Drug Repositioning
Orabase-Formulated Benzalkonium Chloride Effectively Suppressed Oral Potentially Malignant Disorder In Vitro and In Vivo.
Orabase-Formulated Benzalkonium Chloride Effectively Suppressed Oral Potentially Malignant Disorder In Vitro and In Vivo.
ACS Omega. 2020 Mar 31;5(12):7018-7024
Authors: Wang YY, Xiao LY, Chen YK, Wu PC, Chen YH, Hu SC, Yuan SF
Abstract
Oral potentially malignant disorder (OPMD) is associated with an increased risk of progression to oral cancer. Patients with dysplastic changes of the precancerous lesions have a higher malignant transformation rate than those without dysplastic changes. Radiotherapy and surgery are the traditionally preferred choices for OPMD treatment. However, side effects caused by radiotherapy and surgery may reduce the willingness of patients to accept therapy. Therefore, developing an Orabase-formulated drug, which can be non-invasively administered, may provide an alternative treatment choice. To find, verify, and develop a new anti-cancer drug cost a lot of time and money, while drug repurposing can shorten both time and cost. In this study, we utilized high-throughput screening library to identify clinical drugs, which may have new bioactivities. Herein, we report that benzalkonium chloride (BAK), an antimicrobial preservative for pharmaceutical products, significantly induced reactive oxygen species production and cell death in oral precancerous cells. Additionally, our results showed that phosphorylation of STAT3 (Tyr705) and Akt (Ser473) were involved in cell death caused by BAK in DOK cells. According to animal studies, the development of DMBA-induced oral precancerous lesions was inhibited by 2% BAK. In conclusion, Orabase-formulated BAK may be a potential treatment for OPMD in the future.
PMID: 32258937 [PubMed - as supplied by publisher]
Identifying Small Molecule-miRNA Associations Based on Credible Negative Sample Selection and Random Walk.
Identifying Small Molecule-miRNA Associations Based on Credible Negative Sample Selection and Random Walk.
Front Bioeng Biotechnol. 2020;8:131
Authors: Liu F, Peng L, Tian G, Yang J, Chen H, Hu Q, Liu X, Zhou L
Abstract
Recently, many studies have demonstrated that microRNAs (miRNAs) are new small molecule drug targets. Identifying small molecule-miRNA associations (SMiRs) plays an important role in finding new clues for various human disease therapy. Wet experiments can discover credible SMiR associations; however, this is a costly and time-consuming process. Computational models have therefore been developed to uncover possible SMiR associations. In this study, we designed a new SMiR association prediction model, RWNS. RWNS integrates various biological information, credible negative sample selections, and random walk on a triple-layer heterogeneous network into a unified framework. It includes three procedures: similarity computation, negative sample selection, and SMiR association prediction based on random walk on the constructed small molecule-disease-miRNA association network. To evaluate the performance of RWNS, we used leave-one-out cross-validation (LOOCV) and 5-fold cross validation to compare RWNS with two state-of-the-art SMiR association methods, namely, TLHNSMMA and SMiR-NBI. Experimental results showed that RWNS obtained an AUC value of 0.9829 under LOOCV and 0.9916 under 5-fold cross validation on the SM2miR1 dataset, and it obtained an AUC value of 0.8938 under LOOCV and 0.9899 under 5-fold cross validation on the SM2miR2 dataset. More importantly, RWNS successfully captured 9, 17, and 37 SMiR associations validated by experiments among the predicted top 10, 20, and 50 SMiR candidates with the highest scores, respectively. We inferred that enoxacin and decitabine are associated with mir-21 and mir-155, respectively. Therefore, RWNS can be a powerful tool for SMiR association prediction.
PMID: 32258003 [PubMed - as supplied by publisher]
Clinical trials on drug repositioning for COVID-19 treatment.
Clinical trials on drug repositioning for COVID-19 treatment.
Rev Panam Salud Publica. 2020;44:e40
Authors: Rosa SGV, Santos WC
Abstract
The World Health Organization (WHO) was informed on December 2019 about a coronavirus pneumonia outbreak in Wuhan, Hubei province (China). Subsequently, on March 12, 2020, 125,048 cases and 4,614 deaths were reported. Coronavirus is an enveloped RNA virus, from the genus Betacoronavirus, that is distributed in birds, humans, and other mammals. WHO has named the novel coronavirus disease as COVID-19. More than 80 clinical trials have been launched to test coronavirus treatment, including some drug repurposing or repositioning for COVID-19. Hence, we performed a search in March 2020 of the clinicaltrials.gov database. The eligibility criteria for the retrieved studies were: contain a clinicaltrials.gov base identifier number; describe the number of participants and the period for the study; describe the participants' clinical conditions; and utilize interventions with medicines already studied or approved for any other disease in patients infected with the novel coronavirus SARS-CoV-2 (2019-nCoV). It is essential to emphasize that this article only captured trials listed in the clinicaltrials.gov database. We identified 24 clinical trials, involving more than 20 medicines, such as human immunoglobulin, interferons, chloroquine, hydroxychloroquine, arbidol, remdesivir, favipiravir, lopinavir, ritonavir, oseltamivir, methylprednisolone, bevacizumab, and traditional Chinese medicines (TCM). Although drug repurposing has some limitations, repositioning clinical trials may represent an attractive strategy because they facilitate the discovery of new classes of medicines; they have lower costs and take less time to reach the market; and there are existing pharmaceutical supply chains for formulation and distribution.
PMID: 32256547 [PubMed - as supplied by publisher]
On the Role of Artificial Intelligence in Genomics to Enhance Precision Medicine.
On the Role of Artificial Intelligence in Genomics to Enhance Precision Medicine.
Pharmgenomics Pers Med. 2020;13:105-119
Authors: Álvarez-Machancoses Ó, DeAndrés Galiana EJ, Cernea A, Fernández de la Viña J, Fernández-Martínez JL
Abstract
The complexity of orphan diseases, which are those that do not have an effective treatment, together with the high dimensionality of the genetic data used for their analysis and the high degree of uncertainty in the understanding of the mechanisms and genetic pathways which are involved in their development, motivate the use of advanced techniques of artificial intelligence and in-depth knowledge of molecular biology, which is crucial in order to find plausible solutions in drug design, including drug repositioning. Particularly, we show that the use of robust deep sampling methodologies of the altered genetics serves to obtain meaningful results and dramatically decreases the cost of research and development in drug design, influencing very positively the use of precision medicine and the outcomes in patients. The target-centric approach and the use of strong prior hypotheses that are not matched against reality (disease genetic data) are undoubtedly the cause of the high number of drug design failures and attrition rates. Sampling and prediction under uncertain conditions cannot be avoided in the development of precision medicine.
PMID: 32256101 [PubMed - as supplied by publisher]
Targeting DNA Repair in Ovarian Cancer Treatment Resistance.
Targeting DNA Repair in Ovarian Cancer Treatment Resistance.
Clin Oncol (R Coll Radiol). 2020 Apr 03;:
Authors: Wong-Brown MW, van der Westhuizen A, Bowden NA
Abstract
Most patients with advanced high-grade serous ovarian cancer (HGSOC) develop recurrent disease within 3 years and succumb to the disease within 5 years. Standard treatment for HGSOC is cytoreductive surgery followed by a combination of platinum (carboplatin or cisplatin) and taxol (paclitaxel) chemotherapies. Although initial recurrences are usually platinum-sensitive, patients eventually develop resistance to platinum-based chemotherapy. Accordingly, one of the major problems in the treatment of HGSOC and disease recurrence is the development of chemotherapy resistance. One of the causes of chemoresistance may be redundancies in the repair pathways involved in the response to DNA damage caused by chemotherapy. These pathways may be acting in parallel, where if the repair pathway that is responsible for triggering cell death after platinum chemotherapy therapy is deficient, an alternative repair pathway compensates and drives cancer cells to repair the damage, leading to chemotherapy resistance. In addition, if the repair pathways are epigenetically inactivated by DNA methylation, cell death may not be triggered, resulting in accumulation of mutations and DNA damage. There are novel and existing therapies that can drive DNA repair pathways towards sensitivity to platinum chemotherapy or targeted therapy, thus enabling treatment-resistant ovarian cancer to overcome chemotherapy resistance.
PMID: 32253106 [PubMed - as supplied by publisher]
Structural and molecular modeling studies reveal a new mechanism of action of chloroquine and hydroxychloroquine against SARS-CoV-2 infection.
Structural and molecular modeling studies reveal a new mechanism of action of chloroquine and hydroxychloroquine against SARS-CoV-2 infection.
Int J Antimicrob Agents. 2020 Apr 03;:105960
Authors: Fantini J, Scala CD, Chahinian H, Yahi N
Abstract
The recent emergence of the novel pathogenic SARS-coronavirus 2 (SARS-CoV-2) is responsible for a global pandemic. In face of the health emergency, drug repositioning is the most reliable option to design an efficient therapy for infected patients without delay. The first step of the viral replication cycle, i.e. the attachment to the surface of respiratory cells mediated by the spike (S) viral protein, offers several potential therapeutic targets. The S protein uses the ACE-2 receptor for entry, but also sialic acids linked to host cell surface gangliosides. Using a combination of structural and molecular modeling approaches, we showed that chloroquine (CLQ), one of the drugs currently under investigation for SARS-CoV-2 treatment, binds sialic acids and gangliosides with high affinity. We identified a new type of ganglioside-binding domain at the tip of the N-terminal domain of the SARS-CoV-2 spike (S) protein. This domain (aa 111-158), which is fully conserved among clinical isolates worldwide, may improve the attachment of the virus to lipid rafts and facilitate the contact with the ACE-2 receptor. We showed that in presence of CLQ (or of the more active derivative hydroxychloroquine, CLQ-OH), the viral spike is no longer able to bind gangliosides. The identification of this new mechanism of action of CLQ and CLQ-OH supports the use of these repositioned drugs to cure SARS-CoV-2 infected patients and stop the pandemic. Our in silico approaches might also be used to assess the efficiency of a broad range of repositioned and/or innovative drug candidates before their clinical evaluation.
PMID: 32251731 [PubMed - as supplied by publisher]
Cationic Amphiphilic Drugs Boost the Lysosomal Escape of Small Nucleic Acid Therapeutics in a Nanocarrier-Dependent Manner.
Cationic Amphiphilic Drugs Boost the Lysosomal Escape of Small Nucleic Acid Therapeutics in a Nanocarrier-Dependent Manner.
ACS Nano. 2020 Apr 06;:
Authors: Van de Vyver T, Bogaert B, De Backer L, Joris F, Guagliardo R, Van Hoeck J, Merckx P, Van Calenbergh S, Ramishetti S, Peer D, Remaut K, De Smedt SC, Raemdonck K
Abstract
Small nucleic acid (NA) therapeutics, such as small interfering RNA (siRNA), are generally formulated in nanoparticles (NPs) to overcome the multiple extra- and intracellular barriers upon in vivo administration. Interaction with target cells typically triggers endocytosis and sequesters the NPs in endosomes, thus hampering the pharmacological activity of the encapsulated siRNAs that occurs in the cytosol. Unfortunately, for most state-of-the-art NPs, endosomal escape is largely inefficient. As a result, the bulk of the endocytosed NA drug is rapidly trafficked towards the degradative lysosomes that are considered as a dead-end for siRNA nanomedicines. Opposed to this paradigm, we recently reported that cationic amphiphilic drugs (CADs) could strongly promote functional siRNA delivery from the endolysosomal compartment via transient induction of lysosomal membrane permeabilization. However, many questions still remain regarding the broader applicability of such a CAD adjuvant effect on NA delivery. Here, we report a drug repurposing screen (National Institutes of Health Clinical Collection) that allowed identification of 56 CAD adjuvants. We furthermore demonstrate that the CAD adjuvant effect is dependent on the type of nanocarrier, with NPs that generate an appropriate pool of decomplexed siRNA in the endolysosomal compartment being most susceptible to CAD-promoted gene silencing. Finally, the CAD adjuvant effect was verified on human ovarian cancer cells and for antisense oligonucleotides. In conclusion, this study strongly expands our current knowledge on how CADs increase the cytosolic release of small NAs, providing relevant insights to more rationally combine CAD adjuvants with NA-loaded NPs for future therapeutic applications.
PMID: 32250113 [PubMed - as supplied by publisher]
Computational studies of drug repurposing and synergism of lopinavir, oseltamivir and ritonavir binding with SARS-CoV-2 Protease against COVID-19.
Computational studies of drug repurposing and synergism of lopinavir, oseltamivir and ritonavir binding with SARS-CoV-2 Protease against COVID-19.
J Biomol Struct Dyn. 2020 Apr 06;:1-7
Authors: Muralidharan N, Sakthivel R, Velmurugan D, Gromiha MM
Abstract
A novel coronavirus (SARS-CoV-2) has caused a major outbreak in humans all over the world, and it is the latest pandemic in the series of other infectious diseases. The concept of drug repurposing has been used successfully for many years for known diseases. Considering the emergency and urgency, drug repurposing concept is being explored for coronavirus disease (COVID-19) as well. Recently, the combination of three known drugs, lopinavir, oseltamivir and ritonavir has been proposed to control the virulence to a great extent in COVID-19 affected patients within 48 hours. Hence, we tried to understand the effect of synergism of these drugs against the SARS-CoV-2 protease using sequential docking studies. As a result, combination of three drugs showed a better binding energy than that of individual drugs. Further, the complex was subjected to molecular dynamics simulations to get insights into the stability of the complex, considering the simultaneous interactions between three drugs and the protein. The protein complexed with three drugs remained stable during the simulations. Hence, these drugs can be explored further for drug repurposing against the successful inhibition of COVID-19.
PMID: 32248766 [PubMed - as supplied by publisher]
The Treatment of Impaired Wound Healing in Diabetes: Looking among Old Drugs.
The Treatment of Impaired Wound Healing in Diabetes: Looking among Old Drugs.
Pharmaceuticals (Basel). 2020 Apr 01;13(4):
Authors: Spampinato SF, Caruso GI, De Pasquale R, Sortino MA, Merlo S
Abstract
Chronic wounds often occur in patients with diabetes mellitus due to the impairment of wound healing. This has negative consequences for both the patient and the medical system and considering the growing prevalence of diabetes, it will be a significant medical, social, and economic burden in the near future. Hence, the need for therapeutic alternatives to the current available treatments that, although various, do not guarantee a rapid and definite reparative process, appears necessary. We here analyzed current treatments for wound healing, but mainly focused the attention on few classes of drugs that are already in the market with different indications, but that have shown in preclinical and few clinical trials the potentiality to be used in the treatment of impaired wound healing. In particular, repurposing of the antiglycemic agents dipeptidylpeptidase 4 (DPP4) inhibitors and metformin, but also, statins and phenyotin have been analyzed. All show encouraging results in the treatment of chronic wounds, but additional, well designed studies are needed to allow these drugs access to the clinics in the therapy of impaired wound healing.
PMID: 32244718 [PubMed - as supplied by publisher]
Pentamidine sensitizes FDA-approved non-antibiotics for the inhibition of multidrug-resistant Gram-negative pathogens.
Pentamidine sensitizes FDA-approved non-antibiotics for the inhibition of multidrug-resistant Gram-negative pathogens.
Eur J Clin Microbiol Infect Dis. 2020 Apr 02;:
Authors: Wu C, Xia L, Huang W, Xu Y, Gu Y, Liu C, Ji L, Li W, Wu Y, Zhou K, Feng X
Abstract
Pentamidine sensitizes FDA-approved antibiotics to combat Gram-negative pathogens. We screened 1374 FDA-approved non-antibiotics for their ability to be sensitized by pentamidine against Escherichia coli. We identified mitomycin C and mefloquine as potent hits effective against multiple drug-resistant, Gram-negative bacteria. Killing kinetics and an in vivo model with Caenorhabditis elegans (C. elegans) revealed that such combinations produced synergy against colistin-resistant Enterobacter cloacae (E. cloacae). These findings suggest combinations of FDA-approved non-antibiotics, and pentamidine can be repurposed into new antimicrobial agents.
PMID: 32242313 [PubMed - as supplied by publisher]
Drug repurposing in rare diseases: Myths and reality.
Drug repurposing in rare diseases: Myths and reality.
Therapie. 2020 Feb 13;:
Authors: Fetro C, Scherman D
Abstract
While nearly 8000 rare diseases have been identified, only 5 percent have licensed treatments. As most of these diseases are life threatening, it underscores the urgent need for new drugs. Drug repurposing (also called drug repositioning) consists in identifying new uses for approved or investigational drugs that are outside the scope of the original medical indication. It represents an opportunity for rare diseases and patients with unmet needs. It is an alternative option in drug development and is often presented as being a viable, risk-managed strategy for pharmaceutical companies developing orphan drugs. Drug repurposing is presented as offering various advantages over developing an entirely new drug for a given indication: fewer risks, lower costs and shorter timelines. However, matters are not as simple as this. There are notable successes for drug repurposing. Nevertheless, repurposing does not always succeed. The repurposed drug may fail to demonstrate a benefits-harms balance in clinical trials. Moreover, there are legal and regulatory issues which are specific barriers to drug repurposing and which have to be carefully analyzed before any development of repurposed drugs. The objective of this article is to identify major challenges and opportunities of drug repurposing in rare diseases and to separate fact from fiction.
PMID: 32241561 [PubMed - as supplied by publisher]
"drug repositioning" OR "drug repurposing"; +7 new citations
7 new pubmed citations were retrieved for your search. Click on the search hyperlink below to display the complete search results:
"drug repositioning" OR "drug repurposing"
These pubmed results were generated on 2020/04/03
PubMed comprises more than millions of citations for biomedical literature from MEDLINE, life science journals, and online books. Citations may include links to full-text content from PubMed Central and publisher web sites.
Calcium Channels as Novel Therapeutic Targets for Ovarian Cancer Stem Cells.
Calcium Channels as Novel Therapeutic Targets for Ovarian Cancer Stem Cells.
Int J Mol Sci. 2020 Mar 27;21(7):
Authors: Lee H, Kim JW, Kim DK, Choi DK, Lee S, Yu JH, Kwon OB, Lee J, Lee DS, Kim JH, Min SH
Abstract
Drug resistance in epithelial ovarian cancer (EOC) is reportedly attributed to the existence of cancer stem cells (CSC), because in most cancers, CSCs still remain after chemotherapy. To overcome this limitation, novel therapeutic strategies are required to prevent cancer recurrence and chemotherapy-resistant cancers by targeting cancer stem cells (CSCs). We screened an FDA-approved compound library and found four voltage-gated calcium channel blockers (manidipine, lacidipine, benidipine, and lomerizine) that target ovarian CSCs. Four calcium channel blockers (CCBs) decreased sphere formation, viability, and proliferation, and induced apoptosis in ovarian CSCs. CCBs destroyed stemness and inhibited the AKT and ERK signaling pathway in ovarian CSCs. Among calcium channel subunit genes, three L- and T-type calcium channel genes were overexpressed in ovarian CSCs, and downregulation of calcium channel genes reduced the stem-cell-like properties of ovarian CSCs. Expressions of these three genes are negatively correlated with the survival rate of patient groups. In combination therapy with cisplatin, synergistic effect was shown in inhibiting the viability and proliferation of ovarian CSCs. Moreover, combinatorial usage of manidipine and paclitaxel showed enhanced effect in ovarian CSCs xenograft mouse models. Our results suggested that four CCBs may be potential therapeutic drugs for preventing ovarian cancer recurrence.
PMID: 32230901 [PubMed - in process]
COVID-19 and chronological aging: senolytics and other anti-aging drugs for the treatment or prevention of corona virus infection?
COVID-19 and chronological aging: senolytics and other anti-aging drugs for the treatment or prevention of corona virus infection?
Aging (Albany NY). 2020 Mar 30;:
Authors: Sargiacomo C, Sotgia F, Lisanti MP
Abstract
COVID-19, also known as SARS-CoV-2, is a new emerging zoonotic corona virus of the SARS (Severe Acute Respiratory Syndrome) and the MERS (Middle East Respiratory Syndrome) family. COVID-19 originated in China and spread world-wide, resulting in the pandemic of 2020. For some reason, COVID-19 shows a considerably higher mortality rate in patients with advanced chronological age. This begs the question as to whether there is a functional association between COVID-19 infection and the process of chronological aging. Two host receptors have been proposed for COVID-19. One is CD26 and the other is ACE-2 (angiotensin-converting enzyme 2). Interestingly, both CD26 and the angiotensin system show associations with senescence. Similarly, two proposed therapeutics for the treatment of COVID-19 infection are Azithromycin and Quercetin, both drugs with significant senolytic activity. Also, Chloroquine-related compounds inhibit the induction of the well-known senescence marker, Beta-galactosidase. Other anti-aging drugs should also be considered, such as Rapamycin and Doxycycline, as they behave as inhibitors of protein synthesis, blocking both SASP and viral replication. Therefore, we wish to speculate that the fight against COVID-19 disease should involve testing the hypothesis that senolytics and other anti-aging drugs may have a prominent role in preventing the transmission of the virus, as well as aid in its treatment. Thus, we propose that new clinical trials may be warranted, as several senolytic and anti-aging therapeutics are existing FDA-approved drugs, with excellent safety profiles, and would be readily available for drug repurposing efforts. As Azithromycin and Doxycycline are both commonly used antibiotics that inhibit viral replication and IL-6 production, we may want to consider this general class of antibiotics that functionally inhibits cellular protein synthesis as a side-effect, for the treatment and prevention of COVID-19 disease.
PMID: 32229706 [PubMed - as supplied by publisher]
Actinomycin D Downregulates Sox2 and Improves Survival in Preclinical Models of Recurrent Glioblastoma.
Actinomycin D Downregulates Sox2 and Improves Survival in Preclinical Models of Recurrent Glioblastoma.
Neuro Oncol. 2020 Mar 30;:
Authors: Taylor JT, Ellison S, Pandele A, Wood S, Nathan E, Forte G, Parker H, Zindy E, Elvin M, Dickson A, Williams KJ, Karabatsou K, McCabe M, McBain C, Bigger BW
Abstract
BACKGROUND: Glioblastoma (GBM) has been extensively researched over the last few decades, yet despite aggressive multi-modal treatment, recurrence is inevitable and second-line treatment options are limited. Here, we demonstrate how high throughput screening (HTS) in multicellular spheroids can generate physiologically relevant patient chemosensitivity data using patient-derived cells in a rapid and cost-effective manner. Our HTS system identified ACTD to be highly cytotoxic over a panel of twelve patient-derived glioma stem-like cell lines (GSCs). Actinomycin D (ACTD) is antineoplastic antibiotic used in the treatment of childhood cancers. Here, we validate ACTD as a potential repurposed therapeutic for glioblastoma in three-dimensional GSC cultures and patient-derived xenograft models of recurrent glioblastoma.
METHODS: Twelve patient-derived GSCs were screened at 10µM, as multicellular spheroids, in a 384-well serum-free assay with 133 FDA-approved compounds. GSCs were then treated in vitro with ACTD at established IC50 concentrations. Downregulation of Sox2, a stem-cell transcription factor, was investigated via western blot and through immunohistological assessment of murine brain tissue.
RESULTS: Treatment with ACTD was shown to significantly reduce tumor growth in two recurrent GBM (rGBM) patient-derived models and significantly increased survival. ACTD is also shown to specifically downregulate the expression of Sox2 both in vitro and in vivo.
CONCLUSION: These findings indicate that, as predicted by our HTS, ACTD could deplete the cancer stem cell population within the tumor mass, ultimately leading to a delay in tumor progression.
PMID: 32227096 [PubMed - as supplied by publisher]
High-Throughput Screening of the ReFRAME Library Identifies Potential Drug Repurposing Candidates for Trypanosoma cruzi.
High-Throughput Screening of the ReFRAME Library Identifies Potential Drug Repurposing Candidates for Trypanosoma cruzi.
Microorganisms. 2020 Mar 26;8(4):
Authors: Bernatchez JA, Chen E, Hull MV, McNamara CW, McKerrow JH, Siqueira-Neto JL
Abstract
Chagas disease, caused by the kinetoplastid parasite Trypanosoma cruzi, affects between 6 and 7 million people worldwide, with an estimated 300,000 to 1 million of these cases in the United States. In the chronic phase of infection, T. cruzi can cause severe gastrointestinal and cardiac disease, which can be fatal. Currently, only benznidazole is clinically approved by the FDA for pediatric use to treat this infection in the USA. Toxicity associated with this compound has driven the search for new anti-Chagas agents. Drug repurposing is a particularly attractive strategy for neglected diseases, as pharmacological parameters and toxicity are already known for these compounds, reducing costs and saving time in the drug development pipeline. Here, we screened 7680 compounds from the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library, a collection of drugs or compounds with confirmed clinical safety, against T. cruzi. We identified seven compounds of interest with potent in vitro activity against the parasite with a therapeutic index of 10 or greater, including the previously unreported activity of the antiherpetic compound 348U87. These results provide the framework for further development of new T. cruzi leads that can potentially move quickly to the clinic.
PMID: 32224991 [PubMed]
[Multi-targeting drugs: past, present and future].
[Multi-targeting drugs: past, present and future].
Orv Hetil. 2020 Apr;161(14):523-531
Authors: Mátyus P
Abstract
The one disease - one target - one drug paradigm was an almost dominant principle in drug discovery from the 1960s to the 2000s. The stagnation and even decline in the productivity of drug innovation around the turn of the millennium and beyond, the realization of limitations of the one-target approach, especially in the treatment of multifactorial diseases, have drawn attention considerably to the one disease - multiple target - one drug multi-targeting drug concept. In this review, we outline old and new molecular design strategies and their practical implementation with own and other examples that also demonstrate unique therapeutic and diagnostic values and benefits of the multi-targeting approach. Finally, we point out that the full potential of the multi-targeting concept can emerge through data analytics and association methods (such as artificial intelligence) and system-based approach, preferably by linking it to quantitative systems pharmacology. This new systems pharmacology drug approach may also lead to novel breakthrough drugs, drug combinations and drug repositioning. Orv Hetil. 2020; 161(14): 523-531.
PMID: 32223419 [PubMed - in process]
Drug vector representation: a tool for drug similarity analysis.
Drug vector representation: a tool for drug similarity analysis.
Mol Genet Genomics. 2020 Mar 28;:
Authors: Lin L, Wan L, He H, Liu W
Abstract
DrugMatrix is a valuable toxicogenomic dataset, which provides in vivo transcriptome data corresponding to hundreds of chemical drugs. However, the relationships between drugs and how those drugs affect the biological process are still unknown. The high dimensionality of the microarray data hinders its application. The aims of this study are to (1) represent the transcriptome data by lower-dimensional vectors, (2) compare drug similarity, (3) represent drug combinations by adding vectors and (4) infer drug mechanism of action (MoA) and genotoxicity features. We borrowed the latent semantic analysis (LSA) technique from natural language processing to represent treatments (drugs with multiple concentrations and time points) by dense vectors, each dimension of which is an orthogonal biological feature. The gProfiler enrichment tool was used for the 100-dimensional vector feature annotation. The similarity between treatments vectors was calculated by the cosine function. Adding vectors may represent drug combinations, treatment times or treatment doses that are not presented in the original data. Drug-drug interaction pairs had a higher similarity than random drug pairs in the hepatocyte data. The vector features helped to reveal the MoA. Differential feature expression was also implicated for genotoxic and non-genotoxic carcinogens. An easy-to-use Web tool was developed by Shiny Web application framework for the exploration of treatment similarities and drug combinations (https://bioinformatics.fafu.edu.cn/drugmatrix/). We represented treatments by vectors and provided a tool that is useful for hypothesis generation in toxicogenomic, such as drug similarity, drug repurposing, combination therapy and MoA.
PMID: 32222838 [PubMed - as supplied by publisher]
Ribavirin, Remdesivir, Sofosbuvir, Galidesivir, and Tenofovir against SARS-CoV-2 RNA dependent RNA polymerase (RdRp): A molecular docking study.
Ribavirin, Remdesivir, Sofosbuvir, Galidesivir, and Tenofovir against SARS-CoV-2 RNA dependent RNA polymerase (RdRp): A molecular docking study.
Life Sci. 2020 Mar 25;:117592
Authors: Elfiky AA
Abstract
AIMS: A new human coronavirus (HCoV), which has been designated SARS-CoV-2, began spreading in December 2019 in Wuhan City, China causing pneumonia called COVID-19. The spread of SARS-CoV-2 has been faster than any other coronaviruses that have succeeded in crossing the animal-human barrier. There is concern that this new virus will spread around the world as did the previous two HCoVs-Severe Acute Respiratory Syndrome (SARS) and Middle East Respiratory Syndrome (MERS)-each of which caused approximately 800 deaths in the years 2002 and 2012, respectively. Thus far, 11,268 deaths have been reported from the 258,842 confirmed infections in 168 countries.
MAIN METHODS: In this study, the RNA-dependent RNA polymerase (RdRp) of the newly emerged coronavirus is modeled, validated, and then targeted using different anti-polymerase drugs currently on the market that have been approved for use against various viruses.
KEY FINDINGS: The results suggest the effectiveness of Ribavirin, Remdesivir, Sofosbuvir, Galidesivir, and Tenofovir as potent drugs against SARS-CoV-2 since they tightly bind to its RdRp. In addition, the results suggest guanosine derivative (IDX-184), Setrobuvir, and YAK as top seeds for antiviral treatments with high potential to fight the SARS-CoV-2 strain specifically.
SIGNIFICANCE: The availability of FDA-approved anti-RdRp drugs can help treat patients and reduce the danger of the mysterious new viral infection COVID-19. The drugs mentioned above can tightly bind to the RdRp of the SARS-CoV-2 strain and thus may be used to treat the disease. No toxicity measurements are required for these drugs since they were previously tested prior to their approval by the FDA.
PMID: 32222463 [PubMed - as supplied by publisher]
Effects of 5-HT2C receptor modulation and the NA reuptake inhibitor atomoxetine in tests of compulsive and impulsive behaviour.
Effects of 5-HT2C receptor modulation and the NA reuptake inhibitor atomoxetine in tests of compulsive and impulsive behaviour.
Neuropharmacology. 2020 Mar 25;:108064
Authors: Higgins GA, Brown M, St John J, MacMillan C, Silenieks LB, Thevarkunnel S
Abstract
Drug repositioning has gained strategic value as a reaction to high attrition rates of new drugs as they pass through the clinical development process. The 5-HT2C receptor agonist lorcaserin (Belviq®), and the selective NA reuptake inhibitor atomoxetine (Strattera®) represent two drugs FDA approved for obesity and ADHD respectively. Although both drugs are of differing pharmacological class, each share a property of regulating impulsive behaviours in preclinical studies, and thus represent candidates for consideration in clinical conditions labelled as 'impulsive-compulsive disorders'. The present studies investigated both drugs, as well as the highly selective 5-HT2C agonist CP-809101 in two tests of compulsive action: schedule-induced polydipsia (SIP) and increased perseverative [PSV] (and premature [PREM]) responses emitted during an extended ITI 5-choice task. While lorcaserin (0.06-0.6 mg/kg), CP-809101 (0.1-1 mg/kg) and atomoxetine (0.1-1 mg/kg) each reduced both PREM and PSV measures in the 5-choice task, at equivalent doses only lorcaserin and CP-809101 affected excessive water intake in the SIP task, atomoxetine (0.1-2 mg/kg) was essentially ineffective. Further evidence supporting a role of the 5-HT2C receptor as an important regulator of impulsive-compulsive behaviours, the selective antagonist SB-242084 produced the opposing effects to lorcaserin, i.e promoting both impulsive and compulsive behaviours. The profile of atomoxetine may suggest differences in the nature of compulsive action measured either as non-regulatory drinking in the SIP task, and PSV responses made in a 5-choice task. These studies support the consideration of 5-HT2C receptor agonists, typified by lorcaserin, and atomoxetine as potential treatments for clinical conditions categorised as 'impulsive-compulsive disorders'.
PMID: 32222404 [PubMed - as supplied by publisher]