1,244 results
Search Results
2. Economic aspects of treating seizure clusters.
- Author
-
Faught E
- Subjects
- Brain Damage, Chronic, Cost-Benefit Analysis, Employment, Humans, Drug Repositioning economics, Epilepsy, Generalized, Seizures drug therapy, Seizures economics
- Abstract
Seizure clusters may initiate a chain of events that have economic as well as clinical consequences. The potential economic consequences of seizure clusters must be weighed against the cost of medication to attenuate them. This is true both for individual patients and for society. Data needed for economic analyses include the chance that a cluster will progress to an adverse outcome, such as a need for emergency care, the costs of such an outcome, the cost of a rescue medication (RM), and the effectiveness of the RM. Indirect costs, such as lost employment for patients and caregivers, must also be considered. Several types of economic analyses can be used to determine costs and benefits of a medical intervention. There are studies comparing different RMs from an economic perspective, but there is little direct information on the costs of using an RM versus allowing clusters to run their course. However, the high expense of consequences of seizure clusters makes it likely that effective RMs will make economic as well as medical sense for many patients., (© 2022 International League Against Epilepsy.)
- Published
- 2022
- Full Text
- View/download PDF
3. Repurposing the estrogen receptor modulator raloxifene to treat SARS-CoV-2 infection.
- Author
-
Allegretti M, Cesta MC, Zippoli M, Beccari A, Talarico C, Mantelli F, Bucci EM, Scorzolini L, and Nicastri E
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Antiviral Agents therapeutic use, Estradiol therapeutic use, Estrogens metabolism, Female, Humans, Male, SARS-CoV-2 drug effects, Sex Factors, Anti-Inflammatory Agents therapeutic use, Drug Repositioning, Estrogen Receptor Modulators therapeutic use, Raloxifene Hydrochloride therapeutic use, COVID-19 Drug Treatment
- Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) necessitates strategies to identify prophylactic and therapeutic drug candidates to enter rapid clinical development. This is particularly true, given the uncertainty about the endurance of the immune memory induced by both previous infections or vaccines, and given the fact that the eradication of SARS-CoV-2 might be challenging to reach, given the attack rate of the virus, which would require unusually high protection by a vaccine. Here, we show how raloxifene, a selective estrogen receptor modulator with anti-inflammatory and antiviral properties, emerges as an attractive candidate entering clinical trials to test its efficacy in early-stage treatment COVID-19 patients., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
4. The role of new medical treatments for the management of developmental and epileptic encephalopathies: Novel concepts and results.
- Author
-
Johannessen Landmark C, Potschka H, Auvin S, Wilmshurst JM, Johannessen SI, Kasteleijn-Nolst Trenité D, and Wirrell EC
- Subjects
- Cannabidiol therapeutic use, Drug Repositioning trends, Epilepsies, Myoclonic diagnosis, Epilepsies, Myoclonic physiopathology, Everolimus therapeutic use, Fenfluramine therapeutic use, Humans, Lennox Gastaut Syndrome diagnosis, Lennox Gastaut Syndrome physiopathology, Precision Medicine trends, Treatment Outcome, Anticonvulsants therapeutic use, Disease Management, Drug Repositioning methods, Epilepsies, Myoclonic drug therapy, Lennox Gastaut Syndrome drug therapy, Precision Medicine methods
- Abstract
Developmental and epileptic encephalopathies (DEEs) are among the most challenging of all epilepsies to manage, given the exceedingly frequent and often severe seizure types, pharmacoresistance to conventional antiseizure medications, and numerous comorbidities. During the past decade, efforts have focused on development of new treatment options for DEEs, with several recently approved in the United States or Europe, including cannabidiol as an orphan drug in Dravet and Lennox-Gastaut syndromes and everolimus as a possible antiepileptogenic and precision drug for tuberous sclerosis complex, with its impact on the mammalian target of rapamycin pathway. Furthermore, fenfluramine, an old drug, was repurposed as a novel therapy in the treatment of Dravet syndrome. The evolution of new insights into pathophysiological processes of various DEEs provides possibilities to investigate novel and repurposed drugs and to place them into the context of their role in future management of these patients. The purpose of this review is to provide an overview of these new medical treatment options for the DEEs and to discuss the clinical implications of these results for improved treatment., (© 2021 The Authors. Epilepsia published by Wiley Periodicals LLC on behalf of International League Against Epilepsy.)
- Published
- 2021
- Full Text
- View/download PDF
5. Calculating the similarity between prescriptions to find their new indications based on graph neural network.
- Author
-
Han, Xingxing, Xie, Xiaoxia, Zhao, Ranran, Li, Yu, Ma, Pengzhen, Li, Huan, Chen, Fengming, Zhao, Yufeng, and Tang, Zhishu
- Subjects
CHINESE medicine ,DIFFUSION of innovations ,RESEARCH funding ,QUESTIONNAIRES ,DRUG repositioning ,ARTIFICIAL neural networks ,DRUGS ,COMPARATIVE studies - Abstract
Background: Drug repositioning has the potential to reduce costs and accelerate the rate of drug development, with highly promising applications. Currently, the development of artificial intelligence has provided the field with fast and efficient computing power. Nevertheless, the repositioning of traditional Chinese medicine (TCM) is still in its infancy, and the establishment of a reasonable and effective research method is a pressing issue that requires urgent attention. The use of graph neural network (GNN) to compute the similarity between TCM prescriptions to develop a method for finding their new indications is an innovative attempt. Methods: This paper focused on traditional Chinese medicine prescriptions containing ephedra, with 20 prescriptions for treating external cough and asthma taken as target prescriptions. The remaining 67 prescriptions containing ephedra were taken as to-be-matched prescriptions. Furthermore, a multitude of data pertaining to the prescriptions, including diseases, disease targets, symptoms, and various types of information on herbs, was gathered from a diverse array of literature sources, such as Chinese medicine databases. Then, cosine similarity and Jaccard coefficient were calculated to characterize the similarity between prescriptions using graph convolutional network (GCN) with a self-supervised learning method, such as deep graph infomax (DGI). Results: A total of 1340 values were obtained for each of the two calculation indicators. A total of 68 prescription pairs were identified after screening with 0.77 as the threshold for cosine similarity. Following the removal of false positive results, 12 prescription pairs were deemed to have further research value. A total of 5 prescription pairs were screened using a threshold of 0.50 for the Jaccard coefficient. However, the specific results did not exhibit significant value for further use, which may be attributed to the excessive variety of information in the dataset. Conclusions: The proposed method can provide reference for finding new indications of target prescriptions by quantifying the similarity between prescriptions. It is expected to offer new insights for developing a scientific and systematic research methodology for traditional Chinese medicine repositioning. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Converging pathways: bringing community, student initiatives, and a systematic review project in COVID-19 pandemic.
- Author
-
Bomfim Ribeiro, Tatiane, Camila Ramírez, Paula, Maria Pelissari, Daniele, Souza Vieira, Adriano Tito, Santos de Melo, Luís Ricardo, Pereira Persch, Gustavo, Campêlo Brandim de Sá Lopes, João Guilherme, de Sousa Alves, Rafael, Alves Rizzo, Gustavo, Adorno Brito, Elisama, Santos Evangelista, Thiago, Campos Ornelas, Rachel, Tedesco e Silva, Aída Rita, Pires Daneris, Andrea, Ferraz Mota, Larissa, Bento de Moura, Jade, dos Santos França, Júlia, Nascimento Martins, Pedro, Espindula da Silva, Poliana, and Kariny Gomes, Karen
- Subjects
COVID-19 pandemic ,MEDICAL personnel ,DRUG repositioning ,COVID-19 treatment ,UNIVERSITY extension - Abstract
Copyright of Revista Salud UIS is the property of Universidad Industrial de Santander and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
7. Antivirals for monkeypox virus: Proposing an effective machine/deep learning framework.
- Author
-
Hashemi, Morteza, Zabihian, Arash, Hajsaeedi, Masih, and Hooshmand, Mohsen
- Subjects
DEEP learning ,DRUG repositioning ,MONKEYPOX ,MACHINE learning ,PROTEIN structure - Abstract
Monkeypox (MPXV) is one of the infectious viruses which caused morbidity and mortality problems in these years. Despite its danger to public health, there is no approved drug to stand and handle MPXV. On the other hand, drug repurposing is a promising screening method for the low-cost introduction of approved drugs for emerging diseases and viruses which utilizes computational methods. Therefore, drug repurposing is a promising approach to suggesting approved drugs for the MPXV. This paper proposes a computational framework for MPXV antiviral prediction. To do this, we have generated a new virus-antiviral dataset. Moreover, we applied several machine learning and one deep learning method for virus-antiviral prediction. The suggested drugs by the learning methods have been investigated using docking studies. The target protein structure is modeled using homology modeling and, then, refined and validated. To the best of our knowledge, this work is the first work to study deep learning methods for the prediction of MPXV antivirals. The screening results confirm that Tilorone, Valacyclovir, Ribavirin, Favipiravir, and Baloxavir marboxil are effective drugs for MPXV treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Repurposed Medicines: A Scan of the Non-commercial Clinical Research Landscape.
- Author
-
Akinbolade S, Fairbairn R, Inskip A, Potter R, Oliver A, and Craig D
- Subjects
- Humans, Biomedical Research, Clinical Trials as Topic, Databases, Factual, Drug Repositioning methods
- Abstract
Medicine repurposing is a strategy to identify new uses for the existing medicines for the purpose of addressing areas of unmet medical need. This paper aims to provide horizon scanning intelligence on repurposed medicines that are evaluated by non-commercial organizations such as academia and highlights opportunities for further research to improve patient health outcomes. A scan of the clinical landscape of non-commercially sponsored repurposed medicines is routinely conducted by the NIHR Innovation Observatory (IO). This ongoing project involves a horizon scan of clinical trial registries and the IO's internal horizon scanning Medicines Innovation Database to identify potential candidate medicines used as monotherapy or in combination to treat new indications outside the scope of their licensed indication. In addition to making these data publicly available, the output also supports the NHS England Medicines Repurposing Programme. The snapshot scan reported here (trials completing April 2020-March 2023) identified a total of 528 technologies (meaning, a single product or combination of medicinal products targeting a specific indication in one or more related trials). The technologies were classified according to their characteristics and targeted therapeutic indications as well as revealing the least treated disease conditions. The candidate medicines identified in this scan could potentially receive tailored support toward adoption into practice and policy. The NIHR IO regularly provides this scan as a source of intelligence on repurposed medicines. This provides valuable insights into innovation trends, gaps, and areas of unmet clinical need., (© 2024 The Author(s). Pharmacology Research & Perspectives published by British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics and John Wiley & Sons Ltd.)
- Published
- 2025
- Full Text
- View/download PDF
9. MCF-DTI: Multi-Scale Convolutional Local-Global Feature Fusion for Drug-Target Interaction Prediction.
- Author
-
Wang J, He R, Wang X, Li H, and Lu Y
- Subjects
- Humans, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Lung Neoplasms pathology, Algorithms, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Drug Discovery methods, Neural Networks, Computer, Drug Repositioning methods
- Abstract
Predicting drug-target interactions (DTIs) is a crucial step in the development of new drugs and drug repurposing. In this paper, we propose a novel drug-target prediction model called MCF-DTI. The model utilizes the SMILES representation of drugs and the sequence features of targets, employing a multi-scale convolutional neural network (MSCNN) with parallel shared-weight modules to extract features from the drug side. For the target side, it combines MSCNN with Transformer modules to capture both local and global features effectively. The extracted features are then weighted and fused, enabling comprehensive feature representation to enhance the predictive power of the model. Experimental results on the Davis dataset demonstrate that MCF-DTI achieves an AUC of 0.9746 and an AUPR of 0.9542, outperforming other state-of-the-art models. Our case study demonstrates that our model effectively validated several known drug-target relationships in lung cancer and predicted the therapeutic potential of certain preclinical compounds in treating lung cancer. These findings contribute valuable insights for subsequent drug repurposing efforts and novel drug development.
- Published
- 2025
- Full Text
- View/download PDF
10. Chemotherapeutic potential of radotinib against blood and solid tumors: A beacon of hope in drug repurposing.
- Author
-
Manoharan S and Perumal E
- Subjects
- Humans, Molecular Structure, Pyrimidines chemistry, Pyrimidines pharmacology, Pyrimidines chemical synthesis, Pyrimidines therapeutic use, Hematologic Neoplasms drug therapy, Hematologic Neoplasms pathology, Animals, Benzamides, Pyrazines, Drug Repositioning, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors chemical synthesis, Neoplasms drug therapy
- Abstract
Tyrosine kinase inhibitors (TKIs) represent a pivotal class of targeted therapies in oncology, with multiple generations developed to address diverse molecular targets. Imatinib is the first TKI developed to target the BCR-ABL1 chimeric protein, which is the key driver oncogene implicated in Philadelphia chromosome-positive chronic myeloid leukemia (CML). Several second-generation tyrosine kinase inhibitors (2GTKIs), such as nilotinib, dasatinib, bosutinib, and radotinib (RTB), followed the groundbreaking introduction of imatinib. RTB occupies the unique position of being the least explored member of this class. While nilotinib, dasatinib, and bosutinib have garnered significant attention and extensive research focus, RTB remains relatively uncharted in comparison to its counterparts. Fundamental drug characteristics, such as the pharmacokinetic and pharmacodynamic properties of RTB, remain unavailable in existing sources. Compared to other 2GTKIs, RTB has been less utilized in combinatorial drug studies, and no investigations have been reported on its effects on solid tumors to date. However, the effects of RTB have been studied in acute myeloid leukemia (AML), multiple myeloma (MM), Parkinson's disease, and idiopathic pulmonary fibrosis (IPF). Although RTB has been investigated in some conditions, these studies are still in their preliminary stages and are comparatively lesser than studies on other 2GTKIs. This review is the first attempt that extensively presents a compilation of data on RTB and describes its therapeutic potential against blood and solid tumors. Further investigations on RTB could expand its chemotherapeutic usage in various solid tumors and enhance the possibility of drug repurposing in cancer therapy., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
11. Drug repurposing screen for the rare disease ataxia-telangiectasia.
- Author
-
Jayanth N, Mahé G, Campbell M, Lipkin M, Jain S, van de Bospoort R, Thornton J, Margus B, and Fischer DF
- Subjects
- Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells metabolism, High-Throughput Screening Assays methods, Rare Diseases drug therapy, Rare Diseases genetics, DNA Repair drug effects, DNA Repair genetics, Checkpoint Kinase 2 genetics, Checkpoint Kinase 2 metabolism, Mutation genetics, Ataxia Telangiectasia genetics, Ataxia Telangiectasia drug therapy, Drug Repositioning methods, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, DNA Damage drug effects
- Abstract
Ataxia Telangiectasia (A-T) is a rare, autosomal recessive genetic disorder characterized by a variety of symptoms, including progressive neurodegeneration, telangiectasia, immunodeficiency, and an increased susceptibility to cancer. It is caused by bi-allelic mutations impacting a gene encoding a serine/threonine kinase ATM (Ataxia Telangiectasia Mutated), which plays a crucial role in DNA repair and maintenance of genomic stability. The disorder primarily affects the nervous system, leading to a range of neurological issues, including cerebellar ataxia. The cause of neurodegeneration due to mutations in ATM is still an area of investigation, and currently there is no known treatment to slow down or stop the progression of the neurological problems. In this collaboration of the A-T Children's Project (ATCP) with Charles River Discovery, we successfully developed a high-throughput assay using induced pluripotent stem cells (iPSC) from A-T donors to measure DNA damage response (DDR). By measuring the changes in levels of activated phosphorylated CHK2 (p-CHK2), which is a downstream signaling event of ATM, we were able to identify compounds that restore this response in the DDR pathway in A-T derived patient cells. Over 6,000 compounds from small molecule drug repurposing libraries were subsequently screened in the assay developed, leading to identification of several promising in vitro hits. Using the assay developed and the identified hits opens avenues to investigate potential therapeutics for A-T., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Brad Margus reports a relationship with AT Childrens Project that includes: board membership. Jennifer Thornton reports a relationship with AT Childrens Project that includes: employment and non-financial support. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2025
- Full Text
- View/download PDF
12. Discovery of Daclatasvir as a potential PD-L1 inhibitor from drug repurposing.
- Author
-
Sun M, Lv S, Pan Y, Song Q, Ma C, Yu M, Gao X, Guo X, Wang S, Gao Z, Wang S, Meng Q, Zhang L, and Li Y
- Subjects
- Humans, Animals, Mice, Molecular Structure, Dose-Response Relationship, Drug, Molecular Docking Simulation, Drug Discovery, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Cell Proliferation drug effects, Drug Screening Assays, Antitumor, Quantitative Structure-Activity Relationship, Imidazoles chemistry, Imidazoles pharmacology, Valine analogs & derivatives, Valine chemistry, Valine pharmacology, Pyrrolidines chemistry, Pyrrolidines pharmacology, Carbamates chemistry, Carbamates pharmacology, Drug Repositioning, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen metabolism
- Abstract
This study employed a drug repositioning strategy to discover novel PD-L1 small molecule inhibitors. 3D-QSAR pharmacophore models were establishedand subsequently validated through various means to select a robust model, Hypo-1, suitable for virtual screening. Hypo1 was used toscreen a library of 7,475 compounds from the Drugbank database, leading to the identification of 283 molecules following molecular docking with PD-L1.19 compounds underwent HTRF assays, with 15 showing varying degrees of inhibition of the PD-1/PD-L1 interaction. Compounds2202,2204,2207, and2208were further confirmed to bind to PD-L1 using SPR experiments. Among them, compound2204(Daclatasvir, K
D = 11.4 μM) showeda higher affinity for human PD-L1 than the control compound BMS-1. In the HepG2/Jurkat cell co-culture model, Daclatasvir effectively activated Jurkat cells to kill HepG2 cells. In the mouse H22 hepatocellular tumor model, Daclatasvir significantly inhibited tumor growth (TGI = 53.4 % at a dose of 100 mg/kg). Its anti-tumor effect was more pronounced when combined with Lenvatinib (TGI = 85.1 %). Flow cytometry analysis of splenocytes and tumor cells indicated that Daclatasvir activated the immune system in both models. In summary, Daclatasvir was identified as a novel PD-L1small molecule inhibitor., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
13. A promising drug repurposing approach for Alzheimer's treatment: Givinostat improves cognitive behavior and pathological features in APP/PS1 mice.
- Author
-
Gao QC, Liu GL, Wang Q, Zhang SX, Ji ZL, Wang ZJ, Wu MN, Yu Q, and He PF
- Subjects
- Animals, Mice, Humans, Mice, Transgenic, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Mitochondria metabolism, Mitochondria drug effects, Cognition drug effects, Amyloid beta-Peptides metabolism, Hippocampus metabolism, Hippocampus drug effects, Hippocampus pathology, Membrane Potential, Mitochondrial drug effects, Reactive Oxygen Species metabolism, Alzheimer Disease drug therapy, Alzheimer Disease metabolism, Alzheimer Disease pathology, Drug Repositioning, Carbamates pharmacology, Carbamates therapeutic use, Disease Models, Animal
- Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, characterized by memory loss, speech and motor defects, personality changes, and psychological disorders. The exact cause of AD remains unclear. Current treatments focus on maintaining neurotransmitter levels or targeting β-amyloid (Aβ) protein, but these only alleviate symptoms and do not reverse the disease. Developing new drugs is time-consuming, costly, and has a high failure rate. Utilizing multi-omics for drug repositioning has emerged as a new strategy. Based on transcriptomic perturbation data of over 40,000 drugs in human cells from the LINCS-L1000 database, our study employed the Jaccard index and hypergeometric distribution test for reverse transcriptional feature matching analysis, identifying Givinostat as a potential treatment for AD. Our research found that Givinostat improved cognitive behavior and brain pathology in models and enhanced hippocampal synaptic plasticity. Transcriptome sequencing revealed increased expression of mitochondrial respiratory chain complex proteins in the brains of APP/PS1 mice after Givinostat treatment. Functionally, Givinostat restored mitochondrial membrane potential, reduced reactive oxygen species, and increased ATP content in Aβ-induced HT22 cells. Additionally, it improved mitochondrial morphology and quantity in the hippocampus of APP/PS1 mice and enhanced brain glucose metabolic activity. These effects are linked to Givinostat promoting mitochondrial biogenesis and improving mitochondrial function. In summary, Givinostat offers a promising new strategy for AD treatment by targeting mitochondrial dysfunction., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Automatic collaborative learning for drug repositioning
- Author
-
Wang, Yi, Meng, Yajie, Zhou, Chang, Tang, Xianfang, Zeng, Pan, Pan, Chu, Zhu, Qiang, Zhang, Bengong, and Xu, Junlin
- Published
- 2025
- Full Text
- View/download PDF
15. Design and application of a knowledge network for automatic prioritization of drug mechanisms
- Author
-
Núria Queralt-Rosinach, Steinecke D, Roger Tu, Tianhu Li, Mayers, and Andrew I. Su
- Subjects
Statistics and Probability ,Databases, Pharmaceutical ,Computer science ,Machine learning ,computer.software_genre ,Biochemistry ,Code (cryptography) ,Mean reciprocal rank ,Set (psychology) ,Molecular Biology ,Repurposing ,computer.programming_language ,Drug discovery ,business.industry ,Drug Repositioning ,Python (programming language) ,Original Papers ,Computer Science Applications ,Drug repositioning ,Computational Mathematics ,Computational Theory and Mathematics ,Test set ,Artificial intelligence ,business ,computer ,Algorithms - Abstract
Motivation Drug repositioning is an attractive alternative to de novo drug discovery due to reduced time and costs to bring drugs to market. Computational repositioning methods, particularly non-black-box methods that can account for and predict a drug’s mechanism, may provide great benefit for directing future development. By tuning both data and algorithm to utilize relationships important to drug mechanisms, a computational repositioning algorithm can be trained to both predict and explain mechanistically novel indications. Results In this work, we examined the 123 curated drug mechanism paths found in the drug mechanism database (DrugMechDB) and after identifying the most important relationships, we integrated 18 data sources to produce a heterogeneous knowledge graph, MechRepoNet, capable of capturing the information in these paths. We applied the Rephetio repurposing algorithm to MechRepoNet using only a subset of relationships known to be mechanistic in nature and found adequate predictive ability on an evaluation set with AUROC value of 0.83. The resulting repurposing model allowed us to prioritize paths in our knowledge graph to produce a predicted treatment mechanism. We found that DrugMechDB paths, when present in the network were rated highly among predicted mechanisms. We then demonstrated MechRepoNet’s ability to use mechanistic insight to identify a drug’s mechanistic target, with a mean reciprocal rank of 0.525 on a test set of known drug–target interactions. Finally, we walked through repurposing examples of the anti-cancer drug imatinib for use in the treatment of asthma, and metolazone for use in the treatment of osteoporosis, to demonstrate this method’s utility in providing mechanistic insight into repurposing predictions it provides. Availability and implementation The Python code to reproduce the entirety of this analysis is available at: https://github.com/SuLab/MechRepoNet (archived at https://doi.org/10.5281/zenodo.6456335). Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2022
- Full Text
- View/download PDF
16. Inclusion Complexes of Gold(I)‐Dithiocarbamates with β‐Cyclodextrin: A Journey from Drug Repurposing towards Drug Discovery
- Author
-
Nikolas Gunkel, Eberhard Amtmann, Michael Morgen, Piotr Fabrowski, and Aubry K. Miller
- Subjects
Catalysis ,beta-cyclodextrin ,X-Ray Diffraction ,Drug Discovery ,medicine ,Moiety ,Humans ,host–guest interactions ,Cytotoxicity ,inclusion complexes ,chemistry.chemical_classification ,Reactive oxygen species ,Cyclodextrin ,Full Paper ,drug repurposing ,Drug discovery ,Organic Chemistry ,beta-Cyclodextrins ,Drug Repositioning ,General Chemistry ,Full Papers ,Combinatorial chemistry ,Sodium aurothiomalate ,gold dithiocarbamates ,chemistry ,Solubility ,Cancer cell ,Disulfiram ,Gold ,medicine.drug - Abstract
The gold(I)‐dithiocarbamate (dtc) complex [Au(N,N‐diethyl)dtc]2 was identified as the active cytotoxic agent in the combination treatment of sodium aurothiomalate and disulfiram on a panel of cancer cell lines. In addition to demonstrating pronounced differential cytotoxicity to these cell lines, the gold complex showed no cross‐resistance in therapy‐surviving cancer cells. In the course of a medicinal chemistry campaign on this class of poorly soluble gold(I)‐dtc complexes, >35 derivatives were synthesized and X‐ray crystallography was used to examine structural aspects of the dtc moiety. A group of hydroxy‐substituted complexes has an improved solubility profile, and it was found that these complexes form 2 : 1 host–guest inclusion complexes with β‐cyclodextrin (CD), exhibiting a rarely observed “tail‐to‐tail” arrangement of the CD cones. Formulation of a hydroxy‐substituted gold(I)‐dtc complex with excess sulfobutylether‐β‐CD prevents the induction of mitochondrial reactive oxygen species, which is a major burden in the development of metallodrugs., Inclusion complex of a complex: The two drugs disulfiram and aurothiomalate react to produce a gold(I)‐dithiocarbamate complex, which selectively kills cancer cells. Hydroxy‐substituted derivatives of this complex form inclusion complexes with β‐cyclodextrin in a rare “tail‐to‐tail” arrangement. Formulation of one gold complex with a β‐cyclodextrin derivative prevents formation of reactive oxygen species in mitochondria.
- Published
- 2021
17. Atomistic De‐novo Inhibitor Generation‐Guided Drug Repurposing for SARS‐CoV‐2 Spike Protein with Free‐Energy Validation by Well‐Tempered Metadynamics
- Author
-
Amal Vijay, Venkata Sai Sreyas Adury, Reman K. Singh, Arnab Mukherjee, and Rituparno Chowdhury
- Subjects
Models, Molecular ,human ACE2 ,Protein Conformation ,Structural similarity ,Computational biology ,Spike protein ,010402 general chemistry ,Antiviral Agents ,01 natural sciences ,Biochemistry ,Protein structure ,Humans ,Computer Simulation ,Repurposing ,Full Paper ,SARS-CoV-2 ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Drug Repositioning ,Metadynamics ,COVID-19 ,Spike Protein ,de Novo drug design ,General Chemistry ,Full Papers ,free energy ,molecular dynamics ,COVID-19 Drug Treatment ,0104 chemical sciences ,Drug repositioning ,Docking (molecular) ,Drug Design ,Spike Glycoprotein, Coronavirus ,docking ,Thermodynamics ,Spike (software development) ,repurposing therapeutics ,well-tempered metadynamics - Abstract
Computational drug design is increasingly becoming important with new and unforeseen diseases like COVID‐19. In this study, we present a new computational de novo drug design and repurposing method and applied it to find plausible drug candidates for the receptor binding domain (RBD) of SARS‐CoV‐2 (COVID‐19). Our study comprises three steps: atom‐by‐atom generation of new molecules around a receptor, structural similarity mapping to existing approved and investigational drugs, and validation of their binding strengths to the viral spike proteins based on rigorous all‐atom, explicit‐water well‐tempered metadynamics free energy calculations. By choosing the receptor binding domain of the viral spike protein, we showed that some of our new molecules and some of the repurposable drugs have stronger binding to RBD than hACE2. To validate our approach, we also calculated the free energy of hACE2 and RBD, and found it to be in an excellent agreement with experiments. These pool of drugs will allow strategic repurposing against COVID‐19 for a particular prevailing conditions., Schematic diagram of atom‐by‐atom molecule generation, followed by repurposed molecule selection and validation using fre‐energy calculations.
- Published
- 2021
- Full Text
- View/download PDF
18. Drug synergy of combinatory treatment with remdesivir and the repurposed drugs fluoxetine and itraconazole effectively impairs SARS‐CoV‐2 infection in vitro
- Author
-
Linda Brunotte, Stephan Ludwig, Angeles Mecate-Zambrano, Ursula Rescher, Shuyu Zheng, Jing Tang, Sebastian Schloer, Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, and University of Helsinki
- Subjects
0301 basic medicine ,PHARMACOKINETICS ,remdesivir ,Pharmacology ,SARS‐CoV‐2 ,combination therapy ,0302 clinical medicine ,Medicine ,Repurposing ,media_common ,0303 health sciences ,Alanine ,drug repurposing ,Research Papers ,CONCISE GUIDE ,itraconazole ,3. Good health ,Drug repositioning ,Pharmaceutical Preparations ,Drug development ,Synergy ,317 Pharmacy ,030220 oncology & carcinogenesis ,VIRUS ,Research Paper ,medicine.drug ,Drug ,Combination therapy ,Itraconazole ,media_common.quotation_subject ,SARS‐ ,Antiviral Agents ,Virus ,03 medical and health sciences ,CoV‐ ,Humans ,030304 developmental biology ,SARS-CoV-2 ,business.industry ,fluoxetine ,Hepatitis C, Chronic ,Drug interaction ,Adenosine Monophosphate ,COVID-19 Drug Treatment ,030104 developmental biology ,INHIBITORS ,business ,030217 neurology & neurosurgery - Abstract
Background and Purpose The SARS-COV-2 pandemic and the global spread of coronavirus disease 2019 (COVID-19) urgently call for efficient and safe antiviral treatment strategies. A straightforward approach to speed up drug development at lower costs is drug repurposing. Here, we investigated the therapeutic potential of targeting the interface of SARS CoV-2 with the host via repurposing of clinically licensed drugs and evaluated their use in combinatory treatments with virus- and host-directed drugs in vitro. Experimental Approach We tested the antiviral potential of the antifungal itraconazole and the antidepressant fluoxetine on the production of infectious SARS-CoV-2 particles in the polarized Calu-3 cell culture model and evaluated the added benefit of a combinatory use of these host-directed drugs with the direct acting antiviral remdesivir, an inhibitor of viral RNA polymerase. Key Results Drug treatments were well-tolerated and potently impaired viral replication. Importantly, both itraconazole?remdesivir and fluoxetine?remdesivir combinations inhibited the production of infectious SARS-CoV-2 particles?>?90% and displayed synergistic effects, as determined in commonly used reference models for drug interaction. Conclusion and Implications Itraconazole?remdesivir and fluoxetine?remdesivir combinations are promising starting points for therapeutic options to control SARS-CoV-2 infection and severe progression of COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
19. Repurposed Drugs and Plant-Derived Natural Products as Potential Host-Directed Therapeutic Candidates for Tuberculosis.
- Author
-
Raqib R and Sarker P
- Subjects
- Animals, Humans, Mycobacterium tuberculosis drug effects, Mycobacterium tuberculosis pathogenicity, Antitubercular Agents therapeutic use, Antitubercular Agents pharmacology, Biological Products therapeutic use, Biological Products pharmacology, Drug Repositioning, Extensively Drug-Resistant Tuberculosis drug therapy, Extensively Drug-Resistant Tuberculosis microbiology
- Abstract
Tuberculosis (TB) is one of the leading causes of death due to infectious disease. It is a treatable disease; however, conventional treatment requires a lengthy treatment regimen with severe side effects, resulting in poor compliance among TB patients. Intermittent drug use, the non-compliance of patients, and prescription errors, among other factors, have led to the emergence of multidrug-resistant TB, while the mismanagement of multidrug-resistant TB (MDR-TB) has eventually led to the development of extensively drug-resistant tuberculosis (XDR-TB). Thus, there is an urgent need for new drug development, but due to the enormous expenses and time required (up to 20 years) for new drug research and development, new therapeutic approaches to TB are required. Host-directed therapies (HDT) could be a most attractive strategy, as they target the host defense processes instead of the microbe and thereby may prevent the alarming rise of MDR- and XDR-TB. This paper reviews the progress in HDT for the treatment of TB using repurposed drugs which have been investigated in clinical trials (completed or ongoing) and plant-derived natural products that are in clinical or preclinical trial stages. Additionally, this review describes the existing challenges to the development and future research directions in the implementation of HDT.
- Published
- 2024
- Full Text
- View/download PDF
20. Heterogeneous graph contrastive learning with gradient balance for drug repositioning.
- Author
-
Cui H, Duan M, Bi H, Li X, Hou X, and Zhang Y
- Subjects
- Humans, Computational Biology methods, Machine Learning, Algorithms, Semantics, Drug Discovery methods, Drug Repositioning methods
- Abstract
Drug repositioning, which involves identifying new therapeutic indications for approved drugs, is pivotal in accelerating drug discovery. Recently, to mitigate the effect of label sparsity on inferring potential drug-disease associations (DDAs), graph contrastive learning (GCL) has emerged as a promising paradigm to supplement high-quality self-supervised signals through designing auxiliary tasks, then transfer shareable knowledge to main task, i.e. DDA prediction. However, existing approaches still encounter two limitations. The first is how to generate augmented views for fully capturing higher-order interaction semantics. The second is the optimization imbalance issue between auxiliary and main tasks. In this paper, we propose a novel heterogeneous Graph Contrastive learning method with Gradient Balance for DDA prediction, namely GCGB. To handle the first challenge, a fusion view is introduced to integrate both semantic views (drug and disease similarity networks) and interaction view (heterogeneous biomedical network). Next, inter-view contrastive learning auxiliary tasks are designed to contrast the fusion view with semantic and interaction views, respectively. For the second challenge, we adaptively adjust the gradient of GCL auxiliary tasks from the perspective of gradient direction and magnitude for better guiding parameter update toward main task. Extensive experiments conducted on three benchmarks under 10-fold cross-validation demonstrate the model effectiveness., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
21. Knowledge Graphs for drug repurposing: a review of databases and methods.
- Author
-
Perdomo-Quinteiro P and Belmonte-Hernández A
- Subjects
- Humans, Algorithms, Artificial Intelligence, Databases, Factual, Computational Biology methods, Drug Repositioning methods
- Abstract
Drug repurposing has emerged as a effective and efficient strategy to identify new treatments for a variety of diseases. One of the most effective approaches for discovering potential new drug candidates involves the utilization of Knowledge Graphs (KGs). This review comprehensively explores some of the most prominent KGs, detailing their structure, data sources, and how they facilitate the repurposing of drugs. In addition to KGs, this paper delves into various artificial intelligence techniques that enhance the process of drug repurposing. These methods not only accelerate the identification of viable drug candidates but also improve the precision of predictions by leveraging complex datasets and advanced algorithms. Furthermore, the importance of explainability in drug repurposing is emphasized. Explainability methods are crucial as they provide insights into the reasoning behind AI-generated predictions, thereby increasing the trustworthiness and transparency of the repurposing process. We will discuss several techniques that can be employed to validate these predictions, ensuring that they are both reliable and understandable., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
22. Repurposing of antimycobacterium drugs for COVID-19 treatment by targeting SARS CoV-2 main protease: An in-silico perspective.
- Author
-
Chakraborty A, Ghosh R, Soumya Mohapatra S, Barik S, Biswas A, and Chowdhuri S
- Subjects
- Humans, Antiviral Agents pharmacology, Antiviral Agents chemistry, Anti-Bacterial Agents pharmacology, Minocycline pharmacology, Rifampin pharmacology, COVID-19 virology, Computer Simulation, Drug Repositioning, Molecular Docking Simulation, COVID-19 Drug Treatment, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus 3C Proteases metabolism, Coronavirus 3C Proteases chemistry, Molecular Dynamics Simulation
- Abstract
The global mortality rate has been significantly impacted by the COVID-19 pandemic, caused by the SARS CoV-2 virus. Although the pursuit for a potent antiviral is still in progress, experimental therapies based on repurposing of existing drugs is being attempted. One important therapeutic target for COVID-19 is the main protease (Mpro) that cleaves the viral polyprotein in its replication process. Recently minocycline, an antimycobacterium drug, has been successfully implemented for the treatment of COVID-19 patients. But it's mode of action is still far from clear. Furthermore, it remains unresolved whether alternative antimycobacterium drugs can effectively regulate SARS CoV-2 by inhibiting the enzymatic activity of Mpro. To comprehend these facets, eight well-established antimycobacterium drugs were put through molecular docking experiments. Four of the antimycobacterium drugs (minocycline, rifampicin, clofazimine and ofloxacin) were selected by comparing their binding affinities towards Mpro. All of the four drugs interacted with both the catalytic residues of Mpro (His41 and Cys145). Additionally, molecular dynamics experiments demonstrated that the Mpro-minocyline complex has enhanced stability, experiences reduced conformational fluctuations and greater compactness than other three Mpro-antimycobacterium and Mpro-N3/lopinavir complexes. This research furnishes evidences for implementation of minocycline against SARS CoV-2. In addition, our findings also indicate other three antimycobacterium/antituberculosis drugs (rifampicin, clofazimine and ofloxacin) could potentially be evaluated for COVID-19 therapy., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. Repositioning fluphenazine as a cuproptosis-dependent anti-breast cancer drug candidate based on TCGA database.
- Author
-
Zhang X, Shi X, Zhang X, Zhang Y, Yu S, Zhang Y, and Liu Y
- Subjects
- Humans, Female, MCF-7 Cells, Copper, Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Databases, Genetic, Fluphenazine pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Drug Repositioning methods, Cell Survival drug effects
- Abstract
Breast cancer is one of the most prevalent malignancies among women. Enhancing the prognosis is an effective approach to enhance the survival rate of breast cancer. Cuproptosis, a copper-dependent programmed cell death process, has been associated with patient prognosis. Inducing cuproptosis is a promising approach for therapy. However, there is currently no anti-breast cancer drug that induces cuproptosis. In this study, we repositioned the clinical drug fluphenazine as a potential agent for breast cancer treatment by inducing cuproptosis. Firstly, we utilized the Cancer Genome Atlas (TCGA) database and Connectivity Map (CMap) database to identify 22 potential compounds with anti-breast cancer activity through inducing cuproptosis. Subsequently, our findings demonstrated that fluphenazine effectively suppressed the viability of MCF-7 cells. Fluphenazine also significantly inhibited the viability of triple negative breast cancer cells MDA-MB-453 and MDA-MB-231. Furthermore, our study revealed that fluphenazine significantly down-regulated the expression of potential prognostic biomarkers associated with cuproptosis, increased copper ion levels, and reduced intracellular pyruvate accumulation. Additionally, it up-regulated the expression of FDX1 at both the mRNA and protein levels, which has been reported to play a crucial role in the induction of cuproptosis. These findings suggest that fluphenazine has the potential to be used as an anti-breast cancer drug by inducing cuproptosis. Therefore, this research provides an insight for the development of novel cuproptosis-dependent anti-cancer agents., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. DVGEDR: a drug repositioning method based on dual-view fusion and graph enhancement mechanism in heterogeneous networks.
- Author
-
Niu, Dongjiang, Zhang, Lianwei, Zhang, Beiyi, Zhang, Qiang, Ding, Shanyang, Wei, Hai, and Li, Zhen
- Abstract
Drug repositioning, the discovery of new therapeutic uses for existing drugs, is increasingly gaining attention as a cost-effective and high-yield drug discovery strategy. Existing methods integrate diverse biological information into heterogeneous networks, providing a comprehensive framework for understanding complex drug–disease associations, which also will introduces noise into the data and affect the performance of the model. In this paper, first, a novel drug repositioning method, namely DVGEDR, is proposed, which generates two subgraphs of the target drug–disease pair to fuse biological information and integrate drug–disease associations from two distinct perspectives: drug–disease heterogeneous network and similarity networks. Next, a Multiple Attention Graph encoder (MAGencoder) module is designed to learn subgraph features and explore relationships between entities, which also improve the interpretability of the model. Finally, a graph enhancement mechanism is devised to improve the perception of critical information of model, enabling the model to flexibly process different graph structures. Performance comparisons with baseline models on three public datasets validate the state-of-the-art performance of DVGEDR in the field of drug repositioning. In case study, DVGEDR identifies 10 new candidate drugs for breast cancer and COVID-19, demonstrating not only superior performance in experimental settings but also potential therapeutic advantages in clinical environments. Furthermore, we select two sets of instances and further analyzed the attention distribution of the different nodes in the subgraph to explain the decision process of the model. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
25. Progress in the study of mefloquine as an antibiotic adjuvant for combination bacterial inhibition treatment.
- Author
-
Liang, Xiaofang, Liu, Zhihong, Wang, Yulin, Zhang, Yu, Deng, Wenbo, Liu, Qianqian, Lu, Zhangping, Li, Keke, Chang, Yanbing, and Wei, Lianhua
- Subjects
BACTERIAL cell membranes ,ANTITUBERCULAR agents ,MEFLOQUINE ,DRUG repositioning ,MYCOBACTERIUM tuberculosis - Abstract
Antimicrobial resistance is among the greatest threats to public health globally, and drug repurposing strategies may be advantageous to addressing this problem. Mefloquine, a drug traditionally used to treat malaria, has emerged as a promising antibiotic adjuvant, due to its ability to enhance the effectiveness of conventional antibiotics against resistant bacterial strains. In this paper, we first outline the enhancement properties of mefloquine and its mechanisms of action as an adjuvant antibiotic against multidrug-resistant bacteria. Mefloquine exhibits synergistic bacteriostatic effects when combined with colistin, β-lactams, antituberculosis drugs, quinolones, and linezolid. Potential mechanisms underlying its synergistic effects include inhibition of antibiotic efflux, disruption of bacterial cell membrane integrity, and disturbance of biofilm formation. In addition, we explore the bacteriostatic effects of several mefloquine derivatives against Mycobacterium tuberculosis and some fungi. Further, we summarize the findings of recent studies on other aspects of mefloquine activity, including its antiviral and antitumor effects. Finally, the advantages and challenges of mefloquine use as an antibiotic adjuvant in combination with antibiotics for bacterial inhibition are discussed. Overall, mefloquine shows excellent potential as an antibiotic adjuvant therapy against multidrug-resistant bacteria and is a promising candidate for combination therapy; however, further studies are needed to fully elucidate its mechanism of action and address the challenges associated with its clinical application. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Imidazoles and benzimidazoles as putative inhibitors of SARS-CoV-2 B.1.1.7 (Alpha) and P.1 (Gamma) variant spike glycoproteins: A computational approach
- Author
-
Vidyasrilekha Yele, Bharat Kumar Reddy. Sanapalli, and Afzal Azam Mohammed
- Subjects
Benzimidazole ,General Chemical Engineering ,Mutant ,Protein Data Bank (RCSB PDB) ,Flubendazole ,Biochemistry ,Molecular dynamic simulation study ,Industrial and Manufacturing Engineering ,chemistry.chemical_compound ,In vivo ,Materials Chemistry ,SARS-CoV-2 B.1.1.7 lineage (Alpha) ,chemistry.chemical_classification ,Original Paper ,Drug repositioning ,Imidazoles ,General Chemistry ,In vitro ,SARS-CoV-2 P.1 lineage (Gamma) ,Binding affinity ,chemistry ,Molecular docking ,Benzimidazoles ,Glycoprotein - Abstract
Graphic abstract COVID-19 is an unprecedented pandemic threatening global health, and variants were discovered rapidly after the pandemic. The two variants, namely the SARS-CoV-2 B.1.1.7 (Alpha) and P.1 (Gamma), were formed by the mutations in the receptor binding domain of spike glycoprotein (SGP). These two variants are known to possess a high binding affinity with the angiotensin-converting enzyme 2. Amidst the rapid spread of these mutant strains, research and development of novel molecules become tedious and labour-intensive. Imidazole and benzimidazole scaffolds were selected in this study based on their unique structural features and electron-rich environment, resulting in increased affinity against a variety of therapeutic targets. In the current study, imidazole- and benzimidazole-based anti-parasitic drugs are repurposed against SARS-CoV-2 Alpha and Gamma variant spike glycoproteins using computational strategies. Out of the screened 15 molecules, flubendazole and mebendazole have exhibited promising binding features to the two receptors (PDB ID: 7NEH and 7NXC), as evidenced by their glide score and binding free energy. The results are compared with that of the two standard drugs, remdesivir and hydroxychloroquine. Flubendazole and mebendazole have become convenient treatment options against mutant lineages of SARS-CoV-2. The edge of the flubendazole was further established by its stability in MD simulation conducted for 100 ns employing GROMACS software. Further, in vitro and in vivo studies are essential to understand, if flubendazole and mebendazole indeed hold the promise to manage SARS-CoV-2 mutant stains. Supplementary Information The online version contains supplementary material available at 10.1007/s11696-021-01900-8.
- Published
- 2021
27. Tumor microenvironment-based screening repurposes drugs targeting cancer stem cells and cancer-associated fibroblasts
- Author
-
Jia-Hua Lee, Hao Ho, Jeremy J.W. Chen, Wen-Chung Chu, Ker-Chau Li, Chao-Chi Ho, Pan-Chyr Yang, Yi-Chen Juan, Huei-Wen Chen, Hsuan-Yu Chen, Chiu-Hua Lin, Wan-Jiun Chen, Sheng-Fang Su, Gee-Chen Chang, Pei-Jung Lee, and Yi-Hua Lai
- Subjects
cancer stem cells ,Lung Neoplasms ,Drug Evaluation, Preclinical ,Medicine (miscellaneous) ,Metastasis ,Cancer-Associated Fibroblasts ,Cancer stem cell ,Cell Line, Tumor ,Tumor Microenvironment ,Medicine ,Humans ,drug screening ,Lung cancer ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,high-throughput ,Early Detection of Cancer ,Cell Proliferation ,Tumor microenvironment ,business.industry ,Drug Repositioning ,Cancer ,Fibroblasts ,medicine.disease ,Drug development ,Pharmaceutical Preparations ,Cancer cell ,Cancer research ,Neoplastic Stem Cells ,Drug Screening Assays, Antitumor ,business ,Research Paper - Abstract
The tumorous niche may drive the plasticity of heterogeneity and cancer stemness, leading to drug resistance and metastasis, which is the main reason of treatment failure in most cancer patients. The aim of this study was to establish a tumor microenvironment (TME)-based screening to identify drugs that can specifically target cancer stem cells (CSCs) and cancer-associated fibroblasts (CAFs) in the TME. Methods: Lung cancer patient-derived cancer cell and CAFs were utilized to mimic the TME and reproduce the stemness properties of CSCs in vitro and develop a high-throughput drug screening platform with phenotypical parameters. Limiting dilution assay, sphere-forming and ALDH activity assay were utilized to measure the cancer stemness characteristics. In vivo patient-derived xenograft (PDX) models and single-cell RNA sequencing were used to evaluate the mechanisms of the compounds in CSCs and CAFs. Results: The TME-based drug screening platform could comprehensively evaluate the response of cancer cells, CSCs and CAFs to different treatments. Among the 1,524 compounds tested, several drugs were identified to have anti-CAFs, anticancer and anti-CSCs activities. Aloe-emodin and digoxin both show anticancer and anti-CSCs activity in vitro and in vivo, which was further confirmed in the lung cancer PDX model. The combination of digoxin and chemotherapy improved therapeutic efficacy. The single-cell transcriptomics analysis revealed that digoxin could suppress the CSCs subpopulation in CAFs-cocultured cancer cells and cytokine production in CAFs. Conclusions: The TME-based drug screening platform provides a tool to identify and repurpose compounds targeting cancer cells, CSCs and CAFs, which may accelerate drug development and therapeutic application for lung cancer patients.
- Published
- 2021
28. Repurposing existing drugs for the treatment ofCOVID-19/SARS-CoV-2: A review of pharmacological effects and mechanism of action
- Author
-
Liang, Yutong, Quan, Xiaoxiao, Gu, Ruolan, Meng, Zhiyun, Gan, Hui, Wu, Zhuona, Sun, Yunbo, Pan, Huajie, Han, Peng, Liu, Shuchen, and Dou, Guifang
- Published
- 2024
- Full Text
- View/download PDF
29. Antiparasitic mebendazole (MBZ) effectively overcomes cisplatin resistance in human ovarian cancer cells by inhibiting multiple cancer-associated signaling pathways
- Author
-
Hue H. Luu, Tong-Chuan He, Ling Zhao, Connie Chen, Linjuan Huang, Fang He, William Wagstaff, Liangdan Tang, Qing Liu, Na Ni, Jing Zhang, Le Shen, Hui Wang, Deyao Shi, Rex C. Haydon, and Russell R. Reid
- Subjects
Aging ,Cell ,cisplatin ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Carcinoma, Ovarian Epithelial ,chemistry.chemical_compound ,Mice ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Tumor Stem Cell Assay ,Cell Proliferation ,Cisplatin ,Ovarian Neoplasms ,Wound Healing ,drug repurposing ,business.industry ,Cell growth ,Antinematodal Agents ,Drug Repositioning ,Cancer ,chemoresistance ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Drug repositioning ,Mebendazole ,medicine.anatomical_structure ,ovarian cancer ,Paclitaxel ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,Female ,cisplatin resistance ,mebendazole (MBZ) ,business ,Ovarian cancer ,medicine.drug ,Research Paper ,Signal Transduction - Abstract
Ovarian cancer is the third most common cancer and the second most common cause of gynecologic cancer death in women. Its routine clinical management includes surgical resection and systemic therapy with chemotherapeutics. While the first-line systemic therapy requires the combined use of platinum-based agents and paclitaxel, many ovarian cancer patients have recurrence and eventually succumb to chemoresistance. Thus, it is imperative to develop new strategies to overcome recurrence and chemoresistance of ovarian cancer. Repurposing previously-approved drugs is a cost-effective strategy for cancer drug discovery. The antiparasitic drug mebendazole (MBZ) is one of the most promising drugs with repurposing potential. Here, we investigate whether MBZ can overcome cisplatin resistance and sensitize chemoresistant ovarian cancer cells to cisplatin. We first established and characterized two stable and robust cisplatin-resistant (CR) human ovarian cancer lines and demonstrated that MBZ markedly inhibited cell proliferation, suppressed cell wounding healing/migration, and induced apoptosis in both parental and CR cells at low micromole range. Mechanistically, MBZ was revealed to inhibit multiple cancer-related signal pathways including ELK/SRF, NFKB, MYC/MAX, and E2F/DP1 in cisplatin-resistant ovarian cancer cells. We further showed that MBZ synergized with cisplatin to suppress cell proliferation, induce cell apoptosis, and blunt tumor growth in xenograft tumor model of human cisplatin-resistant ovarian cancer cells. Collectively, our findings suggest that MBZ may be repurposed as a synergistic sensitizer of cisplatin in treating chemoresistant human ovarian cancer, which warrants further clinical studies.
- Published
- 2021
30. Drug repurposing against breast cancer by integrating drug-exposure expression profiles and drug–drug links based on graph neural network
- Author
-
Xiaomin Luo, Dingyan Wang, Fu Xiao, Xiaoyu Ding, Lifan Chen, Mingyue Zheng, Hualiang Jiang, Chen Cui, Kaixian Chen, and Tingyang Xu
- Subjects
Statistics and Probability ,Drug ,AcademicSubjects/SCI01060 ,Computer science ,media_common.quotation_subject ,MEDLINE ,Disease ,Computational biology ,Biochemistry ,Causes of cancer ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,medicine ,Molecular Biology ,Repurposing ,030304 developmental biology ,media_common ,0303 health sciences ,Systems Biology ,Cancer ,medicine.disease ,Original Papers ,Computer Science Applications ,Computational Mathematics ,Drug repositioning ,Computational Theory and Mathematics ,030220 oncology & carcinogenesis - Abstract
Motivation Breast cancer is one of the leading causes of cancer deaths among women worldwide. It is necessary to develop new breast cancer drugs because of the shortcomings of existing therapies. The traditional discovery process is time-consuming and expensive. Repositioning of clinically approved drugs has emerged as a novel approach for breast cancer therapy. However, serendipitous or experiential repurposing cannot be used as a routine method. Results In this study, we proposed a graph neural network model GraphRepur based on GraphSAGE for drug repurposing against breast cancer. GraphRepur integrated two major classes of computational methods, drug network-based and drug signature-based. The differentially expressed genes of disease, drug-exposure gene expression data and the drug–drug links information were collected. By extracting the drug signatures and topological structure information contained in the drug relationships, GraphRepur can predict new drugs for breast cancer, outperforming previous state-of-the-art approaches and some classic machine learning methods. The high-ranked drugs have indeed been reported as new uses for breast cancer treatment recently. Availabilityand implementation The source code of our model and datasets are available at: https://github.com/cckamy/GraphRepur and https://figshare.com/articles/software/GraphRepur_Breast_Cancer_Drug_Repurposing/14220050. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2021
- Full Text
- View/download PDF
31. Screening of Clinically Approved and Investigation Drugs as Potential Inhibitors of SARS‐CoV‐2: A Combined in silico and in vitro Study
- Author
-
Timucin Avsar, Berna Dogan, Muge Didem Orhan, Seyma Calis, Serdar Durdagi, Busecan Aksoydan, Necla Birgül Iyison, Aida Shahraki, and Kader Sahin
- Subjects
COVID19 ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Cefpiramide ,Pharmacology ,Antiviral Agents ,Cefotiam ,chemistry.chemical_compound ,Structural Biology ,Drug Discovery ,medicine ,Humans ,Rotigaptide ,Coronavirus 3C Proteases ,Spike/ACE-2 ,Virtual screening ,MD simulations ,Protease ,Ritonavir ,Full Paper ,drug repurposing ,SARS-CoV-2 ,Organic Chemistry ,Drug Repositioning ,Denopamine ,Drugs, Investigational ,Full Papers ,virtual screening ,Computer Science Applications ,COVID-19 Drug Treatment ,Molecular Docking Simulation ,chemistry ,Docking (molecular) ,main protease ,docking ,Spike Glycoprotein, Coronavirus ,Molecular Medicine ,Angiotensin-Converting Enzyme 2 ,medicine.drug - Abstract
In the current study, we used 7922 FDA approved small molecule drugs as well as compounds in clinical investigation from NIH's NPC database in our drug repurposing study. SARS‐CoV‐2 main protease as well as Spike protein/ACE2 targets were used in virtual screening and top‐100 compounds from each docking simulations were considered initially in short molecular dynamics (MD) simulations and their average binding energies were calculated by MM/GBSA method. Promising hit compounds selected based on average MM/GBSA scores were then used in long MD simulations. Based on these numerical calculations following compounds were found as hit inhibitors for the SARS‐CoV‐2 main protease: Pinokalant, terlakiren, ritonavir, cefotiam, telinavir, rotigaptide, and cefpiramide. In addition, following 3 compounds were identified as inhibitors for Spike/ACE2: Denopamine, bometolol, and rotigaptide. In order to verify the predictions of in silico analyses, 4 compounds (ritonavir, rotigaptide, cefotiam, and cefpiramide) for the main protease and 2 compounds (rotigaptide and denopamine) for the Spike/ACE2 interactions were tested by in vitro experiments. While the concentration‐dependent inhibition of the ritonavir, rotigaptide, and cefotiam was observed for the main protease; denopamine was effective at the inhibition of Spike/ACE2 binding.
- Published
- 2021
32. Repurposing an anti‐cancer agent for the treatment of hypertrophic heart disease
- Author
-
Adrian J. Hobbs, Reshma S. Baliga, Gabriela D'Amico, Kenneth Bedi, Matthew Dukinfield, Kenneth B. Margulies, Kairbaan Hodivala-Dilke, Eleni Maniati, Louise E. Reynolds, Oscar Maiques, Jun Wang, Victoria Sanz-Moreno, and Aisah A. Aubdool
- Subjects
Male ,0301 basic medicine ,Heart disease ,Angiogenesis ,heart failure ,cardiomyocyte ,Cilengitide ,Muscle hypertrophy ,angiogenesis ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Myocytes, Cardiac ,Cells, Cultured ,Angiotensin II ,Original Papers ,3. Good health ,Endothelial stem cell ,030220 oncology & carcinogenesis ,ischaemia ,hypertrophy ,Signal Transduction ,Snake Venoms ,Ischemia ,Neovascularization, Physiologic ,Cardiomegaly ,Pathology and Forensic Medicine ,blood vessels ,03 medical and health sciences ,Renin–angiotensin system ,medicine ,Animals ,Humans ,Original Paper ,business.industry ,Drug Repositioning ,Cardiovascular Agents ,Recovery of Function ,Integrin alphaVbeta3 ,medicine.disease ,Fibrosis ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,chemistry ,cilengitide ,Case-Control Studies ,Heart failure ,integrins ,Cancer research ,Transcriptome ,business - Abstract
Coronary microvascular dysfunction combined with maladaptive cardiomyocyte morphology and energetics is a major contributor to heart failure advancement. Thus, dually enhancing cardiac angiogenesis and targeting cardiomyocyte function to slow, or reverse, the development of heart failure is a logical step towards improved therapy. We present evidence for the potential to repurpose a former anti‐cancer Arg‐Gly‐Asp (RGD)‐mimetic pentapeptide, cilengitide, here used at low doses. Cilengitide targets αvβ3 integrin and this protein is upregulated in human dilated and ischaemic cardiomyopathies. Treatment of mice after abdominal aortic constriction (AAC) surgery with low‐dose cilengitide (ldCil) enhances coronary angiogenesis and directly affects cardiomyocyte hypertrophy with an associated reduction in disease severity. At a molecular level, ldCil treatment has a direct effect on cardiac endothelial cell transcriptomic profiles, with a significant enhancement of pro‐angiogenic signalling pathways, corroborating the enhanced angiogenic phenotype after ldCil treatment. Moreover, ldCil treatment of Angiotensin II‐stimulated AngII‐stimulated cardiomyocytes significantly restores transcriptomic profiles similar to those found in normal human heart. The significance of this finding is enhanced by transcriptional similarities between AngII‐treated cardiomyocytes and failing human hearts. Taken together, our data provide evidence supporting a possible new strategy for improved heart failure treatment using low‐dose RGD‐mimetics with relevance to human disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
- Published
- 2019
- Full Text
- View/download PDF
33. COVID-19: molecular pathophysiology, genetic evolution and prospective therapeutics—a review
- Author
-
C. T. Dhanya Raj, Rathinam Arthur James, Raju Rajasabapathy, Ravi Chandra Sekhara Reddy Danduga, and Dinesh Kumar Kandaswamy
- Subjects
Biochemistry ,Dexamethasone ,Lopinavir ,0302 clinical medicine ,Medicine ,Prospective Studies ,030212 general & internal medicine ,Phylogeny ,Repurposing ,media_common ,0303 health sciences ,Alanine ,Drug discovery ,Chloroquine ,General Medicine ,Drug repositioning ,Pyrazines ,Molecular targets ,Hydroxychloroquine ,medicine.drug ,Drug ,medicine.medical_specialty ,Ribavarin ,Drug repositioning/repurposing ,media_common.quotation_subject ,Remdesivir ,Favipiravir ,Antiviral Agents ,Microbiology ,Evolution, Molecular ,03 medical and health sciences ,Ribavirin ,Genetics ,Animals ,Humans ,Intensive care medicine ,Adverse effect ,Pandemics ,Molecular Biology ,COVID-19 Serotherapy ,030304 developmental biology ,QR355 ,Original Paper ,Ritonavir ,SARS-CoV-2 ,business.industry ,Drug Repositioning ,Immunization, Passive ,COVID-19 ,Amides ,R1 ,Adenosine Monophosphate ,QR ,Clinical trial ,business - Abstract
The Covid-19 pandemic is highly contagious and has spread rapidly across the globe. To date there have been no specific treatment options available for this life-threatening disease. During this medical emergency, target-based drug repositioning/repurposing with a continuous monitoring and recording of results is an effective method for the treatment and drug discovery. This review summarizes the recent findings on COVID-19, its genomic organization, molecular evolution through phylogenetic analysis and has recapitulated the drug targets by analyzing the viral molecular machinery as drug targets and repurposing of most frequently used drugs worldwide and their therapeutic applications in COVID-19. Data from solidarity trials have shown that the treatment with Chloroquine, hydroxychloroquine and lopinavir-ritonavir had no effect in reducing the mortality rate and also had adverse side effects. Remdesivir, Favipiravir and Ribavirin might be a safer therapeutic option for COVID-19. Recent clinical trial has revealed that dexamethasone and convalescent plasma treatment can reduce mortality in patients with severe forms of COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
34. Microbial and genetic-based framework identifies drug targets in inflammatory bowel disease
- Author
-
Na Xie, Pan Gao, Qinqin Pu, Canhua Huang, Zhihan Wang, Junguk Hur, Changlong Li, Kai Guo, Shugang Qin, Min Wu, and Ping Lin
- Subjects
0301 basic medicine ,Medicine (miscellaneous) ,Computational biology ,Gut flora ,digestive system ,Inflammatory bowel disease ,Mice ,cyclic GMP-AMP synthase (cGAS) ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,medicine ,Animals ,Humans ,Microbiome ,Colitis ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,brefeldin-a ,Repurposing ,Gene knockout ,host transcriptome-microbiome interaction ,Mice, Knockout ,drug repurposing ,biology ,business.industry ,Inflammatory Bowel Diseases ,medicine.disease ,biology.organism_classification ,Nucleotidyltransferases ,digestive system diseases ,Gastrointestinal Microbiome ,Drug repositioning ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Gene Knockout Techniques ,business ,Biomarkers ,Research Paper - Abstract
Rationale: With increasing incidence and prevalence of inflammatory bowel disease (IBD), it has become one of the major public health threats, and there is an urgent need to develop new therapeutic agents. Although the pathogenesis of IBD is still unclear, previous research has provided evidence for complex interplays between genetic, immune, microbial, and environmental factors. Here, we constructed a gene-microbiota interaction-based framework to discover IBD biomarkers and therapeutics. Methods: We identified candidate biomarkers for IBD by analyzing the publicly available transcriptomic and microbiome data from IBD cohorts. Animal models of IBD and diarrhea were established. The inflammation-correlated microbial and genetic variants in gene knockout mice were identified by 16S rRNA sequences and PCR array. We performed bioinformatic analysis of microbiome functional prediction and drug repurposing. Our validation experiments with cells and animals confirmed anti-inflammatory properties of a drug candidate. Results: We identified the DNA-sensing enzyme cyclic GMP-AMP synthase (cGAS) as a potential biomarker for IBD in both patients and murine models. cGAS knockout mice were less susceptible to DSS-induced colitis. cGAS-associated gut microbiota and host genetic factors relating to IBD pathogenesis were also identified. Using a computational drug repurposing approach, we predicted 43 candidate drugs with high potency to reverse colitis-associated gene expression and validated that brefeldin-a mitigates inflammatory response in colitis mouse model and colon cancer cell lines. Conclusions: By integrating computational screening, microbiota interference, gene knockout techniques, and in vitro and in vivo validation, we built a framework for predicting biomarkers and host-microbe interaction targets and identifying repurposing drugs for IBD, which may be tested further for clinical application. This approach may also be a tool for repurposing drugs for treating other diseases.
- Published
- 2021
- Full Text
- View/download PDF
35. Insights into idarubicin antimicrobial activity against methicillin-resistant Staphylococcus aureus
- Author
-
Zhen Luo, Ti Chen, Jinfeng Liao, Pengfei She, Linying Zhou, Shijia Li, Lanlan Xu, Yaqian Liu, Yong Wu, and Xianghai Zeng
- Subjects
Microbiology (medical) ,topoisomerase ii ,Multidrug tolerance ,Immunology ,Microbial Sensitivity Tests ,idarubicin ,methicillin-resistant staphylococcus aureus ,Infectious and parasitic diseases ,RC109-216 ,Biology ,Molecular Dynamics Simulation ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,Mice ,Antibiotic resistance ,Fosfomycin ,medicine ,Idarubicin ,Animals ,030304 developmental biology ,0303 health sciences ,drug repurposing ,030306 microbiology ,Biofilm ,Drug Repositioning ,Drug Synergism ,biochemical phenomena, metabolism, and nutrition ,Antimicrobial ,Methicillin-resistant Staphylococcus aureus ,skin and soft tissue infections ,Anti-Bacterial Agents ,Specific Pathogen-Free Organisms ,Mice, Inbred C57BL ,Drug repositioning ,Infectious Diseases ,Biofilms ,Community setting ,Parasitology ,Female ,Staphylococcal Skin Infections ,cell membrane ,medicine.drug ,Research Article ,Research Paper - Abstract
Background MRSA is a major concern in community settings and in health care. The emergence of biofilms and persister cells substantially increases its antimicrobial resistance. It is very urgent to develop new antimicrobials to solve this problem. Objective Idarubicin was profiled to assess its antimicrobial effects in vitro and in vivo, and the underlying mechanisms. Methods We investigated the antimicrobial effects of idarubicin against MRSA by time-kill analysis. The antibiofilm efficacy of idarubicin was assessed by crystal violet and XTT staining, followed by laser confocal microscopy observation. The mechanisms underlying the antimicrobial effects were studied by transmission electron microscopy, all-atom molecular dynamic simulations, SYTOX staining, surface plasma resonance, and DNA gyrase inhibition assay. Further, we addressed the antimicrobial efficacy in wound and subcutaneous abscess infection in vivo. Results Idarubicin kills MRSA cells by disrupting the lipid bilayers and interrupting the DNA topoisomerase IIA subunits, and idarubicin shows synergistic antimicrobial effects with fosfomycin. Through synergy with a single dose treatment fosfomycin and the addition of the cell protector amifostine, the cytotoxicity and cardiotoxicity of idarubicin were significantly reduced without affecting its antimicrobial effects. Idarubicin alone or in combination with fosfomycin exhibited considerable efficacy in a subcutaneous abscess mouse model of MRSA infection. In addition, idarubicin also showed a low probability of causing resistance and good postantibiotic effects. Conclusions Idarubicin and its analogs have the potential to become a new class of antimicrobials for the treatment of MRSA-related infections.
- Published
- 2020
36. Digitoxigenin presents an effective and selective antileishmanial action against Leishmania infantum and is a potential therapeutic agent for visceral leishmaniasis
- Author
-
Daniela P. Lage, Rafaella R Costa, Gabriela Silva Ramos, João A. Oliveira-da-Silva, Débora V.C. Mendonça, Rodrigo Maia de Pádua, Bruno Mendes Roatt, Rory Cristiane Fortes de Brito, Fernanda F. Ramos, Priscilla R. V. Campana, Fernão Castro Braga, Camila S. Freitas, Amanda S. Machado, Fernanda Ludolf, Luciana M.R. Antinarelli, Flaviano Melo Ottoni, Miguel A. Chávez-Fumagalli, Elaine Soares Coimbra, Thiago A.R. Reis, Vinicio T.S. Coelho, Maria Victoria Humbert, Jennifer Munkert, Vívian T. Martins, Grasiele S.V. Tavares, and Eduardo A.F. Coelho
- Subjects
Pharmacology ,Parasite Load ,030308 mycology & parasitology ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Amphotericin B ,Amphotericin B deoxycholate ,Leishmania infantum ,Micelles ,Membrane Potential, Mitochondrial ,Visceral leishmaniasis ,Mice, Inbred BALB C ,0303 health sciences ,biology ,General Medicine ,Drug Combinations ,Infectious Diseases ,Liver ,Leishmaniasis, Visceral ,Female ,Deoxycholic Acid ,medicine.drug ,030231 tropical medicine ,Antiprotozoal Agents ,Poloxamer ,Treatment and Prophylaxis - Original Paper ,03 medical and health sciences ,Digitalis lanata ,medicine ,Animals ,Digitoxigenin ,Miltefosine ,General Veterinary ,Macrophages ,Drug repositioning ,biology.organism_classification ,Leishmania ,medicine.disease ,Treatment ,chemistry ,Insect Science ,Parasitology ,Reactive Oxygen Species ,Spleen - Abstract
Treatment for visceral leishmaniasis (VL) is hampered mainly by drug toxicity, their high cost, and parasite resistance. Drug development is a long and pricey process, and therefore, drug repositioning may be an alternative worth pursuing. Cardenolides are used to treat cardiac diseases, especially those obtained from Digitalis species. In the present study, cardenolide digitoxigenin (DIGI) obtained from a methanolic extract of Digitalis lanata leaves was tested for its antileishmanial activity against Leishmania infantum species. Results showed that 50% Leishmania and murine macrophage inhibitory concentrations (IC50 and CC50, respectively) were of 6.9 ± 1.5 and 295.3 ± 14.5 μg/mL, respectively. With amphotericin B (AmpB) deoxycholate, used as a control drug, values of 0.13 ± 0.02 and 0.79 ± 0.12 μg/mL, respectively, were observed. Selectivity index (SI) values were of 42.8 and 6.1 for DIGI and AmpB, respectively. Preliminary studies suggested that the mechanism of action for DIGI is to cause alterations in the mitochondrial membrane potential, to increase the levels of reactive oxygen species and induce accumulation of lipid bodies in the parasites. DIGI was incorporated into Pluronic® F127-based polymeric micelles, and the formula (DIGI/Mic) was used to treat L. infantum–infected mice. Miltefosine was used as a control drug. Results showed that animals treated with either miltefosine, DIGI, or DIGI/Mic presented significant reductions in the parasite load in their spleens, livers, bone marrows, and draining lymph nodes, as well as the development of a specific Th1-type response, when compared with the controls. Results obtained 1 day after treatment were corroborated with data corresponding to 15 days after therapy. Importantly, treatment with DIGI/Mic induced better parasitological and immunological responses when compared with miltefosine- and DIGI-treated mice. In conclusion, DIGI/Mic has the potential to be used as a therapeutic agent to protect against L. infantum infection, and it is therefore worth of consideration in future studies addressing VL treatment.
- Published
- 2020
- Full Text
- View/download PDF
37. Target RNA modification for epigenetic drug repositioning in neuroblastoma: computational omics proximity between repurposing drug and disease
- Author
-
Peigen Xiao, Da-Cheng Hao, Lifeng Yue, Zhihong Wu, Guanhua Du, Lijia Xu, Pei Ma, and Sen Zhang
- Subjects
Aging ,drug repurposing ,DNA repair ,RNA ,Cell Biology ,Computational biology ,Biology ,Cell cycle ,Transcriptome ,L1000 FWD ,Drug repositioning ,neuroblastoma ,Gene expression ,gene expression ,the connectivity map ,Epigenetics ,Gene ,Research Paper - Abstract
RNA modifications modulate most steps of gene expression. However, little is known about its role in neuroblastoma (NBL) and the inhibitors targeting it. We analyzed the RNA-seq (n=122) and CNV data (n=78) from NBL patients in Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. The NBL sub-clusters (cluster1/2) were identified via consensus clustering for expression of RNA modification regulators (RNA-MRs). Cox regression, principle component analysis and chi-square analysis were used to compare differences of survival, transcriptome, and clinicopathology between clusters. Cluster1 showed significantly poor prognosis, of which RNA-MRs' expression and CNV alteration were closely related to pathologic stage. RNA-MRs and functional related prognostic genes were obtained using spearman correlation analysis, and queried in CMap and L1000 FWD database to obtain 88 inhibitors. The effects of 5 inhibitors on RNA-MRs were confirmed in SH-SY5Y cells. The RNA-MRs exhibited two complementary regulation functions: one conducted by TET2 and related to translation and glycolysis; another conducted by ALYREF, NSUN2 and ADARB1 and related to cell cycle and DNA repair. The perturbed proteomic profile of HDAC inhibitors was different from that of others, thus drug combination overcame drug resistance and was potential for NBL therapy with RNA-MRs as therapeutic targets.
- Published
- 2020
38. Coupled immune stratification and identification of therapeutic candidates in patients with lung adenocarcinoma
- Author
-
Lonny Yarmus, Yundi Chen, Yuan Wan, Weilei Hu, Biao Liu, and Guosheng Wang
- Subjects
Drug ,Oncology ,Aging ,medicine.medical_specialty ,Lung Neoplasms ,media_common.quotation_subject ,medicine.medical_treatment ,Clinical Decision-Making ,Adenocarcinoma of Lung ,Antineoplastic Agents ,medicine.disease_cause ,cold tumor ,Immune system ,Cancer immunotherapy ,Predictive Value of Tests ,Internal medicine ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Humans ,Gene Regulatory Networks ,Molecular Targeted Therapy ,Precision Medicine ,media_common ,Tumor microenvironment ,business.industry ,Gene Expression Profiling ,Drug Repositioning ,Cell Biology ,Immunotherapy ,personalized cancer immunotherapies ,patient stratification ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Molecular Docking Simulation ,Drug repositioning ,A549 Cells ,Adenocarcinoma ,Transcriptome ,business ,Carcinogenesis ,Research Paper - Abstract
In recent years, personalized cancer immunotherapy, especially stratification-driven precision treatments have gained significant traction. However, due to the heterogeneity in clinical cohorts, the uncombined analysis of stratification/therapeutics may lead to confusion in determining ideal therapeutic options. We report that the coupled immune stratification and drug repurposing could facilitate identification of therapeutic candidates in patients with lung adenocarcinoma (LUAD). First, we categorized the patients into four groups based on immune gene profiling, associated with distinct molecular characteristics and clinical outcomes. Then, the weighted gene co-expression network analysis (WGCNA) algorithm was used to identify co-expression modules of each groups. We focused on C3 group which is characterized by low immune infiltration (cold tumor) and wild-type EGFR, posing a significant challenge for treatment of LUAD. Five drug candidates against the C3 status were identified which have potential dual functions to correct aberrant immune microenvironment and also halt tumorigenesis. Furthermore, their steady binding affinity against the targets was verified through molecular docking analysis. In sum, our findings suggest that such coupled analysis could be a promising methodology for identification and exploration of therapeutic candidates in the practice of personalized immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
39. Losartan improves the therapeutic effect of metronomic cyclophosphamide in triple negative mammary cancer models
- Author
-
Julian Guercetti, Antonela Del Giúdice, María Virginia Baglioni, O. Graciela Scharovsky, Monica Carolina Grillo, Leandro Ernesto Mainetti, Cintia Daniela Kaufman, María J. Rico, Maria Celeste Capitani, Viviana R. Rozados, and Matias Fusini
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Cyclophosphamide ,losartan ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,mammary cancer ,Internal medicine ,metronomic chemotherapy ,medicine ,Triple-negative breast cancer ,Chemotherapy ,drug repurposing ,business.industry ,Therapeutic effect ,Metronomic Chemotherapy ,Drug repositioning ,Regimen ,030104 developmental biology ,Losartan ,030220 oncology & carcinogenesis ,cyclophosphamide ,business ,medicine.drug ,Research Paper - Abstract
Metronomic chemotherapy refers to the minimum biologically effective doses of a chemotherapy agent given as a continuous regimen without extended rest periods. Drug repurposing is defined as the use of an already known drug for a new medical indication, different from the original one. In oncology the combination of these two therapeutic approaches is called "Metronomics". The aim of this work is to evaluate the therapeutic effect of cyclophosphamide in a metronomic schedule in combination with the repurposed drug losartan in two genetically different mice models of triple negative breast cancer. Our findings showed that adding losartan to metronomic cyclophosphamide significantly improved the therapeutic outcome. In both models the combined treatment increased the mice's survival without sings of toxicity. Moreover, we elucidated some of the mechanisms of action involved, which include a decrease of intratumor hypoxia, stimulation of the immune response and remodeling of the tumor microenvironment. The remarkable therapeutic effect, the lack of toxicity, the low cost of the drugs and its oral administration, strongly suggest its translation to the clinical setting in the near future.
- Published
- 2020
40. Revealing new therapeutic opportunities through drug target prediction: a class imbalance-tolerant machine learning approach
- Author
-
Haiyuan Yu and Siqi Liang
- Subjects
Statistics and Probability ,Drug ,Computer science ,In silico ,media_common.quotation_subject ,Drug target ,Druggability ,Machine learning ,computer.software_genre ,Biochemistry ,Genome ,Machine Learning ,03 medical and health sciences ,Class imbalance ,0302 clinical medicine ,Computer Simulation ,Molecular Biology ,Gene ,030304 developmental biology ,media_common ,0303 health sciences ,business.industry ,Drug Repositioning ,Computational Biology ,Original Papers ,Computer Science Applications ,Computational Mathematics ,Drug repositioning ,Computational Theory and Mathematics ,030220 oncology & carcinogenesis ,Artificial intelligence ,business ,computer ,Classifier (UML) - Abstract
Motivation In silico drug target prediction provides valuable information for drug repurposing, understanding of side effects as well as expansion of the druggable genome. In particular, discovery of actionable drug targets is critical to developing targeted therapies for diseases. Results Here, we develop a robust method for drug target prediction by leveraging a class imbalance-tolerant machine learning framework with a novel training scheme. We incorporate novel features, including drug–gene phenotype similarity and gene expression profile similarity that capture information orthogonal to other features. We show that our classifier achieves robust performance and is able to predict gene targets for new drugs as well as drugs that potentially target unexplored genes. By providing newly predicted drug–target associations, we uncover novel opportunities of drug repurposing that may benefit cancer treatment through action on either known drug targets or currently undrugged genes. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2020
- Full Text
- View/download PDF
41. Repurposing Antiviral Drugs for Orthogonal RNA‐Catalyzed Labeling of RNA
- Author
-
Ann-Kathrin Lenz, Surjendu Dey, Claudia Höbartner, and Mohammad Ghaem Maghami
- Subjects
site-specific RNA labeling ,Guanosine ,ribozymes ,010402 general chemistry ,Antiviral Agents ,01 natural sciences ,Catalysis ,chemistry.chemical_compound ,Transferases ,Ribozymes | Hot Paper ,Transferase ,RNA, Catalytic ,Nucleotide ,in vitro selection ,chemistry.chemical_classification ,biology ,010405 organic chemistry ,Chemistry ,Communication ,Drug Repositioning ,Ribozyme ,RNA ,General Medicine ,General Chemistry ,Communications ,tenofovir ,0104 chemical sciences ,3. Good health ,Biochemistry ,Phosphodiester bond ,Biocatalysis ,biology.protein ,Nucleic acid ,Bioorthogonal chemistry ,antiviral nucleoside analogues - Abstract
In vitro selected ribozymes are promising tools for site‐specific labeling of RNA. Previously known nucleic acid catalysts attached fluorescently labeled adenosine or guanosine derivatives through 2′,5′‐branched phosphodiester bonds to the RNA of interest. Herein, we report new ribozymes that use orthogonal substrates, derived from the antiviral drug tenofovir, and attach bioorthogonal functional groups, as well as affinity handles and fluorescent reporter units through a hydrolytically more stable phosphonate ester linkage. The tenofovir transferase ribozymes were identified by in vitro selection and are orthogonal to nucleotide transferase ribozymes. As genetically encodable functional RNAs, these ribozymes may be developed for potential cellular applications. The orthogonal ribozymes addressed desired target sites in large RNAs in vitro, as shown by fluorescent labeling of E. coli 16S and 23S rRNAs in total cellular RNA., A strategy for illuminating RNA with fluorescent derivatives of the antiviral acyclic nucleoside phosphonate Tenofovir is presented. Tenofovir transferase ribozymes were identified by in vitro selection and are shown to be orthogonal to nucleotidyl transferase ribozymes, thus enabling site‐specific dual labeling of RNA.
- Published
- 2020
- Full Text
- View/download PDF
42. Drug Repurposing for the SARS-CoV-2 Papain-Like Protease
- Author
-
Tyler Lalonde, Ge Yu, Shuhua G Li, Wenshe R. Liu, Kai S Yang, Chia-Chuan Cho, Yuchen Qiao, and Shiqing Xu
- Subjects
Proteases ,medicine.medical_treatment ,Allosteric regulation ,Coronavirus Papain-Like Proteases ,papain-like protease ,Cysteine Proteinase Inhibitors ,Biochemistry ,Antiviral Agents ,Deubiquitinating enzyme ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Drug Discovery ,medicine ,cysteine protease ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Pharmacology ,chemistry.chemical_classification ,Protease ,biology ,Full Paper ,SARS-CoV-2 ,Organic Chemistry ,Drug Repositioning ,COVID-19 ,Full Papers ,Cysteine protease ,Papain ,deubiquitinase ,Enzyme ,chemistry ,biology.protein ,Molecular Medicine ,Cysteine - Abstract
As the pathogen of COVID‐19, SARS‐CoV‐2 encodes two essential cysteine proteases that process the pathogen's two large polypeptide products pp1a and pp1ab in the human cell host to form 15 functionally important, mature nonstructural proteins. One of the two enzymes is papain‐like protease or PLPro. It possesses deubiquitination and deISGylation activities that suppress host innate immune responses toward SARS‐CoV‐2 infection. To repurpose drugs for PLPro, we experimentally screened libraries of 33 deubiquitinase and 37 cysteine protease inhibitors on their inhibition of PLPro. Our results showed that 15 deubiquitinase and 1 cysteine protease inhibitors exhibit strong inhibition of PLPro at 200 μM. More comprehensive characterizations revealed seven inhibitors GRL0617, SJB2‐043, TCID, DUB‐IN‐1, DUB‐IN‐3, PR‐619, and S130 with an IC50 value below 40 μM and four inhibitors GRL0617, SJB2‐043, TCID, and PR‐619 with an IC50 value below 10 μM. Among four inhibitors with an IC50 value below 10 μM, SJB2‐043 is the most unique in that it does not fully inhibit PLPro but has a noteworthy IC50 value of 0.56 μM. SJB2‐043 likely binds to an allosteric site of PLPro to convene its inhibition effect, which needs to be further investigated. As a pilot study, the current work indicates that COVID‐19 drug repurposing by targeting PLPro holds promise, but in‐depth analysis of repurposed drugs is necessary to avoid omitting critical allosteric inhibitors., Papain‐like protease (PLpro) is a cysteine protease with deubiquitination and deISGylation activities, and is essential for SARS‐CoV‐2 replication and pathogenesis. By screening 33 deubiquitinase inhibitors and 37 cysteine protease inhibitors, we discovered that a number of molecules are potent PLpro inhibitors such as TCID, PR‐619, and SJB2‐043. Among these inhibitors, SJB2‐043 has an outstanding IC50 value of 0.56 μM, but only shows partial inhibition of papain‐like protease. It binds likely to an allosteric site of PLpro to convene its inhibition effect.
- Published
- 2021
43. Drug repositioning based on residual attention network and free multiscale adversarial training.
- Author
-
Li G, Li S, Liang C, Xiao Q, and Luo J
- Subjects
- Humans, Computational Biology methods, Algorithms, Neural Networks, Computer, Drug Repositioning methods
- Abstract
Background: Conducting traditional wet experiments to guide drug development is an expensive, time-consuming and risky process. Analyzing drug function and repositioning plays a key role in identifying new therapeutic potential of approved drugs and discovering therapeutic approaches for untreated diseases. Exploring drug-disease associations has far-reaching implications for identifying disease pathogenesis and treatment. However, reliable detection of drug-disease relationships via traditional methods is costly and slow. Therefore, investigations into computational methods for predicting drug-disease associations are currently needed., Results: This paper presents a novel drug-disease association prediction method, RAFGAE. First, RAFGAE integrates known associations between diseases and drugs into a bipartite network. Second, RAFGAE designs the Re_GAT framework, which includes multilayer graph attention networks (GATs) and two residual networks. The multilayer GATs are utilized for learning the node embeddings, which is achieved by aggregating information from multihop neighbors. The two residual networks are used to alleviate the deep network oversmoothing problem, and an attention mechanism is introduced to combine the node embeddings from different attention layers. Third, two graph autoencoders (GAEs) with collaborative training are constructed to simulate label propagation to predict potential associations. On this basis, free multiscale adversarial training (FMAT) is introduced. FMAT enhances node feature quality through small gradient adversarial perturbation iterations, improving the prediction performance. Finally, tenfold cross-validations on two benchmark datasets show that RAFGAE outperforms current methods. In addition, case studies have confirmed that RAFGAE can detect novel drug-disease associations., Conclusions: The comprehensive experimental results validate the utility and accuracy of RAFGAE. We believe that this method may serve as an excellent predictor for identifying unobserved disease-drug associations., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
44. Exploring therapeutic approaches against Naegleria fowleri infections through the COVID box.
- Author
-
Chao-Pellicer J, Arberas-Jiménez I, Sifaoui I, Piñero JE, and Lorenzo-Morales J
- Subjects
- Humans, Amebicides pharmacology, Amebicides therapeutic use, COVID-19, Naegleria fowleri drug effects, Drug Repositioning, Central Nervous System Protozoal Infections drug therapy, Central Nervous System Protozoal Infections parasitology
- Abstract
Naegleria fowleri, known as the brain-eating amoeba, is the pathogen that causes the primary amoebic meningoencephalitis (PAM), a severe neurodegenerative disease with a fatality rate exceeding 95%. Moreover, PAM cases commonly involved previous activities in warm freshwater bodies that allow amoebae-containing water through the nasal passages. Hence, awareness among healthcare professionals and the general public are the key to contribute to a higher and faster number of diagnoses worldwide. Current treatment options for PAM, such as amphotericin B and miltefosine, are limited by potential cytotoxic effects. In this context, the repurposing of existing compounds has emerged as a promising strategy. In this study, the evaluation of the COVID Box which contains 160 compounds demonstrated significant in vitro amoebicidal activity against two type strains of N. fowleri. From these compounds, terconazole, clemastine, ABT-239 and PD-144418 showed a higher selectivity against the parasite compared to the remaining products. In addition, programmed cell death assays were conducted with these four compounds, unveiling compatible metabolic events in treated amoebae. These compounds exhibited chromatin condensation and alterations in cell membrane permeability, indicating their potential to induce programmed cell death. Assessment of mitochondrial membrane potential disruption and a significant reduction in ATP production emphasized the impact of these compounds on the mitochondria, with the identification of increased ROS production underscoring their potential as effective treatment options. This study emphasizes the potential of the mentioned COVID Box compounds against N. fowleri, providing a path for enhanced PAM therapies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
45. Identification of therapeutic drug target of Shigella Flexneri serotype X through subtractive genomic approach and in-silico screening based on drug repurposing.
- Author
-
Ahmed MH, Khan K, Tauseef S, Jalal K, Haroon U, Uddin R, Abdellattif MH, Khan A, and Al-Harrasi A
- Subjects
- Humans, Genome, Bacterial, Computer Simulation, Bacterial Proteins genetics, Bacterial Proteins metabolism, Shigella flexneri drug effects, Shigella flexneri genetics, Drug Repositioning methods, Genomics methods, Serogroup, Anti-Bacterial Agents pharmacology, Dysentery, Bacillary drug therapy, Dysentery, Bacillary microbiology
- Abstract
Shigellosis, induced by Shigella flexneri, constitutes a significant health burden in developing nations, particularly impacting socioeconomically disadvantaged communities. Designated as the second most prevalent cause of diarrheal illness by the World Health Organization (WHO), it precipitates an estimated 212,000 fatalities annually. Within the spectrum of S. flexneri strains, serotype X is notably pervasive and resilient, yet its comprehensive characterization remains deficient. The present investigation endeavors to discern potential pharmacological targets and repurpose existing drug compounds against S. flexneri serotype X. Employing the framework of subtractive genomics, the study interrogates the reference genome of S. flexneri Serotype X (strain 2,002,017; UP000001884) to delineate its proteome into categories of non-homologous, non-paralogous, essential, virulent, and resistant constituents, thereby facilitating the identification of therapeutic targets. Subsequently, a screening of approximately 9000 compounds from the FDA library against the identified drug target aims to delineate efficacious agents for combating S. flexneri serotype X infections. The application of subtractive genomics methodology yields prognostic insights, unveiling non-paralogous proteins (n = 4122), non-homologues (n = 1803), essential (n = 1246), drug-like (n = 389), resistant (n = 167), alongside 42 virulent proteins within the reference proteome. This iterative process culminates in the identification of Serine O-acetyltransferase as a viable drug target. Subsequent virtual screening endeavors to unearth FDA-approved medicinal compounds capable of inhibiting Serine O-acetyltransferase. Noteworthy candidates such as DB12983, DB15085, DB16098, DB16185, and DB16262 emerge, exhibiting potential for mitigating S. flexneri Serotype X. Despite the auspicious findings, diligent scrutiny is imperative to ascertain the efficacy and safety profile of the proposed drug candidates vis-à-vis S. flexneri., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
46. Repurposing anti-cancer porphyrin derivative drugs to target SARS-CoV-2 envelope.
- Author
-
Mendonça DA, Cadima-Couto I, Buga CC, Arnaut ZA, Schaberle FA, Arnaut LG, Castanho MARB, and Cruz-Oliveira C
- Subjects
- Humans, Caco-2 Cells, COVID-19 Drug Treatment, Antineoplastic Agents pharmacology, Viral Envelope drug effects, Animals, Chlorocebus aethiops, Vero Cells, COVID-19 virology, Porphyrins pharmacology, SARS-CoV-2 drug effects, Drug Repositioning, Antiviral Agents pharmacology
- Abstract
Antiviral medicines to treat COVID-19 are still scarce. Porphyrins and porphyrin derivatives (PDs) usually present broad-spectrum antiviral activity with low risk of resistance development. In fact, some PDs are clinically approved to be used in anti-cancer photodynamic therapy and repurposing clinically approved PDs might be an alternative to treat COVID-19. Here, we characterize the ability of temoporfin, verteporfin, talaporfin and redaporfin to inactivate SARS-CoV-2 infectious particles. PDs light-dependent and -independent effect on SARS-CoV-2 infectivity were evaluated. PDs photoactivation successfully inactivated SARS-CoV-2 with very low concentrations and light dose. However, only temoporfin and verteporfin inactivated SARS-CoV-2 in the dark, being verteporfin the most effective. PDs treatment reduced viral load in infected Caco-2 cells, while not inducing cytotoxicity. Furthermore, light-independent treatment with temoporfin and verteporfin act on early stages of viral infection. Using lipid vehicles as membrane models, we characterized PDs interaction to the viral envelope. Verteporfin presented the lowest IC50 for viral inactivation and the highest partition coefficients (K
p ) towards lipid bilayers. Curiously, although temoporfin and redaporfin presented similar Kp s, redaporfin did not present light-independent antiviral activity, and only temoporfin and verteporfin caused lipid membrane disorder. In fact, redaporfin is located closer to the bilayer surface, while temoporfin and verteporfin are located closer to the centre. Our results suggest that viral envelope affinity, with penetration and destabilization of the lipid bilayer, seems critical to mediate PDs antiviral activity. Altogether, these findings open new avenues for the off-label application of temoporfin and verteporfin in the systemic treatment of COVID-19., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
47. DRTerHGAT: A drug repurposing method based on the ternary heterogeneous graph attention network.
- Author
-
He H, Xie J, Huang D, Zhang M, Zhao X, Ying Y, and Wang J
- Subjects
- Humans, Proteins chemistry, Algorithms, Alzheimer Disease drug therapy, Neural Networks, Computer, Computational Biology methods, Software, Drug Repositioning methods
- Abstract
Drug repurposing is an effective method to reduce the time and cost of drug development. Computational drug repurposing can quickly screen out the most likely associations from large biological databases to achieve effective drug repurposing. However, building a comprehensive model that integrates drugs, proteins, and diseases for drug repurposing remains challenging. This study proposes a drug repurposing method based on the ternary heterogeneous graph attention network (DRTerHGAT). DRTerHGAT designs a novel protein feature extraction process consisting of a large-scale protein language model and a multi-task autoencoder, so that protein features can be extracted accurately and efficiently from amino acid sequences. The ternary heterogeneous graph of drug-protein-disease comprehensively considering the relationships among the three types of nodes, including three homogeneous and three heterogeneous relationships. Based on the graph and the extracted protein features, the deep features of the drugs and the diseases are extracted by graph convolutional networks (GCN) and heterogeneous graph node attention networks (HGNA). In the experiments, DRTerHGAT is proven superior to existing advanced methods and DRTerHGAT variants. DRTerHGAT's powerful ability for drug repurposing is also demonstrated in Alzheimer's disease., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
48. The combination therapy using tyrosine kinase receptors inhibitors and repurposed drugs to target patient-derived glioblastoma stem cells.
- Author
-
Kucinska M, Pospieszna J, Tang J, Lisiak N, Toton E, Rubis B, and Murias M
- Subjects
- Humans, Cell Line, Tumor, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases metabolism, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Agents pharmacology, Cell Survival drug effects, Glioblastoma drug therapy, Glioblastoma pathology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Drug Repositioning methods, Protein Kinase Inhibitors pharmacology
- Abstract
The lesson from many studies investigating the efficacy of targeted therapy in glioblastoma (GBM) showed that a future perspective should be focused on combining multiple target treatments. Our research aimed to assess the efficacy of drug combinations against glioblastoma stem cells (GSCs). Patient-derived cells U3042, U3009, and U3039 were obtained from the Human Glioblastoma Cell Culture resource. Additionally, the study was conducted on a GBM commercial U251 cell line. Gene expression analysis related to receptor tyrosine kinases (RTKs), stem cell markers and genes associated with significant molecular targets was performed, and selected proteins encoded by these genes were assessed using the immunofluorescence and flow cytometry methods. The cytotoxicity studies were preceded by analyzing the expression of specific proteins that serve as targets for selected drugs. The cytotoxicity study using the MTS assay was conducted to evaluate the effects of selected drugs/candidates in monotherapy and combinations. The most cytotoxic compounds for U3042 cells were Disulfiram combined with Copper gluconate (DSF/Cu), Dacomitinib, and Foretinib with IC
50 values of 52.37 nM, 4.38 µM, and 4.54 µM after 24 h incubation, respectively. Interactions were assessed using SynergyFinder Plus software. The analysis enabled the identification of the most effective drug combinations against patient-derived GSCs. Our findings indicate that the most promising drug combinations are Dacomitinib and Foretinib, Dacomitinib and DSF/Cu, and Foretinib and AZD3759. Since most tested combinations have not been previously examined against glioblastoma stem-like cells, these results can shed new light on designing the therapeutic approach to target the GSC population., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
49. Novel drug targets and molecular mechanisms for sarcopenia based on systems biology.
- Author
-
Ceyhan AB, Ozcan M, Kim W, Li X, Altay O, Zhang C, and Mardinoglu A
- Subjects
- Humans, Aged, Animals, Gene Regulatory Networks drug effects, Male, Mice, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Female, Cell Line, Troglitazone, Molecular Targeted Therapy, Leupeptins pharmacology, Leupeptins therapeutic use, Systems Biology, Sarcopenia drug therapy, Sarcopenia metabolism, Sarcopenia genetics, Drug Repositioning methods
- Abstract
Sarcopenia is a major public health concern among older adults, leading to disabilities, falls, fractures, and mortality. This study aimed to elucidate the pathophysiological mechanisms of sarcopenia and identify potential therapeutic targets using systems biology approaches. RNA-seq data from muscle biopsies of 24 sarcopenic and 29 healthy individuals from a previous cohort were analysed. Differential expression, gene set enrichment, gene co-expression network, and topology analyses were conducted to identify target genes implicated in sarcopenia pathogenesis, resulting in the selection of 6 hub genes (PDHX, AGL, SEMA6C, CASQ1, MYORG, and CCDC69). A drug repurposing approach was then employed to identify new pharmacological treatment options for sarcopenia (clofibric-acid, troglitazone, withaferin-a, palbociclib, MG-132, bortezomib). Finally, validation experiments in muscle cell line (C2C12) revealed MG-132 and troglitazone as promising candidates for sarcopenia treatment. Our approach, based on systems biology and drug repositioning, provides insight into the molecular mechanisms of sarcopenia and offers potential new treatment options using existing drugs., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper, (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
50. Pathways for non-manufacturers to drive generic drug repurposing for cancer in the U.S.
- Author
-
Crittenden, Devon, Gallagher, Raquel, del Bosch, Fernanda Milans, Fox, David M., and Kleiman, Laura B.
- Subjects
OFF-label use (Drugs) ,DRUG approval ,GENERIC drugs ,DRUG repositioning ,DRUG accessibility ,DRUG labeling ,GENERIC drug manufacturing - Abstract
Repurposing generic drugs as new treatments for life-threatening diseases such as cancer is an exciting yet largely overlooked opportunity due to a lack of market-driven incentives. Nonprofit organizations and other non-manufacturers have been ramping up efforts to repurpose widely available generic drugs and rapidly expand affordable treatment options for patients. However, these nonmanufacturers find it difficult to obtain regulatory approval in the U.S. Without a straightforward path for approval and updating drug labeling, non-manufacturers have relied on off-label use of repurposed drugs. This limits the broad clinical adoption of these drugs and patient access. In this paper, we explore the regulatory landscape for repurposing of small molecule generic drugs within the U.S. We describe case studies of repurposed drugs that have been successfully incorporated into clinical treatment guidelines for cancer without regulatory approval. To encourage greater adoption of generic drugs in clinical practice-that is, to encourage the repurposing of these drugs-we examine existing Food and Drug Administration (FDA) pathways for approval of new uses or indications for generic drugs. We show how non-manufacturers, who are generally more active in generic drug repurposing than manufacturers, could utilize existing regulatory authorities and pathways, and we describe the challenges they face. We propose an extension of the existing 505(b)(2) new drug application (NDA) approval pathway, called a "labeling-only" 505(b)(2) NDA, that would enable non-manufacturers to seek approval of new indications for well-established small molecule drugs when multiple generic products are already available. It would not require new chemistry, manufacturing, and controls (CMC) data or introducing new drug products into the marketplace. This pathway would unlock innovation broadly and enable patients to benefit from the enormous potential of low-cost generic drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.