Abstract
Better drugs are needed for common epilepsies. Drug repurposing offers the potential of significant savings in the time and cost of developing new treatments. In order to select the best candidate drug(s) to repurpose for a disease, it is desirable to predict the relative clinical efficacy that drugs will have against the disease. Common epilepsy can be divided into different types and syndromes. Different antiseizure medications are most effective for different types and syndromes of common epilepsy. For predictions of antiepileptic efficacy to be clinically translatable, it is essential that the predictions are specific to each form of common epilepsy, and reflect the patterns of drug efficacy observed in clinical studies and practice. These requirements are not fulfilled by previously published drug predictions for epilepsy. We developed a novel method for predicting the relative efficacy of drugs against any common epilepsy, by using its Genome-Wide Association Study summary statistics and drugs’ activity data. The methodological advancement in our technique is that the drug predictions for a disease are based upon drugs’ effects on the function and abundance of proteins, and the magnitude and direction of those effects, relative to the importance, degree and direction of the proteins’ dysregulation in the disease. We used this method to predict the relative efficacy of all drugs, licensed for any condition, against each of the major types and syndromes of common epilepsy. Our predictions are concordant with findings from real-world experience and randomized clinical trials. Our method predicts the efficacy of existing antiseizure medications against common epilepsies; in this prediction, our method outperforms the best alternative existing method: area under receiver operating characteristic curve (mean ± standard deviation) 0.83 ± 0.03 and 0.63 ± 0.04, respectively. Importantly, our method predicts which antiseizure medications are amongst the more efficacious in clinical practice, and which antiseizure medications are amongst the less efficacious in clinical practice, for each of the main syndromes of common epilepsy, and it predicts the distinct order of efficacy of individual antiseizure medications in clinical trials of different common epilepsies. We identify promising candidate drugs for each of the major syndromes of common epilepsy. We screen five promising predicted drugs in an animal model: each exerts a significant dose-dependent effect upon seizures. Our predictions are a novel resource for selecting suitable candidate drugs that could potentially be repurposed for each of the major syndromes of common epilepsy. Our method is potentially generalizable to other complex diseases.
Keywords: epilepsy, drug repurposing, GWAS, genomics
Mirza et al. develop and use a novel genomic method to predict the relative efficacy of drugs for each of the major types and syndromes of common epilepsy. This dataset is a novel and valuable resource for selecting the best candidate drug(s) to repurpose for these epilepsies.
Graphical Abstract
Introduction
A total of 50 million people are affected by epilepsy.1 Current drug treatments for epilepsy fail to control seizures in ∼30% of patients2,3 and cause adverse effects in ∼88% of patients4,5; ∼20% of people with newly diagnosed epilepsy discontinue their first antiseizure medication (ASM) because of intolerable adverse effects.6 Hence, there is a need for new ASMs with higher efficacy and/or lower toxicity. Drug repurposing—treating a disease using drugs already licensed for other conditions—offers the potential of significant savings in the time and cost of developing new therapies. Numerous drugs licensed for other conditions have the potential of antiepileptic efficacy.7 In order to select the best candidate drug(s) to repurpose for epilepsy, it is desirable to predict the relative clinical efficacy that drugs will have in people with epilepsy. One established strategy for discovering potentially effective drugs is to, first, identify the proteins that underlie a disease and, then, identify the drugs that affect the disease-proteins. In such analyses, genes associated with a disease are routinely used as proxies for disease-proteins.8
Genetic factors can contribute to the development of epilepsies, either as single-gene mutations in rare monogenic epilepsies, or as multiple genetic variants in common epilepsies.9 Common epilepsies are complex traits with a polygenic origin, which means that the combined effect of many common risk variants contributes to their genetic risk.9 Common epilepsies are divided into different types and syndromes10; for brevity, we use ‘forms’ as a general term for both types and syndromes. Different forms of common epilepsy have important differences in their genetic determinants,11 clinical manifestations and response to medications.12 Hence, to be most useful for common epilepsies, methods of drug prediction must use the specific genes/proteins underlying a particular form of common epilepsy, to make drug predictions that are specific for that particular form of common epilepsy. This has not been achieved by any of the published drug prediction studies for epilepsy.11,13–17 Some studies have pooled genes/proteins associated with different forms of epilepsy (including rare epilepsies), to produce a single list of drug predictions for all forms of epilepsy15–17; these methods are not readily adaptable to individual common epilepsies, as they require a large number of genes/proteins definitively associated with a disease. Other studies have used genome-wide transcriptomic analysis of human brain tissue from epilepsy surgery14,15; such tissue is only available for a very limited number of epilepsy syndromes, and its analysis is hindered by the lack of suitable control brain tissue that is comparable, normal and has been exposed to ASMs. Of course, any transcriptomic changes detected in epileptic brain tissue could be a consequence, rather than a cause, of disease.
The Genome-Wide Association Study (GWAS) is becoming an increasingly powerful tool for revealing the distinct genetic determinants of different common epilepsies.11,18–20 GWAS results are routinely used to predict new candidate drugs for complex diseases. In the standard approach, significant variants from the GWAS are mapped to genes; drugs that are known to affect the (protein products) of the genes, are predicted to affect the disease.21 This simplistic approach has a number of methodological deficiencies. It reflects neither the polygenicity of common diseases, nor the polypharmacology of common drugs. It ignores drugs’ effects on disease-protein abundance, even though, in order to exert their therapeutic effect, drugs rectify the activity of disease-proteins by modulating their function or abundance or both.22–24 It disregards the magnitude and direction of change in disease-proteins’ activity, and drugs’ effects upon it. Potential causal variants below the genome-wide disease significance threshold are ignored. Practically, it produces an unordered and unranked pool of drug names, with no indication of the relative predicted efficacy of the compounds, to enable selection of the most promising candidates. Ultimately, it is liable to producing poor results. Some limitations of the standard approach are addressed by recently developed enhanced techniques for using GWAS results to identify effective drugs,25–28 but these newer methods and their drug predictions for common epilepsy still leave room for improvement. None of the existing methods make drug predictions for a disease based upon drugs’ effects on the function and abundance of proteins, and the magnitude and direction of those effects, relative to the importance, degree and direction of the proteins’ dysregulation in the disease. Our aim was to develop such a method, and to use this method to predict the relative efficacy of drugs for each of the major types and syndromes of common epilepsy, and to make our predictions available as a novel resource for selecting suitable candidate drugs that could potentially be repurposed for each of the major types and syndromes of common epilepsy.
Materials and methods
Methods are summarized below; further details can be found in the Supplementary methods.
Overview
Epilepsy types and syndromes
The common epilepsies are divided into different types, which are further subdivided into different syndromes.10 In the current work, we included the main types and syndromes analysed in the most recent epilepsy GWAS11:
All epilepsy, which is comprised of generalized, focal and unclassified epilepsies
The two main types of all epilepsy: generalized epilepsy (GE) and focal epilepsy (FE)
Two GE syndromes: juvenile myoclonic epilepsy (JME) and childhood absence epilepsy (CAE)
A FE syndrome: FE with hippocampal sclerosis (HS)
Method summary
Genetic variants cause disease by modifying the function or abundance (or both) of proteins derived from the variant genes.29 Drugs exert a therapeutic effect by rectifying the abnormal function or abundance (or both) of the proteins underlying a disease.22–24 To predict the relative efficacy of drugs against a disease, we developed (Fig. 1) a novel score for drugs’ relative ability to affect the protein function and abundance changes caused by common genetic variations associated with the disease: the disease-protein function and abundance modulation (FAM) score.
For method development and benchmarking, we used the all epilepsy GWAS. Then, we applied the developed method to the GWAS for specific epilepsy types and syndromes.
It should be noted that, to aid brevity and readability, we use the expressions ‘disease-associated proteins’ and ‘disease-proteins’ as proxies for ‘proteins encoded by genes bearing variations associated with the disease’, and we use the expression ‘protein abundance changes’ as proxy for ‘changes in gene expression’.
The disease-protein FAM score: creation and benchmarking
The steps taken in developing the method for calculating the FAM score are detailed in Supplementary material. Below, we summarize the method (Fig. 1) we developed for calculating the FAM score.
The FAM score is calculated by aggregating its two constituent scores:
The disease-protein function modulation (FM) score
The disease-protein abundance correction (AC) score
FM score
The FM score is based on the following premise: A drug’s ability to affect a disease can be predicted from:
the degree of disease-association of each protein whose function is affected by the drug, and
the strength of the drug’s effect on the function of each of those proteins
The degree of disease-association of proteins is derived from GWAS gene-based P-values. The strength with which drugs affect proteins’ function is derived from drug-target affinity data. Figure 1 presents a conceptual explanation of how the FM score is calculated from these two types of data. A more detailed explanation can be found in the Supplementary material.
AC score
The AC score is based upon the following premise: A drug is more likely to be effective for a disease if it is better able to rectify the protein abundance changes underlying the disease.30 Disease- and drug-induced transcriptomes were compared in order to predict each drug’s relative ability to rectify disease-associated protein abundance changes, as previously described11 and detailed in the Supplementary material. Briefly, the AC score for a drug is calculated as follows: For each disease-associated protein, the algorithm compares the magnitude and direction of change in the protein’s abundance found in the disease, with the magnitude and direction of change in the protein’s abundance caused by the drug. Then, drugs are ranked in accordance with their overall predicted corrective effect on the abundance of all disease-associated proteins. To measure the overall effect, a metric called ‘cosine distance’ is used.31
Aggregating the FM and AC sores to generate the FAM score
The FM and AC scores were converted into their respective z-scores. The FAM score for each is calculated by averaging its FM and AC z-scores (see Supplementary material for details).
Comparing our method with existing alternative advanced methods
We compared our results with the results from two existing and contrasting advanced methods for GWAS-based drug predictions.
Network-based method
An approach employed in a number of studies is to identify the drugs that target genome-wide significant disease-proteins and, in addition, the drugs that target the proteins interacting with genome-wide significant disease-proteins.32–34 We used the GUILDify v2.0 Web Server35 to identify such drugs.
Gene-set analysis method
In this method,36 GWAS gene-based P-values are first converted to z-statistics and, then, a single-sided two-sample t-test is used to determine if the mean z-statistic of the genes that are altered in function by a drug is lower than the mean z-statistic of the genes that are not.
Validation of the FAM score
For in silico validation of the FAM score, we examined the following hypotheses:
The FAM score for all epilepsy specifically prioritizes the drugs that are effective in people with epilepsy: when drugs are ranked by their FAM score for all epilepsy, drugs used to treat epilepsy are ranked higher than drugs used to treat any other human disease
The FAM score predicts which ASMs are more clinically effective, and which ASMs are less clinically effective, for each common epilepsy syndrome studied
The FAM score predicts the observed patterns of relative efficacy of individual clinically-effective ASMs for each common epilepsy syndrome studied
The above hypotheses are further detailed in Results and in Supplementary methods.
To test the above hypotheses, we used the following metrics:
Identification of effective drugs: we used area under receiver operated characteristics curve (AUROC) analysis to determine the accuracy with drugs’ scores discriminate ASMs from all other drugs, or discriminate more from less clinically-effective subsets of ASMs. AUROC was calculated using the package PRROC (version 1.3.1)37 in R (version 3.4.3). In assessing the discrimination of ASMs from all other drugs, there is a marked class imbalance, because a very small fraction of all drugs are ASMs. To correct for this imbalance, we employed the standard technique of random under-sampling, which is commonly used in published studies (see Supplementary material for further details and references). Specifically, AUROC was calculated using the set of ASMs and a randomly selected set of other drugs equal in number to the ASMs. This process was repeated 1000 times, and mean (± standard deviation) AUROC was calculated. When discriminating more from less effective ASMs, class imbalance is not an issue and, hence, random under-sampling was not employed.
Prioritization of effective drugs: amongst all the drug predictions for a phenotype, we determined the average rank of ASMs, or compared the average rank of more clinically-effective and less clinically-effective ASMs. To ease conceptualization and interpretation of results, we converted ranks to percentile ranks. For example, a drug with a percentile rank of 90 is ranked higher/better than 90% of all drugs. Like numerous published studies, we used the median in order to compute the average of ranks, as it is less liable to skewing by outliers (see Supplementary material for further details and references).
Statistical analysis
We determined the statistical significance of drug identification and prioritization results by comparing the results to those from a null distribution generated by performing 106 random permutations of the scores assigned to drugs.
Determining whether the drug predictions are driven by individual highly disease-associated proteins
For each epilepsy, FAM scores were re-calculated after excluding, one at a time, the top 10 most strongly disease-associated proteins (Supplementary Table 3). Drug ranks obtained after excluding a protein were compared with the original drug ranks, using Kendall’s τ. Kendall’s τ is a commonly used measure of rank correlation.38 Kendall’s τ ranges from +1 to –1, where +1 means that two ranked lists are identical to each other, –1 means that they are the exact inverse of each other, and 0 means that there is no relationship between them.
Further details about this analysis can be found in the Supplementary material.
Top candidate drugs
To aid the selection of suitable candidate drugs for experimental validation and clinical evaluation, we demarcated the most promising candidate drugs for each phenotype: the topmost drug predictions with the greatest enrichment of (more) effective ASMs for that phenotype. A manually curated selection of top candidate drugs for different forms of common epilepsy was also produced.
Testing top candidate compounds in an animal model
As we used complex genetic data to make our drug predictions, we used a complex genetic model to test our drug predictions. We used a rodent model with a complex genetic seizure disorder39–42 that manifests as audiogenic generalized seizures: the DBA/2 mouse. We tested the five most highly ranked predictions for GE, after filtering out known ASMs, compounds with existing published evidence in the DBA/2 mouse model, drugs lacking evidence of blood–brain barrier permeability, drugs lacking evidence of safe long-term oral use in humans, compounds insoluble in water or saline and ‘controlled substances’ that require exceptional legal authorization for procurement under the laws of France, where the animal experiments were performed by a contract research organization.
The animal experiment protocol followed the method described by Dürmüller et al.43 The study was conducted in compliance with Animal Health regulations, in particular:
Council Directive No. 2010/63/UE of 22 September 2010 on the protection of animals used for scientific purposes and French decree No. 2013-118 of 1 February 2013 on the protection of animals;
Porsolt facility accreditation for experimentation (E 53 1031, renewed on 19 April 2016);
The recommendations of the Association for Assessment and Accreditation of Laboratory Animal Care of which the accreditation was granted in June 2012 and renewed in 2018.
Porsolt has an in-house ethics programme, which covers animal care and use within the facility.
Additional experimental details about the animal model testing can be found in the Supplementary material.
Code availability
The R code for computing FM and FAM scores is available at https://figshare.com/projects/Using_common_variants_to_find_drugs_for_common_epilepsies/78330. The code is for non-commercial use only.
Data availability
The following datasets are available for download from the project’s data repository page (https://figshare.com/projects/Using_common_variants_to_find_drugs_for_common_epilepsies/78330):
GWAS gene-based and tissue-wide association study (TWAS) datasets used in our analyses.
Ranked list of the top predicted drugs for each phenotype.
Our complete set of predictions, listing each drug and its FAM score, for each phenotype.
Results
The standard method is inadequate for predicting drugs effective against common epilepsies
In the standard method, drugs are predicted to be efficacious if they modulate the function of proteins that are associated with the disease, according to the GWAS, at a genome-wide level of significance.21 For all epilepsy, GE and FE, SCN1A is the only gene that both (i) reaches genome-wide level of disease-significance, and (ii) produces a protein that is known to be altered in function by any existing compound. For CAE, JME and HS, there are no genes that both (i) reach genome-wide level of disease-significance and (ii) produce a protein that is known to be altered in function by any existing compound. Predicting candidate compounds for epilepsy based upon their ability to affect the function of sodium channel protein Type 1 subunit alpha (the protein product of SCN1A) yields a recall (from amongst all ASMs, the fraction predicted to be effective) of 35% and precision (from amongst all drugs predicted to be effective, the fraction that are ASMs) of 32%, which equates to an F-score (harmonic mean of the precision and recall) of 33%. The standard method of drug prediction produces an unordered and unranked set of candidate drugs, with no metrics for the relative predicted efficacy of the candidate compounds. This precludes method evaluation based upon predicted drug rankings and AUROC, and hampers the selection of the most promising candidate drugs for experimental validation. The same set of ASMs is predicted to be effective for the two divergent phenotypes of GE and FE, even though some seizure types in the former are aggravated by the ASMs that are most effective for the latter. Hence, for different common epilepsies, this method either fails to identify the majority of known effective drugs, or identifies no candidate drugs at all, or identifies potentially aggravating drugs. By extension, applying the standard approach to common epilepsies will yield no or few candidates for repurposing, will not prioritize amongst the candidates, will fail to identify any or most of the efficacious compounds and will potentially identify aggravating drugs.
Creating and benchmarking a new method for predicting the relative efficacy of drugs against common epilepsies
To predict the relative efficacy of drugs against common epilepsies, we devised the disease-protein FAM score, which is calculated using the method illustrated in Fig. 1.
For benchmarking, we used the FAM score and alternative existing advanced methods to predict drugs for all epilepsy, and compared the methods’ performance. For the identification and prioritization of ASMs, the FAM score achieved AUROC (mean ± standard deviation) of 0.83 ± 0.03 and average percentile of 94, respectively. In comparison, the best performing alternative method achieved AUROC (mean ± standard deviation) of 0.63 ± 0.04 and average percentile of 77. Results of all comparator alternative approaches are shown in Supplementary Table 1.
Validating the FAM score
Next, we present results of the analyses performed to test the validity of the predictions made using the FAM score.
The FAM score for all epilepsy specifically prioritizes the drugs that are effective in people with epilepsy
When drugs are ranked by their FAM score for all epilepsy, drugs used to treat epilepsy are ranked higher than drugs used to treat any other human disease. The median rank of drugs used to treat epilepsy is at least seven percentiles higher than that of drug-sets used to treat other human diseases. Permutation-based P-value = 1 × 10−4 that ASMs are ranked highest, and so much higher than all other drug-sets used to treat all other human diseases.
The FAM score predicts which ASMs are more clinically effective, and which ASMs are less clinically effective, for each common epilepsy syndrome
Different ASMs are most effective for different syndromes of common epilepsy. Clinical studies and experience show that, for each common epilepsy syndrome, some ASMs can be classified into a more clinically-effective subset and some into a less clinically-effective subset. For each common epilepsy syndrome, the FAM score predicts which ASMs are amongst the more efficacious in clinical practice, and which ASMs are amongst the less efficacious in clinical practice (Table 1). Specifically, for each common epilepsy syndrome, the FAM score (i) distinguishes the more from the less clinically-effective ASMs and (ii) prioritizes the more clinically-effective ASMs higher than the less clinically-effective ASMs (Table 1).
Table 1.
Epi | Identification of AEDs (AUROC) |
Prioritisation of AEDs (average percentile) |
P | |||
---|---|---|---|---|---|---|
More effective AEDs from all drugs (mean ± SD) | Less effective AEDs from all drugs (mean ± SD) | More from less effective AEDs | More effective AEDs | Less effective AEDs | ||
HS | 0.65 ± 0.13 | 0.36 ± 0.18 | 0.87 | 73 | 27 | 8 × 10–3 |
GE | 0.85 ± 0.04 | 0.69 ± 0.09 | 0.71 | 93 | 70 | <1 × 10–6 |
JME | 0.88 ± 0.04 | 0.76 ± 0.08 | 0.72 | 96 | 86 | <1 × 10–6 |
CAE | 0.75 ± 0.05 | 0.45 ± 0.15 | 0.79 | 85 | 48 | 2.9 × 10–5 |
Constituents of the ‘More effective AEDs’ and ‘Less effective AEDs’ drug-sets are specific to each phenotype. ‘Less effective AEDs’ comprise the set of less effective, ineffective or aggravating AEDs for that phenotype. AUROC is calculated using drugs’ FAM scores. AUROC for identifying AEDs from all drugs is computed using the technique of random under-sampling, and presented as mean ± standard deviation (see Supplementary methods). Prioritization is calculated using drugs’ ranks, when all drugs have been ranked from highest to lowest predicted effect on the phenotype. Prioritization result shown is the average (median) rank of AEDs, expressed as a percentile; it is equivalent to the percentage of all drugs ranked below the middle-ranked AED (see Supplementary methods). AUROC, area under the receiver operating characteristics; CAE, childhood absence epilepsy; Epi, epilepsy type or syndrome; GE, generalized epilepsy; HS, focal epilepsy with hippocampal sclerosis; JME, juvenile myoclonic epilepsy; P, permutation-based P-value after Benjamini–Hochberg correction; SD, standard deviation.
In order to predict which ASMs are more clinically-effective and which ASMs are less clinically-effective for a syndrome, the best results are obtained by using the FAM score for that syndrome. To illustrate this, we show that the ASMs that are more effective for CAE are favoured over the ASMs that are less effective for CAE, only when drugs are predicted using the FAM scores for CAE (AUROC: 0.79), and not when drugs are predicted using the FAM scores for all epilepsy, GE, JME, FE or HS (max AUROC: 0.49); permutation-based P-value = 1 × 10−5 that the AUROC values for CAE and other phenotypes are so contrasting.
For FE, current ASMs are not readily classified into more clinically-effective and less clinically-effective subsets. The FE FAM score identifies and prioritizes ASMs: AUROC (mean ± standard deviation) of 0.85 ± 0.03 and average percentile of 94; the FAM score’ performance is statistically significant (permutation-based P-value = 1 × 10−6), and superior to that of its constituent scores.
When considering the ability to distinguish more effective ASMs from all drugs and from less effective ASMs, the FAM score outperforms its constituent scores (Supplementary Table 2).
The FAM score predicts the observed patterns of relative efficacy of individual clinically-effective ASMs
We tested our predictions against the following observed patterns of relative efficacy of individual clinically-effective ASMs.
Valproate is the most effective ASM for GE, whereas carbamazepine is the most effective ASM for FE
It is recognized that the efficacy of valproate for generalized onset seizures is ‘unsurpassed’,44 whist for focal onset seizures, ‘no other drug has been shown to be more effective’ than carbamazepine.45 In our predictions for GE, valproate is ranked highest of all current ASMs. In our predictions for FE, carbamazepine is ranked highest of all current ASMs. Valproate and carbamazepine are amongst the top two of all drugs in our predictions for GE and FE, respectively; permutation-based P-value = 5.6 × 10−6 for both valproate and carbamazepine being ranked so highly in our predictions for GE and FE, respectively.
The predicted order of efficacy of ASMs for FE matches that seen in the SANAD trials
The SANAD studies are the largest published head-to-head comparison of multiple ASMs for FE, and the largest published randomized controlled trial of ASMs for FE.46,47
Five ASMs were compared in the FE arm of SANAD I: carbamazepine, gabapentin, lamotrigine, oxcarbazepine and topiramate. These drugs’ predicted order of efficacy for FE matches the observed order of efficacy in the SANAD I trial. The finding that these drugs are ranked as highly and in the correct order is unlikely to occur by chance (P < 1 × 10−6 by permutation).
Carbamazepine and gabapentin are effective ASMs but, in the FE arm of the SANAD I trial, carbamazepine was significantly more efficacious than gabapentin. Carbamazepine and gabapentin are ranked high in our predictions for FE (percentile ranks 100 and 79, respectively), but carbamazepine is ranked significantly higher than gabapentin (permutation-based P-value = 1 × 10−4 for the ranks of both drugs being as high but as disparate as observed).
The ASMs compared in the FE arm of SANAD II were lamotrigine, levetiracetam and zonisamide. These drugs’ predicted order of efficacy for FE matches the observed order of efficacy in the SANAD II trial. The finding that these drugs are ranked as highly and in the correct order is unlikely to occur by chance (P < 1 × 10−6 by permutation).
The prioritized order of efficacy of ASMs for GE matches that seen in the SANAD I trial
The SANAD studies are the largest published head-to-head comparison of multiple ASMs for GE, and the largest published randomized controlled trial of ASMs for GE.48,49
The ASMs compared in the GE arm of SANAD I were lamotrigine, topiramate and valproate. These drugs’ predicted order of efficacy matches the clinically observed order of efficacy in the SANAD I trial. The finding that these drugs are ranked as highly and in the correct order is unlikely to occur by chance (permutation-based P-value = 1 × 10−5).
Valproate and lamotrigine are effective ASMs but, in the GE arm of the SANAD I trial, valproate was significantly more efficacious than lamotrigine. Valproate and lamotrigine are ranked high in our predictions for GE (percentile ranks 100 and 81, respectively), but valproate is ranked significantly higher than lamotrigine (permutation-based P-value = 3 × 10−4 for the ranks of both drugs being as high but as disparate as observed).
The ASMs compared in the GE arm of SANAD II were levetiracetam and valproate. Valproate and levetiracetam are effective ASMs but, in the GE arm of the SANAD II trial, valproate was significantly more efficacious than levetiracetam. Valproate and levetiracetam are ranked high in our predictions for GE (ranks 1 and 15, respectively), but valproate is ranked significantly higher than levetiracetam (permutation-based P-value < 1 × 10−5 for the ranks of both drugs being as high but as disparate as observed).
Topiramate is more effective than lamotrigine for GE, but lamotrigine is more effective than topiramate for FE, in concordance with the SANAD I trial
Lamotrigine and topiramate are the only two ASMs included in both the FE and GE arms of the SANAD I study. In the GE arm of SANAD I, topiramate was more efficacious than lamotrigine, whereas in the FE arm, lamotrigine was more efficacious then topiramate. In our predictions for FE, lamotrigine is ranked higher than topiramate, while for GE, topiramate is ranked higher than lamotrigine. The contrasting ranks of lamotrigine and topiramate for FE and GE are unlikely to occur by chance (permutation-based P-value = 1 × 10−4).
For JME, valproate is most effective
Valproate is thought to be the most efficacious broad-spectrum ASM for JME50–52 but this is based on anecdotal data and retrospective analyses. Amongst our predictions for JME, valproate was amongst the highest ranked drugs (percentile rank 98), but not the highest. The highest ranked prediction was primidone. In the longest retrospective cohort study of JME to date, primidone was most effective, with a 5-year terminal remission rate of 73.3, compared to 50% with valproate.53
For CAE, valproate and ethosuximide are most effective
Valproate and ethosuximide are most effective for CAE; both are similarly effective for CAE.54 In our predictions for CAE, valproate is ranked highest of all drugs. Ethosuximide is not ranked highly, but higher than average, amongst our predictions (median percentile rank 58). The P-value for the two drugs being ranked so favourably is 5 = 1 × 10−4. Ethosuximide is ascribed a particularly low FM score for CAE, which places it in the 20th percentile of predictions for the phenotype. One possible explanation of ethosuximide’s low FM score is that its mechanism of action is poorly understood, as it is not an extensively studied compound. Indeed, ethosuximide is one of the least studied of the current ASMs: there are 343 MEDLINE articles with the word ethosuximide in their title, compared to a mean of ∼1765 for the other current ASMs that are also found in our datasets (as of 2 September 2021; single-sample one-tailed t-test t = 3.7 and P-value = 6.9 × 10−4).
The drug predictions are not driven by individual highly disease-associated proteins
The relative predicted efficacy of drugs does not change significantly after excluding, one at a time, the top 10 most strongly disease-associated proteins that contribute to the FAM score for that epilepsy. The predicted ranks of drugs for each epilepsy remained significantly stable after excluding, one at a time, the top 10 most strongly disease-associated proteins that contribute to the FAM score for that epilepsy. For each epilepsy, FAM scores were re-calculated after excluding, one at a time, the top 10 most strongly disease-associated proteins (Supplementary Table 3) that contribute to the FAM score for that epilepsy. When drug ranks obtained after excluding a protein were compared with the original drug ranks, Kendall’s τ ranged from 0.80 to 0.93, with all corrected P-values <1 × 10−200. In contrast, comparing the predicted drug rankings for two unrelated epilepsies—CAE and HS—yields a Kendall’s τ of 0.04 (P = 0.10).
Top candidate drugs
Ranked lists of the top drugs predicted to be effective for each phenotype, which are most enriched with the drugs that are known to be (more) effective for the phenotype, are available for download (see Data availability). For each phenotype, the top candidate drugs are significantly (Benjamini–Hochberg P-value <0.05) enriched with the ASMs that are (more) effective for the phenotype, except for HS. For HS, there was no significant enrichment of (more) effective ASMs, which may be a reflection of the often drug-resistant nature of HS, or of the lower power of the HS GWAS, or the relatively smaller size of the more effective subset of ASMs for HS, or a combination of these factors.
A manually curated selection of top candidate drugs that could potentially be repurposed for different forms of common epilepsy is shown in the Table 2.
Table 2.
Epi | Drugs | Evidence of antiseizure efficacy in | Indication | Mode of action |
---|---|---|---|---|
CAE | Clomipramine | Animal models1 and humans2 | Depression | Serotonin–noradrenaline reuptake inhibitor |
CAE | Doxepin | Animal models3,4 | Depression | Tricyclic antidepressant |
CAE | Pentoxifylline | Animal models5 | Peripheral vascular disease | Haemorheological agent, increases leukocyte deformability |
CAE | Phenelzine | Animal models6 | Depression | Monoamine oxidase inhibitor |
CAE | Sulindac | Animal models7 | Pain | Non-steroidal anti-inflammatory |
CAE | Tolbutamide | Animal models8 | Diabetes mellitus | Sulphonylurea |
CAE | Tranylcypromine | Animal models9 | Depression | Monoamine oxidase inhibitor |
FE | Chlorzoxazone | Rat hippocampal neurons10 | Muscle spasms | Calcium and potassium channel inhibitor |
FE | Hydrochlorothiazide | Animal models11, 12 and human12 | Hypertension | ACEII antagonist |
FE | Thalidomide | Animal models16-18 | Multiple myeloma | Immunomodulation, unspecified |
FE | Zaleplon | Animal models19 | Insomnia | GABA-BZ agonist |
FE | Zolpidem | Animal models20-22 | Insomnia | GABA-BZ/GABA-A agonist |
HS | Amiodarone | Animal models23 | Arrhythmia | Potassium channel blocker |
HS | Clonidine | Animal models24-44 | Hypertension | Alpha-2 adrenoceptor agonist |
HS | Methoxamine | Animal models45 | Hypotension | Alpha-1 adrenergic receptor agonist |
HS | Pergolide | Animal models46 | Parkinson’s disease | D2 agonist |
HS | Thioridazine | Animal models47 | Psychosis | D1/D2 antagonist |
HS | Tizanidine | Animal models40 | Muscle spasticity | Alpha-2 adrenergic receptor antagonist |
JME | Aliskiren | Animal48, 49 | Hypertension | Renin inhibitor |
JME | Baclofen | Animal models39, 50-76 | Muscle spasticity | GABA-B receptor agonist |
JME | Diazoxide | Animal models77, 78 | Hypoglycaemia | Potassium channel agonist, inhibits insulin release |
JME | Icosapent | Animals79, 80 and humans81-85 | Hypertriglyceridaemia | 20-carbon omega-3 fatty acid |
JME | Iloprost | Animal models86, 87 | Pulmonary arterial hypertension | Synthetic analogue of prostacyclin PGI2 |
JME | Nicotinamide | Animal models94-103 | Pellagra | Water-soluble form of Vitamin B3 |
JME | Pranlukast | Animal models104 and humans105 | Asthma | Cysteinyl leukotriene receptor-1 antagonist |
JME | Riluzole | Animal models106-109 | Amyotrophic lateral sclerosis | Glutamate antagonist |
Candidate drugs for GE, which we tested in an animal model, are listed in Table 3. References, for the evidence cited here, can be found in the Supplementary material. CAE, childhood absence epilepsy; Epi, epilepsy type or syndrome; HS, focal epilepsy with hippocampal sclerosis; JME, juvenile myoclonic epilepsy.
Predicted drugs have a significant dose-dependent effect on seizures in an animal model
After excluding drugs that are toxic or otherwise unsuitable, the top five predicted drugs for GE were tested in a mouse model with a complex genetic seizure disorder that manifests as audiogenic generalized seizures. Each of the drugs had a significant dose-dependent effect on tonic and clonic convulsions (Table 3). Whilst four of the drugs had a significant dose-dependent anti-convulsant effect, one of the compounds (betahistine) had a significant dose-dependent pro-convulsant effect.
Table 3.
Drug | Latency (s) to convulsions (mean±s.e.m) | P |
---|---|---|
Vehicle (i.p.) | 10.9 ± 2.6 | – |
Orphenadrine (12.5 mg/kg i.p.) | 40.0 ± 5.6 | 6.10 × 10–5 |
Orphenadrine (25 mg/kg i.p.) | 53.4 ± 3.7 | 5.40 × 10–7 |
Orphenadrine (50 mg/kg i.p.) | 60.0 ± 0.0 | 4.14 × 10–7 |
Dyclonine (5 mg/kg i.p.) | 31.5 ± 6.2 | 1.77 × 10–2 |
Dyclonine (10 mg/kg i.p.) | 44.7 ± 5.4 | 2.16 × 10–4 |
Dyclonine (20 mg/kg i.p.) | 57.7 ± 2.4 | 4.14 × 10–7 |
Trimeprazine (2.5 mg/kg i.p.) | 11.0 ± 20.6 | 6.52 × 10–1 |
Trimeprazine (5 mg/kg i.p.) | 18.1 ± 4.1 | 1.77 × 10–2 |
Trimeprazine (10 mg/kg i.p.) | 44.5 ± 5.3 | 4.06 × 10–6 |
Acamprosate (125 mg/kg i.p.) | 8.7 ± 0.4 | 6.40 × 10–1 |
Acamprosate (250 mg/kg i.p.) | 9.2 ± 0.2 | 4.56 × 10–1 |
Acamprosate (500 mg/kg i.p.) | 14.3 ± 2.5 | 1.20 × 10–2 |
Betahistine (75 mg/kg i.p.) | 9.1 ± 0.5 | 4.53 × 10–1 |
Betahistine (150 mg/kg i.p.) | 6.9 ± 0.4 | 2.83 × 10–2 |
Betahistine (300 mg/kg i.p.) | 5.3 ± 0.3 | 4.48 × 10–5 |
Valproate (180 mg/kg i.p.) | 57.7 ± 1.4 | 4.89 × 10–7 |
After activation of a bell, latency to the occurrence of tonic convulsions and clonic convulsions was measured. P, Benjamini–Hochberg-corrected P-value from two-sided Mann–Whitney U test; s.e.m, standard error of the mean.
Discussion
We present the relative predicted efficacy of drugs against each of the main types and syndromes of common epilepsy. This dataset is a novel and valuable resource for selecting the best candidate drug(s) to repurpose for any of the main types and syndromes of common epilepsy. Of course, our predicted candidate drugs require further animal model and/or human clinical trial evidence before being considered for deployment in clinical practice.
To generate our predictions, we created a novel method. Our method possesses several strengths that are lacking in previously published approaches. Common epilepsies, like other complex diseases, develop when many different proteins display abnormal activity due to pathological changes in their abundance or function.55 Our method prioritizes drugs according to their relative ability to modulate changes in both the abundance and the function of disease-proteins. Furthermore, drugs are prioritized on the basis of their ability to correct disease-protein abnormalities that are found in people with the disease, rather than in animal models, and that are not consequential to or compensatory for the disease, as they are driven by germline variations. We use genetic variation data specific to each form of common epilepsy, to make drug predictions specific to that form of common epilepsy. The ASMs that are more clinically-effective for a syndrome and the ASMs that are less clinically-effective for a syndrome are predicted more effective and less effective, respectively, for that syndrome only, but not for any other epilepsy type or syndrome—this suggests that our predictions are not systemically biased in favour of a particular set or type of drugs. The methodology is based upon a polygenic model of disease and a multi-targeted approach to treatment, which are desirable for complex diseases. We utilize conventional canonical low-throughput single-target functional drug activity data, and high-throughput genome-wide transcriptomic drug activity data, so that prioritization of drugs is informed by their on-target and off-target effects, and by their affinities for individual proteins and effects upon genome-wide gene expression. The directionality of drugs’ effects on protein activity also helps inform drug prioritization. Rather than dichotomous categorization of compounds into drugs that are predicted to be effective or ineffective, our method ranks drugs individually according to relative predicted efficacy, which aids candidate selection for in vivo validation and for development.
Our method produces accurate drug predictions for epilepsy syndromes even if their GWAS results include few genome-wide significant loci. Even excluding the most strongly disease-associated proteins does not significantly change the relative efficacy of drugs predicted by our method (as we show in the Results, under subheading ‘The drug predictions are not driven by individual highly disease-associated proteins’). This is because our method is not reliant on individual highly disease-associated proteins. Instead, our method leverages the gene-set analysis approach, where each gene-set is the set of genes affected by each drug. The disease association of all the genes in a gene-set, even those below the genome-wide significant threshold, is combined; the gene-sets that are more disease-associated overall are more biologically relevant. The gene-set approach is a long-established and widely-used method in all areas of genomic analysis,56 including post-GWAS analysis generally57 and GWAS-based drug repurposing analysis specifically.27,28 Utilizing the full distribution of all genetic associations for gene-set analysis is a validated, established and accepted approach, which has been implemented in numerous widely-used post-GWAS analysis tools, for example FUMA,58 MAGMA,58 MAGENTA,59 INRICH60 and DEPICT,61 each of which has been employed in a multitude of published GWAS-based studies.
Alongside these strengths, our method has some limitations, discussed below.
Our drug prediction method, like all previously published genetics- or genomics-based drug prediction methods, predicts the efficacy of drugs for a disease. However, the most efficacious drug for a disease in not always the most appropriate drug for every individual with the disease. Important factors to consider when choosing a drug for an individual include the potential of undesirable interactions with other medications and the possible side-effects. Our method, like all previously published genetics- or genomics-based drug prediction methods, does not predict drugs’ interactions with other medications and side-effects. Indeed, the success of an ASM is determined as much by its tolerability as by its efficacy.59–62 As the drugs we have predicted for epilepsies are already being used for other diseases, their side-effect profiles are known. This allows researchers to select for further development those candidate compounds whose side-effects are less deleterious or even desirable.
Our method predicts drugs effective for a disease from the proteins underlying the disease, after identifying the proteins underlying the disease from the common genetic variations associated with the disease. However, some proteins become dysfunctional or dysregulated not because of common genetic variations, but because of rare genetic variations, or copy number variations, or abnormalities of epigenetic, post-transcriptional or post-translational mechanisms, or because of environmental insults. Such protein changes do not inform our predictions, which could affect their accuracy, commensurate with the contribution of those proteins to the causal mechanism underlying an epilepsy and/or to the mechanism of action of a drug. We are not aware of any existing drug prediction methods which take into account the multiple potential pathogenic factors that influence proteins; the development of such methods might lead to improved accuracy of drug predictions.
Our analysis uses data from a GWAS that employed imputation to improve genomic coverage. The GWAS gene-level data used in this analysis offers coverage of genes across the genome, and it is corrected for the lengths and single nucleotide polymorphism-densities of genes. However, if a gene is not (adequately) covered by the genotyping array and the imputation, but the gene is of importance in epilepsy and affected by drug(s), the accuracy of our drug predictions could be adversely affected. Hence, improved coverage of future epilepsy GWAS analyses could help to improve the accuracy of drug predictions.
Our drug predictions are based upon two scores: the FM and AC scores. The FM score relies upon knowledge of the proteins changed in function by drugs. At present, knowledge of the proteins that are changed in function by each drug is incomplete, and it is more incomplete for some drugs than for others. The more incomplete the knowledge of the proteins changed in function by a drug, the more likely it is that the drug’s FM score will be underestimated. By extension, the FM score is more likely to be underestimated for drugs that are less studied, as their modes of action are less analysed and, hence, knowledge of the proteins changed in function by them is less complete. This may explain the relatively low FM sore and, hence, FAM score and ranking for ethosuximide. The AC score is free of this limitation, as the AC score is based upon profiles of drug-induced transcriptomic changes assayed by using the same standardized pipeline for each drug. With over 44 000 compounds already analysed on this platform (http://lincsportal.ccs.miami.edu/SmallMolecules/catalog; accessed on 1 February 2021), transcriptomic profiles are available for the vast majority of drugs of interest. However, a small number of interesting drugs (for example, brivaracetam and cenobamate) have not been assayed, which means that an AC score and, hence, a FAM score cannot be calculated for them. The platform and pipeline used for generating drugs’ transcriptomic profiles are in the public domain, and have been used by researchers to generate profiles for any compounds of interest not already found in the database, albeit for industrial-scale projects.62 In addition, there is active ongoing development of computational methods for using knowledge of drugs’ structures to predict the proteins that they change in function and/or abundance,63,64 which is another potential future strategy for predicting the relative efficacy of compounds whose molecular effects are still unknown.
It is noted that the FM score does not predict the ‘directionality’ of drugs’ effects (that is, beneficial or harmful) on disease-protein function. Therefore, drugs predicted by the FM score to affect a phenotype may be alleviating or aggravating for the phenotype. This is a recognized limitation of methods that use data for the ability of drugs to alter the function of genetically-associated disease-proteins in order to predict drugs that can affect the disease,16,17,65 as the direction of change in protein activity occurring in the disease is unknown. On the other hand, the AC score does predict the ‘directionality’ of drugs’ effects (that is, beneficial or harmful) on individual disease-proteins and, thereby, the overall ‘directionality’ of drugs’ effects (that is, beneficial or harmful) on the disease. The AC score takes into account the magnitude and direction of change in proteins’ abundance underlying disease, and the magnitude and direction of change in proteins’ abundance caused by drugs. Thereby, the AC score proposes to predict the drugs with a beneficial effect on disease-protein abundance and clinical phenotype. Hence, inclusion of the AC score, with the FM score, in our final FAM score, is expected to help mitigate the risk of deleterious compounds with high FM scores being included in our candidate drugs. Still, it is possible that some aggravating drugs are included in our candidate compounds. Hence, experimental validation of candidate drugs is essential before clinical use, as with all in silico drug prediction methods. We tested five of our candidate compounds in a rodent model: all five compounds had a significant dose-dependent effect on seizures. Interestingly, one of the candidate compounds (betahistine) had a significant dose-dependent pro-convulsant effect in the animal model. This finding could be explained by the possibility that some of our predicted compounds are aggravating, as discussed. However, it is also possible that the pro-convulsant effect of betahistine in our study is a reflection of species- or model-specific behaviour. Indeed, a recent study (published after our animal experiments had ended) showed that betahistine has a significant antiepileptogenic and anticonvulsant effect on pentylenetetrazole-induced generalized seizures in a different mouse strain.66
Whilst acknowledging these limitations and some aberrant predictions, we note that our method outperforms alternative methods for predicting drugs that have efficacy against common epilepsies in clinical studies and experience. Our method also predicts which ASMs are amongst the more efficacious in clinical practice, and which ASMs are amongst the less efficacious in clinical practice, for each of the main syndromes of common epilepsy, and it predicts the distinct order of efficacy of individual ASMs in clinical trials of different common epilepsies. This aspect is key to the clinical translation of drug predictions for common epilepsies, but is missing from previously published studies that have predicted drugs for epilepsy.13–17
In this study, we have used the tissue-wide association study method to identify the protein abundance changes underlying disease. A closely-related alternative method is to use Mendelian randomization. In future studies, both methods could be compared and/or combined in order to determine if this improves the drug predictions. Mendelian randomization is discussed at greater length in the Supplementary material.
As our method uses GWAS data, it cannot be applied to monogenic diseases. It is conceivable that this method could be adapted to make it applicable to monogenic diseases, and we plan to explore this possibility in a future study dedicated to this objective.
We have used results from the latest epilepsy GWAS mega-analysis, which includes previously published and unpublished epilepsy GWAS analyses, making it the largest epilepsy GWAS to date.11 However, compared to other common neurological diseases, even the largest epilepsy GWAS had a modest sample size, with 15 212 cases and 29 677 controls, and produced a modest number of discoveries, with 16 loci identified. The latest schizophrenia GWAS, for example, included 36 989 cases and 113 075 controls, resulting in the identification of 108 risk loci.67 It is hoped that expanded cohort sizes of future epilepsy GWAS analyses will increase power and improve drug predictions. In this analysis, we predicted drugs for the main epilepsy syndromes that had risk loci identified in the latest epilepsy GWAS. It is hoped that future epilepsy GWAS will be large enough to report results for additional epilepsy syndromes, and drugs can be predicted for them using the method presented here. Finally, it is likely that our method can be applied to the GWAS results of other common complex phenotypes.
Supplementary material
Supplementary material is available at Brain Communications online.
Supplementary Material
Acknowledgements
Some of the data reported in this paper were collected as part of a project undertaken by the International League against Epilepsy (ILAE) and some of the authors are experts selected by the ILAE. Opinions expressed by the authors, however, do not necessarily represent the policy or position of the ILAE.
Funding
This work was supported by a Medical Research Council Confidence in Concept Award to N.M. B.P.C.K. is supported by funds from the Friends of Wilhemina KinderZiekenhuis MING funds.
Competing interests
The authors report no competing interests.
Glossary
- AC score =
abundance correction score
- ASM =
antiseizure medication
- CAE =
childhood absence epilepsy
- FAM score =
function and abundance modulation score
- FE =
focal epilepsy
- FM score =
function modulation score
- GE =
generalized epilepsy
- GWAS =
Genome-Wide Association Study
- HS =
hippocampal sclerosis
- ILAE =
International League against Epilepsy
- JME =
juvenile myoclonic epilepsy
- ROC =
area under receiver operated characteristics curve
Appendix I
The International League Against Epilepsy Consortium on Complex Epilepsies author names are shown below. Members are listed in alphabetical order. Author affiliations can be found in the Supplementary material.
Bassel Abou-Khalil, Pauls Auce, Andreja Avbersek, Melanie Bahlo, David J. Balding, Thomas Bast, Larry Baum, Albert J. Becker, Felicitas Becker Bianca Berghuis, Samuel F. Berkovic, Katja E. Boysen, Jonathan P. Bradfield, Lawrence C. Brody, Russell J. Buono, Ellen Campbell, Gregory D. Cascino, Claudia B. Catarino, Gianpiero L. Cavalleri, Stacey S. Cherny, Krishna Chinthapalli, Alison J. Coffey, Alastair Compston, Antonietta Coppola, Patrick Cossette, John J. Craig, Gerrit-Jan de Haan, Peter De Jonghe, Carolien G. F. de Kovel, Norman Delanty, Chantal Depondt, Orrin Devinsky, Dennis J. Dlugos, Colin P. Doherty, Christian E. Elger, Johan G. Eriksson, Thomas N. Ferraro, Martha Feucht, Ben Francis, Andre Franke, Jacqueline A. French, Saskia Freytag, Verena Gaus, Eric B. Geller, Christian Gieger, Tracy Glauser, Simon Glynn, David B. Goldstein, Hongsheng Gui, Youling Guo, Kevin F. Haas, Hakon Hakonarson, Kerstin Hallmann, Sheryl Haut, Erin L. Heinzen, Ingo Helbig, Christian Hengsbach, Helle Hjalgrim, Michele Iacomino, Andrés Ingason, Jennifer Jamnadas-Khoda, Michael R. Johnson, Reetta Kälviäinen, Anne-Mari Kantanen, Dalia Kasperavičiūte, Dorothee Kasteleijn-Nolst Trenite, Heidi E. Kirsch, Robert C. Knowlton, Bobby P. C. Koeleman, Roland Krause, Martin Krenn, Wolfram S. Kunz, Ruben Kuzniecky, Patrick Kwan, Dennis Lal, Yu-Lung Lau, Holger Lerche, Costin Leu, Wolfgang Lieb, Dick Lindhout, Warren D. Lo, Iscia Lopes-Cendes, Daniel H. Lowenstein, Alberto Malovini, Anthony G. Marson, Thomas Mayer, Mark McCormack, James L. Mills, Nasir Mirza, Martina Moerzinger, Rikke S. Møller, Anne M. Molloy, Hiltrud Muhle, Mark Newton, Ping-Wing Ng, Markus M. Nöthen, Peter Nürnberg, Terence J. O’Brien, Karen L. Oliver, Aarno Palotie, Faith Pangilinan, Sarah Peter, Slavé Petrovski, Annapurna Poduri, Michael Privitera, Rodney Radtke, Sarah Rau, Philipp S. Reif, Eva M. Reinthaler, Felix Rosenow, Josemir W. Sander, Thomas Sander, Theresa Scattergood, Steven C. Schachter, Christoph J. Schankin, Ingrid E. Scheffer, Bettina Schmitz, Susanne Schoch, Pak C. Sham, Jerry J. Shih, Graeme J. Sills, Sanjay M. Sisodiya, Lisa Slattery, Alexander Smith, David F. Smith, Michael C. Smith, Philip E. Smith, Anja C. M. Sonsma, Doug Speed, Michael R. Sperling, Bernhard J. Steinhoff, Ulrich Stephani, Remi Stevelink, Konstantin Strauch, Pasquale Striano, Hans Stroink, Rainer Surges, K. Meng Tan, Liu Lin Thio, G. Neil Thomas, Marian Todaro, Rossana Tozzi, Maria S. Vari, Eileen P. G. Vining, Frank Visscher, Sarah von Spiczak, Nicole M. Walley, Yvonne G. Weber, Zhi Wei, Judith Weisenberg, Christopher D. Whelan, Peter Widdess-Walsh, Markus Wolff, Stefan Wolking, Wanling Yang, Federico Zara and Fritz Zimprich.
Contributor Information
International League Against Epilepsy Consortium on Complex Epilepsies:
Bassel Abou-Khalil, Pauls Auce, Andreja Avbersek, Melanie Bahlo, David J Balding, Thomas Bast, Larry Baum, Albert J Becker, Felicitas Becker Bianca Berghuis, Samuel F Berkovic, Katja E Boysen, Jonathan P Bradfield, Lawrence C Brody, Russell J Buono, Ellen Campbell, Gregory D Cascino, Claudia B Catarino, Gianpiero L Cavalleri, Stacey S Cherny, Krishna Chinthapalli, Alison J Coffey, Alastair Compston, Antonietta Coppola, Patrick Cossette, John J Craig, Gerrit-Jan de Haan, Peter De Jonghe, Carolien G. F de Kovel, Norman Delanty, Chantal Depondt, Orrin Devinsky, Dennis J Dlugos, Colin P Doherty, Christian E Elger, Johan G Eriksson, Thomas N Ferraro, Martha Feucht, Ben Francis, Andre Franke, Jacqueline A French, Saskia Freytag, Verena Gaus, Eric B Geller, Christian Gieger, Tracy Glauser, Simon Glynn, David B Goldstein, Hongsheng Gui, Youling Guo, Kevin F Haas, Hakon Hakonarson, Kerstin Hallmann, Sheryl Haut, Erin L Heinzen, Ingo Helbig, Christian Hengsbach, Helle Hjalgrim, Michele Iacomino, Andrés Ingason, Jennifer Jamnadas-Khoda, Michael R Johnson, Reetta Kälviäinen, Anne-Mari Kantanen, Dalia Kasperavičiūte, Dorothee Kasteleijn-Nolst Trenite, Heidi E Kirsch, Robert C Knowlton, Bobby P. C Koeleman, Roland Krause, Martin Krenn, Wolfram S Kunz, Ruben Kuzniecky, Patrick Kwan, Dennis Lal, Yu-Lung Lau, Holger Lerche, Costin Leu, Wolfgang Lieb, Dick Lindhout, Warren D Lo, Iscia Lopes-Cendes, Daniel H Lowenstein, Alberto Malovini, Anthony G Marson, Thomas Mayer, Mark McCormack, James L Mills, Nasir Mirza, Martina Moerzinger, Rikke S Møller, Anne M Molloy, Hiltrud Muhle, Mark Newton, Ping-Wing Ng, Markus M Nöthen, Peter Nürnberg, Terence J O’Brien, Karen L Oliver, Aarno Palotie, Faith Pangilinan, Sarah Peter, Slavé Petrovski, Annapurna Poduri, Michael Privitera, Rodney Radtke, Sarah Rau, Philipp S Reif, Eva M Reinthaler, Felix Rosenow, Josemir W Sander, Thomas Sander, Theresa Scattergood, Steven C Schachter, Christoph J Schankin, Ingrid E Scheffer, Bettina Schmitz, Susanne Schoch, Pak C Sham, Jerry J Shih, Graeme J Sills, Sanjay M Sisodiya, Lisa Slattery, Alexander Smith, David F Smith, Michael C Smith, Philip E Smith, Anja C. M Sonsma, Doug Speed, Michael R Sperling, Bernhard J Steinhoff, Ulrich Stephani, Remi Stevelink, Konstantin Strauch, Pasquale Striano, Hans Stroink, Rainer Surges, K. Meng Tan, Liu Lin Thio, G. Neil Thomas, Marian Todaro, Rossana Tozzi, Maria S Vari, Eileen P. G Vining, Frank Visscher, Sarah von Spiczak, Nicole M Walley, Yvonne G Weber, Zhi Wei, Judith Weisenberg, Christopher D Whelan, Peter Widdess-Walsh, Markus Wolff, Stefan Wolking, Wanling Yang, Federico Zara, and Fritz Zimprich
References
- 1. GBD 2016 Epilepsy Collaborators. Global, regional, and national burden of epilepsy, 1990-2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(4):357–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Shorvon SD. The epidemiology and treatment of chronic and refractory epilepsy. Epilepsia. 1996;37 Suppl 2:S1–S3. [DOI] [PubMed] [Google Scholar]
- 3. Chen Z, Brodie MJ, Liew D, Kwan P.. Treatment outcomes in patients with newly diagnosed epilepsy treated with established and new antiepileptic drugs: A 30-year longitudinal cohort study. JAMA Neurol. 2018;75(3):279–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Baker GA, Jacoby A, Buck D, Stalgis C, Monnet D.. Quality of life of people with epilepsy: A European study. Epilepsia. 1997;38(3):353–362. [DOI] [PubMed] [Google Scholar]
- 5. Perucca P, Carter J, Vahle V, Gilliam FG.. Adverse antiepileptic drug effects: Toward a clinically and neurobiologically relevant taxonomy. Neurology. 2009;72(14):1223–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Alsfouk BAA, Brodie MJ, Walters M, Kwan P, Chen Z.. Tolerability of antiseizure medications in individuals with newly diagnosed epilepsy. JAMA Neurol. 2020;77(5):574–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Sivapalarajah S, Krishnakumar M, Bickerstaffe H, et al. The prescribable drugs with efficacy in experimental epilepsies (PDE3) database for drug repurposing research in epilepsy. Epilepsia. 2018;59(2):492–501. [DOI] [PubMed] [Google Scholar]
- 8. Kamb A, Harper S, Stefansson K.. Human genetics as a foundation for innovative drug development. Nat Biotechnol. 2013;31(11):975–978. [DOI] [PubMed] [Google Scholar]
- 9. Koeleman BPC. What do genetic studies tell us about the heritable basis of common epilepsy? Polygenic or complex epilepsy? Neurosci Lett. 2018;667:10–16. [DOI] [PubMed] [Google Scholar]
- 10. Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification of the epilepsies: Position paper of the ILAE Commission for Classification and Terminology. Epilepsia. 2017;58(4):512–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. International League Against Epilepsy Consortium on Complex Epilepsies. Genome-wide mega-analysis identifies 16 loci and highlights diverse biological mechanisms in the common epilepsies. Nat Commun. 2018;9(1):5269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Bourgeois BF. Chronic management of seizures in the syndromes of idiopathic generalized epilepsy. Epilepsia. 2003;44 Suppl 2:27–32. [DOI] [PubMed] [Google Scholar]
- 13. Mirza N, Sills GJ, Pirmohamed M, Marson AG.. Identifying new antiepileptic drugs through genomics-based drug repurposing. Hum Mol Genet. 2017;26(3):ddw410. [DOI] [PubMed] [Google Scholar]
- 14. Brueggeman L, Sturgeon ML, Martin RM, et al. Drug repositioning in epilepsy reveals novel antiseizure candidates. Ann Clin Transl Neurol. 2019;6(2):295–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Delahaye-Duriez A, Srivastava P, Shkura K, et al. Rare and common epilepsies converge on a shared gene regulatory network providing opportunities for novel antiepileptic drug discovery. Genome Biol. 2016;17(1):245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Himmelstein DS, Lizee A, Hessler C, et al. Systematic integration of biomedical knowledge prioritizes drugs for repurposing. Elife. 2017;6:e26726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Guney E, Menche J, Vidal M, Barabasi AL.. Network-based in silico drug efficacy screening. Nat Commun. 2016;7:10331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Steffens M, Leu C, Ruppert A-K, et al. ; EMINet Consortium. Genome-wide association analysis of genetic generalized epilepsies implicates susceptibility loci at 1q43, 2p16.1, 2q22.3 and 17q21.32. Hum Mol Genet. 2012;21(24):5359–5372. [DOI] [PubMed] [Google Scholar]
- 19. International League Against Epilepsy Consortium on Complex Epilepsies. Electronic address: epilepsy-austin@unimelb.edu.au. Genetic determinants of common epilepsies: A meta-analysis of genome-wide association studies. Lancet Neurol. 2014;13(9):893–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Kasperaviciute D, Catarino CB, Matarin M, et al. ; UK Brain Expression Consortium. Epilepsy, hippocampal sclerosis and febrile seizures linked by common genetic variation around SCN1A. Brain. 2013;136(Pt 10):3140–3150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Sanseau P, Agarwal P, Barnes MR, et al. Use of genome-wide association studies for drug repositioning. Nat Biotechnol. 2012;30(4):317–320. [DOI] [PubMed] [Google Scholar]
- 22. Young MP, Zimmer S, Whitmore AV, Morphy J, Harris C.. Drug molecules and biology: Network and systems aspects. Cambridge, UK: Designing multi-target drugs Royal Society of Chemistry; 2012:32–49. [Google Scholar]
- 23. Iskar M, Campillos M, Kuhn M, Jensen LJ, van Noort V, Bork P.. Drug-induced regulation of target expression. PLoS Comput Biol. 2010;6(9):e1000925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Bantscheff M, Scholten A, Heck AJ.. Revealing promiscuous drug-target interactions by chemical proteomics. Drug Discov Today. 2009;14(21-22):1021–1029. [DOI] [PubMed] [Google Scholar]
- 25. So HC, Chau CK, Lau A, Wong SY, Zhao K.. Translating GWAS findings into therapies for depression and anxiety disorders: Gene-set analyses reveal enrichment of psychiatric drug classes and implications for drug repositioning. Psychol Med. 2019;49(16):2692–2708. [DOI] [PubMed] [Google Scholar]
- 26. So HC, Chau CK, Chiu WT, et al. Analysis of genome-wide association data highlights candidates for drug repositioning in psychiatry. Nat Neurosci. 2017;20(10):1342–1349. [DOI] [PubMed] [Google Scholar]
- 27. Lau A, So HC.. Turning genome-wide association study findings into opportunities for drug repositioning. Comput Struct Biotechnol J. 2020;18:1639–1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Reay WR, Cairns MJ.. Advancing the use of genome-wide association studies for drug repurposing. Nat Rev Genet. 2021;22(10):658–671. [DOI] [PubMed] [Google Scholar]
- 29. Gallagher MD, Chen-Plotkin AS.. The post-GWAS era: From association to function. Am J Hum Genet. 2018;102(5):717–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Sirota M, Dudley JT, Kim J, et al. Discovery and preclinical validation of drug indications using compendia of public gene expression data. Sci Transl Med. 2011;3(96):96ra77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Cheng J, Xie Q, Kumar V, et al. Evaluation of analytical methods for connectivity map data. Pac Symp Biocomput. 2013;5–16. [PubMed] [Google Scholar]
- 32. Okada Y, Wu D, Trynka G, et al. ; GARNET Consortium. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature. 2014;506(7488):376–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wang YF, Zhang Y, Zhu Z, et al. Identification of ST3AGL4, MFHAS1, CSNK2A2 and CD226 as loci associated with systemic lupus erythematosus (SLE) and evaluation of SLE genetics in drug repositioning. Ann Rheum Dis. 2018;77(7):1078–1084. [DOI] [PubMed] [Google Scholar]
- 34. Wain LV, Shrine N, Artigas MS, et al. ; Geisinger-Regeneron DiscovEHR Collaboration. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat Genet. 2017;49(3):416–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Aguirre-Plans J, Pinero J, Sanz F, et al. GUILDify v2.0: A tool to identify molecular networks underlying human diseases, their comorbidities and their druggable targets. J Mol Biol. 2019;431(13):2477–2484. [DOI] [PubMed] [Google Scholar]
- 36. So HC, Chau CK, Lau A, Wong SY, Zhao K.. Translating GWAS findings into therapies for depression and anxiety disorders: Gene-set analyses reveal enrichment of psychiatric drug classes and implications for drug repositioning. Psychol Med. 2019;49:2692–2708. [DOI] [PubMed] [Google Scholar]
- 37. Grau J, Grosse I, Keilwagen J.. PRROC: Computing and visualizing precision-recall and receiver operating characteristic curves in R. Bioinformatics. 2015;31(15):2595–2597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Abdi H. The Kendall rank correlation coefficient. Thousand Oaks, CA: Encyclopedia of Measurement and Statistics Sage; 2007:508–510. [Google Scholar]
- 39. Misawa H, Sherr EH, Lee DJ, et al. Identification of a monogenic locus (jams1) causing juvenile audiogenic seizures in mice. J Neurosci. 2002;22(23):10088–10093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Italiano D, Striano P, Russo E, et al. Genetics of reflex seizures and epilepsies in humans and animals. Epilepsy Res. 2016;121:47–54. [DOI] [PubMed] [Google Scholar]
- 41. Frankel WN. Genetics of complex neurological disease: Challenges and opportunities for modeling epilepsy in mice and rats. Trends Genet. 2009;25(8):361–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Neumann PE, Seyfried TN.. Mapping of two genes that influence susceptibility to audiogenic seizures in crosses of C57BL/6J and DBA/2J mice. Behav Genet. 1990;20(2):307–323. [DOI] [PubMed] [Google Scholar]
- 43. Dürmüller N, Smith SE, Meldrum BS.. Proconvulsant and anticonvulsant effects of Evans blue dye in rodents. Neuroreport. 1993;4(6):683–686. [DOI] [PubMed] [Google Scholar]
- 44. Marson AG, Sills GJ.. Valproate. In: Shorvon, S., Perucca, E. and Engel Jr, J. eds. The treatment of epilepsy. John Wiley & Sons; 2015:652–666. [Google Scholar]
- 45. Tomson T, Johannessen SI.. Carbamazepine. In: Shorvon, S., Perucca, E. and Engel Jr, J. eds. The treatment of epilepsy. John Wiley & Sons; 2015:431–446. [Google Scholar]
- 46. Marson AG, Al-Kharusi AM, Alwaidh M, et al. ; SANAD Study Group. The SANAD study of effectiveness of carbamazepine, gabapentin, lamotrigine, oxcarbazepine, or topiramate for treatment of partial epilepsy: An unblinded randomised controlled trial. Lancet. 2007;369(9566):1000–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Marson A, Burnside G, Appleton R, et al. ; SANAD II Orators. The SANAD II study of the effectiveness and cost-effectiveness of levetiracetam, zonisamide, or lamotrigine for newly diagnosed focal epilepsy: An open-label, non-inferiority, multicentre, phase 4, randomised controlled trial. Lancet. 2021;397(10282):1363–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Marson AG, Al-Kharusi AM, Alwaidh M, et al. ; SANAD Study Group. The SANAD study of effectiveness of valproate, lamotrigine, or topiramate for generalised and unclassifiable epilepsy: An unblinded randomised controlled trial. Lancet. 2007;369(9566):1016–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Marson A, Burnside G, Appleton R, et al. ; SANAD II Orators. The SANAD II study of the effectiveness and cost-effectiveness of valproate versus levetiracetam for newly diagnosed generalised and unclassifiable epilepsy: An open-label, non-inferiority, multicentre, phase 4, randomised controlled trial. Lancet. 2021;397(10282):1375–1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Brodie MJ. Modern management of juvenile myoclonic epilepsy. Expert Rev Neurother. 2016;16(6):681–688. [DOI] [PubMed] [Google Scholar]
- 51. Crespel A, Gelisse P, Reed RC, et al. Management of juvenile myoclonic epilepsy. Epilepsy Behav. 2013;28 Suppl 1:S81–S86. [DOI] [PubMed] [Google Scholar]
- 52. Nicolson A, Marson AG.. When the first antiepileptic drug fails in a patient with juvenile myoclonic epilepsy. Pract Neurol. 2010;10(4):208–218. [DOI] [PubMed] [Google Scholar]
- 53. Senf P, Schmitz B, Holtkamp M, Janz D.. Prognosis of juvenile myoclonic epilepsy 45 years after onset: Seizure outcome and predictors. Neurology. 2013;81(24):2128–2133. [DOI] [PubMed] [Google Scholar]
- 54. Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy. N Engl J Med. 2010;362(9):790–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Speed D, O’Brien TJ, Palotie A, et al. Describing the genetic architecture of epilepsy through heritability analysis. Brain. 2014;137(Pt 10):2680–2689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Luo L, Peng G, Zhu Y, Dong H, Amos CI, Xiong M.. Genome-wide gene and pathway analysis. Eur J Hum Genet. 2010;18(9):1045–1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Watanabe K, Taskesen E, van Bochoven A, Posthuma D.. Functional mapping and annotation of genetic associations with FUMA. Nat Commun. 2017;8(1):1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Segre AV; DIAGRAM Consortium, MAGIC Investigators, et al. Common inherited variation in mitochondrial genes is not enriched for associations with type 2 diabetes or related glycemic traits. PLoS Genet. 2010;6(8):e1001058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Lee PH, O’Dushlaine C, Thomas B, Purcell SM.. INRICH: Interval-based enrichment analysis for genome-wide association studies. Bioinformatics. 2012;28(13):1797–1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Pers TH, Karjalainen JM, Chan Y, et al. ; Genetic Investigation of ANthropometric Traits (GIANT) Consortium. Biological interpretation of genome-wide association studies using predicted gene functions. Nat Commun. 2015;6:5890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. De Wolf H, Cougnaud L, Van Hoorde K, et al. High-throughput gene expression profiles to define drug similarity and predict compound activity. Assay Drug Dev Technol. 2018;16(3):162–176. [DOI] [PubMed] [Google Scholar]
- 63. Hu S, Zhang C, Chen P, Gu P, Zhang J, Wang B.. Predicting drug-target interactions from drug structure and protein sequence using novel convolutional neural networks. BMC Bioinformatics. 2019;20(Suppl 25):689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Hodos R, Zhang P, Lee HC, et al. Cell-specific prediction and application of drug-induced gene expression profiles. Pac Symp Biocomput. 2018;23:32–43. [PMC free article] [PubMed] [Google Scholar]
- 65. Cheng F, Desai RJ, Handy DE, et al. Network-based approach to prediction and population-based validation of in silico drug repurposing. Nat Commun. 2018;9(1):2691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Yazdi A, Doostmohammadi M, Pourhossein Majarshin F, Beheshti S.. Betahistine, prevents kindling, ameliorates the behavioral comorbidities and neurodegeneration induced by pentylenetetrazole. Epilepsy Behav. 2020;105:106956. [DOI] [PubMed] [Google Scholar]
- 67. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511(7510):421–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The following datasets are available for download from the project’s data repository page (https://figshare.com/projects/Using_common_variants_to_find_drugs_for_common_epilepsies/78330):
GWAS gene-based and tissue-wide association study (TWAS) datasets used in our analyses.
Ranked list of the top predicted drugs for each phenotype.
Our complete set of predictions, listing each drug and its FAM score, for each phenotype.