in Figure 2 allow the identification of key structural and functional factors that determine each of the ECG biomarkers and provide a deeper and more precise understanding of the information each of the biomarkers conveys.42 The new knowledge also aids in the identification of more selective and specific biomarkers for specific disease conditions with complex functional and structural signatures such as HCM or myocardial infarction. Body surface ECGs combined with imaging data using mathematical algorithms to non-invasively reconstruct the electrical activity on the epicardial surface of the heart in vivo as demonstrated for example in references.4345
Computer simulation of the human heart electrophysiology from ion channel to body surface potentials and the electrocardiogram. Simulations are conducted using human biophysically detailed models considering heterogeneity in specific ionic properties (left, colour scale correspond to the maximum conductance of the slow component of the delayed rectifying current) to determine their effect on the spatiotemporal evolution of electrical potentials across the whole torso (middle, extracellular potentials throughout the torso) and on the ECG (right, main leads displayed).
Figure 2

Computer simulation of the human heart electrophysiology from ion channel to body surface potentials and the electrocardiogram. Simulations are conducted using human biophysically detailed models considering heterogeneity in specific ionic properties (left, colour scale correspond to the maximum conductance of the slow component of the delayed rectifying current) to determine their effect on the spatiotemporal evolution of electrical potentials across the whole torso (middle, extracellular potentials throughout the torso) and on the ECG (right, main leads displayed).

Human ex vivo and in vitro cardiovascular science

Prof. Andras Varro began by discussing the knowledge we have of human electrophysiology largely through classic techniques such as conventional microelectrode and more modern patch clamp techniques, protein and RNA expression approaches using human cardiomyocytes from biopsies or donor hearts.4648 Obtaining good representative experimental human data is restricted by a number of practical problems. Sources of tissue particularly ventricular cells are difficult to obtain and isolation of cells is complicated by disease. The source of undiseased donor hearts is particularly limited. The process of cell isolation may also lead to changes from properties studied in multicellular tissue preparations. Furthermore, the separation of each of the ionic currents illustrated in Figure 3 is problematic since the available pharmacological inhibitors are not totally selective. Finally, Prof. Varro highlighted the importance of human-based studies rather than highlighted the use of animal models. It is often underappreciated that there are substantial species differences between human and even large mammals, such as the dog, considered to be a representative model.49,50 For example, marked differences in ventricular repolarization reserve have been reported between human and dog,49 with larger rapid component of the delayed rectifying current (IKr) in human but stronger slow component (IKs) and inward rectifying current (IK1) in the dog. Consequently, APD prolongation caused by selective IKr block is three-fold larger in human than in dog, which suggests caution in translation of animal findings to human. Species-specific differences in pharmacological action between animal models have been also described,50 with rabbit exhibiting larger APD prolongation and proneness to repolarization abnormalities upon selective IKr block compared to others. Computational modelling and simulation can facilitate interspecies comparison by identifying and addressing differences and maximizing the re-use of experimental data from human hearts.
Simulation of the human ventricular action potential and the underlying ionic currents. From top to bottom, time course of the action potential, sodium current (INa), L-type calcium current, the rapid and slow component of the delayed rectifying current (IKr, IKs) and the inward rectifying current (IK1), the transient outward current (Ito) and the sodium potassium pump (INaK) and the sodium calcium exchanger (INaCa).
Figure 3

Simulation of the human ventricular action potential and the underlying ionic currents. From top to bottom, time course of the action potential, sodium current (INa), L-type calcium current, the rapid and slow component of the delayed rectifying current (IKr, IKs) and the inward rectifying current (IK1), the transient outward current (Ito) and the sodium potassium pump (INaK) and the sodium calcium exchanger (INaCa).

Dr Oliver Britton described how the complex electrophysiological datasets obtained from ex vivo human hearts have been integrated in multiscale computer models, specifically focusing on human data over the past decade5154 as examples. He described the recent construction of a population of human ventricular cell computational models that captures the inter-subject variability seen in action potential recordings from human ventricular tissue preparations from Prof Varro's laboratory. The models in the population have a wide range of different configurations of ionic current strengths to mimic variability in a population as in reference 55. The team investigated whether there were configurations that were particularly vulnerable to developing repolarization abnormalities such as alternans and early afterdepolarizations (EADs), in response to blockade of different combinations of currents known to be important in repolarization—rapid delayed rectifying (IKr), slow delayed rectifying (IKs), inward rectifying (IK1), and late calcium (ICaL) current. The computational approach therefore integrates and extends experimental recordings, generating predictions and refining hypotheses about proarrhythmic mechanisms that can then be tested experimentally. Importantly, the in silico human models provide a multiscale framework to investigate with high spatiotemporal resolution key ionic mechanisms in drug safety and efficacy in human, with high degree of flexibility in the possible interventions (such as heart rate, concentrations, and adrenergic challenge) compared to experiments.

Prof. Matt Daniels reviewed the status of human cardiomyocytes derived from induced pluripotent stem cells (hiPSC-CMs). He saw three uses for these cells:
  • screening for cardiotoxicity of novel therapeutics,

  • modelling and drug discovery for inherited cardiac conditions and

  • in the medium to long term, regenerative cell therapy.

While there is progress in scaling up production of these cells,56 functional maturity is still a major impediment. Stem cell derivatives in many ways (force generation, gene expression, sarcomeric maturity, electrophysiological properties, etc.) resemble immature neonatal cardiac substrates.57 However, there is one key difference that is rather unique to these cells and best illustrated by comparison to the existing alternatives. Adult cardiomyocytes are consistently mature; neonatal cardiomyocytes are consistently immature. By contrast, stem cell derivatives display an inconsistent immaturity (Figure 4) such that in any experimental test to date, variability within the sample is typically one order of magnitude of measures such as APD and cycle length as shown in reference 58. This will complicate comparisons between samples, which are typically made on small numbers of cells.
Stem-cell-derived cardiomyocytes have variable phenotypes: current methods of stem cell differentiation produce mixed populations at two distinct levels—gene expression, and post-transcription. This is demonstrated for sarcomeric morphology here, with the panel on the left showing two cells in the field of view positive for the z-disc marker alpha-actinin (white), and the thin filament protein troponin I (magenta). However, further heterogeneity exists even within the cell populations expressing both markers, as only some cells demonstrate ordered sarcomeric units with clear cell polarity (panel on the right). Methods to eliminate (or compensate for) this will be needed to enable small differences between samples to be identified above the noise of the difference within the sample. Scale bar 10 µm. The human ES line OXF2 was grown to confluency on Matrigel and differentiated as described in reference 59. Cells were dissociated by incubation with trypsin/EDTA (0.05%, Lifetech) for 15 min at room temperature prior to seeding onto 0.1% gelatin coated glass coverslips. Ten days after seeding, cells were fixed in 4% PFA (10 min, room temperature), permeabilized (0.1% Triton X-100 in Tris-buffered saline), and blocked with 2% BSA plus 0.001% sodium azide in TBS-T (1 h RT) and incubated with Primary antibodies (mouse monoclonal anti alpha-actinin, (sigma), and rabbit polyclonal anti-troponin T, prior to washing and incubation with Fab fragment anti-mouse 488, and anti-rabbit 568 (molecular probes). Images were acquired on an upright Leica SP5 confocal with a 63× lens.
Figure 4

Stem-cell-derived cardiomyocytes have variable phenotypes: current methods of stem cell differentiation produce mixed populations at two distinct levels—gene expression, and post-transcription. This is demonstrated for sarcomeric morphology here, with the panel on the left showing two cells in the field of view positive for the z-disc marker alpha-actinin (white), and the thin filament protein troponin I (magenta). However, further heterogeneity exists even within the cell populations expressing both markers, as only some cells demonstrate ordered sarcomeric units with clear cell polarity (panel on the right). Methods to eliminate (or compensate for) this will be needed to enable small differences between samples to be identified above the noise of the difference within the sample. Scale bar 10 µm. The human ES line OXF2 was grown to confluency on Matrigel and differentiated as described in reference 59. Cells were dissociated by incubation with trypsin/EDTA (0.05%, Lifetech) for 15 min at room temperature prior to seeding onto 0.1% gelatin coated glass coverslips. Ten days after seeding, cells were fixed in 4% PFA (10 min, room temperature), permeabilized (0.1% Triton X-100 in Tris-buffered saline), and blocked with 2% BSA plus 0.001% sodium azide in TBS-T (1 h RT) and incubated with Primary antibodies (mouse monoclonal anti alpha-actinin, (sigma), and rabbit polyclonal anti-troponin T, prior to washing and incubation with Fab fragment anti-mouse 488, and anti-rabbit 568 (molecular probes). Images were acquired on an upright Leica SP5 confocal with a 63× lens.

Dr Stefano Severi showed how a computational approach can be supportive and complementary to the functional in vitro study of hiPSC-CMs. In exploiting hiPSC-CMs as in vitro models for the electrophysiological effects of evolving or new drugs, a detailed understanding of the electrophysiological properties of hiPSC-CMs is necessary. In silico models of hiPSC-CMs action potential have been constructed for control60 and some genetic mutations, such as those causing long QT (LQT) syndrome type 1 (LQT1),61 type 2, and type 3,62 based on recent electrophysiological measurements6366 and validated against drug administration. Computer simulations showed that in principle hiPSC-CMs are qualitatively consistent with adult CMs in response to many current blockers,67 but differences also emerged. Moreover, hiPSC-CMs show a highly variable electrophysiological behaviour, namely a variable and depolarized resting potential and diverse rates of spontaneous action potentials. Such high variability can be perceived as a limiting factor to the application of the computational approach to hiPSC-CMs. Indeed, computational models of cell electrophysiology are usually developed on the basis of the average values of quantities measured in in vitro experiments. The underlying idea is that the predictions obtained with the model of the ‘average cell’ can apply to all the cells with a tolerance that is of the same order of the variability of the data on which the model is based. Therefore, high variability in the data results in predictions with lower reliability. One way to overcome this limitation is to include the variability within the model itself (which is no longer a model of the ‘average cell’) in order to take it into account both in the investigation of physiological mechanisms and in model-based predictions. The aforementioned population of models approach is a relevant and promising example. Furthermore, when experimental data show high variability, computational models can help to identify the causes of such heterogeneity and potentially help to reduce it. As a relevant example, computational analysis can be used to assess to what extent the variability is due to (i) different experimental systems (e.g. cell lines) that could then be described by different, specific, computational models (ii) differences in the expression of ionic channels from cell to cell, which is eventually much larger than in adult cardiomyocytes or (iii) lack of robustness in the electrical activity of incompletely mature hiPSC-CMs. An example of the latter case are the effects of small changes in depolarizing currents, which can lead to dramatic changes in the rate of spontaneous beating in hiPSC-CMs, whereas they lead to only minor changes in resting potential in adult cardiomyocytes. In this sense, the variability can be reduced if observed in the model parameter's space since cells showing very different APs could be quite close in terms of their ionic current maximal conductances.

Dr Ivan Kopljar continued the theme looking at experimental studies on hiPS-CMs in safety assessments in the drug development setting, with an emphasis on the investigation of long-term drug effects. Currently, various technologies such as multi-electrode array (MEA), Ca2+ transient, impedance, and optical action potential measurements are applied on hiPSC-CMs for their characterization. Their potential has been highlighted by the CiPA initiative launched by the FDA. On the other hand, drug-induced delayed and chronic cardiotoxicity is one of the main risk for drug withdrawal from the market. Therefore, Dr Kopljar described investigations of the acute and delayed (5 days) effects of various oncological compounds on hiPS-CMs using an impedance-based functional assay. Different functional parameters such as beat rate, cell index and incidence of arrhythmia-like events were evaluated, and indicate that the hiPS-CMs can be used to detect different levels of acute and chronic cardiotoxicity and could be valuable in drug safety.

In another industrial perspective, Dr Najah Abi-Gerges discussed currently employed strategies in the pharmaceutical industry for cardiotoxicity screening. He gave an overview of the issue pointing out the high attrition rate of new molecules during the iterative drug discovery process: a significant number of these are caused by cardiotoxicity.68 He highlighted the extent to which some safe new drugs are not developed because of these potential concerns. The new CiPA initiative attempts to address this issue.6 CiPA proposes a paradigm based on screening new molecules against specific cardiac ion channels combined with integrative computer modelling to predict the proarrhythmic potential of new drugs. This is subsequently combined with non-rodent and early human clinical studies to assess drug effects on QTc measurements. Dr Abi-Gerges highlighted the need for further developing robust and predictive in silico models that represent native myocyte physiology and the heart of healthy volunteers and patients. Such models will predict acute and chronic drug effects with high predictive value on ECG abnormalities other than QT/proarrhythmia, heart rate, contractility, blood pressure, and cardiac structure. Dr Abi-Gerges concluded by advocating that scientists, modellers, regulators, and pharmaceutical industry are currently well positioned to shape how future cardiac modelling would positively impact drug development.

Arrhythmia mechanisms and biomarkers

A fourth session presented the use of experimental and computational methods for investigations into arrhythmia mechanisms and biomarkers. Dr Stephen Harmer continued the theme of iPSC technologies, in this case for investigations on mechanisms of disease pathogenesis in LQT1 using iPSC technology to model the hereditary cardiac arrhythmia syndrome. Both heterologous (HEK293/CHO-K1 cell expression systems) and iPSC-based cellular models are used to investigate the underlying disease mechanisms, and a comparison is conducted to evaluate whether the disease mechanisms are similar in both cell types. Results show differences on IKs channel function and trafficking in heterologous systems for five LQT1 patient mutations. The results are being modelled computationally to investigate the implications of IKs channel mutations on action potential duration and morphology, and the penetrance of the mutation in human cell populations such as those described in the previous session.

Dr Sara Dutta presented a state-of-the-art multiscale human whole heart computational framework to investigate arrhythmic effects of drugs and disease, and specifically hERG block in acute myocardial ischaemia. The framework presented here consists of a human anatomically based model with biophysically detailed representation of membrane kinetics including ionic current and concentration dynamics, as well as fibre orientation and ventricular heterogeneity. The human multiscale model is parameterized using experimental data, from voltage clamp at the ionic level to MRI scans at the whole heart level. They provide a multiscale platform to dissect and analyse specific ischaemic and arrhythmic processes with high spatiotemporal resolution, which is not possible to obtain through experiments alone, especially in human. The new insights provided by the human model could help in the design of further experimental and clinical studies to improve patient risk stratification, as well as decisions about drug dose during management of anti-arrhythmic therapy. This study69 can be extended to explore the mechanisms under different conditions, such as varying sizes and locations of the ischemic region, different drug compounds and their multichannel effects, and inter-subject variability in ionic currents and repolarization patterns. The presentation therefore highlighted the power of human multiscale simulations using anatomically based heart models to investigate safety and efficacy of pharmacological action in lethal disease conditions.

Using a different computational approach, Dr Julien Oster presented advances on the computational detection of arrhythmia episodes in ECGs. The development of mobile technologies (mobile phones, tablets, etc.) for health services (mHealth) is currently rapidly growing for two main reasons: (i) cost reduction and (ii) access to resource-scarce communities.70 The importance for the automatic or semi-automatic detection of arrhythmias on ECG recordings was highlighted in the presentation, given the simplicity of data acquisitions and therefore the multiplication of such data. Manual expert analysis would be more a burden for the clinicians than help for the diagnosis of cardiovascular diseases. Machine-learning approaches have been demonstrated to be a powerful tool for such an analysis for several applications, such as atrial fibrillation (AF) episodes or ventricular ectopic beat detection. Many Holter softwares already require electrophysiologists or laboratory technicians to annotate beats clustered together automatically. Llamedo and Martinez71 recently suggested a technique requesting expert labelling of the clusters, outputted based on both morphological- and rhythm-based features. Other techniques for rhythm classification or AF episodes detection, based uniquely on the heart rhythm, have also been presented, and are implemented in implantable devices.72

Dr Oster's presentation focused on two major arrhythmic types: AF and premature ventricular contraction (PVC). A machine-learning approach has recently been proposed, through the implementation of a Support Vector Machine73 on a mobile phone.74 PVC is another type of ventricular arrhythmia, identified as a predictor for mortality after myocardial ischaemia.75 The application of an ECG morphology model-based Bayesian filtering76,77 was shown to be effective for PVC detection.78

Ms Xin Zhou presented computational investigations into the ionic mechanisms underlying repolarization alternans in a population of human ventricular models calibrated using in vivo recordings such as those presented by Prof. Taggart. Previous research into cardiac alternans has mainly been carried out in animals rather than in human. Ms Zhou presented the construction and calibration of a population of over 2000 human ventricular cell electrophysiology models to mimic the action potential variability exhibited in in vivo electrophysiological recordings from 41 patients.79 The in silico human ventricular cell population is illustrated in Figure 5 and it was shown to reproduce two types of alternans restitution curves also observed in human in vivo recordings. By analysing the population of human in silico models, she dissected the mechanisms underlying cardiac alternans and how the complex interaction between sarcolemmal currents and calcium dynamics contribute to the initiation and maintenance of each alternans type. Therefore, in this presentation, multiscale human in silico models were used to capture key repolarization properties of in vivo recordings, to investigate the ionic mechanisms underlying the occurrence of proarrhythmic repolarization alternans and to identify potential anti-arrhythmic targets.
Population of human ventricular action potential models calibrated using in vivo electrogram recordings. Each simulated action potential in the population is generated using the O'Hara–Rudy model with ionic conductances sampled in a wide range of possible values. Calibration is then conducted using the in vivo electrograms by selecting the models that yield action potentials with properties such as action potential duration consistent with the electrograms (red traces, accepted models), and rejecting those that are outside range. In this figure, the action potential for the original O’Hara–Rudy model is shown in black.
Figure 5

Population of human ventricular action potential models calibrated using in vivo electrogram recordings. Each simulated action potential in the population is generated using the O'Hara–Rudy model with ionic conductances sampled in a wide range of possible values. Calibration is then conducted using the in vivo electrograms by selecting the models that yield action potentials with properties such as action potential duration consistent with the electrograms (red traces, accepted models), and rejecting those that are outside range. In this figure, the action potential for the original O’Hara–Rudy model is shown in black.

Dr Hua Rong Lu described a new, non-invasive and translational biomarker—the index of cardiac electrophysiological balance (iCEB, the ratio between QT and QRS)–in drug and ischaemia-induced cardiac arrhythmias and in genetic LQT syndrome and Brugada syndrome. Currently used biomarkers may not be adequate to detect all types of drug-induced cardiac arrhythmias. Furthermore, there is also a need for a new biomarker to detect cardiac risks in patients with gene-defects in the heart such as LQT syndrome and Brugada syndrome, in patients with heart failure and to detect cardiac risks in sportsmen and women. iCEB was successfully developed and introduced in 2013,80 it may detect potential risks for drug-induced cardiac arrhythmias beyond LQT and Torsade de Pointes. It may also a be better than currently used biomarkers derived from animal models, such as the isolated rabbit left-ventricular wedge model, because iCEB is also detecting additional drug-induced potential cardiac arrhythmias by slowing conduction and QT-shortening.80 iCEB was also found to be significantly increased in patients in genetic LQT syndrome and significantly decreased in patients with Brugada syndrome.

New perspectives/beyond the human heart

Dr Jean-Pierre Valentin introduced the background of compound testing strategies in response to ICH guidelines and how the CiPA initiative6,8183 will develop a new non-clinical paradigm for cardiac safety evaluation of new drugs by shifting the focus away from QT prolongation to an assessment of proarrhythmia to mitigate against the thorough QT study. The proposals would be to consider using ion channel effects in tandem with utilizing the emerging technology of in silico assessment and stem-cell-derived cardiomyocyte effects, but not at the expense of clinical ECGs or an understanding of pharmacokinetics/pharmacodynamics.6 The initiative aims to deliver an implementation of recommendations initially by mid-2016 onwards; however, this is dependent on community support in understanding more about which experimental inputs, which models and which levels of predictive capacity are required.6 Finally, Valentin reminded the audience of a need to keep QT in perspective and to consider cardiac effects beyond QT as it only accounts for 4% of cardiac safety-related drug attrition.84,85 Among the adverse events that are observed are arrhythmia, tachycardia and changes in blood pressure.84,85

Dr Andre Ghetti set out the problem facing the development of pharmaceutical drugs in the translation between in vitro and animal studies to the clinical setting and the critical need to have stronger models at the non-clinical phase for improving success and understanding of drug action. The approach that Dr Ghetti's company AnaBios will be taking is to use primary human tissue to derive cells that can be used more reliably in predicting later clinical effects than in vitro or animal studies can alone. This work is potentially to be used in parallel to the development of in silico approaches that are better informed (parameterized) by the data being generated from these isolated human primary cells. Taken together with a much improved characterization of donors, including medical history, allows us to consider the concept of incorporating real-world-type data into models.

Dr Rebecca Burton presented her recent work on developing a cell culture model of neurally mediated arrhythmogenesis and non-invasive optical imaging methods being pursued at Oxford.86,87 Biological models with varying degrees of complexity have been developed to shed light on re-entrant arrhythmias and cardiac monolayers are one of the simplest models. The next stages of this research is to pursue remote monitoring of in vitro cell cultures that would increase experimental access, reduce the need to sacrifice additional animals, and spur the adoption by other laboratories working in allied research areas. There has been a proliferation of remote access platforms that offer promising functionality. Remote access imaging offers advantages to both ‘wet-lab’ and ‘dry’ experimentation (computational and mathematical modelling), and there is a need for platforms which offer secure and reliable implementation.

Key challenges moving forward

In order to succeed in a programme of research and implementation for drug discovery and testing that takes full advantage of the state of the art in cardiovascular science, the workshop discussions identified five key challenges to be met

  1. Each of the human-based methodologies and techniques presented at the workshop has advantages and disadvantages, as each is able to provide specific forms of data and information, while also leaving some gaps and therefore having limitations. For example,

    • while in vivo human experiments are in many respects the best form of data, there are severe practical and ethical restrictions on acquiring this form of data;

    • ex vivo human data suffer from many similar practical and ethical restrictions, but also from the fact that the techniques (such as cell isolation and voltage clamp) used can sometimes bring about non-trivial effects that need to be compensated for interpreting results;

    • The utility of in vitro experiments on stem-cell-derived cardiomyocytes depends on the quality of the cell type produced by differentiation and subsequent maturation. Currently, this restricts their meaningful use to certain questions depending on the functional integrity of key components which may need to be proven rather than assumed to be intact.

  2. Computational approaches have the potential to bridge these different methodologies, by extracting more value from the data acquired from each, filling in gaps, and facilitating comparison and complementarity between them. This can be achieved through computational approaches for data analysis (signal and image processing to automatically capture and quantify properties, through the use of mathematics and computer science) and through multiscale modelling, which allows for the exploration of mechanisms of disease at different scales, and through making predictions, in the form of new hypotheses for experimental approaches to investigate further.

    Computational modelling and simulation can meet their potential only insofar as they interact in meaningful ways with experimental methodologies and techniques. From an industry perspective, prediction would likely be predominantly used early in drug discovery for prioritization and aiding decision-making in compound selection. Later in drug discovery, the utility is likely to shift to providing a mechanistic understanding of in vivo results either to mitigate safety concerns or provide insights into mechanisms for efficacy in, e.g. antiarrhythmia indications. If computational approaches are developed in a close dialogue with experimental, clinical, and newly emerging digital techniques, they act as mediators between the different data forms and methodologies, and help to forge links between them.

  3. The success of the integration of complementary forms of data and of methods depends upon achieving a true interdisciplinarity of approaches and people. There is a need for expertise from a wide spectrum of disciplines, together with the development of skills at reaching across disciplines and for communicating with researchers who are experts in different methods, and who have different perspectives and priorities. New ways of communicating are called for, as well as new approaches to training next-generation researchers who are capable of interdisciplinarity. In this respect, interdisciplinarity should be widened to include social studies of science in order to get a better grip on institutional, organizational, and social barriers.

  4. Complementarity between different approaches requires input and investment from the scientific community, who are key to defining the criteria to be met for the assessment of drugs and models, through establishing benchmarks, as well as through a reconsideration of different methods and approaches to model validation. A consensus for how the pharma and biotech industry should respond is required and ideally supported by compelling data that draw on a retrospective analysis supporting the reasons to change. The motivation to change in pharma will be aided by the proposed reduced requirements for thorough QT studies and for the opportunity to reconsider compounds that previously have been discontinued due to, e.g. QT prolongation. Implementation will be somewhat dependent upon existing capabilities within the company, e.g. those capable of ion channel screening assays, stem cell assays, and in silico modelling. Even for those where this technology is established, the choice of protocols and standards is key, especially given the laboratory-to-laboratory variance in experimental measurements. From the modelling perspective, which model and which parameter set(s) need to be carefully defined; it would also be necessary to consider CRO partners for these organizations where ion channel screening, stem cell assays, and in silico modelling are impractical.

  5. Finally, further measures to ensure a robust community formed around strong partnerships across different sectors need to be taken, including co-funding strategies that are targeted at developing the required research capability in people and organizations. Robust community formation requires attention to the social elements of the community as well as the scientific research questions.88 The further development of human-based methods have ethical as well as scientific advantages, since they aim to contribute to the 3Rs of animal experimentation; however, they are associated with other forms of social, ethical and legal constraints, and depend on successful engagement with the wider non-scientific community.

Conclusion

In conclusion, the workshop identified key challenges in developing an integrative human-based approach to pharmacology and cardiology through the combination of in silico, in vivo, ex vivo, and in vitro approaches. A clear outcome of the discussions is that these challenges must be tackled synergistically, through joint efforts and discussions across the sectors and stakeholders. A creative strategy that is able to exploit complementarities between approaches needs to be designed and implemented, in a concerted community endeavour that is fully interdisciplinary and intersectoral.

Funding

Financial support was provided by the Knowledge Exchange Fund of the University of Oxford, Wellcome Trust fellowships to B.R. (100246/Z/12/Z) and M.J.D. (WT098519MA), a British Heart Foundation Intermediate Basic Science Research Fellowship to S.H. (FS/12/59/29756), scholarships to S.D., L.C.N. O.J.B., and A.M. from the EPSRC, to A.L. from the British Heart Foundation Centre of Research Excellence, and to X.Z. from the China Scholarship Council. Funding to pay the Open Access publication charges for this article was provided by the Wellcome Trust.

Acknowledgements

The authors are grateful for the contribution of all participants to the workshop.

References

1

Laverty
H
,
Benson
C
,
Cartwright
E
,
Cross
M
,
Garland
C
,
Hammond
T
et al. .
How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines?
Br J Pharmacol
2011
;
163
:
675
93
.

2

Ewart
L
,
Aylott
M
,
Deurinck
M
,
Engwall
M
,
Gallacher
DJ
,
Geys
H
et al. .
The concordance between nonclinical and phase I clinical cardiovascular assessment from a cross-company data sharing initiative
.
Toxicol Sci
2014
;
142
:
427
35
.

3

Koerner
J
,
Valentin
JP
,
Willard
J
,
Park
EJ
,
Bi
D
,
Link
WT
et al. .
Predictivity of non-clinical repolarization assay data for clinical TQT data in the FDA database
.
Int J Toxicol
2013
;
32
:
63
.

4

Severi
S
,
Rodriguez
B
,
Zaza
A
.
Computational cardiac electrophysiology is moving towards translation medicine
.
Europace
2014
;
16
:
703
4
.

5

Severi
S
,
Rodriguez
B
,
Zaza
A
.
Computational cardiac electrophysiology is ready for prime time
.
Europace
2014
;
16
:
382
3
.

6

Sager
PT
,
Gintant
G
,
Turner
JR
,
Pettit
S
,
Stockbridge
N
.
Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium
.
Am Heart J
2014
;
167
:
292
300
.

7

Russell
WMS
,
Burch
RL
.
The Principles of Humane Experimental Technique
.
London
:
Methuen
;
1959
.

8

Plank
G
,
Burton
RAB
,
Hales
P
,
Bishop
M
,
Mansoori
T
,
Bernabeu
MO
et al. .
Generation of histo-anatomically representative models of the individual heart: tools and application
.
Philos Trans A Math Phys Eng Sci
2009
;
367
:
2257
92
.

9

Zemzemi
N
,
Bernabeu
MO
,
Saiz
J
,
Cooper
J
,
Pathmanathan
P
,
Mirams
GR
et al. .
Computational assessment of drug-induced effects on the electrocardiogram: from ion channel to body surface potentials
.
Br J Pharmacol
2013
;
168
:
718
33
.

10

Taggart
P
,
Sutton
P
,
Runnalls
M
,
O'Brien
W
,
Donaldson
R
,
Hayward
R
et al. .
Use of monophasic action potential recordings during routine coronary–artery bypass surgery as an index of localised myocardial ischaemia
.
Lancet
1986
;
1
:
1462
5
.

11

Taggart
P
,
Sutton
PM
,
Treasure
T
,
Lab
M
,
O'Brien
W
,
Runnalls
M
et al. .
Monophasic action potentials at discontinuation of cardiopulmonary bypass: evidence for contraction–excitation feedback in man
.
Circulation
1988
;
77
:
1266
75
.

12

Taggart
P
,
Sutton
PM
,
Boyett
MR
,
Lab
M
,
Swanton
H
.
Human ventricular action potential duration during short and long cycles. Rapid modulation by ischemia
.
Circulation
1996
;
94
:
2526
34
.

13

Taggart
P
,
Sutton
PM
,
Opthof
T
,
Coronel
R
,
Trimlett
R
,
Pugsley
W
et al. .
Inhomogeneous transmural conduction during early ischaemia in patients with coronary artery disease
.
J Mol Cell Cardiol
2000
;
32
:
621
30
.

14

Taggart
P
.
Transmural repolarisation in the left ventricle in humans during normoxia and ischaemia
.
Cardiovasc Res
2001
;
50
:
454
62
.

15

Taggart
P
,
Sutton
P
,
Chalabi
Z
,
Boyett
MR
,
Simon
R
,
Elliott
D
et al. .
Effect of adrenergic stimulation on action potential duration restitution in humans
.
Circulation
2003
;
107
:
285
9
.

16

Taggart
P
,
Sutton
P
.
Load dependence of ventricular repolarisation
. In
Kohl
P
,
Sachs
F
,
Franz
MR
(eds).
Card. Mechano-Electric Coupling
. 2nd ed.
Philadelphia, USA: Elsevier Saunders
;
2011
, p.
269
73
.

17

Taggart
P
,
Sutton
P
.
Termination of arrhythmias by haemodynamic unloading
. In
Kohl
P
,
Sachs
F
,
Franz
MR
(eds).
Card. Mechano-Electric Coupling
.
Philadelphia, USA: Elsevier Saunders
;
2011
, p.
369
73
.

18

Nash
MP
,
Mourad
A
,
Clayton
RH
,
Sutton
PM
,
Bradley
CP
,
Hayward
M
et al. .
Evidence for multiple mechanisms in human ventricular fibrillation
.
Circulation
2006
;
114
:
536
42
.

19

Bradley
CP
,
Clayton
RH
,
Nash
MP
,
Mourad
A
,
Hayward
M
,
Paterson
DJ
et al. .
Human ventricular fibrillation during global ischemia and reperfusion: paradoxical changes in activation rate and wavefront complexity
.
Circ Arrhythm Electrophysiol
2011
;
4
:
684
91
.

20

Coronel
R
,
Janse
MJ
,
Opthof
T
,
Wilde
AA
,
Taggart
P
.
Postrepolarization refractoriness in acute ischemia and after antiarrhythmic drug administration: action potential duration is not always an index of the refractory period
.
Heart Rhythm
2012
;
9
:
977
82
.

21

Maron
BJ
,
Gardin
JM
,
Flack
JM
,
Gidding
SS
,
Kurosaki
TT
,
Bild
DE
.
Prevalence of hypertrophic cardiomyopathy in a general population of young adults. Echocardiographic analysis of 4111 subjects in the CARDIA Study. Coronary Artery Risk Development in (Young) Adults
.
Circulation
1995
;
92
:
785
9
.

22

Burke
AP
,
Farb
A
,
Virmani
R
,
Goodin
J
,
Smialek
JE
.
Sports-related and non-sports-related sudden cardiac death in young adults
.
Am Heart J
1991
;
121
:
568
75
.

23

Force
T
,
Bonow
RO
,
Houser
SR
,
Solaro
RJ
,
Hershberger
RE
,
Adhikari
B
et al. .
Research priorities in hypertrophic cardiomyopathy: report of a Working Group of the National Heart, Lung, and Blood Institute
.
Circulation
2010
;
122
:
1130
3
.

24

Rubinshtein
R
,
Glockner
JF
,
Ommen
SR
,
Araoz
PA
,
Ackerman
MJ
,
Sorajja
P
et al. .
Characteristics and clinical significance of late gadolinium enhancement by contrast-enhanced magnetic resonance imaging in patients with hypertrophic cardiomyopathy
.
Circ Heart Fail
2010
;
3
:
51
8
.

25

Petersen
SE
,
Jerosch-Herold
M
,
Hudsmith
LE
,
Robson
MD
,
Francis
JM
,
Doll
HA
et al. .
Evidence for microvascular dysfunction in hypertrophic cardiomyopathy: new insights from multiparametric magnetic resonance imaging
.
Circulation
2007
;
115
:
2418
25
.

26

Karamitsos
TD
,
Dass
S
,
Suttie
J
,
Sever
E
,
Birks
J
,
Holloway
CJ
et al. .
Blunted myocardial oxygenation response during vasodilator stress in patients with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
2013
;
61
:
1169
76
.

27

Dass
S
,
Suttie
J
,
Karamitsos
T
,
Watkins
H
,
Neubauer
S
.
081 Acute derangement of cardiac energy metabolism and oxygenation during stress in hypertrophic cardiomyopathy: a potential mechanism for sudden cardiac death
.
Heart
2012
;
98
:
A45
6
.

28

Tunnicliffe
EM
,
Scott
AD
,
Ferreira
P
,
Ariga
R
,
McGill
L-A
,
Nielles-Vallespin
S
et al. .
Intercentre reproducibility of cardiac apparent diffusion coefficient and fractional anisotropy in healthy volunteers
.
J Cardiovasc Magn Reson
2014
;
16
:
31
.

29

Abozguia
K
,
Elliott
P
,
McKenna
W
,
Phan
TT
,
Nallur-Shivu
G
,
Ahmed
I
et al. .
Metabolic modulator perhexiline corrects energy deficiency and improves exercise capacity in symptomatic hypertrophic cardiomyopathy
.
Circulation
2010
;
122
:
1562
9
.

30

Hsu
EW
,
Muzikant
AL
,
Matulevicius
SA
,
Penland
RC
,
Henriquez
CS
.
Magnetic resonance myocardial fiber-orientation mapping with direct histological correlation
.
Am J Physiol
1998
;
274
:
H1627
34
.

31

Scollan
DF
,
Holmes
A
,
Winslow
R
,
Forder
J
.
Histological validation of myocardial microstructure obtained from diffusion tensor magnetic resonance imaging
.
Am J Physiol
1998
;
275
:
H2308
18
.

32

Lohezic
M
,
Teh
I
,
Bollensdorff
C
,
Peyronnet
R
,
Hales
PW
,
Grau
V
et al. .
Interrogation of living myocardium in multiple static deformation states with diffusion tensor and diffusion spectrum imaging
.
Prog Biophys Mol Biol
2014
;
115
:
213
25
.

33

Gibb
M
,
Burton
RAB
,
Bollensdorff
C
,
Afonso
C
,
Mansoori
T
,
Schotten
U
et al. .
Resolving the three-dimensional histology of the heart
.
Comput Methods Syst Biol
2012
,
Lecture Notes in Computer Science 7605; pp. 2–16. doi: 10.1007/978-3-642-33636-2_2
.

34

Bates
J
,
Teh
I
,
Kohl
P
,
Schneider
JE
,
Grau
V
.
Sensitivity Analysis of Diffusion Tensor MRI in Simulated Rat Myocardium
.
FIMH. Switzerland: Springer International Publishing
;
2015
. pp.
120
8
.

35

Carapella
V
,
Bordas
R
,
Pathmanathan
P
,
Lohezic
M
,
Schneider
JE
,
Kohl
P
et al. .
Quantitative study of the effect of tissue microstructure on contraction in a computational model of rat left ventricle
.
PLoS ONE
2014
;
9
:
e92792
.

36

Gonzales
MJ
,
Vincent
KP
,
Rappel
W-J
,
Narayan
SM
,
McCulloch
AD
.
Structural contributions to fibrillatory rotors in a patient-derived computational model of the atria
.
Europace
2014
;
16
(Suppl. 4)
:
iv3
10
.

37

Burton
RAB
,
Lee
P
,
Casero
R
,
Garny
A
,
Siedlecka
U
,
Schneider
JE
et al. .
Three-dimensional histology: tools and application to quantitative assessment of cell-type distribution in rabbit heart
.
Europace
2014
;
16
(Suppl. 4)
:
iv86
95
.

38

Sörnmo
L
,
Laguna
P
.
Bioelectrical Signal Processing in Cardiac and Neurological Applications
. 1st ed.
Burlington, MA, USA: Elsevier Academic Press
;
2005
.

39

Mincholé
A
,
Ariga
R
,
Neubauer
S
,
Watkins
H
,
Rodriguez
B
.
Electrocardiographic abnormalities in hypertrophic cardiomyopathy
.
Comput Cardiol
2014
;
41
;
377
80
.

40

Lyon
A
,
Mincholé
A
,
Ariga
R
,
Laguna
P
,
Neubauer
S
,
Watkins
H
et al. .
Extraction of morphological QRS-based biomarkers in hypertrophic cardiomyopathy for risk stratification using L1 regularized logistic regression
.
Comput Cardiol
2015
;
42
.

41

Zemzemi
N
,
Rodriguez
B
.
Effects of L-type calcium channel and human ether-a-go-go related gene blockers on the electrical activity of the human heart: a simulation study
.
Europace
2014
;
17
:
326
33
.

42

Potse
M
,
Krause
D
,
Kroon
W
,
Murzilli
R
,
Muzzarelli
S
,
Regoli
F
et al. .
Patient-specific modelling of cardiac electrophysiology in heart-failure patients
.
Europace
2014
;
16
(Suppl. 4)
:
iv56
61
.

43

Ramanathan
C
,
Jia
P
,
Ghanem
R
,
Ryu
K
,
Rudy
Y
.
Activation and repolarization of the normal human heart under complete physiological conditions
.
Proc Natl Acad Sci USA
2006
;
103
:
6309
14
.

44

Rudy
Y
.
Noninvasive electrocardiographic imaging of arrhythmogenic substrates in humans
.
Circ Res
2013
;
112
:
863
74
.

45

Zhang
J
,
Sacher
F
,
Hoffmayer
K
,
O'Hara
T
,
Strom
M
,
Cuculich
P
et al. .
Cardiac electrophysiological substrate underlying the ECG phenotype and electrogram abnormalities in Brugada syndrome patients
.
Circulation
2015
;
131
:
1950
9
.

46

Sivagangabalan
G
,
Nazzari
H
,
Bignolais
O
,
Maguy
A
,
Naud
P
,
Farid
T
et al. .
Regional ion channel gene expression heterogeneity and ventricular fibrillation dynamics in human hearts
.
PLoS ONE
2014
;
9
:
e82179
.

47

Jost
N
,
Virág
L
,
Bitay
M
,
Takács
J
,
Lengyel
C
,
Biliczki
P
et al. .
Restricting excessive cardiac action potential and QT prolongation: a vital role for IKs in human ventricular muscle
.
Circulation
2005
;
112
:
1392
9
.

48

Holzem
KM
,
Madden
EJ
,
Efimov
IR
.
Human cardiac systems electrophysiology and arrhythmogenesis: iteration of experiment and computation
.
Europace
2014
;
16
(Suppl. 4)
:
iv77
85
.

49

Jost
N
,
Virág
L
,
Comtois
P
,
Ordög
B
,
Szuts
V
,
Seprényi
G
et al. .
Ionic mechanisms limiting cardiac repolarization reserve in humans compared to dogs
.
J Physiol
2013
;
591
:
4189
206
.

50

Lu
HR
,
Mariën
R
,
Saels
A
,
De Clerck
F
.
Species plays an important role in drug-induced prolongation of action potential duration and early afterdepolarizations in isolated Purkinje fibers
.
J Cardiovasc Electrophysiol
2001
;
12
:
93
102
.

51

O'Hara
T
,
Virág
L
,
Varró
A
,
Rudy
Y
.
Simulation of the undiseased human cardiac ventricular action potential: model formulation and experimental validation
.
PLoS Comput Biol
2011
;
7
:
e1002061
.

52

Grandi
E
,
Pasqualini
FS
,
Bers
DM
.
A novel computational model of the human ventricular action potential and Ca transient
.
J Mol Cell Cardiol
2010
;
48
:
112
21
.

53

Ten Tusscher
KHWJ
,
Panfilov
AV
.
Alternans and spiral breakup in a human ventricular tissue model
.
Am J Physiol Heart Circ Physiol
2006
;
291
:
H1088
100
.

54

Ten Tusscher
KHWJ
,
Noble
D
,
Noble
PJ
,
Panfilov
AV
.
A model for human ventricular tissue
.
Am J Physiol Heart Circ Physiol
2004
;
286
:
H1573
89
.

55

Britton
OJ
,
Bueno-Orovio
A
,
Van Ammel
K
,
Lu
HR
,
Towart
R
,
Gallacher
DJ
et al. .
Experimentally calibrated population of models predicts and explains intersubject variability in cardiac cellular electrophysiology
.
Proc Natl Acad Sci USA
2013
;
110
:
E2098
105
.

56

Burridge
PW
,
Matsa
E
,
Shukla
P
,
Lin
ZC
,
Churko
JM
,
Ebert
AD
et al. .
Chemically defined generation of human cardiomyocytes
.
Nat Methods
2014
;
11
:
855
60
.

57

Yang
X
,
Pabon
L
,
Murry
CE
.
Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes
.
Circ Res
2014
;
114
:
511
23
.

58

Doss
MX
,
Di Diego
JM
,
Goodrow
RJ
,
Wu
Y
,
Cordeiro
JM
,
Nesterenko
VV
et al. .
Maximum diastolic potential of human induced pluripotent stem cell-derived cardiomyocytes depends critically on IKr
.
PLoS ONE
2012
;
7
:
e40288
.

59

Lian
X
,
Hsiao
C
,
Wilson
G
,
Zhu
K
,
Hazeltine
LB
,
Azarin
SM
et al. .
Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling
.
Proc Natl Acad Sci USA
2012
;
109
:
E1848
57
.

60

Paci
M
,
Hyttinen
J
,
Aalto-Setälä
K
,
Severi
S
.
Computational models of ventricular- and atrial-like human induced pluripotent stem cell derived cardiomyocytes
.
Ann Biomed Eng
2013
;
41
:
2334
48
.

61

Paci
M
,
Hyttinen
J
,
Severi
S
.
Computational modelling of LQT1 in human induced pluripotent stem cell derived cardiomyocytes
.
Comput Cardiol
2013
;
40
:
1239
42
.

62

Paci
M
,
Hyttinen
J
,
Severi
S
.
Computational modeling supports induced pluripotent stem cell-derived cardiomyocytes reliability as a model for human LQT3
.
Comput Cardiol
2014
;
41
:
69
72
.

63

Ma
J
,
Guo
L
,
Fiene
SJ
,
Anson
BD
,
Thomson
JA
,
Kamp
TJ
et al. .
High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents
.
Am J Physiol Heart Circ Physiol
2011
;
301
:
H2006
17
.

64

Moretti
A
,
Bellin
M
,
Welling
A
,
Jung
CB
,
Lam
JT
,
Bott-Flügel
L
et al. .
Patient-specific induced pluripotent stem-cell models for long-QT syndrome
.
N Engl J Med
2010
;
363
:
1397
409
.

65

Bellin
M
,
Casini
S
,
Davis
RP
,
D'Aniello
C
,
Haas
J
,
Ward-van Oostwaard
D
et al. .
Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome
.
EMBO J
2013
;
32
:
3161
75
.

66

Ma
D
,
Wei
H
,
Zhao
Y
,
Lu
J
,
Li
G
,
Sahib
NBE
et al. .
Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells
.
Int J Cardiol
2013
;
168
:
5277
86
.

67

Paci
M
,
Hyttinen
J
,
Rodriguez
B
,
Severi
S
.
Human induced pluripotent stem cell-derived versus adult cardiomyocytes: an in silico electrophysiological study on ionic current block effects
.
Br J Pharmacol
2015
;
doi:10.1111/bph.13282. [Epub ahead of print]

68

Cook
D
,
Brown
D
,
Alexander
R
,
March
R
,
Morgan
P
,
Satterthwaite
G
et al. .
Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework
.
Nat Rev Drug Discov
2014
;
13
:
419
31
.

69

Dutta
S
,
Mincholé
A
,
Quinn
TA
,
Rodriguez
B
.
Class III drugs in human ischemic ventricles: anti- or pro-arrhythmic action
.
Europace
2014
;
16
(Suppl. 2)
:
686
90
.

70

Clifford
GD
,
Clifton
D
.
Wireless technology in disease management and medicine
.
Annu Rev Med
2012
;
63
:
479
92
.

71

Llamedo
M
,
Martinez
JP
.
An automatic patient-adapted ECG heartbeat classifier allowing expert assistance
.
IEEE Trans Biomed Eng
2012
;
59
:
2312
20
.

72

Sarkar
S
,
Ritscher
D
,
Mehra
R
.
A detector for a chronic implantable atrial tachyarrhythmia monitor
.
IEEE Trans Biomed Eng
2008
;
55
:
1219
24
.

73

Colloca
R
,
Johnson
AEW
,
Mainardi
L
,
Clifford
GD
.
A Support Vector Machine approach for reliable detection of atrial fibrillation events
.
Comput Cardiol
2013
;
40
;
1047
50
.

74

Oster
J
,
Behar
J
,
Colloca
R
,
Qichen
Li
,
Qiao
Li
,
Clifford
GD
.
Open source Java-based ECG analysis software and Android app for atrial fibrillation screening
.
Comput Cardiol
2013
;
40
;
731
4
.

75

Schmidt
G
,
Malik
M
,
Barthel
P
,
Schneider
R
,
Ulm
K
,
Rolnitzky
L
et al. .
Heart-rate turbulence after ventricular premature beats as a predictor of mortality after acute myocardial infarction
.
Lancet
1999
;
353
:
1390
6
.

76

McSharry
PE
,
Clifford
GD
,
Tarassenko
L
,
Smith
LA
.
A dynamical model for generating synthetic electrocardiogram signals
.
IEEE Trans Biomed Eng
2003
;
50
:
289
94
.

77

Sameni
R
,
Shamsollahi
MB
,
Jutten
C
,
Clifford
GD
.
A nonlinear Bayesian filtering framework for ECG denoising
.
IEEE Trans Biomed Eng
2007
;
54
:
2172
85
.

78

Oster
J
,
Behar
J
,
Sayadi
O
,
Nemati
S
,
Johnson
A
,
Clifford
G
.
Semi-supervised ECG ventricular beat classification with novelty detection based on switching Kalman filters
.
IEEE Trans Biomed Eng
2015
;
62
:
2125
34
.

79

Zhou
X
,
Bueno-Orovio
A
,
Orini
M
,
Hanson
B
,
Haywood
M
,
Taggart
P
et al. .
Population of human ventricular cell models calibrated with in vivo measurements unravels ionic mechanisms of cardiac alternans
.
Comput Cardiol
2013
;
40
;
855
8
.

80

Lu
HR
,
Yan
G-X
,
Gallacher
DJ
.
A new biomarker-index of cardiac electrophysiological balance (iCEB)-plays an important role in drug-induced cardiac arrhythmias: beyond QT-prolongation and Torsades de Pointes (TdPs)
.
J Pharmacol Toxicol Methods
2013
;
68
:
250
9
.

81

Anonymous
.
The Clinical Evaluation of QT/QTc Interval Prolongation and Proarrhythmic Potential for Non-Antiarrhythmic Drugs (ICH-E14) 2005
.

82

Anonymous
.
The Non-Clinical Evaluation of the Potential for Delayed Ventricular Repolarization (QT interval Prolongation) by Human Pharmaceuticals (ICH-S7B) 2005
.

83

Pollard
CE
,
Abi Gerges
N
,
Bridgland-Taylor
MH
,
Easter
A
,
Hammond
TG
,
Valentin
J-P
.
An introduction to QT interval prolongation and non-clinical approaches to assessing and reducing risk
.
Br J Pharmacol
2010
;
159
:
12
21
.

84

Redfern
WS
,
Ewart
L
,
Hammond
TG
,
Bialecki
R
,
Kinter
L
,
Lindgren
S
et al. .
Impact and frequency of different toxicities throughout the pharmaceutical life cycle
.
Toxicol
2010
;
114
(Suppl. 1)
:
1081
.

85

Valentin
J-P
,
Kenna
JG
,
Lainée
P
,
Redfern
WS
,
Roberts
S
,
Hammond
TG
.
A literature-based analysis of cardiovascular adverse events impacting on drug development
.
J Pharmacol Toxicol Methods
2012
;
66
:
173
.

86

Burton
RAB
,
Stephens
G
,
Sharkey
A
,
Bilton
S
,
Larsen
H
,
Kramer
H
et al. .
Spatiotemporal transitions in cardiac neuronal co-cultures
.
Biophys J
2014
;
106
:
630a
.

87

Bub
G
,
Burton
R-AB
.
Macro-micro imaging of cardiac-neural circuits in co-cultures from normal and diseased hearts
.
J Physiol
2014
;
593
:
3047
53.

88

Leonelli
S
,
Ankeny
R
.
Repertoires: how to transform a project into a research community
.
Bioscience
2015
:.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.