Drug Repositioning

Drug repurposing screen identifies vidofludimus calcium and pyrazofurin as novel chemical entities for the development of hepatitis E interventions

Mon, 2023-11-20 06:00

Virol Sin. 2023 Nov 18:S1995-820X(23)00138-4. doi: 10.1016/j.virs.2023.11.006. Online ahead of print.

ABSTRACT

Hepatitis E virus (HEV) infection can cause severe complications and high mortality, particularly in pregnant women, organ transplant recipients, individuals with pre-existing liver disease and immunosuppressed patients. However, there are still unmet needs for treating chronic HEV infections. Herein, we screened a best-in-class drug repurposing library consisting of 262 drugs/compounds. Upon screening, we identified vidofludimus calcium and pyrazofurin as novel anti-HEV entities. Vidofludimus calcium is the next-generation dihydroorotate dehydrogenase (DHODH) inhibitor in the phase 3 pipeline to treat autoimmune diseases or SARS-CoV-2 infection. Pyrazofurin selectively targets uridine monophosphate synthetase (UMPS). Their anti-HEV effects were further investigated in a range of cell culture models and human liver organoids models with wild type HEV strains and ribavirin treatment failure-associated HEV strains. Encouragingly, both drugs exhibited a sizeable therapeutic window against HEV. For instance, the IC50 value of vidofludimus calcium is 4.6-7.6-fold lower than the current therapeutic doses in patients. Mechanistically, their anti-HEV mode of action depends on the blockage of pyrimidine synthesis. Notably, two drugs robustly inhibited ribavirin treatment failure-associated HEV mutants (Y1320H, G1634R). Their combination with IFN-α resulted in synergistic antiviral activity. In conclusion, we identified vidofludimus calcium and pyrazofurin as potent candidates for the treatment of HEV infections. Based on their antiviral potency, and also the favorable safety profile identified in clinical studies, our study supports the initiation of clinical studies to repurpose these drugs for treating chronic hepatitis E.

PMID:37984761 | DOI:10.1016/j.virs.2023.11.006

Categories: Literature Watch

Discovery of New <em>Fusobacterium nucleatum</em> Inhibitors to Attenuate Migratory Capability of Colon Cancer Cells by the Drug Repositioning Strategy

Mon, 2023-11-20 06:00

J Med Chem. 2023 Nov 20. doi: 10.1021/acs.jmedchem.3c00281. Online ahead of print.

ABSTRACT

Recent studies revealed that intestinal microbiota played important roles in colorectal cancer (CRC) carcinogenesis. Particularly, Fusobacterium nucleatum was confirmed to promote the proliferation and metastasis of CRC. Therefore, targeting F. nucleatum may be a potential preventive and therapeutic approach for CRC. Herein, 2,272 off-patent drugs were screened inhibitory activity against F. nucleatum. Among the hits, nitisinone was identified as a promising anti-F. nucleatum lead compound. Further optimization of nitisinone led to the discovery of more potent derivatives. Particularly, compounds 19q and 22c showed potent anti-F. nucleatum activity (MIC50 = 1 and 2 μg/mL, respectively) with low cytotoxicity. Among them, compound 19q effectively attenuated the migratory ability of MC-38 cells induced by F. nucleatum. Preliminary mechanism studies suggested that nitisinone and its derivatives might act by downregulating nitroreductase and tryptophanase. Thus, the development of small molecule F. nucleatum inhibitors represents an effective strategy to treat CRC.

PMID:37983010 | DOI:10.1021/acs.jmedchem.3c00281

Categories: Literature Watch

Drug repurposing of rimantadine for treatment of cancer

Mon, 2023-11-20 06:00

Pharm Pat Anal. 2023 Nov 20. doi: 10.4155/ppa-2023-0019. Online ahead of print.

ABSTRACT

Repurposing of approved drugs allows strong savings in time and investment. Rimantadine is an FDA-approved drug for prevention and treatment of influenza A infection. Patent US2021330605 describes the use of rimantadine, an adamantane derivative, for the treatment of melanoma, breast cancer and head and neck squamous cell carcinoma. Rimantadine inhibited proliferation of cell lines of melanoma, breast cancer, and head and neck squamous cell carcinoma, increased the survival of mice injected with cancer cell lines and restores the expression of MHC class I. Rimantadine has the potential to be used successfully in the treatment of head and neck squamous cell carcinoma.

PMID:37982658 | DOI:10.4155/ppa-2023-0019

Categories: Literature Watch

Novel amodiaquine analogues to treat cervical cancer and microbial infection in the future

Mon, 2023-11-20 06:00

Future Med Chem. 2023 Nov 20. doi: 10.4155/fmc-2023-0245. Online ahead of print.

ABSTRACT

Aim: To synthesize and explore the therapeutic potential of amodiaquine analogues. Methodology: New promising analogues were synthesized by nucleophilic substitution at the 4-amino position and were characterized using 1H NMR, 13C NMR and FT-IR spectroscopic techniques. Results: Antibacterial and cytotoxic screening revealed the high potency of these compounds; analogue AS1 had a 34.3 ± 0.18 mm zone of inhibition against Pseudomonas aeruginosa. Excellent activity against fungal strains, that is, Candida albicans (39.6 ± 0.23 mm) was shown by analogue AS2. Analogue AS1 had a IC50 = 4.2 μg/ml against the HeLa cell line (cervical cancer) and binding energy against 5GWK (-8.32688 kcal/mol), 1PFK (-6.4780 kcal/mol) and 1TUP (-6.5279 kcal/mol) in the docking study. Conclusion: The obtained results reveal that these analogues exhibit potent antimicrobial and cytotoxic potential.

PMID:37982232 | DOI:10.4155/fmc-2023-0245

Categories: Literature Watch

Pangenome diversification and resistance gene characterization in Salmonella Typhi prioritized RfaJ as a significant therapeutic marker

Fri, 2023-11-17 06:00

J Genet Eng Biotechnol. 2023 Nov 17;21(1):125. doi: 10.1186/s43141-023-00591-w.

ABSTRACT

BACKGROUND: Salmonella Typhi stands as the etiological agent responsible for the onset of human typhoid fever. The pressing demand for innovative therapeutic targets against S. Typhi is underscored by the escalating prevalence of this pathogen and the severe nature of its infections. Consequently, this study employs pangenome analysis to scrutinize 119 S. Typhi-resistant strains, aiming to identify the most promising therapeutic targets originating from its core genome.

RESULTS: Subtractive genomics was employed to systematically eliminate non-homologous (n=1147), essential (n=551), drug-like (n=80), and pathogenicity-related (n=18) proteins from the initial pool of 3351 core genome proteins. Consequently, lipopolysaccharide 1,2-glucosyltransferase RfaJ was designated as the optimal pharmacological target due to its potential versatility. Furthermore, a compendium of 9000 FDA-approved compounds was repurposed for evaluation against the RfaJ drug target, with the specific intent of prioritizing novel, high-potency therapeutic candidates for combating S. Typhi. Ultimately, four compounds, namely DB00549 (Zafirlukast), DB15637 (Fluzoparib), DB15688 (Zavegepant), and DB12411 (Bemcentinib), were singled out as potential inhibitors based on the ligand-protein binding affinity (indicated by the lowest anticipated binding energy) and the overall stability of these compounds. Notably, molecular dynamics simulations, conducted over a 50 nanosecond interval, convincingly demonstrated the stability of these compounds in the context of the RfaJ protein.

CONCLUSION: In summary, the present findings hold significant promise as an initial stride in the broader drug discovery endeavor against S. Typhi infections. However, the experimental validation of the identified drug target and drug candidate is further required to increase the effectiveness of the applied methodology.

PMID:37975995 | DOI:10.1186/s43141-023-00591-w

Categories: Literature Watch

Unveiling the anti-glioma potential of a marine derivative, Fucoidan: its synergistic cytotoxicity with Temozolomide-an in vitro and in silico experimental study

Fri, 2023-11-17 06:00

3 Biotech. 2023 Dec;13(12):397. doi: 10.1007/s13205-023-03814-6. Epub 2023 Nov 14.

ABSTRACT

Glioma coined as a "butterfly" tumor associated with a dismal prognosis. Marine algal compounds with the richest sources of bioactive components act as significant anti-tumor therapeutics. However, there is a paucity of studies conducted on Fucoidan to enhance the anti-glioma efficacy of Temozolomide. Therefore, the present study aimed to evaluate the synergistic anti-proliferative, anti-inflammatory and pro-apoptotic effects of Fucoidan with Temozolomide in in vitro and in silico experimental setup. The anti-proliferative effects of Temozolomide and Fucoidan were evaluated on C6 glioma cells by MTT and migration assay. Modulation of inflammatory markers and apoptosis induction was affirmed at the morphological and transcriptional level by dual staining and gene expression. Molecular docking (MD) and molecular dynamics simulation (MDS) studies were performed against the targets to rationalize the inhibitory effect. The dual-drug combination significantly reduced the cell viability and migration of glioma cells in a synergistic dose-dependent manner. At the molecular level, the dual-drug combination significantly down-regulated inflammatory genes with a concomitant upregulation of pro-apoptotic marker. In consensus with our in vitro findings, molecular docking and simulation studies revealed that the anti-tumor ligands: Temozolomide, Fucoidan with 5-(3-Methy1-trizeno)-imidazole-4-carboxamide (MTIC), and 4-amino-5-imidazole-carboxamide (AIC) had the potency to bind to the inflammatory proteins at their active sites, mediated by H-bonds and other non-covalent interactions. The dual-drug combinatorial treatment synergistically inhibited the proliferation, migration of glioma cells and promoted apoptosis; conversely with the down-regulation of inflammatory genes. However, pre-clinical experimental evidence is warranted for the possible translation of this combination.

SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s13205-023-03814-6.

PMID:37974928 | PMC:PMC10645720 | DOI:10.1007/s13205-023-03814-6

Categories: Literature Watch

Correction to: Drug repurposing using computational methods to identify therapeutic options for COVID-19

Thu, 2023-11-16 06:00

J Diabetes Metab Disord. 2023 Jul 10;22(2):1815. doi: 10.1007/s40200-023-01248-8. eCollection 2023 Dec.

ABSTRACT

[This corrects the article DOI: 10.1007/s40200-020-00546-9.].

PMID:37969922 | PMC:PMC10638279 | DOI:10.1007/s40200-023-01248-8

Categories: Literature Watch

Changing human hair fibre colour and shape from the follicle

Wed, 2023-11-15 06:00

J Adv Res. 2023 Nov 13:S2090-1232(23)00350-8. doi: 10.1016/j.jare.2023.11.013. Online ahead of print.

ABSTRACT

INTRODUCTION: Natural hair curvature and colour are genetically determined human traits, that we intentionally change by applying thermal and chemical treatments to the fibre. Presently, those cosmetic methodologies act externally and their recurrent use is quite detrimental to hair fibre quality and even to our health.

OBJECTIVES: This work represents a disruptive concept to modify natural hair colour and curvature. We aim to model the fibre phenotype as it is actively produced in the follicle through the topical delivery of specific bioactive molecules to the scalp.

METHODS: Transcriptome differences between curly and straight hairs were identified by microarray. In scalp samples, the most variable transcripts were mapped by in situ hybridization. Then, by using appropriate cellular models, we screened a chemical library of 1200 generic drugs, searching for molecules that could lead to changes in either fibre colour or curvature. A pilot-scale, single-centre, investigator-initiated, prospective, blind, bilateral (split-scalp) placebo-controlled clinical study with the intervention of cosmetics was conducted to obtain a proof of concept (RNEC n.92938).

RESULTS: We found 85 genes transcribed significantly different between curly and straight hair, not previously associated with this human trait. Next, we mapped some of the most variable genes to the inner root sheath of follicles, reinforcing the role of this cell layer in fibre shape moulding. From the drug library screening, we selected 3 and 4 hits as modulators of melanin synthesis and gene transcription, respectively, to be further tested in 33 volunteers. The intentional specific hair change occurred: 8 of 14 volunteers exhibited colour changes, and 16 of 19 volunteers presented curvature modifications, by the end of the study.

CONCLUSION: The promising results obtained are the first step towards future cosmetics, complementary or alternative to current methodologies, taking hair styling to a new level: changing hair from the inside out.

PMID:37967812 | DOI:10.1016/j.jare.2023.11.013

Categories: Literature Watch

Integrated genomic network analysis revealed potential of a druggable target for hemorrhoid treatment

Wed, 2023-11-15 06:00

Saudi Pharm J. 2023 Dec;31(12):101831. doi: 10.1016/j.jsps.2023.101831. Epub 2023 Oct 20.

ABSTRACT

Hemorrhoids are a prevalent medical condition that necessitates effective treatment options. The current options for treatment consist of oral medications, topical applications, or surgery, yet a scarcity of highly effective drugs still exists. Genetic markers provide promising avenues for investigating the treatment of hemorrhoids, as they may reveal intricate biological mechanisms and targeted drug therapies, ultimately enhancing more precise treatment tailored to the patient. This study aims to identify new drug candidates for treating hemorrhoids through a meticulous bioinformatics approach and integrated with genomic network analysis. After extracting 21 druggable target genes using DrugBank from 293 genes connected to hemorrhoids, 87 possible drugs were selected. Three of these drugs (ketamine, methylene blue, and fulvestrant) hold potential in addressing issues associated with hemorrhoids and have been supported by clinical or preclinical studies. Eighty-four compounds present new therapeutic possibilities for managing hemorrhoids. We highlight that our findings indicate that NOX1 and NOS3 genes are promising biomarkers, with NOS3 gaining significance owing to its robust systemic functional annotations. Sapropterin, an existing drug, is closely associated with NOS3, providing a clear target for biomarker-driven interventions. This study illustrates the potential of combining genomic network analysis with bioinformatics to repurpose drugs for treating hemorrhoids. Subsequent research will explore the mechanisms for utilizing NOS3 targeting in the treatment of hemorrhoids.

PMID:37965490 | PMC:PMC10641558 | DOI:10.1016/j.jsps.2023.101831

Categories: Literature Watch

Exploring disease-causing traits for drug repurposing in critically ill COVID-19 patients: A causal inference approach

Wed, 2023-11-15 06:00

iScience. 2023 Oct 12;26(11):108185. doi: 10.1016/j.isci.2023.108185. eCollection 2023 Nov 17.

ABSTRACT

Despite recent development of vaccines to prevent SARS-CoV-2 infection, treatment of critically ill COVID-19 patients remains an important goal. In principle, genome-wide association studies (GWASs) provide a shortcut to the clinical evidence needed to repurpose existing drugs; however, genes identified frequently lack a causal disease link. We report an alternative method for finding drug repurposing targets, focusing on disease-causing traits beyond immediate disease genetics. Sixty blood cell types and biochemistries, and body mass index, were screened on a cohort of critically ill COVID-19 cases and controls that exhibited mild symptoms after infection, yielding high neutrophil cell count as a possible causal trait for critical illness. Our methodology identified CDK6 and janus kinase (JAK) inhibitors as treatment targets that were validated in an ex vivo neutrophil extracellular trap (NET) formation assay. Our methodology demonstrates the increased power for drug target identification by leveraging large disease-causing trait datasets.

PMID:37965141 | PMC:PMC10641251 | DOI:10.1016/j.isci.2023.108185

Categories: Literature Watch

The interplay of drug therapeutics and immune responses to SARS-CoV-2

Tue, 2023-11-14 06:00

Cell Mol Immunol. 2023 Nov 14. doi: 10.1038/s41423-023-01098-7. Online ahead of print.

ABSTRACT

The SARS-CoV-2 pandemic has necessitated rapid therapeutic and preventative responses. While vaccines form the frontline of defense, antiviral treatments such as nirmatrelvir have emerged as vital adjunctive measures, particularly for those unable or unwilling to be vaccinated. This review delves into the potential influence of nirmatrelvir on enduring immunity. In parallel, the potential of drug repurposing is explored, with bisphosphonates being examined for their possible effects against COVID-19 due to their immunomodulatory properties. The importance of rigorous clinical trials and careful interpretation of preliminary data is emphasized.

PMID:37964122 | DOI:10.1038/s41423-023-01098-7

Categories: Literature Watch

Drug Repurposing for Cancers With Limited Survival: Protocol for a Retrospective Cohort Study

Tue, 2023-11-14 06:00

JMIR Res Protoc. 2023 Nov 14;12:e48925. doi: 10.2196/48925.

ABSTRACT

BACKGROUND: Only 5% of the molecules tested in oncology phase 1 trials reach the market after an average of 7.5 years of waiting and at a cost of tens of millions of dollars. To reduce the cost and shorten the time of discovery of new treatments, "drug repurposing" (research with molecules already approved for another indication) and the use of secondary data (not collected for the purpose of research) have been proposed. Due to advances in informatics in clinical care, secondary data can, in some cases, be of equal quality to primary data generated through prospective studies.

OBJECTIVE: The objective of this study is to identify drugs currently marketed for other indications that may have an effect on the prognosis of patients with cancer.

METHODS: We plan to monitor a cohort of patients with high-lethality cancers treated in the public health system of Catalonia between 2006 and 2012, retrospectively, for survival for 5 years after diagnosis or until death. A control cohort, comprising people without cancer, will also be retrospectively monitored for 5 years. The following study variables will be extracted from different population databases: type of cancer (patients with cancer cohort), date and cause of death, pharmacological treatment, sex, age, and place of residence. During the first stage of statistical analysis of the patients with cancer cohort, the drugs consumed by the long-term survivors (alive at 5 years) will be compared with those consumed by nonsurvivors. In the second stage, the survival associated with the consumption of each relevant drug will be analyzed. For the analyses, groups will be matched for potentially confounding variables, and multivariate analyses will be performed to adjust for residual confounding variables if necessary. The control cohort will be used to verify whether the associations found are exclusive to patients with cancer or whether they also occur in patients without cancer.

RESULTS: We anticipate discovering multiple significant associations between commonly used drugs and the survival outcomes of patients with cancer. We expect to publish the initial results in the first half of 2024.

CONCLUSIONS: This retrospective study may identify several commonly used drugs as candidates for repurposing in the treatment of various cancers. All analyses are considered exploratory; therefore, the results will have to be confirmed in subsequent clinical trials. However, the results of this study may accelerate drug discovery in oncology.

INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/48925.

PMID:37962929 | DOI:10.2196/48925

Categories: Literature Watch

Correction to Network module-based drug repositioning for pulmonary arterial hypertension

Tue, 2023-11-14 06:00

CPT Pharmacometrics Syst Pharmacol. 2023 Nov 14. doi: 10.1002/psp4.13080. Online ahead of print.

NO ABSTRACT

PMID:37962018 | DOI:10.1002/psp4.13080

Categories: Literature Watch

Modeling Path Importance for Effective Alzheimer's Disease Drug Repurposing

Tue, 2023-11-14 06:00

ArXiv. 2023 Oct 27:arXiv:2310.15211v2. Preprint.

ABSTRACT

Recently, drug repurposing has emerged as an effective and resource-efficient paradigm for AD drug discovery. Among various methods for drug repurposing, network-based methods have shown promising results as they are capable of leveraging complex networks that integrate multiple interaction types, such as protein-protein interactions, to more effectively identify candidate drugs. However, existing approaches typically assume paths of the same length in the network have equal importance in identifying the therapeutic effect of drugs. Other domains have found that same length paths do not necessarily have the same importance. Thus, relying on this assumption may be deleterious to drug repurposing attempts. In this work, we propose MPI (Modeling Path Importance), a novel network-based method for AD drug repurposing. MPI is unique in that it prioritizes important paths via learned node embeddings, which can effectively capture a network's rich structural information. Thus, leveraging learned embeddings allows MPI to effectively differentiate the importance among paths. We evaluate MPI against a commonly used baseline method that identifies anti-AD drug candidates primarily based on the shortest paths between drugs and AD in the network. We observe that among the top-50 ranked drugs, MPI prioritizes 20.0% more drugs with anti-AD evidence compared to the baseline. Finally, Cox proportional-hazard models produced from insurance claims data aid us in identifying the use of etodolac, nicotine, and BBB-crossing ACE-INHs as having a reduced risk of AD, suggesting such drugs may be viable candidates for repurposing and should be explored further in future studies.

PMID:37961739 | PMC:PMC10635281

Categories: Literature Watch

Colloidal aggregation confounds cell-based Covid-19 antiviral screens

Tue, 2023-11-14 06:00

bioRxiv. 2023 Oct 30:2023.10.27.564435. doi: 10.1101/2023.10.27.564435. Preprint.

ABSTRACT

Colloidal aggregation is one of the largest contributors to false-positives in early drug discovery and chemical biology. Much work has focused on its impact on pure-protein screens; here we consider aggregations role in cell-based infectivity assays in Covid-19 drug repurposing. We began by investigating the potential aggregation of 41 drug candidates reported as SARs-CoV-2 entry inhibitors. Of these, 17 formed colloidal-particles by dynamic light scattering and exhibited detergent-dependent enzyme inhibition. To evaluate antiviral efficacy of the drugs in cells we used spike pseudotyped lentivirus and pre-saturation of the colloids with BSA. The antiviral potency of the aggregators was diminished by at least 10-fold and often entirely eliminated in the presence of BSA, suggesting antiviral activity can be attributed to the non-specific nature of the colloids. In confocal microscopy, the aggregates induced fluorescent puncta of labeled spike protein, consistent with sequestration of the protein on the colloidal particles. Addition of either non-ionic detergent or of BSA disrupted these puncta. These observations suggest that colloidal aggregation is common among cell-based anti-viral drug repurposing, and perhaps cell-based assays more broadly, and offers rapid counter-screens to detect and eliminate these artifacts, allowing the community invest resources in compounds with true potential as a Covid-19 therapeutic.

PMID:37961552 | PMC:PMC10634915 | DOI:10.1101/2023.10.27.564435

Categories: Literature Watch

Integrating population-level and cell-based signatures for drug repositioning

Tue, 2023-11-14 06:00

bioRxiv. 2023 Oct 26:2023.10.25.564079. doi: 10.1101/2023.10.25.564079. Preprint.

ABSTRACT

Drug repositioning presents a streamlined and cost-efficient way to expand the range of therapeutic possibilities. Furthermore, drugs with genetic evidence are more likely to progress successfully through clinical trials towards FDA approval. Exploiting these developments, single gene-based drug repositioning methods have been implemented, but approaches leveraging the entire spectrum of molecular signatures are critically underexplored. Most multi-gene-based approaches rely on differential gene expression (DGE) analysis, which is prone to identify the molecular consequence of disease and renders causal inference challenging. We propose a framework TReD (Transcriptome-informed Reversal Distance) that integrates population-level disease signatures robust to reverse causality and cell-based drug-induced transcriptome response profiles. TReD embeds the disease signature and drug profile in a high-dimensional normed space, quantifying the reversal potential of candidate drugs in a disease-related cell screen assay. The robustness is ensured by evaluation in additional cell screens. For an application, we implement the framework to identify potential drugs against COVID-19. Taking transcriptome-wide association study (TWAS) results from four relevant tissues and three DGE results as disease features, we identify 37 drugs showing potential reversal roles in at least four of the seven disease signatures. Notably, over 70% (27/37) of the drugs have been linked to COVID-19 from other studies, and among them, eight drugs are supported by ongoing/completed clinical trials. For example, TReD identifies the well-studied JAK1/JAK2 inhibitor baricitinib, the first FDA-approved immunomodulatory treatment for COVID-19. Novel potential candidates, including enzastaurin, a selective inhibitor of PKC-beta which can be activated by SARS-CoV-2, are also identified. In summary, we propose a comprehensive genetics-anchored framework integrating population-level signatures and cell-based screens that can accelerate the search for new therapeutic strategies.

PMID:37961219 | PMC:PMC10634827 | DOI:10.1101/2023.10.25.564079

Categories: Literature Watch

Nonspecific membrane bilayer perturbations by ivermectin underlie SARS-CoV-2 in vitro activity

Tue, 2023-11-14 06:00

bioRxiv. 2023 Oct 24:2023.10.23.563088. doi: 10.1101/2023.10.23.563088. Preprint.

ABSTRACT

Since it was proposed as a potential host-directed antiviral agent for SARS-CoV-2, the antiparasitic drug ivermectin has been investigated thoroughly in clinical trials, which have provided insufficient support for its clinical efficacy. To examine the potential for ivermectin to be repurposed as an antiviral agent, we therefore undertook a series of preclinical studies. Consistent with early reports, ivermectin decreased SARS-CoV-2 viral burden in in vitro models at low micromolar concentrations, five- to ten-fold higher than the reported toxic clinical concentration. At similar concentrations, ivermectin also decreased cell viability and increased biomarkers of cytotoxicity and apoptosis. Further mechanistic and profiling studies revealed that ivermectin nonspecifically perturbs membrane bilayers at the same concentrations where it decreases the SARS-CoV-2 viral burden, resulting in nonspecific modulation of membrane-based targets such as G-protein coupled receptors and ion channels. These results suggest that a primary molecular mechanism for the in vitro antiviral activity of ivermectin may be nonspecific membrane perturbation, indicating that ivermectin is unlikely to be translatable into a safe and effective antiviral agent. These results and experimental workflow provide a useful paradigm for performing preclinical studies on (pandemic-related) drug repurposing candidates.

PMID:37961094 | PMC:PMC10634736 | DOI:10.1101/2023.10.23.563088

Categories: Literature Watch

Attention Mechanism-Based Graph Neural Network Model for Effective Activity Prediction of SARS-CoV-2 Main Protease Inhibitors: Application to Drug Repurposing as Potential COVID-19 Therapy

Tue, 2023-11-14 06:00

J Chem Inf Model. 2023 Nov 13. doi: 10.1021/acs.jcim.3c01280. Online ahead of print.

ABSTRACT

Compared to de novo drug discovery, drug repurposing provides a time-efficient way to treat coronavirus disease 19 (COVID-19) that is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 main protease (Mpro) has been proved to be an attractive drug target due to its pivotal involvement in viral replication and transcription. Here, we present a graph neural network-based deep-learning (DL) strategy to prioritize the existing drugs for their potential therapeutic effects against SARS-CoV-2 Mpro. Mpro inhibitors were represented as molecular graphs ready for graph attention network (GAT) and graph isomorphism network (GIN) modeling for predicting the inhibitory activities. The result shows that the GAT model outperforms the GIN and other competitive models and yields satisfactory predictions for unseen Mpro inhibitors, confirming its robustness and generalization. The attention mechanism of GAT enables to capture the dominant substructures and thus to realize the interpretability of the model. Finally, we applied the optimal GAT model in conjunction with molecular docking simulations to screen the Drug Repurposing Hub (DRH) database. As a result, 18 drug hits with best consensus prediction scores and binding affinity values were identified as the potential therapeutics against COVID-19. Both the extensive literature searching and evaluations on adsorption, distribution, metabolism, excretion, and toxicity (ADMET) illustrate the premium drug-likeness and pharmacokinetic properties of the drug candidates. Overall, our work not only provides an effective GAT-based DL prediction tool for inhibitory activity of SARS-CoV-2 Mpro inhibitors but also provides theoretical guidelines for drug discovery in the COVID-19 treatment.

PMID:37960886 | DOI:10.1021/acs.jcim.3c01280

Categories: Literature Watch

Repurposing Drugs for Inhibition against ALDH2 via a 2D/3D Ligand-Based Similarity Search and Molecular Simulation

Tue, 2023-11-14 06:00

Molecules. 2023 Oct 29;28(21):7325. doi: 10.3390/molecules28217325.

ABSTRACT

Aldehyde dehydrogenase-2 (ALDH2) is a crucial enzyme participating in intracellular aldehyde metabolism and is acknowledged as a potential therapeutic target for the treatment of alcohol use disorder and other addictive behaviors. Using previously reported ALDH2 inhibitors of Daidzin, CVT-10216, and CHEMBL114083 as reference molecules, here we perform a ligand-based virtual screening of world-approved drugs via 2D/3D similarity search methods, followed by the assessments of molecular docking, toxicity prediction, molecular simulation, and the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) analysis. The 2D molecular fingerprinting of ECFP4 and FCFP4 and 3D molecule-shape-based USRCAT methods show good performances in selecting compounds with a strong binding behavior with ALDH2. Three compounds of Zeaxanthin (q = 0), Troglitazone (q = 0), and Sequinavir (q = +1 e) are singled out as potential inhibitors; Zeaxanthin can only be hit via USRCAT. These drugs displayed a stronger binding strength compared to the reported potent inhibitor CVT-10216. Sarizotan (q = +1 e) and Netarsudil (q = 0/+1 e) displayed a strong binding strength with ALDH2 as well, whereas they displayed a shallow penetration into the substrate-binding tunnel of ALDH2 and could not fully occupy it. This likely left a space for substrate binding, and thus they were not ideal inhibitors. The MM-PBSA results indicate that the selected negatively charged compounds from the similarity search and Vina scoring are thermodynamically unfavorable, mainly due to electrostatic repulsion with the receptor (q = -6 e for ALDH2). The electrostatic attraction with positively charged compounds, however, yielded very strong binding results with ALDH2. These findings reveal a deficiency in the modeling of electrostatic interactions (in particular, between charged moieties) in the virtual screening via the 2D/3D similarity search and molecular docking with the Vina scoring system.

PMID:37959744 | DOI:10.3390/molecules28217325

Categories: Literature Watch

Unveiling the Molecular Landscape of <em>FOXA1</em> Mutant Prostate Cancer: Insights and Prospects for Targeted Therapeutic Strategies

Tue, 2023-11-14 06:00

Int J Mol Sci. 2023 Oct 31;24(21):15823. doi: 10.3390/ijms242115823.

ABSTRACT

Prostate cancer continues to pose a global health challenge as one of the most prevalent malignancies. Mutations of the Forkhead box A1 (FOXA1) gene have been linked to unique oncogenic features in prostate cancer. In this study, we aimed to unravel the intricate molecular characteristics of FOXA1 mutant prostate cancer through comprehensive in silico analysis of transcriptomic data from The Cancer Genome Atlas (TCGA). A comparison between FOXA1 mutant and control groups unearthed 1525 differentially expressed genes (DEGs), which map to eight intrinsic and six extrinsic signaling pathways. Interestingly, the majority of intrinsic pathways, but not extrinsic pathways, were validated using RNA-seq data of 22Rv1 cells from the GEO123619 dataset, suggesting complex biology in the tumor microenvironment. As a result of our in silico research, we identified novel therapeutic targets and potential drug candidates for FOXA1 mutant prostate cancer. KDM1A, MAOA, PDGFB, and HSP90AB1 emerged as druggable candidate targets, as we found that they have approved drugs throughout the drug database CADDIE. Notably, as most of the approved drugs targeting MAOA and KDM1A were monoamine inhibitors used for mental illness or diabetes, we suggest they have a potential to cure FOXA1 mutant primary prostate cancer without lethal side effects.

PMID:37958805 | DOI:10.3390/ijms242115823

Categories: Literature Watch

Pages