“Transcriptomic Profiling Unveils Novel Therapeutic Options for Drug-Resistant Temporal Lobe Epilepsy” ========================================================================================================== * Patricia Sánchez-Jiménez * Lola Alonso-Guirado * Laura Cerrada-Gálvez * Marcos Elizalde-Horcada * Inmaculada Granero-Cremades * Paolo Maietta * Antonio Gómez-Martín * Francisco Abad-Santos * Cristina Virginia Torres-Díaz * Maria de Toledo * Natalia Frade-Porto * Patricia Gonzalez-Tarno * Paloma Pulido * María C. Ovejero-Benito ## ABSTRACT **Background** Epilepsy drug treatments fail in 25-30% of patients, who then develop drug resistance. Temporal lobe epilepsy is the most prevalent subtype associated with drug resistance. Classical drug discovery is a long and extremely costly process with a high rate of failure in clinical trials. Drug repurposing is a more cost- and time-effective strategy. Hence, the main objective of this study is to propose drug candidates for the treatment of drug-resistant temporal lobe epilepsy (DR-TLE) through drug repurposing based on transcriptomic profiling. **Methods** Total RNA-sequencing (RNA-Seq) was performed on 45 formalin-fixed paraffin-embedded (FFPE) hippocampi of DR-TLE patients and 36 FFPE hippocampi of post-mortem biobank donors. RNA-Seq was carried out in an Illumina NovaSeq 6000 platform in 100bp paired-end. Drug repurposing based on transcriptomic analysis top candidates was performed against these databases: Pandrugs2, PharmOmics, DGIdb, ToppGene, L1000CDS2 and Connectivity Map. **Results** We found 887 genes differentially expressed between DR-TLE patients and post-mortem controls. We observed 74 potential drug candidates in at least two independent databases. Of these, we selected only the 11 which can cross the blood-brain barrier: cobimetinib, panobinostat, melphalan, rucaparib, alectinib, ponatinib, danazol, carboplatin, vandetanib, erlotinib, and gefitinib. After analyzing their safety and efficacy profile based on previous publications, we provide a list of the top 5 candidates. **Conclusions** We therefore propose erlotinib, danazol, rucaparib, ponatinib, and panobinostat as potential therapies for DR-TLE based on differential RNA-Seq profiling. Keywords * Drug repurposing * refractory epilepsy * drug-resistant epilepsy * transcriptomics * RNA-Seq * drug discovery ## INTRODUCTION Epilepsy is a chronic neurological disease, often difficult to detect, frequently of unknown etiology and challenging to control. The number of approved antiseizure drugs (ASDs) has increased considerably over the last few years 1,2. However, 25-33% of epileptic patients do not respond adequately to these medications 1, and they develop drug-resistant epilepsy (DRE). Frequently, neurologists observe DRE appearing in focal epilepsies, especially in those associated with temporal lobe epilepsy (TLE), the most common subtype of epilepsy 3. Nowadays, the etiology of DRE is still unknown, but it has been found that drug resistance often occurs nine years after starting pharmacological treatment 4 although the cause of this latency is still unknown. Several hypotheses try to explain DRE, such as genetic variations, disease-related and drug-related mechanisms 5. It has been hypothesized that environmental factors and epigenetic mechanisms are involved in the development of drug resistance 6–11. Epigenetic modifications can modulate gene expression, and different studies have explored the connection between gene expression and drug resistance 12–17. There are several therapeutic alternatives for DRE patients, such as the highly effective neurosurgical resection of the epileptogenic zone 18. However, this is only applicable to focal epilepsies such as TLE 19. In addition, there is an unmet need for disease-modifying ASDs that reduce the severity or delay the onset of seizures 20. Discovery of new ASDs is a long and incredibly costly process, with a high failure rate in clinical trials due to inefficacy 21. The failure of new drugs for epilepsy in clinical trials may be partly explained by the fact that ASDs are tested in preclinical models such as maximal electroshock seizure (MES) and s.c. pentylenetetrazole (PTZ). These could be considered models of seizure rather than models of epilepsy 22. Drug discovery is implementing computation models and new technologies to accelerate the process 23–26. However, the time period from candidate drug to approved drug is still long and with a low success rate 27. In recent years drug repurposing strategies have been used to identify new indications for approved drugs 28. Drug repurposing accelerates the approval of these new treatments by 3 to 12 years and reduces costs and risk by 50%. The approval rate is higher in repurposed drugs (30%) than in new compounds (10%) 2. Drug repurposing is based on the fact that drugs may have multiple targets, while different diseases may have in common the same genetic factors, molecular pathways, and/or clinical manifestations. Therefore, a drug can potentially modulate these common factors and may benefit different diseases 29. Drug repurposing has been successfully used for diverse diseases such as cancer or COVID-19 30–32. Systematic analysis of classical drug discovery pipelines showed that drugs developed with supporting human genetic data had a double probability of being approved 21,33. This approach has not been deeply explored in the epilepsy field, since, to our knowledge, there is only one study of drug repurposing based on transcriptomics in epilepsy 34. The main objective of this study is the identification of approved drug candidates that can modulate factors and/or molecular pathways involved in drug-resistant temporal lobe epilepsy (DR-TLE) based on the differential gene expression data. ## MATERIALS AND METHOD ### Study population A total of 46 hippocampal formalin-fixed paraffin-embedded (FFPE) samples from DR-TLE patients subjected to neurosurgery in the previous ten years were analyzed. All the recruited patients agreed with the protocol and signed the informed consent approved by the Independent Clinical Research Ethics Committee of Hospital Universitario de La Princesa. The study was carried out according to the revised Declaration of Helsinki and STROBE guidelines. Thirty-five hippocampal FFPE samples were provided as control samples from Spanish biobanks: “Biobanco en Red de la Región de Murcia” (BIOBANC-MUR), “Biobanco del Hospital Universitario Puerta de Hierro” (HUPHM), “Biobanco del Instituto de Investigaciones Biomédicas August Pi i Sunyer” (IDIBAPS), and “Biobanco de Navarrabiomed” (Navarrabiomed). Moreover, they were processed following standard operating procedures with the approval of their Ethics and Scientific Committees. None of them suffered any psychiatric, neurological, or neurodegenerative diseases, and all were analyzed by anatomopathologist to discard pathological features. Controls were of similar age to the patients. ### Sample processing and RNA isolation A total of 81 hippocampal FFPE blocks were cut on a 15μm scroll. RNA was isolated with the truXTRAC® FFPE total NA kit - Column (Covaris, USA), according to the manufacturer’s instructions. Then, DNA-free RNA was obtained with the RNase-Free DNase Set (Qiagen, Germany) and the RNeasy® MinElute® Cleanup kit (Qiagen, Germany). Quantification and quality control were performed before and after those steps using the 4200 TapeStation system (Agilent, USA) and the Qubit 2.0 Fluorometer (Thermo Fisher Scientific, USA). ### Library Preparation and Sequencing Libraries were prepared using the SMARTer® Stranded Total RNA-Seq kit v2-Pico Input Mammalian (Takara Bio, Japan) following the manufacturer’s protocol to process FFPE samples. Libraries were sequenced to achieve an average output of 50M paired-end reads (100bp pair-end) on the Illumina NovaSeqTM 6000 platform. Read quality was assessed by fastp 35. Raw reads were aligned against GRCh37/hg19 reference genome with HISAT2 36 and StringTie 37 to obtain a counts matrix according to the protocol described by 38. ### Differential Expression Analysis using DESeq2 We performed a differential expression analysis of genes using the DESeq2 package (version 1.34.0) in the R software (R version 4.2.2). Raw gene counts matrix was pre-processed before the analysis. In the quality control checks performed, samples with an alignment rate lower than 5% were discarded (13 controls and 1 patient). The sample size was reduced from 81 (35 controls and 46 patients) to 67 subjects (22 controls and 45 patients) to satisfy our integrity and accuracy quality thresholds (Figure 1). ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/26/2024.06.26.24309519/F1/graphic-1.medium.gif) [](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/F1/graphic-1) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/26/2024.06.26.24309519/F1/graphic-2.medium.gif) [](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/F1/graphic-2) Figure 1: Workflow of the experiments and analysis carried out. Abbreviations: BBB: blood-brain barrier; CMap: Connectivity Map; DGIdb: Drug Gene Interaction Database; DR-TLE: drug-resistant-temporal lobe epilepsy. We loaded the complete phenotypes dataset from ENSEMBL 75, and we filtered out nuclear and mitochondrial RNA-ribosomes, snoRA (small nucleolar RNAs), and SNORD (small nucleolar RNA-derived) genes 39 from the gene raw counts dataframe. The raw count data was then normalized using the DESeq2 normalization methods. As “metadata” information about the samples, we had alignment rate, cohort information, sex or batch. To control the potential effect of geographical regions on the study outcomes, a new variable called “region” was included in the contrast parameters. Thus, the contrast parameters used for the DESeq2 analysis were the variables “sex”, “region”, “age”, and “treatment”. Then, we obtained the list of differentially expressed genes (DEGs) with an absolute lfcThreshold=0.5 and a FDR-adjusted p-value lower than 0.05. ### Enrichment analysis Signaling pathways and biological functions associated with DR-TLE were analyzed using the following tools: Enrichr 40, FUMA GWAS 41, g:Profiler 42 and ToppGene 43. We analyzed separately for the upregulated and downregulated genes in patients with respect to controls (Figure 1). Only significant results with a FDR-adjusted p-value <0.05 were considered. Protein network analysis was performed by STRING 44 and Cytoscape software version 3.10.1. 45. Markov Clustering Algorithm was applied to determine groups of nodes within the network using a 0.4 cutoff and 4 points of granularity. ### Drug repurposing Drug repurposing was performed using the following tools: Pandrugs2 46; PharmOmics 47; Drug Gene Interaction Database (DGIdb) 48; ToppGene Suite 43; L1000CDS2 49; Connectivity Map (CMap) 50 (Figure 1). All these algorithms score the association between genes and drug targets. Some drug candidate lists were filtered more restrictively to select the top candidates. In Pandrugs2, just the best candidates for therapies were selected. The functions of the proteins encoded by downregulated genes in DR-TLE patients with respect to controls are decreased in these patients. Therefore, we filtered the list of drugs proposed by DGIdb to modulate downregulated genes, and discarded drugs classified as blockers, inhibitors, or antagonists. In Connectivity Map (CMap), the drug candidates with negative Raw_cs and a base 10 logarithm of FDR (fdr_q_nlog10) higher than 15.35, were selected. CMap allows a maximum input of only 150 upregulated and 150 downregulated DEGs. To perform this analysis, we selected those genes with the lowest and highest log2fc values and that were included in the CMap database. Venn Diagram plots were performed with the tool Interactive Venn ([https://www.interactivenn.net/](https://www.interactivenn.net/)). To increase the reliability of our results, the webtool SwissADME 51 was employed to predict whether potential drugs could cross blood-brain barrier BBB and access the target tissue. This tool also analyzes whether drugs are substrates of Glycoprotein-P that reduces the amount of drugs that reach the brain 5,52, and the ability of these drugs to inhibit CYP450 superfamily enzymes. Pan-Assay Interference compounds (PAINS) are promiscuous molecules that form unspecific interactions with many different proteins. For this reason, PAINS could give rise to false positives in drug repurposing studies 53. We filtered out the PAINS identified with the webtool Hit Dexter 3 53. ## RESULTS ### Study population Initially, 81 samples were recruited (46 DR-TLE patients and 35 controls). Our final cohort after having discarded the samples that did not reach quality standards included 67 subjects: 45 DR-TLE patients, with a mean age of 46.20 ± 10.36 years old, and 22 post-mortem controls, with a mean age of 49.09 ± 14.80 years old (Table 1). The percentage of men in the patient group was 46%, while in the control group was 74% (Table 1), representing a significant difference (p=0.045). This imbalance increased because some samples were removed from the initial cohort after quality control checks. The main clinical and demographic characteristics of patients and controls are described in Table 1. View this table: [Table 1:](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/T1) Table 1: Summary of the clinical and demographic characteristics of the study population and the controls. ### Differentially expressed genes associated with DR-TLE We observed 887 DEGs between DR-TLE patients and *post-mortem* controls: 471 upregulated and 416 downregulated in patients with respect to controls (Figure 2A, Supplementary Table 1). The top 10 upregulated and downregulated genes in DR-TLE patients with respect to controls are shown in Table 2. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/26/2024.06.26.24309519/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/F2) Figure 2. **A)** Volcano plots of the DEGs associated with DR-TLE in the hippocampus. The logarithm of the association p-value for each DEGs is displayed along the Y-axis while the logarithm of fold change is shown along the X-axis. Green and red dots show DEGs over-expressed and under-expressed in patients with respect to controls, respectively. Blue dots show DEGs that do not have a significant association with DR-TLE. Only the names of the 40 DEGs with the lowest adjusted p-value are displayed. B) Enrichment analysis performed with FUMA GWAS analyzing functions of the DEGs in the hippocampus according to the GO molecular function and GO biological processes libraries. Red bars represent the proportion of overlapping genes in the gene set. Blue bars show the enrichment p-value, represented as the logarithm of the FDR adjusted p-value. Orange squares show the genes involved in every enrichment term. Abbreviation: FDR, false discovery rate. **B)** Enrichment analysis of the DEGs upregulated in patients with respect to controls. **C)** Enrichment analysis of the DEGs downregulated in patients with respect to controls. Red bars represent the proportion of overlapping genes in gene set. Blue bars show the enrichment p-value, represented as the negative logarithm of the FDR adjusted p-value. Yellow squares show the genes involved in every enrichment term. Abbreviation: DEGs: Differentially Expressed Genes; DR-TLE: drug-resistant temporal lobe epilepsy; FDR: false discovery rate. View this table: [Table 2:](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/T2) Table 2: Top 10 upregulated and downregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. ### Enrichment analysis To determine the biological functions and the signaling pathways where the DEGs associated with DR-TLE were involved, we performed an enrichment study. Genes upregulated in DR-TLE were enriched in methyltransferase activity, oxidoreductase activity, catabolic processes, lipid metabolism and immune system activation (Figure 2B, Supplementary Figure 1B and 1C). In contrast, downregulated genes were enriched in alternative splicing, calcium ion binding, and Cajal bodies (Figure 2C, Supplementary Figure 1A). Furthermore, we performed a clustering analysis with Cytoscape of the proteins encoded by DEGs associated with DR-TLE. This software grouped proteins in clusters of 5 or more, related to specific functions (Figure 3, Supplementary Table 2). Upregulated genes were clustered in 8 groups (Supplementary Table 2A), with the four largest involved in functions such histone modifications (Figure 3A-1), metabolic pathways (Figure 3A-2), inflammatory response (Figure 3A-3), and cysteine and methionine metabolism (Figure 3A-4). Downregulated genes were grouped in 5 clusters (Supplementary Table 2B). The four biggest clusters were implicated in functions such as calcium signaling (Figure 3B-1), cytoplasmic ribonucleoprotein granule (Figure 3B-2), PI3K-Akt signaling pathway (Figure 3B-3), and eosinophil percentage of leukocytes (Figure 3B-4, Supplementary Table 2B). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/26/2024.06.26.24309519/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/F3) Figure 3. Cytoscape network of proteins expressed by DEGs associated with DR-TLE. A) Top 4 largest clusters of the proteins encoded by the genes upregulated in DR-TLE patients with respect to controls. B) Top 4 largest clusters of the proteins encoded by the downregulated in DR-TLE patients with respect to controls. Abbreviation: DEGs: Differentially Expressed Genes; DR-TLE: drug-resistant temporal lobe epilepsy. ### Drug repurposing We found 42 potential drug candidates in L1000CDS2, and 979 in CMap, associated with both upregulated and downregulated genes (Supplementary Figure 2A, Supplementary Table 3A). In the case of **upregulated** DEGs in DR-TLE patients with respect to controls, we observed 140 potential drug candidates in the Pandrugs2 database, 13 in PharmOmics, 67 in DGIdb and 48 in ToppGene (Supplementary Table 3A). Then, we analyzed the 54 candidates identified by at least two different databases (Supplementary Table 3B). We observed 35 potential drug candidates in Pandrugs2 and DGIdb; 1 in both Pandrugs2 and L1000CDS2; 5 in Pandrugs2 and CMap; 5 in L1000CDS2 and CMap; 2 in both DGIdb and CMap; 2 in both ToppGene and CMap and 4 in Pandrugs, DGIdb and CMap (Supplementary Figure 2B, Supplementary Table 3B). Only 9 of these potential drug candidates cross the BBB (Supplementary Table 3C). After that, we searched for potential drugs that could modulate **downregulated** genes in DR-TLE patients with respect to controls. We obtained 166 potential drug candidates in Pandrugs2; 101 in PharmOmics; and 54 in DGIdb (Supplementary Figure 2B, Supplementary Table 3D). ToppGene did not propose any drug candidate for downregulated genes. Again, we focused on the 52 candidates proposed by at least 2 different databases. We obtained 7 potential drug candidates in PharmOmics and Pandrugs2; 2 in both PharmOmics and DGIdb; 1 in PharmOmics and CMap; 23 in Pandrugs2 and DGIdb; 4 in Pandrugs2 and CMap; 5 in L1000CDS2 and CMap; and 1 in DGIdb and CMap (Supplementary Figure 2B, Supplementary Table 3E). We also found 8 potential drug candidates proposed by 3 databases: 5 drugs in both PharmOmics, Pandrugs2 and DGIdb; 1 in Pandrugs2, DGIdb, and L1000CDS2; and 2 in Pandrugs2, DGIdb, and CMap (Supplementary Figure 2B, Supplementary Table 3E). We discovered 1 potential drug candidate in common in 4 databases: docetaxel was proposed by PharmOmics, Pandrugs2, DGIdb, and CMap (Supplementary Figure B, Supplementary Table 3E). From all the mentioned drug candidates, we selected only the 9 drugs that cross the BBB (Supplementary Table 3F). We then explored the pharmacokinetics, pharmacodynamics, and toxicology properties of the drug candidates. To select the best candidate drugs for treating DR-TLE, we focused on those which can reach the brain. We studied the therapeutic indication, target, and mechanism of action of these drugs (Supplementary Table 4). As the repurposed drugs may be used in combination with other ASDs, we analyzed the ability of the candidate drugs to inhibit the main enzymes of cytochrome P450 superfamily (Supplementary Table 4). Finally, we described the most common adverse drug reactions (ADRs) that appear in one in ten patients (Very Common ADRs) (Supplementary Table 4). Based on their pharmacological and toxicological profile, we obtained a list of the top 5 candidates for treating DR-TLE: erlotinib, danazol, rucaparib, ponatinib, and panobinostat (Table 3). View this table: [Table 3:](http://medrxiv.org/content/early/2024/06/26/2024.06.26.24309519/T3) Table 3: List of the top five drug candidates proposed for drug-resistant temporal lobe epilepsy treatment. ## DISCUSSION This study applied next-generation sequencing to the epileptogenic zone of DR-TLE patients in order to guide the selection of drug candidates, already approved to treat other conditions, for the treatment of this disease. We observed 471 upregulated and 416 downregulated genes in DR-TLE patients compared to *post-mortem* non-epilepsy controls. We observed that genes upregulated in DR-TLE were involved in different processes such as histone modifications, methyltransferase activity, metabolic pathways, inflammatory response, alternative splicing, lipid oxidation or cysteine metabolism. Most of these processes have been previously associated with DRE 6,54–56. In fact, the over-expression of histone modification and methyltransferases may support the epigenetic hypothesis of DRE 6,57–59. There is wide evidence that inflammatory response is deeply involved in DRE 5,7,8,60–62. Moreover, mutations on splicing sites are associated with myoclonic epilepsy 56 or epileptic encephalopathies 63. In addition, lipid metabolism and fatty acid oxidation were overexpressed in brain samples of DRE patients 55,64. Cysteine and methionine metabolism were upregulated in DR-TLE patients. Surprisingly, cysteine levels are increased in blood samples of epilepsy patients 54. On the other hand, genes downregulated in DR-TLE patients play roles related to DRE such as calcium signaling 5, senescence or PI3K signaling 65. There are previous transcriptomic analysis of DRE and DR-TLE 12–15,17,61,66–68. Some of these took a different approach from this study, focusing on specific features such as hippocampal sclerosis 14,17, febrile seizure 68, or electric activity 69. Other studies analyzed different brain regions such as the neocortex 15,69. Discrepancies in the number of DEGs found in these studies could be explained partially by diverse factors, such as patients’ ethnicity 12, the technique used 69, or the conservation method (paraffin-embedded and fixed versus fresh tissue) 14,15,17. We used FFPE tissue based on the availability of these samples in the anatomical pathology unit of the hospital and biobanks. However, paraffin may act as an intercalant in nucleic acids, modifying nucleotides and affecting RNA integrity and stability 70. Thus, we had to discard 14 samples (13 controls and 1 DRE patient) because their alignment rate was lower than 5%. Even after this patient exclusion, we had a sample of 46 patients, a higher number than in previous studies 12,14,61. Drug repurposing based on transcriptomics is a powerful, time- and cost-effective strategy 30–32. In epilepsy, the principal strategy to discover new candidates for drug therapy was genome-wide association studies 30,71. In this study, we used the transcriptomic strategy for drug repurposing and employed six repurposing databases. We selected drugs proposed by at least two independent databases to increase the robustness of the results 32. Epilepsy treatment is challenging because BBB limits the number of drugs that reach the brain 72. However, we found 11 drug candidates that can cross the BBB: alectinib, carboplatin, cobimetinib, danazol, erlotinib, gefitinib, melphalan, panobinostat, ponatinib, rucaparib and vandetanib. To prioritize the best candidates for DR-TLE, we thoroughly explored their safety profile and the information derived from the previous use of these drugs in epilepsy. For instance, there are clinical case reports and studies showing patients who developed seizures after being treated with melphalan for myeloma 73, alectinib for lung tumors 74, or vandetinib or gefitinib for glioma 75,76. Moreover, gefitinib showed brain toxicity in a chronic epilepsy drug screening 77. In addition, it was reported that carboplatin can induce the development of reversible posterior leukoencephalopathy syndrome, which courses with seizures 78 and was not suitable for treating chronic epilepsy in a drug screening 77. Cobimetinib does not have anticonvulsant effects by itself because, for the treatment of advanced metastatic melanoma, it must be used in combination with carbamazepine to control seizures 79. Therefore, based on the previous publications and their safety profile (Supplementary Table 4), we do not recommend validating alectinib, carboplatin, cobimetinib, gefitinib, melphalan, or vandetanib in DR-TLE. On the other hand, previous publications suggest that there are drug candidates that might be effective for DR-TLE. For instance, erlotinib is approved for tumoral epilepsy 80 and is not a substrate of P-glycoprotein, so most of the drug dose will reach the brain. Panobinostat is approved for the treatment of refractory multiple myeloma. It is a histone deacetylase inhibitor, in the same way as valproic acid, and was proposed as a possible antiseizure therapy 10,81 and a therapeutic alternative for neuromuscular disease 82. Moreover, this drug is promising for DR-TLE since it could modify gene expression and prevent epileptogenesis 10,81. Danazol is approved for fibrocystic breast cancer. In one case study danazol was applied to treat cerebral endometriosis after brain surgery and the patient remained seizure-free with this treatment 83. Rucaparib, indicated for recurrent epithelial ovarian cancer, was proposed to treat inflammatory diseases, including neurological disorders 84, and was even proposed for visual epilepsy in a drug repurposing study 85. Ponatinib is used to treat chronic myeloid leukemia. In an *in-silico* study, ponatinib was suggested for the treatment of functional seizures 86, and it also reduced seizure severity in a kainate-induced status epilepticus rat model 87. Most of the ASDs are taken in polytherapy for DRE 88. Therefore, we studied the ability of the selected drug candidates to inhibit the main members of the CYP450 superfamily (CYP1A2, CYP2C9, CYP3C19, CYP2D6, and CYP3A4). These drugs inhibit many of these enzymes, so they may cause interactions when used in combination with other ASDs. However, it would be safe to combine them with drugs that are not metabolized by the liver, such as levetiracetam 89. Another factor we considered was whether our drug candidates are substrates of P-glycoprotein that pumps drugs out of the brain 90,91. We found that panabinostat, danazol, ponatinib, and rucaparib are substrates, which may require dose adjustment to achieve the therapeutic dose in the brain. Although these drug candidates may be promising treatment for DR-TLE, they are all part of pharmacological chemotherapies that can cause a wide range of very common ADRs. This may be due to the databases used, such as Pandrugs2, which prioritize anti-tumor drugs 46. However, for treating epilepsy, the therapeutic dose may be lower than that required to treat tumors, thus reducing the drugs’ toxicity. Further studies will be needed to determine the adequate dose to treat DR-TLE, but given the severity of DR-TLE, if these drugs decrease the frequency of seizures, the benefits may outweigh the risks. There is only one other publication that performed drug repurposing based on transcriptomics in epilepsy 34. We observed that 67 drugs proposed were common to both studies, such as metformin 92 and nifedipine, which were validated in a zebrafish model 34. We did not have the chance to validate the results in an animal model which is the main limitation of this study. However, we used 6 different repurposing databases, we studied their ability to cross the BBB, as well as their safety profile, and we relied on a larger cohort than the previous publication (N=46 (our study) vs N=6 34). In conclusion, we provide insights into the changes in gene expression associated with DR-TLE. This study proposes promising potential drug candidates to treat DR-TLE: danazol, rucaparib, erlotinib, ponatinib, and panobinostat. Nevertheless, further research is needed to validate these results in a preclinical model. ## Supporting information Supplementary Figure 1 [[supplements/309519_file06.jpg]](pending:yes) Supplementary Figure 2 [[supplements/309519_file07.jpg]](pending:yes) Supplementary Table 1 [[supplements/309519_file08.xlsx]](pending:yes) Supplementary Table 2 [[supplements/309519_file09.xlsx]](pending:yes) Supplementary Table 3 [[supplements/309519_file10.xlsx]](pending:yes) Supplementary Table 4 [[supplements/309519_file11.xlsx]](pending:yes) ## SUPPLEMENTARY TABLE **Supplementary Table 1.** Differentially expressed genes (DEGs) associated with drug-resistant temporal lobe epilepsy in the hippocampus sorted by their logarithm of fold change. Genes under-expressed in drug-resistant temporal lobe epilepsy patients with respect to controls have a negative value of Log2FC and are shown in red. Genes over-expressed in drug-resistant temporal lobe epilepsy patients with respect to controls have a positive value of Log2FC are shown in green. Abbreviations: Log2FC: logarithm of fold change. **Supplementary Table 2.** Definition of the main clusters created by Cytoscape. A) Clusters of proteins encoded by upregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. B) Clusters of proteins encoded by downregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. **Supplementary Table 3.** Detailed information about the repurposed drugs obtained with the different databases. A) Drugs proposed by each database based on upregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. B) Selection of the drugs proposed by at least 2 different databases based on upregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. C) Characteristic of the drugs proposed by at least 2 different databases based on upregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. D) Drugs proposed by each database based on downregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. E) Selection of the drugs proposed by at least 2 different databases based on downregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. F) Characteristics of the drugs proposed by two different databases using downregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. We tested drugs’ ability to cross the BBB, whether they were substrates of PgP or PAINS and we showed their SMILES code. Abbreviations: BBB: blood-brain barrier, CMap: Connectivity Map; DGIdb: Drug Gene Interaction Database; PgP: P-glycoprotein, PAINS: Pan-Assay Interference compounds; SMILES: Simplified Molecular Input Line Entry System. Drugs that can cross the blood-brain barrier were highlighted in pink. **Supplementary Table 4:** Detailed information of the pharmacodynamics, pharmacokinetics and toxicological characteristics of drug candidates. Only very common drug reactions, affecting more than 10% of the population are shown in this table. Promising drug candidates for drug-resistant temporal lobe epilepsy, based on previous publications and safety were shown in green. Abbreviations: BBB: blood-brain barrier, CYP: cytochrome P 450; GI: gastrointestinal; P-gp: P-glycoprotein. **Supplementary Figure 1. A)** Enrichment analysis of the downregulated with a logarithm of fold change in drug-resistant temporal lobe epilepsy patients with respect to controls lower than 0.5 using g:Profiler. **B)** Enrichment analysis of the upregulated with a logarithm of fold change in drug-resistant temporal lobe epilepsy patients with respect to controls higher than 0.5 using G-profiler. **C**) Enrichment analysis performed with FUMA GWAS analyzing functions of the DEGs in the hippocampus according to the immunological signature libraries. Red bars represent the proportion of overlapping genes in the gene set. Blue bars show the enrichment p-value, represented as the logarithm of the FDR adjusted p-value. Abbreviation: FDR, false discovery rate. **Supplementary Figure 2.** Venn diagrams of the comparison and matching of repurposed drugs among different databases. A) Upregulated genes B) Downregulated genes in drug-resistant temporal lobe epilepsy patients with respect to controls. Abbreviations: CMap: Connectivity Map; DGIdb: Drug Gene Interaction Database. ## DATA AVAILABILITY All data produced in the present study are available upon reasonable request to the authors. ## FUNDING STATEMENT This study was supported by Instituto de Salud Carlos III: PI2020/01391. PSJ is funded by Industrial PhD grant from ‘Consejeria de Educación e Investigación’ of ‘Comunidad de Madrid’ developed in NIMGenetics and in Hospital Universitario de La Princesa (CAM.IND2017/BMD-7578). ## CONFLICT OF INTERESTS DISCLOSURE F Abad-Santos has been a consultant or investigator in clinical trials sponsored by the following pharmaceutical companies: Abbott, Alter, aptaTargerts, Chemo, Farmalíder, Ferrer, GlaxoSmithKline, Gilead, Janssen-Cilag, Kern, Normon, Novartis, Servier, Teva, and Zambon. MC Ovejero-Benito has potential conflicts of interest (research support) with Leo Pharma. None of the conflicts of interest disclosed relate to this publication. The rest of the authors have no relevant financial or non-financial interests to disclose. ## ETHICAL DECLARATION The protocol and the Informed Consent Form were approved by the Independent Clinical Research Ethics Committee of the Hospital Universitario de La Princesa. The study followed the STROBE guidelines and the Revised Declaration of Helsinki. ## PATIENT CONSENT STATEMENT All the recruited patients agreed with the protocol and signed the informed consent approved by the Independent Clinical Research Ethics Committee of Hospital Universitario de La Princesa. ## ACKNOWLEDGMENTS We are particularly grateful for the generous contribution of the patients and the different Spanish Biobanks: “Biobanco del Hospital Universitario Puerta de Hierro” (HUPHM), “Biobanco del Instituto de Investigaciones Biomédicas August Pi i Sunyer” (IDIBAPS), and “Biobanco de Navarrabiomed” (Navarrabiomed) and “Biobanco en Red de la Región de Murcia” (BIOBANC-MUR). BIOBANC-MUR (Collaboration of Biobank Network of the Region of Murcia) is registered on the Registro Nacional de Biobancos with registration number B.0000859. BIOBANC-MUR is supported by the “Instituto de Salud Carlos III (proyecto PT20/00109), by “Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, IMIB” and by “Consejeria de Salud de la Comunidad Autónoma de la Región de Murcia”. We would like to thank the following scientists for their help with this study: Dr. Javier Fraga, Dr. Juan Cruz Cigudosa and Gemma Benito. We would like to thank Dr. Gillian Moody for her valuable comments on this manuscript. * Received June 26, 2024. * Revision received June 26, 2024. * Accepted June 26, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## REFERENCES 1. 1.Amin U, Benbadis SR. Avoiding complacency when treating uncontrolled seizures: why and how? Expert Rev Neurother. 2020;20(3):227–235. doi:10.1080/14737175.2020.1713100 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/14737175.2020.1713100&link_type=DOI) 2. 2.Löscher W. Single-Target Versus Multi-Target Drugs Versus Combinations of Drugs With Multiple Targets: Preclinical and Clinical Evidence for the Treatment or Prevention of Epilepsy. Front Pharmacol. 2021;12. doi:10.3389/fphar.2021.730257 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fphar.2021.730257&link_type=DOI) 3. 3.Schmidt D, Löscher W. Drug resistance in epilepsy: putative neurobiologic and clinical mechanisms. Epilepsia. 2005;46(6):858–877. doi:10.1111/j.1528-1167.2005.54904.x [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1528-1167.2005.54904.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15946327&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000229802200007&link_type=ISI) 4. 4.Janmohamed M, Brodie MJ, Kwan P. Pharmacoresistance – Epidemiology, mechanisms, and impact on epilepsy treatment. Neuropharmacology. 2020;168:107790. doi:10.1016/j.neuropharm.2019.107790 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuropharm.2019.107790&link_type=DOI) 5. 5.Löscher W, Potschka H, Sisodiya SM, Vezzani A. Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options. Barker EL, ed. Pharmacol Rev. 2020;72(3):606–638. doi:10.1124/pr.120.019539 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoicGhhcm1yZXYiO3M6NToicmVzaWQiO3M6ODoiNzIvMy82MDYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wNi8yNi8yMDI0LjA2LjI2LjI0MzA5NTE5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 6. 6.Kobow K, Reid CA, van Vliet EA, et al. Epigenetics explained: a topic “primer” for the epilepsy community by the ILAE Genetics/Epigenetics Task Force. Epileptic Disord. 2020;22(2):127–141. doi:10.1684/epd.2020.1143 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1684/epd.2020.1143&link_type=DOI) 7. 7.Martins-Ferreira R, Leal B, Chaves J, et al. Epilepsy progression is associated with cumulative DNA methylation changes in inflammatory genes. Prog Neurobiol. 2022;209:102207. doi:10.1016/j.pneurobio.2021.102207 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pneurobio.2021.102207&link_type=DOI) 8. 8.Sánchez-Jiménez P, Elizalde-Horcada M, Sanz-García A, et al. DNA Methylation Description of Hippocampus, Cortex, Amygdala, and Blood of Drug-Resistant Temporal Lobe Epilepsy. Mol Neurobiol. 2023;60(4):2070–2085. doi:10.1007/s12035-022-03180-z [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12035-022-03180-z&link_type=DOI) 9. 9.Xiao W, Liu C, Zhong K, et al. CpG methylation signature defines human temporal lobe epilepsy and predicts drug-resistant. CNS Neurosci Ther. 2020;26(10):1021–1030. doi:10.1111/cns.13394 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/cns.13394&link_type=DOI) 10. 10.Wang Z, Ren D, Zheng P. The role of Rho/ROCK in epileptic seizure-related neuronal damage. Metab Brain Dis. 2022;37(4):881–887. doi:10.1007/s11011-022-00909-6 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11011-022-00909-6&link_type=DOI) 11. 11.Zhang W, Wang H, Liu B, et al. Differential DNA Methylation Profiles in Patients with Temporal Lobe Epilepsy and Hippocampal Sclerosis ILAE Type I. J Mol Neurosci. 2021;71(9):1951–1966. doi:10.1007/s12031-020-01780-9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12031-020-01780-9&link_type=DOI) 12. 12.Dixit AB, Banerjee J, Srivastava A, et al. RNA-seq analysis of hippocampal tissues reveals novel candidate genes for drug refractory epilepsy in patients with MTLE-HS. Genomics. 2016;107(5):178–188. doi:10.1016/j.ygeno.2016.04.001 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygeno.2016.04.001&link_type=DOI) 13. 13.Dixit AB, Sharma D, Tripathi M, et al. Genome-wide DNA Methylation and RNAseq Analyses Identify Aberrant Signalling Pathways in Focal Cortical Dysplasia (FCD) Type II. Sci Rep. 2018;8(1):17976. doi:10.1038/s41598-018-35892-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-018-35892-5&link_type=DOI) 14. 14.Griffin NG, Wang Y, Hulette CM, et al. Differential gene expression in dentate granule cells in mesial temporal lobe epilepsy with and without hippocampal sclerosis. Epilepsia. 2016;57(3):376–385. doi:10.1111/epi.13305 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/epi.13305&link_type=DOI) 15. 15.Guelfi S, Botia JA, Thom M, et al. Transcriptomic and genetic analyses reveal potential causal drivers for intractable partial epilepsy. Brain. 2019;142(6):1616–1630. doi:10.1093/brain/awz074 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awz074&link_type=DOI) 16. 16.Kjær C, Barzaghi G, Bak LK, et al. Transcriptome analysis in patients with temporal lobe epilepsy. Brain. 2019;142(10):e55–e55. doi:10.1093/brain/awz265 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awz265&link_type=DOI) 17. 17.Miller-Delaney SFC, Bryan K, Das S, et al. Differential DNA methylation profiles of coding and non-coding genes define hippocampal sclerosis in human temporal lobe epilepsy. Brain. 2015;138(3):616–631. doi:10.1093/brain/awu373 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awu373&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25552301&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 18. 18.Torres CV, Pastor J, García-Navarrete E, Pulido-Rivas P, Sola RG. Classification of structural lesions in magnetic resonance imaging. Surgical implications in drug-resistant epilepsy patients. Rev Neurol. 2015;61(6):241–248. 19. 19.Roessler K, Kasper BS, Shawarba J, et al. Operative variations in temporal lobe epilepsy surgery and seizure and memory outcome in 226 patients suffering from hippocampal sclerosis. Neurol Res. 2021;43(11):884–893. doi:10.1080/01616412.2021.1942407 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/01616412.2021.1942407&link_type=DOI) 20. 20.Jacobs MP, Leblanc GG, Brooks-Kayal A, et al. Curing epilepsy: Progress and future directions. Epilepsy Behav. 2009;14(3):438–445. doi:10.1016/j.yebeh.2009.02.036 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.yebeh.2009.02.036&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19341977&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265040200003&link_type=ISI) 21. 21.Nelson MR, Tipney H, Painter JL, et al. The support of human genetic evidence for approved drug indications. Nat Genet. 2015;47(8):856–860. doi:10.1038/ng.3314 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3314&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26121088&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 22. 22.Löscher W. Critical review of current animal models of seizures and epilepsy used in the discovery and development of new antiepileptic drugs. Seizure. 2011;20(5):359–368. doi:10.1016/j.seizure.2011.01.003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.seizure.2011.01.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21292505&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291295600002&link_type=ISI) 23. 23.Alshehri FF. Integrated virtual screening, molecular modeling and machine learning approaches revealed potential natural inhibitors for epilepsy. Saudi Pharm J. 2023;31(12):101835. doi:10.1016/j.jsps.2023.101835 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jsps.2023.101835&link_type=DOI) 24. 24.Palestro PH, Enrique N, Goicoechea S, et al. Searching for New Leads To Treat Epilepsy: Target-Based Virtual Screening for the Discovery of Anticonvulsant Agents. J Chem Inf Model. 2018;58(7):1331–1342. doi:10.1021/acs.jcim.7b00721 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1021/acs.jcim.7b00721&link_type=DOI) 25. 25.Chauhan A, Singh J, Sangwan N, et al. Designing the 5HT2BR structure and its modulation as a therapeutic target for repurposing approach in drug-resistant epilepsy. Epilepsy Res. 2023;194:107168. doi:10.1016/j.eplepsyres.2023.107168 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.eplepsyres.2023.107168&link_type=DOI) 26. 26.Couyoupetrou M, E. Gantner M, E. Di Ianni M, et al. Computer-Aided Recognition of ABC Transporters Substrates and Its Application to the Development of New Drugs for Refractory Epilepsy. Mini Rev Med Chem. 2017;17(3):205–215. 27. 27.Sadybekov AV, Katritch V. Computational approaches streamlining drug discovery. Nature. 2023;616(7958):673-685. doi:10.1038/s41586-023-05905-z [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-023-05905-z&link_type=DOI) 28. 28.Chong CR, Sullivan DJ. New uses for old drugs. Nature. 2007;448(7154):645-646. doi:10.1038/448645a [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/448645a&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17687303&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248598000020&link_type=ISI) 29. 29.Hodos RA, Kidd BA, Shameer K, Readhead BP, Dudley JT. In silico methods for drug repurposing and pharmacology. WIREs Syst Biol Med. 2016;8(3):186–210. doi:10.1002/wsbm.1337 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/wsbm.1337&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27080087&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 30. 30.He H, Duo H, Hao Y, et al. Computational drug repurposing by exploiting large-scale gene expression data: Strategy, methods and applications. Comput Biol Med. 2023;155:106671. doi:10.1016/j.compbiomed.2023.106671 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.compbiomed.2023.106671&link_type=DOI) 31. 31.Mousavi SZ, Rahmanian M, Sami A. A connectivity map-based drug repurposing study and integrative analysis of transcriptomic profiling of SARS-CoV-2 infection. Infect Genet Evol. 2020;86:104610. doi:10.1016/j.meegid.2020.104610 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.meegid.2020.104610&link_type=DOI) 32. 32.Karatzas E, Kolios G, Spyrou GM. An Application of Computational Drug Repurposing Based on Transcriptomic Signatures. In: Vanhaelen Q, ed. Computational Methods for Drug Repurposing. Springer; 2019:149–177. doi:10.1007/978-1-4939-8955-3_9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/978-1-4939-8955-3_9&link_type=DOI) 33. 33.King EA, Davis JW, Degner JF. Are drug targets with genetic support twice as likely to be approved? Revised estimates of the impact of genetic support for drug mechanisms on the probability of drug approval. PLOS Genet. 2019;15(12):e1008489. doi:10.1371/journal.pgen.1008489 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pgen.1008489&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31830040&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 34. 34.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:10.1002/acn3.703 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/acn3.703&link_type=DOI) 35. 35.Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884–i890. doi:10.1093/bioinformatics/bty560 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/bty560&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30423086&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 36. 36.Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37(8):907–915. doi:10.1038/s41587-019-0201-4 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41587-019-0201-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31375807&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 37. 37.Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33(3):290–295. doi:10.1038/nbt.3122 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nbt.3122&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25690850&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 38. 38.Pertea M, Kim D, Pertea GM, Leek JT, Salzberg SL. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat Protoc. 2016;11(9):1650–1667. doi:10.1038/nprot.2016.095 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nprot.2016.095&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27560171&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 39. 39.Pease LI, Clegg PD, Proctor CJ, Shanley DJ, Cockell SJ, Peffers MJ. Cross platform analysis of transcriptomic data identifies ageing has distinct and opposite effects on tendon in males and females. Sci Rep. 2017;7(1):14443. doi:10.1038/s41598-017-14650-z [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-017-14650-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29089527&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 40. 40.Kuleshov MV, Jones MR, Rouillard AD, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90–97. doi:10.1093/nar/gkw377 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkw377&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27141961&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 41. 41.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:10.1038/s41467-017-01261-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-017-01261-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 42. 42.Kolberg L, Raudvere U, Kuzmin I, Adler P, Vilo J, Peterson H. g:Profiler— interoperable web service for functional enrichment analysis and gene identifier mapping (2023 update). Nucleic Acids Res. 2023;51(W1):W207–W212. doi:10.1093/nar/gkad347 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkad347&link_type=DOI) 43. 43.Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009;37(suppl_2):W305–W311. doi:10.1093/nar/gkp427 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkp427&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19465376&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000267889100054&link_type=ISI) 44. 44.Szklarczyk D, Kirsch R, Koutrouli M, et al. The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 2023;51(D1):D638–D646. doi:10.1093/nar/gkac1000 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkac1000&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=36370105&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 45. 45.Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–2504. doi:10.1101/gr.1239303 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjEwOiIxMy8xMS8yNDk4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDYvMjYvMjAyNC4wNi4yNi4yNDMwOTUxOS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 46. 46.Jiménez-Santos MJ, Nogueira-Rodríguez A, Piñeiro-Yáñez E, et al. PanDrugs2: prioritizing cancer therapies using integrated individual multi-omics data. Nucleic Acids Res. 2023;51(W1):W411–W418. doi:10.1093/nar/gkad412 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkad412&link_type=DOI) 47. 47.Chen YW, Diamante G, Ding J, et al. PharmOmics: A species- and tissue-specific drug signature database and gene-network-based drug repositioning tool. iScience. 2022;25(4). doi:10.1016/j.isci.2022.104052 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.isci.2022.104052&link_type=DOI) 48. 48.Freshour SL, Kiwala S, Cotto KC, et al. Integration of the Drug–Gene Interaction Database (DGIdb 4.0) with open crowdsource efforts. Nucleic Acids Res. 2021;49(D1):D1144–D1151. doi:10.1093/nar/gkaa1084 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkaa1084&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 49. 49.Duan Q, Reid SP, Clark NR, et al. L1000CDS2: LINCS L1000 characteristic direction signatures search engine. Npj Syst Biol Appl. 2016;2(1):1–12. doi:10.1038/npjsba.2016.15 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/npjsba.2016.15&link_type=DOI) 50. 50.Lamb J, Crawford ED, Peck D, et al. The Connectivity Map: Using Gene-Expression Signatures to Connect Small Molecules, Genes, and Disease. Science. 2006;313(5795):1929–1935. doi:10.1126/science.1132939 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzMTMvNTc5NS8xOTI5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDYvMjYvMjAyNC4wNi4yNi4yNDMwOTUxOS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 51. 51.Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7(1):42717. doi:10.1038/srep42717 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/srep42717&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28256516&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 52. 52.Wolking S, Schaeffeler E, Lerche H, Schwab M, Nies AT. Impact of Genetic Polymorphisms of ABCB1 (MDR1, P-Glycoprotein) on Drug Disposition and Potential Clinical Implications: Update of the Literature. Clin Pharmacokinet. 2015;54(7):709–735. doi:10.1007/s40262-015-0267-1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s40262-015-0267-1&link_type=DOI) 53. 53.Stork C, Mathai N, Kirchmair J. Computational prediction of frequent hitters in target-based and cell-based assays. Artif Intell Life Sci. 2021;1:100007. doi:10.1016/j.ailsci.2021.100007 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ailsci.2021.100007&link_type=DOI) 54. 54.Abid F, Al-Ajeeli F, Salim Al-Kawari A, Rasool K. Reversed phase HPLC determination of total homocysteine, cysteine, cysteinyl glycine, glutathione in plasma of epileptic patients. 2023;5:34–45. 55. 55.Ajith A, Mondal S, Chattopadhyay S, et al. Mass Spectrometry Imaging Deciphers Dysregulated Lipid Metabolism in the Human Hippocampus Affected by Temporal Lobe Epilepsy. ACS Chem Neurosci. 2021;12(21):4187–4194. doi:10.1021/acschemneuro.1c00587 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1021/acschemneuro.1c00587&link_type=DOI) 56. 56.Poirier K, Hubert L, Viot G, et al. *CSNK2B* splice site mutations in patients cause intellectual disability with or without myoclonic epilepsy. Hum Mutat. 2017;38(8):932–941. doi:10.1002/humu.23270 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/humu.23270&link_type=DOI) 57. 57.Conboy K, Henshall DC, Brennan GP. Epigenetic principles underlying epileptogenesis and epilepsy syndromes. Neurobiol Dis. 2021;148:105179. doi:10.1016/j.nbd.2020.105179 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.nbd.2020.105179&link_type=DOI) 58. 58.Hauser RM, Henshall DC, Lubin FD. The Epigenetics of Epilepsy and Its Progression. The Neuroscientist. 2018;24(2):186–200. doi:10.1177/1073858417705840 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/1073858417705840&link_type=DOI) 59. 59.Kobow K, Blümcke I. The emerging role of DNA methylation in epileptogenesis. Epilepsia. 2012;53 Suppl 9:11–20. doi:10.1111/epi.12031 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/epi.12031&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23216575&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 60. 60.Hodges SL, Lugo JN. Therapeutic role of targeting mTOR signaling and neuroinflammation in epilepsy. Epilepsy Res. 2020;161:106282. doi:10.1016/j.eplepsyres.2020.106282 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.eplepsyres.2020.106282&link_type=DOI) 61. 61.Mills JD, van Vliet EA, Chen BJ, et al. Coding and non-coding transcriptome of mesial temporal lobe epilepsy: Critical role of small non-coding RNAs. Neurobiol Dis. 2020;134:104612. doi:10.1016/j.nbd.2019.104612 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.nbd.2019.104612&link_type=DOI) 62. 62.Volmering E, Niehusmann P, Peeva V, et al. Neuropathological signs of inflammation correlate with mitochondrial DNA deletions in mesial temporal lobe epilepsy. Acta Neuropathol (Berl*)*. 2016;132(2):277–288. doi:10.1007/s00401-016-1561-1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00401-016-1561-1&link_type=DOI) 63. 63.Lebrun N, Lebon S, Jeannet PY, Jacquemont S, Billuart P, Bienvenu T. Early-onset encephalopathy with epilepsy associated with a novel splice site mutation in SMC1A. Am J Med Genet A. 2015;167A(12):3076–3081. doi:10.1002/ajmg.a.37364 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ajmg.a.37364&link_type=DOI) 64. 64.Lalwani AM, Yilmaz A, Bisgin H, Ugur Z, Akyol S, Graham SF. The Biochemical Profile of Post-Mortem Brain from People Who Suffered from Epilepsy Reveals Novel Insights into the Etiopathogenesis of the Disease. Metabolites. 2020;10(6):261. doi:10.3390/metabo10060261 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/metabo10060261&link_type=DOI) 65. 65.Duan W, Chen Y, Wang XR. MicroRNALJ155 contributes to the occurrence of epilepsy through the PI3K/Akt/mTOR signaling pathway. Int J Mol Med. 2018;42(3):1577–1584. doi:10.3892/ijmm.2018.3711 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3892/IJMM.2018.3711&link_type=DOI) 66. 66.Bando SY, Alegro MC, Amaro E, et al. Hippocampal CA3 transcriptome signature correlates with initial precipitating injury in refractory mesial temporal lobe epilepsy. PloS One. 2011;6(10):e26268. doi:10.1371/journal.pone.0026268 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0026268&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22022585&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 67. 67.Bando SY, Silva FN, Costa L da F, et al. Complex network analysis of CA3 transcriptome reveals pathogenic and compensatory pathways in refractory temporal lobe epilepsy. PloS One. 2013;8(11):e79913. doi:10.1371/journal.pone.0079913 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0079913&link_type=DOI) 68. 68.Patterson KP, Baram TZ, Shinnar S. Origins of Temporal Lobe Epilepsy: Febrile Seizures and Febrile Status Epilepticus. Neurotherapeutics. 2014;11(2):242–250. doi:10.1007/s13311-014-0263-4 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s13311-014-0263-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24604424&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 69. 69.Arion D, Sabatini M, Unger T, et al. Correlation of transcriptome profile with electrical activity in temporal lobe epilepsy. Neurobiol Dis. 2006;22(2):374–387. doi:10.1016/j.nbd.2005.12.012 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.nbd.2005.12.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16480884&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000237378000017&link_type=ISI) 70. 70.Srinivasan M, Sedmak D, Jewell S. Effect of Fixatives and Tissue Processing on the Content and Integrity of Nucleic Acids. Am J Pathol. 2002;161(6):1961–1971. doi:10.1016/S0002-9440(10)64472-0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0002-9440(10)64472-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12466110&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000179663400001&link_type=ISI) 71. 71.Mirza N, Stevelink R, Taweel B, Koeleman BPC, Marson AG, International League Against Epilepsy Consortium on Complex Epilepsies. Using common genetic variants to find drugs for common epilepsies. Brain Commun. 2021;3(4):fcab287. doi:10.1093/braincomms/fcab287 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/braincomms/fcab287&link_type=DOI) 72. 72.Löscher W, Friedman A. Structural, Molecular, and Functional Alterations of the Blood-Brain Barrier during Epileptogenesis and Epilepsy: A Cause, Consequence, or Both? Int J Mol Sci. 2020;21(2):E591. doi:10.3390/ijms21020591 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/ijms21020591&link_type=DOI) 73. 73.Hequet O, Dumontet C, El Jaafari-Corbin A, et al. Epileptic seizures after autologous peripheral blood progenitor infusion in a patient treated with high-dose chemotherapy for myeloma. Bone Marrow Transplant. 2002;29(6):544–544. doi:10.1038/sj.bmt.1703383 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.bmt.1703383&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11960281&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000175018500019&link_type=ISI) 74. 74.Sasaki K, Yokota Y, Isojima T, et al. Enteral lorlatinib after alectinib as a treatment option in anaplastic lymphoma kinase-positive non-small cell lung cancer with triple problems: carcinomatous meningitis, poor performance status, and dysphagia—a case report. Respirol Case Rep. 2021;9(7):e00796. doi:10.1002/rcr2.796 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/rcr2.796&link_type=DOI) 75. 75.Kreins AY, Bonfanti P, Davies EG. Current and Future Therapeutic Approaches for Thymic Stromal Cell Defects. Front Immunol. 2021;12:655354. doi:10.3389/fimmu.2021.655354 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2021.655354&link_type=DOI) 76. 76.Brown N, McBain C, Nash S, et al. Multi-Center Randomized Phase II Study Comparing Cediranib plus Gefitinib with Cediranib plus Placebo in Subjects with Recurrent/Progressive Glioblastoma. PLOS ONE. 2016;11(5):e0156369. doi:10.1371/journal.pone.0156369 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0156369&link_type=DOI) 77. 77.Berdichevsky Y, Saponjian Y, Park KI, et al. Staged anticonvulsant screening for chronic epilepsy. Ann Clin Transl Neurol. 2016;3(12):908–923. doi:10.1002/acn3.364 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/acn3.364&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28097203&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 78. 78.Kandemir M, Kucukkaya B, Tepe MS, Yalciner ZB, Salepci NT. Reversible Posterior Leukoencephalopathy Syndrome Due to Carboplatin and Paclitaxel Therapy. Balk Med J. 2015;32(4):421–425. doi:10.5152/balkanmedj.2015.15487 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.5152/balkanmedj.2015.15487&link_type=DOI) 79. 79.García-Muñoz C, Rodríguez-Quesada PP, Ferrari-Piquero JM. Use of oral antineoplastic in special situations in a third level hospital: real life results. Farm Hosp. 2018;42(1):5–9. doi:10.7399/fh.10856 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7399/fh.10856&link_type=DOI) 80. 80.Perucca E. Optimizing antiepileptic drug treatment in tumoral epilepsy. Epilepsia. 2013;54(s9):97–104. doi:10.1111/epi.12452 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/epi.12452&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24328881&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 81. 81.Younus I, Reddy DS. Epigenetic interventions for epileptogenesis: A new frontier for curing epilepsy. Pharmacol Ther. 2017;177:108–122. doi:10.1016/j.pharmthera.2017.03.002 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pharmthera.2017.03.002&link_type=DOI) 82. 82.Kobow K, Blümcke I. The methylation hypothesis: Do epigenetic chromatin modifications play a role in epileptogenesis? Epilepsia. 2011;52(s4):15–19. doi:10.1111/j.1528-1167.2011.03145.x [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1528-1167.2011.03145.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21732935&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 83. 83.Ichida M, Gomi A, Hiranouchi N, et al. A case of cerebral endometriosis causing catamenial epilepsy. Neurology. 1993;43(12):2708–2708. doi:10.1212/WNL.43.12.2708 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.43.12.2708&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8255485&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F26%2F2024.06.26.24309519.atom) 84. 84.Mekhaeil M, Dev KK, Conroy MJ. Existing Evidence for the Repurposing of PARP-1 Inhibitors in Rare Demyelinating Diseases. Cancers. 2022;14(3):687. doi:10.3390/cancers14030687 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/cancers14030687&link_type=DOI) 85. 85.Ayuso-Muñoz A, Prieto-Santamaría L, Álverez-Pérez A, Otero-Carrasco B, Serrano E, Rodríguez-González A. Enhancing drug repurposing on graphs by integrating drug molecular structure as feature. IEEE. Published online 2023:192–197. 86. 86.Asadi-Pooya AA, Malekpour M, Zamiri B, Kashkooli M, Firouzabadi N. FKBP5 blockade may provide a new horizon for the treatment of stress-associated disorders: An in-silico study. Epilepsia Open. 2023;8(2):633–640. doi:10.1002/epi4.12749 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/epi4.12749&link_type=DOI) 87. 87.Zhang Y, Zhang M, Zhu W, et al. Role of Elevated Thrombospondin-1 in Kainic Acid-Induced Status Epilepticus. Neurosci Bull. 2020;36(3):263–276. doi:10.1007/s12264-019-00437-x [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12264-019-00437-x&link_type=DOI) 88. 88.Verrotti A, Tambucci R, Di Francesco L, et al. The role of polytherapy in the management of epilepsy: suggestions for rational antiepileptic drug selection. Expert Rev Neurother. 2020;20(2):167–173. doi:10.1080/14737175.2020.1707668 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/14737175.2020.1707668&link_type=DOI) 89. 89.Wright C, Downing J, Mungall D, et al. Clinical pharmacology and pharmacokinetics of levetiracetam. Front Neurol. 2013;4:192. doi:10.3389/fneur.2013.00192 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fneur.2013.00192&link_type=DOI) 90. 90.Löscher W, Potschka H, Sisodiya SM, Vezzani A. Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options. Pharmacol Rev. 2020;72(3):606–638. doi:10.1124/pr.120.019539 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoicGhhcm1yZXYiO3M6NToicmVzaWQiO3M6ODoiNzIvMy82MDYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wNi8yNi8yMDI0LjA2LjI2LjI0MzA5NTE5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 91. 91.Wolking S, Schaeffeler E, Lerche H, Schwab M, Nies AT. Impact of Genetic Polymorphisms of ABCB1 (MDR1, P-Glycoprotein) on Drug Disposition and Potential Clinical Implications: Update of the Literature. Clin Pharmacokinet. 2015;54(7):709–735. doi:10.1007/s40262-015-0267-1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s40262-015-0267-1&link_type=DOI) 92. 92.Sanz P, Serratosa JM, Sánchez MP. Beneficial Effects of Metformin on the Central Nervous System, with a Focus on Epilepsy and Lafora Disease. Int J Mol Sci. 2021;22(10):5351. doi:10.3390/ijms22105351 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/ijms22105351&link_type=DOI)