The Comprehensive In Vitro Proarrhythmia Assay Initiative

, Dilip R. Karnad2 and Snehal Kothari3



(1)
Cardiac Safety Services Quintiles, Durham, North Carolina, USA

(2)
Research Team, Cardiac Safety Services Quintiles, Mumbai, India

(3)
Cardiac Safety Services Global Head, Cardiac Safety Center of Excellence Quintiles, Mumbai, India

 



These new strategies have the potential to improve sensitivity and specificity in the early detection of genuine cardiotoxicity risks, thereby reducing the likelihood of mistakenly discarding viable drug candidates and speeding the progression of worthy drugs into clinical trials (Gintant et al. 2016).



9.1 Introduction


This chapter discusses the Comprehensive In Vitro Proarrhythmia Assay (CiPA) initiative, an integrated set of investigations that may lead to modifications of the nonclinical proarrhythmic cardiac safety regulatory landscape. Underlying this initiative is the fact that drug-induced I Kr reduction and QTc interval prolongation are far from ideal surrogates for actual proarrhythmic risk, the true concern in the domain of proarrhythmic cardiac safety. The link between drug-induced QTc prolongation and the rare (but potentially lethal) arrhythmia torsades appears to be multifaceted, and occurrence of drug-induced torsades typically requires multiple contributing factors to be present simultaneously. Some of these clinical risk factors include female sex, structural heart disease, metabolic and electrolyte abnormalities, bradycardia and pauses, increased concentrations of “culprit” drugs, and inherited syndromes causing QT prolongation (Vlachos et al. 2016). Sager and colleagues (2014) therefore observed that, given our advancing knowledge and understanding on multiple fronts, it is incumbent upon us to provide a more comprehensive evaluation of actual proarrhythmic risk. These advances are evident in our knowledge of mechanisms responsible for torsades, our ability to evaluate drug effects on human cardiac ion channels, successes in in silico modeling of human ventricular electrical activity, and the evolving employment of isolated human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs).

The CiPA paradigm is comprised of three fundamental components: a two-component core in vitro strategy assessing effects on isolated cardiac currents and integrated drug responses from hiPSC-CMs and in silico reconstructions of cellular electrophysiological activity. An additional step is the definition of the proarrhythmic risk of a select group of drugs used clinically, thereby defining a “gold standard” for comparison of nonclinical and clinical findings. Organized as a public–private collaboration, CiPA represents a mechanistic-based set of investigations that will provide a better understanding of a drug’s proarrhythmic liability. The initiative is being driven by an international consortium comprising multiple collaborators including the FDA, EMA, Health Canada, PMDA, CSRC, the Health and Environmental Sciences Institute (HESI), the Safety Pharmacology Society (SPS), Japan National Institute of Health Sciences, Japan iPS Cardiac Safety Assessment, academics, in silico modelers, and partners from contract research organizations, the pharmaceutical industry, stem cell providers, and medical device companies (Fermini et al. 2016).

The first major presentation and discussion of CiPA occurred at a July 2013 Think Tank Meeting sponsored by CSRC, HESI, and FDA: a report was provided by Sager and colleagues (2014). Following that meeting, workstreams were created to focus on various aspects of CiPA, with each workstream operating under the auspices of a collaborating organization. The Ion Channel Working Group was established by SPS, the In Silico Working Group operates under the direction of FDA, the Cardiac Myocyte Working Group is sponsored by HESI, and the Clinical Translation Working Group operates under joint direction from the CSRC and HESI. A subsequent Think Tank Meeting sponsored by FDA, CSRC, HESI, and SPS provided an update on their work in December 2014, and additional discussion has been presented by Fermini and colleagues (Fermini et al. 2016).


9.2 Advantages of the Proposed Paradigm


The CiPA paradigm addresses many of the concerns related to the traditional ICH S7B and ICH E14 approaches. Importantly, CiPA focuses on the assessment of potential ventricular proarrhythmia risk via a mechanistically robust set of input data rather than employment of the surrogates of I Kr reduction and QTc prolongation. Drugs with increased proarrhythmic risk would be assessed for their potential to enhance vulnerability to disrupt repolarization rather than simply to delay repolarization. Moving the mainstay of the assessment of proarrhythmic risk away from later (clinical) drug development to earlier stages of drug discovery, hence permitting its deployment in candidate selection, will likely prevent the current common scenario that a small QTc prolongation signal seen in drug development constitutes uncertain risk that is to be avoided, resulting in the premature discontinuation of a compound that may have been therapeutically beneficial and had an acceptable benefit–risk balance.

A second advantage is that the CiPA paradigm is not dependent on dichotomous categorization, i.e., prolongation vs. no prolongation of QTc. A graduated risk scale that would permit improved benefit–risk assessments is envisioned. Some degree of proarrhythmic risk may be considered acceptable for some drugs addressing unmet medical needs, e.g., serious oncologic diseases and chronic, debilitating diseases, but not for other drugs such as those for allergic rhinitis. The vision for CiPA is a nonbinary output in which novel compounds are given a proarrhythmic risk score predicated on a continuous scale that has been calibrated against a test set of clinical drugs spanning the range of proarrhythmic risk.

This paradigm also offers the facilitation of more informative package inserts at the time of a new drug’s marketing approval and also the relabeling of those compounds that currently have QTc warnings and precautions but for which a true low proarrhythmic risk can now be identified.


9.3 Cardiac Ionic Currents of Particular Interest


Numerous overlapping ionic currents contribute to the determination of the morphology and duration of the ventricular action potential: those discussed in this chapter are summarized in Table 9.1.


Table 9.1
Ionic currents and their influence on each phase of the action potential































Ionic current

Influence

I Na[fast]

Rapid inward sodium current responsible for depolarization and rising phase of the action potential (phase 0),

I to

Transient outward repolarizing current comprised of multiple overlapping outward repolarizing potassium currents (phase 1)

I Na[late]

Late sodium (depolarizing) current governing the plateau phase of the action potential (phase 2)

I CaL

L-type calcium (depolarizing) current governing the plateau phase of the action potential (phase 2)

I Ks

Slow component of the delayed rectifier potassium current especially prominent in supporting repolarization (phase 3) with beta-adrenergic stimulation

I Kr

Rapid component of the delayed rectifier potassium current responsible for the transition from plateau to terminal (phase 3) repolarization

I K1

Inward rectifier potassium current (an outward repolarizing current) responsible for terminal repolarization of the action potential and sustaining the resting membrane potential of ventricular cardiomyocytes during diastole (phase 4)

While there are multiple repolarizing potassium currents, I Kr has attracted particular attention in two contexts: the inherited channelopathy LQT2 (Bunch and Ackerman 2007; McBride et al. 2013; Smith et al. 2013) (recall discussions in Chap. 3) and drug-induced (acquired) QTc prolongation. In LQT2, an abnormal variant of the hERG gene produces hERG channels with decreased expression or function, leading to a decrease in repolarizing I Kr. In the case of drug-induced hERG channel blockade, a small-molecule drug typically binds to the inner walls of the central pore of the channel and impedes I Kr. Many noncardiac drugs block the hERG channel in preference to other cardiac potassium channels as a result of differences in its intracellular channel vestibule and pore structure, characteristics that have led to the channel being described as “an unusually promiscuous target among potassium channels” (Lagrutta et al. 2008). While the mechanism of action is different (inherited vs. acquired), hERG blockade therefore results in the same cascade of occurrences as does LQT2, i.e., a reduction of repolarizing I Kr and delayed ventricular repolarization manifesting as QT prolongation. Given this commonality, drug-induced QT prolongation, like LQT2, is also of considerable clinical concern.

All other influences being kept equal, a drug-induced decrease in I Kr will lead to a reduction in overall (net) repolarizing current. However, the relationships between the amount of hERG block, the extent of delayed repolarization/QT prolongation, and the proclivity and incidence of proarrhythmia/torsades are uncertain and likely dependent on the kinetics and extent of I Kr block, block of other cardiac channels during repolarization (termed multi-channel block), and the underlying electrophysiologic substrate (which may be altered in the setting of cardiac disease). Two possibilities arise. The same drug molecule can affect one or more of the multiple other cardiac ion channels, thereby affecting multiple ionic currents to affect net outward current. Depending on the channels affected, as well as kinetics and extent of block, multi-channel block may effectively minimize or cancel out the decrease in repolarizing influence due to hERG channel block, leading to minimal (if any) proarrhythmic effects. Alternatively, decreases in both I Kr and I Ks could have additive/synergistic effects not appreciated by evaluating hERG channel block alone. This notion is embodied in the term repolarization reserve, which recognizes that both I Kr and I Ks define ventricular repolarization: with one current (e.g., I Ks) already reduced, block of the second (I Kr) will have greater effects on repolarization and potentially convey more proarrhythmic liability in the presence of reduced baseline repolarizing current. Malik (2016) commented on the term repolarization reserve as follows:

It suggests that the interplay between different ion channels that maintain myocardial repolarization is to some extent redundant and that this redundancy offers mechanisms protecting against externally induced abnormalities including drug-induced anomalies. The susceptibility to arrhythmic consequences of drug-induced repolarization changes markedly increases when other pathological processes reduce this built-in protection.

Advances in the capabilities and adoption of higher-throughput automated voltage clamp patch platforms will facilitate more efficient characterization of drug effects on multiple cardiac currents (Dale et al. 2007; Castle et al. 2009; Ma et al. 2011; Farre and Fertig 2012; Di Veroli et al. 2013). Employing higher-throughput automated patch techniques will also provide sufficient sample size and statistical power to facilitate parameterization of subsequent in silico reconstruction efforts and to determine IC50 and other characteristics of block as deemed necessary to provide reliable, reproducible characterization of integrated electrophysiological effects.


9.4 Electrophysiological Principles Underlying CiPA


As previously noted, the relationships between the degree of hERG block, the extent of delayed repolarization/QT prolongation, and the proclivity and incidence of proarrhythmia/torsades are uncertain. Not all drugs that prolong QTc are proarrhythmic; examples include ranolazine, phenobarbital, and tolterodine. Verapamil is a potent hERG current blocker, but it does not cause QTc prolongation (except possibly at very high intravenous exposures), likely as a result of its concomitant blockade of calcium current (Zhang et al. 1999). Amiodarone is an example of a drug that causes marked QTc prolongation (not infrequently to lengths >550 msec) and yet only very rarely causes torsades.

It is likely that drug effects on multiple cardiac currents, especially reductions of inward (depolarizing) calcium and sodium currents during the action potential plateau, provide protection from proarrhythmia when coupled with a decrease of I Kr. The concept that block of non-hERG currents may mitigate proarrhythmic effects of hERG current block is not new: it has been known for some years that combining block of repolarizing potassium current with either sodium or calcium channel block may reduce or reverse early after-depolarization (EAD) formation (Bril et al. 1996; Martin et al. 2004), a topic discussed in the following section. A review of the potency of I Kr block (relative to clinical exposures) and TQT study results for 39 drugs demonstrated the need for additional nonclinical assays addressing drug effects on other currents to assess more comprehensively the risk of QTc prolongation (Gintant 2011). More recently, a logistic-regression approach involving assessment of drug effects on three cardiac channels, Kv11.1 or hERG (I Kr), fast sodium Nav1.5 (I Na fast), and Cav1.2 (I CaL), showed a significant reduction in false-positive and false-negative classifications for 55 drugs from multiple classes (32 torsadogenic and 23 non-torsadogenic drugs) as compared with predictions based on I Kr block alone (Kramer et al. 2013).

These observations reinforce the need to consider drug effects on multiple cardiac currents when assessing proarrhythmic liabilities. Some companies are already screening multiple cardiac ion channels in drug discovery (Davies et al. 2012). The ionic current studies component of the CiPA paradigm would provide standardized, best practice assays to ensure valuable data sets that inform decisions regarding cardiac safety early in drug discovery, provide guidance on first-in-human studies, and generate valuable information for regulatory considerations.


9.4.1 Early After-Depolarizations


Early after-depolarizations (EADs) are slowly rising depolarizations that occur during the later phases of an action potential after the initial depolarization (termed the “triggering event”) that inscribe a second depolarizing upstroke (hence the term “after-depolarization”) that occurs prior to full repolarization (hence an “early” after-depolarization) (see Chang et al. 2012). A stylistic representation of an EAD can be seen as Figure 1B in an Open Access paper by Sager and colleagues (2014). If they are of sufficiently large amplitude and occur at specific times during repolarization, EADs can trigger single or multiple premature ventricular depolarizations that may propagate throughout the ventricles. In the setting of enhanced dispersion of repolarization (a phenomenon that can occur with nonuniform drug-induced prolongation of repolarization in different regions of the ventricular myocardium), and sometimes coupled to rhythm disturbances, EAD-triggered responses may give rise to torsades. CiPA therefore focuses on cellular electrophysiological effects of drugs that modify repolarization to enable EADs. This focus is in line with generally increasing emphasis being placed on early EADs and the microenvironments that elicit and support their morphology and dynamics (Qu et al. 2013b).

Outward potassium current, mostly I Kr, promotes repolarization and suppresses re-excitation during the plateau of each action potential until terminal repolarization ensues with the subsequent contribution of I K1. When repolarization is impaired, EADs can arise, initiating triggered activity likely resulting from I CaL “reactivation” or window current. The dynamic balance of inward vs. outward currents predisposes to triggered activity. Arrhythmias can only arise where net inward current during repolarization occurs, allowing the early activity of one part of the heart to affect activity in another part with a delay, which then eventually makes its way back to the first area with sufficient delay to set up a loop or reentrant circuit. Although the exact circumstances that give rise to the conduction loop are not well characterized, the vulnerability resulting from a drug’s effects on various ion channels leading to triggered activity can be assessed with great precision using in silico reconstructions based on human ventricular activity (Sager et al. 2014).


9.5 CiPA’s Core In Vitro Strategy


The core in vitro strategy focuses on the evaluation of drug effects on multiple isolated human cardiac currents via heterologous expression systems assessed electrically using voltage/patch clamp techniques. As we have seen, multiple currents define the cardiac action potential, and knowledge gleaned from inherited LQTS and drug-induced proarrhythmia convincingly demonstrate that repolarizing and depolarizing currents must both be considered to understand proarrhythmia. I Kr represents only one of multiple potassium and sodium currents that, when differing from normal, are associated with long QT and proarrhythmia. Studies of acquired LQT syndromes and proarrhythmia demonstrate that fast inward sodium current and enhanced inward current (I Na-Late or reactivation of calcium current during the action potential plateau) are also involved in proarrhythmia. Thus, a more comprehensive in vitro set of ion current assays could conceivably explore I Kr, I Ks, and I K1, as well as I Na-Fast, I Na-Late, and I Ca-L for drug effects. The specific currents to be evaluated to generate a sufficiently comprehensive and predictive data set are currently under discussion by various workstreams.

The use of voltage clamp studies for unbiased and standardized decision-making in arrhythmia evaluation will necessitate the development of consensus on best practices and/or standardization of protocols, positive/negative controls, and experimental conditions. This effort will reduce variability, allow comparisons across assays and laboratories, and generate movement towards more uniform data quality for purposes of decision-making, both by sponsors internally and by regulatory agencies. As one example, multiple studies demonstrate that the potency of I Kr blockade is, at least for some drugs, affected by the experimental temperature, i.e., room temperature vs. physiologic temperature (37oC). It is becoming clear that the potency of drugs that demonstrate prominent temperature-dependent effects is uncertain, necessitating an evaluation of hERG blocking potency at physiologic temperature.

Within CiPA, the potency of current block (based on IC50 values) relative to free drug plasma concentration will be a key component in evaluating a drug’s proarrhythmic liability. Further characterization of the kinetics of block, e.g., including voltage, time, and concentration dependence, might be critical for some currents, likely including I Kr: comparison of in silico studies incorporating conductance block models with those incorporating kinetics of drug block and unblock will guide future discussions (Di Veroli et al. 2013).


9.6 CiPA’s In Silico Modeling Component


In silico models of cellular human ventricular activity are employed to integrate drug effects on multiple cardiac currents, providing reconstructions of cellular electrical activity. Electrophysiological models have been used since the pioneering work of Hodgkin and Huxley to reconstruct neuronal excitability of squid giant axons based on contributions of overlapping voltage- and time-dependent sodium and potassium currents (Hodgkin and Huxley 1952; Krouchev et al. 2015). In the CiPA paradigm, voltage clamp data describing a drug’s effects on multiple ionic currents, based on the O’Hara–Rudy model (O’Hara et al. 2011), will describe effects on ventricular repolarization that are not easily understood from effects on any individual cardiac current.

It is envisioned that in silico reconstructions will provide information on two fronts: drug effects related to the ability to elicit EADs during Phase 3 repolarization based on measures of net current during repolarization and evaluation of the robustness of repolarization based on the ability of depolarizing currents applied during the action potential plateau to amplify delayed repolarization and EAD activity. A scoring system may be necessary to rank order proarrhythmic risk based on measures of repolarization instability calibrated against a training set of compounds affecting multiple cardiac ion channels that have been ranked according to clinical torsades risk. This continuous scoring system could then be used to rank order drug candidates’ risk of torsades proarrhythmia in the context of therapeutic margins, e.g., therapeutic concentration and plasma protein binding.

In support of the use of integrative in silico models, a study by Mirams and colleagues (Mirams et al. 2011) employing in silico modeling to measure action potential prolongation demonstrated that evaluation of drug effects across three human ion channels (Kv11.1, Nav1.5, and Cav1.2) improved prediction of torsadogenic risk compared with evaluations based solely on hERG channel block. Numerous studies have described the general utility of in silico ventricular reconstructions in evaluating overall delayed repolarization liabilities and/or proarrhythmic risk (Valentin and Hammond 2008; Fletcher et al. 2011; Mirams et al. 2011, 2012; Kramer et al. 2013; Beattie et al. 2013).

The best in silico cellular model(s) for reconstructions will have to be selected and then made available to users in a standardized format to provide meaningful ranking of proarrhythmia across different laboratories or, alternatively, be made widely available on a centralized cloud-based resource.


9.7 Effects on hiPSC-CMs


CiPA’s third component is the evaluation of drug effects on the electrical activity of hiPSC-CMs. This approach yields a cell-based integrated electrophysiological drug response, providing a check on the adequacy of the voltage clamp data and in silico reconstructions of ventricular electrical activity: it is critical to ensure that drug effects not detected in voltage clamp-based ionic current assays and in silico models are detected and evaluated. Isolation and propagation of human-induced pluripotent stem cells and hiPSC-CMs has provided a useful source of cells for applications in drug discovery and cardiotoxicity screening (Mordwinkin et al. 2013).

Voltage clamp studies of hiPSC-CMs have demonstrated the presence of currents expected in adult ventricular myocytes (Ma et al. 2011; Hoekstra et al. 2012) and effects on repolarization consistent with human responses (Hoekstra et al. 2012; Peng et al. 2010). However, studies have shown that hiPSC-CMs demonstrate a relatively immature phenotype compared with adult human myocytes (Jonsson et al. 2012; Qu et al. 2013a). Despite these limitations, numerous studies have demonstrated their ability to detect responses consistent with clinical findings. Thus, while they represent a model system, human ventricular myocytes presently do not fully recapitulate the native adult ventricular myocyte in all functional aspects. Efforts are ongoing in multiple laboratories to provide more representative, or mature, ventricular myocytes. Once successful, fully mature hiPSC-CMs (expressing all the ionic currents with the same densities and characteristics as adult human ventricular myocytes) should replace most in vitro proarrhythmia testing approaches. Furthermore, it should also be possible to test evolving diseased ventricular myocyte models in vitro to evaluate drug effects on at-risk populations (the so-called disease in a dish studies).

A critical assessment of present practices and data yielded from hiPSC-CMs will be necessary in defining the most appropriate experimental methodologies and their limitations. As hiPSC-CMs represent a relatively new and rapidly evolving area of investigation, it is necessary to characterize these preparations more fully and build consensus on their ability to provide consistent data across laboratories and methods. As is the case for all new in vitro preparations, the selection of hiPSC-CMs and the experimental conditions in which they are employed will need to be rigorously defined to facilitate subsequent standardization for use in CiPA.


9.8 Updates from the CiPA Working Groups


Updates from each working group are provided in turn.


9.8.1 The Ion Channel Working Group


The Ion Channel Working Group utilizes the expertise and experience of its members in the fields of ion channel biophysics and pharmacology and in the translation from in vitro to in vivo models. The group is tasked with “bringing together expertise and resources required to deliver best practice recommendations for generating ion channel data needed for in silico human cardiac action potential reconstructions of proarrhythmic liabilities” (Fermini et al. 2016).

Since its launch in January 2014, this working group has focused on addressing several important questions related to best practices, including the following: Which ion channels are necessary to best support in silico action potential modeling? Which characteristics of drug block should be studied (e.g., IC50 determinations, kinetics of block and unblock, rate/use/voltage dependence of block)? What is required to deliver robust, reliable, and reproducible ion channel data in a high-throughput screening environment in support of in silico action potential reconstruction?

As a first step, the group distributed a survey to active members of SPS to collect frequency/type data on the commonly used ion channels in their laboratories to obtain qualitative information on their relevance to drug-induced cardiac safety concerns, with a specific focus on proarrhythmia. The survey was critical in identifying seven ionic currents that are routinely studied because of perceived safety concerns: I Kr, I Ks, I to, I K1, I Ca, I Na[fast], and I Na[late]: these currents were described in Table 9.1. Consequently, protocols are being developed for each of these ionic currents, with the intent of gathering key data to be used for in silico action potential reconstructions.

The channels used for cardiac ionic current studies are typically human clones overexpressed in heterologous expression systems, typically human embryonic kidney (HEK) or Chinese hamster ovary (CHO) cells. While these models are useful, it is recognized that comparisons of the characteristics and drug sensitivities of these currents with currents expressed in native adult ventricular myocytes are lacking. This lack, however, is not considered a significant impediment. Therefore, experiments evaluating drug effects on ionic currents will focus on the following candidate human recombinant channels:



  • Nav1.5 (through which the rapid I Na current flows)


  • Toxin-modified (ATX II) Nav1.5 (I Na[late])


  • Cav1.2 (I CaL)


  • Kv4.3 + KChIP2 (I to)


  • hERG (I Kr)


  • KCNQ1 + KCNE1 (I Ks)


  • Kir2.1 (I K1)

Emphasis is first being placed on I Kr, consistent with its prominent role in defining ventricular repolarization and the ability of many drugs to block this current. Currents will be studied individually using protocols that will assess potencies of block and kinetics that might prove critical in understanding a compound’s potential proarrhythmic liability. Ultimately, only the most informative protocols will be retained (and then standardized) for the CiPA paradigm. It is therefore likely that the list of seven targeted channels just provided will narrow once their roles as proarrhythmic markers are, or are not, confirmed. It is also expected that this work will facilitate the establishment of best practices for the employment of automated patch systems in studies characterizing drug effects (Fermini et al. 2016).


9.8.2 The In Silico Working Group


The In Silico Working Group is responsible for “the development and validation of the best in silico model of human ventricular electrophysiology for the action potential reconstruction of drug effects on the individual ion channel, as determined by the work of the Ion Channel Working Group” (Fermini et al. 2016). It will do so by integrating results from that group’s work on drug effects on individual ion currents. In silico modeling offers the potential “to provide integrative, cost-effective, and high-throughput solutions to predict drug-induced changes in action potential duration” (Fermini et al. 2016). While the O’Hara–Rudy model of the human ventricle was selected by leading in silico modelers in July 2013, further details of drug effects on current kinetics are being incorporated into the model. This model offers several advantages:
Jun 25, 2017 | Posted by in CARDIOLOGY | Comments Off on The Comprehensive In Vitro Proarrhythmia Assay Initiative

Full access? Get Clinical Tree

Get Clinical Tree app for offline access